WO2024130119A2 - Synbiotic compositions for short chain fatty acid production - Google Patents
Synbiotic compositions for short chain fatty acid production Download PDFInfo
- Publication number
- WO2024130119A2 WO2024130119A2 PCT/US2023/084290 US2023084290W WO2024130119A2 WO 2024130119 A2 WO2024130119 A2 WO 2024130119A2 US 2023084290 W US2023084290 W US 2023084290W WO 2024130119 A2 WO2024130119 A2 WO 2024130119A2
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- lacto
- propionate
- human milk
- producing bacterium
- strain
- Prior art date
Links
- 239000000203 mixture Substances 0.000 title claims abstract description 474
- 238000004519 manufacturing process Methods 0.000 title claims abstract description 49
- 150000004666 short chain fatty acids Chemical class 0.000 title abstract description 9
- 235000019722 synbiotics Nutrition 0.000 title description 5
- 235000013406 prebiotics Nutrition 0.000 claims abstract description 337
- XBDQKXXYIPTUBI-UHFFFAOYSA-M Propionate Chemical compound CCC([O-])=O XBDQKXXYIPTUBI-UHFFFAOYSA-M 0.000 claims abstract description 333
- 241000894006 Bacteria Species 0.000 claims abstract description 329
- 235000020256 human milk Nutrition 0.000 claims abstract description 299
- 210000004251 human milk Anatomy 0.000 claims abstract description 299
- 229920001542 oligosaccharide Polymers 0.000 claims abstract description 259
- 150000002482 oligosaccharides Chemical class 0.000 claims abstract description 257
- 238000000034 method Methods 0.000 claims abstract description 160
- 241000186015 Bifidobacterium longum subsp. infantis Species 0.000 claims abstract description 129
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 claims abstract description 124
- 244000005700 microbiome Species 0.000 claims abstract description 76
- 208000035475 disorder Diseases 0.000 claims abstract description 55
- 230000000968 intestinal effect Effects 0.000 claims abstract description 46
- 208000015181 infectious disease Diseases 0.000 claims abstract description 45
- 230000007140 dysbiosis Effects 0.000 claims abstract description 37
- 208000027244 Dysbiosis Diseases 0.000 claims abstract description 36
- 208000026935 allergic disease Diseases 0.000 claims abstract description 31
- 206010020751 Hypersensitivity Diseases 0.000 claims abstract description 29
- 230000007815 allergy Effects 0.000 claims abstract description 28
- 206010061218 Inflammation Diseases 0.000 claims abstract description 23
- 230000004054 inflammatory process Effects 0.000 claims abstract description 23
- 208000026278 immune system disease Diseases 0.000 claims abstract description 20
- 241000186000 Bifidobacterium Species 0.000 claims description 243
- 239000012466 permeate Substances 0.000 claims description 92
- 201000010099 disease Diseases 0.000 claims description 69
- FERIUCNNQQJTOY-UHFFFAOYSA-M Butyrate Chemical compound CCCC([O-])=O FERIUCNNQQJTOY-UHFFFAOYSA-M 0.000 claims description 61
- FERIUCNNQQJTOY-UHFFFAOYSA-N Butyric acid Natural products CCCC(O)=O FERIUCNNQQJTOY-UHFFFAOYSA-N 0.000 claims description 61
- 208000024891 symptom Diseases 0.000 claims description 47
- 241001148134 Veillonella Species 0.000 claims description 45
- 241000282414 Homo sapiens Species 0.000 claims description 33
- 241000894007 species Species 0.000 claims description 33
- HWHQUWQCBPAQQH-BWRPKUOHSA-N 2-fucosyllactose Chemical compound O[C@H]1[C@H](O)[C@H](O)[C@H](C)O[C@H]1O[C@@H]1[C@@H](O)[C@@H](O)[C@@H](CO)O[C@H]1O[C@H]([C@H](O)CO)[C@H](O)[C@@H](O)C=O HWHQUWQCBPAQQH-BWRPKUOHSA-N 0.000 claims description 30
- 241001148135 Veillonella parvula Species 0.000 claims description 30
- 239000008194 pharmaceutical composition Substances 0.000 claims description 30
- 229940062827 2'-fucosyllactose Drugs 0.000 claims description 27
- HWHQUWQCBPAQQH-UHFFFAOYSA-N 2-O-alpha-L-Fucosyl-lactose Natural products OC1C(O)C(O)C(C)OC1OC1C(O)C(O)C(CO)OC1OC(C(O)CO)C(O)C(O)C=O HWHQUWQCBPAQQH-UHFFFAOYSA-N 0.000 claims description 27
- AUNPEJDACLEKSC-ZAYDSPBTSA-N 3-fucosyllactose Chemical compound O[C@H]1[C@H](O)[C@H](O)[C@H](C)O[C@H]1O[C@@H]1[C@@H](O)[C@H](O[C@@H]2[C@H](OC(O)[C@H](O)[C@H]2O)CO)O[C@H](CO)[C@@H]1O AUNPEJDACLEKSC-ZAYDSPBTSA-N 0.000 claims description 27
- SNFSYLYCDAVZGP-UHFFFAOYSA-N UNPD26986 Natural products OC1C(O)C(O)C(C)OC1OC1C(OC2C(OC(O)C(O)C2O)CO)OC(CO)C(O)C1O SNFSYLYCDAVZGP-UHFFFAOYSA-N 0.000 claims description 27
- 208000001395 Acute radiation syndrome Diseases 0.000 claims description 26
- 241000588724 Escherichia coli Species 0.000 claims description 26
- 206010068142 Radiation sickness syndrome Diseases 0.000 claims description 26
- TYALNJQZQRNQNQ-UHFFFAOYSA-N #alpha;2,6-sialyllactose Natural products O1C(C(O)C(O)CO)C(NC(=O)C)C(O)CC1(C(O)=O)OCC1C(O)C(O)C(O)C(OC2C(C(O)C(O)OC2CO)O)O1 TYALNJQZQRNQNQ-UHFFFAOYSA-N 0.000 claims description 24
- AXQLFFDZXPOFPO-UHFFFAOYSA-N UNPD216 Natural products O1C(CO)C(O)C(OC2C(C(O)C(O)C(CO)O2)O)C(NC(=O)C)C1OC(C1O)C(O)C(CO)OC1OC1C(O)C(O)C(O)OC1CO AXQLFFDZXPOFPO-UHFFFAOYSA-N 0.000 claims description 24
- TYALNJQZQRNQNQ-JLYOMPFMSA-N alpha-Neup5Ac-(2->6)-beta-D-Galp-(1->4)-beta-D-Glcp Chemical compound O1[C@@H]([C@H](O)[C@H](O)CO)[C@H](NC(=O)C)[C@@H](O)C[C@@]1(C(O)=O)OC[C@@H]1[C@H](O)[C@H](O)[C@@H](O)[C@H](O[C@H]2[C@@H]([C@@H](O)[C@H](O)O[C@@H]2CO)O)O1 TYALNJQZQRNQNQ-JLYOMPFMSA-N 0.000 claims description 24
- AXQLFFDZXPOFPO-UNTPKZLMSA-N beta-D-Galp-(1->3)-beta-D-GlcpNAc-(1->3)-beta-D-Galp-(1->4)-beta-D-Glcp Chemical compound O([C@@H]1O[C@H](CO)[C@H](O)[C@@H]([C@H]1O)O[C@H]1[C@@H]([C@H]([C@H](O)[C@@H](CO)O1)O[C@H]1[C@@H]([C@@H](O)[C@@H](O)[C@@H](CO)O1)O)NC(=O)C)[C@H]1[C@H](O)[C@@H](O)[C@H](O)O[C@@H]1CO AXQLFFDZXPOFPO-UNTPKZLMSA-N 0.000 claims description 24
- USIPEGYTBGEPJN-UHFFFAOYSA-N lacto-N-tetraose Natural products O1C(CO)C(O)C(OC2C(C(O)C(O)C(CO)O2)O)C(NC(=O)C)C1OC1C(O)C(CO)OC(OC(C(O)CO)C(O)C(O)C=O)C1O USIPEGYTBGEPJN-UHFFFAOYSA-N 0.000 claims description 24
- 241000935148 Veillonella infantium Species 0.000 claims description 23
- CILYIEBUXJIHCO-UHFFFAOYSA-N 102778-91-6 Natural products O1C(C(O)C(O)CO)C(NC(=O)C)C(O)CC1(C(O)=O)OC1C(O)C(OC2C(C(O)C(O)OC2CO)O)OC(CO)C1O CILYIEBUXJIHCO-UHFFFAOYSA-N 0.000 claims description 21
- DVGKRPYUFRZAQW-UHFFFAOYSA-N 3 prime Natural products CC(=O)NC1OC(CC(O)C1C(O)C(O)CO)(OC2C(O)C(CO)OC(OC3C(O)C(O)C(O)OC3CO)C2O)C(=O)O DVGKRPYUFRZAQW-UHFFFAOYSA-N 0.000 claims description 21
- LKOHREGGXUJGKC-UHFFFAOYSA-N Lactodifucotetraose Natural products OC1C(O)C(O)C(C)OC1OC1C(OC2C(C(O)C(O)OC2CO)OC2C(C(O)C(O)C(C)O2)O)OC(CO)C(O)C1O LKOHREGGXUJGKC-UHFFFAOYSA-N 0.000 claims description 21
- CILYIEBUXJIHCO-UITFWXMXSA-N N-acetyl-alpha-neuraminyl-(2->3)-beta-D-galactosyl-(1->4)-beta-D-glucose Chemical compound O1[C@@H]([C@H](O)[C@H](O)CO)[C@H](NC(=O)C)[C@@H](O)C[C@@]1(C(O)=O)O[C@@H]1[C@@H](O)[C@H](O[C@H]2[C@@H]([C@@H](O)[C@H](O)O[C@@H]2CO)O)O[C@H](CO)[C@@H]1O CILYIEBUXJIHCO-UITFWXMXSA-N 0.000 claims description 21
- OIZGSVFYNBZVIK-UHFFFAOYSA-N N-acetylneuraminosyl-D-lactose Natural products O1C(C(O)C(O)CO)C(NC(=O)C)C(O)CC1(C(O)=O)OC1C(O)C(OC(C(O)CO)C(O)C(O)C=O)OC(CO)C1O OIZGSVFYNBZVIK-UHFFFAOYSA-N 0.000 claims description 21
- 241001341704 Veillonella rogosae Species 0.000 claims description 21
- FZIVHOUANIQOMU-YIHIYSSUSA-N alpha-L-Fucp-(1->2)-beta-D-Galp-(1->3)-beta-D-GlcpNAc-(1->3)-beta-D-Galp-(1->4)-D-Glcp Chemical compound O[C@H]1[C@H](O)[C@H](O)[C@H](C)O[C@H]1O[C@H]1[C@H](O[C@@H]2[C@H]([C@H](O[C@@H]3[C@H]([C@H](O[C@@H]4[C@H](OC(O)[C@H](O)[C@H]4O)CO)O[C@H](CO)[C@@H]3O)O)O[C@H](CO)[C@H]2O)NC(C)=O)O[C@H](CO)[C@H](O)[C@@H]1O FZIVHOUANIQOMU-YIHIYSSUSA-N 0.000 claims description 21
- 230000003115 biocidal effect Effects 0.000 claims description 21
- 239000003814 drug Substances 0.000 claims description 21
- FZIVHOUANIQOMU-UHFFFAOYSA-N lacto-N-fucopentaose I Natural products OC1C(O)C(O)C(C)OC1OC1C(OC2C(C(OC3C(C(OC4C(OC(O)C(O)C4O)CO)OC(CO)C3O)O)OC(CO)C2O)NC(C)=O)OC(CO)C(O)C1O FZIVHOUANIQOMU-UHFFFAOYSA-N 0.000 claims description 21
- 208000009329 Graft vs Host Disease Diseases 0.000 claims description 19
- 208000002389 Pouchitis Diseases 0.000 claims description 19
- 208000024908 graft versus host disease Diseases 0.000 claims description 19
- WJPIUUDKRHCAEL-UHFFFAOYSA-N 3FL Natural products OC1C(O)C(O)C(C)OC1OC1C(OC2C(C(O)C(O)C(CO)O2)O)C(CO)OC(O)C1O WJPIUUDKRHCAEL-UHFFFAOYSA-N 0.000 claims description 18
- 206010012735 Diarrhoea Diseases 0.000 claims description 18
- 241001533204 Veillonella dispar Species 0.000 claims description 18
- RQNFGIWYOACERD-OCQMRBNYSA-N alpha-L-Fucp-(1->4)-[alpha-L-Fucp-(1->2)-beta-D-Galp-(1->3)]-beta-D-GlcpNAc-(1->3)-beta-D-Galp-(1->4)-D-Glcp Chemical compound O[C@H]1[C@H](O)[C@H](O)[C@H](C)O[C@H]1O[C@H]1[C@H](O[C@H]2[C@@H]([C@@H](CO)O[C@@H](O[C@@H]3[C@H]([C@H](O[C@@H]4[C@H](OC(O)[C@H](O)[C@H]4O)CO)O[C@H](CO)[C@@H]3O)O)[C@@H]2NC(C)=O)O[C@H]2[C@H]([C@H](O)[C@H](O)[C@H](C)O2)O)O[C@H](CO)[C@H](O)[C@@H]1O RQNFGIWYOACERD-OCQMRBNYSA-N 0.000 claims description 18
- FCIROHDMPFOSFG-LAVSNGQLSA-N disialyllacto-N-tetraose Chemical compound O1[C@@H]([C@H](O)[C@H](O)CO)[C@H](NC(=O)C)[C@@H](O)C[C@@]1(C(O)=O)OC[C@@H]1[C@@H](O)[C@H](O[C@H]2[C@@H]([C@@H](O[C@]3(O[C@H]([C@H](NC(C)=O)[C@@H](O)C3)[C@H](O)[C@H](O)CO)C(O)=O)[C@@H](O)[C@@H](CO)O2)O)[C@@H](NC(C)=O)[C@H](O[C@@H]2[C@H]([C@H](O[C@H]3[C@@H]([C@@H](O)C(O)O[C@@H]3CO)O)O[C@H](CO)[C@@H]2O)O)O1 FCIROHDMPFOSFG-LAVSNGQLSA-N 0.000 claims description 18
- 229940079593 drug Drugs 0.000 claims description 18
- RQNFGIWYOACERD-UHFFFAOYSA-N lacto-N-Difucosylhexaose I Natural products OC1C(O)C(O)C(C)OC1OC1C(OC2C(C(CO)OC(OC3C(C(OC4C(OC(O)C(O)C4O)CO)OC(CO)C3O)O)C2NC(C)=O)OC2C(C(O)C(O)C(C)O2)O)OC(CO)C(O)C1O RQNFGIWYOACERD-UHFFFAOYSA-N 0.000 claims description 18
- OQIUPKPUOLIHHS-UHFFFAOYSA-N lacto-N-difucohexaose I Natural products OC1C(O)C(O)C(C)OC1OC1C(OC2C(C(CO)OC(OC3C(C(OC(C(O)CO)C(O)C(O)C=O)OC(CO)C3O)O)C2NC(C)=O)OC2C(C(O)C(O)C(C)O2)O)OC(CO)C(O)C1O OQIUPKPUOLIHHS-UHFFFAOYSA-N 0.000 claims description 18
- 241000193403 Clostridium Species 0.000 claims description 17
- 241000588921 Enterobacteriaceae Species 0.000 claims description 17
- 230000005865 ionizing radiation Effects 0.000 claims description 17
- 238000001356 surgical procedure Methods 0.000 claims description 17
- BRHHWBDLMUBZQQ-JZEMXWCPSA-N Lactodifucotetraose Chemical compound O[C@H]1[C@H](O)[C@H](O)[C@H](C)O[C@H]1O[C@@H]([C@H](O)[C@H](O)CO)[C@H](C=O)O[C@@H]1[C@H](O[C@H]2[C@H]([C@H](O)[C@H](O)[C@H](C)O2)O)[C@@H](O)[C@@H](O)[C@@H](CO)O1 BRHHWBDLMUBZQQ-JZEMXWCPSA-N 0.000 claims description 15
- 206010028980 Neoplasm Diseases 0.000 claims description 15
- 201000011510 cancer Diseases 0.000 claims description 15
- IEQCXFNWPAHHQR-UHFFFAOYSA-N lacto-N-neotetraose Natural products OCC1OC(OC2C(C(OC3C(OC(O)C(O)C3O)CO)OC(CO)C2O)O)C(NC(=O)C)C(O)C1OC1OC(CO)C(O)C(O)C1O IEQCXFNWPAHHQR-UHFFFAOYSA-N 0.000 claims description 15
- 229940062780 lacto-n-neotetraose Drugs 0.000 claims description 15
- RBMYDHMFFAVMMM-PLQWBNBWSA-N neolactotetraose Chemical compound O([C@H]1[C@H](O)[C@H]([C@@H](O[C@@H]1CO)O[C@@H]1[C@H]([C@H](O[C@H]([C@H](O)CO)[C@H](O)[C@@H](O)C=O)O[C@H](CO)[C@@H]1O)O)NC(=O)C)[C@@H]1O[C@H](CO)[C@H](O)[C@H](O)[C@H]1O RBMYDHMFFAVMMM-PLQWBNBWSA-N 0.000 claims description 15
- 241001505572 Anaerostipes caccae Species 0.000 claims description 14
- 208000035143 Bacterial infection Diseases 0.000 claims description 14
- 208000022559 Inflammatory bowel disease Diseases 0.000 claims description 14
- 241000872832 Roseburia hominis Species 0.000 claims description 14
- 241001533207 Veillonella atypica Species 0.000 claims description 14
- 208000022362 bacterial infectious disease Diseases 0.000 claims description 14
- 239000002773 nucleotide Substances 0.000 claims description 14
- 125000003729 nucleotide group Chemical group 0.000 claims description 14
- 208000011580 syndromic disease Diseases 0.000 claims description 14
- 241000398180 Roseburia intestinalis Species 0.000 claims description 13
- 230000006378 damage Effects 0.000 claims description 13
- 230000008569 process Effects 0.000 claims description 13
- 241001464894 Blautia producta Species 0.000 claims description 12
- 238000001914 filtration Methods 0.000 claims description 12
- 208000002551 irritable bowel syndrome Diseases 0.000 claims description 12
- 235000020183 skimmed milk Nutrition 0.000 claims description 12
- 241000604448 Megasphaera elsdenii Species 0.000 claims description 11
- 241000352296 Megasphaera massiliensis Species 0.000 claims description 11
- 206010049416 Short-bowel syndrome Diseases 0.000 claims description 11
- 208000023275 Autoimmune disease Diseases 0.000 claims description 10
- 241000605980 Faecalibacterium prausnitzii Species 0.000 claims description 10
- 206010071061 Small intestinal bacterial overgrowth Diseases 0.000 claims description 10
- 241001531188 [Eubacterium] rectale Species 0.000 claims description 10
- 230000002496 gastric effect Effects 0.000 claims description 10
- 230000007142 small intestinal bacterial overgrowth Effects 0.000 claims description 10
- 238000001228 spectrum Methods 0.000 claims description 10
- 241000194033 Enterococcus Species 0.000 claims description 9
- 241000588748 Klebsiella Species 0.000 claims description 9
- 241000604449 Megasphaera Species 0.000 claims description 9
- 241000186569 [Clostridium] leptum Species 0.000 claims description 9
- DUKURNFHYQXCJG-JEOLMMCMSA-N alpha-L-Fucp-(1->4)-[beta-D-Galp-(1->3)]-beta-D-GlcpNAc-(1->3)-beta-D-Galp-(1->4)-D-Glcp Chemical compound O[C@H]1[C@H](O)[C@H](O)[C@H](C)O[C@H]1O[C@H]1[C@H](O[C@H]2[C@@H]([C@@H](O)[C@@H](O)[C@@H](CO)O2)O)[C@@H](NC(C)=O)[C@H](O[C@@H]2[C@H]([C@H](O[C@@H]3[C@H](OC(O)[C@H](O)[C@H]3O)CO)O[C@H](CO)[C@@H]2O)O)O[C@@H]1CO DUKURNFHYQXCJG-JEOLMMCMSA-N 0.000 claims description 9
- FKADDOYBRRMBPP-UHFFFAOYSA-N lacto-N-fucopentaose II Natural products OC1C(O)C(O)C(C)OC1OC1C(OC2C(C(O)C(O)C(CO)O2)O)C(NC(C)=O)C(OC2C(C(OC(C(O)CO)C(O)C(O)C=O)OC(CO)C2O)O)OC1CO FKADDOYBRRMBPP-UHFFFAOYSA-N 0.000 claims description 9
- 241000356993 Anaerotignum Species 0.000 claims description 8
- 241000319947 Anaerotignum lactatifermentans Species 0.000 claims description 8
- 241000217846 Bacteroides caccae Species 0.000 claims description 8
- 241000606124 Bacteroides fragilis Species 0.000 claims description 8
- 241000606161 Chlamydia Species 0.000 claims description 8
- 241001464948 Coprococcus Species 0.000 claims description 8
- 241000220677 Coprococcus catus Species 0.000 claims description 8
- 208000004262 Food Hypersensitivity Diseases 0.000 claims description 8
- 241000191940 Staphylococcus Species 0.000 claims description 8
- 241000191967 Staphylococcus aureus Species 0.000 claims description 8
- 241000191981 Streptococcus cristatus Species 0.000 claims description 8
- 108010059993 Vancomycin Proteins 0.000 claims description 8
- 208000037976 chronic inflammation Diseases 0.000 claims description 8
- 208000037893 chronic inflammatory disorder Diseases 0.000 claims description 8
- 235000020932 food allergy Nutrition 0.000 claims description 8
- MYPYJXKWCTUITO-LYRMYLQWSA-N vancomycin Chemical compound O([C@@H]1[C@@H](O)[C@H](O)[C@@H](CO)O[C@H]1OC1=C2C=C3C=C1OC1=CC=C(C=C1Cl)[C@@H](O)[C@H](C(N[C@@H](CC(N)=O)C(=O)N[C@H]3C(=O)N[C@H]1C(=O)N[C@H](C(N[C@@H](C3=CC(O)=CC(O)=C3C=3C(O)=CC=C1C=3)C(O)=O)=O)[C@H](O)C1=CC=C(C(=C1)Cl)O2)=O)NC(=O)[C@@H](CC(C)C)NC)[C@H]1C[C@](C)(N)[C@H](O)[C@H](C)O1 MYPYJXKWCTUITO-LYRMYLQWSA-N 0.000 claims description 8
- 229960003165 vancomycin Drugs 0.000 claims description 8
- MYPYJXKWCTUITO-UHFFFAOYSA-N vancomycin Natural products O1C(C(=C2)Cl)=CC=C2C(O)C(C(NC(C2=CC(O)=CC(O)=C2C=2C(O)=CC=C3C=2)C(O)=O)=O)NC(=O)C3NC(=O)C2NC(=O)C(CC(N)=O)NC(=O)C(NC(=O)C(CC(C)C)NC)C(O)C(C=C3Cl)=CC=C3OC3=CC2=CC1=C3OC1OC(CO)C(O)C(O)C1OC1CC(C)(N)C(O)C(C)O1 MYPYJXKWCTUITO-UHFFFAOYSA-N 0.000 claims description 8
- 241000606125 Bacteroides Species 0.000 claims description 7
- 208000015943 Coeliac disease Diseases 0.000 claims description 7
- 206010016946 Food allergy Diseases 0.000 claims description 7
- 241000192142 Proteobacteria Species 0.000 claims description 7
- 241000194017 Streptococcus Species 0.000 claims description 7
- 241001488326 Veillonella ratti Species 0.000 claims description 7
- 241000070775 Veillonella seminalis Species 0.000 claims description 7
- 241001592639 Veillonella tobetsuensis Species 0.000 claims description 7
- 208000019902 chronic diarrheal disease Diseases 0.000 claims description 7
- 238000002271 resection Methods 0.000 claims description 7
- 241001584951 Anaerostipes hadrus Species 0.000 claims description 6
- OAXMVFUPLMUHGJ-JUZXSSEISA-N Difucosyllacto-N-hexaose a Chemical compound O[C@H]1[C@H](O)[C@H](O)[C@H](C)O[C@@H]1O[C@H]1[C@H](OC2[C@H]([C@H](O[C@H]3[C@H]([C@@H](CO[C@H]4[C@@H]([C@@H](O[C@H]5[C@H]([C@H](O)[C@H](O)[C@H](O)O5)O)[C@H](O[C@@H]5[C@@H]([C@@H](O)[C@@H](O)[C@@H](CO)O5)O)[C@@H](CO)O4)NC(C)=O)O[C@@H](OC([C@H](O)CO)[C@H](O)[C@@H](O)C=O)[C@@H]3C)O)O[C@H](CO)[C@H]2O)NC(C)=O)O[C@H](CO)[C@H](O)[C@@H]1O OAXMVFUPLMUHGJ-JUZXSSEISA-N 0.000 claims description 6
- 108090000204 Dipeptidase 1 Proteins 0.000 claims description 6
- 241000588747 Klebsiella pneumoniae Species 0.000 claims description 6
- 241000058827 Megasphaera hominis Species 0.000 claims description 6
- 241001116693 Megasphaera micronuciformis Species 0.000 claims description 6
- 241000607142 Salmonella Species 0.000 claims description 6
- 241000607768 Shigella Species 0.000 claims description 6
- SFMRPVLZMVJKGZ-JRZQLMJNSA-N Sialyllacto-N-tetraose b Chemical compound O1[C@@H]([C@H](O)[C@H](O)CO)[C@H](NC(=O)C)[C@@H](O)C[C@@]1(C(O)=O)OC[C@@H]1[C@@H](O)[C@H](O[C@H]2[C@@H]([C@@H](O)[C@@H](O)[C@@H](CO)O2)O)[C@@H](NC(C)=O)[C@H](O[C@@H]2[C@H]([C@H](O[C@H]([C@H](O)CO)[C@H](O)[C@@H](O)C=O)O[C@H](CO)[C@@H]2O)O)O1 SFMRPVLZMVJKGZ-JRZQLMJNSA-N 0.000 claims description 6
- 241000193462 [Clostridium] innocuum Species 0.000 claims description 6
- LKOHREGGXUJGKC-GXSKDVPZSA-N alpha-L-Fucp-(1->3)-[alpha-L-Fucp-(1->2)-beta-D-Galp-(1->4)]-beta-D-Glcp Chemical compound C[C@@H]1O[C@@H](O[C@@H]2[C@@H](O)[C@@H](O)[C@@H](CO)O[C@H]2O[C@@H]2[C@@H](CO)O[C@@H](O)[C@H](O)[C@H]2O[C@@H]2O[C@@H](C)[C@@H](O)[C@@H](O)[C@@H]2O)[C@@H](O)[C@H](O)[C@@H]1O LKOHREGGXUJGKC-GXSKDVPZSA-N 0.000 claims description 6
- CMQZRJBJDCVIEY-JEOLMMCMSA-N alpha-L-Fucp-(1->3)-[beta-D-Galp-(1->4)]-beta-D-GlcpNAc-(1->3)-beta-D-Galp-(1->4)-D-Glcp Chemical compound O[C@H]1[C@H](O)[C@H](O)[C@H](C)O[C@H]1O[C@H]1[C@H](O[C@H]2[C@@H]([C@@H](O)[C@@H](O)[C@@H](CO)O2)O)[C@@H](CO)O[C@@H](O[C@@H]2[C@H]([C@H](O[C@@H]3[C@H](OC(O)[C@H](O)[C@H]3O)CO)O[C@H](CO)[C@@H]2O)O)[C@@H]1NC(C)=O CMQZRJBJDCVIEY-JEOLMMCMSA-N 0.000 claims description 6
- PDWGIAAFQACISG-QZBWVFMZSA-N beta-D-Gal-(1->3)-beta-D-GlcNAc-(1->3)-[beta-D-Gal-(1->4)-beta-D-GlcNAc-(1->6)]-beta-D-Gal-(1->4)-D-Glc Chemical compound O([C@H]1[C@H](O)[C@H]([C@@H](O[C@@H]1CO)OC[C@@H]1[C@@H]([C@H](O[C@H]2[C@@H]([C@@H](O[C@H]3[C@@H]([C@@H](O)[C@@H](O)[C@@H](CO)O3)O)[C@H](O)[C@@H](CO)O2)NC(C)=O)[C@@H](O)[C@H](O[C@@H]2[C@H](OC(O)[C@H](O)[C@H]2O)CO)O1)O)NC(=O)C)[C@@H]1O[C@H](CO)[C@H](O)[C@H](O)[C@H]1O PDWGIAAFQACISG-QZBWVFMZSA-N 0.000 claims description 6
- DMYPRRDPOMGEAK-XWDFSUOISA-N beta-D-Galp-(1->3)-[alpha-L-Fucp-(1->4)]-beta-D-GlcpNAc-(1->3)-beta-D-Galp-(1->4)-[alpha-L-Fucp-(1->3)]-D-Glcp Chemical compound O[C@H]1[C@H](O)[C@H](O)[C@H](C)O[C@H]1O[C@H]1[C@H](O[C@H]2[C@@H]([C@@H](O[C@H]3[C@@H]([C@@H](O[C@H]4[C@@H]([C@@H](O)[C@@H](O)[C@@H](CO)O4)O)[C@H](O[C@H]4[C@H]([C@H](O)[C@H](O)[C@H](C)O4)O)[C@@H](CO)O3)NC(C)=O)[C@@H](O)[C@@H](CO)O2)O)[C@@H](CO)OC(O)[C@@H]1O DMYPRRDPOMGEAK-XWDFSUOISA-N 0.000 claims description 6
- 102000006635 beta-lactamase Human genes 0.000 claims description 6
- BCENFJUGBDUPJS-ZVOZPCDCSA-N difucosyllacto-n-hexaose b Chemical compound O[C@H]1[C@H](O)[C@H](O)[C@H](C)O[C@H]1O[C@H]1[C@H](O[C@H]2[C@@H]([C@@H](O)[C@@H](O)[C@@H](CO)O2)O)[C@@H](NC(C)=O)[C@H](OC2[C@H]([C@H](O[C@H]([C@H](O)CO)[C@H](O)[C@@H](O)C=O)OC(CO[C@H]3[C@@H]([C@@H](O[C@H]4[C@H]([C@H](O)[C@H](O)[C@H](C)O4)O)[C@H](O[C@@H]4[C@@H]([C@@H](O)[C@@H](O)[C@@H](CO)O4)O)[C@@H](CO)O3)NC(C)=O)[C@@H]2O)O)O[C@@H]1CO BCENFJUGBDUPJS-ZVOZPCDCSA-N 0.000 claims description 6
- DMYPRRDPOMGEAK-UHFFFAOYSA-N lacto-N-difucohexaose II Natural products OC1C(O)C(O)C(C)OC1OC1C(OC2C(C(OC3C(C(OC4C(C(O)C(O)C(CO)O4)O)C(OC4C(C(O)C(O)C(C)O4)O)C(CO)O3)NC(C)=O)C(O)C(CO)O2)O)C(CO)OC(O)C1O DMYPRRDPOMGEAK-UHFFFAOYSA-N 0.000 claims description 6
- CMQZRJBJDCVIEY-UHFFFAOYSA-N lacto-N-fucopentaose III Natural products OC1C(O)C(O)C(C)OC1OC1C(OC2C(C(O)C(O)C(CO)O2)O)C(CO)OC(OC2C(C(OC3C(OC(O)C(O)C3O)CO)OC(CO)C2O)O)C1NC(C)=O CMQZRJBJDCVIEY-UHFFFAOYSA-N 0.000 claims description 6
- 241000589876 Campylobacter Species 0.000 claims description 5
- 241000589875 Campylobacter jejuni Species 0.000 claims description 5
- 241000193163 Clostridioides difficile Species 0.000 claims description 5
- 241000305071 Enterobacterales Species 0.000 claims description 5
- 208000005577 Gastroenteritis Diseases 0.000 claims description 5
- 206010025323 Lymphomas Diseases 0.000 claims description 5
- 241000440950 Megasphaera indica Species 0.000 claims description 5
- 241000607598 Vibrio Species 0.000 claims description 5
- 241000607626 Vibrio cholerae Species 0.000 claims description 5
- 241000607734 Yersinia <bacteria> Species 0.000 claims description 5
- 150000001413 amino acids Chemical group 0.000 claims description 5
- 229940124307 fluoroquinolone Drugs 0.000 claims description 5
- 201000011587 gastric lymphoma Diseases 0.000 claims description 5
- 210000003958 hematopoietic stem cell Anatomy 0.000 claims description 5
- ZDZMLVPSYYRJNI-CYQYEHMMSA-N p-lacto-n-hexaose Chemical compound O([C@H]1[C@H](O)[C@@H](CO)O[C@H]([C@@H]1N=C(C)O)O[C@@H]1[C@@H](O)[C@H](O[C@H]([C@H](O)CO)[C@H](O)[C@@H](O)C=O)OC([C@@H]1O)CO[C@H]1[C@@H]([C@H](C(O[C@H]2[C@@H]([C@@H](O)[C@@H](O)[C@@H](CO)O2)O)[C@@H](CO)O1)O)N=C(O)C)[C@@H]1O[C@H](CO)[C@H](O)[C@H](O)[C@H]1O ZDZMLVPSYYRJNI-CYQYEHMMSA-N 0.000 claims description 5
- SXMGGNXBTZBGLU-UHFFFAOYSA-N sialyllacto-n-tetraose c Chemical compound OCC1OC(OC2C(C(OC(C(O)CO)C(O)C(O)C=O)OC(CO)C2O)O)C(NC(=O)C)C(O)C1OC(C(C(O)C1O)O)OC1COC1(C(O)=O)CC(O)C(NC(C)=O)C(C(O)C(O)CO)O1 SXMGGNXBTZBGLU-UHFFFAOYSA-N 0.000 claims description 5
- 229940118696 vibrio cholerae Drugs 0.000 claims description 5
- 241000607528 Aeromonas hydrophila Species 0.000 claims description 4
- 208000012657 Atopic disease Diseases 0.000 claims description 4
- 241000193830 Bacillus <bacterium> Species 0.000 claims description 4
- 241000193755 Bacillus cereus Species 0.000 claims description 4
- 241000186016 Bifidobacterium bifidum Species 0.000 claims description 4
- 241000588807 Bordetella Species 0.000 claims description 4
- 241000589562 Brucella Species 0.000 claims description 4
- 241001453380 Burkholderia Species 0.000 claims description 4
- 241000589874 Campylobacter fetus Species 0.000 claims description 4
- 241000588919 Citrobacter freundii Species 0.000 claims description 4
- 241000588917 Citrobacter koseri Species 0.000 claims description 4
- 241000193155 Clostridium botulinum Species 0.000 claims description 4
- 241000193468 Clostridium perfringens Species 0.000 claims description 4
- 241000186216 Corynebacterium Species 0.000 claims description 4
- 241001445332 Coxiella <snail> Species 0.000 claims description 4
- 241000605314 Ehrlichia Species 0.000 claims description 4
- 241000588697 Enterobacter cloacae Species 0.000 claims description 4
- 241000194032 Enterococcus faecalis Species 0.000 claims description 4
- 241000194031 Enterococcus faecium Species 0.000 claims description 4
- 241000588722 Escherichia Species 0.000 claims description 4
- 241001646719 Escherichia coli O157:H7 Species 0.000 claims description 4
- 241000589601 Francisella Species 0.000 claims description 4
- 241000606790 Haemophilus Species 0.000 claims description 4
- 241000590002 Helicobacter pylori Species 0.000 claims description 4
- 241000588915 Klebsiella aerogenes Species 0.000 claims description 4
- 241000588749 Klebsiella oxytoca Species 0.000 claims description 4
- 240000001929 Lactobacillus brevis Species 0.000 claims description 4
- 241000589248 Legionella Species 0.000 claims description 4
- 208000007764 Legionnaires' Disease Diseases 0.000 claims description 4
- 241000589902 Leptospira Species 0.000 claims description 4
- 241000186781 Listeria Species 0.000 claims description 4
- 241000186779 Listeria monocytogenes Species 0.000 claims description 4
- 241000588771 Morganella <proteobacterium> Species 0.000 claims description 4
- 241000588772 Morganella morganii Species 0.000 claims description 4
- 241000186359 Mycobacterium Species 0.000 claims description 4
- 241000204031 Mycoplasma Species 0.000 claims description 4
- 241000588653 Neisseria Species 0.000 claims description 4
- 241000984031 Orientia Species 0.000 claims description 4
- 241000588769 Proteus <enterobacteria> Species 0.000 claims description 4
- 241000588770 Proteus mirabilis Species 0.000 claims description 4
- 241000589516 Pseudomonas Species 0.000 claims description 4
- 241000606701 Rickettsia Species 0.000 claims description 4
- 241000531795 Salmonella enterica subsp. enterica serovar Paratyphi A Species 0.000 claims description 4
- 241000293871 Salmonella enterica subsp. enterica serovar Typhi Species 0.000 claims description 4
- 241000607715 Serratia marcescens Species 0.000 claims description 4
- 241000191963 Staphylococcus epidermidis Species 0.000 claims description 4
- 241000194008 Streptococcus anginosus Species 0.000 claims description 4
- 241000176094 Streptococcus australis Species 0.000 claims description 4
- 241001291896 Streptococcus constellatus Species 0.000 claims description 4
- 241000194026 Streptococcus gordonii Species 0.000 claims description 4
- 241000960363 Streptococcus infantis Species 0.000 claims description 4
- 241001134658 Streptococcus mitis Species 0.000 claims description 4
- 241000194019 Streptococcus mutans Species 0.000 claims description 4
- 241000194025 Streptococcus oralis Species 0.000 claims description 4
- 241000782300 Streptococcus oralis subsp. tigurinus Species 0.000 claims description 4
- 241000193991 Streptococcus parasanguinis Species 0.000 claims description 4
- 241000960362 Streptococcus peroris Species 0.000 claims description 4
- 241000193998 Streptococcus pneumoniae Species 0.000 claims description 4
- 241001403829 Streptococcus pseudopneumoniae Species 0.000 claims description 4
- 241000194024 Streptococcus salivarius Species 0.000 claims description 4
- 241000194023 Streptococcus sanguinis Species 0.000 claims description 4
- 241000193987 Streptococcus sobrinus Species 0.000 claims description 4
- 241000194051 Streptococcus vestibularis Species 0.000 claims description 4
- 241000589886 Treponema Species 0.000 claims description 4
- 241000607272 Vibrio parahaemolyticus Species 0.000 claims description 4
- 241000607265 Vibrio vulnificus Species 0.000 claims description 4
- 241000607447 Yersinia enterocolitica Species 0.000 claims description 4
- 230000003872 anastomosis Effects 0.000 claims description 4
- 208000006673 asthma Diseases 0.000 claims description 4
- 229940092559 enterobacter aerogenes Drugs 0.000 claims description 4
- 229940032049 enterococcus faecalis Drugs 0.000 claims description 4
- 230000000369 enteropathogenic effect Effects 0.000 claims description 4
- 230000000688 enterotoxigenic effect Effects 0.000 claims description 4
- 229940037467 helicobacter pylori Drugs 0.000 claims description 4
- 229940076266 morganella morganii Drugs 0.000 claims description 4
- 229940031000 streptococcus pneumoniae Drugs 0.000 claims description 4
- 229940098232 yersinia enterocolitica Drugs 0.000 claims description 4
- OWEGMIWEEQEYGQ-UHFFFAOYSA-N 100676-05-9 Natural products OC1C(O)C(O)C(CO)OC1OCC1C(O)C(O)C(O)C(OC2C(OC(O)C(O)C2O)CO)O1 OWEGMIWEEQEYGQ-UHFFFAOYSA-N 0.000 claims description 3
- FNCPZGGSTQEGGK-DRSOAOOLSA-N 3'-Sialyl-3-fucosyllactose Chemical compound O[C@H]1[C@H](O)[C@H](O)[C@H](C)O[C@H]1O[C@H]([C@@H](O)C=O)[C@@H]([C@H](O)CO)O[C@H]1[C@H](O)[C@@H](O[C@]2(O[C@H]([C@H](NC(C)=O)[C@@H](O)C2)[C@H](O)[C@H](O)CO)C(O)=O)[C@@H](O)[C@@H](CO)O1 FNCPZGGSTQEGGK-DRSOAOOLSA-N 0.000 claims description 3
- ODDPRQJTYDIWJU-UHFFFAOYSA-N 3'-beta-D-galactopyranosyl-lactose Natural products OC1C(O)C(O)C(CO)OC1OC1C(O)C(OC2C(OC(O)C(O)C2O)CO)OC(CO)C1O ODDPRQJTYDIWJU-UHFFFAOYSA-N 0.000 claims description 3
- 241000607534 Aeromonas Species 0.000 claims description 3
- 241000588923 Citrobacter Species 0.000 claims description 3
- 206010012438 Dermatitis atopic Diseases 0.000 claims description 3
- 241000588914 Enterobacter Species 0.000 claims description 3
- TVVLIFCVJJSLBL-SEHWTJTBSA-N Lacto-N-fucopentaose V Chemical compound O[C@H]1C(O)C(O)[C@H](C)O[C@H]1OC([C@@H](O)C=O)[C@@H](C(O)CO)O[C@H]1[C@H](O)[C@@H](OC2[C@@H](C(OC3[C@@H](C(O)C(O)[C@@H](CO)O3)O)[C@H](O)[C@@H](CO)O2)NC(C)=O)[C@@H](O)[C@@H](CO)O1 TVVLIFCVJJSLBL-SEHWTJTBSA-N 0.000 claims description 3
- ZHYLZEPTSHTOHM-UHFFFAOYSA-N Lacto-N-neo-difucohexaose II Natural products CC1OC(OC2C(O)C(O)OC(CO)C2OC3OC(CO)C(O)C(OC4OC(CO)C(OC5OC(CO)C(O)C(O)C5O)C(OC6OC(C)C(O)C(O)C6O)C4NC(=O)C)C3O)C(O)C(O)C1O ZHYLZEPTSHTOHM-UHFFFAOYSA-N 0.000 claims description 3
- 208000008589 Obesity Diseases 0.000 claims description 3
- 241000607000 Plesiomonas Species 0.000 claims description 3
- QUOQJNYANJQSDA-MHQSSNGYSA-N Sialyllacto-N-tetraose a Chemical compound O1C([C@H](O)[C@H](O)CO)[C@H](NC(=O)C)[C@@H](O)C[C@@]1(C(O)=O)O[C@@H]1[C@@H](O)[C@H](OC2[C@H]([C@H](OC3[C@H]([C@H](O[C@H]([C@H](O)CO)[C@H](O)[C@@H](O)C=O)O[C@H](CO)[C@@H]3O)O)O[C@H](CO)[C@H]2O)NC(C)=O)O[C@H](CO)[C@@H]1O QUOQJNYANJQSDA-MHQSSNGYSA-N 0.000 claims description 3
- 201000008937 atopic dermatitis Diseases 0.000 claims description 3
- NPPRJALWPIXIHO-PNCMPRLYSA-N beta-D-Gal-(1->4)-beta-D-GlcNAc-(1->3)-[beta-D-Gal-(1->4)-beta-D-GlcNAc-(1->6)]-beta-D-Gal-(1->4)-D-Glc Chemical compound O([C@H]1[C@H](O)[C@H]([C@@H](O[C@@H]1CO)OC[C@@H]1[C@@H]([C@H](O[C@H]2[C@@H]([C@@H](O)[C@H](O[C@H]3[C@@H]([C@@H](O)[C@@H](O)[C@@H](CO)O3)O)[C@@H](CO)O2)NC(C)=O)[C@@H](O)[C@H](O[C@@H]2[C@H](OC(O)[C@H](O)[C@H]2O)CO)O1)O)NC(=O)C)[C@@H]1O[C@H](CO)[C@H](O)[C@H](O)[C@H]1O NPPRJALWPIXIHO-PNCMPRLYSA-N 0.000 claims description 3
- ODDPRQJTYDIWJU-OAUIKNEUSA-N beta-D-Galp-(1->3)-beta-D-Galp-(1->4)-beta-D-Glcp Chemical compound O[C@@H]1[C@@H](O)[C@@H](O)[C@@H](CO)O[C@H]1O[C@@H]1[C@@H](O)[C@H](O[C@@H]2[C@H](O[C@@H](O)[C@H](O)[C@H]2O)CO)O[C@H](CO)[C@@H]1O ODDPRQJTYDIWJU-OAUIKNEUSA-N 0.000 claims description 3
- KNWXPODBRXAWBX-PFNBIPCHSA-N disialyllacto-n-fucopentaose ii Chemical compound OC1[C@H](O)[C@H](O)[C@H](C)O[C@H]1O[C@H]1[C@H](O[C@H]2[C@@H]([C@@H](O[C@]3(O[C@H]([C@H](NC(C)=O)[C@@H](O)C3)[C@H](O)[C@H](O)CO)C(O)=O)[C@@H](O)[C@@H](CO)O2)O)[C@@H](NC(C)=O)[C@H](OC2[C@H]([C@H](O[C@H]([C@H](O)CO)[C@H](O)[C@@H](O)C=O)O[C@H](CO)[C@@H]2O)O)O[C@@H]1CO[C@@]1(C(O)=O)O[C@@H]([C@H](O)[C@H](O)CO)[C@H](NC(C)=O)[C@@H](O)C1 KNWXPODBRXAWBX-PFNBIPCHSA-N 0.000 claims description 3
- 230000002045 lasting effect Effects 0.000 claims description 3
- 235000020824 obesity Nutrition 0.000 claims description 3
- 208000035473 Communicable disease Diseases 0.000 claims description 2
- 208000001072 type 2 diabetes mellitus Diseases 0.000 claims description 2
- 241000408655 Dispar Species 0.000 claims 2
- 240000000249 Morus alba Species 0.000 claims 1
- 238000011282 treatment Methods 0.000 abstract description 87
- 239000006041 probiotic Substances 0.000 abstract description 86
- 235000018291 probiotics Nutrition 0.000 abstract description 86
- 230000000529 probiotic effect Effects 0.000 abstract description 74
- -1 propionate Chemical class 0.000 abstract description 52
- 241001331543 Veillonella sp. Species 0.000 abstract description 46
- 235000021391 short chain fatty acids Nutrition 0.000 abstract description 4
- 230000006806 disease prevention Effects 0.000 abstract description 3
- 239000011885 synergistic combination Substances 0.000 abstract description 2
- 210000001035 gastrointestinal tract Anatomy 0.000 description 50
- 150000007523 nucleic acids Chemical class 0.000 description 44
- 108090000623 proteins and genes Proteins 0.000 description 39
- 108091028043 Nucleic acid sequence Proteins 0.000 description 36
- 108010005774 beta-Galactosidase Proteins 0.000 description 27
- JVTAAEKCZFNVCJ-UHFFFAOYSA-M Lactate Chemical compound CC(O)C([O-])=O JVTAAEKCZFNVCJ-UHFFFAOYSA-M 0.000 description 25
- 229940001447 lactate Drugs 0.000 description 25
- 108010059881 Lactase Proteins 0.000 description 24
- GUBGYTABKSRVRQ-QKKXKWKRSA-N Lactose Natural products OC[C@H]1O[C@@H](O[C@H]2[C@H](O)[C@@H](O)C(O)O[C@@H]2CO)[C@H](O)[C@@H](O)[C@H]1O GUBGYTABKSRVRQ-QKKXKWKRSA-N 0.000 description 21
- 239000008101 lactose Substances 0.000 description 21
- 102000004190 Enzymes Human genes 0.000 description 18
- 108090000790 Enzymes Proteins 0.000 description 18
- 229940088598 enzyme Drugs 0.000 description 18
- 235000013336 milk Nutrition 0.000 description 18
- 239000008267 milk Substances 0.000 description 18
- 210000004080 milk Anatomy 0.000 description 18
- 238000000108 ultra-filtration Methods 0.000 description 18
- 239000012528 membrane Substances 0.000 description 17
- 230000007423 decrease Effects 0.000 description 16
- 230000012010 growth Effects 0.000 description 16
- 208000018522 Gastrointestinal disease Diseases 0.000 description 15
- 208000010643 digestive system disease Diseases 0.000 description 14
- 208000018685 gastrointestinal system disease Diseases 0.000 description 14
- 210000004379 membrane Anatomy 0.000 description 14
- 102100026189 Beta-galactosidase Human genes 0.000 description 13
- 229940116108 lactase Drugs 0.000 description 13
- QTBSBXVTEAMEQO-UHFFFAOYSA-M Acetate Chemical compound CC([O-])=O QTBSBXVTEAMEQO-UHFFFAOYSA-M 0.000 description 12
- 238000011529 RT qPCR Methods 0.000 description 12
- 239000003242 anti bacterial agent Substances 0.000 description 12
- 229940088710 antibiotic agent Drugs 0.000 description 12
- 244000052616 bacterial pathogen Species 0.000 description 12
- 239000002552 dosage form Substances 0.000 description 12
- 239000002207 metabolite Substances 0.000 description 12
- 238000011269 treatment regimen Methods 0.000 description 12
- 241000699670 Mus sp. Species 0.000 description 11
- 230000000694 effects Effects 0.000 description 11
- 235000013305 food Nutrition 0.000 description 11
- 238000009472 formulation Methods 0.000 description 11
- 235000021140 nondigestible carbohydrates Nutrition 0.000 description 11
- 206010061598 Immunodeficiency Diseases 0.000 description 10
- 230000008859 change Effects 0.000 description 10
- 230000002354 daily effect Effects 0.000 description 10
- 235000016709 nutrition Nutrition 0.000 description 10
- 230000002829 reductive effect Effects 0.000 description 10
- 239000003826 tablet Substances 0.000 description 10
- 238000004458 analytical method Methods 0.000 description 9
- 230000001580 bacterial effect Effects 0.000 description 9
- 238000011534 incubation Methods 0.000 description 9
- 239000007788 liquid Substances 0.000 description 9
- 238000001728 nano-filtration Methods 0.000 description 9
- 238000011002 quantification Methods 0.000 description 9
- 238000013268 sustained release Methods 0.000 description 9
- 239000012730 sustained-release form Substances 0.000 description 9
- 238000012360 testing method Methods 0.000 description 9
- 206010017943 Gastrointestinal conditions Diseases 0.000 description 8
- 229920002472 Starch Polymers 0.000 description 8
- 230000001332 colony forming effect Effects 0.000 description 8
- 230000002550 fecal effect Effects 0.000 description 8
- 229920001223 polyethylene glycol Polymers 0.000 description 8
- 208000012868 Overgrowth Diseases 0.000 description 7
- 238000011319 anticancer therapy Methods 0.000 description 7
- 238000002512 chemotherapy Methods 0.000 description 7
- 239000006071 cream Substances 0.000 description 7
- 239000000796 flavoring agent Substances 0.000 description 7
- 239000000463 material Substances 0.000 description 7
- 238000002360 preparation method Methods 0.000 description 7
- 235000019698 starch Nutrition 0.000 description 7
- 239000008107 starch Substances 0.000 description 7
- 229940032147 starch Drugs 0.000 description 7
- 239000000725 suspension Substances 0.000 description 7
- 230000001225 therapeutic effect Effects 0.000 description 7
- 241001608472 Bifidobacterium longum Species 0.000 description 6
- 239000002202 Polyethylene glycol Substances 0.000 description 6
- 206010040047 Sepsis Diseases 0.000 description 6
- 206010047700 Vomiting Diseases 0.000 description 6
- 239000002775 capsule Substances 0.000 description 6
- 229910052799 carbon Inorganic materials 0.000 description 6
- 235000008504 concentrate Nutrition 0.000 description 6
- 239000012141 concentrate Substances 0.000 description 6
- 235000009508 confectionery Nutrition 0.000 description 6
- 238000002474 experimental method Methods 0.000 description 6
- 235000019634 flavors Nutrition 0.000 description 6
- 244000052769 pathogen Species 0.000 description 6
- 230000001717 pathogenic effect Effects 0.000 description 6
- 239000000843 powder Substances 0.000 description 6
- 235000018102 proteins Nutrition 0.000 description 6
- 102000004169 proteins and genes Human genes 0.000 description 6
- 239000000243 solution Substances 0.000 description 6
- 239000000126 substance Substances 0.000 description 6
- 230000004083 survival effect Effects 0.000 description 6
- 239000003981 vehicle Substances 0.000 description 6
- 208000004998 Abdominal Pain Diseases 0.000 description 5
- 241001202853 Blautia Species 0.000 description 5
- OKTJSMMVPCPJKN-UHFFFAOYSA-N Carbon Chemical compound [C] OKTJSMMVPCPJKN-UHFFFAOYSA-N 0.000 description 5
- SHZGCJCMOBCMKK-DHVFOXMCSA-N L-fucopyranose Chemical compound C[C@@H]1OC(O)[C@@H](O)[C@H](O)[C@@H]1O SHZGCJCMOBCMKK-DHVFOXMCSA-N 0.000 description 5
- 241000736262 Microbiota Species 0.000 description 5
- 206010028813 Nausea Diseases 0.000 description 5
- 206010037660 Pyrexia Diseases 0.000 description 5
- 241000605947 Roseburia Species 0.000 description 5
- 241000192031 Ruminococcus Species 0.000 description 5
- 230000001363 autoimmune Effects 0.000 description 5
- 235000013361 beverage Nutrition 0.000 description 5
- 210000001185 bone marrow Anatomy 0.000 description 5
- YZBQHRLRFGPBSL-RXMQYKEDSA-N carbapenem Chemical compound C1C=CN2C(=O)C[C@H]21 YZBQHRLRFGPBSL-RXMQYKEDSA-N 0.000 description 5
- 210000004027 cell Anatomy 0.000 description 5
- 238000005119 centrifugation Methods 0.000 description 5
- 239000003795 chemical substances by application Substances 0.000 description 5
- 238000005352 clarification Methods 0.000 description 5
- 238000000576 coating method Methods 0.000 description 5
- 206010009887 colitis Diseases 0.000 description 5
- 238000013270 controlled release Methods 0.000 description 5
- 230000029087 digestion Effects 0.000 description 5
- 239000008187 granular material Substances 0.000 description 5
- 230000003394 haemopoietic effect Effects 0.000 description 5
- 230000003301 hydrolyzing effect Effects 0.000 description 5
- 238000001727 in vivo Methods 0.000 description 5
- 230000004048 modification Effects 0.000 description 5
- 238000012986 modification Methods 0.000 description 5
- 230000008693 nausea Effects 0.000 description 5
- 239000000047 product Substances 0.000 description 5
- 210000002966 serum Anatomy 0.000 description 5
- CCEKAJIANROZEO-UHFFFAOYSA-N sulfluramid Chemical group CCNS(=O)(=O)C(F)(F)C(F)(F)C(F)(F)C(F)(F)C(F)(F)C(F)(F)C(F)(F)C(F)(F)F CCEKAJIANROZEO-UHFFFAOYSA-N 0.000 description 5
- 238000002560 therapeutic procedure Methods 0.000 description 5
- 230000008673 vomiting Effects 0.000 description 5
- 108020004465 16S ribosomal RNA Proteins 0.000 description 4
- 108010078791 Carrier Proteins Proteins 0.000 description 4
- 206010009900 Colitis ulcerative Diseases 0.000 description 4
- 208000011231 Crohn disease Diseases 0.000 description 4
- SHZGCJCMOBCMKK-UHFFFAOYSA-N D-mannomethylose Natural products CC1OC(O)C(O)C(O)C1O SHZGCJCMOBCMKK-UHFFFAOYSA-N 0.000 description 4
- 206010013975 Dyspnoeas Diseases 0.000 description 4
- PNNNRSAQSRJVSB-SLPGGIOYSA-N Fucose Natural products C[C@H](O)[C@@H](O)[C@H](O)[C@H](O)C=O PNNNRSAQSRJVSB-SLPGGIOYSA-N 0.000 description 4
- 208000007465 Giant cell arteritis Diseases 0.000 description 4
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 4
- 235000010469 Glycine max Nutrition 0.000 description 4
- 241000282412 Homo Species 0.000 description 4
- 229920002774 Maltodextrin Polymers 0.000 description 4
- 239000005913 Maltodextrin Substances 0.000 description 4
- 241001465754 Metazoa Species 0.000 description 4
- HSHXDCVZWHOWCS-UHFFFAOYSA-N N'-hexadecylthiophene-2-carbohydrazide Chemical compound CCCCCCCCCCCCCCCCNNC(=O)c1cccs1 HSHXDCVZWHOWCS-UHFFFAOYSA-N 0.000 description 4
- 206010035664 Pneumonia Diseases 0.000 description 4
- VYPSYNLAJGMNEJ-UHFFFAOYSA-N Silicium dioxide Chemical compound O=[Si]=O VYPSYNLAJGMNEJ-UHFFFAOYSA-N 0.000 description 4
- 208000031981 Thrombocytopenic Idiopathic Purpura Diseases 0.000 description 4
- 201000006704 Ulcerative Colitis Diseases 0.000 description 4
- 230000001154 acute effect Effects 0.000 description 4
- 239000013566 allergen Substances 0.000 description 4
- 208000022531 anorexia Diseases 0.000 description 4
- 201000003710 autoimmune thrombocytopenic purpura Diseases 0.000 description 4
- 229950002916 avelumab Drugs 0.000 description 4
- 230000009286 beneficial effect Effects 0.000 description 4
- 235000010980 cellulose Nutrition 0.000 description 4
- 229920002678 cellulose Polymers 0.000 description 4
- 239000001913 cellulose Substances 0.000 description 4
- 210000003169 central nervous system Anatomy 0.000 description 4
- 239000011248 coating agent Substances 0.000 description 4
- 210000001072 colon Anatomy 0.000 description 4
- 206010061428 decreased appetite Diseases 0.000 description 4
- 230000003247 decreasing effect Effects 0.000 description 4
- 238000001514 detection method Methods 0.000 description 4
- 229950009791 durvalumab Drugs 0.000 description 4
- 230000003203 everyday effect Effects 0.000 description 4
- 239000008103 glucose Substances 0.000 description 4
- 229920003088 hydroxypropyl methyl cellulose Polymers 0.000 description 4
- 235000010979 hydroxypropyl methyl cellulose Nutrition 0.000 description 4
- 239000001866 hydroxypropyl methyl cellulose Substances 0.000 description 4
- 229910052500 inorganic mineral Inorganic materials 0.000 description 4
- JVTAAEKCZFNVCJ-UHFFFAOYSA-N lactic acid Chemical compound CC(O)C(O)=O JVTAAEKCZFNVCJ-UHFFFAOYSA-N 0.000 description 4
- 229940035034 maltodextrin Drugs 0.000 description 4
- 238000002705 metabolomic analysis Methods 0.000 description 4
- 230000001431 metabolomic effect Effects 0.000 description 4
- 235000010755 mineral Nutrition 0.000 description 4
- 239000011707 mineral Substances 0.000 description 4
- 150000002772 monosaccharides Chemical class 0.000 description 4
- 229960003301 nivolumab Drugs 0.000 description 4
- 239000008188 pellet Substances 0.000 description 4
- 229960002621 pembrolizumab Drugs 0.000 description 4
- 239000000546 pharmaceutical excipient Substances 0.000 description 4
- 230000002265 prevention Effects 0.000 description 4
- 238000001223 reverse osmosis Methods 0.000 description 4
- 101150090202 rpoB gene Proteins 0.000 description 4
- 239000000523 sample Substances 0.000 description 4
- 238000012163 sequencing technique Methods 0.000 description 4
- 239000007787 solid Substances 0.000 description 4
- 210000000130 stem cell Anatomy 0.000 description 4
- 210000002784 stomach Anatomy 0.000 description 4
- 235000000346 sugar Nutrition 0.000 description 4
- 150000008163 sugars Chemical class 0.000 description 4
- 239000006188 syrup Substances 0.000 description 4
- 235000020357 syrup Nutrition 0.000 description 4
- 206010043207 temporal arteritis Diseases 0.000 description 4
- 235000013618 yogurt Nutrition 0.000 description 4
- DNIAPMSPPWPWGF-GSVOUGTGSA-N (R)-(-)-Propylene glycol Chemical compound C[C@@H](O)CO DNIAPMSPPWPWGF-GSVOUGTGSA-N 0.000 description 3
- OIZGSVFYNBZVIK-FHHHURIISA-N 3'-sialyllactose Chemical compound O1[C@@H]([C@H](O)[C@H](O)CO)[C@H](NC(=O)C)[C@@H](O)C[C@@]1(C(O)=O)O[C@@H]1[C@@H](O)[C@H](O[C@H]([C@H](O)CO)[C@H](O)[C@@H](O)C=O)O[C@H](CO)[C@@H]1O OIZGSVFYNBZVIK-FHHHURIISA-N 0.000 description 3
- 108010006533 ATP-Binding Cassette Transporters Proteins 0.000 description 3
- 102000005416 ATP-Binding Cassette Transporters Human genes 0.000 description 3
- QTBSBXVTEAMEQO-UHFFFAOYSA-N Acetic acid Chemical compound CC(O)=O QTBSBXVTEAMEQO-UHFFFAOYSA-N 0.000 description 3
- 241001148536 Bacteroides sp. Species 0.000 description 3
- 208000009299 Benign Mucous Membrane Pemphigoid Diseases 0.000 description 3
- 241001655328 Bifidobacteriales Species 0.000 description 3
- 241000123777 Blautia obeum Species 0.000 description 3
- 241001051189 Blautia schinkii Species 0.000 description 3
- OYPRJOBELJOOCE-UHFFFAOYSA-N Calcium Chemical compound [Ca] OYPRJOBELJOOCE-UHFFFAOYSA-N 0.000 description 3
- 208000002881 Colic Diseases 0.000 description 3
- 241000711810 Coprococcus sp. Species 0.000 description 3
- 108020004414 DNA Proteins 0.000 description 3
- 208000005156 Dehydration Diseases 0.000 description 3
- 208000000059 Dyspnea Diseases 0.000 description 3
- 241000186394 Eubacterium Species 0.000 description 3
- 208000010201 Exanthema Diseases 0.000 description 3
- 101150010169 FUT2 gene Proteins 0.000 description 3
- 244000068988 Glycine max Species 0.000 description 3
- 206010072579 Granulomatosis with polyangiitis Diseases 0.000 description 3
- 241001134638 Lachnospira Species 0.000 description 3
- 241001576959 Megasphaera sp. Species 0.000 description 3
- 208000030852 Parasitic disease Diseases 0.000 description 3
- 229920002732 Polyanhydride Polymers 0.000 description 3
- 229920000954 Polyglycolide Polymers 0.000 description 3
- 229930006000 Sucrose Natural products 0.000 description 3
- CZMRCDWAGMRECN-UGDNZRGBSA-N Sucrose Chemical compound O[C@H]1[C@H](O)[C@@H](CO)O[C@@]1(CO)O[C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](CO)O1 CZMRCDWAGMRECN-UGDNZRGBSA-N 0.000 description 3
- ZMANZCXQSJIPKH-UHFFFAOYSA-N Triethylamine Chemical compound CCN(CC)CC ZMANZCXQSJIPKH-UHFFFAOYSA-N 0.000 description 3
- 208000024780 Urticaria Diseases 0.000 description 3
- 208000036142 Viral infection Diseases 0.000 description 3
- 208000002552 acute disseminated encephalomyelitis Diseases 0.000 description 3
- 238000000540 analysis of variance Methods 0.000 description 3
- 239000000427 antigen Substances 0.000 description 3
- 108091007433 antigens Proteins 0.000 description 3
- 102000036639 antigens Human genes 0.000 description 3
- 102000005936 beta-Galactosidase Human genes 0.000 description 3
- 230000015572 biosynthetic process Effects 0.000 description 3
- 210000004369 blood Anatomy 0.000 description 3
- 239000008280 blood Substances 0.000 description 3
- 239000011575 calcium Substances 0.000 description 3
- 229910052791 calcium Inorganic materials 0.000 description 3
- 235000013351 cheese Nutrition 0.000 description 3
- RQFQJYYMBWVMQG-IXDPLRRUSA-N chitotriose Chemical compound O[C@@H]1[C@@H](N)[C@H](O)O[C@H](CO)[C@H]1O[C@H]1[C@H](N)[C@@H](O)[C@H](O[C@H]2[C@@H]([C@@H](O)[C@H](O)[C@@H](CO)O2)N)[C@@H](CO)O1 RQFQJYYMBWVMQG-IXDPLRRUSA-N 0.000 description 3
- 230000001684 chronic effect Effects 0.000 description 3
- 150000001875 compounds Chemical class 0.000 description 3
- 239000000470 constituent Substances 0.000 description 3
- HEBKCHPVOIAQTA-NGQZWQHPSA-N d-xylitol Chemical compound OC[C@H](O)C(O)[C@H](O)CO HEBKCHPVOIAQTA-NGQZWQHPSA-N 0.000 description 3
- 230000018044 dehydration Effects 0.000 description 3
- 238000006297 dehydration reaction Methods 0.000 description 3
- 230000002255 enzymatic effect Effects 0.000 description 3
- 201000005884 exanthem Diseases 0.000 description 3
- 206010016256 fatigue Diseases 0.000 description 3
- 235000021001 fermented dairy product Nutrition 0.000 description 3
- 235000003599 food sweetener Nutrition 0.000 description 3
- 235000021255 galacto-oligosaccharides Nutrition 0.000 description 3
- 150000003271 galactooligosaccharides Chemical class 0.000 description 3
- 229930182830 galactose Natural products 0.000 description 3
- 244000005709 gut microbiome Species 0.000 description 3
- 238000010438 heat treatment Methods 0.000 description 3
- 238000009169 immunotherapy Methods 0.000 description 3
- 238000011221 initial treatment Methods 0.000 description 3
- 230000000977 initiatory effect Effects 0.000 description 3
- DLRVVLDZNNYCBX-RTPHMHGBSA-N isomaltose Chemical compound O[C@@H]1[C@@H](O)[C@H](O)[C@@H](CO)O[C@@H]1OC[C@@H]1[C@@H](O)[C@H](O)[C@@H](O)C(O)O1 DLRVVLDZNNYCBX-RTPHMHGBSA-N 0.000 description 3
- 239000008176 lyophilized powder Substances 0.000 description 3
- 230000004060 metabolic process Effects 0.000 description 3
- 239000003094 microcapsule Substances 0.000 description 3
- DNIAPMSPPWPWGF-UHFFFAOYSA-N monopropylene glycol Natural products CC(O)CO DNIAPMSPPWPWGF-UHFFFAOYSA-N 0.000 description 3
- 239000002088 nanocapsule Substances 0.000 description 3
- 239000002105 nanoparticle Substances 0.000 description 3
- 208000008795 neuromyelitis optica Diseases 0.000 description 3
- 230000007935 neutral effect Effects 0.000 description 3
- 210000000056 organ Anatomy 0.000 description 3
- 238000009928 pasteurization Methods 0.000 description 3
- 230000007170 pathology Effects 0.000 description 3
- 229920000747 poly(lactic acid) Polymers 0.000 description 3
- 229920000642 polymer Polymers 0.000 description 3
- 238000012545 processing Methods 0.000 description 3
- 235000013772 propylene glycol Nutrition 0.000 description 3
- 206010037844 rash Diseases 0.000 description 3
- 230000004044 response Effects 0.000 description 3
- 239000012465 retentate Substances 0.000 description 3
- 150000003839 salts Chemical group 0.000 description 3
- 235000010356 sorbitol Nutrition 0.000 description 3
- 239000000600 sorbitol Substances 0.000 description 3
- 239000005720 sucrose Substances 0.000 description 3
- 239000003765 sweetening agent Substances 0.000 description 3
- 238000003786 synthesis reaction Methods 0.000 description 3
- 230000009885 systemic effect Effects 0.000 description 3
- 235000019640 taste Nutrition 0.000 description 3
- 208000019206 urinary tract infection Diseases 0.000 description 3
- 230000003612 virological effect Effects 0.000 description 3
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 3
- 102100023990 60S ribosomal protein L17 Human genes 0.000 description 2
- 208000008190 Agammaglobulinemia Diseases 0.000 description 2
- 229920001817 Agar Polymers 0.000 description 2
- GUBGYTABKSRVRQ-XLOQQCSPSA-N Alpha-Lactose Chemical compound O[C@@H]1[C@@H](O)[C@@H](O)[C@@H](CO)O[C@H]1O[C@@H]1[C@@H](CO)O[C@H](O)[C@H](O)[C@H]1O GUBGYTABKSRVRQ-XLOQQCSPSA-N 0.000 description 2
- 241000193457 Anaerocolumna aminovalerica Species 0.000 description 2
- 241000511612 Anaerofilum Species 0.000 description 2
- 208000003343 Antiphospholipid Syndrome Diseases 0.000 description 2
- 208000019901 Anxiety disease Diseases 0.000 description 2
- 208000006820 Arthralgia Diseases 0.000 description 2
- 208000031212 Autoimmune polyendocrinopathy Diseases 0.000 description 2
- 108010074708 B7-H1 Antigen Proteins 0.000 description 2
- 208000009137 Behcet syndrome Diseases 0.000 description 2
- 241000186560 Blautia coccoides Species 0.000 description 2
- 241000194002 Blautia hansenii Species 0.000 description 2
- 241000520742 Blautia hydrogenotrophica Species 0.000 description 2
- 241000028537 Blautia luti Species 0.000 description 2
- 206010006002 Bone pain Diseases 0.000 description 2
- 241000589968 Borrelia Species 0.000 description 2
- 241000605902 Butyrivibrio Species 0.000 description 2
- 108010021064 CTLA-4 Antigen Proteins 0.000 description 2
- 102000008203 CTLA-4 Antigen Human genes 0.000 description 2
- 229940045513 CTLA4 antagonist Drugs 0.000 description 2
- 208000030939 Chronic inflammatory demyelinating polyneuropathy Diseases 0.000 description 2
- 206010010904 Convulsion Diseases 0.000 description 2
- 229920002261 Corn starch Polymers 0.000 description 2
- 206010011224 Cough Diseases 0.000 description 2
- FBPFZTCFMRRESA-FSIIMWSLSA-N D-Glucitol Natural products OC[C@H](O)[C@H](O)[C@@H](O)[C@H](O)CO FBPFZTCFMRRESA-FSIIMWSLSA-N 0.000 description 2
- FBPFZTCFMRRESA-JGWLITMVSA-N D-glucitol Chemical compound OC[C@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-JGWLITMVSA-N 0.000 description 2
- 201000004624 Dermatitis Diseases 0.000 description 2
- 241001143779 Dorea Species 0.000 description 2
- 241001531200 Dorea formicigenerans Species 0.000 description 2
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 2
- 241001531274 Faecalicatena contorta Species 0.000 description 2
- 241001109643 Faecalicatena fissicatena Species 0.000 description 2
- 241000186589 Faecalicatena orotica Species 0.000 description 2
- 241001134569 Flavonifractor plautii Species 0.000 description 2
- 206010017367 Frequent bowel movements Diseases 0.000 description 2
- 108010010803 Gelatin Proteins 0.000 description 2
- 241000589989 Helicobacter Species 0.000 description 2
- 208000035186 Hemolytic Autoimmune Anemia Diseases 0.000 description 2
- 241001304190 Hungatella Species 0.000 description 2
- 241001674997 Hungatella hathewayi Species 0.000 description 2
- 206010020983 Hypogammaglobulinaemia Diseases 0.000 description 2
- 201000009794 Idiopathic Pulmonary Fibrosis Diseases 0.000 description 2
- 206010021245 Idiopathic thrombocytopenic purpura Diseases 0.000 description 2
- DGAQECJNVWCQMB-PUAWFVPOSA-M Ilexoside XXIX Chemical compound C[C@@H]1CC[C@@]2(CC[C@@]3(C(=CC[C@H]4[C@]3(CC[C@@H]5[C@@]4(CC[C@@H](C5(C)C)OS(=O)(=O)[O-])C)C)[C@@H]2[C@]1(C)O)C)C(=O)O[C@H]6[C@@H]([C@H]([C@@H]([C@H](O6)CO)O)O)O.[Na+] DGAQECJNVWCQMB-PUAWFVPOSA-M 0.000 description 2
- 206010021639 Incontinence Diseases 0.000 description 2
- 208000015710 Iron-Deficiency Anemia Diseases 0.000 description 2
- 206010023126 Jaundice Diseases 0.000 description 2
- 208000003456 Juvenile Arthritis Diseases 0.000 description 2
- 206010059176 Juvenile idiopathic arthritis Diseases 0.000 description 2
- 240000007472 Leucaena leucocephala Species 0.000 description 2
- 235000010643 Leucaena leucocephala Nutrition 0.000 description 2
- 208000012309 Linear IgA disease Diseases 0.000 description 2
- 208000016604 Lyme disease Diseases 0.000 description 2
- 235000019759 Maize starch Nutrition 0.000 description 2
- 208000002720 Malnutrition Diseases 0.000 description 2
- 241000124008 Mammalia Species 0.000 description 2
- 241000425724 Marvinbryantia formatexigens Species 0.000 description 2
- 101710093383 Methylmalonyl-CoA decarboxylase subunit alpha Proteins 0.000 description 2
- 101710167629 Methylmalonyl-CoA decarboxylase subunit beta Proteins 0.000 description 2
- 208000003250 Mixed connective tissue disease Diseases 0.000 description 2
- 101100407308 Mus musculus Pdcd1lg2 gene Proteins 0.000 description 2
- 208000007101 Muscle Cramp Diseases 0.000 description 2
- 241001446611 Papillibacter cinnamivorans Species 0.000 description 2
- 208000000733 Paroxysmal Hemoglobinuria Diseases 0.000 description 2
- 206010064229 Pelvic discomfort Diseases 0.000 description 2
- 206010034277 Pemphigoid Diseases 0.000 description 2
- 102100036050 Phosphatidylinositol N-acetylglucosaminyltransferase subunit A Human genes 0.000 description 2
- DLRVVLDZNNYCBX-UHFFFAOYSA-N Polydextrose Polymers OC1C(O)C(O)C(CO)OC1OCC1C(O)C(O)C(O)C(O)O1 DLRVVLDZNNYCBX-UHFFFAOYSA-N 0.000 description 2
- RJKFOVLPORLFTN-LEKSSAKUSA-N Progesterone Chemical compound C1CC2=CC(=O)CC[C@]2(C)[C@@H]2[C@@H]1[C@@H]1CC[C@H](C(=O)C)[C@@]1(C)CC2 RJKFOVLPORLFTN-LEKSSAKUSA-N 0.000 description 2
- 108700030875 Programmed Cell Death 1 Ligand 2 Proteins 0.000 description 2
- 102100024216 Programmed cell death 1 ligand 1 Human genes 0.000 description 2
- 102100024213 Programmed cell death 1 ligand 2 Human genes 0.000 description 2
- 101710089372 Programmed cell death protein 1 Proteins 0.000 description 2
- 208000003251 Pruritus Diseases 0.000 description 2
- 206010057071 Rectal tenesmus Diseases 0.000 description 2
- 229920000294 Resistant starch Polymers 0.000 description 2
- 208000037656 Respiratory Sounds Diseases 0.000 description 2
- 241001394655 Roseburia inulinivorans Species 0.000 description 2
- UIIMBOGNXHQVGW-UHFFFAOYSA-M Sodium bicarbonate Chemical compound [Na+].OC([O-])=O UIIMBOGNXHQVGW-UHFFFAOYSA-M 0.000 description 2
- UIIMBOGNXHQVGW-DEQYMQKBSA-M Sodium bicarbonate-14C Chemical compound [Na+].O[14C]([O-])=O UIIMBOGNXHQVGW-DEQYMQKBSA-M 0.000 description 2
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 2
- DBMJMQXJHONAFJ-UHFFFAOYSA-M Sodium laurylsulphate Chemical compound [Na+].CCCCCCCCCCCCOS([O-])(=O)=O DBMJMQXJHONAFJ-UHFFFAOYSA-M 0.000 description 2
- 241000194046 Streptococcus intermedius Species 0.000 description 2
- 206010042276 Subacute endocarditis Diseases 0.000 description 2
- 206010043561 Thrombocytopenic purpura Diseases 0.000 description 2
- 238000010162 Tukey test Methods 0.000 description 2
- 208000025851 Undifferentiated connective tissue disease Diseases 0.000 description 2
- 208000017379 Undifferentiated connective tissue syndrome Diseases 0.000 description 2
- 206010046851 Uveitis Diseases 0.000 description 2
- 206010047924 Wheezing Diseases 0.000 description 2
- 241001464870 [Ruminococcus] torques Species 0.000 description 2
- 206010000059 abdominal discomfort Diseases 0.000 description 2
- 239000002253 acid Substances 0.000 description 2
- 239000008272 agar Substances 0.000 description 2
- 235000010443 alginic acid Nutrition 0.000 description 2
- 229920000615 alginic acid Polymers 0.000 description 2
- 108010061314 alpha-L-Fucosidase Proteins 0.000 description 2
- 102000012086 alpha-L-Fucosidase Human genes 0.000 description 2
- 239000003708 ampul Substances 0.000 description 2
- 238000010171 animal model Methods 0.000 description 2
- 230000036506 anxiety Effects 0.000 description 2
- 230000004596 appetite loss Effects 0.000 description 2
- 238000013459 approach Methods 0.000 description 2
- 238000003556 assay Methods 0.000 description 2
- 229960003852 atezolizumab Drugs 0.000 description 2
- 208000029560 autism spectrum disease Diseases 0.000 description 2
- 208000027625 autoimmune inner ear disease Diseases 0.000 description 2
- WQZGKKKJIJFFOK-VFUOTHLCSA-N beta-D-glucose Chemical compound OC[C@H]1O[C@@H](O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-VFUOTHLCSA-N 0.000 description 2
- SQVRNKJHWKZAKO-UHFFFAOYSA-N beta-N-Acetyl-D-neuraminic acid Natural products CC(=O)NC1C(O)CC(O)(C(O)=O)OC1C(O)C(O)CO SQVRNKJHWKZAKO-UHFFFAOYSA-N 0.000 description 2
- 230000000975 bioactive effect Effects 0.000 description 2
- 208000037815 bloodstream infection Diseases 0.000 description 2
- 239000000872 buffer Substances 0.000 description 2
- 235000010418 carrageenan Nutrition 0.000 description 2
- 229920001525 carrageenan Polymers 0.000 description 2
- 239000000679 carrageenan Substances 0.000 description 2
- 229940113118 carrageenan Drugs 0.000 description 2
- 238000004113 cell culture Methods 0.000 description 2
- 238000006243 chemical reaction Methods 0.000 description 2
- 201000005795 chronic inflammatory demyelinating polyneuritis Diseases 0.000 description 2
- 239000002577 cryoprotective agent Substances 0.000 description 2
- 230000013872 defecation Effects 0.000 description 2
- 201000001981 dermatomyositis Diseases 0.000 description 2
- 235000005911 diet Nutrition 0.000 description 2
- 230000037213 diet Effects 0.000 description 2
- 239000004205 dimethyl polysiloxane Substances 0.000 description 2
- 235000013870 dimethyl polysiloxane Nutrition 0.000 description 2
- LOKCTEFSRHRXRJ-UHFFFAOYSA-I dipotassium trisodium dihydrogen phosphate hydrogen phosphate dichloride Chemical compound P(=O)(O)(O)[O-].[K+].P(=O)(O)([O-])[O-].[Na+].[Na+].[Cl-].[K+].[Cl-].[Na+] LOKCTEFSRHRXRJ-UHFFFAOYSA-I 0.000 description 2
- 239000008298 dragée Substances 0.000 description 2
- 239000003937 drug carrier Substances 0.000 description 2
- 230000004064 dysfunction Effects 0.000 description 2
- 230000006862 enzymatic digestion Effects 0.000 description 2
- 210000003608 fece Anatomy 0.000 description 2
- 239000000835 fiber Substances 0.000 description 2
- 239000000945 filler Substances 0.000 description 2
- 235000015203 fruit juice Nutrition 0.000 description 2
- 238000003304 gavage Methods 0.000 description 2
- 239000000499 gel Substances 0.000 description 2
- 239000008273 gelatin Substances 0.000 description 2
- 229920000159 gelatin Polymers 0.000 description 2
- 239000007903 gelatin capsule Substances 0.000 description 2
- 235000019322 gelatine Nutrition 0.000 description 2
- 235000011852 gelatine desserts Nutrition 0.000 description 2
- 230000036541 health Effects 0.000 description 2
- 235000015243 ice cream Nutrition 0.000 description 2
- 229940126546 immune checkpoint molecule Drugs 0.000 description 2
- 230000008676 import Effects 0.000 description 2
- 238000000338 in vitro Methods 0.000 description 2
- 230000002779 inactivation Effects 0.000 description 2
- 230000002757 inflammatory effect Effects 0.000 description 2
- 229960005386 ipilimumab Drugs 0.000 description 2
- 230000000366 juvenile effect Effects 0.000 description 2
- 239000004310 lactic acid Substances 0.000 description 2
- 235000014655 lactic acid Nutrition 0.000 description 2
- 238000012006 liquid chromatography with tandem mass spectrometry Methods 0.000 description 2
- 238000001294 liquid chromatography-tandem mass spectrometry Methods 0.000 description 2
- 235000021266 loss of appetite Nutrition 0.000 description 2
- 208000019017 loss of appetite Diseases 0.000 description 2
- 206010025135 lupus erythematosus Diseases 0.000 description 2
- HQKMJHAJHXVSDF-UHFFFAOYSA-L magnesium stearate Chemical compound [Mg+2].CCCCCCCCCCCCCCCCCC([O-])=O.CCCCCCCCCCCCCCCCCC([O-])=O HQKMJHAJHXVSDF-UHFFFAOYSA-L 0.000 description 2
- 238000012423 maintenance Methods 0.000 description 2
- 206010025482 malaise Diseases 0.000 description 2
- 230000001071 malnutrition Effects 0.000 description 2
- 235000000824 malnutrition Nutrition 0.000 description 2
- 238000005374 membrane filtration Methods 0.000 description 2
- 238000002156 mixing Methods 0.000 description 2
- 108020004707 nucleic acids Proteins 0.000 description 2
- 102000039446 nucleic acids Human genes 0.000 description 2
- 208000015380 nutritional deficiency disease Diseases 0.000 description 2
- WGNAKZGUSRVWRH-UHFFFAOYSA-N p-cresol sulfate Chemical compound CC1=CC=C(OS(O)(=O)=O)C=C1 WGNAKZGUSRVWRH-UHFFFAOYSA-N 0.000 description 2
- 235000016236 parenteral nutrition Nutrition 0.000 description 2
- 201000003045 paroxysmal nocturnal hemoglobinuria Diseases 0.000 description 2
- 230000036961 partial effect Effects 0.000 description 2
- 235000010987 pectin Nutrition 0.000 description 2
- 229920001277 pectin Polymers 0.000 description 2
- 239000001814 pectin Substances 0.000 description 2
- 229960000292 pectin Drugs 0.000 description 2
- 230000002085 persistent effect Effects 0.000 description 2
- 239000002953 phosphate buffered saline Substances 0.000 description 2
- 239000006187 pill Substances 0.000 description 2
- 229920000435 poly(dimethylsiloxane) Polymers 0.000 description 2
- 229920001592 potato starch Polymers 0.000 description 2
- 230000002035 prolonged effect Effects 0.000 description 2
- 230000000069 prophylactic effect Effects 0.000 description 2
- 238000000746 purification Methods 0.000 description 2
- 208000002574 reactive arthritis Diseases 0.000 description 2
- 230000001105 regulatory effect Effects 0.000 description 2
- 235000021254 resistant starch Nutrition 0.000 description 2
- 230000004043 responsiveness Effects 0.000 description 2
- 201000000980 schizophrenia Diseases 0.000 description 2
- 230000035807 sensation Effects 0.000 description 2
- 235000019615 sensations Nutrition 0.000 description 2
- 208000013220 shortness of breath Diseases 0.000 description 2
- 239000000377 silicon dioxide Substances 0.000 description 2
- 239000011734 sodium Substances 0.000 description 2
- 229910052708 sodium Inorganic materials 0.000 description 2
- 235000019333 sodium laurylsulphate Nutrition 0.000 description 2
- 208000008467 subacute bacterial endocarditis Diseases 0.000 description 2
- 239000004094 surface-active agent Substances 0.000 description 2
- 239000000375 suspending agent Substances 0.000 description 2
- 230000002194 synthesizing effect Effects 0.000 description 2
- 229940066453 tecentriq Drugs 0.000 description 2
- 208000012271 tenesmus Diseases 0.000 description 2
- 229940124597 therapeutic agent Drugs 0.000 description 2
- 230000004797 therapeutic response Effects 0.000 description 2
- 231100000331 toxic Toxicity 0.000 description 2
- 230000002588 toxic effect Effects 0.000 description 2
- 230000001052 transient effect Effects 0.000 description 2
- 238000007492 two-way ANOVA Methods 0.000 description 2
- 241001148471 unidentified anaerobic bacterium Species 0.000 description 2
- 235000015112 vegetable and seed oil Nutrition 0.000 description 2
- 239000008158 vegetable oil Substances 0.000 description 2
- 239000002699 waste material Substances 0.000 description 2
- 230000004580 weight loss Effects 0.000 description 2
- 208000016261 weight loss Diseases 0.000 description 2
- 229940055760 yervoy Drugs 0.000 description 2
- UHVMMEOXYDMDKI-JKYCWFKZSA-L zinc;1-(5-cyanopyridin-2-yl)-3-[(1s,2s)-2-(6-fluoro-2-hydroxy-3-propanoylphenyl)cyclopropyl]urea;diacetate Chemical compound [Zn+2].CC([O-])=O.CC([O-])=O.CCC(=O)C1=CC=C(F)C([C@H]2[C@H](C2)NC(=O)NC=2N=CC(=CC=2)C#N)=C1O UHVMMEOXYDMDKI-JKYCWFKZSA-L 0.000 description 2
- 150000003952 β-lactams Chemical class 0.000 description 2
- HDTRYLNUVZCQOY-UHFFFAOYSA-N α-D-glucopyranosyl-α-D-glucopyranoside Natural products OC1C(O)C(O)C(CO)OC1OC1C(O)C(O)C(O)C(CO)O1 HDTRYLNUVZCQOY-UHFFFAOYSA-N 0.000 description 1
- OMDQUFIYNPYJFM-XKDAHURESA-N (2r,3r,4s,5r,6s)-2-(hydroxymethyl)-6-[[(2r,3s,4r,5s,6r)-4,5,6-trihydroxy-3-[(2s,3s,4s,5s,6r)-3,4,5-trihydroxy-6-(hydroxymethyl)oxan-2-yl]oxyoxan-2-yl]methoxy]oxane-3,4,5-triol Chemical compound O[C@@H]1[C@@H](O)[C@@H](O)[C@@H](CO)O[C@@H]1OC[C@@H]1[C@@H](O[C@H]2[C@H]([C@@H](O)[C@H](O)[C@@H](CO)O2)O)[C@H](O)[C@H](O)[C@H](O)O1 OMDQUFIYNPYJFM-XKDAHURESA-N 0.000 description 1
- LNAZSHAWQACDHT-XIYTZBAFSA-N (2r,3r,4s,5r,6s)-4,5-dimethoxy-2-(methoxymethyl)-3-[(2s,3r,4s,5r,6r)-3,4,5-trimethoxy-6-(methoxymethyl)oxan-2-yl]oxy-6-[(2r,3r,4s,5r,6r)-4,5,6-trimethoxy-2-(methoxymethyl)oxan-3-yl]oxyoxane Chemical compound CO[C@@H]1[C@@H](OC)[C@H](OC)[C@@H](COC)O[C@H]1O[C@H]1[C@H](OC)[C@@H](OC)[C@H](O[C@H]2[C@@H]([C@@H](OC)[C@H](OC)O[C@@H]2COC)OC)O[C@@H]1COC LNAZSHAWQACDHT-XIYTZBAFSA-N 0.000 description 1
- FVVCFHXLWDDRHG-UPLOTWCNSA-N (2s,3r,4s,5r,6r)-2-[(2r,3s,4r,5r,6r)-6-[(2s,3s,4s,5r)-3,4-dihydroxy-2,5-bis(hydroxymethyl)oxolan-2-yl]oxy-4,5-dihydroxy-2-(hydroxymethyl)oxan-3-yl]oxy-6-(hydroxymethyl)oxane-3,4,5-triol Chemical compound O[C@H]1[C@H](O)[C@@H](CO)O[C@@]1(CO)O[C@@H]1[C@H](O)[C@@H](O)[C@H](O[C@H]2[C@@H]([C@@H](O)[C@@H](O)[C@@H](CO)O2)O)[C@@H](CO)O1 FVVCFHXLWDDRHG-UPLOTWCNSA-N 0.000 description 1
- LUEWUZLMQUOBSB-FSKGGBMCSA-N (2s,3s,4s,5s,6r)-2-[(2r,3s,4r,5r,6s)-6-[(2r,3s,4r,5s,6s)-4,5-dihydroxy-2-(hydroxymethyl)-6-[(2r,4r,5s,6r)-4,5,6-trihydroxy-2-(hydroxymethyl)oxan-3-yl]oxyoxan-3-yl]oxy-4,5-dihydroxy-2-(hydroxymethyl)oxan-3-yl]oxy-6-(hydroxymethyl)oxane-3,4,5-triol Chemical compound O[C@H]1[C@@H](O)[C@H](O)[C@@H](CO)O[C@H]1O[C@@H]1[C@@H](CO)O[C@@H](O[C@@H]2[C@H](O[C@@H](OC3[C@H](O[C@@H](O)[C@@H](O)[C@H]3O)CO)[C@@H](O)[C@H]2O)CO)[C@H](O)[C@H]1O LUEWUZLMQUOBSB-FSKGGBMCSA-N 0.000 description 1
- MINDHVHHQZYEEK-UHFFFAOYSA-N (E)-(2S,3R,4R,5S)-5-[(2S,3S,4S,5S)-2,3-epoxy-5-hydroxy-4-methylhexyl]tetrahydro-3,4-dihydroxy-(beta)-methyl-2H-pyran-2-crotonic acid ester with 9-hydroxynonanoic acid Natural products CC(O)C(C)C1OC1CC1C(O)C(O)C(CC(C)=CC(=O)OCCCCCCCCC(O)=O)OC1 MINDHVHHQZYEEK-UHFFFAOYSA-N 0.000 description 1
- 230000006269 (delayed) early viral mRNA transcription Effects 0.000 description 1
- IXPNQXFRVYWDDI-UHFFFAOYSA-N 1-methyl-2,4-dioxo-1,3-diazinane-5-carboximidamide Chemical compound CN1CC(C(N)=N)C(=O)NC1=O IXPNQXFRVYWDDI-UHFFFAOYSA-N 0.000 description 1
- IIZPXYDJLKNOIY-JXPKJXOSSA-N 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine Chemical compound CCCCCCCCCCCCCCCC(=O)OC[C@H](COP([O-])(=O)OCC[N+](C)(C)C)OC(=O)CCC\C=C/C\C=C/C\C=C/C\C=C/CCCCC IIZPXYDJLKNOIY-JXPKJXOSSA-N 0.000 description 1
- PUNGSQUVTIDKNU-UHFFFAOYSA-N 2,4,6,8,10-pentamethyl-1,3,5,7,9,2$l^{3},4$l^{3},6$l^{3},8$l^{3},10$l^{3}-pentaoxapentasilecane Chemical compound C[Si]1O[Si](C)O[Si](C)O[Si](C)O[Si](C)O1 PUNGSQUVTIDKNU-UHFFFAOYSA-N 0.000 description 1
- MIJDSYMOBYNHOT-UHFFFAOYSA-N 2-(ethylamino)ethanol Chemical compound CCNCCO MIJDSYMOBYNHOT-UHFFFAOYSA-N 0.000 description 1
- XGILAAMKEQUXLS-UHFFFAOYSA-N 3-(indol-3-yl)lactic acid Chemical compound C1=CC=C2C(CC(O)C(O)=O)=CNC2=C1 XGILAAMKEQUXLS-UHFFFAOYSA-N 0.000 description 1
- SATHPVQTSSUFFW-UHFFFAOYSA-N 4-[6-[(3,5-dihydroxy-4-methoxyoxan-2-yl)oxymethyl]-3,5-dihydroxy-4-methoxyoxan-2-yl]oxy-2-(hydroxymethyl)-6-methyloxane-3,5-diol Chemical compound OC1C(OC)C(O)COC1OCC1C(O)C(OC)C(O)C(OC2C(C(CO)OC(C)C2O)O)O1 SATHPVQTSSUFFW-UHFFFAOYSA-N 0.000 description 1
- 108010024655 4-hydroxybutyrate CoA-transferase Proteins 0.000 description 1
- FHVDTGUDJYJELY-UHFFFAOYSA-N 6-{[2-carboxy-4,5-dihydroxy-6-(phosphanyloxy)oxan-3-yl]oxy}-4,5-dihydroxy-3-phosphanyloxane-2-carboxylic acid Chemical compound O1C(C(O)=O)C(P)C(O)C(O)C1OC1C(C(O)=O)OC(OP)C(O)C1O FHVDTGUDJYJELY-UHFFFAOYSA-N 0.000 description 1
- 206010000087 Abdominal pain upper Diseases 0.000 description 1
- 244000215068 Acacia senegal Species 0.000 description 1
- 108030002957 Acetate CoA-transferases Proteins 0.000 description 1
- 241000251468 Actinopterygii Species 0.000 description 1
- 208000032194 Acute haemorrhagic leukoencephalitis Diseases 0.000 description 1
- 208000026872 Addison Disease Diseases 0.000 description 1
- 208000032671 Allergic granulomatous angiitis Diseases 0.000 description 1
- 235000019489 Almond oil Nutrition 0.000 description 1
- 201000004384 Alopecia Diseases 0.000 description 1
- 206010001935 American trypanosomiasis Diseases 0.000 description 1
- QGZKDVFQNNGYKY-UHFFFAOYSA-O Ammonium Chemical compound [NH4+] QGZKDVFQNNGYKY-UHFFFAOYSA-O 0.000 description 1
- 241001522777 Anaerostipes butyraticus Species 0.000 description 1
- 241000100356 Anaerostipes rhamnosivorans Species 0.000 description 1
- 241000428313 Anaerotruncus colihominis Species 0.000 description 1
- 206010002199 Anaphylactic shock Diseases 0.000 description 1
- 208000028185 Angioedema Diseases 0.000 description 1
- 206010002556 Ankylosing Spondylitis Diseases 0.000 description 1
- 239000001904 Arabinogalactan Substances 0.000 description 1
- 229920000189 Arabinogalactan Polymers 0.000 description 1
- 244000105624 Arachis hypogaea Species 0.000 description 1
- 241000203069 Archaea Species 0.000 description 1
- 206010003267 Arthritis reactive Diseases 0.000 description 1
- 241000416162 Astragalus gummifer Species 0.000 description 1
- 208000032116 Autoimmune Experimental Encephalomyelitis Diseases 0.000 description 1
- 206010071576 Autoimmune aplastic anaemia Diseases 0.000 description 1
- 206010003827 Autoimmune hepatitis Diseases 0.000 description 1
- 206010071577 Autoimmune hyperlipidaemia Diseases 0.000 description 1
- 206010064539 Autoimmune myocarditis Diseases 0.000 description 1
- 206010069002 Autoimmune pancreatitis Diseases 0.000 description 1
- 208000022106 Autoimmune polyendocrinopathy type 2 Diseases 0.000 description 1
- 206010003840 Autonomic nervous system imbalance Diseases 0.000 description 1
- 235000007319 Avena orientalis Nutrition 0.000 description 1
- 241000209763 Avena sativa Species 0.000 description 1
- 235000007558 Avena sp Nutrition 0.000 description 1
- 235000017166 Bambusa arundinacea Nutrition 0.000 description 1
- 235000017491 Bambusa tulda Nutrition 0.000 description 1
- 208000023328 Basedow disease Diseases 0.000 description 1
- 241000186018 Bifidobacterium adolescentis Species 0.000 description 1
- 241000901051 Bifidobacterium animalis subsp. animalis Species 0.000 description 1
- 241000901050 Bifidobacterium animalis subsp. lactis Species 0.000 description 1
- 241000186011 Bifidobacterium catenulatum Species 0.000 description 1
- 241000185999 Bifidobacterium longum subsp. longum Species 0.000 description 1
- 208000008439 Biliary Liver Cirrhosis Diseases 0.000 description 1
- 208000033222 Biliary cirrhosis primary Diseases 0.000 description 1
- 241001140799 Blautia caecimuris Species 0.000 description 1
- 241000335561 Blautia faecis Species 0.000 description 1
- 241001449840 Blautia glucerasea Species 0.000 description 1
- 241001603665 Blautia hominis Species 0.000 description 1
- 241000772275 Blautia sp. Species 0.000 description 1
- 241000335560 Blautia stercoris Species 0.000 description 1
- 241001038648 Blautia wexlerae Species 0.000 description 1
- 241000167854 Bourreria succulenta Species 0.000 description 1
- 241000589893 Brachyspira hyodysenteriae Species 0.000 description 1
- 206010006784 Burning sensation Diseases 0.000 description 1
- 108700024126 Butyrate kinases Proteins 0.000 description 1
- 241000193174 Butyrivibrio crossotus Species 0.000 description 1
- 241000634280 Caproiciproducens Species 0.000 description 1
- 229920002134 Carboxymethyl cellulose Polymers 0.000 description 1
- 208000031229 Cardiomyopathies Diseases 0.000 description 1
- 208000005024 Castleman disease Diseases 0.000 description 1
- 229920000623 Cellulose acetate phthalate Polymers 0.000 description 1
- 240000008886 Ceratonia siliqua Species 0.000 description 1
- 235000013912 Ceratonia siliqua Nutrition 0.000 description 1
- 208000024699 Chagas disease Diseases 0.000 description 1
- 206010008609 Cholangitis sclerosing Diseases 0.000 description 1
- 206010008631 Cholera Diseases 0.000 description 1
- 208000006344 Churg-Strauss Syndrome Diseases 0.000 description 1
- 241000207199 Citrus Species 0.000 description 1
- 235000005979 Citrus limon Nutrition 0.000 description 1
- 244000131522 Citrus pyriformis Species 0.000 description 1
- 241001112696 Clostridia Species 0.000 description 1
- 208000010007 Cogan syndrome Diseases 0.000 description 1
- 208000011038 Cold agglutinin disease Diseases 0.000 description 1
- 206010009868 Cold type haemolytic anaemia Diseases 0.000 description 1
- 206010056979 Colitis microscopic Diseases 0.000 description 1
- 206010009944 Colon cancer Diseases 0.000 description 1
- 206010010071 Coma Diseases 0.000 description 1
- 208000013586 Complex regional pain syndrome type 1 Diseases 0.000 description 1
- 241000949098 Coprococcus comes Species 0.000 description 1
- 241001464949 Coprococcus eutactus Species 0.000 description 1
- 206010011258 Coxsackie myocarditis Diseases 0.000 description 1
- 208000014997 Crohn colitis Diseases 0.000 description 1
- 208000019707 Cryoglobulinemic vasculitis Diseases 0.000 description 1
- 201000003883 Cystic fibrosis Diseases 0.000 description 1
- FBPFZTCFMRRESA-KVTDHHQDSA-N D-Mannitol Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-KVTDHHQDSA-N 0.000 description 1
- 206010012468 Dermatitis herpetiformis Diseases 0.000 description 1
- 206010048768 Dermatosis Diseases 0.000 description 1
- 206010064687 Device related infection Diseases 0.000 description 1
- 239000004375 Dextrin Substances 0.000 description 1
- 229920001353 Dextrin Polymers 0.000 description 1
- 206010012741 Diarrhoea haemorrhagic Diseases 0.000 description 1
- 241000016537 Dorea longicatena Species 0.000 description 1
- 208000021866 Dressler syndrome Diseases 0.000 description 1
- 241001438869 Eisenbergiella tayi Species 0.000 description 1
- 206010014418 Electrolyte imbalance Diseases 0.000 description 1
- 241000196324 Embryophyta Species 0.000 description 1
- 241000442100 Emergencia timonensis Species 0.000 description 1
- 201000009273 Endometriosis Diseases 0.000 description 1
- 241000792859 Enema Species 0.000 description 1
- 206010014954 Eosinophilic fasciitis Diseases 0.000 description 1
- 208000018428 Eosinophilic granulomatosis with polyangiitis Diseases 0.000 description 1
- 206010064212 Eosinophilic oesophagitis Diseases 0.000 description 1
- 241000186588 Erysipelatoclostridium ramosum Species 0.000 description 1
- 206010015226 Erythema nodosum Diseases 0.000 description 1
- 102100029106 Ethylmalonyl-CoA decarboxylase Human genes 0.000 description 1
- 241001531192 Eubacterium ventriosum Species 0.000 description 1
- 208000004332 Evans syndrome Diseases 0.000 description 1
- 101150074294 FUT3 gene Proteins 0.000 description 1
- 241000870027 Faecalicatena Species 0.000 description 1
- 241000870020 Faecalimonas Species 0.000 description 1
- 241000192125 Firmicutes Species 0.000 description 1
- 206010016717 Fistula Diseases 0.000 description 1
- 206010016952 Food poisoning Diseases 0.000 description 1
- 208000019331 Foodborne disease Diseases 0.000 description 1
- 235000016623 Fragaria vesca Nutrition 0.000 description 1
- 240000009088 Fragaria x ananassa Species 0.000 description 1
- 235000011363 Fragaria x ananassa Nutrition 0.000 description 1
- 241000233866 Fungi Species 0.000 description 1
- 229920000926 Galactomannan Polymers 0.000 description 1
- 206010018364 Glomerulonephritis Diseases 0.000 description 1
- 229920001503 Glucan Polymers 0.000 description 1
- 229920002581 Glucomannan Polymers 0.000 description 1
- 208000024869 Goodpasture syndrome Diseases 0.000 description 1
- 208000015023 Graves' disease Diseases 0.000 description 1
- 229920002907 Guar gum Polymers 0.000 description 1
- 208000035895 Guillain-Barré syndrome Diseases 0.000 description 1
- 229920000084 Gum arabic Polymers 0.000 description 1
- 208000030836 Hashimoto thyroiditis Diseases 0.000 description 1
- 206010019263 Heart block congenital Diseases 0.000 description 1
- 201000004331 Henoch-Schoenlein purpura Diseases 0.000 description 1
- 206010019617 Henoch-Schonlein purpura Diseases 0.000 description 1
- 206010019670 Hepatic function abnormal Diseases 0.000 description 1
- 206010019939 Herpes gestationis Diseases 0.000 description 1
- 108010000540 Hexosaminidases Proteins 0.000 description 1
- 102000002268 Hexosaminidases Human genes 0.000 description 1
- 101000801619 Homo sapiens Long-chain-fatty-acid-CoA ligase ACSBG1 Proteins 0.000 description 1
- 241001523756 Hungatella effluvii Species 0.000 description 1
- 208000001953 Hypotension Diseases 0.000 description 1
- 208000031814 IgA Vasculitis Diseases 0.000 description 1
- 208000010159 IgA glomerulonephritis Diseases 0.000 description 1
- 206010021263 IgA nephropathy Diseases 0.000 description 1
- 208000021330 IgG4-related disease Diseases 0.000 description 1
- 208000014919 IgG4-related retroperitoneal fibrosis Diseases 0.000 description 1
- 229940076838 Immune checkpoint inhibitor Drugs 0.000 description 1
- 208000029462 Immunodeficiency disease Diseases 0.000 description 1
- 208000031781 Immunoglobulin G4 related sclerosing disease Diseases 0.000 description 1
- 208000004187 Immunoglobulin G4-Related Disease Diseases 0.000 description 1
- 206010022557 Intermediate uveitis Diseases 0.000 description 1
- 208000005615 Interstitial Cystitis Diseases 0.000 description 1
- 229920001202 Inulin Polymers 0.000 description 1
- 206010023230 Joint stiffness Diseases 0.000 description 1
- HNDVDQJCIGZPNO-YFKPBYRVSA-N L-histidine Chemical compound OC(=O)[C@@H](N)CC1=CN=CN1 HNDVDQJCIGZPNO-YFKPBYRVSA-N 0.000 description 1
- 241001134640 Lachnospira multipara Species 0.000 description 1
- 241001134642 Lachnospira pectinoschiza Species 0.000 description 1
- 241001112693 Lachnospiraceae Species 0.000 description 1
- 241000685814 Lactonifactor longoviformis Species 0.000 description 1
- 201000010743 Lambert-Eaton myasthenic syndrome Diseases 0.000 description 1
- 241001148567 Lawsonia intracellularis Species 0.000 description 1
- 208000032514 Leukocytoclastic vasculitis Diseases 0.000 description 1
- 206010024434 Lichen sclerosus Diseases 0.000 description 1
- 108090001030 Lipoproteins Proteins 0.000 description 1
- 102000004895 Lipoproteins Human genes 0.000 description 1
- 229920000161 Locust bean gum Polymers 0.000 description 1
- 102100033564 Long-chain-fatty-acid-CoA ligase ACSBG1 Human genes 0.000 description 1
- 240000002129 Malva sylvestris Species 0.000 description 1
- 235000006770 Malva sylvestris Nutrition 0.000 description 1
- 229930195725 Mannitol Natural products 0.000 description 1
- 208000027530 Meniere disease Diseases 0.000 description 1
- 201000009906 Meningitis Diseases 0.000 description 1
- 241001601860 Merdimonas faecis Species 0.000 description 1
- 108010085747 Methylmalonyl-CoA Decarboxylase Proteins 0.000 description 1
- 108700011259 MicroRNAs Proteins 0.000 description 1
- 229920000168 Microcrystalline cellulose Polymers 0.000 description 1
- 206010049567 Miller Fisher syndrome Diseases 0.000 description 1
- 208000024599 Mooren ulcer Diseases 0.000 description 1
- 208000012192 Mucous membrane pemphigoid Diseases 0.000 description 1
- 241000151756 Murimonas intestini Species 0.000 description 1
- 241000699666 Mus <mouse, genus> Species 0.000 description 1
- 208000010428 Muscle Weakness Diseases 0.000 description 1
- 206010028372 Muscular weakness Diseases 0.000 description 1
- 241000238367 Mya arenaria Species 0.000 description 1
- 208000000112 Myalgia Diseases 0.000 description 1
- 241000186367 Mycobacterium avium Species 0.000 description 1
- 201000002481 Myositis Diseases 0.000 description 1
- OVRNDRQMDRJTHS-UHFFFAOYSA-N N-acelyl-D-glucosamine Natural products CC(=O)NC1C(O)OC(CO)C(O)C1O OVRNDRQMDRJTHS-UHFFFAOYSA-N 0.000 description 1
- OVRNDRQMDRJTHS-FMDGEEDCSA-N N-acetyl-beta-D-glucosamine Chemical compound CC(=O)N[C@H]1[C@H](O)O[C@H](CO)[C@@H](O)[C@@H]1O OVRNDRQMDRJTHS-FMDGEEDCSA-N 0.000 description 1
- SQVRNKJHWKZAKO-PFQGKNLYSA-N N-acetyl-beta-neuraminic acid Chemical compound CC(=O)N[C@@H]1[C@@H](O)C[C@@](O)(C(O)=O)O[C@H]1[C@H](O)[C@H](O)CO SQVRNKJHWKZAKO-PFQGKNLYSA-N 0.000 description 1
- MBLBDJOUHNCFQT-LXGUWJNJSA-N N-acetylglucosamine Natural products CC(=O)N[C@@H](C=O)[C@@H](O)[C@H](O)[C@H](O)CO MBLBDJOUHNCFQT-LXGUWJNJSA-N 0.000 description 1
- 206010029216 Nervousness Diseases 0.000 description 1
- 208000027626 Neurocognitive disease Diseases 0.000 description 1
- 208000029726 Neurodevelopmental disease Diseases 0.000 description 1
- 206010071579 Neuronal neuropathy Diseases 0.000 description 1
- 208000003435 Optic Neuritis Diseases 0.000 description 1
- 208000007117 Oral Ulcer Diseases 0.000 description 1
- 241000843248 Oscillibacter Species 0.000 description 1
- 206010053869 POEMS syndrome Diseases 0.000 description 1
- 241001446614 Papillibacter Species 0.000 description 1
- 206010048705 Paraneoplastic cerebellar degeneration Diseases 0.000 description 1
- 208000026681 Paratuberculosis Diseases 0.000 description 1
- 208000004788 Pars Planitis Diseases 0.000 description 1
- 208000008223 Pemphigoid Gestationis Diseases 0.000 description 1
- 241000721454 Pemphigus Species 0.000 description 1
- 208000031845 Pernicious anaemia Diseases 0.000 description 1
- 102000009097 Phosphorylases Human genes 0.000 description 1
- 108010073135 Phosphorylases Proteins 0.000 description 1
- 244000082204 Phyllostachys viridis Species 0.000 description 1
- 235000015334 Phyllostachys viridis Nutrition 0.000 description 1
- 208000000766 Pityriasis Lichenoides Diseases 0.000 description 1
- 206010048895 Pityriasis lichenoides et varioliformis acuta Diseases 0.000 description 1
- 244000134552 Plantago ovata Species 0.000 description 1
- 235000003421 Plantago ovata Nutrition 0.000 description 1
- 241000606999 Plesiomonas shigelloides Species 0.000 description 1
- 229920001054 Poly(ethylene‐co‐vinyl acetate) Polymers 0.000 description 1
- 206010065159 Polychondritis Diseases 0.000 description 1
- 229920001100 Polydextrose Polymers 0.000 description 1
- 229920001710 Polyorthoester Polymers 0.000 description 1
- 229920001213 Polysorbate 20 Polymers 0.000 description 1
- 208000031732 Post-Lyme Disease Syndrome Diseases 0.000 description 1
- 208000004347 Postpericardiotomy Syndrome Diseases 0.000 description 1
- ZLMJMSJWJFRBEC-UHFFFAOYSA-N Potassium Chemical compound [K] ZLMJMSJWJFRBEC-UHFFFAOYSA-N 0.000 description 1
- 208000012654 Primary biliary cholangitis Diseases 0.000 description 1
- 206010036774 Proctitis Diseases 0.000 description 1
- 206010036783 Proctitis ulcerative Diseases 0.000 description 1
- 208000037534 Progressive hemifacial atrophy Diseases 0.000 description 1
- 241001528479 Pseudoflavonifractor capillosus Species 0.000 description 1
- 201000004681 Psoriasis Diseases 0.000 description 1
- 201000001263 Psoriatic Arthritis Diseases 0.000 description 1
- 208000036824 Psoriatic arthropathy Diseases 0.000 description 1
- 239000009223 Psyllium Substances 0.000 description 1
- 208000003670 Pure Red-Cell Aplasia Diseases 0.000 description 1
- 208000012322 Raynaud phenomenon Diseases 0.000 description 1
- 201000001947 Reflex Sympathetic Dystrophy Diseases 0.000 description 1
- 208000033464 Reiter syndrome Diseases 0.000 description 1
- 208000005793 Restless legs syndrome Diseases 0.000 description 1
- 206010038979 Retroperitoneal fibrosis Diseases 0.000 description 1
- 208000025747 Rheumatic disease Diseases 0.000 description 1
- 241001052237 Robinsoniella peoriensis Species 0.000 description 1
- 241000872831 Roseburia faecis Species 0.000 description 1
- 241000711837 Roseburia sp. Species 0.000 description 1
- 241000192029 Ruminococcus albus Species 0.000 description 1
- 241000123753 Ruminococcus bromii Species 0.000 description 1
- 241000291327 Ruminococcus faecis Species 0.000 description 1
- 241000202356 Ruminococcus lactaris Species 0.000 description 1
- 241000134861 Ruminococcus sp. Species 0.000 description 1
- 206010039705 Scleritis Diseases 0.000 description 1
- 206010039710 Scleroderma Diseases 0.000 description 1
- 208000021386 Sjogren Syndrome Diseases 0.000 description 1
- 239000004141 Sodium laurylsulphate Substances 0.000 description 1
- 206010062255 Soft tissue infection Diseases 0.000 description 1
- 229920002125 Sokalan® Polymers 0.000 description 1
- 241000168515 Sporobacter Species 0.000 description 1
- 235000021355 Stearic acid Nutrition 0.000 description 1
- 206010072148 Stiff-Person syndrome Diseases 0.000 description 1
- 241001580973 Subdoligranulum variabile Species 0.000 description 1
- 208000031650 Surgical Wound Infection Diseases 0.000 description 1
- 208000002286 Susac Syndrome Diseases 0.000 description 1
- 206010042674 Swelling Diseases 0.000 description 1
- 206010042742 Sympathetic ophthalmia Diseases 0.000 description 1
- 241001656784 Syntrophococcus Species 0.000 description 1
- 241001656782 Syntrophococcus sucromutans Species 0.000 description 1
- 208000001871 Tachycardia Diseases 0.000 description 1
- 208000001106 Takayasu Arteritis Diseases 0.000 description 1
- FEWJPZIEWOKRBE-UHFFFAOYSA-N Tartaric Acid Chemical class [H+].[H+].[O-]C(=O)C(O)C(O)C([O-])=O FEWJPZIEWOKRBE-UHFFFAOYSA-N 0.000 description 1
- 206010071574 Testicular autoimmunity Diseases 0.000 description 1
- 244000299461 Theobroma cacao Species 0.000 description 1
- 206010051526 Tolosa-Hunt syndrome Diseases 0.000 description 1
- 229920001615 Tragacanth Polymers 0.000 description 1
- 108090000992 Transferases Proteins 0.000 description 1
- 102000004357 Transferases Human genes 0.000 description 1
- HDTRYLNUVZCQOY-WSWWMNSNSA-N Trehalose Natural products O[C@@H]1[C@@H](O)[C@@H](O)[C@@H](CO)O[C@@H]1O[C@@H]1[C@H](O)[C@@H](O)[C@@H](O)[C@@H](CO)O1 HDTRYLNUVZCQOY-WSWWMNSNSA-N 0.000 description 1
- 235000021307 Triticum Nutrition 0.000 description 1
- 244000098338 Triticum aestivum Species 0.000 description 1
- 241000223109 Trypanosoma cruzi Species 0.000 description 1
- 206010067584 Type 1 diabetes mellitus Diseases 0.000 description 1
- 108700036309 Type I Plasminogen Deficiency Proteins 0.000 description 1
- 241001147795 Tyzzerella nexilis Species 0.000 description 1
- 206010064996 Ulcerative keratitis Diseases 0.000 description 1
- 208000003443 Unconsciousness Diseases 0.000 description 1
- 235000009499 Vanilla fragrans Nutrition 0.000 description 1
- 244000263375 Vanilla tahitensis Species 0.000 description 1
- 235000012036 Vanilla tahitensis Nutrition 0.000 description 1
- 206010047115 Vasculitis Diseases 0.000 description 1
- 241001048169 Veillonella caviae Species 0.000 description 1
- 241001050689 Veillonella denticariosi Species 0.000 description 1
- 241001142820 Veillonella magna Species 0.000 description 1
- 241001116698 Veillonella montpellierensis Species 0.000 description 1
- 241001048168 Veillonella rodentium Species 0.000 description 1
- 208000009470 Ventilator-Associated Pneumonia Diseases 0.000 description 1
- 241000700605 Viruses Species 0.000 description 1
- 206010047642 Vitiligo Diseases 0.000 description 1
- 235000009754 Vitis X bourquina Nutrition 0.000 description 1
- 235000012333 Vitis X labruscana Nutrition 0.000 description 1
- 240000006365 Vitis vinifera Species 0.000 description 1
- 235000014787 Vitis vinifera Nutrition 0.000 description 1
- 206010048038 Wound infection Diseases 0.000 description 1
- 208000027418 Wounds and injury Diseases 0.000 description 1
- HCHKCACWOHOZIP-UHFFFAOYSA-N Zinc Chemical compound [Zn] HCHKCACWOHOZIP-UHFFFAOYSA-N 0.000 description 1
- 241001509492 [Clostridium] aerotolerans Species 0.000 description 1
- 241001118737 [Clostridium] aldenense Species 0.000 description 1
- 241000360105 [Clostridium] algidixylanolyticum Species 0.000 description 1
- 241001350461 [Clostridium] amygdalinum Species 0.000 description 1
- 241000342876 [Clostridium] asparagiforme Species 0.000 description 1
- 241001246487 [Clostridium] bolteae Species 0.000 description 1
- 241001147804 [Clostridium] celerecrescens Species 0.000 description 1
- 241001147772 [Clostridium] cellulosi Species 0.000 description 1
- 241001118742 [Clostridium] citroniae Species 0.000 description 1
- 241000186561 [Clostridium] clostridioforme Species 0.000 description 1
- 241000696045 [Clostridium] glycyrrhizinilyticum Species 0.000 description 1
- 241000030493 [Clostridium] hylemonae Species 0.000 description 1
- 241000985249 [Clostridium] indolis Species 0.000 description 1
- 241001098250 [Clostridium] lavalense Species 0.000 description 1
- 241001656794 [Clostridium] saccharolyticum Species 0.000 description 1
- 241001147801 [Clostridium] scindens Species 0.000 description 1
- 241001147717 [Clostridium] sphenoides Species 0.000 description 1
- 241000186586 [Clostridium] sporosphaeroides Species 0.000 description 1
- 241000193450 [Clostridium] symbiosum Species 0.000 description 1
- 241000215449 [Clostridium] viride Species 0.000 description 1
- 241001147712 [Clostridium] xylanolyticum Species 0.000 description 1
- 241001531273 [Eubacterium] eligens Species 0.000 description 1
- 241001531197 [Eubacterium] hallii Species 0.000 description 1
- 241001531189 [Eubacterium] siraeum Species 0.000 description 1
- 241001464867 [Ruminococcus] gnavus Species 0.000 description 1
- 208000020560 abdominal swelling Diseases 0.000 description 1
- 235000010489 acacia gum Nutrition 0.000 description 1
- 239000000205 acacia gum Substances 0.000 description 1
- WDJHALXBUFZDSR-UHFFFAOYSA-M acetoacetate Chemical compound CC(=O)CC([O-])=O WDJHALXBUFZDSR-UHFFFAOYSA-M 0.000 description 1
- 230000002378 acidificating effect Effects 0.000 description 1
- 239000004480 active ingredient Substances 0.000 description 1
- 239000013543 active substance Substances 0.000 description 1
- 229940072056 alginate Drugs 0.000 description 1
- 239000000783 alginic acid Substances 0.000 description 1
- 229960001126 alginic acid Drugs 0.000 description 1
- 150000004781 alginic acids Chemical class 0.000 description 1
- 230000000735 allogeneic effect Effects 0.000 description 1
- 239000008168 almond oil Substances 0.000 description 1
- 208000004631 alopecia areata Diseases 0.000 description 1
- HDTRYLNUVZCQOY-LIZSDCNHSA-N alpha,alpha-trehalose Chemical compound O[C@@H]1[C@@H](O)[C@H](O)[C@@H](CO)O[C@@H]1O[C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](CO)O1 HDTRYLNUVZCQOY-LIZSDCNHSA-N 0.000 description 1
- WQZGKKKJIJFFOK-PHYPRBDBSA-N alpha-D-galactose Chemical compound OC[C@H]1O[C@H](O)[C@H](O)[C@@H](O)[C@H]1O WQZGKKKJIJFFOK-PHYPRBDBSA-N 0.000 description 1
- 230000004075 alteration Effects 0.000 description 1
- XAGFODPZIPBFFR-UHFFFAOYSA-N aluminium Chemical compound [Al] XAGFODPZIPBFFR-UHFFFAOYSA-N 0.000 description 1
- 229910052782 aluminium Inorganic materials 0.000 description 1
- 206010002022 amyloidosis Diseases 0.000 description 1
- 208000003455 anaphylaxis Diseases 0.000 description 1
- 150000001450 anions Chemical class 0.000 description 1
- 239000002246 antineoplastic agent Substances 0.000 description 1
- 239000008135 aqueous vehicle Substances 0.000 description 1
- 235000019312 arabinogalactan Nutrition 0.000 description 1
- 208000010216 atopic IgE responsiveness Diseases 0.000 description 1
- 208000010668 atopic eczema Diseases 0.000 description 1
- 201000000448 autoimmune hemolytic anemia Diseases 0.000 description 1
- 208000006424 autoimmune oophoritis Diseases 0.000 description 1
- 201000009780 autoimmune polyendocrine syndrome type 2 Diseases 0.000 description 1
- 206010071578 autoimmune retinopathy Diseases 0.000 description 1
- 208000010928 autoimmune thyroid disease Diseases 0.000 description 1
- 230000003376 axonal effect Effects 0.000 description 1
- 235000008452 baby food Nutrition 0.000 description 1
- 239000011425 bamboo Substances 0.000 description 1
- 230000004888 barrier function Effects 0.000 description 1
- 239000011324 bead Substances 0.000 description 1
- 230000008901 benefit Effects 0.000 description 1
- 239000003613 bile acid Substances 0.000 description 1
- 210000000013 bile duct Anatomy 0.000 description 1
- 239000011230 binding agent Substances 0.000 description 1
- 238000002306 biochemical method Methods 0.000 description 1
- 229920002988 biodegradable polymer Polymers 0.000 description 1
- 239000004621 biodegradable polymer Substances 0.000 description 1
- 238000004820 blood count Methods 0.000 description 1
- 235000008429 bread Nutrition 0.000 description 1
- 210000000481 breast Anatomy 0.000 description 1
- 235000010634 bubble gum Nutrition 0.000 description 1
- 239000006189 buccal tablet Substances 0.000 description 1
- 239000000337 buffer salt Substances 0.000 description 1
- 239000006172 buffering agent Substances 0.000 description 1
- 239000004067 bulking agent Substances 0.000 description 1
- 208000000594 bullous pemphigoid Diseases 0.000 description 1
- CRFNGMNYKDXRTN-CITAKDKDSA-N butyryl-CoA Chemical compound O[C@@H]1[C@H](OP(O)(O)=O)[C@@H](COP(O)(=O)OP(O)(=O)OCC(C)(C)[C@@H](O)C(=O)NCCC(=O)NCCSC(=O)CCC)O[C@H]1N1C2=NC=NC(N)=C2N=C1 CRFNGMNYKDXRTN-CITAKDKDSA-N 0.000 description 1
- 235000010410 calcium alginate Nutrition 0.000 description 1
- 239000000648 calcium alginate Substances 0.000 description 1
- 229960002681 calcium alginate Drugs 0.000 description 1
- NKWPZUCBCARRDP-UHFFFAOYSA-L calcium bicarbonate Chemical compound [Ca+2].OC([O-])=O.OC([O-])=O NKWPZUCBCARRDP-UHFFFAOYSA-L 0.000 description 1
- 229910000020 calcium bicarbonate Inorganic materials 0.000 description 1
- FUFJGUQYACFECW-UHFFFAOYSA-L calcium hydrogenphosphate Chemical compound [Ca+2].OP([O-])([O-])=O FUFJGUQYACFECW-UHFFFAOYSA-L 0.000 description 1
- 239000001506 calcium phosphate Substances 0.000 description 1
- 229910000389 calcium phosphate Inorganic materials 0.000 description 1
- 235000011010 calcium phosphates Nutrition 0.000 description 1
- OKHHGHGGPDJQHR-YMOPUZKJSA-L calcium;(2s,3s,4s,5s,6r)-6-[(2r,3s,4r,5s,6r)-2-carboxy-6-[(2r,3s,4r,5s,6r)-2-carboxylato-4,5,6-trihydroxyoxan-3-yl]oxy-4,5-dihydroxyoxan-3-yl]oxy-3,4,5-trihydroxyoxane-2-carboxylate Chemical compound [Ca+2].O[C@@H]1[C@H](O)[C@H](O)O[C@@H](C([O-])=O)[C@H]1O[C@H]1[C@@H](O)[C@@H](O)[C@H](O[C@H]2[C@H]([C@@H](O)[C@H](O)[C@H](O2)C([O-])=O)O)[C@H](C(O)=O)O1 OKHHGHGGPDJQHR-YMOPUZKJSA-L 0.000 description 1
- 238000004422 calculation algorithm Methods 0.000 description 1
- 238000004364 calculation method Methods 0.000 description 1
- 235000010948 carboxy methyl cellulose Nutrition 0.000 description 1
- 239000001768 carboxy methyl cellulose Substances 0.000 description 1
- 239000008112 carboxymethyl-cellulose Substances 0.000 description 1
- 210000000748 cardiovascular system Anatomy 0.000 description 1
- 239000000969 carrier Substances 0.000 description 1
- 230000015556 catabolic process Effects 0.000 description 1
- 125000002091 cationic group Chemical class 0.000 description 1
- 150000001768 cations Chemical class 0.000 description 1
- 230000001413 cellular effect Effects 0.000 description 1
- 229940081734 cellulose acetate phthalate Drugs 0.000 description 1
- 238000007385 chemical modification Methods 0.000 description 1
- 235000019693 cherries Nutrition 0.000 description 1
- 239000007910 chewable tablet Substances 0.000 description 1
- 229940068682 chewable tablet Drugs 0.000 description 1
- 229940112822 chewing gum Drugs 0.000 description 1
- 235000015218 chewing gum Nutrition 0.000 description 1
- 235000019219 chocolate Nutrition 0.000 description 1
- 208000013116 chronic cough Diseases 0.000 description 1
- 208000025302 chronic primary adrenal insufficiency Diseases 0.000 description 1
- 201000010002 cicatricial pemphigoid Diseases 0.000 description 1
- 235000020971 citrus fruits Nutrition 0.000 description 1
- 229920001688 coating polymer Polymers 0.000 description 1
- 229940110456 cocoa butter Drugs 0.000 description 1
- 235000019868 cocoa butter Nutrition 0.000 description 1
- 208000008609 collagenous colitis Diseases 0.000 description 1
- 208000029742 colonic neoplasm Diseases 0.000 description 1
- 239000003086 colorant Substances 0.000 description 1
- 238000004040 coloring Methods 0.000 description 1
- 201000004395 congenital heart block Diseases 0.000 description 1
- 239000000356 contaminant Substances 0.000 description 1
- 230000036461 convulsion Effects 0.000 description 1
- 238000012937 correction Methods 0.000 description 1
- 201000003278 cryoglobulinemia Diseases 0.000 description 1
- 235000015140 cultured milk Nutrition 0.000 description 1
- 230000006341 curative response Effects 0.000 description 1
- 230000001351 cycling effect Effects 0.000 description 1
- 238000006731 degradation reaction Methods 0.000 description 1
- 230000000593 degrading effect Effects 0.000 description 1
- 230000003111 delayed effect Effects 0.000 description 1
- 230000003210 demyelinating effect Effects 0.000 description 1
- 208000002925 dental caries Diseases 0.000 description 1
- 230000001419 dependent effect Effects 0.000 description 1
- 238000011161 development Methods 0.000 description 1
- 230000018109 developmental process Effects 0.000 description 1
- 235000019425 dextrin Nutrition 0.000 description 1
- 206010012601 diabetes mellitus Diseases 0.000 description 1
- 238000003745 diagnosis Methods 0.000 description 1
- 235000019700 dicalcium phosphate Nutrition 0.000 description 1
- 235000015872 dietary supplement Nutrition 0.000 description 1
- 239000003085 diluting agent Substances 0.000 description 1
- 238000007907 direct compression Methods 0.000 description 1
- 239000007884 disintegrant Substances 0.000 description 1
- 239000007919 dispersible tablet Substances 0.000 description 1
- 239000002270 dispersing agent Substances 0.000 description 1
- 201000008243 diversion colitis Diseases 0.000 description 1
- 235000021113 dry cheese Nutrition 0.000 description 1
- 206010013781 dry mouth Diseases 0.000 description 1
- 210000001198 duodenum Anatomy 0.000 description 1
- 208000019479 dysautonomia Diseases 0.000 description 1
- 208000001848 dysentery Diseases 0.000 description 1
- 230000002526 effect on cardiovascular system Effects 0.000 description 1
- 239000007938 effervescent tablet Substances 0.000 description 1
- 235000013601 eggs Nutrition 0.000 description 1
- 239000003995 emulsifying agent Substances 0.000 description 1
- 230000001804 emulsifying effect Effects 0.000 description 1
- 239000000839 emulsion Substances 0.000 description 1
- 206010014599 encephalitis Diseases 0.000 description 1
- 239000007920 enema Substances 0.000 description 1
- 229940079360 enema for constipation Drugs 0.000 description 1
- 239000003623 enhancer Substances 0.000 description 1
- 239000002662 enteric coated tablet Substances 0.000 description 1
- 239000002702 enteric coating Substances 0.000 description 1
- 238000009505 enteric coating Methods 0.000 description 1
- 201000000708 eosinophilic esophagitis Diseases 0.000 description 1
- 210000003238 esophagus Anatomy 0.000 description 1
- 150000002148 esters Chemical class 0.000 description 1
- 235000019441 ethanol Nutrition 0.000 description 1
- BEFDCLMNVWHSGT-UHFFFAOYSA-N ethenylcyclopentane Chemical compound C=CC1CCCC1 BEFDCLMNVWHSGT-UHFFFAOYSA-N 0.000 description 1
- 210000003527 eukaryotic cell Anatomy 0.000 description 1
- 210000001808 exosome Anatomy 0.000 description 1
- 238000000605 extraction Methods 0.000 description 1
- 208000002980 facial hemiatrophy Diseases 0.000 description 1
- 239000003925 fat Substances 0.000 description 1
- 235000019197 fats Nutrition 0.000 description 1
- 235000021107 fermented food Nutrition 0.000 description 1
- 239000000706 filtrate Substances 0.000 description 1
- 235000019688 fish Nutrition 0.000 description 1
- 230000003890 fistula Effects 0.000 description 1
- 239000013568 food allergen Substances 0.000 description 1
- 238000005755 formation reaction Methods 0.000 description 1
- 238000004108 freeze drying Methods 0.000 description 1
- 235000011389 fruit/vegetable juice Nutrition 0.000 description 1
- 230000033581 fucosylation Effects 0.000 description 1
- 230000002538 fungal effect Effects 0.000 description 1
- ZZUFCTLCJUWOSV-UHFFFAOYSA-N furosemide Chemical compound C1=C(Cl)C(S(=O)(=O)N)=CC(C(O)=O)=C1NCC1=CC=CO1 ZZUFCTLCJUWOSV-UHFFFAOYSA-N 0.000 description 1
- 230000030136 gastric emptying Effects 0.000 description 1
- 208000018090 giant cell myocarditis Diseases 0.000 description 1
- 208000024693 gingival disease Diseases 0.000 description 1
- 229940046240 glucomannan Drugs 0.000 description 1
- 235000001727 glucose Nutrition 0.000 description 1
- 125000005456 glyceride group Chemical group 0.000 description 1
- 238000000227 grinding Methods 0.000 description 1
- 235000010417 guar gum Nutrition 0.000 description 1
- 239000000665 guar gum Substances 0.000 description 1
- 229960002154 guar gum Drugs 0.000 description 1
- 229920000591 gum Polymers 0.000 description 1
- 230000007149 gut brain axis pathway Effects 0.000 description 1
- 208000024963 hair loss Diseases 0.000 description 1
- 230000003676 hair loss Effects 0.000 description 1
- 239000007902 hard capsule Substances 0.000 description 1
- 230000008821 health effect Effects 0.000 description 1
- 208000035861 hematochezia Diseases 0.000 description 1
- 238000011134 hematopoietic stem cell transplantation Methods 0.000 description 1
- 208000007475 hemolytic anemia Diseases 0.000 description 1
- 239000012676 herbal extract Substances 0.000 description 1
- 238000012165 high-throughput sequencing Methods 0.000 description 1
- HNDVDQJCIGZPNO-UHFFFAOYSA-N histidine Natural products OC(=O)C(N)CC1=CN=CN1 HNDVDQJCIGZPNO-UHFFFAOYSA-N 0.000 description 1
- 238000001794 hormone therapy Methods 0.000 description 1
- 239000000416 hydrocolloid Substances 0.000 description 1
- 150000004679 hydroxides Chemical class 0.000 description 1
- 201000006362 hypersensitivity vasculitis Diseases 0.000 description 1
- 230000036543 hypotension Effects 0.000 description 1
- 210000003405 ileum Anatomy 0.000 description 1
- 239000012729 immediate-release (IR) formulation Substances 0.000 description 1
- 230000001900 immune effect Effects 0.000 description 1
- 230000008102 immune modulation Effects 0.000 description 1
- 239000012274 immune-checkpoint protein inhibitor Substances 0.000 description 1
- 230000007813 immunodeficiency Effects 0.000 description 1
- 230000002163 immunogen Effects 0.000 description 1
- 208000015446 immunoglobulin a vasculitis Diseases 0.000 description 1
- 230000004957 immunoregulator effect Effects 0.000 description 1
- 239000012535 impurity Substances 0.000 description 1
- 238000000099 in vitro assay Methods 0.000 description 1
- 238000005462 in vivo assay Methods 0.000 description 1
- 201000008319 inclusion body myositis Diseases 0.000 description 1
- 238000010348 incorporation Methods 0.000 description 1
- 239000003701 inert diluent Substances 0.000 description 1
- 230000002458 infectious effect Effects 0.000 description 1
- 208000027866 inflammatory disease Diseases 0.000 description 1
- 238000001802 infusion Methods 0.000 description 1
- 239000004615 ingredient Substances 0.000 description 1
- 238000002347 injection Methods 0.000 description 1
- 239000007924 injection Substances 0.000 description 1
- 208000014674 injury Diseases 0.000 description 1
- 238000011081 inoculation Methods 0.000 description 1
- 239000002198 insoluble material Substances 0.000 description 1
- 239000000543 intermediate Substances 0.000 description 1
- 208000036971 interstitial lung disease 2 Diseases 0.000 description 1
- 210000000936 intestine Anatomy 0.000 description 1
- JYJIGFIDKWBXDU-MNNPPOADSA-N inulin Chemical compound O[C@H]1[C@H](O)[C@@H](CO)O[C@@]1(CO)OC[C@]1(OC[C@]2(OC[C@]3(OC[C@]4(OC[C@]5(OC[C@]6(OC[C@]7(OC[C@]8(OC[C@]9(OC[C@]%10(OC[C@]%11(OC[C@]%12(OC[C@]%13(OC[C@]%14(OC[C@]%15(OC[C@]%16(OC[C@]%17(OC[C@]%18(OC[C@]%19(OC[C@]%20(OC[C@]%21(OC[C@]%22(OC[C@]%23(OC[C@]%24(OC[C@]%25(OC[C@]%26(OC[C@]%27(OC[C@]%28(OC[C@]%29(OC[C@]%30(OC[C@]%31(OC[C@]%32(OC[C@]%33(OC[C@]%34(OC[C@]%35(OC[C@]%36(O[C@@H]%37[C@@H]([C@@H](O)[C@H](O)[C@@H](CO)O%37)O)[C@H]([C@H](O)[C@@H](CO)O%36)O)[C@H]([C@H](O)[C@@H](CO)O%35)O)[C@H]([C@H](O)[C@@H](CO)O%34)O)[C@H]([C@H](O)[C@@H](CO)O%33)O)[C@H]([C@H](O)[C@@H](CO)O%32)O)[C@H]([C@H](O)[C@@H](CO)O%31)O)[C@H]([C@H](O)[C@@H](CO)O%30)O)[C@H]([C@H](O)[C@@H](CO)O%29)O)[C@H]([C@H](O)[C@@H](CO)O%28)O)[C@H]([C@H](O)[C@@H](CO)O%27)O)[C@H]([C@H](O)[C@@H](CO)O%26)O)[C@H]([C@H](O)[C@@H](CO)O%25)O)[C@H]([C@H](O)[C@@H](CO)O%24)O)[C@H]([C@H](O)[C@@H](CO)O%23)O)[C@H]([C@H](O)[C@@H](CO)O%22)O)[C@H]([C@H](O)[C@@H](CO)O%21)O)[C@H]([C@H](O)[C@@H](CO)O%20)O)[C@H]([C@H](O)[C@@H](CO)O%19)O)[C@H]([C@H](O)[C@@H](CO)O%18)O)[C@H]([C@H](O)[C@@H](CO)O%17)O)[C@H]([C@H](O)[C@@H](CO)O%16)O)[C@H]([C@H](O)[C@@H](CO)O%15)O)[C@H]([C@H](O)[C@@H](CO)O%14)O)[C@H]([C@H](O)[C@@H](CO)O%13)O)[C@H]([C@H](O)[C@@H](CO)O%12)O)[C@H]([C@H](O)[C@@H](CO)O%11)O)[C@H]([C@H](O)[C@@H](CO)O%10)O)[C@H]([C@H](O)[C@@H](CO)O9)O)[C@H]([C@H](O)[C@@H](CO)O8)O)[C@H]([C@H](O)[C@@H](CO)O7)O)[C@H]([C@H](O)[C@@H](CO)O6)O)[C@H]([C@H](O)[C@@H](CO)O5)O)[C@H]([C@H](O)[C@@H](CO)O4)O)[C@H]([C@H](O)[C@@H](CO)O3)O)[C@H]([C@H](O)[C@@H](CO)O2)O)[C@@H](O)[C@H](O)[C@@H](CO)O1 JYJIGFIDKWBXDU-MNNPPOADSA-N 0.000 description 1
- 229940029339 inulin Drugs 0.000 description 1
- JJWLVOIRVHMVIS-UHFFFAOYSA-N isopropylamine Chemical compound CC(C)N JJWLVOIRVHMVIS-UHFFFAOYSA-N 0.000 description 1
- 230000007803 itching Effects 0.000 description 1
- 235000015110 jellies Nutrition 0.000 description 1
- 239000008274 jelly Substances 0.000 description 1
- 201000002215 juvenile rheumatoid arthritis Diseases 0.000 description 1
- 235000015141 kefir Nutrition 0.000 description 1
- 239000000832 lactitol Substances 0.000 description 1
- 235000010448 lactitol Nutrition 0.000 description 1
- VQHSOMBJVWLPSR-JVCRWLNRSA-N lactitol Chemical compound OC[C@H](O)[C@@H](O)[C@@H]([C@H](O)CO)O[C@@H]1O[C@H](CO)[C@H](O)[C@H](O)[C@H]1O VQHSOMBJVWLPSR-JVCRWLNRSA-N 0.000 description 1
- 229960003451 lactitol Drugs 0.000 description 1
- 108010088312 lacto-N-biosidase Proteins 0.000 description 1
- JCQLYHFGKNRPGE-FCVZTGTOSA-N lactulose Chemical compound OC[C@H]1O[C@](O)(CO)[C@@H](O)[C@@H]1O[C@H]1[C@H](O)[C@@H](O)[C@@H](O)[C@@H](CO)O1 JCQLYHFGKNRPGE-FCVZTGTOSA-N 0.000 description 1
- 229960000511 lactulose Drugs 0.000 description 1
- PFCRQPBOOFTZGQ-UHFFFAOYSA-N lactulose keto form Natural products OCC(=O)C(O)C(C(O)CO)OC1OC(CO)C(O)C(O)C1O PFCRQPBOOFTZGQ-UHFFFAOYSA-N 0.000 description 1
- 210000002429 large intestine Anatomy 0.000 description 1
- 239000012633 leachable Substances 0.000 description 1
- 235000010445 lecithin Nutrition 0.000 description 1
- 239000000787 lecithin Substances 0.000 description 1
- 229940067606 lecithin Drugs 0.000 description 1
- 201000011486 lichen planus Diseases 0.000 description 1
- 206010071570 ligneous conjunctivitis Diseases 0.000 description 1
- 230000000670 limiting effect Effects 0.000 description 1
- 150000002632 lipids Chemical class 0.000 description 1
- 239000002502 liposome Substances 0.000 description 1
- 239000012669 liquid formulation Substances 0.000 description 1
- 210000004185 liver Anatomy 0.000 description 1
- 208000019423 liver disease Diseases 0.000 description 1
- 239000000711 locust bean gum Substances 0.000 description 1
- 238000001325 log-rank test Methods 0.000 description 1
- 235000011475 lollipops Nutrition 0.000 description 1
- 239000000314 lubricant Substances 0.000 description 1
- 208000004341 lymphocytic colitis Diseases 0.000 description 1
- 235000019359 magnesium stearate Nutrition 0.000 description 1
- 229940057948 magnesium stearate Drugs 0.000 description 1
- 230000014759 maintenance of location Effects 0.000 description 1
- 235000010355 mannitol Nutrition 0.000 description 1
- 239000000594 mannitol Substances 0.000 description 1
- 239000011159 matrix material Substances 0.000 description 1
- 210000001809 melena Anatomy 0.000 description 1
- 230000002503 metabolic effect Effects 0.000 description 1
- 230000037353 metabolic pathway Effects 0.000 description 1
- 229920000609 methyl cellulose Polymers 0.000 description 1
- 125000002496 methyl group Chemical group [H]C([H])([H])* 0.000 description 1
- 235000010270 methyl p-hydroxybenzoate Nutrition 0.000 description 1
- 235000010981 methylcellulose Nutrition 0.000 description 1
- 239000001923 methylcellulose Substances 0.000 description 1
- 230000000813 microbial effect Effects 0.000 description 1
- 230000002906 microbiologic effect Effects 0.000 description 1
- 229940016286 microcrystalline cellulose Drugs 0.000 description 1
- 235000019813 microcrystalline cellulose Nutrition 0.000 description 1
- 239000008108 microcrystalline cellulose Substances 0.000 description 1
- 206010063344 microscopic polyangiitis Diseases 0.000 description 1
- 235000020166 milkshake Nutrition 0.000 description 1
- 208000018962 mouth sore Diseases 0.000 description 1
- 238000011201 multiple comparisons test Methods 0.000 description 1
- 201000006417 multiple sclerosis Diseases 0.000 description 1
- 229960003128 mupirocin Drugs 0.000 description 1
- 229930187697 mupirocin Natural products 0.000 description 1
- DDHVILIIHBIMQU-YJGQQKNPSA-L mupirocin calcium hydrate Chemical compound O.O.[Ca+2].C[C@H](O)[C@H](C)[C@@H]1O[C@H]1C[C@@H]1[C@@H](O)[C@@H](O)[C@H](C\C(C)=C\C(=O)OCCCCCCCCC([O-])=O)OC1.C[C@H](O)[C@H](C)[C@@H]1O[C@H]1C[C@@H]1[C@@H](O)[C@@H](O)[C@H](C\C(C)=C\C(=O)OCCCCCCCCC([O-])=O)OC1 DDHVILIIHBIMQU-YJGQQKNPSA-L 0.000 description 1
- 206010028417 myasthenia gravis Diseases 0.000 description 1
- 229950006780 n-acetylglucosamine Drugs 0.000 description 1
- 201000003631 narcolepsy Diseases 0.000 description 1
- 201000008383 nephritis Diseases 0.000 description 1
- 208000015122 neurodegenerative disease Diseases 0.000 description 1
- 230000000626 neurodegenerative effect Effects 0.000 description 1
- 230000001123 neurodevelopmental effect Effects 0.000 description 1
- 201000001119 neuropathy Diseases 0.000 description 1
- 230000007823 neuropathy Effects 0.000 description 1
- 208000004235 neutropenia Diseases 0.000 description 1
- 239000002687 nonaqueous vehicle Substances 0.000 description 1
- 235000015097 nutrients Nutrition 0.000 description 1
- 230000035764 nutrition Effects 0.000 description 1
- 235000014571 nuts Nutrition 0.000 description 1
- QIQXTHQIDYTFRH-UHFFFAOYSA-N octadecanoic acid Chemical compound CCCCCCCCCCCCCCCCCC(O)=O QIQXTHQIDYTFRH-UHFFFAOYSA-N 0.000 description 1
- OQCDKBAXFALNLD-UHFFFAOYSA-N octadecanoic acid Natural products CCCCCCCC(C)CCCCCCCCC(O)=O OQCDKBAXFALNLD-UHFFFAOYSA-N 0.000 description 1
- CXQXSVUQTKDNFP-UHFFFAOYSA-N octamethyltrisiloxane Chemical compound C[Si](C)(C)O[Si](C)(C)O[Si](C)(C)C CXQXSVUQTKDNFP-UHFFFAOYSA-N 0.000 description 1
- 208000015200 ocular cicatricial pemphigoid Diseases 0.000 description 1
- 229940095353 oral granules Drugs 0.000 description 1
- 229940042126 oral powder Drugs 0.000 description 1
- 239000006191 orally-disintegrating tablet Substances 0.000 description 1
- 238000001139 pH measurement Methods 0.000 description 1
- 235000019629 palatability Nutrition 0.000 description 1
- 201000005580 palindromic rheumatism Diseases 0.000 description 1
- 208000014965 pancolitis Diseases 0.000 description 1
- 230000007918 pathogenicity Effects 0.000 description 1
- 230000004963 pathophysiological condition Effects 0.000 description 1
- 235000020232 peanut Nutrition 0.000 description 1
- 230000000149 penetrating effect Effects 0.000 description 1
- 230000035515 penetration Effects 0.000 description 1
- 208000033808 peripheral neuropathy Diseases 0.000 description 1
- 230000035699 permeability Effects 0.000 description 1
- 239000008196 pharmacological composition Substances 0.000 description 1
- CTYRPMDGLDAWRQ-UHFFFAOYSA-N phenyl hydrogen sulfate Chemical compound OS(=O)(=O)OC1=CC=CC=C1 CTYRPMDGLDAWRQ-UHFFFAOYSA-N 0.000 description 1
- 230000000704 physical effect Effects 0.000 description 1
- 230000035790 physiological processes and functions Effects 0.000 description 1
- 239000000419 plant extract Substances 0.000 description 1
- 238000004987 plasma desorption mass spectroscopy Methods 0.000 description 1
- 229920000191 poly(N-vinyl pyrrolidone) Polymers 0.000 description 1
- 229920001200 poly(ethylene-vinyl acetate) Polymers 0.000 description 1
- 229920003229 poly(methyl methacrylate) Polymers 0.000 description 1
- 201000006292 polyarteritis nodosa Diseases 0.000 description 1
- 235000013856 polydextrose Nutrition 0.000 description 1
- 239000001259 polydextrose Substances 0.000 description 1
- 229940035035 polydextrose Drugs 0.000 description 1
- 239000004633 polyglycolic acid Substances 0.000 description 1
- 229920002338 polyhydroxyethylmethacrylate Polymers 0.000 description 1
- 229920000193 polymethacrylate Polymers 0.000 description 1
- 239000004926 polymethyl methacrylate Substances 0.000 description 1
- 208000005987 polymyositis Diseases 0.000 description 1
- 235000010486 polyoxyethylene sorbitan monolaurate Nutrition 0.000 description 1
- 239000000256 polyoxyethylene sorbitan monolaurate Substances 0.000 description 1
- 229920001282 polysaccharide Polymers 0.000 description 1
- 239000005017 polysaccharide Substances 0.000 description 1
- 150000004804 polysaccharides Chemical class 0.000 description 1
- 229940068977 polysorbate 20 Drugs 0.000 description 1
- 229920002451 polyvinyl alcohol Polymers 0.000 description 1
- 239000011591 potassium Substances 0.000 description 1
- 229910052700 potassium Inorganic materials 0.000 description 1
- 229940116317 potato starch Drugs 0.000 description 1
- 239000002243 precursor Substances 0.000 description 1
- 230000002028 premature Effects 0.000 description 1
- 239000003755 preservative agent Substances 0.000 description 1
- 238000002203 pretreatment Methods 0.000 description 1
- 208000018290 primary dysautonomia Diseases 0.000 description 1
- 201000000742 primary sclerosing cholangitis Diseases 0.000 description 1
- MFDFERRIHVXMIY-UHFFFAOYSA-N procaine Chemical compound CCN(CC)CCOC(=O)C1=CC=C(N)C=C1 MFDFERRIHVXMIY-UHFFFAOYSA-N 0.000 description 1
- 229960004919 procaine Drugs 0.000 description 1
- 206010036784 proctocolitis Diseases 0.000 description 1
- 208000017048 proctosigmoiditis Diseases 0.000 description 1
- 229960003387 progesterone Drugs 0.000 description 1
- 239000000186 progesterone Substances 0.000 description 1
- 235000010232 propyl p-hydroxybenzoate Nutrition 0.000 description 1
- QELSKZZBTMNZEB-UHFFFAOYSA-N propylparaben Chemical class CCCOC(=O)C1=CC=C(O)C=C1 QELSKZZBTMNZEB-UHFFFAOYSA-N 0.000 description 1
- 229940070687 psyllium Drugs 0.000 description 1
- 235000011962 puddings Nutrition 0.000 description 1
- 208000005069 pulmonary fibrosis Diseases 0.000 description 1
- 230000000541 pulsatile effect Effects 0.000 description 1
- 208000009954 pyoderma gangrenosum Diseases 0.000 description 1
- 230000005855 radiation Effects 0.000 description 1
- 238000001959 radiotherapy Methods 0.000 description 1
- 210000000664 rectum Anatomy 0.000 description 1
- 230000000306 recurrent effect Effects 0.000 description 1
- 230000009467 reduction Effects 0.000 description 1
- 208000009169 relapsing polychondritis Diseases 0.000 description 1
- 230000000241 respiratory effect Effects 0.000 description 1
- 230000002441 reversible effect Effects 0.000 description 1
- 230000000552 rheumatic effect Effects 0.000 description 1
- 201000003068 rheumatic fever Diseases 0.000 description 1
- 206010039073 rheumatoid arthritis Diseases 0.000 description 1
- 206010039083 rhinitis Diseases 0.000 description 1
- 229940100486 rice starch Drugs 0.000 description 1
- 201000000306 sarcoidosis Diseases 0.000 description 1
- 208000010157 sclerosing cholangitis Diseases 0.000 description 1
- 238000000926 separation method Methods 0.000 description 1
- 235000015170 shellfish Nutrition 0.000 description 1
- SQVRNKJHWKZAKO-OQPLDHBCSA-N sialic acid Chemical compound CC(=O)N[C@@H]1[C@@H](O)C[C@@](O)(C(O)=O)OC1[C@H](O)[C@H](O)CO SQVRNKJHWKZAKO-OQPLDHBCSA-N 0.000 description 1
- 230000009450 sialylation Effects 0.000 description 1
- 208000017520 skin disease Diseases 0.000 description 1
- 206010040872 skin infection Diseases 0.000 description 1
- 239000002002 slurry Substances 0.000 description 1
- 210000000813 small intestine Anatomy 0.000 description 1
- 206010041232 sneezing Diseases 0.000 description 1
- 235000015424 sodium Nutrition 0.000 description 1
- 235000010413 sodium alginate Nutrition 0.000 description 1
- 239000000661 sodium alginate Substances 0.000 description 1
- 229940005550 sodium alginate Drugs 0.000 description 1
- WXMKPNITSTVMEF-UHFFFAOYSA-M sodium benzoate Chemical compound [Na+].[O-]C(=O)C1=CC=CC=C1 WXMKPNITSTVMEF-UHFFFAOYSA-M 0.000 description 1
- 235000010234 sodium benzoate Nutrition 0.000 description 1
- 239000004299 sodium benzoate Substances 0.000 description 1
- 229960003885 sodium benzoate Drugs 0.000 description 1
- 235000017557 sodium bicarbonate Nutrition 0.000 description 1
- 229910000030 sodium bicarbonate Inorganic materials 0.000 description 1
- 239000011780 sodium chloride Substances 0.000 description 1
- 229920003109 sodium starch glycolate Polymers 0.000 description 1
- 239000008109 sodium starch glycolate Substances 0.000 description 1
- 229940079832 sodium starch glycolate Drugs 0.000 description 1
- 239000007901 soft capsule Substances 0.000 description 1
- 239000012439 solid excipient Substances 0.000 description 1
- 235000010199 sorbic acid Nutrition 0.000 description 1
- 239000004334 sorbic acid Substances 0.000 description 1
- 229940075582 sorbic acid Drugs 0.000 description 1
- 235000021262 sour milk Nutrition 0.000 description 1
- 235000011496 sports drink Nutrition 0.000 description 1
- 239000007921 spray Substances 0.000 description 1
- 238000001694 spray drying Methods 0.000 description 1
- 239000003381 stabilizer Substances 0.000 description 1
- 230000000087 stabilizing effect Effects 0.000 description 1
- 238000007619 statistical method Methods 0.000 description 1
- 239000008117 stearic acid Substances 0.000 description 1
- 239000008223 sterile water Substances 0.000 description 1
- 238000003860 storage Methods 0.000 description 1
- 239000006228 supernatant Substances 0.000 description 1
- 239000000829 suppository Substances 0.000 description 1
- 239000002511 suppository base Substances 0.000 description 1
- 230000008961 swelling Effects 0.000 description 1
- 206010042772 syncope Diseases 0.000 description 1
- 201000000596 systemic lupus erythematosus Diseases 0.000 description 1
- 230000006794 tachycardia Effects 0.000 description 1
- 239000000454 talc Substances 0.000 description 1
- 229910052623 talc Inorganic materials 0.000 description 1
- 229940033134 talc Drugs 0.000 description 1
- 235000012222 talc Nutrition 0.000 description 1
- 235000010491 tara gum Nutrition 0.000 description 1
- 239000000213 tara gum Substances 0.000 description 1
- 238000002626 targeted therapy Methods 0.000 description 1
- 230000008685 targeting Effects 0.000 description 1
- 235000002906 tartaric acid Nutrition 0.000 description 1
- 238000010257 thawing Methods 0.000 description 1
- 231100001274 therapeutic index Toxicity 0.000 description 1
- 230000008719 thickening Effects 0.000 description 1
- 239000002562 thickening agent Substances 0.000 description 1
- 230000000699 topical effect Effects 0.000 description 1
- 235000021476 total parenteral nutrition Nutrition 0.000 description 1
- 230000001988 toxicity Effects 0.000 description 1
- 231100000419 toxicity Toxicity 0.000 description 1
- 235000010487 tragacanth Nutrition 0.000 description 1
- 239000000196 tragacanth Substances 0.000 description 1
- 229940116362 tragacanth Drugs 0.000 description 1
- 230000009261 transgenic effect Effects 0.000 description 1
- 230000005945 translocation Effects 0.000 description 1
- 208000009174 transverse myelitis Diseases 0.000 description 1
- QORWJWZARLRLPR-UHFFFAOYSA-H tricalcium bis(phosphate) Chemical compound [Ca+2].[Ca+2].[Ca+2].[O-]P([O-])([O-])=O.[O-]P([O-])([O-])=O QORWJWZARLRLPR-UHFFFAOYSA-H 0.000 description 1
- 229940086542 triethylamine Drugs 0.000 description 1
- 210000004881 tumor cell Anatomy 0.000 description 1
- 230000002568 urticarial effect Effects 0.000 description 1
- 229940070710 valerate Drugs 0.000 description 1
- NQPDZGIKBAWPEJ-UHFFFAOYSA-N valeric acid Chemical compound CCCCC(O)=O NQPDZGIKBAWPEJ-UHFFFAOYSA-N 0.000 description 1
- 210000001835 viscera Anatomy 0.000 description 1
- 235000013343 vitamin Nutrition 0.000 description 1
- 239000011782 vitamin Substances 0.000 description 1
- 229940088594 vitamin Drugs 0.000 description 1
- 229930003231 vitamin Natural products 0.000 description 1
- 235000012431 wafers Nutrition 0.000 description 1
- 230000003442 weekly effect Effects 0.000 description 1
- 239000000080 wetting agent Substances 0.000 description 1
- 229940100445 wheat starch Drugs 0.000 description 1
- 235000008939 whole milk Nutrition 0.000 description 1
- 235000010493 xanthan gum Nutrition 0.000 description 1
- 229940082509 xanthan gum Drugs 0.000 description 1
- 230000022814 xenobiotic metabolic process Effects 0.000 description 1
- 229920001221 xylan Polymers 0.000 description 1
- 150000004823 xylans Chemical class 0.000 description 1
- 239000011701 zinc Substances 0.000 description 1
- 229910052725 zinc Inorganic materials 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K35/00—Medicinal preparations containing materials or reaction products thereof with undetermined constitution
- A61K35/66—Microorganisms or materials therefrom
- A61K35/74—Bacteria
- A61K35/741—Probiotics
- A61K35/742—Spore-forming bacteria, e.g. Bacillus coagulans, Bacillus subtilis, clostridium or Lactobacillus sporogenes
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/70—Carbohydrates; Sugars; Derivatives thereof
- A61K31/702—Oligosaccharides, i.e. having three to five saccharide radicals attached to each other by glycosidic linkages
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K35/00—Medicinal preparations containing materials or reaction products thereof with undetermined constitution
- A61K35/66—Microorganisms or materials therefrom
- A61K35/74—Bacteria
- A61K35/741—Probiotics
- A61K35/744—Lactic acid bacteria, e.g. enterococci, pediococci, lactococci, streptococci or leuconostocs
- A61K35/745—Bifidobacteria
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P1/00—Drugs for disorders of the alimentary tract or the digestive system
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P29/00—Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P3/00—Drugs for disorders of the metabolism
- A61P3/04—Anorexiants; Antiobesity agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P3/00—Drugs for disorders of the metabolism
- A61P3/08—Drugs for disorders of the metabolism for glucose homeostasis
- A61P3/10—Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P37/00—Drugs for immunological or allergic disorders
- A61P37/02—Immunomodulators
Definitions
- compositions, methods, strategies, kits, and articles of manufacture that are useful, inter alia, in the treatment or prevention of diseases, disorders, or conditions that may be associated with inflammation, infection, allergy, immune dysfunction, or dysbiosis of the intestinal microbiome.
- the invention provides a synergistic combination of a prebiotic, e.g., a mixture of human milk oligosaccharides, and probiotic strains of bacteria, such as one or more strains capable of internalizing and consuming the prebiotic, e.g., Bifidobacterium longum subsp. infantis, and one or more strains capable of producing short chain fatty acids such as propionate, e.g., Veillonella sp.
- the microorganisms that colonize the human gastrointestinal tract also referred to as the gut microbiota, comprise a complex community that plays an important role in the maintenance of health and resistance to disease.
- One promising approach to treat or prevent disease is direct manipulation of the dysbiotic microbiome.
- Such therapies include live biotherapeutic products (LBPs) that may “engraft” or durably persist in the recipient gastrointestinal tract and thereby lead to positive outcomes. While LBPs conceptually hold promise as potentially effective therapeutic tools, control of their engraftment and accurate prediction of their efficacy and effects on co-resident microbes remain relatively unpredictable. Not only is selection of microbial species still a challenge, but questions remain about the duration and success of engraftment of introduced strains into the human intestinal microbiome.
- LBPs live biotherapeutic products
- compositions, kits, articles of manufacture, and methods of use thereof that are or include a prebiotic mixture, e.g., of human milk oligosaccharides, at least one Bifidobacterium, e.g., B. longum subsp. inf antis, and at least one propionate producing bacterium.
- a prebiotic mixture e.g., of human milk oligosaccharides
- Bifidobacterium e.g., B. longum subsp. inf antis
- propionate producing bacterium e.g., a propionate producing bacterium.
- the provided prebiotic mixture, Bifidobacteria, and propionate producing bacteria are, inter alia, surprisingly effective for the treatment or prevention of diseases involving an infectious or inflammatory component, or for preventing negative health effects of a dysbiotic intestinal microbiome.
- a method of treating or preventing a disease, disorder, or condition associated with one or more of inflammation, immune dysfunction, cancer, allergy, or dysbiosis of the intestinal microbiome in a subject in need thereof comprising administering to the subject i) a prebiotic mixture comprising one or more human milk oligosaccharides, ii) at least one Bifidobacterium capable of consuming the one or more human milk oligosaccharides; and iii) at least one propionate producing bacterium.
- a prebiotic mixture and probiotic strains of bacteria for the treatment or prevention of a disease, disorder, or condition associated with one or more of inflammation, immune dysfunction, cancer, allergy, or dysbiosis of the intestinal microbiome in a subject in need thereof.
- the prebiotic mixture comprises one or more human milk oligosaccharides.
- the probiotic strains of bacteria comprise at least one Bifidobacterium capable of consuming one or more human milk oligosaccharides and at least one propionate producing bacterium.
- prebiotic mixtures and probiotic strains of bacteria for use in the manufacture of medicaments for the treatment or prevention of a disease, disorder, or condition associated with one or more of inflammation, immune dysfunction, cancer, allergy, or dysbiosis of the intestinal microbiome in a subject in need thereof.
- probiotic strains of bacteria for use in the manufacture of medicaments for the treatment or prevention of a disease, disorder, or condition associated with one or more of inflammation, immune dysfunction, cancer, allergy, or dysbiosis of the intestinal microbiome to a subject in need thereof in conjunction with the administration of a prebiotic mixture.
- the prebiotic mixture comprises one or more human milk oligosaccharides.
- the probiotic strains of bacteria comprise at least one Bifidobacterium capable of consuming one or more human milk oligosaccharides and at least one propionate producing bacterium.
- a disease, disorder, or condition associated with one or more of inflammation, immune dysfunction, cancer, allergy, or dysbiosis of the intestinal microbiome in a subject in need thereof comprising administering to the subject i) a prebiotic mixture comprising one or more human milk oligosaccharides, ii) at least one Bifidobacterium capable of consuming the one or more human milk oligosaccharides; and iii) at least one propionate producing bacterium.
- the subject has received or will receive an allogenic hematopoietic stem cell transplant, and wherein the disease, condition, or disorder comprises graft versus host disease.
- the disease, disorder, or condition comprises one or more of obesity, type II diabetes, a chronic inflammatory disease, an autoimmune disease, an infection, an infectious disease domination, bowel resection, or a condition associated with chronic diarrhea.
- the disease, disorder, or condition comprises one or more of irritable bowel syndrome (IBS), inflammatory bowel disease (IBD), short bowel syndrome (SBS), celiac disease, small intestinal bacterial overgrowth (SIBO), gastroenteritis, leaky gut syndrome, pouchitis, or gastric lymphoma.
- the disease, disorder, or condition comprises an atopic disease.
- the atopic disease comprises atopic dermatitis, food allergy, and/or asthma.
- the disease, condition, or disorder is associated with an infection.
- the infection comprises a bacterial infection or gut domination.
- the bacterial infection or gut domination comprises an infection or gut domination by one or more species, subspecies, or strains of Aeromonas, Bacillus, Blautia, Bordetella, Borrelia, Brucella, Burkholderia, Campylobacter, Chlamydia, Chlamydophila, Citrobacter, Clostridium, Corynebacterium, Coxiella, Ehrlichia, Enterobacter, Enterobacteriaceae, Enterococcus, Escherichia, Faecalicatena, Francisella, Haemophilus, Helicobacter, Hungatella, Klebsiella, Lachnospiraceae, Legionella, Leptospira, Listeria, Morganella, Mycobacterium, Mycoplasma, Neisseria, Orientia, Plesi
- coli enterotoxigenic Escherichia coli, Escherichia coli 0157:H7, Helicobacter pylori, Klebsiellia pneumonia, Lysteria monocytogenes, Salmonella paratyphi, Salmonella typhi, Staphylococcus aureus, Vibrio cholerae, Vibrio parahaemolyticus, Vibrio vulnificus, or Yersinia enter ocolitica.
- the disease, condition, or disorder is associated with an infection.
- the infection comprises a bacterial infection or gut domination.
- the bacterial infection or gut domination comprises an infection or gut domination by one or more species, subspecies, or strains of Aeromonas, Bacillus, Bordetella, Brucella, Burkholderia, Campylobacter, Chlamydia, Chlamydophila, Citrobacter, Clostridium, Corynebacterium, Coxiella, Ehrlichia, Enterobacter, Enterobacteriaceae, Enterococcus, Escherichia, Francisella, Haemophilus, Klebsiella, Legionella, Leptospira, Listeria, Morganella, Mycobacterium, Mycoplasma, Neisseria, Orientia, Plesiomonas, Proteus, Pseudomonas, Rickettsia, Salmonella, Shigella
- coli enterotoxigenic Escherichia coli, Escherichia coli 0157:H7, Helicobacter pylori, Klebsiella pneumoniae, Listeria monocytogenes, Salmonella paratyphi, Salmonella typhi, Staphylococcus aureus, Vibrio cholerae, Vibrio parahaemolyticus, Vibrio vulnificus, or Yersinia enterocolitica.
- the bacterial infection or gut domination comprises an infection or gut domination by one or more of Citrobacter freundii, Citrobacter koseri, Enterobacter aerogenes, Enterobacter cloacae, Enterococcus faecalis, Enterococcus faecium, Escherichia coli, Klebsiella oxytoca, Klebsiella pneumoniae, Morganella morganii, Proteus mirabilis, Serratia marcescens, Staphylococcus aureus, Staphylococcus epidermidis, Streptococcus anginosus, Streptococcus australis, Streptococcus constellatus, Streptococcus cristatus, Streptococcus gordonii, Streptococcus infantis, Streptococcus intermedins, Streptococcus mitis, Streptococcus mutans, Streptobacter freundii
- the bacterial infection or gut domination comprises an infection or gut domination by drug -resistant bacteria.
- the drug-resistant bacteria comprise one or more of antibiotic-resistant bacterium (ARB), Antibiotic-resistant Proteobacteria, Carbapenem-resistant Ewterotocterzaceae (CRE), Extended Spectrum Beta-Lactamase producing Enterobacteriaceae (ESBL-E), fluoroquinolone-resistant Enter obacteriaceae, extended spectrum beta-lactam resistant Enterococci (ESBL), vancomycin-resistant Enterococci (VRE), multi-drug resistant A. coli, or multi-drug resistant Klebsiella.
- ARB antibiotic-resistant bacterium
- CRE Carbapenem-resistant Ewterotocterzaceae
- ESBL-E Extended Spectrum Beta-Lactamase producing Enterobacteriaceae
- ESBL extended spectrum beta-lactam resistant Enterococci
- VRE vancomycin-resistant Enterococci
- the bacterial infection or gut domination comprises an infection or gut domination by drug-resistant bacteria.
- the drugresistant bacteria comprises one or more of antibiotic-resistant bacterium (ARB), Antibioticresistant Proteobacteria, Carbapenem-resistant Enterobacteriaceae (CRE), Extended Spectrum Beta-Lactamase producing Enterobacterales (ESBL-E), fluoroquinolone-resistant Enterobacteriaceae, vancomycin-resistant Enterococci (VRE), multi-drug resistant A. coli, or multi-drug resistant Klebsiella.
- the subject has undergone or will undergo an ileal pouch-anal anastomosis (IPAA) surgery, and wherein the disease, condition, or disorder comprises pouchitis.
- AIPAA ileal pouch-anal anastomosis
- a method for increasing propionate in the gut of a subject in need thereof comprising administering to the subject i) a prebiotic mixture comprising one or more human milk oligosaccharides, ii) at least one Bifidobacterium capable of consuming the one or more human milk oligosaccharides; and iii) at least one propionate producing bacterium.
- the at least one propionate producing bacterium comprises a strain that is isolated from a human intestinal microbiome and/or is capable of engrafting within the human intestinal microbiome.
- the at least one propionate producing bacterium comprises one or more strains of the genus Veillonella.
- the at least one propionate producing bacterium comprises one or more of Veillonella atypica (V. atypica), Veillonella dispar (V. dispar), Veillonella infantium (V. infantium), Veillonella nakazawae (V. nakazawae), Veillonella parvula (V. parvula), Veillonella ratti (V. ratti), Veillonella rogosae (V. rogosae), Veillonella seminalis (V. seminalis), and/or Veillonella tobetsuensis (V.
- the at least one propionate producing bacterium comprises V. atypica, V. dispar, V. infantium, V. nakazawae, V. parvula, and/or V. rogosae.
- the at least one propionate producing bacterium comprises V. infantium, V. nakazawae, V. parvula, and/or V. rogosae.
- the at least one propionate producing bacterium comprises V. infantium, V. dispar, and/or V. nakazawae .
- the at least one propionate producing bacterium comprises V. parvula.
- the at least one propionate producing bacterium comprises V. rogosae.
- the at least one propionate producing bacterium comprises one or more strains of the genus Megasphaera.
- the at least one propionate producing bacterium comprises one or more of Megasphaera elsdenii (M. elsdenii), Megasphaera hominis (M. hominis), Megasphaera indica (M. indica), Megasphaera massiliensis (M. massiliensis), and/ or Megasphaera micronuciformis (M. micronuciformis).
- the at least one propionate producing bacterium comprises one or more of M. elsdenii ndJo M. massiliensis.
- the at least one propionate producing bacterium comprises one or more strains of the genus Anaerotignum. In certain embodiments, the at least one propionate producing bacterium comprises Anaerotignum lactatifermentans (A. lactatifermentans). In particular embodiments, the at least one propionate producing bacterium comprises one or more strains of the genus Bacteroides. In some embodiments, the at least one propionate producing bacterium comprises Bacteroides fragilis (B. fragilis o Bacteroides caccae (B. caccae). In certain embodiments, the at least one propionate producing bacterium comprises one or more strains of the genus Coprococcus.
- the at least one propionate producing bacterium comprises Coprococcus catus (C. catus). In some embodiments, the at least one propionate producing bacterium comprises one or more strains of the genus Merdimmobilis . In certain embodiments, the at least one propionate producing bacterium comprises Merdimmobilis hominis.
- the propionate producing bacterium comprises a nucleotide sequence with at least 97%, 98%, or 99% sequence identity to any of SEQ ID NOS: 40-49 or 52-58 and/or an amino acid sequence with at least 97%, 98%, or 99% sequence identity to SEQ ID NO: 50 or 51.
- the Bifidobacterium comprises B. breve, B. bifidum, or B. longum subsp. infantis. In certain embodiments, the Bifidobacterium comprises B. longum subsp. infantis.
- the method further comprising at least one butyrate producing strain of bacteria.
- the at least one butyrate producing strain comprises a strain of Clostridium Cluster IV or Clostridium Cluster XlVa bacteria.
- the at least one butyrate producing strain comprises one or more of Agathobacter rectalis, Anaerobutyricum hallii, Anaerostipes caccae, Blautia producta, Clostridium leptum, Faecalibacterium prausnitzii, Anaerobutyricum soehngenii, Roseburia hominis, or Roseburia intestinalis.
- the at least one butyrate producing strain comprises one or more of Anaerostipes caccae, Clostridium innocuum, Roseburia hominis, or Roseburia intestinalis.
- the prebiotic mixture comprises one or more of 2'- fucosyllactose, 3-fucosyllactose, difucosyllactose, 3’-sialyllactose, 6'-sialyllactose, lacto-N- tetraose, lacto-N-neo-tetraose, lacto-N-fucopentaose I, lacto-N-fucopentaose II, lacto-N- fucopentaose III, sialyl-lacto-N-tetraose a, sialyl-lacto-N-tetraose b, sialyl-lacto-N-tetraose c, lacto-N-difuco-hexaose I, lacto-N-difuco-hexaose II, lacto-N-hexaose, para
- the prebiotic mixture comprises one or more of 2'- fucosyllactose, 3-fucosyllactose, 3'-sialyllactose, 6'-sialyllactose, lacto-N-tetraose, lacto-N- difucohexaose I, lactodifucotetraose, lacto-N-fucopentaose I, sialylacto-N-tetraose c, sialylacto-N-tetraose b, or disialyllacto-N-tetraose.
- the prebiotic mixture comprises one or more of 2'-fucosyl-lactose, 3-fucosyllactose, 3’-sialyllactose, 6'- sialyllactose, lacto-N-tetraose, lacto-N-neotetraose, or difucosyllactose.
- the prebiotic mixture comprises one or more of 2'-fucosyllactose, 3- fucosyllactose, lacto-N-tetraose, or lacto-N-neotetraose.
- the prebiotic mixture comprises one or both of 2'-fucosyllactose and lacto-N-neotetraose. In particular embodiments, the prebiotic mixture comprises at least 10, at least 25, at least 50, at least 100, or at least 150 human milk oligosaccharides.
- the prebiotic mixture comprises 2'-fucosyllactose, 3-fucosyllactose, 3’-sialyllactose, 6'-sialyllactose, lacto-N- tetraose, lacto-N-difucohexaose I, lactodifucotetraose, lacto-N-fucopentaose I, sialylacto-N- tetraose c, sialylacto-N-tetraose b, and disialyllacto-N-tetraose.
- the prebiotic mixture is, is derived from, or comprises a concentrated human milk permeate, wherein the concentrated human milk permeate is obtained by a process comprising the steps of ultra-filtering human skim milk to obtain human milk permeate and concentrating the human milk oligosaccharide content of the human milk permeate.
- the human skim milk is obtained from human milk pooled from at least 25, 50, or 100 individual donors.
- kits comprising i) a prebiotic mixture comprising one or more human milk oligosaccharides, ii) at least one Bifidobacterium capable of consuming the one or more human milk oligosaccharides; and iii) at least one propionate producing bacterium.
- the at least one propionate producing bacterium comprises a strain that is isolated from a human intestinal microbiome and/or is capable of engrafting within the human intestinal microbiome.
- the kit further comprises at least one butyrate producing strain of bacteria.
- a method for increasing propionate concentration and/or propionate production in the gut of a subject in need thereof comprising administering to the subject the prebiotic mixture, the at least one Bifidobacterium, and the at least one propionate producing bacterium any of the kits described herein.
- Also provided herein is a method of treating or preventing a disease, disorder, or condition associated with one or more of inflammation, immune dysfunction, cancer, allergy, or dysbiosis of the intestinal microbiome in a subject in need thereof, the method comprising administering to the subject the prebiotic mixture, the at least one Bifidobacterium, and the at least one propionate producing bacterium of any of the kits described herein.
- a method of ameliorating a symptom of a disease, disorder, or condition associated with one or more of inflammation, immune dysfunction, cancer, allergy, or dysbiosis of the intestinal microbiome in a subject in need thereof comprising administering to the subject the prebiotic mixture, the at least one Bifidobacterium, and the at least one propionate producing bacterium of any of the kits described herein.
- a pharmaceutical composition comprising i) a prebiotic mixture comprising one or more human milk oligosaccharides, ii) at least one Bifidobacterium capable of consuming the one or more human milk oligosaccharides; and iii) at least one propionate producing bacterium.
- the at least one Bifidobacterium comprises B. longum subsp. inf antis and the at least one propionate producing bacterium comprises one or more strains of the genus Veillonella.
- the pharmaceutical composition further comprises at least one butyrate producing strain of bacteria, optionally wherein the at least one butyrate producing strain comprises one or more of Agathobacter rectalis, Anaerobutyricum hallii, Anaerostipes caccae, Blautia producta, Clostridium leptum, Faecalibacterium prausnitzii, Anaerobutyricum soehngenii, Roseburia hominis, or Roseburia intestinalis.
- the at least one butyrate producing strain comprises one or more of Agathobacter rectalis, Anaerobutyricum hallii, Anaerostipes caccae, Blautia producta, Clostridium leptum, Faecalibacterium prausnitzii, Anaerobutyricum soehngenii, Roseburia hominis, or Roseburia intestinalis.
- the at least one Bifidobacterium comprises B. longum subsp. infantis.
- the method further comprises administering to the subject in need thereof iii) at least one propionate producing bacterium.
- the at least one propionate producing bacterium comprises one or more strains of the genus Veillonella.
- the one or more symptoms of acute radiation syndrome comprise gastrointestinal damage.
- the subject was exposed to an ionizing radiation dose of at least 0.3 Gray (Gy), optionally at least 0.7 Gy, 1 Gy, 2 Gy, 5 Gy, 6 Gy, or 10 Gy over a period of time lasting under 60 minutes, optionally under 30 minutes, 15 minutes, 10 minutes, 5 minutes, 3 minutes, 2 minutes, or 1 minute.
- kits described herein comprising any of the kits described herein and instructions for use describing any of the methods described herein.
- FIG. 1 provides a schematic of the study described in Example 2.
- FIGS. 2A-2D show graphs displaying time courses of B. longum subsp. infantis abundance during a clinical study.
- FIG. 2A shows B. infantis abundance over time as determined by qPCR and plotted as the average for subjects in each of the three cohorts. Circles indicate Cohort 1, triangles indicate Cohort 2, and squares indicate Cohort 3. Data are geometric means with 95% confidence intervals. The line labeled LOD represents the limit of detection (27 copies/ng DNA). Significant differences were calculated using mixed effects analysis for repeated measures of log-transformed data; significance between kinetic curves were determined using the time versus treatment factor and significances at individual timepoints were determined using Sidak’s post-test.
- FIG. 2B shows B.
- FIG. 2C shows B. infantis abundance over time as determined by qPCR for subjects in the cohort that received B. infantis and HMO (Cohort 3) comparing those who were engrafted and not engrafted.
- Subjects were deemed “engrafted” (dark lines) if B. infantis signal was two geometric standard deviations above the geometric mean of signal on days 3-5 (equivalent to 5.4 x 10 3 copies/ng DNA) for at least two consecutive time points; subjects below the geometric mean of the B. infantis only cohort on any one of those days were deemed “not engrafted” (light lines). Traces represent individual subjects.
- FIG. 2C shows B. infantis abundance over time as determined by qPCR for subjects in the cohort that received B.
- FIG. 2D shows the abundance of B. infantis within the microbiome as determined by whole metagenome sequencing (WMS) using a strain tracking algorithm. Circles indicate Cohort 1, triangles indicate Cohort 2, and squares indicate Cohort 3. Each point represents the median of the indicated cohort subpopulation, and error bars represent the interquartile range. Undetected values were imputed with the LOD. The LOD represents the lowest detected value.
- FIGS. 3A-3F show graphs summarizing whole metagenomic sequencing (WMS) from genomic DNA extracted from stool samples collected from subjects as described in Example 2.
- FIG. 3A displays Shannon Entropy of observed reads rarefied to 76,000 reads used as a measure of the alpha diversity of samples over time. Data are median and 95% confidence interval. Significant differences were calculated using mixed effects analysis for repeated measures; significance between kinetic curves was determined using the time versus treatment factor. Significances between individual timepoints were determined using Sidak’s post-test for each individual cohort and asterisks indicate significance across both cohorts.
- FIG. 3A displays Shannon Entropy of observed reads rarefied to 76,000 reads used as a measure of the alpha diversity of samples over time. Data are median and 95% confidence interval. Significant differences were calculated using mixed effects analysis for repeated measures; significance between kinetic curves was determined using the time versus treatment factor. Significances between individual timepoints were determined using Sidak’s post-test for each individual cohort and asterisks indicate
- FIG. 3B shows Bray-Curtis dissimilarity for each sample with rarefied sequences aggregated to the genus taxonomic level and displayed by cohort and day in a principal coordinate analysis (PcoA) plot noting the trajectory from pre-treatment (day 1) and end (day 5) of antibiotic treatment (arrow).
- FIG. 3C contains stacked bar charts displaying aggregated families (and two at the order level) present at >1% abundance for each cohort on each day of the study. Taxa ⁇ 1% in each cohort were aggregated to “Other”. For the antibiotics + B. infantis + HMO cohort (Cohort 3), subjects are separated into Engrafted and Not Engrafted (NE).
- 3D shows PcoA plots of Bray-Curtis dissimilarity calculated with rarefied sequences aggregated to the genus taxonomic level for days 1, 5 and 14 only.
- the antibiotics only cohort was compared to engrafted subjects in the antibiotics + B. infantis + HMO cohort, including (top row) and excluding (bottom row) taxa assigned to the genus Bifidobacterium.
- FIG. 3E shows a volcano plot of aggregated genera >1% across cohorts comparing abundance in Cohort 3 with Cohort 1.
- FIG. 3F shows the relative abundance of Veillonella across study days comparing Cohort 1 (left) and Cohort 3 (right) showing statistical differences calculated using the CLR transformed abundances in ALDEx2.
- FIGS. 4A-4F show graphs summarizing gut metabolites in stool samples of antibiotic-treated subjects.
- FIGS. 4A and 4C show levels of acetic acid (FIG. 4A) and lactic acid (FIG. 4C) as measured in stool samples from subjects in the antibiotics only cohort (Cohort 1, circles) and engrafted subjects in the antibiotics + B. infantis + HMO cohort (Cohort 3, squares). Dotted lines represent limit of detection. Significant differences were calculated using mixed effects analysis for repeated measures of log-transformed data; significance between kinetic curves was determined using the time versus treatment factor. In FIG. 4A, significance between day 1 and day 5 was determined using Sidak’s post-test for each individual cohort and was significant across both cohorts. In FIG.
- FIG. 4C significances between cohorts at individual timepoints were determined using Sidak’s post-test.
- FIG. 4B shows Kaplan-Meier curve plotting the day on which stool acetate for each subject returned to baseline levels, where baseline is the average of subjects at Day 1. Significance between groups was calculated using log-rank test.
- FIG. 4E shows levels of indolelactate in stool at day 14 (left) and in serum at days 1, 5, 14, 28, and 35 (right).
- FIG. 4F shows levels of p-cresol sulfate in stool at day 14 (left) and in serum at days 1, 5, 14, 28, and 35 (right).
- horizontal lines are geometric mean; dotted lines are the imputed value for samples without detected signal, and significances are derived from two-way repeated measures ANOVA calculated for the entire global metabolomics dataset as described in FIG. 4D.
- Timecourse data in FIGS. 4E and 4F are geometric mean and 95% confidence interval; dotted lines are limit of detection; significances are calculated as in FIG. 4B.
- FIGS. 5A-5D show graphs summarizing in vitro cross-feeding of three different Veillonella species by B. infantis + HMO.
- the indicated propionate-producing species of Veillonella were grown with no addition, B. infantis, HMO, both B. infantis and HMO, or lactate alone as indicated.
- Dotted lines labeled “LOQ” indicate the limits of quantitation.
- FIG. 5A shows propionate quantified using liquid chromatography with tandem mass spectrometry from media collected after 30 hours of culture with media alone or the noted concentration of lactate.
- FIG 5B shows propionate quantified from media collected after 30 hours using liquid chromatography with tandem mass spectrometry.
- FIGS. 5C and 5D show abundance of B. infantis (FIG.
- FIGS. 6A-6D depict in vivo cross-feeding of Veillonella species by B. infantis + HMO.
- FIG. 6A provides a study schematic. Germ-free mice were divided into three groups, two of which were inoculated with Veillonella parvula and the third with B. infantis as a control. After one week, the two V. parvula-associated groups were given a single gavage of either PBS or B. infantis + HMO; B. infantis-associated animals were gavaged with HMO only. Over the subsequent three days, animals received once-daily gavages of PBS or HMO.
- FIG. 6B shows quantification of Veillonella in feces.
- FIG. 6C shows the levels of propionate quantified in contents of the indicated intestinal segments using LC-MS/MS.
- FIG. 6D shows the levels of lactate quantified in contents of the indicated intestinal segments using LC-MS/MS. Significance of differences was calculated by Sidak’s multiple comparison test from mixed effects analysis for repeated measures of log- transformed data.
- FIGS. 7A and 7B show relative abundance of B. infantis as determined by qPCR.
- FIG. 7A shows data from FIG. 2 A replotted to exclude the non-engrafted subjects in the B. infantis + HMO cohort; significances were calculated as in FIG. 2A.
- FIG. 7B shows B. infantis relative abundance in stool samples determined using qPCR to measure B. infantis (shown in FIG. 2 A) and 16S rRNA and then normalizing B. infantis levels to 16S rRNA levels. Each point represents the median, and error bars represent the interquartile regions.
- the dotted line represents limit of detection (LOD) calculated across the entire study using the following calculation: [LOD for B. infantis copy number per reaction] / [median 16S rRNA gene copy number per reaction across all samples],
- FIGS. 8A-8H depict microbiome changes after antibiotic perturbation.
- FIG. 8A shows number of observed reads whole metagenomic sequencing (WMS) data from stool samples over time; significances were calculated as in FIG. 3 A.
- FIG. 8B shows the geometric mean of 16S rRNA gene copy number per gram of stool for each cohort over time. Each dot represents the geometric mean and error bars represent the 95% confidence intervals. Significances were calculated as in FIG. 3 A except that data were log-transformed prior to statistical analysis.
- FIG. 8C provides stacked bar charts of metagenomic sequencing at the family level for subjects receiving B. infantis only (Cohort 2) over time. Each study day is separated into engrafted (E) or not engrafted.
- 8D-I are box and whisker plots of the relative abundance of bacteria highlighted in the ALDEx2 analysis shown in FIG. 3E and Bacteroides over time comparing subjects in the antibiotics only cohort (Cohort 1, left) and the B. Infantis + HMO (engrafted only) cohort (Cohort 3, right).
- FIGS. 9A-9N depict changes in gut metabolites after antibiotic perturbation.
- FIGS. 9A and 9B show levels of butyrate (FIG. 9A) and propionate (FIG. 9B) measured in stool samples collected on days 1, 5, 9, 11, 14, 17, 28, and 35 from subjects that received antibiotics only (Cohort 1, circles) and engrafted subjects who also received B. infantis and HMO (Cohort 3, squares). Dots represent the geometric mean and error bars represent the 95% confidence intervals. Significances were calculated as in FIG 4A.
- FIG. 9C shows pH measurements of fecal samples within cohorts over time. Data show average pH with error bars representing standard error. A mixed effects model with Sidak’s multiple comparisons test was used for comparisons.
- FIGS. 9A-9N depict changes in gut metabolites after antibiotic perturbation.
- FIGS. 9A and 9B show levels of butyrate (FIG. 9A) and propionate (FIG. 9B) measured in stool samples collected on days 1, 5, 9, 11, 14,
- FIG. 9D-9I show volcano plots of fecal global metabolomic data comparing cohorts (FIG. 9F and FIG. 91) or study days within each cohort (FIGS. 9D, 9E, 9G, and 9H).
- FIG. 9J shows Bray-Curtis dissimilarity calculated for each sample and displayed by cohort and day in a PcoA plot.
- FIG. 9K shows HMOs from the stool metabolomics analysis on day 5 represented in batch-normalized imputed peak area values. Each dot represents a subject in the Antibiotics only cohort (Cohort 1, left) or the engrafted subjects in the B. infantis + HMO cohort (Cohort 3, right), with a line at the median.
- FIG. 9L is a volcano plot showing fold change in serum metabolites in the Antibiotics Only cohort (Cohort 1) between Day 5 and Day 1 against significance of the observed change. Significances were calculated with a repeated measures test on log-transformed data and Sidak’s post-test to correct for multiple comparisons. Relevant metabolites are noted in color and labeled.
- FIG. 9M is a volcano plot showing fold change in serum metabolites on Day 14 between engrafted subjects (Cohort 3) and subjects that only received antibiotics (Cohort 1) plotted against significance of the observed change, calculated as in FIG. 9L.
- FIG. 9N shows levels of phenol sulfate measured in serum samples over time for each cohort. Data and significances are presented as in FIG. 9A.
- FIGS. 10A-10G show in vitro cross-feeding of Veillonella species by B. infantis + HMO. The indicated species of Veillonella were grown in the absence or presence of B. infantis, HMO, both, or lactate.
- FIGS. 10A-10C are growth curves of the three different Veillonella strains presented in FIG. 5 A with varying concentrations of lactate.
- FIGS. 10D-10G show quantification of acetate (FIGS. 10D and 10F) and lactate (FIGS. 10E and 10G) in cultures presented in FIG. 5. Data represent geometric mean and standard deviation of 3 independent experiments. Within each experiment, triplicate culture wells were pooled for analysis. Dotted lines indicate the limit of quantitation. Statistics reflect a two-way ANOVA with Tukey’s test for multiple comparisons to assess acetate production (FIGS. 10D and 10F) or lactate consumption (FIGS. 10E and 10G) under each set of growth conditions, in all cases using log-transformed data (* indicates adjusted p ⁇ 0.05, “ns” indicates adjusted p >0.05). Lines in FIG. 10E and 10G indicate the displayed comparisons between levels of lactate in cultures containing Veillonella and the corresponding Veillonella-V culture.
- FIGS. 11A-11C depict in vivo cross-feeding of Veillonella species with B. infantis + HMO in mice described in Example 12.
- FIGS. HA and 11B show quantification of Veillonella (FIG. 11 A) and B. infantis (FIG. 11B) in mice using qPCR. Data are plotted as the geometric mean and standard deviation.
- FIG. 11C shows concentration of acetate isolated intestinal sections and feces from mice of the experiments shown in FIG. 6.
- FIG. 12 shows a scatterplot depicting concentrations of propionate (Y axis) and lactate (x axis) of various test strains following a 48 hour incubation with lactate as a carbon source. Starting lactate concentration is indicated by the vertical dotted line.
- FIGS. 13A-13C show quantification of Veillonella sp. (left) and B. longum subsp. infantis (B. infantis,' right) in stool collected at various timepoints from germ free mice inoculated with Veillonella sp., B. infantis, or both and administered a human milk oligosaccharide composition (HMO) or a vehicle control.
- Veillonella sp. included a strain of V. parvula (FIG. 13A) and two Veillonella strains (FIGS. 13B and 13C) isolated as described in Example 11. Strains were quantified using qPCR. Data are plotted as the geometric mean and standard deviation.
- FIGS. 14A-14C show quantification of propionate levels detected in ileal, cecal, rectal, and fecal samples collected from germ free mice inoculated with Veillonella sp. (V sp.), B. infantis (BI), or both and administered a human milk oligosaccharide composition (HMO) or a vehicle control.
- V sp. Veillonella sp. included a strain of V. parvula (FIG. 14A) and two Veillonella strains (FIGS. 14B and 14C) isolated as described in Example 11. Data are plotted as the geometric mean and standard deviation. Data are plotted as the geometric mean and standard deviation.
- FIGS. 15A-15C show quantification of lactate levels detected in ileal, cecal, rectal, and fecal samples collected from germ free mice inoculated with Veillonella sp. (V sp.), B. infantis (BI), or both and administered a human milk oligosaccharide composition (HMO) or a vehicle control.
- V sp. Veillonella sp. included a strain of V. parvula (FIG. 15A) and two Veillonella strains (FIGS. 15B and 15C) isolated as described in Example 11. Data are plotted as the geometric mean and standard deviation. Data are plotted as the geometric mean and standard deviation.
- FIGS. 16A-16C show quantification of acetate levels detected in ileal, cecal, rectal, and fecal samples collected from germ free mice inoculated with Veillonella sp. (V sp.), B. infantis (BI), or both and administered a human milk oligosaccharide composition (HMO) or a vehicle control.
- V sp. Veillonella sp. included a strain of V. parvula (FIG. 16A) and two Veillonella strains (FIGS. 16B and 16C) isolated as described in Example 11. Data are plotted as the geometric mean and standard deviation. Data are plotted as the geometric mean and standard deviation.
- FIGS. 17A-17F depict experimental results following inoculation of strains in germ free mice.
- FIG. 17A show quantification of B. longum subsp. infantis (B. infantis; left) and Megasphaera elsdenii (M. elsdenii; right) in stool collected at various timepoints from germ free mice inoculated with A/, elsdenii, B. infantis, or both and administered a human milk oligosaccharide composition (HMO) or a vehicle control.
- HMO human milk oligosaccharide composition
- FIGS. 17B-17F show quantification of butyrate (FIG. 17B), propionate (FIG. 17C), valerate (FIG. 17D), lactate (FIG. 17E), and acetate (FIG. 17F) levels detected in cecal samples collected from these mice. Data are plotted as the geometric mean and standard deviation.
- compositions, kits, and articles of manufacture as well as methods of use thereof.
- the provided compositions, kits, and articles of manufacture are or include prebiotics and at least two probiotic strains of bacteria.
- the prebiotics are or include a mixture, e.g., of oligosaccharides such as human milk oligosaccharides.
- the at least two probiotic strains of bacteria are or include one or more strains of Bifidobacteria, for example a strain of Bifibacterium capable of consuming the prebiotics and/or human milk oligosaccharides, and a strain of bacteria capable of producing the short chain fatty acid propionate, e.g., a strain of Veillonella sp.
- the provided compositions, kits, and articles of manufacture may be administered to a subject to treat or prevent dysbiosis, e.g., of the intestinal microbiome, as well as conditions, diseases, or disorders that may originate from or cause dysbiosis.
- the provided compositions, kits, and articles of manufacture may be administered to a subject to treat or prevent conditions, diseases, or disorders that are, include, or contribute to inflammation, infection, allergy, or immune dysfunction.
- the maintenance of a healthy human metabolism depends on a symbiotic consortium among bacteria, archaea, viruses, fungi, and host eukaryotic cells throughout the human gastrointestinal tract.
- microbial communities may provide enzymatic machinery and metabolic pathways that contribute to food digestion, xenobiotic metabolism, and production of a variety of bioactive molecules. Disturbances to the microbiome may result in a microbial imbalance (dysbiosis) characterized by phylumlevel changes in the microbiota composition, including a marked decrease in the representation of obligate anaerobic bacteria and an increased relative abundance of facultative anaerobic bacteria. While dysbiosis is associated with numerous diseases and conditions, successfully treating dysbiosis is difficult, particularly in vulnerable or immunocompromised patients.
- compositions, methods, kits, and articles of manufacture address these needs.
- the present invention includes specific combinations of prebiotics, such as human milk oligosaccharides, and probiotics, such as Bifidobacterium including B. longum subspecies (subsp.) infantis and bacteria capable of producing propionate, e.g., Veillonella sp., that are particularly safe and effective for treating, ameliorating, or reducing dysbiosis in the gut microbiome as well as effective in treating, ameliorating, or preventing diseases or disorders that may be accompanied by dysbiosis, such including but not limited to diseases associated with immune disorders, inflammatory disorders, or infection.
- prebiotics such as human milk oligosaccharides
- probiotics such as Bifidobacterium including B. longum subspecies (subsp.) infantis and bacteria capable of producing propionate, e.g., Veillonella sp.
- compositions, kits, and articles of manufacture that are or include i) a prebiotic mixture that are or include one or more oligosaccharides, ii) at least one strain of bacteria from the genus Bifidobacterium that is capable of consuming oligosaccharides, e.g., the oligosaccharides of the mixture and/or human milk oligosaccharides; and iii) one or more strains of bacteria capable of producing propionate.
- the prebiotic mixture is or includes human milk oligosaccharides.
- the Bifidobacteria is or includes B. longum subsp. infantis.
- the one or more strains of bacteria capable of producing propionate are or include strains of bacteria from the genus Veillonella.
- the Bifidobacteria capable of consuming human milk oligosaccharides, the strain capable of producing propionate, and the prebiotic mixture are or are included in separate compositions, e.g., are administered separately to a subject.
- kits and articles of manufacture that include (i) a composition that is or includes a strain from the genus Bifidobacteria capable of consuming human milk oligosaccharides (ii) a composition that is or includes at least one propionate producing strain of bacterium (also referred to herein and used interchangeably with “strain of bacteria capable of producing propionate” or “propionate producing bacterium” unless otherwise indicated), and (iii) a composition that is or includes the prebiotic mixture, e.g., of human milk oligosaccharides.
- kits and articles of manufacture that are or include two compositions, a probiotic composition that is or includes the Bifidobacteria, e.g., B. longum subsp. infantis, and bacteria capable of producing propionate, e.g., Veillonella, and a composition that is or includes the prebiotic mixture, e.g., of human milk oligosaccharides.
- kits and articles of manufacture that include a composition that is or includes all of the Bifidobacteria, e.g., B. longum subsp. infantis, the strain of bacteria capable of producing propionate, e.g., Veillonella, and the prebiotic mixture, e.g., of human milk oligosaccharides.
- the provided compositions, kits, and articles of manufacture are or include any of the strains of bacteria capable of producing propionate, (also referred to herein as a “propionate producing strain of bacteria,” “propionate producing strain” or “propionate producing bacterium”) described throughout the application, including in Section I-A.
- the provided compositions, kits, and articles of manufacture are or include any of the one or more strains of the genus Bifidobacterium, e.g., a strain of Bifidobacterium capable of consuming human milk oligosaccharides, described throughout the application including in Section I-B.
- the provided compositions, kits, and articles of manufacture are or include any of the prebiotic mixtures, e.g., of human milk oligosaccharides, described throughout the application, including at least at Section I-C.
- the provided kits and articles of manufacture may also include labels or instructions for use. In some embodiments, such labels or instructions for use may describe any of the uses or methods provided herein, such as those described in Section II.
- the at least one propionate producing bacterium is capable of consuming lactate and/or acetate. In certain embodiments, the at least one propionate producing bacterium is capable of producing, generating, and/or creating propionate in the presence of lactate and/or acetate. In certain embodiments, the at least one propionate producing bacterium is capable of growing or expanding in the presence of a strain of Bifidobacterium, e.g., a strain of Bifidobacterium probiotic strain described herein such as in Section I-B. In some embodiments, the at least one propionate producing strain of bacterium is capable of growing or expanding in the presence of B. longum subsp. infantis.
- the at least one propionate producing bacterium is or includes at least one, two, three, four, five, six, seven, eight, nine, ten, or more species, subspecies, or strains of bacteria capable of producing propionate.
- the propionate producing bacterium is or includes at least three strains of bacteria capable of producing propionate.
- the propionate producing strain is or includes between one and five species, subspecies, or strains of bacteria capable of producing propionate.
- the at least one propionate producing bacterium has or contains one or more functional genes that contribute to the production, generation, synthesis, or making of propionate.
- the at least one propionate producing bacterium is or includes a strain of bacterium from the genus Veillonella.
- the at least one propionate producing bacterium is or includes Veillonella sp.
- the at least one strain of bacterium from the genus Veillonella is capable of converting lactate to propionate.
- the one or more propionate producing strain of bacteria is or includes Veillonella agrestimuris, Veillonella alcalescens, Veillonella atypica, Veillonella caviae, Veillonella cricetid, Veillonella denticariosi, Veillonella dispar, Veillonella hominis, Veillonella infantium, Veillonella intestinalis, Veillonella magna, Veillonella montpellierensis, Veillonella nakazawae, Veillonella parvula, Veillonella ratti, Veillonella rodentium, Veillonella rogosae, Veillonella seminalis, or Veillonella tobetsuensis.
- the propionate producing bacterium is or includes Veillonella atypica (V. atypica), Veillonella dispar (V. dispar), Veillonella infantium (V. infantium), Veillonella nakazawae (V. nakazawae), or Veillonella parvula (V. parvula).
- the propionate producing bacterium is or includes V. infantium, V. dispar, or V. nakazawae.
- the propionate producing bacterium is or includes V. parvula.
- the propionate producing bacterium is or includes V. rogosae.
- the propionate producing bacterium is or includes a strain of Veillonella sp. that has or includes a nucleic acid of at least 200, 300, 400, 500, 600, 700, 800, 900, 1,000, 1,200, or 1,400 nucleotides in length with at least 85%, 90%, 95%, 97%, 97.5%, 98%, 99% or 100% sequence identity to any of SEQ ID NOS: 40-49.
- the propionate producing bacterium is or includes a strain of Veillonella sp.
- the propionate producing bacterium is or includes a strain of Veillonella sp. that has or includes a nucleic acid sequence with at least 85%, 90%, 95%, 97%, 97.5%, 98%, 99% or 100% sequence identity to any of SEQ ID NOS: 40-49.
- the propionate producing bacterium is or includes a strain of Veillonella sp. that has or includes a nucleic acid sequence with at least 85%, 90%, 95%, 97%, 97.5%, 98%, 99% or 100% sequence identity to any of SEQ ID NOS: 40-49.
- the propionate producing bacterium is or includes a strain of Veillonella sp. that has or includes a nucleic acid sequence with at least 85%, 90%, 95%, 97%, 97.5%, 98%, 99% or 100% sequence identity to any of SEQ ID NOS: 40-43.
- the propionate producing bacterium is or includes a strain of Veillonella sp. that has or includes a nucleic acid sequence encoding an rpoB gene having a sequence of at least 200, 300, 400, 500, 600, 700, 800, 900, 1,000, 1,200, or 1,400 nucleotides in length with at least 95%, 97%, 97.5%, 98%, 99% or 100% sequence identity to SEQ ID NO: 40 or 41.
- the propionate producing bacterium is or includes a strain of Veillonella sp. that has or includes a nucleic acid sequence encoding an rpoB gene with at least 95%, 97%, 97.5%, 98%, 99% or 100% sequence identity to SEQ ID NO: 40 or 41.
- the propionate producing bacterium is or includes a strain of Veillonella sp. that has or includes a nucleic acid sequence encoding an 16S gene having a sequence of at least 200, 300, 400, 500, 600, 700, 800, 900, 1,000, 1,200, or 1,400 nucleotides in length with at least 95%, 97%, 97.5%, 98%, 99% or 100% sequence identity to any of SEQ ID NOS: 42-49.
- the propionate producing bacterium is or includes a strain of Veillonella sp.
- the propionate producing bacterium is or includes a strain of Veillonella sp. that has or includes a nucleic acid sequence encoding an 16S gene having a sequence of at least 200, 300, 400, 500, 600, 700, 800, 900, 1,000, 1,200, or 1,400 nucleotides in length with at least 95%, 97%, 97.5%, 98%, 99% or 100% sequence identity to SEQ ID NO: 42 or 43.
- the propionate producing bacterium is or includes a strain of Veillonella sp. that has or includes a nucleic acid sequence encoding an 16S gene with at least 95%, 97%, 97.5%, 98%, 99% or 100% sequence identity to SEQ ID NO: 42 or 43.
- the propionate producing bacterium is or includes a strain of Veillonella sp. that has or includes a nucleic acid sequence encoding an rpoB gene with at least 97%, 97.5%, 98%, 99% or 100% sequence identity to SEQ ID NO: 40 and a nucleic acid sequence encoding a 16S gene with at least 97%, 97.5%, 98%, 99% or 100% sequence identity to SEQ ID NO: 42.
- the propionate producing bacterium is or includes a strain of Veillonella sp.
- the propionate producing bacterium is or includes a strain of bacteria that has, includes, or expresses a methylmalonyl-CoA decarboxylase subunit alpha and/or a methylmalonyl-CoA decarboxylase subunit beta protein and/or has or includes one or more genes encoding a methylmalonyl-CoA decarboxylase subunit alpha and/or a methylmalonyl-CoA decarboxylase subunit beta.
- the methylmalonyl-CoA decarboxylase alpha and beta subunits are from and/or are identical to alpha and beta subunits found in and/or expressed by a Veillonella sp.
- the propionate producing bacterium is or includes a strain of bacteria that has, includes, or expresses a protein having an amino acid sequence with at least 85%, 90%, 95%, 98%, or 99% identity to SEQ ID NO: 50 or 51.
- the propionate producing bacterium is or includes a strain of bacteria that has, includes, or expresses a protein having an amino acid sequence with 100% identity to SEQ ID NO: 50 or 51.
- the at least one propionate producing bacteria is or includes at least one strain of bacterium from the genus Megasphaera.
- the at least one strain of bacterium from the genus Megasphaera is capable of converting lactate to propionate.
- the at least one strain of bacterium from the genus Megasphaera is capable of converting lactate to propionate and butyrate.
- the at least one propionate producing bacterium is or includes at least one strain of Megasphaera sp.
- the propionate producing bacterium is or includes Megasphaera elsdenii (M.
- the propionate producing bacterium is or includes Megasphaera elsdenii and/ or Megasphaera massiliensis.
- the propionate producing bacterium is or includes a strain of Megasphaera sp. that has or includes a nucleic acid sequence encoding an 16S gene having a sequence of at least 200, 300, 400, 500, 600, 700, 800, 900, 1,000, 1,200, or 1,400 nucleotides in length with at least 95%, 97%, 97.5%, 98%, 99% or 100% sequence identity to SEQ ID NO: 52 or 53.
- the propionate producing bacterium is or includes a strain of Megasphaera sp. that has or includes a nucleic acid sequence encoding an 16S gene with at least 95%, 97%, 97.5%, 98%, 99% or 100% sequence identity to SEQ ID NO: 52 or 53.
- the at least one propionate producing bacteria is or includes at least one strain of bacterium from the genus Anaerotignum.
- the at least one strain of bacterium from the genus Anaerotignum is capable of converting lactate to propionate and butyrate.
- the at least one propionate producing bacterium is or includes at least one strain of Anaerotignum sp.
- the propionate producing bacterium is or includes Anaerotignum lactatifermentans (A. lactatifermentans).
- the propionate producing bacterium is or includes a strain of Anaerotignum sp. that has or includes a nucleic acid sequence encoding an 16S gene having a sequence of at least 200, 300, 400, 500, 600, 700, 800, 900, 1,000, 1,200, or 1,400 nucleotides in length with at least 95%, 97%, 97.5%, 98%, 99% or 100% sequence identity to SEQ ID NO: 54.
- the propionate producing bacterium is or includes a strain of Anaerotignum sp. that has or includes a nucleic acid sequence encoding an 16S gene with at least 95%, 97%, 97.5%, 98%, 99% or 100% sequence identity to SEQ ID NO: 54.
- the at least one propionate producing bacteria is or includes at least one strain of bacterium from the genus Bacteroides.
- the at least one strain of bacterium from the genus Bacteroides is capable of converting lactate to propionate and butyrate.
- the at least one propionate producing bacterium is or includes at least one strain of Bacteroides sp.
- the propionate producing bacterium is or includes Bacteroides fragilis (B. fragilis and/ or Bacteroides caccae (B. caccae).
- the propionate producing bacterium is or includes a strain of Bacteroides sp. that has or includes a nucleic acid sequence encoding an 16S gene having a sequence of at least 200, 300, 400, 500, 600, 700, 800, 900, 1,000, 1,200, or 1,400 nucleotides in length with at least 95%, 97%, 97.5%, 98%, 99% or 100% sequence identity to SEQ ID NO: 55 and/or 57.
- the propionate producing bacterium is or includes a strain of Bacteroides sp. that has or includes a nucleic acid sequence encoding an 16S gene with at least 95%, 97%, 97.5%, 98%, 99% or 100% sequence identity to SEQ ID NO: 55 and/or 57.
- the at least one propionate producing bacteria is or includes at least one strain of bacterium from the genus Coprococcus.
- the at least one strain of bacterium from the genus Coprococcus is capable of converting lactate to propionate.
- the at least one propionate producing bacterium is or includes at least one strain of Coprococcus sp.
- the propionate producing bacterium is or includes Coprococcus catus (C catus).
- the propionate producing bacterium is or includes a strain of Coprococcus sp. that has or includes a nucleic acid sequence encoding an 16S gene having a sequence of at least 200, 300, 400, 500, 600, 700, 800, 900, 1,000, 1,200, or 1,400 nucleotides in length with at least 95%, 97%, 97.5%, 98%, 99% or 100% sequence identity to SEQ ID NO: 56.
- the propionate producing bacterium is or includes a strain of Coprococcus sp. that has or includes a nucleic acid sequence encoding an 16S gene with at least 95%, 97%, 97.5%, 98%, 99% or 100% sequence identity to SEQ ID NO: 56.
- the at least one propionate producing bacteria is or includes at least one strain of bacterium from the genus Merdimmobilis.
- the at least one strain of bacterium from the genus Merdimmobilis is capable of converting lactate to propionate.
- the at least one propionate producing bacterium is or includes at least one strain of Merdimmobilis sp.
- the propionate producing bacterium is or includes Merdimmobilis hominis).
- the propionate producing bacterium is or includes a strain of Merdimmobilis sp. that has or includes a nucleic acid sequence encoding an 16S gene having a sequence of at least 200, 300, 400, 500, 600, 700, 800, 900, 1,000, 1,200, or 1,400 nucleotides in length with at least 95%, 97%, 97.5%, 98%, 99% or 100% sequence identity to SEQ ID NO: 58.
- the propionate producing bacterium is or includes a strain of Merdimmobilis sp. that has or includes a nucleic acid sequence encoding an 16S gene with at least 95%, 97%, 97.5%, 98%, 99% or 100% sequence identity to SEQ ID NO: 58.
- the at least one propionate producing bacteria is or includes at least one strain of bacterium from the genus Blautia.
- the at least one strain of bacterium from the genus Blautia is capable of converting human milk oligosaccharides and/or 1,2-propanediol to propionate and/or butyrate.
- the at least one propionate producing bacterium is or includes at least one strain of Blautia sp.
- the propionate producing bacterium is or includes Blautia obeum.
- the at least one propionate producing bacteria is or includes at least one strain of bacterium from the genus Ruminococcus.
- the at least one strain of bacterium from the genus Ruminococcus is capable of converting human milk oligosaccharides and/or 1,2-propanediol to propionate and/or butyrate.
- the at least one propionate producing bacterium is or includes at least one strain of Ruminococcus sp.
- the propionate producing bacterium is or includes Ruminococcus torques.
- the at least one propionate producing bacteria is or includes at least one strain of bacterium from the genus Roseburia.
- the at least one strain of bacterium from the genus Roseburia is capable of converting human milk oligosaccharides and/or 1,2-propanediol to propionate and/or butyrate.
- the at least one propionate producing bacterium is or includes at least one strain of Roseburia sp.
- the propionate producing bacterium is or includes Roseburia inulinivorans .
- compositions that are or include at least one strain of the genus Bifidobacteria, e.g., a strain of Bifidobacteria capable of consuming human milk oligosaccharides such as B. longum subsp. infantis.
- Bifidobacteria is or includes two or more strains or species of Bifidobacteria, such as any two or more of those described herein, e.g., in Section I-B.
- Some suitable Bifidobacteria strains may be described in PCT App. Pub. No. W02021061991, hereby incorporated by reference in its entirety.
- the at least one strain of Bifidobacterium is capable of consuming or metabolizing non-digestible carbohydrates, e.g., oligosaccharides such as human milk oligosaccharides.
- at least one strain of Bifidobacterium is capable of consuming or metabolizing oligosaccharides such as human milk oligosaccharides.
- at least one strain of Bifidobacterium is capable of utilizing the prebiotics of the prebiotic mixture, e.g., human milk oligosaccharides, as a carbon source.
- human milk oligosaccharides are preferentially consumed or metabolized by the at least one strain of Bifidobacterium, e.g., as compared to other microbes or bacteria present in the gut or microbiome.
- the at least one strain of Bifidobacterium is capable of consuming or metabolizing one or more prebiotics of the mixture, including those of any of the prebiotic mixtures described herein, e.g., in Section I-C.
- the at least one strain of Bifidobacterium is capable of consuming or metabolizing all or essentially all of the oligosaccharides of the prebiotic mixture.
- the at least one strain of Bifidobacterium is capable of consuming or metabolizing human milk oligosaccharides.
- strains of Bifidobacteria that consume or metabolize human milk oligosaccharides are known and may be identified by routine techniques such as those described in Gotoh et al. Sci Rep. 2018 Sep 18;8(1): 13958, incorporated by reference herein in its entirety.
- the at least one strain of Bifidobacterium contains one or more enzymes capable of hydrolyzing the prebiotics of the mixture. In certain embodiments, the at least one strain of Bifidobacterium contains one or more enzymes capable of hydrolyzing the human milk oligosaccharides. In particular embodiments, the one or more enzymes hydrolyze external oligosaccharides, e.g., oligosaccharides such as human milk oligosaccharides that are outside of the Bifidobacterial cell. In some embodiments, the one or more enzymes hydrolyze oligosaccharides such as human milk oligosaccharides internally or within the Bifidobacterial cell. In certain embodiments, the one or more enzymes hydrolyze internalized human milk oligosaccharides.
- the at least one strain of Bifidobacterium contains one or more enzymes capable of hydrolyzing one or more human milk oligosaccharides.
- the one or more enzymes hydrolyze external human milk oligosaccharides.
- the one or more enzymes hydrolyze human milk oligosaccharides that are outside of the Bifidobacterial cell.
- the one or more enzymes hydrolyze human milk oligosaccharides internally.
- the one or more enzymes hydrolyze human milk oligosaccharides within the probiotic cell.
- the one or more enzymes hydrolyze internalized human milk oligosaccharides.
- the at least one strain of Bifidobacterium is capable of internalizing human milk oligosaccharides. In certain embodiments, the at least one strain of Bifidobacterium internalizes human milk oligosaccharides prior to hydrolyzing the human milk oligosaccharides. In various embodiments, the at least one strain of Bifidobacterium selectively or exclusively utilizes human milk oligosaccharides as a carbon source.
- the at least one strain of Bifidobacterium is administered to the subject and/or has engrafted, e.g., within the subject’s microbiome (such as the intestinal microbiome)
- the at least one strain of Bifidobacterium is present, expands, or increases in amount within the subject’s microbiome when human milk oligosaccharides are administered to and/or ingested by the subject, and, in certain embodiments, the at least one strain of Bifidobacterium is no longer present and/or decreases in amount within the subject’s microbiome when the human milk oligosaccharides are no longer ingested or administered.
- the at least one strain of Bifidobacterium is capable of internalizing oligosaccharides, such as to consume or metabolize the oligosaccharides.
- the at least one strain of Bifidobacterium is capable of internalizing one or more oligosaccharides of the mixture, including those of any of the oligosaccharides or mixtures described herein, e.g., in Section I-C.
- the probiotic strain is capable of internalizing human milk oligosaccharides.
- the at least one strain of Bifidobacterium is or includes a strain of B. longum subsp. infantis, B. adolescentis, B. animalis subsp. animalis, B. animalis subsp. lactis B. bifidum, B. breve, B. catenulatum, B. longum subsp. longum, B. pseudocatanulatum, or B. pseudoIongum.
- the species or subspecies of a given strain of Bifidobacteria may be identified by routine techniques. For example, in some embodiments, the species or subspecies is identified by assessing the sequence similarity of one or more genes to corresponding sequences of known members of bacterial species or subspecies. In certain embodiments, a probiotic strain falls within a species or subspecies if all or a portion of its 16S gene has at least 97% sequence identity to all or a portion of a known 16S sequence of a known strain falling within the species.
- a probiotic strain falls within a species or subspecies if all or a portion of its 16S gene has at least 97% sequence identity to all or a portion of a known 16S sequence of a known strain falling within the species.
- Exemplary full or partial 16S sequences are summarized in Table 1.
- the at least one strain of Bifidobacterium has or includes a nucleic acid sequence with at least 97%, at least 98%, at least 99%, or at least 99.5% identity to a nucleic acid sequence set forth in any of SEQ ID NOS: 1-16 or 20-39.
- the at least one strain of Bifidobacterium has or includes a nucleic acid sequence with at least 97%, at least 98%, at least 99%, or at least 99.5% identity to a nucleic acid sequence set forth in any of SEQ ID NOS: 1-7 or 20-39.
- the at least one strain of Bifidobacterium has or includes a nucleic acid sequence with at least 97%, at least 98%, at least 99%, or at least 99.5% identity to a nucleic acid sequence set forth in any of SEQ ID NOS: 1-7 or 11. In certain embodiments, the at least one strain of Bifidobacterium has or includes a nucleic acid sequence with at least 97%, at least 98%, at least 99%, or at least 99.5% identity to a nucleic acid sequence set forth in any of SEQ ID NOS: 1-16.
- the at least one strain of Bifidobacterium has or includes a nucleic acid sequence with at least 97%, at least 98%, at least 99%, or at least 99.5% identity to a nucleic acid sequence set forth in any of SEQ ID NOS: 1-7.
- the at least one strain of Bifidobacterium is or includes a strain of B. longum subsp. infantis.
- the strain of B. longum subsp. infantis has or includes a nucleic acid sequence with at least 97%, at least 98%, at least 99%, or at least 99.5% identity to a nucleic acid sequence set forth in any of SEQ ID NOS: 1-7 or 20-39.
- infantis has or includes a nucleic acid sequence of at least 200, 300, 400, 500, 600, 700, 800, 900, 1,000, 1,200, or 1,500 nucleotides in length with at least 60%, 70%, 80%, 90%, 95%, 99%, or 99.9% sequence identity to a nucleic acid sequence set forth in SEQ ID NOS: 1-7 or 20-39.
- the strain of B. longum subsp. infantis has or includes nucleic acid sequences having at least 90%, 95%, or 99% sequence identity to one or more of the nucleic acid sequences set forth in SEQ ID NOS: 20-39.
- infantis has or includes the nucleic acid sequences set forth in one or more of SEQ ID NOS: 20-39.
- the strain of B. longum subsp. infantis has or includes nucleic acid sequences having at least 90%, 95%, or 99% sequence identity to all of the nucleic acid sequences set forth in SEQ ID NOS: 20-39.
- the strain of B. longum subsp. infantis has or includes the nucleic acid sequences set forth in SEQ ID NOS: 20-39.
- At least one strain of Bifidobacterium capable of consuming, metabolizing, and/or internalizing human milk oligosaccharides HMO cannot be metabolized by the host, e.g., mammals such as humans, or most bacteria, including most bacteria commonly found in the microbiome of adult humans.
- some strains, species, or subspecies of Bifidobacterium, such as B. longum subsp. infantis have enzymatic activity able to degrade specific alpha and beta bonds of human milk oligosaccharides.
- Bifidobacterium Five monosaccharides can be found in different HMO structures, glucose, galactose, N-acetyl glucosamine, fucose, and sialic acid (also referred to herein as N-acetyl neuraminic acid). Some strains, species, or subspecies of Bifidobacterium are able to fully degrade HMO intracellularly. Such Bifidobacterium possess genes encoding specific transporters (e.g., ABC transporters such as those described in Sela et al. PNAS (2008) 105 (48) 18964-18969; Schell, et al. PNAS. (2002) 99(22): 14422-14427 and LoCascio et al.
- specific transporters e.g., ABC transporters such as those described in Sela et al. PNAS (2008) 105 (48) 18964-18969; Schell, et al. PNAS. (2002) 99(22): 14422
- the probiotic strain is at least one strain of bacterium having one or more genes encoding all or a portion of a transporter, e.g., an ABC transporter, capable of internalizing an oligosaccharide such as an HMO.
- the probiotic strain is a bacterium having one or more genes encoding one or more enzymes, e.g., alpha-fucosidase, alpha-sialidase, beta-galactosidase, and beta-N-hexosaminidase, capable degrading an oligosaccharide such as an HMO.
- the probiotic strain is at least one strain of Bifidobacterium or Bacteroides having one or more genes encoding all or a portion of a transporter, e.g., an ABC transporter, capable of internalizing an oligosaccharide, e.g., an HMO.
- a transporter e.g., an ABC transporter
- the at least one strain of Bifidobacterium is B. longum subsp. infantis.
- B. longum subsp. infantis is known and readily identifiable by those of skill in the art using routine techniques.
- B. longum subsp. infantis including its genome and biology, are known and for example have been described, including in Sela et al. PNAS (2008) 105 (48) 18964- 18969; Underwood et al., Pediatr Res. (2015) 77(0): 229-235, incorporated by reference herein.
- Bifidobacterium e.g., B. longum subsp.
- infantis may be isolated using known selective microbiological media, e.g., De Man, Rogosa and Sharpe agar (MRS), optionally in combination with mupirocin, or those described in O’Sullivan et al., J Appl Microbiol. 2011 Aug;l 11(2):467-73, incorporated by reference herein.
- suitable sources for isolating Bifidobacterium e.g., B. longum subsp. infantis
- suitable sources for isolating Bifidobacterium e.g., B. longum subsp. infantis
- suitable sources for isolating Bifidobacterium e.g., B. longum subsp. infantis
- suitable sources for isolating Bifidobacterium e.g., B. longum subsp. infantis
- bacterial colonies may be identified or characterized by routine biochemical techniques, such as PCR.
- infantis is identified by taqman qPCR, such as described in Lawley et al., PeerJ. 2017 May 25;5:e3375. e.g., as performed with forward primer sequence ATACAGCAGAACCTTGGCCT (SEQ ID NO: 17), reverse primer sequence GCGATCACATGGACGAGAAC (SEQ ID NO: 18) and probe sequence [FAM dye] -TTTCACGGA - [ZEN quencher] - TCACCGGACCATACG - [3IABkFQ quencher] (SEQ ID NO: 19).
- a strain may be confirmed as B. longum subsp.
- the prebiotic mixture is a mixture of non-digestible carbohydrates, e.g., oligosaccharides such as human milk oligosaccharides (HMOs), that promotes the growth or expansion of the probiotic strain, e.g., in vivo such as in the human gut and/or within the human gut microbiome.
- HMOs human milk oligosaccharides
- the prebiotic mixture e.g., of non-digestible carbohydrates such as human milk oligosaccharides, promotes, e.g., selectively or exclusively, the colonization, expansion, extension, or increased presence of the probiotic strain within the microbiome.
- the mixture of non- digestible carbohydrates promotes the growth or expansion of a probiotic strain of Bifidobacterium such as B. longum subsp. infantis, e.g., in vivo such as in the human gut.
- the prebiotic mixture is a mixture of oligosaccharides, e.g., human milk oligosaccharides, that promote, e.g., selectively or exclusively, the colonization, expansion, extension, or increased presence of one or more strains of Bifidobacterium, e.g., B. longum subsp. infanlis, within the microbiome.
- the prebiotic mixture is or includes a mixture of non- digestible carbohydrates. In various embodiments, the prebiotic mixture is or includes a mixture of oligosaccharides. In particular embodiments, the prebiotic mixture is a mixture of one or more human milk oligosaccharides.
- the non-digestible carbohydrates are or include oligosaccharides.
- the non-digestible carbohydrates are or include milk oligosaccharides.
- the non-digestible carbohydrates are or include human milk oligosaccharides (human milk oligosaccharides).
- the prebiotic mixture is a mixture of non-digestible carbohydrates that are or include human milk oligosaccharides.
- the prebiotic mixture is a mixture of human milk oligosaccharides, such as those that are obtained or derived from permeate, e.g., permeate derived or obtained from pooled human milk.
- the provided mixture may contain any oligosaccharide that may be internalized by one or more strains of Bifidobacterium such as a strain of B. longum subsp. infantis.
- the oligosaccharides of the mixture may include one or more of a fructo-oligosacharide (FOS), galactooligosaccharide (GOS), transgalactooligosaccharide (TOS), gluco-oligosaccharide, xylo-oligosaccharide (XOS), chitosan oligosaccharide (COS), soy oligosaccharide (SOS), isomalto-oligosaccharide (IMOS), or derivatives thereof.
- FOS fructo-oligosacharide
- GOS galactooligosaccharide
- TOS transgalactooligosaccharide
- XOS xylo-oligosaccharide
- COS chitosan oli
- such derivatives include those with modifications that may increase the likelihood or probability of consumption, metabolism, and/or internalization (such as by transport or import) of the oligosaccharide by the probiotic strain, e.g., B. longum subsp. infantis.
- modifications may include but are not limited to fucosylation or sialylation.
- the oligosaccharides of the mixture may include one or more of a FOS, GOS, TOS, gluco-oligosaccharide, XOS, COS, SOS, IMOS, or derivatives or any or all of the foregoing, that are capable of being metabolized, consumed, and/or internalized by one or more strains, species, or subspecies of Bifidobacterium, e.g., B. longum subsp. infantis.
- the oligosaccharides of the mixture include one or more oligosaccharides that are obtained or derived from a resistant starch, pectin, psyllium, arabinogalactan, glucomannan, galactomannan, xylan, lactosucrose, lactulose, lactitol and various other types of gums such as tara gum, acacia, carob, oat, bamboo, citrus fibers, such as by treatment with enzymes that hydrolyze fiber or polysaccharides.
- the one or more oligosaccharides of the mixture that are obtained by these means are capable of being consumed, metabolized, and/or internalized by at least one strain of Bifidobacterium such as B. longum subsp. infantis.
- the prebiotic mixture is a mixture that is or includes at least one HMO.
- the prebiotic mixture is a mixture of human milk oligosaccharides that is or includes a plurality of human milk oligosaccharides.
- the prebiotic mixture is or includes a plurality of, of about, or at least 2, 3, 5, 10, 25, 50, 75, 100, 125, 150 different individual human milk oligosaccharides, e.g., human milk oligosaccharides with different individual chemical formulas or chemical structures.
- the prebiotic mixture is or includes a plurality of, of about, or at least 10, 25, 50, 75, 100, 125, 150 different individual human milk oligosaccharides. In some embodiments, the prebiotic mixture is or includes a plurality of, of about, or at least 25 different individual human milk oligosaccharides. In some embodiments, the prebiotic mixture is or includes a plurality of, of about, or of at least 80 different individual human milk oligosaccharides.
- an oligosaccharide is an HMO, such as if the oligosaccharide has a chemical formula and structure that is identical to an oligosaccharide that is found in human milk, as a matter of routine.
- the prebiotic mixture contains one or more synthetic human milk oligosaccharides, e.g., human milk oligosaccharides that are obtained, purified, or synthesized from a source other than human milk.
- synthetic human milk oligosaccharides as well as methods for synthesizing oligosaccharides and human milk oligosaccharides, are known, and include but are not limited to those described in PCT Publication Nos.: W02017101958, WO2015197082, WO2015032413, WO2014167538, WO2014167537, WO2014135167, W02013190531, W02013190530, WO2013139344, WO2013182206, WO2013044928, W02019043029, W02019008133, WO2018077892, WO2017042382, WO2015150328, WO2015106943, WO2015049331, WO2015036138, and W0
- the prebiotic mixture includes some or all of 2'- fucosyllactose, 3’ -fucosy llactose, 3’-sialyllactose, 6'-sialyllactose, Lacto-N-tetraose, lacto-N- difucohexaose I, lactodifucotetraose, lacto-N-fucopentaose I, sialylacto-N-tetraose c, sialylacto-N-tetraose b, and disialyllacto-N-tetraose.
- the mixture includes all of 2'-fucosyllactose, 3’-fucosyllactose, 3’-sialyllactose, 6'-sialyllactose, lacto-N- tetraose, lacto-N-difucohexaose I, lactodifucotetraose, lacto-N-fucopentaose I, sialylacto-N- tetraose c, sialylacto-N-tetraose b, and disialyllacto-N-tetraose.
- the prebiotic mixture includes some or all of 2- fucosyllactose, lacto-N-tetraorose, 3-sialyllactose, 3-fucosyllactose, lacto-N-fucopentaose I, lacto-N-fucopentaose II, and 6’sialyllactose.
- the prebiotic mixture includes some or all of 2'-fucosyl-lactose, 3’-fucosyl-lactose, 3’-sialyl-lactose, 6'- sialyl-lactose, lacto-N-tetraose, lacto-N-neo-tetraose, lacto-N-fucopentaose I, lacto-N- fucopentaose II, lacto-N-fucopentaose III, sialyl-lacto-N-tetraose b, sialyl-lacto-N-tetraose c, lacto-N-difuco-hexaose I, lacto-N-difuco-hexaose II, lacto-N-hexaose, para-lacto-N-hexaose, disialyllacto-N-tetraose
- the prebiotic mixture contains at least 25, 50, 100, 125, or 150 human milk oligosaccharides which include all of 2-fucosyllactose, lacto-N- tetraorose, 3-sialyllactose, 3-fucosyllactose, lacto-N-fucopentaose I, lacto-N-fucopentaose II, and 6’sialyllactose.
- the prebiotic mixture contains at least 25, 50, 100, 125, or 150 human milk oligosaccharides which include all of 2'-fucosyl-lactose, 3’- fucosyl-lactose, 3’-sialyl-lactose, 6'-sialyl-lactose, lacto-N-tetraose, lacto-N-neo-tetraose, lacto-N-fucopentaose I, lacto-N-fucopentaose II, lacto-N-fucopentaose III, sialyl-lacto-N- tetraose b, sialyl-lacto-N-tetraose c, lacto-N-difuco-hexaose I, lacto-N-difuco-hexaose II, lacto-N-hexaose, para-lacto-N-
- the prebiotic mixture is a mixture of human milk oligosaccharides that is, includes, or is obtained or derived from human milk or a fraction thereof. In certain embodiments, the prebiotic mixture is or includes a mixture of human milk oligosaccharides that is or is obtained from an ultra-filtered permeate from human skim milk. In various embodiments, the prebiotic mixture is, includes, or is derived or produced from a concentrated human milk permeate. In some embodiments, the mixture of human milk oligosaccharides or the concentrated human milk permeate is or is obtained from a process described herein, e.g., in Section I-C-i.
- the prebiotic mixture is a concentrated human milk permeate, such as those described in U.S. Pat. No. 8,927,027 or in PCT Application No. WO 2018053535, incorporated herein by reference.
- the prebiotic mixture is a prebiotic mixture produced or resulting from any of the methods described herein, e.g., in Section I-C- (i).
- the prebiotic mixture is or includes a concentrated human milk permeate that contains a plurality of human milk oligosaccharides.
- the human milk permeate is obtained by filtering human skim milk that was obtained by separating cream from whole human milk.
- the permeate was ultrafiltered from the human skim milk.
- the permeate is further concentrated, e.g., by reverse osmosis or nanofiltration, to increase the HMO content within the permeate.
- the human milk permeate e.g., the concentrated human milk permeate, has a concentration of at least 0.5%, 1%, 2.5%, 5%, or 10% w/v HMO.
- the human milk permeate may undergo additional processing steps, such as to digest or remove sugars, e.g., lactose, prior to its formulation or incorporate as a prebiotic mixture.
- the concentrated human milk permeate includes a plurality of, of about, or of at least 1, 2, 3, 5, 10, 25, 50, 75, 100, 125, 150 different individual human milk oligosaccharides, e.g., human milk oligosaccharides with different individual chemical formulas or chemical structures.
- the prebiotic mixture is or includes a plurality of, of about, or of at least 10, 25, 50, 75, 100, 125, 150 different individual human milk oligosaccharides.
- the prebiotic mixture is or includes a plurality of, of about, or of at least 25 different individual human milk oligosaccharides.
- the prebiotic mixture is or includes a plurality of, of about, or of at least 80 different individual human milk oligosaccharides.
- the prebiotic mixture is or includes a concentrated human milk permeate and contains at least 10, 25, 50, 75, 100, 125, 150 different individual human milk oligosaccharides.
- the concentrated human milk permeate contains at least 10, 25, 50, 100, 125, or 150 human milk oligosaccharides which include all of 2- fucosyllactose, lacto-N-tetraorose, 3-sialyllactose, 3-fucosyllactose, lacto-N-fucopentaose I, lacto-N-fucopentaose II, and 6’sialyllactose.
- the concentrated human milk permeate and/or the prebiotic mixture has an increased amount, level, or concentration of one or more human milk oligosaccharides as compared to what is typically found human milk.
- the prebiotic mixture has an increased amount, level, or concentration of one or more human milk oligosaccharides as compared to what is typically found in untreated human milk permeate, e.g., permeate resulting from ultrafiltration of pooled human skim milk.
- the prebiotic mixture is or includes at least 25, 50, 75, 100, 125, 150, of the different human milk oligosaccharides found, present, or detected in pooled human milk or in permeate (e.g., permeate resulting from ultra-filtering skim) obtained from pooled human milk.
- the prebiotic mixture is or includes at least 50%, 60%, 70%, 80%, 85%, 90%, 95%, 97%, 99%, or 99.9% of the different human milk oligosaccharides found, present, or detected in pooled human milk or in permeate (e.g., permeate resulting from ultra-filtering skim) obtained from pooled human milk.
- the prebiotic mixture is or includes at least 50%, 60%, 70%, 80%, 85%, 90%, 95%, 97%, 99%, or 99.9% of the individual human milk oligosaccharides that may be found, present, or detected across samples of human milk, e.g., samples of milk obtained from different individuals.
- the prebiotic mixture of human milk oligosaccharides is or includes the same or substantially the same human milk oligosaccharides found, present, or detected in pooled human milk or in permeate (e.g., permeate resulting from ultra-filtering skim) obtained from pooled human milk.
- the prebiotic mixture is or includes a human milk permeate resulting from the ultrafiltration of human whole or skim milk pooled from at milk collected from at least 10, 25, 50, or 100 individual human milk donors that is further concentrated, e.g., by nanofiltration or reverse osmosis, to increase the concentration of total HMO (e.g., by w/w).
- the prebiotic mixture is free or essentially free of oligosaccharides that are not human milk oligosaccharides.
- the prebiotic mixture contains human milk permeate, e.g., concentrated human milk permeate, and one or more synthetic human milk oligosaccharides, e.g., one or more of synthetically derived 2'-fucosyl-lactose, 3’-fucosyl-lactose, 3’-sialyl-lactose, 6'-sialyl-lactose, lacto-N- tetraose, lacto-N-neo-tetraose, lacto-N-fucopentaose I, lacto-N-fucopentaose II, lacto-N- fucopentaose III, sialyl-lacto-N-tetraose b, sialyl-lacto-N-t
- Prebiotic mixtures containing human milk oligosaccharides for use in the compositions and methods disclosed herein may be obtained according to methods known in the art, including, but not limited to, chemical synthesis and purification from human milk.
- processes to obtain HMO compositions from human milk are described below and are detailed in PCT Pub. Nos. WO/2010/065652 and WO/2018/053535, the contents of which are hereby incorporated in their entirety.
- the prebiotic mixture is a mixture of human milk oligosaccharides that are or are derived from a concentrated ultra-filtered human milk permeate, e.g., any ultra-filtered human milk permeate described herein such as in Section I- C-(i).
- the prebiotic mixtures are mixtures of human milk oligosaccharides having an HMO profile that is substantially similar both structurally and functionally to the profile of human milk oligosaccharides observed across the population of whole human milk. That is to say, in some aspects, since the prebiotic mixtures may be obtained from a source of human milk derived from a pool of donors rather than an individual donor, the array of human milk oligosaccharides will be more diverse than in any one typical individual, and will represent or more closely represent the spectrum of human milk oligosaccharides that are found among human milk across a population as opposed to the spectrum of human milk oligosaccharides that are found or typically found in the human milk produced by any particular individual. Thus, in some embodiments, the prebiotic mixture and the concentrated human milk permeate have more individual HMO species than what can be found in human milk obtained from an individual donor.
- the ratio of the amount or concentration of individual human milk oligosaccharide species to total human milk oligosaccharides of the prebiotic mixture or the concentrated human milk permeate may be different from what would be observed in whole human milk or pooled human milk.
- the prebiotic mixture is or includes a greater amount of different individual human milk oligosaccharides than the number of different individual human milk oligosaccharides found in human milk from an individual donor.
- the prebiotic mixture includes at least 1, 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50 more individual human milk oligosaccharides than the number of different individual human milk oligosaccharides found in human milk from an individual donor.
- a prebiotic mixture is or includes a greater amount of different individual human milk oligosaccharides than the mean or median number of different individual human milk oligosaccharides found in a plurality of human milk samples from individual donors.
- the prebiotic mixture includes at least 1, 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50 more individual human milk oligosaccharides than the number of different individual human milk oligosaccharides found in human milk from an individual donor.
- one of the biggest variables in HMO diversity derives from the mother’s Lewis blood group and specifically whether or not she has an active fucosyltrasferase 2 (FUT2) and/or fucosyltrasferase 3 (FUT3) gene.
- FUT2 fucosyltrasferase 2
- FUT3 fucosyltrasferase 3
- those without an active FUT2 gene may comprise a more varied array of, for example al, -4 linked oligosaccharides (as compared to secretors), but comprise an overall decrease in diversity since they are unable to synthesize a major component of the secretor’ s HMO repertoire.
- the prebiotic mixture includes human milk oligosaccharides that include al-2 linked fucose and human milk oligosaccharides that include al -4 linked fucose.
- the prebiotic mixture is or includes at least 5% total HMO (w/w). In particular embodiments, the prebiotic mixture is or includes least 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 15%, 20%, 25%, or 50% total HMO (w/w). In certain embodiments, the prebiotic mixture is or includes between 5% and 15%, 7.5% and 12.5%, 8% and 12%, 8.5% and 11%, or 8.4% and 10.6% total HMO (w/w). In certain embodiments, the prebiotic mixture is or includes between 8.5% and 11% total HMO (w/w). In some embodiments, the prebiotic mixture is or includes between 8.4% and 10.6% total HMO (w/w).
- the prebiotic mixture has a pH of between 4.0 and 5.5. In certain embodiments, the prebiotic mixture has less than 10%, 5%, 1%, or 0.1% lactose (w/w). In some embodiments, the prebiotic mixture has less than 10%, 5%, 1%, or 0.1% glucose (w/w). In particular embodiments, the prebiotic mixture has less than 10%, 5%, 1%, or 0.1% galactose (w/w). In certain embodiments, the prebiotic mixture has less than 10% galactose, less than 10% glucose, and less than 0.1% lactose.
- the prebiotic mixture is a liquid formulation.
- the prebiotic mixture is in powdered form, e.g., a lyophilized or spray dried composition.
- the prebiotic mixture is incorporated into a dosage form that is a separate composition from the Bifidobacterium strain.
- the prebiotic mixture is incorporated into a dosage form that is a separate composition from the propionate producing strain.
- the prebiotic mixture is or includes human milk oligosaccharides (HMOs) obtained or purified from ultra-filtered permeate from donated human milk.
- the donated human milk is pooled to provide a pool of human milk.
- a pool of human milk comprises milk from two or more (e.g., ten or more) donors.
- the pooled human milk contains milk from at least 50, 75, 100, 150, or 200 individual donors.
- the pooled human milk contains human milk from at least 100 individual donors or between 100 and 300 individual donors.
- the pooled human milk contains milk from at least ten, at least twenty -five, at least fifty, at least seventy-five, at least one hundred, or at least one hundred fifty individual human milk donors.
- the human milk oligosaccharides that are contained or included in the prebiotic mixture are synthetic human milk oligosaccharides, such as those derived from non-human milk sources, e.g., derived or obtained as oligosaccharides or precursors from transgenic microorganisms and/or chemically synthesized.
- synthetic oligosaccharides and human milk oligosaccharides are known, and include but are not limited to those described in PCT Publication Nos.: W02017101958, WO2015197082, WO2015032413, WO2014167538, WO2014167537, WO2014135167, WO2013190531, WO2013190530, WO2013139344, WO2013182206, WO2013044928, W02019043029, W02019008133, WO2018077892, WO2017042382, WO2015150328, WO2015106943, WO2015049331, WO2015036138, and W02012097950, each of which is incorporated by reference herein in its entirety.
- one or more synthetically derived human milk oligosaccharides are added to a concentrated human milk permeate to arrive at a prebiotic mixture.
- the mixture of oligosaccharides described herein are produced from human milk permeate, e.g., concentrated ultra-filtered permeate from pooled human milk.
- the mixture of oligosaccharides described herein contain or are formulated with human milk permeate, e.g., concentrated ultra-filtered permeate from pooled human milk.
- the concentrated ultra-filtered permeate may be made according to any suitable method or technique known in the art.
- suitable methods and techniques include those described in U.S. Pat. No. 8,927,027m PCT Pub. No. WO2018053535, and PCT Pub. No. WO 2021061991, hereby incorporated by reference in their entirety. Exemplary methods and techniques for producing the human milk compositions are briefly summarized herein.
- the prebiotic mixture is or includes human milk permeate that has been generated or produced by a method described herein.
- whole human milk is pooled from multiple donors, and then cream and skim are separated by any suitable technique known in the art, e.g., centrifugation; and then the skim milk is filtered, e.g., ultra-filtered, to obtain human milk permeate and retentate.
- the human milk permeate may be further processed, such as to remove or digest one or more components, e.g., lactose, or to increase the concentration of human milk oligosaccharides, such as by nanofiltration or reverse osmosis.
- the donor milk is received frozen, and when desired, is thawed and pooled. In some embodiments, donor milk is then screened, e.g., to identify contaminants, by one or more of the methods discussed herein.
- the pooled milk is filtered, e.g., through about a 200- micron filter.
- the pooled milk is heated, e.g., at about 63°C or greater for about 30 minutes or more.
- the milk is transferred to a separator, e.g., a centrifuge, to separate the cream from the skim.
- the cream may go through separation once again to yield more skim.
- a desired amount of cream is added to the skim prior to ultra-filtration.
- material that that did not pass through the filter is collected as the retentate fraction, and material that passes through the filter is collected as the permeate fraction.
- the skim fraction undergoes ultra-filtration.
- the ultrafiltration is performed with a filter between 1 kDa and 1000 kDa to obtain a protein rich retentate and the HMO-containing permeate. Details of this process can be found, for example, in US 8,545,920; US 7,914,822; 7,943,315; 8,278,046; 8,628,921; and 9,149,052, each of which is hereby incorporated by reference in its entirety.
- the ultra-filtration is performed with a filter that is between 1 kDa and 100 kDa, 5 kDa and 50 kDa, or 10 kDa and 25 kDa.
- the filter is about or at least 5 kDa, 10 kDa, 20 kDa, 25 kDa, 50 kDa, or 100 kDa. In some embodiments, the skim fraction undergoes ultrafiltration with a filter that is about 10 kDa. In certain embodiments, the skim fraction undergoes ultrafiltration with a filter that is about 25 kDa. In particular embodiments, the skim fraction undergoes ultrafiltration with a filter that is about 50 kDa.
- the ultra-filtered permeate undergoes a process for reducing lactose.
- a process for producing a concentrated human milk permeate composition with substantially reduced levels of lactose is provided.
- the substantial reduction includes or requires the biochemical and/or enzymatic removal of lactose from the lactose-rich human milk permeate fraction, without loss of yield or change in molecular profile of the HMO content of human milk permeate. And, in particular embodiments, without leaving residual inactivated foreign protein, if enzymatic digestion is used to reduce lactose.
- the permeate is free or essentially free of lactose following the enzymatic digestion.
- the process for reducing lactose from human milk permeate includes one or more of the steps of a) adjusting the pH of the permeate mixture; b) heating the pH adjusted mixture; c) adding lactase enzyme to the heated permeate mixture to create a permeate/lactase mixture and incubating a period of time; d) removing the lactase from the mixture and filtering the mixture to remove lactase; and e) concentrating human milk oligosaccharides.
- the order of when steps (a)-(c) are performed may be varied.
- the steps may be performed in the order of (a)-(b)-(c); (a)-(c)-(b); (c)-(b)-(a); (c)-(a)-(b); (b)-(a)-(c); or (b)-(c)-(a), such that, for example, the lactase enzyme may be added prior to heating the mixture, or, alternatively at any point during the heating process. Similarly, and also by way of example only, the mixture may be heated prior to adjustment of the pH. Furthermore, several steps may be grouped into a single step, for example “enzymatically digesting lactose” or “lactases digestion of lactose” involves steps (a)-(c) as described. These steps may be performed concurrently or consecutive in any order. Therefore, as used herein “lactose digestion” refers to the performance of at least these three steps, in any order, consecutively or concurrently.
- the pH of the permeate is adjusted to a pH of about 3 to about 7.5. In some embodiments, the pH is adjusted to a pH of about 3.5 to about 7.0. In particular embodiments, the pH is adjusted to a pH of about 3.0 to about 6.0. In certain embodiments, the pH is adjusted to a pH of about 4 to about 6.5. In some embodiments, pH is adjusted to a pH of about 4.5 to about 6.0. In particular embodiments, the pH is adjusted to a pH of about 5.0 to about 5.5. In certain embodiments, the pH is adjusted to a pH of about 4.3 to about 4.7, preferably 4.5.
- the pH may be adjusted by adding acid or base. In some embodiments, pH is adjusted by adding acid, for example HC1. In particular embodiments, pH is adjusted by adding IN HC1 and mixing for a period of time, e.g., about 15 minutes.
- the pH-adjusted permeate is heated to a temperature of about of about 25°C to about 60°C. In certain embodiments, the permeate is heated to a temperature of about 30°C to about 55°C. In some embodiments, the permeate is heated to a temperature of about 40°C to about 50°C. In certain embodiments, the permeate is heated to a temperature of about 48°C to about 50°C. In some embodiments, the permeate is heated to a temperature about 50°C. In some embodiments, the permeate is heated to a temperature less than or equal to about 40°C.
- lactase enzyme is added to the pH-adjusted, heated permeate to create a permeate/lactase mixture.
- lactose within the permeate/lactase mixture is broken down into monosaccharides.
- lactase enzyme is added at about 0.1% w/w to about 0.5% w/w concentration.
- lactase enzyme is added at about 0.1% w/w, or 0.2% or 0.3% or 0.4% or 0.5% w/w.
- lactase enzyme may be derived from any origin (e.g., fungal or bacterial in origin).
- the pH-adjusted, heated permeate is incubated with the lactase enzyme for about 5 to about 225 minutes.
- the incubation time is about 15 min to about 90 min.
- the incubation time is about 30 minutes to about 90 minutes.
- the incubation time is about 60 minutes.
- pH, temperature, and enzyme incubation conditions are what work optimally for the process described herein, one of skill in the art would understand that modifications may be made to one or more of these variables to achieve similar results. For example, if less enzyme is used than the about 0.1% w/w to about 0.5% w/w described herein, the incubation time may need to be extended to achieve the same level of lactose digestion. Similar adjustments may be made to both the temperature and pH variables as well.
- the permeate/lactase mixture is cooled to a temperature of about 20°C to about 30°C. In a particular embodiment, the permeate/lactase mixture is cooled to a temperature of about 25°C.
- the permeate/lactase mixture is clarified to remove insoluble constituents.
- insoluble material may form throughout the change in pH and temperature.
- it may be necessary or beneficial to clarify the mixture to remove these insoluble constituents for example, through a depth filter.
- the filters may be 0.1 to 10 micron filters. In some embodiments, the filters are about 1 to about 5 micron filters.
- removal of insoluble constituents can be achieved through a centrifugation process or a combination of centrifugation and membrane filtration.
- the clarification step is not essential for the preparation of a diverse HMO composition, as described herein, rather, this optional step aids in obtaining a more purified permeate composition.
- the clarification step is important in the reusability of the filtration membranes and thus to the scalability of the process.
- more or less stringent clarification may be performed at this stage in order to produce more or less purified permeate compositions, depending on formulation and application. For example, precipitated minerals may be less of a problem for a formulation destined for lyophilization.
- the spent and excess lactase enzyme is removed from the clarified permeate/lactase mixture.
- the inactivated foreign protein will carry no biological risk and therefore the added steps of lactase removal or even inactivation may not be necessary.
- the spent and excess lactase is inactivated, for example by high temperature, pressure, or both. In some embodiments, the inactivated lactase is not removed from the composition.
- lactase enzyme removal may be accomplished by ultrafiltration.
- ultrafiltration is accomplished using an ultrafiltration membrane, for example using a membrane with molecular weight cut-off of ⁇ 50,000 Dalton, e.g., a BIOMAX-50K.
- the molecular weight cut-off less than or equal to about 10 kDa. In certain embodiments, the molecular weight cut-off less than or equal to about 25 kDa. In particular embodiments, the molecular weight cut-off less than or equal to about 50 kDa.
- an additional ultrafiltration is performed through a smaller membrane than the initial membrane with molecular weight cut-off of ⁇ 50,000 Dalton.
- the additional ultrafiltration is performed with a membrane with a molecular weight cut-off of between 10 kDa and 50 kDa, 1 kDa and 10 kDa, 1 kDa and 5 kDa, or 2 kDa and 3 kDa.
- the additional ultrafiltration is performed with a membrane with a molecular weight cut-off of between 2 kDa and 3 kDa.
- an additional ultrafiltration is not performed.
- the additional filtration step is performed, such as to aid in the overall purity of the permeate product, such as by assisting in the removal of smaller potentially bioactive and/or immunogenic factors such as microRNAs and exosomes.
- the clarified mixture that has undergone at least one, and in some cases two or more rounds of ultrafiltration (or alternative lactase removal means) is further filtered to purify and concentrate human milk oligosaccharides and to reduce the mineral and monosaccharides content.
- filtration can be accomplished using a nanofiltration membrane.
- the membrane has a molecular weight cut-off of ⁇ 1,000 Dalton. In certain embodiments, the membrane has a molecular weight cut-off of between 1 kDa and 1,000 kDa. In certain embodiments, the membrane has a molecular weight cut-off of less than 600 Da. In certain embodiments, the membrane has a molecular weight cut-off between 400 Da and 500 Da.
- the additional nanofiltration removes monosaccharides, minerals, particularly calcium, and smaller molecules to produce the final purified HMO composition.
- additional or alternative steps may be taken for the removal of minerals.
- Such an additional step may include, for example, centrifugation, membrane clarification ( ⁇ 0.6 micron), or combination of centrifugation and membrane filtration of heated (> 40°C) or refrigerated/frozen and thawing of HMO Concentrate.
- the collected supernatant or filtrate of these additional or alternative steps in some embodiments, is concentrated further using a nanofiltration membrane.
- the nanofiltration comprises filtration through a membrane with a molecular cut-off of ⁇ 600 Dalton.
- these additional steps may be performed at any stage of the process, including but not limited to prior to or after pasteurization.
- the physical property of nanofiltration membranes can be modified, such as chemical modification, to selectively concentrate sialylated human milk oligosaccharides, for example, allowing greater efficiency of neutral human milk oligosaccharides removal from HMO concentrate, in instances where concentrated sialylated human milk oligosaccharides are preferred.
- the permeate may be further processed, e.g., concentrated or diluted.
- the permeate may be concentrated by a suitable process such as nanofiltration, reverse osmosis, or dried, e.g., lyophilized.
- the permeate is treated to reduce bioburden, such as by any means known in the art.
- the purified HMO composition is pasteurized. In some aspects, pasteurization is accomplished at > 63°C for a minimum of 30 minutes. Following pasteurization, the composition is cooled to about 25°C to about 30°C and clarified through a 0.2 micron filter to remove any residual precipitated material.
- the mixture of oligosaccharides is formulated with an ultra-filtered permeate obtained from human milk. In some embodiments, the mixture of oligosaccharides is formulated with permeate that has been ultra filtered from the skim fraction of pooled human milk. In certain embodiments, lactose is removed, e.g., enzymatically degraded, prior to formulation into the mixture of oligosaccharides. D. Butyrate producing bacteria
- a butyrate producing strain of bacteria is administered along with or in addition to the provided prebiotic mixture, e.g., of human milk oligosaccharides, at least one Bifidobacterium and at least one propionate producing bacterium.
- the at least one butyrate producing strain is capable of consuming lactate and/or acetate.
- the at least one butyrate producing strain is capable of producing, generating, and/or creating butyrate in the presence of lactate and/or acetate.
- the at least one butyrate producing strain is capable of producing, generating, and/or creating butyrate in the presence of the at least one Bifidobacterium, e.g., any Bifidobacterium described herein such as in Section I-B, and the prebiotic mixture, e.g., a prebiotic mixture described herein such as in Section I-C.
- the prebiotic mixture e.g., a prebiotic mixture described herein such as in Section I-C.
- Suitable butyrate producing strains are described in PCT App. Pub. No. WO2022036225, hereby incorporated by reference in its entirety.
- the at least one butyrate producing strain is or includes at least one, two, three, four, five, six, seven, eight, nine, ten, or more species, subspecies, or strains of bacteria capable of producing butyrate.
- the butyrate producing strain is or includes at least three strains of bacteria capable of producing butyrate.
- the butyrate producing strain is or includes between one and five species, subspecies, or strains of bacteria capable of producing butyrate.
- the at least one butyrate producing strain has one or more genes that contribute to the production, generation, or making of butyrate.
- the at least one butyrate producing strain has a functional butyryl-Co A: acetate CoA-transferase (but) gene.
- the at least one butyrate producing strain has a functional butyrate kinase (buk) gene.
- the at least one butyrate producing strain has a functional butyryl-CoA:4-hydroxybutyrate CoA transferase (4Hbf) gene.
- the at least one butyrate producing strain has a functional butyryl-Co A: acetoacetate CoA transferase (Ato) gene.
- the at least one butyrate producing strain is or includes one or more species, subspecies, or strains of Clostridium Cluster IV bacteria. In certain embodiments, the at least one butyrate producing strain is or includes one or more species, subspecies, or strains of Clostridium Cluster XlVa bacteria. In certain embodiments, the at least one butyrate producing strain is or includes one or more species, subspecies, or strains of bacteria belonging to the Clostridium, Eubacterium, Ruminococcus, Coprococcus, Dorea, Lachnospira, Roseburia, Butyrivibrio, or Anaerofilum genera.
- the at least one butyrate producing strain is or includes one or more species, subspecies, or strains of bacteria belonging to the Clostridium, Eubacterium, Ruminococcus, Coprococcus, Dorea, Lachnospira, Roseburia or Butyrivibrio genera.
- the at least one butyrate producing strain is or includes one or more species, subspecies, or strains of bacteria belonging to the Clostridium, Eubacterium, Ruminococcus ov Anaerofilum genera.
- the at least one butyrate producing strain is or includes one or more species, subspecies, or strains of Agathobacter rectalis (also referred to as Eubacterium rectale), Anaerobutyricum hallii (also referred to as Eubacterium hallii), Anaerobutyricum soehngenii, Anaerocolumna aminovalerica (also referred to as Clostridium aminovalericum), Anaerostipes butyraticus, Anaerostipes caccae, Anaerostipes hadrus (also referred to as Eubacterium hadrum), Anaerostipes rhamnosivorans, Anaerotruncus colihominis, Blautia argi, Blautia caecimuris, Blautia coccoides (also referred to as Clostridium coccoides), Blautia faecicola, Blautia faecis, Blautia
- the at least one butyrate producing strain is or includes one or more species, subspecies, or strains of Agathobacter rectalis, Anaerobutyricum hallii, Anaerostipes caccae, Blautia producta, Clostridium leptum, Faecalibacterium prausnitzii, Anaerobutyricum soehngenii, Roseburia hominis, or Roseburia intestinalis.
- the at least one butyrate producing strain is or includes one or more species, subspecies, or strains of Anaerostipes caccae, Clostridium innocuum, Roseburia hominis, or Roseburia intestinalis.
- compositions, kits, and articles of manufacture are or include a combination of prebiotics, e.g., prebiotic mixtures of non- digestible carbohydrates such as human milk oligosaccharides, and probiotics that include one or more strains of Bifidobacteria such as B. longum subsp. infantis and one or more strains of propionate producing bacteria, e.g., Veillonella sp.
- prebiotics e.g., prebiotic mixtures of non- digestible carbohydrates such as human milk oligosaccharides
- probiotics that include one or more strains of Bifidobacteria such as B. longum subsp. infantis and one or more strains of propionate producing bacteria, e.g., Veillonella sp.
- the prebiotic mixture and probiotics may be formulated as a pharmaceutical composition or a nutritional composition.
- the provided composition includes or incorporates the prebiotic mixture and the probiotic strains.
- the probiotic strains may be formulated as a pharmaceutical composition or a nutritional composition.
- the prebiotic mixture and the probiotic are formulated or manufactured as separate compositions.
- kits or articles of manufacture that are or include separate prebiotic and probiotic compositions.
- kits or articles of manufacture that are or include prebiotic mixtures that are or include one or more human milk oligosaccharides, and probiotics that include one or more strains of Bifidobacteria and one or more strains of propionate producing bacteria.
- the prebiotic mixture is or includes any of the prebiotic mixtures described herein, e.g., in Section I-C.
- the prebiotic mixture contains at least 2, at least 5, at least 10, at least 25, at least 50, at least 100, or at least 150 human milk oligosaccharides, e.g., at least two, three, four, five, or more than five synthetic human milk oligosaccharides or at least 10, at least 25, at least 50, or at least 100 human milk oligosaccharides from a human milk source such as human milk permeate.
- the one or more strains of Bifidobacteria are or include one or more strains of Bifidobacteria described herein, e.g., in Section I-B.
- the one or more strains of Bifidobacteria are capable of consuming, internalizing, and/or hydrolyzing human milk oligosaccharides.
- the one or more strains of propionate producing bacteria are or include any of the propionate producing bacteria described herein, e.g., in Section I-A.
- the one or more strains of propionate producing bacteria are or include at least one strain from the genus Veillonella or Megasphaera.
- kits or articles of manufacture are or include i) prebiotic mixtures that are or include at least one or more of 2'-fucosyllactose, 3’- fucosyllactose, 3’-sialyllactose, 6'-sialyllactose, lacto-N-tetraose, lacto-N-difucohexaose I, lactodifucotetraose, lacto-N-fucopentaose I, sialylacto-N-tetraose c, sialylacto-N-tetraose b, and/or disialyllacto-N-tetraose; ii) at least one Bifidobacterium that is or includes B.
- kits or articles of manufacture are or include i) prebiotic mixtures that are or include at least one, some, or all of 2'-fucosyllactose, 3’-fucosyllactose, 3’-sialyllactose, 6'-sialyllactose, lacto-N-tetraose, lacto-N-difucohexaose I, lactodifucotetraose, lacto-N-fucopentaose I, sialylacto-N-tetraose c, sialylacto-N-tetraose b, and/or disialyllacto-N-tetraose; ii) at least one B
- kits or articles of manufacture include one or more strains of butyrate producing bacteria, e.g., any of the butyrate producing bacteria described herein such as in Section I-D.
- the butyrate producing bacteria is or includes one or more of Agathobacter rectalis, Anaerobutyricum hallii, Anaerostipes caccae, Blautia producta, Clostridium leptum, Faecalibacterium prausnitzii, Anaerobutyricum soehngenii, Roseburia hominis, or Roseburia intestinalis.
- articles of manufacture that are or include prebiotic mixtures that are or include one or more human milk oligosaccharides, probiotics that include one or more strains of Bifidobacteria and one or more strains of propionate producing bacteria, and instructions for use, e.g., describing any method herein such as the methods described in Section II.
- the articles of manufacture are or include i) prebiotic mixtures that are or include at least one or more of 2'- fucosyllactose, 3’ -fucosy llactose, 3’-sialyllactose, 6'-sialyllactose, lacto-N-tetraose, lacto-N- difucohexaose I, lactodifucotetraose, lacto-N-fucopentaose I, sialylacto-N-tetraose c, sialylacto-N-tetraose b, and/or disialyllacto-N-tetraose; ii) at least one Bifidobacterium that is or includes B.
- longum subsp. infantis iii) at least one propionate producing bacterium that is or includes one or more strains of the genus of the genus Veillonella or Megasphaera, and iv) instructions for use describing one or more methods included herein in Section II.
- the articles of manufacture are or include i) prebiotic mixtures that are or include at least one, some, or all of 2'-fucosyllactose, 3’-fucosyllactose, 3’- sialyllactose, 6'-sialyllactose, lacto-N-tetraose, lacto-N-difucohexaose I, lactodifucotetraose, lacto-N-fucopentaose I, sialylacto-N-tetraose c, sialylacto-N-tetraose b, and/or disialyllacto-N-tetraose; ii) at least one Bifidobacterium that is or includes B.
- the articles of manufacture include one or more strains of butyrate producing bacteria described herein in Section I-D, such as one or more of Agathobacter rectalis, Anaerobutyricum hallii, Anaerostipes caccae, Blautia producta, Clostridium leptum, Faecalibacterium prausnitzii, Anaerobutyricum soehngenii, Roseburia hominis, or Roseburia intestinalis.
- strains of butyrate producing bacteria described herein in Section I-D such as one or more of Agathobacter rectalis, Anaerobutyricum hallii, Anaerostipes caccae, Blautia producta, Clostridium leptum, Faecalibacterium prausnitzii, Anaerobutyricum soehngenii, Roseburia hominis, or Roseburia intestinalis.
- provided herein are methods for treating, preventing, or ameliorating one or more diseases, disorders, or conditions in a subject in need thereof.
- the method is or includes steps for administering to the subject a combination of probiotic bacteria that is or includes at least one strain of Bifidobacterium, e.g., such as any described herein, e.g., in Section I-B, and a strain of bacterium capable of producing propionate, such as any of the propionate producing bacterium described herein, e.g., in Section I-A, and a prebiotic mixture, such as any described herein, e.g., in Section I-C.
- the prebiotic mixture, the Bifidobacterium, and the propionate producing bacterium are administered together, such as at the same time, on the same treatment days, and/or within the same dosage formula.
- the prebiotic mixture, the Bifidobacterium, and the propionate producing bacterium are administered separately, such as at different times, on different treatment days, and/or within different dosage formulations.
- the treatment may include administering the prebiotic mixture, the Bifidobacterium, and the propionate producing bacterium together during certain days or phases of the treatment and then separately during other certain days or phases of the treatment.
- provided herein are methods for treating, preventing, or ameliorating one or more diseases, disorders, or conditions that are or may be associated with dysbiosis, e.g., of the intestinal microbiome, in a subject in need thereof.
- the intestinal microbiome is involved in or associated with a number of physiological functions including digestion, metabolism, extraction of nutrients, synthesis of vitamins, prevention of pathogen colonization, and immune modulation.
- alterations or changes in composition and biodiversity of the intestinal microbiome may be associated with or exacerbate various metabolic states, gastrointestinal disorders, and other pathophysiological conditions.
- conditions, diseases, or disorders with inflammatory components or components relating to infection, allergy, or immune dysfunction may be exacerbated by dysbiosis or may have an underlying contribution of dysbiosis to the pathology.
- targeting the microbiome with the provided prebiotic and probiotic compositions may successfully treat, alleviate, or prevent a wide range of conditions, diseases, and disorders.
- a method for treating, reducing, ameliorating, or preventing dysbiosis is or includes steps for administering to the subject at least one strain of Bifidobacterium, e.g., such as any described herein, e.g., in Section I-B, a strain of bacterium capable of producing propionate, such as any of the propionate producing bacterium described herein, e.g., in Section I-A, and a prebiotic mixture, such as any described herein, e.g., in Section I-C.
- Bifidobacterium e.g., such as any described herein, e.g., in Section I-B
- a strain of bacterium capable of producing propionate such as any of the propionate producing bacterium described herein, e.g., in Section I-A
- a prebiotic mixture such as any described herein, e.g., in Section I-C.
- the one or more diseases, disorders, or conditions is, includes, or is associated with dysbiosis, e.g., of the intestinal microbiome.
- the microbiome is an intestinal microbiome of a human.
- the microbiome is the intestinal microbiome of an infant or young child.
- the microbiome is an intestinal microbiome of an adult human.
- the method is or includes steps for administering to the subject at least one strain of Bifidobacterium, e.g., such as any described herein, e.g., in Section I-B, a strain of bacterium capable of producing propionate, such as any of the propionate producing bacterium described herein, e.g., in Section I-A, and a prebiotic mixture, such as any described herein, e.g., in Section I-C for the treatment of one or more diseases, disorders, or conditions associated with inflammation, infection, allergy, immune dysfunction, or dysbiosis of the intestinal microbiome.
- the one or more conditions, diseases, or disorders is, includes, or is associated with dysbiosis.
- the one or more conditions, diseases, or disorders is, includes, or is associated with inflammation.
- the one or more condition, disease, or disorder is, includes, or is associated with an autoimmune disease.
- the one or more conditions, diseases, or disorders is or is associated with an allergy.
- the prebiotic mixture e.g., of human milk oligosaccharides, the Bifidobacterium, e.g., B. longum subsp. infantis, and the propionate producing bacteria, e.g., Veillonella sp., are administered to prevent a disease, disorder, or condition.
- the prebiotic mixture and the probiotic strain prevent a condition described herein, e.g, in Section II-B.
- the prebiotic mixture and the probiotic strain reduce the risk, likelihood, or probability of the disease, disorder, or condition, and/or of experiencing one or more symptoms associated with the disease, disorder, or condition.
- the risk, likelihood, or probability is reduced by at least 10%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 90%, 99%, or 99.9% as compared to alternative treatments or no treatments, or as compared to administration of the probiotic strain or prebiotic mixture alone.
- the subject is a human.
- the subject is an infant, a child, a juvenile, or an adult.
- the subject is at least 1 month, 3 months, 6 months, 12 months, 18 months, or 24 months of age.
- the subject is at least 1 year, 2 years, 5 years, 10 years, 12 years, 16 years, or at least 18 years of age.
- the subject is at least 12 years old.
- the subject is at least 18 years old.
- the subject is an adult.
- the subject is elderly, e.g, at least 65 or 75 years of age.
- the provided methods include one or more treatment phases that are or include administration of one or more of the Bifidobacterium, e.g., B. longum subsp. infantis, the propionate producing bacterium, e.g., Veillonella sp. and the prebiotic mixture, e.g., of human milk oligosaccharides.
- the method is or includes a treatment where all three of the propionate producing bacterium, the Bifidobacterium, and the prebiotic mixture are administered.
- the method is or includes a treatment phase where the Bifidobacterium and the prebiotic mixture are administered, e.g., in the absence of the propionate producing bacterium.
- the method is or includes a treatment phase where the prebiotic mixture is administered, e.g., in the absence of the propionate producing bacterium and the Bifidobacterium. In some embodiments, the method includes two or more treatment phases.
- the prebiotic mixture is administered daily for at least 2, 3, 4, 5, 7, 10, 14, 21, or 28 days, e.g., consecutive days.
- the prebiotic mixture is administered in an amount of at least 0.001 g, 0.01 g, 0.1 g, 1 g, 2 g, 3 g, 4 g, 5 g, 6 g, 7.5 g, 8 g, 9 g, 10 g, 12 g, 16 g, 18 g, 20 g, 25 g, or 50 g per day, e.g., total weight of the prebiotics such as non-digestible carbohydrates such as human milk oligosaccharides.
- the prebiotic mixture in an amount of at least 0.001 g, 0.01 g, 0.1 g, 1 g, 2 g, 3 g, 4 g, 5 g, 6 g, 7.5 g, 8 g, 9 g, 10 g, 12 g, 16 g, 18 g, 20 g, 25 g, or 50 g total human milk oligosaccharides per day.
- the prebiotic mixture is administered in an amount of between 0.1 g and 50 g; 0.5 g and 25 g, 1 g and 20 g, 2 g and 18 g, 1 g and 5 g, 2 g and 3 g, 3 g and 6 g, 4 g and 5 g, 5 g and 10 g, 8 g and 10 g, 10 g and 20 g, 15 g and 20 g, or 17 g and 19 g total human milk oligosaccharides per day.
- the prebiotic mixture is administered in an amount of, of about, or of at least 2 g, 4.5 g, 6 g, 9 g, 12 g, 16 g, or 18 g total human milk oligosaccharides per day. In some embodiments, the prebiotic mixture is administered in a single dose per day. In various embodiments, the prebiotic mixture is administered in multiple doses per day, e.g., at or at least two, three, four, five, or six or more doses per day.
- a daily amount of human milk oligosaccharides e.g., 2 g, 4.5 g, 6 g, 9 g, 12 g, 16 g, or 18 g total human milk oligosaccharides per day, are administered over two, three, four, five, six, or more than six doses per day to achieve the total amount.
- doses of 9 g of total human milk oligosaccharides are administered twice daily for a total of 18 g/day.
- the at least one Bifidobacterium is administered daily for at least 2, 3, 4, 5, 7, 10, 14, 21, or 28 days, e.g., consecutive days. In some embodiments, the at least one Bifidobacterium is administered in an amount of at least 1 x 10 1 , 5 x 10 1 ,! x 10 2 , 1 x 10 3 , 1 x 10 4 , 1 x 10 5 , 1 x 10 6 , 5 x 10 6 , 1 x 10 7 , 1 x 10 7 , 5 x 10 7 , 1 x 10 8 , or 5 x 10 8 colony forming units (CFU) per day.
- CFU colony forming units
- the at least one Bifidobacterium is administered in an amount of at least 1 x 10 1 , 1 x 10 2 , 1 x 10 3 , 1 x 10 4 , 1 x 10 5 , 1 x 10 6 , 5 x 10 6 , 1 x 10 7 , 5 x 10 7 , l x 10 8 , or 5 x 10 8 colony forming units (CFU) per dose.
- the at least one Bifidobacterium is administered in an amount of between 1 x 10 6 and 1 x 10 12 , 5 x 10 6 and 1 x 10 10 , 1 x 10 7 and 1 x 10 9 , or 1 x 10 7 and 1 x 10 8 CFU per day.
- the Bifidobacterium strain is administered in an amount of, of about, or at least 5 x 10 6 colony forming units (CFU) per dose or per day. In some embodiments, the Bifidobacterium strain is administered in an amount of, of about, or at least 8 x 10 7 colony forming units (CFU) per dose or per day.
- the at least one propionate producing bacterium is administered daily for at least 2, 3, 4, 5, 7, 10, 14, 21, or 28 days, e.g., consecutive days. In some embodiments, the at least one propionate producing bacterium is administered in an amount of at least 1 x 10 1 , 5 x 10 1 ,! x 10 2 , 1 x 10 3 , 1 x 10 4 , 1 x 10 5 ,l x 10 6 , 5 x 10 6 , 1 x 10 7 , 5 x 10 7 , 1 x 10 8 , or 5 x 10 8 colony forming units (CFU) per day.
- CFU colony forming units
- the at least one propionate producing bacterium is administered in an amount of at least 1 x 10 1 , 1 x 10 2 , 1 x 10 3 , 1 x 10 4 , 1 x 10 5 , 1 x 10 6 , 5 x 10 6 , 1 x 10 7 , 5 x 10 7 , 1 x 10 8 , or 5 x 10 8 colony forming units (CFU) per dose.
- the at least one propionate producing bacterium is administered in an amount of between 1 x 10 6 and 1 x 10 12 , 5 x 10 6 and 1 x IO 10 , 1 x 10 7 and 1 x 10 9 , or 1 x 10 7 and 1 x 10 8 CFU per day.
- the at least one propionate producing bacterium is administered in an amount of, of about, or at least 5 x 10 6 colony forming units (CFU) per dose or per day.
- the probiotic strain is administered in an amount of, of about, or at least 8 x 10 7 colony forming units (CFU) per dose or per day.
- the prebiotic mixture is administered at least once within 3 months, 2 months, 1 month, 60 days, 45 days, 30 days, 6 weeks, 5 weeks, 4 weeks, 3 weeks, 28 days, 21 days, 14 days, 10 days, 7 days, 5 days, 3 days, or 1 day prior to administration of the at least one Bifidobacterium.
- the prebiotic mixture is administered at least once daily for at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 21, or 28 consecutive days prior to the at least one Bifidobacterium.
- the prebiotic mixture is administered at least once within 3 months, 2 months, 1 month, 60 days, 45 days, 30 days, 6 weeks, 5 weeks, 4 weeks, 3 weeks, 28 days, 21 days, 14 days, 10 days, 7 days, 5 days, 3 days, or 1 day after administration of the at least one Bifidobacterium.
- the prebiotic mixture is administered at least once daily for at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 21, or 28 consecutive days after the at least one Bifidobacterium.
- the prebiotic mixture is administered prior to and after the at least one Bifidobacterium.
- the prebiotic mixture, the at least one Bifidobacterium, and the at least one propionate producing bacterium are administered together or separately during the same treatment regimen.
- the treatment regimen has separate treatment phases.
- the treatment regimen may include separate treatment phases with different combinations, doses, or timing of doses for some or all of the prebiotic mixture, the at least one Bifidobacterium, and the at least one propionate producing bacterium are administered to the subject.
- all of the prebiotic mixture, the at least one Bifidobacterium, and the at least one propionate producing bacterium are administered during a treatment phase that occurs during the treatment regimen, and, in a different treatment phase, only one or two of the prebiotic mixture, the at least one Bifidobacterium, and the at least one propionate producing bacterium are administered in a different treatment phase that occurs during the same treatment regimen.
- a treatment phase has a duration of at least one, two, three, four, five, six, seven, eight, nine, or ten days, or for at least one, two, three, four, five, or six weeks, or for at least one, two, three, four, five, or six months.
- a treatment phase is or is at least seven days.
- a treatment phase is or is at least fourteen days.
- the treatment regimen includes more than one treatment phase.
- one treatment phase e.g., a first treatment phase
- the prebiotic mixture and the at least one Bifidobacterium strain are administered in one treatment phase, e.g., a first treatment phase.
- the prebiotic mixture and the at least one Bifidobacterium strain are administered within the same composition during the treatment phase, e.g., the first treatment phase.
- the prebiotic mixture and the at least one Bifidobacterium strain are administered as separate compositions during the treatment phase, e.g., the first treatment phase.
- one or both of the at least one Bifidobacterium strain and the prebiotic mixture is administered at least once, at least twice, at least three times, at least once per week, at least twice per week, at least three time per week, every other day, and/or every day during the treatment phase, e.g., the first treatment phase.
- the prebiotic mixture and the at least one Bifidobacterium strain are administered on the same day for at least one, some, or all of the days of the treatment phase, e.g., the first treatment phase.
- the at least one propionate producing strain is also administered during the treatment phase, e.g., the first treatment phase.
- the at least one propionate producing strain is administered at least once, at least twice, at least three times, at least once per week, at least twice per week, at least three time per week, every other day, and/or every day during the treatment phase, e.g., the first treatment phase.
- the at least one propionate producing strain is administered on the same days as the at least one Bifidobacterium strain during the treatment phase, e.g., the first treatment phase.
- the treatment phase e.g., the first treatment phase
- the treatment regimen includes a treatment phase, e.g., a second treatment phase, where the prebiotic mixture and not the at least one Bifidobacterium strain is administered.
- the prebiotic mixture is administered at least once, at least twice, at least three times, at least once per week, at least twice per week, at least three time per week, every other day, and/or every day during the treatment phase, e.g., the second treatment phase.
- the at least one propionate producing strain is also administered during the second treatment phase.
- the at least one propionate producing strain is administered at least once, at least twice, at least three times, at least once per week, at least twice per week, at least three time per week, every other day, and/or every day during the second treatment phase. In certain embodiments, the at least one propionate producing strain is not administered during the second treatment phase.
- the second treatment phase has a duration of at least one, two, three, four, five, six, seven, ten, fourteen days, or at least one, two, three, four, five, or six weeks, or from one day to fourteen days or from three days to seven days.
- the first and second treatment phases occur once during a treatment regimen.
- the first and second treatment phases occur more than once during a treatment regimen, such as cycling or repeating throughout the duration of the treatment regimen.
- there may be a gap or duration e.g., for at least one, two, three, five, seven, ten, or fourteen, days, with no treatments between cycles, e.g., after the end of the second treatment phase and before the beginning of a subsequent first treatment phase.
- the prebiotic mixture and the at least one Bifidobacterium strain are administered together or separately for a period of time, such as in a treatment regimen.
- the administration of the prebiotic mixture e.g., of HMOs, allows for the engraftment and expansion of the Bifidobacterium strain, e.g., B. longum subsp. infantis.
- the Bifidobacterium strain is exogenous to the subject’s microbiome, e.g., intestinal microbiome.
- the Bifidobacterium strain is not present within the subject’s microbiome prior to administration.
- the prebiotic mixture is administered concurrently with and/or subsequently to administration of the at least one Bifidobacterium strain.
- the at least one Bifidobacterium strain is present and/or expands within the subject’s microbiome during a time period in which the prebiotic mixture is administered.
- the presence or amount of the at least one Bifidobacterium strain within the microbiome is reduced when administration of the prebiotic mixture ends, is ceased or is terminated.
- the Bifidobacterium strain is absent and/or undetectable following the termination or end of administration of the prebiotic mixture.
- the presence of the Bifidobacterium strain e.g., B. infantis, is transient and is regulated by administration of the prebiotic mixture.
- At least one Bifidobacterium strain is capable of consuming or metabolizing some or all of the oligosaccharides, e.g., HMOs, of the prebiotic mixture.
- the timing or dosing for administering the at least one Bifidobacterium strain and the prebiotic mixture achieves a growth or expansion of the Bifidobacterium strain in vivo, e.g., within the microbiome of the subject.
- the administered oligosaccharides selectively or exclusively serve as a carbon source for the at least one Bifidobacterium strain, e.g., as opposed to other bacterial strains present in the gut or microbiome.
- the oligosaccharides of the mixture selectively or exclusively serve as an energy source for the at least one Bifidobacterium strain e.g., as opposed to other bacterial strains present in the gut or microbiome.
- subjects are administered (e.g., at least once daily) all of the at least one Bifidobacterium strain, e.g., B. longum subsp. infantis, the at least one propionate producer, and the prebiotic mixture, e.g., of human milk oligosaccharides, during a first or initial treatment phase, e.g., for at least 1, 3, 7, or 14 days, and then are administered the prebiotic mixture alone during a subsequent treatment phase, e.g., such that occurs immediately after the first or initial treatment phase.
- administration of the prebiotic mixture extends the duration of the colonization of the Bifidobacterium strain within the subject’s gut and/or microbiome.
- administering the prebiotic mixture regulates the expansion, level, or amount of the Bifidobacterium strain, e.g., B. longum subsp. infantis, and/or the production or generation of metabolites, e.g., lactate and/or acetate, by the Bifidobacterium strain.
- expansion of the Bifidobacterium strain and/or generation or production of metabolites by the Bifidobacterium strain promotes the engraftment and/or the expansion of the propionate producing strain and/or promotes the production or generation of propionate by the propionate producing strain.
- the Bifidobacterium and propionate producing strains are administered to the subject, and the concurrent or subsequent administration of the prebiotic mixture may be adjusted to provide a therapeutic response, e.g., to promote growth or expansion of beneficial microbiota and/or to promote the generation or production of propionate within the subject’s gut or microbiome.
- the dosage and/or duration of treatment with the prebiotic mixture e.g., of HMOs, can depend on several factors, including severity and responsiveness of the disease, route of administration, time course of treatment (days to months to years), and time to amelioration of the disease.
- the subject is an infant, a child, a juvenile, or an adult. In certain embodiments, the subject is at least 1 month, 3 months, 6 months, 12 months, 18 months, or 24 months of age. In certain embodiments, the subject is at least 1 year, 2 years, 5 years, 10 years, 12 years, 16 years, or at least 18 years of age. In some embodiments, the subject is at least 12 years old. In certain embodiments, the subject is at least 18 years old. In some embodiments, the subject is an adult.
- a method for treating, reducing, ameliorating, or preventing dysbiosis is or includes steps for administering to the subject a prebiotic mixture, such as any described herein e.g., in Section I-C, at least one Bifidobacterium strain, such as a Bifidobacterium strain described herein, e.g., in Section I-B or listed in Table 1, and a propionate producing bacterium, e.g., as described in such as in Section I-A.
- the method treats or prevents one or more diseases or conditions that include or are associated with dysbiosis, e.g., of the intestinal microbiome.
- the microbiome is an intestinal microbiome of a human.
- provided herein are methods for treating, preventing, or ameliorating one or more diseases, disorders, or conditions that are or may be associated with dysbiosis, e.g., of the intestinal microbiome, in a subject in need thereof.
- administration of the at least one strain of Bifidobacterium, e.g., such as any described herein, e.g., in Section I-B, the strain of bacterium capable of producing propionate, such as any of the propionate producing bacterium described herein, e.g., in Section I- A, and the prebiotic mixture, such as any described herein, e.g., in Section I-C, are useful to treat, ameliorate, remedy, or prevent diseases, disorders, or conditions such as obesity, inflammatory bowel disease (IBD), celiac disease, irritable bowel syndrome (IBS), colon cancer, diabetes, liver disorders, cystic fibrosis, and allergies.
- IBD inflammatory bowel disease
- IBS irritable bowel syndrome
- colon cancer diabetes, liver disorders, cystic fibrosis, and allergies.
- the at least one strain of Bifidobacterium e.g., B. longum subsp. infantis
- the strain of bacterium capable of producing propionate e.g., Veillonella sp.
- the prebiotic mixture e.g., of human milk oligosaccharides
- the gastrointestinal condition, disease, or disorder is or includes one or more of a chronic inflammatory disease, an autoimmune disease, an infection, bowel resection, and/or a condition associated with chronic diarrhea.
- the gastrointestinal condition, disease, or disorder is or includes one or more of irritable bowel syndrome (IBS), inflammatory bowel disease (IBD) including Crohn's Disease and colitis, short bowel syndrome (SBS), celiac disease, small intestinal bacterial overgrowth (SIBO), gastroenteritis, leaky gut syndrome, and gastric lymphoma.
- IBS irritable bowel syndrome
- IBD inflammatory bowel disease
- SIBO small intestinal bacterial overgrowth
- gastroenteritis small intestinal bacterial overgrowth
- leaky gut syndrome and gastric lymphoma.
- the gastrointestinal condition, disease, or disorder is associated with a bacterial, viral or parasitic infection or overgrowth.
- the disease or disorder is associated with infection by drug-resistant bacteria, e.g., vancomycin-resistant enterococcus (VRE).
- VRE vancomycin-resistant enterococcus
- administration of the at least one prebiotic mixture, e.g., of human milk oligosaccharides, the strain of bacterium capable of producing propionate, e.g., Veillonella sp., and the at least one probiotic strain, e.g., B. longum subsp. infantis prevents, reduces, or ameliorates one or more symptoms of the gastrointestinal condition.
- the at least one strain of Bifidobacterium e.g., B. longum subsp. infantis, the strain of bacterium capable of producing propionate, e.g., Veillonella sp., and the prebiotic mixture, e.g., of human milk oligosaccharides, are administered to a subject with an immune dysfunction.
- the subject is immunocompromised.
- the administration prevents, reduces, treats, or ameliorates an infection in the immunocompromised subject.
- the administration prevents, reduces, treats, or ameliorates overgrowth or domination of pathogenic bacteria.
- the immunocompromised subject has undergone one or more treatments for cancer.
- the treatments are or include chemotherapy.
- the treatment is or includes an allogenic transplant, e.g, a hematopoietic stem cell transplant or bone marrow transplant.
- the immunocompromised subject is in an ICU, has received an organ transplant, is elderly (e.g., at least 65 or 75 years old) and/or has been on prolonged antibiotic treatment (e.g., for at least 2, 3, 4, 6, 8, 10, or 12 weeks, or at least 1, 2, 3, 6, 12, 18, or 24 months).
- the administration prevents or reduces the probability or likelihood of a systemic infection by, by about, or by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 90%, or 95%, e.g., as compared to a subject administered an alternative treatment and/or not administered the probiotic strains (e.g., the Bifidobacterium and the propionate producing bacterium) and/or the prebiotic mixture.
- the probiotic strains e.g., the Bifidobacterium and the propionate producing bacterium
- the at least one strain of Bifidobacterium e.g., B. longum subsp. infantis, the strain of bacterium capable of producing propionate, e.g., Veillonella sp., and the prebiotic mixture, e.g., of human milk oligosaccharides, are administered to treat or prevent overgrowth or domination of pathogenic bacteria (also referred to herein as gut domination).
- domination of pathogenic bacteria refers to the presence of a species of bacteria (e.g, a pathogenic species), of at least 1%, 5%, 10%, 20%, or 30%, relative to the bacteria present in the subject’s gut or intestinal microbiome.
- overgrowth or domination may be determined by routine techniques in the art, such as including but not limited to PCR or high throughput sequencing.
- the at least one strain of Bifidobacterium e.g., B. longum subsp. infantis, the strain of bacterium capable of producing propionate, e.g., Veillonella sp., and the prebiotic mixture, e.g., of human milk oligosaccharides, are administered to a subject having, suspected of having, or at risk of having dysbiosis, e.g., of the intestinal microbiome.
- the transient presence, engraftment, or expansion of the Bifidobacterium strain e.g., B. longum subsp.
- infantis and the propionate producing strain, e.g., Veilloinella sp. reduces, decreases, or ameliorates the dysbiosis.
- propionate producing strain e.g., Veilloinella sp. reduces, decreases, or ameliorates the dysbiosis.
- particular embodiments contemplate that the presence, engraftment, or expansion of the Bifidobacterium strain, e.g., B. longum subsp.
- infantis, and the propionate producing strain e.g., Veilloinella sp., creates, promotes, or generates an environment and/or one or more conditions that (i) promotes the presence, growth, or expansion of beneficial microbiota; (ii) decreases the presence, growth, or expansion of pathogenic microbiota; (iii) promotes diversity of microbiota present within the microbiome; or (iv) any or all of (i) through (iii).
- administration of the Bifidobacterium strain, e.g., B. longum subsp. infantis, and the propionate producing strain, e.g., Veilloinella sp. reduces the presence or abundance of pathogenic bacteria in the subject’s gut.
- administration of the Bifidobacterium strain, e.g., B. longum subsp. infantis, and the propionate producing strain, e.g., Veilloinella sp. reduces gut domination by pathogenic taxa (e.g., Enterob acteriaceae, Enterococcus, Staphylococcus).
- pathogenic taxa e.g., Enterob acteriaceae, Enterococcus, Staphylococcus
- the growth of the Bifidobacterium strain, e.g., B. longum subsp. infantis, and the propionate producing strain, e.g., Veilloinella sp., within the gut or microbiome reduces the abundance, level, activity, or presence of pathogenic taxa.
- the prebiotic mixture e.g., of human milk oligosaccharides
- the growth of the Bifidobaclerium ivVm e.g., B. longum subsp. infantis
- the prebiotic mixture e.g., of human milk oligosaccharides, the propionate producing strain, e.g., Veilloinella sp., and the Bifidobacterium strain, e.g., B. longum subsp. infantis, are administered to a subject who is at risk of an infection or gut domination, e.g., by pathogenic bacteria.
- the subject has an increased risk of infection or gut domination, e.g., as compared to the general population.
- the subject is immunocompromised, undergoing an extended antibiotic treatment regimen (e.g., lasting at least 2, 3, 4, 5, 6, 8 10, or 12 weeks or 2, 3, 6, 12, 18, or 24 months), is elderly, is hospitalized e.g., in an intensive care unit (ICU), has received an organ transplant, and/or is immunosuppressed.
- an extended antibiotic treatment regimen e.g., lasting at least 2, 3, 4, 5, 6, 8 10, or 12 weeks or 2, 3, 6, 12, 18, or 24 months
- ICU intensive care unit
- the subject will undergo or has received a medical procedure such as a surgery or a chemotherapy that may increase the risk, likelihood, or probability of infection.
- administration of the prebiotic mixture, the propionate producing strain, and the Bifidobacterium strain reduces the risk, likelihood, or probability of infection, e.g, by pathogenic bacteria, is reduced by at least 10%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 90%, 99%, or 99.9% as compared to alternative treatments or no treatments, or as compared to administration of the probiotic strains or prebiotic mixture alone.
- the prebiotic mixture and the probiotic strains are administered at least once at least 1 hour, 2 hours, 3 hours, 4 hours, 6 hours, 8 hours, 12 hours, 18 hours, 24 hours, 1 day, 2 days, 3 days, 4 days, 5 days, 7 days, 10 days, 1 week, 2 weeks, 4 weeks, 6 weeks, one month, or two months prior to the medical procedure, e.g, surgery or chemotherapy.
- the prebiotic mixture and the probiotic strains are administered at least once during the medical procedure, e.g., surgery or chemotherapy.
- the prebiotic mixture and the probiotic strains are administered at least once at least 1 hour, 2 hours, 3 hours, 4 hours, 6 hours, 8 hours, 12 hours, 18 hours, 24 hours, 1 day, 2 days, 3 days, 4 days, 5 days, 7 days, 10 days, 1 week, 2 weeks, 4 weeks, 6 weeks, one month, or two months after to the medical procedure, e.g., surgery or chemotherapy.
- Pathogenic bacteria may include known microbes with pathogenicity for the gastrointestinal tract, e.g., from esophagus down to rectum.
- pathogenic bacteria are or include one or more species, subspecies, or strains of Proteobacteria.
- the pathogenic bacteria may include, but are not limited to strains, species, subspecies, or strains of one or more of Firmicutes, Clostridium, Enter obacteriaceae, Enterococcus, Staphylococcus, Corynebacteria, Salmonella, Shigella, Staphylococcus, Campylobacter (e.g., Campylobacter jejuni), Clostridia, Escherichia coli, Yersinia, Vibrio cholerae, Mycobacterium avium subspecies paratuberculosis, Brachyspira hyodysenteriae, or Law sonia intracellularis .
- administration of the prebiotic mixture and the probiotic strains reduces or decreases the presence, growth, or abundance of pathogenic bacteria within the gut.
- administration of the prebiotic mixture the propionate producing strain, and the Bifidobacterium strain impairs the growth of one or more pathogens.
- pathogens treated by the provided methods include, but are not limited to, Aeromonas hydr ophila, Bacillus, e.g., Bacillus cereus, Bifidobacterium, Bordetella, Borrelia, Brucella, Burkholderia, C.
- E. coli enterotoxigenic Escherichia coli (such as but not limited to LT and/or ST), Escherichia coli 0157:H7, and multi-drug resistant bacteria
- E. coli Francisella, Haemophilus, Helicobacter, e.g., Helicobacter pylori, Klebsiella, e.g., Klebsiellia pneumonia and multi-drug resistant bacteria Klebsiella, Legionella, Leptospira, Listeria, e.g., Lysteria monocytogenes, Morganella, Mycobacterium, Mycoplasma, Neisseria, Orientia, Plesiomonas shigelloides, Antibiotic-resistant Proteobacteria, Proteus, Pseudomonas, Rickettsia, Salmonella, e.g., Salmonella paratyphi, Salmonella spp., and Salmonella typhi, Shigella, e.g
- At least one of the one or more pathogens can be an antibioticresistant bacterium (ARB), e.g., Antibiotic-resistant Proteobacteria, Vancomycin Resistant Enterococcus (VRE), Carbapenem Resistant Enterobacteriaceae (CRE), fluoroquinoloneresistant Enterobacteriaceae, or Extended Spectrum Beta-Lactamase producing Enterobacterales (ESBL-E).
- ARB antibioticresistant bacterium
- VRE Vancomycin Resistant Enterococcus
- CRE Carbapenem Resistant Enterobacteriaceae
- ESBL-E Extended Spectrum Beta-Lactamase producing Enterobacterales
- administration of the prebiotic mixture the propionate producing strain, and the Bifidobacterium strain impairs the growth of of antibiotic-resistant bacterium (ARB), Antibiotic-resistant Proteobacteria, Carbapenem-resistant Enterobacteriaceae (CRE), Extended Spectrum Beta-Lactamase producing Enterobacterales (ESBL-E), fluoroquinolone-resistant Enterobacteriaceae, vancomycin-resistant Enterococci (VRE), multi-drug resistant A. coli, or multi-drug resistant Klebsiella.
- ARB antibiotic-resistant bacterium
- CRE Carbapenem-resistant Enterobacteriaceae
- ESBL-E Extended Spectrum Beta-Lactamase producing Enterobacterales
- VRE vancomycin-resistant Enterococci
- multi-drug resistant A. coli or multi-drug resistant Klebsiella.
- administration of the prebiotic mixture, the propionate producing strain, and the Bifidobacterium strain treat, prevent, or ameliorate an infection or gut domination by one or more of Caproiciproducens, Citrobacter freundii, Citrobacter koseri, Enterobacter aerogenes, Enterobacter cloacae, Enterococcus faecalis, Enterococcus faecium, Escherichia coli, Klebsiella oxytoca, Klebsiella pneumoniae, Morganella morganii, Papillibacter cinnamivorans, Papillibacter, Proteus mirabilis, Serratia marcescens, Sporobacter, Staphylococcus aureus, Staphylococcus epidermidis, Streptococcus anginosus, Streptococcus australis, Streptococcus constellatus, Streptococcus cristat
- administration of the prebiotic mixture, the propionate producing strain, and the Bifidobacterium strain treat, prevent, or ameliorate an infection or gut domination by one or more of Citrobacter freundii, Citrobacter koseri, Enterobacter aerogenes, Enterobacter cloacae, Enterococcus faecalis, Enterococcus faecium, Escherichia coli, Klebsiella oxytoca, Klebsiella pneumoniae, Morganella morganii, Proteus mirabilis, Serratia marcescens, Staphylococcus aureus, Staphylococcus epidermidis, Streptococcus anginosus, Streptococcus australis, Streptococcus constellatus, Streptococcus cristatus, Streptococcus gordonii, Streptococcus infantis, Streptococcus intermedius, Streptoc
- the condition, disease, or disorder is an immune dysfunction that is an autoimmune disorder.
- the autoimmune disorder includes, but is not limited to, acute disseminated encephalomyelitis (ADEM), acute necrotizing hemorrhagic leukoencephalitis, Addison's disease, agammaglobulinemia, alopecia areata, amyloidosis, ankylosing spondylitis, anti-GBM/anti-TBM nephritis, antiphospholipid syndrome (APS), autoimmune angioedema, autoimmune aplastic anemia, autoimmune dysautonomia, autoimmune hemolytic anemia, autoimmune hepatitis, autoimmune hyperlipidemia, autoimmune immunodeficiency, autoimmune inner ear disease (AIED), autoimmune myocarditis, autoimmune oophoritis, autoimmune pancreatitis, autoimmune retinopathy, autoimmune thrombocytopenic purpura (ATP), autoimmune thyroid disease
- ADAM acute disseminated
- the condition, disease, or disorder is a diarrheal disease including, but not limited to, acute bloody diarrhea (e.g., dysentery), acute watery diarrhea (e.g., cholera), checkpoint inhibitor-associated colitis, diarrhea due to food poisoning, persistent diarrhea, and traveler's diarrhea.
- acute bloody diarrhea e.g., dysentery
- acute watery diarrhea e.g., cholera
- checkpoint inhibitor-associated colitis e.g., cholera
- diarrhea due to food poisoning e.g., chronic diarrhea, and traveler's diarrhea.
- administration of the prebiotic mixture, the propionate producing strain, and the Bifidobacterium strain treats or prevents various GI disorders known to result from or be associated or accompanied with dysbiosis of the intestinal microbiome.
- administration of the prebiotic mixture, the propionate producing strain, and the Bifidobacterium strain reduces GI immunoactivation and/or inflammation.
- GI immunoactivation and inflammation may be assessed by known methods that are routine in the art.
- the condition, disease, or disorder is an inflammatory bowel disease (IBD) or related disease including, but not limited to, Behcet's disease, collagenous colitis, Crohn's disease, diversion colitis, fulminant colitis, intermediate colitis, left-sided colitis, lymphocytic colitis, pancolitis, pouchitis, proctosigmoiditis, short bowel syndrome, ulcerative colitis, and ulcerative proctitis.
- IBD inflammatory bowel disease
- Behcet's disease collagenous colitis, Crohn's disease, diversion colitis, fulminant colitis, intermediate colitis, left-sided colitis, lymphocytic colitis, pancolitis, pouchitis, proctosigmoiditis, short bowel syndrome, ulcerative colitis, and ulcerative proctitis.
- administration of the prebiotic mixture, the propionate producing strain, and the Bifidobacterium strain treats or prevents various bloodstream infections (BSI).
- BAI bloodstream infections
- administration of the probiotic strains and the prebiotic mixture treats or prevents catheter or intravascular-line infections (e.g., central-line infections).
- administration of the probiotic strains and the prebiotic mixture treats or prevents chronic inflammatory diseases.
- administration of the prebiotic mixture, the propionate producing strain, and the Bifidobacterium strain treats or prevents meningitis; pneumonia, e.g., ventilator-associated pneumonia; skin and soft tissue infections; surgical- site infections; urinary tract infections (e.g., antibiotic-resistant urinary tract infections and catheter-associated urinary tract infections); wound infections; and/or antibiotic-resistant infections and antibiotic-sensitive infections.
- pneumonia e.g., ventilator-associated pneumonia
- skin and soft tissue infections e.g., surgical- site infections
- urinary tract infections e.g., antibiotic-resistant urinary tract infections and catheter-associated urinary tract infections
- wound infections e.g., antibiotic-resistant infections and antibiotic-sensitive infections.
- administration of the prebiotic mixture, the propionate producing strain, and the Bifidobacterium strain treats or prevents diseases or disorders relating to the "gut-brain axis", including neurodegenerative, neurodevelopmental, and neurocognitive disorders, such as anorexia, anxiety, autism-spectrum disorder, depression, Parkinson's, and Schizophrenia.
- administration of the probiotic strains and the prebiotic mixture reduces one or more symptoms associated with anorexia, anxiety, autism-spectrum disorder, depression, Parkinson's, and/or Schizophrenia.
- administration of the prebiotic mixture, the propionate producing strain, and the Bifidobacterium strain treats or prevents a side effect of an anticancer therapy and/or increases efficacy of an anti -cancer therapeutic agent and/or anticancer therapy.
- the anti -cancer therapy is surgery, radiation therapy, chemotherapy (including hormonal therapy) and/or targeted therapy (including an immunotherapy).
- Illustrative chemotherapeutics agents are provided elsewhere herein.
- the immunotherapy binds to and/or recognizes a tumor-cell antigen and/or a cancer-cell antigen, e.g., CTLA-4, PD-1, PD-L1, or PD-L2.
- the immunotherapy comprises administration of Keytruda (Pembrolizumab), Opdivo (Nivolumab), Yervoy (Ipilimumab), Tecentriq (atezolizumab), Bavencio (avelumab), and Imfinzi (durvalumab).
- the subject is refractory and/or non-responsive to an anti-cancer therapy.
- the prebiotic mixture, the propionate producing strain, and the Bifidobacterium strain treats a subject that presents a non-curative response, a limited response, or no response to the anti-cancer therapy, or even progress, after 12 weeks or so of receiving the anti -cancer therapy.
- the provided probiotic strains and prebiotic mixture of the present invention can rescue subjects that are refractory and/or non-responsive to the anti-cancer therapy.
- the subject is refractory and/or non-responsive to a treatment directed to a checkpoint molecule, e.g., CTLA-4, PD-1, PD-L1, and/or PD-L2.
- a treatment directed to a checkpoint molecule comprises administration of Keytruda (Pembrolizumab), Opdivo (Nivolumab), Yervoy (Ipilimumab), Tecentriq (atezolizumab), Bavencio (avelumab), or Imfinzi (durvalumab).
- the prebiotic mixture c.g, of human milk oligosaccharides, the propionate producing strain, e.g., Veilloinella sp., and the at least one Bifidobacterium strain, e.g, B. longum subsp. infantis, are administered to an immunocompromised subject.
- the administration prevents, reduces, treats, or ameliorates an infection in the immunocompromised subject.
- the administration prevents, reduces, treats, or ameliorates overgrowth or domination of pathogenic bacteria.
- the immunocompromised subject has undergone one or more treatments for cancer.
- the treatments are or include chemotherapy.
- the treatment is or includes an allogenic transplant, e.g., a hematopoietic stem cell transplant or bone marrow transplant.
- the administration prevents or reduces the probability or likelihood of a systemic infection by, by about, or by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 90%, or 95%, e.g., as compared to an alternative treatment or treatment with either the probiotic strains or prebiotic mixture alone.
- the prebiotic mixture e.g., of human milk oligosaccharides, the propionate producing strain, e.g., Veilloinella sp., and the at least one Bifidobacterium strain, e.g, B. longum subsp. infantis, are administered to a subject who has or is at risk of sepsis.
- the probability or likelihood of sepsis is reduced or decreased by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 99%, e.g, as compared to a subject (e.g., who has or is at risk for sepsis) not administered the prebiotic mixture or the probiotics.
- the administration of the prebiotic mixture and the probiotics improves or increases the survival of the subject over 6 months, 12 months, 18 months, 1 year, 2 years, 5 years, 10 years, and/or 20 years or more by, by about, or by at least 5%, 10%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 90%, 95%, 100%, or 1-fold, 2-fold, 3-fold, 4-fold, or 5-fold greater than in subjects (e.g., who have or are at risk for sepsis) not administered the prebiotic mixture and the probiotic strains.
- administration of the prebiotic mixture, the propionate producing strain, and the Bifidobacterium strain prevents, reduces, decreases, remedies, or ameliorates one or more symptoms associated with a gastrointestinal condition, disease, or disorder.
- the one or more symptoms associated with gastrointestinal condition, disease, or disorder may include, but are not limited to, diarrhea, fever, fatigue, abdominal pain and cramping, blood in stool, mouth sores, weight loss, fistula, inflammation (of skin, eyes, or joints), inflamed liver or bile ducts, delayed growth (in children).
- administration of the prebiotic mixture, the propionate producing strain, and the Bifidobacterium strain reduces the risk or probability for the subject of experiencing one or more symptoms associated with the gastrointestinal condition, disease, or disorder by, by about, or by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 90%, or 95%, e.g., as compared to a subject not administered the probiotic strains and/or the prebiotic mixture.
- administration of the prebiotic mixture and the probiotic strains increases probability or likelihood for remission by, by about, or by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 90%, 100%, or 1-fold, 2-fold, 5-fold, 10- fold, 20-fold, 50-fold, or 100-fold e.g., as compared to a subject not administered the probiotic strains and/or the prebiotic mixture.
- administration of the prebiotic mixture, the propionate producing strain, and the Bifidobacterium strain increases probability or likelihood for remission within 12 weeks, 10 weeks, 8 weeks, 6 weeks, 4 weeks, or less than 4 weeks, e.g., from the initiation or termination of the administration.
- the prebiotic mixture e.g., of human milk oligosaccharides, the propionate producing strain, e.g., Veilloinella sp., and the at least one Bifidobacterium strain, e.g, B. longum subsp. infantis, are administered to a subject to treat, ameliorate, remedy, or prevent a chronic inflammatory disease, an autoimmune disease, an infection, bowel resection, and/or a condition associated with chronic diarrhea.
- the prebiotic mixture e.g., of human milk oligosaccharides
- the propionate producing strain e.g., Veilloinella sp.
- the at least one Bifidobacterium strain e.g, B. longum subsp. infantis
- the pathology is selected from the group consisting of: irritable bowel syndrome (IBS), inflammatory bowel disease (IBD), short bowel syndrome (SBS), celiac disease, small intestinal bacterial overgrowth (SIBO), gastroenteritis, leaky gut syndrome, and gastric lymphoma.
- IBS irritable bowel syndrome
- IBD inflammatory bowel disease
- SBS short bowel syndrome
- SIBO small intestinal bacterial overgrowth
- gastroenteritis small intestinal bacterial overgrowth
- leaky gut syndrome and gastric lymphoma
- gastric lymphoma irritable bowel syndrome
- the disease or disorder is associated with a bacterial, viral, or parasitic infection or overgrowth, e.g., by drug-resistant bacteria.
- administration of the prebiotic mixture the propionate producing strain, and the Bifidobacterium strain increases probability or likelihood for cure or remission of the chronic inflammatory disease, autoimmune disease, infection, bowel resection, and/or chronic diarrhea for by, by about, or by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 90%, 100%, or 1-fold, 2-fold, 5-fold, 10-fold, 20-fold, 50-fold, or 100-fold e.g, as compared to a subject not administered the probiotic strains and/or the prebiotic mixture.
- administration of the prebiotic mixture the propionate producing strain, and the Bifidobacterium strain increases probability or likelihood for the cure or remission within 12 weeks, 10 weeks, 8 weeks, 6 weeks, 4 weeks, or less than 4 weeks, e.g., from the initiation or termination of the administration.
- the prebiotic mixture, the propionate producing strain, and the Bifidobacterium strain are administered to a subject to treat, prevent, or ameliorate an allergy.
- the allergy is a food allergy.
- the food allergy is or includes a chronic or acute immunological hypersensitivity reaction (e.g., a type I hypersensitivity reaction) elicited in a mammal in response to an ingested material or food antigen (also referred to in the art as a “food allergen”). Identification and diagnosis of food allergy is routine among persons of ordinary skill in the art.
- Food allergies may include, but are not limited to, allergies to nuts, peanuts, shellfish, fish, milk, eggs, wheat, or soybeans.
- the prebiotic mixture the propionate producing strain, and the Bifidobacterium strain are administered to treat or ameliorate an allergy, e.g., a food allergy.
- the prebiotic mixture, the propionate producing strain, and the Bifidobacterium strain reduce or decrease the severity of the allergic response to the allergen, e.g., as compared to the allergic response prior to any treatment with the probiotic strains and prebiotic mixture.
- the prebiotic mixture the propionate producing strain, and the Bifidobacterium strain attenuates or reduces the severity or intensity of one or more symptoms or clinical manifestations of the allergy, e.g., food allergy, to subsequent exposures to the allergen, e.g., as compared to symptoms or clinical manifestations observed prior to treatment with the probiotic strains and prebiotic mixture.
- the symptoms or clinical manifestations of the allergy may include, but are not limited to rash, eczema, atopic dermatitis, hives, urticaria, angiodema, asthma, rhinitis, wheezing, sneezing, dyspnea, swelling of the airways, shortness of breath, other respiratory symptoms, abdominal pain, cramping, nausea, vomiting, diarrhea, melena, tachycardia, hypotension, syncope, seizures, and anaphylactic shock.
- the prebiotic mixture, the propionate producing strain, and the Bifidobacterium strain are administered to a subject, e.g., a subject at risk of having or developing an allergy, to prevent or reduce or decrease the probability or likelihood experiencing an allergic response.
- administration of the probiotic strains and prebiotic mixture reduce the likelihood or probability of having an allergic response within the next month, 3 months, 6 months, 12 months, 18 months, year, 2 years, 3 years, 5 years, 10 years, or 20 years.
- the probability or likelihood of developing the allergy is reduced by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 90%, or 99% as compared to a subject with a similar risk profile who is not administered the probiotic strains and the prebiotic mixture.
- administration of the probiotic strains and prebiotic mixture reduces the severity of one or more symptoms or clinical manifestations of an allergic response following exposure to the allergen over the next month, 3 months, 6 months, 12 months, 18 months, year, 2 years, 3 years, 5 years, 10 years, or 20 years, e.g., as compared to exposure of the allergen to a subject with the same or similar allergy who was not administered the probiotic strains and the prebiotic mixture.
- the prebiotic mixture e.g., of human milk oligosaccharides, the propionate producing strain, e.g., Veilloinella sp., and the at least one Bifidobacterium strain, e.g., B. longum subsp. infantis, are administered to a subject to treat, ameliorate, remedy, or prevent pouchitis.
- pouchitis is inflammation that occurs in the lining of a pouch created during surgery to treat ulcerative colitis or certain other diseases.
- the surgery is or includes removal of a diseased colon or portion thereof.
- the surgery is a J pouch surgery (ileoanal anastomosis — IPAA).
- the prebiotic mixture e.g., of human milk oligosaccharides, the propionate producing strain, e.g., Veilloinella sp., and the at least one Bifidobacterium strain, e.g., B. longum subsp. infantis, are administered to a subject to treat, ameliorate, remedy, or prevent pouchitis in a subject in need thereof, e.g., a subject who has undergone an IPAA surgery.
- administration of the prebiotic mixture and the probiotic strains prevents, reduces, decreases, remedies, or ameliorates one or more symptoms associated with pouchitis.
- the one or more symptoms associated with pouchitis may include, but are not limited to, increased stool frequency, tenesmus, straining during defecation, blood in the stool, incontinence, seepage of waste matter during sleep, abdominal cramps, pelvic or abdominal discomfort, or tail bone pain.
- symptoms associated with more severe pouchitis include, but are not limited to, fever, dehydration, malnutrition, fatigue, iron-deficiency anemia, or joint pain.
- administration of the prebiotic mixture and the at least one probiotic strain reduces the risk or probability for the subject of experiencing pouchitis by, by about, or by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 90%, or 95%, e.g., as compared to a subject not administered the probiotic strains and/or the prebiotic mixture.
- the prebiotic mixture e.g., of human milk oligosaccharides, the propionate producing strain, e.g., Veilloinella sp., and the at least one Bifidobacterium strain, e.g., B. longum subsp. infantis, are administered to a subject to treat, ameliorate, remedy, or prevent a chronic inflammatory disease, an autoimmune disease, an infection, bowel resection, and/or a condition associated with chronic diarrhea.
- the prebiotic mixture e.g., of human milk oligosaccharides
- the propionate producing strain e.g., Veilloinella sp.
- the at least one Bifidobacterium strain e.g., B. longum subsp. infantis
- Such pathology includes, but is not limited to: irritable bowel syndrome (IBS), inflammatory bowel disease (IBD), short bowel syndrome (SBS), celiac disease, small intestinal bacterial overgrowth (SIBO), gastroenteritis, leaky gut syndrome, and gastric lymphoma.
- IBS irritable bowel syndrome
- IBD inflammatory bowel disease
- SBS short bowel syndrome
- SIBO small intestinal bacterial overgrowth
- gastroenteritis small intestinal bacterial overgrowth
- leaky gut syndrome and gastric lymphoma.
- the disease or disorder is associated with a bacterial, viral, or parasitic infection or overgrowth, e.g., by drug-resistant bacteria.
- administration of the prebiotic mixture and the probiotic strains increases probability or likelihood for cure or remission of the chronic inflammatory disease, autoimmune disease, infection, bowel resection, and/or chronic diarrhea for by, by about, or by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 90%, 100%, or 1-fold, 2-fold, 5-fold, 10-fold, 20-fold, 50-fold, or 100-fold e.g., as compared to a subject not administered the probiotic strains and prebiotic mixture and/or a subject administered an alternative therapy.
- administration of the prebiotic mixture and the probiotic strains increases probability or likelihood for the cure or remission within 12 weeks, 10 weeks, 8 weeks, 6 weeks, 4 weeks, or less than 4 weeks, e.g., from the initiation or termination of the administration e.g., as compared to a subject not administered the probiotic strains and prebiotic mixture and/or a subject administered an alternative therapy.
- the prebiotic mixture e.g., of human milk oligosaccharides, the propionate producing strain, e.g., Veilloinella sp., the Bifidobacterium strain, e.g., B. longum subsp. infantis
- the prebiotic mixture e.g., of human milk oligosaccharides, the propionate producing strain, e.g., Veilloinella sp., the Bifidobacterium strain, e.g., B. longum subsp. infantis
- pouchitis is inflammation that occurs in the lining of a pouch created during surgery to treat ulcerative colitis or certain other diseases.
- the surgery is or includes removal of a diseased colon or portion thereof.
- the surgery is a J pouch surgery (ileoanal anastomosis — IPAA).
- administration of the prebiotic mixture, the Bifidobacterium strain, and the propionate producing strain reduces the risk or probability for the subject of experiencing pouchitis by, by about, or by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 90%, or 95%, e.g, as compared to a subject not administered the probiotic strains and/or the prebiotic mixture.
- the first dose of the prebiotic mixture or the at least one probiotic strain is administered within 12 weeks, 8 weeks, 6 weeks, 4 weeks, 3 weeks, 2 weeks, 14 days, 12 days 10 days, 7 days, 5 days, 4 days, 3 days, 2 days, or 1 day prior to receiving the allogenic stem cell transplant.
- provided herein are methods for treating, preventing, or ameliorating GVHD in a subject in need thereof. In certain embodiments, provided herein are methods for treating, preventing, or ameliorating a condition or disease associated or accompanied with GVHD in a subject in need thereof. In certain embodiments, provided herein are methods for treating, preventing, reducing, decreasing, or ameliorating the severity or presence of one or more symptoms associated with GVHD or a disease or condition associated or accompanied with GVHD in a subject in need thereof.
- administration improves or increases the survival of the subject over 6 months, 12 months, 18 months, 1 year, 2 years, 5 years, 10 years, and/or 20 years or more by, by about, or by at least 5%, 10%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 90%, 95%, 100%, or 1-fold, 2-fold, 3-fold, 4-fold, or 5-fold greater than in subjects (e.g., subjects who received an allogenic transplant, e.g., BMT or HSCT) not administered the prebiotic mixture and the probiotic strains.
- an allogenic transplant e.g., BMT or HSCT
- the Bifidobacterium e.g., B. longum subsp. infantis
- the prebiotic mixture e.g., of human milk oligosaccharides
- the at least one propionate producing bacterium e.g., Veillonella sp.
- the symptoms are or include symptoms associated with hematopoietic or bone marrow syndrome and/or damage, gastrointestinal syndrome and/or damage, and/or cardiovascular/central nervous system (CNS) syndrome and/or damage.
- CNS cardiovascular/central nervous system
- the subject has been exposed to a high dose of ionizing radiation.
- ionizing radiation doses of 0.3 Gray (Gy) or 30 rads may be sufficient to cause or induce symptoms associated with ARS, and doses of at least 0.7 Gray (Gy) or 70 rads may generally be considered as sufficient to cause or induce ARS.
- the dose is at least 0.3 Gy, 0.5 Gy, 0.7 Gy, 1.0 Gy, 2.0 Gy, 3.0 Gy, 4.0 Gy, 5.0 Gy, 6.0 Gy, 7.0 Gy, 8.0 Gy, 9.0 Gy, 10 Gy, 15 Gy, 20 Gy, 25 Gy, 30 Gy, 40 Gy, or at least 50 Gy.
- the administration improves survival in subjects that have or are suspected of having ARS and/or that have been exposed or are suspected of having been exposed to a high dose of ionizing radiation.
- the subjects’ survival rate is about or at least 50%, 60%, 75%, 80%, 90%, or at least 95% or more after about, at least, or at least about two weeks, four weeks, six weeks, eight weeks, ten weeks, twelve weeks, two months, three months, four months, six months, nine months, twelve months, sixteen months, eighteen months, one year, two years, or three years from the exposure to the dose of ionizing radiation and/or the initial onset of one or more ARS symptoms.
- the provided at least one Bifidobacteria e.g., B. longum subsp. infantis, the provided propionate producing bacterium, e.g., Veillonella sp., and the provided prebiotic mixture, e.g, of human milk oligosaccharides, are formulated together or separately, e.g, for administering to a human subject.
- the at least one Bifidobacteria, the provided propionate producing bacterium, and the provided prebiotic mixture are formulated together in the same pharmaceutical composition.
- the at least one Bifidobacterium and the prebiotic mixture are formulated together into the same pharmaceutical composition.
- the at least one Bifidobacterium and the at least one propionate producing bacterium are formulated together into the same pharmaceutical composition.
- the at least one Bifidobacteria, the provided propionate producing bacterium, and the provided prebiotic mixture are formulated separately into separate pharmaceutical compositions.
- the probiotic strains e.g., the provided Bifidobacterium and/or the provided propionate producing bacterium
- prebiotic mixture described herein may be formulated into pharmaceutical compositions in any suitable dosage form (e.g., liquids, capsules, sachet, hard capsules, soft capsules, tablets, enteric coated tablets, suspension powders, granules, or matrix sustained release formations for oral administration) and for any suitable type of administration (e.g., oral, topical, injectable, immediate-release, pulsatile-release, delay ed- release, or sustained release).
- any suitable dosage form e.g., liquids, capsules, sachet, hard capsules, soft capsules, tablets, enteric coated tablets, suspension powders, granules, or matrix sustained release formations for oral administration
- suitable type of administration e.g., oral, topical, injectable, immediate-release, pulsatile-release, delay ed- release, or sustained release.
- the dosing can be higher; for example, 100 mg to 20 g, 100 mg to 30 g, 500 mg to 15 g, 1 g to 10 g, or 2.5 g to 7.5 g per dose.
- the dosing can be reduced; for example, in certain embodiments, to 20 mg to 10 g per dose, 100 mg to 7.5 g per dose, 500 mg to 2.5 g per dose, 750 mg to 1.5 g per dose, 20 mg to 20 g per dose, 100 mg to 10 g per dose, 500 mg to 7.5 g per dose, or 750 mg to 5 g per dose.
- the prebiotic mixture is administered to the subject in an amount of or about 5 g per day.
- a dose of the prebiotic mixture is administered at least once per month, once per week, or once per day. In some embodiments, a dose of the prebiotic mixture is administered at least once, twice, three times, four times, five times, six times, eight times, ten times, or twelve times daily.
- the pharmaceutical compositions containing the provided at least one Bifidobacterium, the at least one propionate producing bacterium, and the prebiotic mixture may be administered orally and formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions, etc.
- Pharmacological compositions for oral use can be made using a solid excipient, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries if desired, to obtain tablets or dragee cores.
- Suitable excipients include, but are not limited to, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose compositions such as maize starch, wheat starch, rice starch, potato starch, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carbomethylcellulose; and/or physiologically acceptable polymers such as) polyethylene glycol (PEG).
- Fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol
- cellulose compositions such as maize starch, wheat starch, rice starch, potato starch, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carbomethylcellulose
- physiologically acceptable polymers such as) polyethylene glycol (PEG).
- Disintegrating agents may also be added, such as cross-linked agar, alginic acid or a salt thereof such as sodium alginate.
- Tablets or capsules can be prepared by conventional means with pharmaceutically acceptable excipients such as binding agents (e.g, pregelatinized maize starch, hydroxypropyl methylcellulose, carboxymethyl cellulose, polyethylene glycol, sucrose, glucose, sorbitol, starch, gum, and tragacanth); fillers (e.g, lactose, microcrystalline cellulose, or calcium hydrogen phosphate); lubricants (e.g., calcium, aluminum, zinc, stearic acid, polyethylene glycol, sodium lauryl sulfate, starch, sodium benzoate, magnesium stearate, talc, or silica); disintegrants (e.g., starch, potato starch, sodium starch glycolate, sugars, cellulose derivatives, silica powders); or wetting agents (e.g., sodium lauryl sulphate).
- binding agents e.g, pregelatinized maize starch, hydroxypropyl methylcellulose, carboxymethyl cellulose, polyethylene
- the at least one Bifidobacterium, the at least one propionate producing bacterium, and/or the prebiotic mixture are enterically coated, such as in order to remain viable during transit through the stomach, reduce contact with bile acids in the small intestine, or for release into the gut or a particular region of the gut, for example, the large intestine.
- the typical pH profile from the stomach to the colon is about 1-4 (stomach), 5.5-6 (duodenum), 7.3-8.0 (ileum), and 5.5-6.5 (colon).
- the pH profile may be modified.
- the coating is degraded in specific pH environments in order to specify the site of release.
- at least two coatings are used.
- the outside coating and the inside coating are degraded at different pH levels.
- a composition suitable for administration to pediatric subjects may also be suitable for administration to adults.
- the pharmaceutical composition that is suitable for administration to pediatric subjects may include a solution, syrup, suspension, elixir, powder for reconstitution as suspension or solution, dispersible/effervescent tablet, chewable tablet, gummy candy, lollipop, freezer pop, troche, chewing gum, oral thin strip, orally disintegrating tablet, sachet, soft gelatin capsule, sprinkle oral powder, or granules.
- the composition is a gummy candy, which is made from a gelatin base, giving the candy elasticity, desired chewy consistency, and longer shelf-life.
- the gummy candy may also comprise sweeteners or flavors.
- the at least one Bifidobacterium, the at least one propionate producing bacterium, and the prebiotic mixture may, together or separately, be orally administered, such as with an inert diluent or an assimilable edible carrier.
- the pharmaceutical composition may also be enclosed in a hard or soft-shell gelatin capsule, a hydroxypropylmethyl cellulose (HPMC) capsule, compressed into tablets, or incorporated directly into the subject's diet.
- HPMC hydroxypropylmethyl cellulose
- the composition containing the at least one Bifidobacterium, the at least one propionate producing bacterium, and/or the prebiotic mixture may be a nutritional or a comestible product, e.g., a food product or nutritional composition.
- the composition is a nutritional composition such as food product.
- the probiotic strain of the invention e.g., B. longum subsp. infantis strain
- the food product is a beverage.
- the beverage is a fruit juice-based beverage or a beverage containing plant or herbal extracts.
- the food product or nutritional composition is a jelly or a pudding.
- Other food products suitable for administration of the probiotic strain and prebiotic mixtures provided herein are known, such as those described in U.S. Application Nos. 2015/0359894 and 2015/0238545.
- the pharmaceutical composition of the invention is injected into, sprayed onto, or sprinkled onto a food product, such as bread, yogurt, or cheese.
- the composition e.g, pharmaceutical composition, that includes the at least one Bifidobacterium, the at least one propionate producing bacterium, and/or the prebiotic mixture is formulated for intraintestinal administration, intrajejunal administration, intraduodenal administration, intraileal administration, gastric shunt administration, or intracolic administration, via nanoparticles, nanocapsules, microcapsules, or microtablets, which are enterically coated or uncoated.
- the compositions may also be formulated in rectal compositions such as suppositories or retention enemas, using, e.g., conventional suppository bases such as cocoa butter or other glycerides.
- the compositions may be suspensions, solutions, or emulsions in oily or aqueous vehicles, and may contain suspending, stabilizing and/or dispersing agents.
- compositions containing the at least one Bifidobacterium, the at least one propionate producing bacterium, and/or the prebiotic mixture in single dosage forms may be in a liquid or a solid form. Single dosage forms may be administered directly to a subject without modification or may be diluted or reconstituted prior to administration. In certain embodiments, a single dosage form may be administered in bolus form, e.g., single injection, single oral dose, including an oral dose that comprises multiple tablets, capsule, pills, etc. In alternate embodiments, a single dosage form may be administered over a period of time, e.g., by infusion.
- Single dosage forms of the pharmaceutical composition containing the at least one Bifidobacterium, the at least one propionate producing bacterium, and/or the prebiotic mixture may be prepared by portioning the pharmaceutical composition into smaller aliquots, single dose containers, single dose liquid forms, or single dose solid forms, such as tablets, granulates, nanoparticles, nanocapsules, microcapsules, microtablets, pellets, or powders, which may be enterically coated or uncoated.
- a single dose in a solid form may be reconstituted by adding liquid, typically sterile water or saline solution, prior to administration to a subject.
- polymers used in sustained release formulations include, but are not limited to, poly((2 -hydroxy ethyl methacrylate), poly(methyl methacrylate), poly(acrylic acid), poly(ethylene-co-vinyl acetate), poly(methacrylic acid), polyglycolides (PLG), polyanhydrides, poly(N-vinyl pyrrolidone), poly(vinyl alcohol), poly(ethylene glycol), polylactides (PLA), poly(lactide-co-glycolides) (PLGA), and polyorthoesters.
- the polymer used in a sustained release formulation may be inert, free of leachable impurities, stable on storage, sterile, and biodegradable.
- a controlled or sustained release system can be placed in proximity of the prophylactic or therapeutic target, thus requiring only a fraction of the systemic dose. Any suitable technique known to one of skill in the art may be used.
- Dosage regimens of the at least one Bifidobacterium, the at least one propionate producing bacterium, and/or the prebiotic mixture may be adjusted to provide a therapeutic response, e.g., to improve or maintain propionate production. Dosing can depend on several factors, including severity and responsiveness of the disease, route of administration, time course of treatment (days to months to years), and time to amelioration of the disease. For example, a single bolus of one or both of the mixture and the probiotic strain may be administered at one time, several divided doses may be administered over a predetermined period of time, or the dose may be reduced or increased as indicated by the therapeutic situation.
- ingredients are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water-free concentrate in a hermetically sealed container such as an ampoule or sachet indicating the quantity of active agent.
- one or more of the prophylactic or therapeutic agents or pharmaceutical compositions is supplied as a dry sterile lyophilized powder in a hermetically sealed container stored between 2° C. and 8° C. and administered within 1 hour, within 3 hours, within 5 hours, within 6 hours, within 12 hours, within 24 hours, within 48 hours, within 72 hours, or within one week after being reconstituted.
- Cryoprotectants can be included for a lyophilized dosage form, principally 0- 10% sucrose (optimally 0.5- 1.0%).
- Other suitable cryoprotectants include trehalose and lactose.
- suitable bulking agents include poly dextrose, dextrins (e.g., maltodextrin (e.g., a native maltodextrin or a resistant maltodextrin)), inulin, P-glucan, resistant starches (e.g., resistant maltodextrin), hydrocolloids (e.g., one or more of gum Arabic, pectin, guar gum, alginate, carrageenan, xanthan gum and cellulose gum), corn syrup solids and the like and polysorbate 80.-. Additional surfactants include but are not limited to polysorbate 20 and BRIJ surfactants.
- the pharmaceutical composition may be prepared as an injectable solution and can further comprise an agent useful as an adjuvant, such as those used to increase absorption or dispersion, e.g., hyaluronidase.
- the pharmaceutical compositions e.g., containing one or both of the probiotic strain and prebiotic mixtures are administered with food.
- the pharmaceutical composition is administered before or after eating food.
- the pharmaceutical compositions may be administered in combination with one or more dietary modifications, e.g., low-protein diet and amino acid supplementation.
- the dosage of the pharmaceutical compositions and the frequency of administration may be selected based on the severity of the symptoms and the progression of the disorder. The appropriate therapeutically effective dose and/or frequency of administration can be selected by a treating clinician.
- composition means, for example, a mixture or formulation containing a specified amount, e.g., a therapeutically effective amount, of an active ingredient such as a human milk fraction, in a pharmaceutically acceptable carrier to be administered to a mammal, e.g., a human.
- the term "pharmaceutically acceptable” refers to those compounds, materials, compositions and/or dosage forms, which are, within the scope of sound medical judgment, suitable for contact with the tissues of mammals, especially humans, without excessive toxicity, irritation, allergic response, and other problem complications commensurate with a reasonable benefit/risk ratio. Such reasonable benefit/risk ratios may be determined by one of skill as a matter of routine.
- human milk oligosaccharide(s) (also referred to herein as “HMO(s)”) is meant a family of structurally diverse unconjugated glycans that are found in human breast milk.
- human milk oligosaccharides include oligosaccharides found in human milk that contain lactose at the reducing end and, typically, fucose, sialic acid or N- acetylglucosamine at the non-reducing end (Morrow et al., J. Nutri. 2005 135: 1304-1307).
- human milk oligosaccharides also encompass 3'-sialyllactose (3'- SL) and 6'-sialyllactose (6'-SL) oligosaccharides that are found in human milk.
- 3'- SL 3'-sialyllactose
- 6'-SL 6'-sialyllactose
- a number of human milk oligosaccharides e.g., “at least 5 human milk oligosaccharides,” refers to the number of unique species of human milk oligosaccharides, e.g., human milk oligosaccharides having different chemical structures or formulas.
- compositions containing particular recited components refers to compositions containing particular recited components while excluding other major bioactive factors.
- the term "dysbiosis” as used herein refers to a state of the microbiota of the gut or other body area in a subject, in which the normal diversity and/or function of the microbial populations is disrupted. This unhealthy state can be due to a decrease in diversity, the overgrowth of one or more pathogens or pathobionts, symbiotic organisms able to cause disease only when certain genetic and/or environmental conditions are present in a subject, or the shift to an ecological microbial network that no longer provides an essential function to the host subject, and therefore no longer promotes health.
- essential functions may include enhancement of the gut mucosal barrier, direct or indirect reduction and elimination of invading pathogens, enhancement of the absorption of specific substances, and suppression of GI inflammation.
- gut microbiome and “intestinal microbiome” are used interchangeably unless otherwise noted.
- Results are displayed in FIGS. 2A-2C.
- subjects from Cohort 1 antibiotics alone
- the antibiotic dosing period Days 1-5)
- low levels of B. longum subsp. infantis were detected in the DNA from stool of subjects in both cohorts that received B. longum subsp. infantis.
- These levels were consistent with a “pass through” reflecting the daily consumption of the bacteria, as B. longum subsp. infantis is sensitive to both antibiotics used in this study and was unlikely to remain viable in the gut.
- Sequence analyses followed an established pipeline (Diversigen Inc.). Briefly, sequences were aligned to a curated database containing all representative genomes in RefSeq for bacteria with additional manually curated strains. Alignments were made at 97% identity against all reference genomes. Every input sequence was compared to every reference sequence in the Diversigen Venti database using fully gapped alignment with BURST 123. Ties were broken by minimizing the overall number of unique Operational Taxonomic Units (OTUs). For taxonomy assignment, each input sequence was assigned the lowest common ancestor that was consistent across at least 80% of all reference sequences tied for best hit. Samples with fewer than 10,000 sequences were discarded.
- OFT Operational Taxonomic Unit
- an OTU table was calculated with the read counts per genome per sample.
- Alpha diversity metrics observed reads and Shannon entropy
- Bray-Curtis dissimilarity was calculated on the same filtered data, aggregated at the genus taxonomic level and rarefied to 76,000 reads using the R package phyloseq (vl.41.0124).
- the filtered OTU table aggregated at the Family taxonomic level was used to calculate the top Families among cohorts. All taxa >1% abundance in any cohort were kept and all taxa ⁇ 1% among taxa were grouped into an “Other” category.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Pharmacology & Pharmacy (AREA)
- Veterinary Medicine (AREA)
- Chemical & Material Sciences (AREA)
- Public Health (AREA)
- General Health & Medical Sciences (AREA)
- Medicinal Chemistry (AREA)
- Animal Behavior & Ethology (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- General Chemical & Material Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Organic Chemistry (AREA)
- Engineering & Computer Science (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Mycology (AREA)
- Diabetes (AREA)
- Microbiology (AREA)
- Molecular Biology (AREA)
- Epidemiology (AREA)
- Immunology (AREA)
- Obesity (AREA)
- Hematology (AREA)
- Endocrinology (AREA)
- Pain & Pain Management (AREA)
- Rheumatology (AREA)
- Emergency Medicine (AREA)
- Child & Adolescent Psychology (AREA)
- Zoology (AREA)
- Medicines Containing Material From Animals Or Micro-Organisms (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
Provided herein are compositions, methods, strategies, kits, and articles of manufacture that are useful, inter alia, in the treatment or prevention of diseases, disorders, or conditions that may be associated with inflammation, infection, allergy, immune dysfunction, or dysbiosis of the intestinal microbiome. In some aspects, the invention provides a synergistic combination of a prebiotic, e.g., a mixture of human milk oligosaccharides, and probiotic strains of bacteria, such as one or more strains capable of internalizing and consuming the prebiotic, e.g., Bifidobacterium longum subsp. infantis, and one or more strains capable of producing short chain fatty acids such as propionate, e.g., Veillonella sp.
Description
SYNBIOTIC COMPOSITIONS FOR SHORT CHAIN FATTY ACID PRODUCTION
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This application claims priority from U.S. provisional application No. 63/387,807 filed December 16, 2022, entitled “SYNBIOTIC COMPOSITIONS FOR SHORT CHAIN FATTY ACID PRODUCTION” and U.S. provisional application No. 63/490,283 filed March 15, 2023, entitled “SYNBIOTIC COMPOSITIONS FOR SHORT CHAIN FATTY ACID PRODUCTION,” the contents of each of which is incorporated by reference in their entirety.
INCORPORATION BY REFERENCE OF SEQUENCE LISTING
[0002] The present application is being filed along with a Sequence Listing in electronic format. The Sequence Listing is provided as a file entitled PROL04604WOSEQLIST ST25, created December 12, 2023, which is 113 kilobytes in size. The information in the electronic format of the Sequence Listing is incorporated by reference in its entirety.
FIELD OF THE INVENTION
[0003] Provided herein are compositions, methods, strategies, kits, and articles of manufacture that are useful, inter alia, in the treatment or prevention of diseases, disorders, or conditions that may be associated with inflammation, infection, allergy, immune dysfunction, or dysbiosis of the intestinal microbiome. In some aspects, the invention provides a synergistic combination of a prebiotic, e.g., a mixture of human milk oligosaccharides, and probiotic strains of bacteria, such as one or more strains capable of internalizing and consuming the prebiotic, e.g., Bifidobacterium longum subsp. infantis, and one or more strains capable of producing short chain fatty acids such as propionate, e.g., Veillonella sp.
BACKGROUND OF THE INVENTION
[0004] The microorganisms that colonize the human gastrointestinal tract, also referred to as the gut microbiota, comprise a complex community that plays an important role in the maintenance of health and resistance to disease. A multitude of factors, including host diet, environmental exposures, and antibiotic usage, influence the composition of the microbiome, sometimes leading to dysbiosis and negative health consequences. One
promising approach to treat or prevent disease is direct manipulation of the dysbiotic microbiome. Such therapies include live biotherapeutic products (LBPs) that may “engraft” or durably persist in the recipient gastrointestinal tract and thereby lead to positive outcomes. While LBPs conceptually hold promise as potentially effective therapeutic tools, control of their engraftment and accurate prediction of their efficacy and effects on co-resident microbes remain relatively unpredictable. Not only is selection of microbial species still a challenge, but questions remain about the duration and success of engraftment of introduced strains into the human intestinal microbiome.
[0005] What is needed in the art are defined live biotherapeutic products capable of predictably, controllably, and reversibly engrafting within the host intestinal microbiome that reliably benefit the host and treat or prevent disease.
SUMMARY OF THE INVENTION
[0001] Provided herein compositions, kits, articles of manufacture, and methods of use thereof, that are or include a prebiotic mixture, e.g., of human milk oligosaccharides, at least one Bifidobacterium, e.g., B. longum subsp. inf antis, and at least one propionate producing bacterium. The provided prebiotic mixture, Bifidobacteria, and propionate producing bacteria are, inter alia, surprisingly effective for the treatment or prevention of diseases involving an infectious or inflammatory component, or for preventing negative health effects of a dysbiotic intestinal microbiome.
[0002] Provided herein is a method of treating or preventing a disease, disorder, or condition associated with one or more of inflammation, immune dysfunction, cancer, allergy, or dysbiosis of the intestinal microbiome in a subject in need thereof, the method comprising administering to the subject i) a prebiotic mixture comprising one or more human milk oligosaccharides, ii) at least one Bifidobacterium capable of consuming the one or more human milk oligosaccharides; and iii) at least one propionate producing bacterium.
[0003] Also provided herein are uses for a prebiotic mixture and probiotic strains of bacteria for the treatment or prevention of a disease, disorder, or condition associated with one or more of inflammation, immune dysfunction, cancer, allergy, or dysbiosis of the intestinal microbiome in a subject in need thereof. In certain embodiments, the prebiotic mixture comprises one or more human milk oligosaccharides. In certain embodiments, the probiotic strains of bacteria comprise at least one Bifidobacterium capable of consuming one or more human milk oligosaccharides and at least one propionate producing bacterium.
[0004] Additionally provided herein are prebiotic mixtures and probiotic strains of bacteria for use in the manufacture of medicaments for the treatment or prevention of a disease, disorder, or condition associated with one or more of inflammation, immune dysfunction, cancer, allergy, or dysbiosis of the intestinal microbiome in a subject in need thereof. In particular embodiments, provided herein are probiotic strains of bacteria for use in the manufacture of medicaments for the treatment or prevention of a disease, disorder, or condition associated with one or more of inflammation, immune dysfunction, cancer, allergy, or dysbiosis of the intestinal microbiome to a subject in need thereof in conjunction with the administration of a prebiotic mixture. In certain embodiments, the prebiotic mixture comprises one or more human milk oligosaccharides. In particular embodiments, the probiotic strains of bacteria comprise at least one Bifidobacterium capable of consuming one or more human milk oligosaccharides and at least one propionate producing bacterium.
[0005] Provided herein are methods of treating or preventing a disease, disorder, or condition associated with one or more of inflammation, immune dysfunction, cancer, allergy, or dysbiosis of the intestinal microbiome in a subject in need thereof, the method comprising administering to the subject i) a prebiotic mixture comprising one or more human milk oligosaccharides, ii) at least one Bifidobacterium capable of consuming the one or more human milk oligosaccharides; and iii) at least one propionate producing bacterium. Also provided are methods of ameliorating a symptom of a disease, disorder, or condition associated with one or more of inflammation, immune dysfunction, cancer, allergy, or dysbiosis of the intestinal microbiome in a subject in need thereof, the method comprising administering to the subject i) a prebiotic mixture comprising one or more human milk oligosaccharides, ii) at least one Bifidobacterium capable of consuming the one or more human milk oligosaccharides; and iii) at least one propionate producing bacterium.
[0006] In particular embodiments, the subject has received or will receive an allogenic hematopoietic stem cell transplant, and wherein the disease, condition, or disorder comprises graft versus host disease. In some embodiments, the disease, disorder, or condition comprises one or more of obesity, type II diabetes, a chronic inflammatory disease, an autoimmune disease, an infection, an infectious disease domination, bowel resection, or a condition associated with chronic diarrhea. In certain embodiments, the disease, disorder, or condition comprises one or more of irritable bowel syndrome (IBS), inflammatory bowel disease (IBD), short bowel syndrome (SBS), celiac disease, small intestinal bacterial overgrowth (SIBO), gastroenteritis, leaky gut syndrome, pouchitis, or gastric lymphoma. In
particular embodiments, the disease, disorder, or condition comprises an atopic disease. In some embodiments, the atopic disease comprises atopic dermatitis, food allergy, and/or asthma.
[0007] In certain embodiments, the disease, condition, or disorder is associated with an infection. In particular embodiments, the infection comprises a bacterial infection or gut domination. In some embodiments, the bacterial infection or gut domination comprises an infection or gut domination by one or more species, subspecies, or strains of Aeromonas, Bacillus, Blautia, Bordetella, Borrelia, Brucella, Burkholderia, Campylobacter, Chlamydia, Chlamydophila, Citrobacter, Clostridium, Corynebacterium, Coxiella, Ehrlichia, Enterobacter, Enterobacteriaceae, Enterococcus, Escherichia, Faecalicatena, Francisella, Haemophilus, Helicobacter, Hungatella, Klebsiella, Lachnospiraceae, Legionella, Leptospira, Listeria, Morganella, Mycobacterium, Mycoplasma, Neisseria, Orientia, Plesiomonas, Proteus, Pseudomonas, Rickettsia, Salmonella, Shigella, Staphylococcus, Streptococcus, Treponema, Vibrio, or Yersinia, optionally one or more of Aeromonas hydrophila, Bacillus cereus, Campylobacter fetus, Campylobacter jejuni, Clostridium botulinum, Clostridium difficile, Clostridium perfringens, enteroaggregative Escherichia coli, enter ohemorrhagic Escherichia coli, enteroinvasive Escherichia coli, enteropathogenic E. coli, enterotoxigenic Escherichia coli, Escherichia coli 0157:H7, Helicobacter pylori, Klebsiellia pneumonia, Lysteria monocytogenes, Salmonella paratyphi, Salmonella typhi, Staphylococcus aureus, Vibrio cholerae, Vibrio parahaemolyticus, Vibrio vulnificus, or Yersinia enter ocolitica.
[0008] In certain embodiments, the disease, condition, or disorder is associated with an infection. In particular embodiments, the infection comprises a bacterial infection or gut domination. In some embodiments, the bacterial infection or gut domination comprises an infection or gut domination by one or more species, subspecies, or strains of Aeromonas, Bacillus, Bordetella, Brucella, Burkholderia, Campylobacter, Chlamydia, Chlamydophila, Citrobacter, Clostridium, Corynebacterium, Coxiella, Ehrlichia, Enterobacter, Enterobacteriaceae, Enterococcus, Escherichia, Francisella, Haemophilus, Klebsiella, Legionella, Leptospira, Listeria, Morganella, Mycobacterium, Mycoplasma, Neisseria, Orientia, Plesiomonas, Proteus, Pseudomonas, Rickettsia, Salmonella, Shigella, Staphylococcus, Streptococcus, Treponema, Vibrio, or Yersinia, optionally one or more of Aeromonas hydrophila, Bacillus cereus, Campylobacter fetus, Campylobacter jejuni, Clostridium botulinum, Clostridium difficile, Clostridium perfringens, enteroaggregative
Escherichia coli, enterohemorrhagic Escherichia coli, enteroinvasive Escherichia coli, enteropathogenic E. coli, enterotoxigenic Escherichia coli, Escherichia coli 0157:H7, Helicobacter pylori, Klebsiella pneumoniae, Listeria monocytogenes, Salmonella paratyphi, Salmonella typhi, Staphylococcus aureus, Vibrio cholerae, Vibrio parahaemolyticus, Vibrio vulnificus, or Yersinia enterocolitica.
[0009] In certain embodiments, the bacterial infection or gut domination comprises an infection or gut domination by one or more of Citrobacter freundii, Citrobacter koseri, Enterobacter aerogenes, Enterobacter cloacae, Enterococcus faecalis, Enterococcus faecium, Escherichia coli, Klebsiella oxytoca, Klebsiella pneumoniae, Morganella morganii, Proteus mirabilis, Serratia marcescens, Staphylococcus aureus, Staphylococcus epidermidis, Streptococcus anginosus, Streptococcus australis, Streptococcus constellatus, Streptococcus cristatus, Streptococcus gordonii, Streptococcus infantis, Streptococcus intermedins, Streptococcus mitis, Streptococcus mutans, Streptococcus oligofermentans, Streptococcus oralis, Streptococcus parasanguinis, Streptococcus peroris, Streptococcus pneumoniae, Streptococcus pseudopneumoniae, Streptococcus salivarius, Streptococcus sanguinis, Streptococcus sobrinus, Streptococcus tigurinus, or Streptococcus vestibularis.
[0010] In particular embodiments, wherein the bacterial infection or gut domination comprises an infection or gut domination by drug -resistant bacteria. In some embodiments, the drug-resistant bacteria comprise one or more of antibiotic-resistant bacterium (ARB), Antibiotic-resistant Proteobacteria, Carbapenem-resistant Ewterotocterzaceae (CRE), Extended Spectrum Beta-Lactamase producing Enterobacteriaceae (ESBL-E), fluoroquinolone-resistant Enter obacteriaceae, extended spectrum beta-lactam resistant Enterococci (ESBL), vancomycin-resistant Enterococci (VRE), multi-drug resistant A. coli, or multi-drug resistant Klebsiella.
[0011] In certain embodiments, the bacterial infection or gut domination comprises an infection or gut domination by drug-resistant bacteria. In particular embodiments, the drugresistant bacteria comprises one or more of antibiotic-resistant bacterium (ARB), Antibioticresistant Proteobacteria, Carbapenem-resistant Enterobacteriaceae (CRE), Extended Spectrum Beta-Lactamase producing Enterobacterales (ESBL-E), fluoroquinolone-resistant Enterobacteriaceae, vancomycin-resistant Enterococci (VRE), multi-drug resistant A. coli, or multi-drug resistant Klebsiella.
[0012] In some embodiments, the subject has undergone or will undergo an ileal pouch-anal anastomosis (IPAA) surgery, and wherein the disease, condition, or disorder comprises pouchitis.
[0013] In certain embodiments, provided herein is a method for increasing propionate in the gut of a subject in need thereof, the method comprising administering to the subject i) a prebiotic mixture comprising one or more human milk oligosaccharides, ii) at least one Bifidobacterium capable of consuming the one or more human milk oligosaccharides; and iii) at least one propionate producing bacterium. In certain embodiments, the at least one propionate producing bacterium comprises a strain that is isolated from a human intestinal microbiome and/or is capable of engrafting within the human intestinal microbiome.
[0014] In particular embodiments, the at least one propionate producing bacterium comprises one or more strains of the genus Veillonella. In some embodiments, the at least one propionate producing bacterium comprises one or more of Veillonella atypica (V. atypica), Veillonella dispar (V. dispar), Veillonella infantium (V. infantium), Veillonella nakazawae (V. nakazawae), Veillonella parvula (V. parvula), Veillonella ratti (V. ratti), Veillonella rogosae (V. rogosae), Veillonella seminalis (V. seminalis), and/or Veillonella tobetsuensis (V. tobetsuensis). In certain embodiments, the at least one propionate producing bacterium comprises V. atypica, V. dispar, V. infantium, V. nakazawae, V. parvula, and/or V. rogosae. In particular embodiments, the at least one propionate producing bacterium comprises V. infantium, V. nakazawae, V. parvula, and/or V. rogosae. In some embodiments, the at least one propionate producing bacterium comprises V. infantium, V. dispar, and/or V. nakazawae . In certain embodiments, the at least one propionate producing bacterium comprises V. parvula. In particular embodiments, the at least one propionate producing bacterium comprises V. rogosae.
[0015] In some embodiments, the at least one propionate producing bacterium comprises one or more strains of the genus Megasphaera. In certain embodiments, the at least one propionate producing bacterium comprises one or more of Megasphaera elsdenii (M. elsdenii), Megasphaera hominis (M. hominis), Megasphaera indica (M. indica), Megasphaera massiliensis (M. massiliensis), and/ or Megasphaera micronuciformis (M. micronuciformis). In particular embodiments, the at least one propionate producing bacterium comprises one or more of M. elsdenii ndJo M. massiliensis. In some embodiments, the at least one propionate producing bacterium comprises one or more strains of the genus Anaerotignum. In certain embodiments, the at least one propionate producing bacterium
comprises Anaerotignum lactatifermentans (A. lactatifermentans). In particular embodiments, the at least one propionate producing bacterium comprises one or more strains of the genus Bacteroides. In some embodiments, the at least one propionate producing bacterium comprises Bacteroides fragilis (B. fragilis o Bacteroides caccae (B. caccae). In certain embodiments, the at least one propionate producing bacterium comprises one or more strains of the genus Coprococcus. In particular embodiments, the at least one propionate producing bacterium comprises Coprococcus catus (C. catus). In some embodiments, the at least one propionate producing bacterium comprises one or more strains of the genus Merdimmobilis . In certain embodiments, the at least one propionate producing bacterium comprises Merdimmobilis hominis.
[0016] In particular embodiments, the propionate producing bacterium comprises a nucleotide sequence with at least 97%, 98%, or 99% sequence identity to any of SEQ ID NOS: 40-49 or 52-58 and/or an amino acid sequence with at least 97%, 98%, or 99% sequence identity to SEQ ID NO: 50 or 51. In some embodiments, the Bifidobacterium comprises B. breve, B. bifidum, or B. longum subsp. infantis. In certain embodiments, the Bifidobacterium comprises B. longum subsp. infantis.
[0017] In particular embodiments, the method further comprising at least one butyrate producing strain of bacteria. In some embodiments, the at least one butyrate producing strain comprises a strain of Clostridium Cluster IV or Clostridium Cluster XlVa bacteria. In certain embodiments, the at least one butyrate producing strain comprises one or more of Agathobacter rectalis, Anaerobutyricum hallii, Anaerostipes caccae, Blautia producta, Clostridium leptum, Faecalibacterium prausnitzii, Anaerobutyricum soehngenii, Roseburia hominis, or Roseburia intestinalis. In particular embodiments, the at least one butyrate producing strain comprises one or more of Anaerostipes caccae, Clostridium innocuum, Roseburia hominis, or Roseburia intestinalis.
[0018] In some embodiments, the prebiotic mixture comprises one or more of 2'- fucosyllactose, 3-fucosyllactose, difucosyllactose, 3’-sialyllactose, 6'-sialyllactose, lacto-N- tetraose, lacto-N-neo-tetraose, lacto-N-fucopentaose I, lacto-N-fucopentaose II, lacto-N- fucopentaose III, sialyl-lacto-N-tetraose a, sialyl-lacto-N-tetraose b, sialyl-lacto-N-tetraose c, lacto-N-difuco-hexaose I, lacto-N-difuco-hexaose II, lacto-N-hexaose, para-lacto-N-hexaose, disialyllacto-N-tetraose, Fucosyl-lacto-N-hexaose, difucosyl-lacto-N-hexaose a, difucosyl- lacto-N-hexaose b, lactodifucotetraose, 6’galactosyllactose, 3 ’galactosyllactose, 3-sialyl-3- fucosyllactose, sialylfucosyllacto-N-tetraose, sialyllacto-N-fucopentaose V, disialyl-lacto-n-
fucopentaose II, disialyl-lacto-n-fucopentaose V, lacto-N-neo-difucohexaose II, 3-Fucosyl- sialylacto-N-tetraose c, para-lacto-N-neohexose, lacto-N-octaose, lacto-N-neooctaose, lacto- N-neohexaose, lacto-N-fucopentaose V, iso-lacto-N-octaose, para-lacto-N-octaose, lacto- decaose, or sialyl-lacto-N-fucopentaose I.
[0019] In certain embodiments, the prebiotic mixture comprises one or more of 2'- fucosyllactose, 3-fucosyllactose, 3'-sialyllactose, 6'-sialyllactose, lacto-N-tetraose, lacto-N- difucohexaose I, lactodifucotetraose, lacto-N-fucopentaose I, sialylacto-N-tetraose c, sialylacto-N-tetraose b, or disialyllacto-N-tetraose. In particular embodiments, the prebiotic mixture comprises one or more of 2'-fucosyl-lactose, 3-fucosyllactose, 3’-sialyllactose, 6'- sialyllactose, lacto-N-tetraose, lacto-N-neotetraose, or difucosyllactose. In some embodiments, the prebiotic mixture comprises one or more of 2'-fucosyllactose, 3- fucosyllactose, lacto-N-tetraose, or lacto-N-neotetraose. In certain embodiments, the prebiotic mixture comprises one or both of 2'-fucosyllactose and lacto-N-neotetraose. In particular embodiments, the prebiotic mixture comprises at least 10, at least 25, at least 50, at least 100, or at least 150 human milk oligosaccharides. In some embodiments, the prebiotic mixture comprises 2'-fucosyllactose, 3-fucosyllactose, 3’-sialyllactose, 6'-sialyllactose, lacto-N- tetraose, lacto-N-difucohexaose I, lactodifucotetraose, lacto-N-fucopentaose I, sialylacto-N- tetraose c, sialylacto-N-tetraose b, and disialyllacto-N-tetraose.
[0020] In certain embodiments, the prebiotic mixture is, is derived from, or comprises a concentrated human milk permeate, wherein the concentrated human milk permeate is obtained by a process comprising the steps of ultra-filtering human skim milk to obtain human milk permeate and concentrating the human milk oligosaccharide content of the human milk permeate. In particular embodiments, the human skim milk is obtained from human milk pooled from at least 25, 50, or 100 individual donors.
[0021] Also provided herein is a kit comprising i) a prebiotic mixture comprising one or more human milk oligosaccharides, ii) at least one Bifidobacterium capable of consuming the one or more human milk oligosaccharides; and iii) at least one propionate producing bacterium. In some embodiments, the at least one propionate producing bacterium comprises a strain that is isolated from a human intestinal microbiome and/or is capable of engrafting within the human intestinal microbiome. In certain embodiments, the kit further comprises at least one butyrate producing strain of bacteria.
[0022] Further provided herein is a method for increasing propionate concentration and/or propionate production in the gut of a subject in need thereof, the method comprising
administering to the subject the prebiotic mixture, the at least one Bifidobacterium, and the at least one propionate producing bacterium any of the kits described herein.
[0023] Also provided herein is a method of treating or preventing a disease, disorder, or condition associated with one or more of inflammation, immune dysfunction, cancer, allergy, or dysbiosis of the intestinal microbiome in a subject in need thereof, the method comprising administering to the subject the prebiotic mixture, the at least one Bifidobacterium, and the at least one propionate producing bacterium of any of the kits described herein.
[0024] Additionally provided herein is a method of ameliorating a symptom of a disease, disorder, or condition associated with one or more of inflammation, immune dysfunction, cancer, allergy, or dysbiosis of the intestinal microbiome in a subject in need thereof, the method comprising administering to the subject the prebiotic mixture, the at least one Bifidobacterium, and the at least one propionate producing bacterium of any of the kits described herein.
[0025] Also provided is a pharmaceutical composition comprising i) a prebiotic mixture comprising one or more human milk oligosaccharides, ii) at least one Bifidobacterium capable of consuming the one or more human milk oligosaccharides; and iii) at least one propionate producing bacterium. In particular embodiments, the at least one Bifidobacterium comprises B. longum subsp. inf antis and the at least one propionate producing bacterium comprises one or more strains of the genus Veillonella. In some embodiments, the pharmaceutical composition further comprises at least one butyrate producing strain of bacteria, optionally wherein the at least one butyrate producing strain comprises one or more of Agathobacter rectalis, Anaerobutyricum hallii, Anaerostipes caccae, Blautia producta, Clostridium leptum, Faecalibacterium prausnitzii, Anaerobutyricum soehngenii, Roseburia hominis, or Roseburia intestinalis.
[0026] Also provided herein is a method for treating one or more symptoms of acute radiation syndrome in a subject in need thereof, the method comprising administering to the subject i) a prebiotic mixture comprising one or more human milk oligosaccharides and ii) at least one Bifidobacterium capable of consuming the one or more human milk oligosaccharides, thereby treating the symptoms of acute radiation syndrome. In certain embodiments, the at least one Bifidobacterium comprises B. longum subsp. infantis. In particular embodiments, the method further comprises administering to the subject in need thereof iii) at least one propionate producing bacterium. In some embodiments, the at least
one propionate producing bacterium comprises one or more strains of the genus Veillonella. In certain embodiments, the one or more symptoms of acute radiation syndrome comprise gastrointestinal damage. In particular embodiments, the subject was exposed to an ionizing radiation dose of at least 0.3 Gray (Gy), optionally at least 0.7 Gy, 1 Gy, 2 Gy, 5 Gy, 6 Gy, or 10 Gy over a period of time lasting under 60 minutes, optionally under 30 minutes, 15 minutes, 10 minutes, 5 minutes, 3 minutes, 2 minutes, or 1 minute.
[0027] Also provided herein is an article of manufacture, comprising any of the kits described herein and instructions for use describing any of the methods described herein.
BRIEF DESCRIPTION OF THE DRAWINGS
[0028] FIG. 1 provides a schematic of the study described in Example 2.
[0029] FIGS. 2A-2D show graphs displaying time courses of B. longum subsp. infantis abundance during a clinical study. FIG. 2A shows B. infantis abundance over time as determined by qPCR and plotted as the average for subjects in each of the three cohorts. Circles indicate Cohort 1, triangles indicate Cohort 2, and squares indicate Cohort 3. Data are geometric means with 95% confidence intervals. The line labeled LOD represents the limit of detection (27 copies/ng DNA). Significant differences were calculated using mixed effects analysis for repeated measures of log-transformed data; significance between kinetic curves were determined using the time versus treatment factor and significances at individual timepoints were determined using Sidak’s post-test. FIG. 2B shows B. infantis abundance over time as determined by qPCR for subjects in the cohort that received B. infantis and HMO (Cohort 3) comparing those who were engrafted and not engrafted. Subjects were deemed “engrafted” (dark lines) if B. infantis signal was two geometric standard deviations above the geometric mean of signal on days 3-5 (equivalent to 5.4 x 103 copies/ng DNA) for at least two consecutive time points; subjects below the geometric mean of the B. infantis only cohort on any one of those days were deemed “not engrafted” (light lines). Traces represent individual subjects. FIG. 2C shows B. infantis abundance over time as determined by qPCR for subjects in the cohort that received B. infantis alone (Cohort 2) comparing those who were engrafted (dark lines) and not engrafted (light lines), using the definitions of “engrafted” and “not engrafted” in FIG. 2B. FIG. 2D shows the abundance of B. infantis within the microbiome as determined by whole metagenome sequencing (WMS) using a strain tracking algorithm. Circles indicate Cohort 1, triangles indicate Cohort 2, and squares
indicate Cohort 3. Each point represents the median of the indicated cohort subpopulation, and error bars represent the interquartile range. Undetected values were imputed with the LOD. The LOD represents the lowest detected value.
[0030] FIGS. 3A-3F show graphs summarizing whole metagenomic sequencing (WMS) from genomic DNA extracted from stool samples collected from subjects as described in Example 2. FIG. 3A displays Shannon Entropy of observed reads rarefied to 76,000 reads used as a measure of the alpha diversity of samples over time. Data are median and 95% confidence interval. Significant differences were calculated using mixed effects analysis for repeated measures; significance between kinetic curves was determined using the time versus treatment factor. Significances between individual timepoints were determined using Sidak’s post-test for each individual cohort and asterisks indicate significance across both cohorts. FIG. 3B shows Bray-Curtis dissimilarity for each sample with rarefied sequences aggregated to the genus taxonomic level and displayed by cohort and day in a principal coordinate analysis (PcoA) plot noting the trajectory from pre-treatment (day 1) and end (day 5) of antibiotic treatment (arrow). FIG. 3C contains stacked bar charts displaying aggregated families (and two at the order level) present at >1% abundance for each cohort on each day of the study. Taxa <1% in each cohort were aggregated to “Other”. For the antibiotics + B. infantis + HMO cohort (Cohort 3), subjects are separated into Engrafted and Not Engrafted (NE). FIG. 3D shows PcoA plots of Bray-Curtis dissimilarity calculated with rarefied sequences aggregated to the genus taxonomic level for days 1, 5 and 14 only. The antibiotics only cohort was compared to engrafted subjects in the antibiotics + B. infantis + HMO cohort, including (top row) and excluding (bottom row) taxa assigned to the genus Bifidobacterium. FIG. 3E shows a volcano plot of aggregated genera >1% across cohorts comparing abundance in Cohort 3 with Cohort 1. Log2 fold change (CLR transformed, sampled 150 times from a Dirichlet model) and p-values (Wilcoxon’s Rank Sum Test with Bejamini -Hochberg correction for multiple comparisons) were calculated using ALDEx2 (v 1.29.1). Values with an adjusted p value < 0.05 and greater than 0.6-fold change are highlighted and colored by taxon. FIG. 3F shows the relative abundance of Veillonella across study days comparing Cohort 1 (left) and Cohort 3 (right) showing statistical differences calculated using the CLR transformed abundances in ALDEx2.
[0031] FIGS. 4A-4F show graphs summarizing gut metabolites in stool samples of antibiotic-treated subjects. FIGS. 4A and 4C show levels of acetic acid (FIG. 4A) and lactic acid (FIG. 4C) as measured in stool samples from subjects in the antibiotics only cohort
(Cohort 1, circles) and engrafted subjects in the antibiotics + B. infantis + HMO cohort (Cohort 3, squares). Dotted lines represent limit of detection. Significant differences were calculated using mixed effects analysis for repeated measures of log-transformed data; significance between kinetic curves was determined using the time versus treatment factor. In FIG. 4A, significance between day 1 and day 5 was determined using Sidak’s post-test for each individual cohort and was significant across both cohorts. In FIG. 4C, significances between cohorts at individual timepoints were determined using Sidak’s post-test. FIG. 4B shows Kaplan-Meier curve plotting the day on which stool acetate for each subject returned to baseline levels, where baseline is the average of subjects at Day 1. Significance between groups was calculated using log-rank test. FIG. 4D provides a Volcano plot showing log2 fold change in fecal metabolites on day 14 between engrafted subjects in the antibiotics + B. infantis + HMO cohort (Cohort 3) and the antibiotics only cohort (Cohort 1) plotted against significance of the observed change, calculated via two-way repeated measures ANOVA. The dotted line represents the threshold of p = 0.05. Metabolites at a significantly different abundance are colored to indicate categorization. Notable metabolites are labeled. FIG. 4E shows levels of indolelactate in stool at day 14 (left) and in serum at days 1, 5, 14, 28, and 35 (right). FIG. 4F shows levels of p-cresol sulfate in stool at day 14 (left) and in serum at days 1, 5, 14, 28, and 35 (right). In FIGS. 4E and 4F scatterplot data, horizontal lines are geometric mean; dotted lines are the imputed value for samples without detected signal, and significances are derived from two-way repeated measures ANOVA calculated for the entire global metabolomics dataset as described in FIG. 4D. Timecourse data in FIGS. 4E and 4F are geometric mean and 95% confidence interval; dotted lines are limit of detection; significances are calculated as in FIG. 4B.
[0032] FIGS. 5A-5D show graphs summarizing in vitro cross-feeding of three different Veillonella species by B. infantis + HMO. The indicated propionate-producing species of Veillonella were grown with no addition, B. infantis, HMO, both B. infantis and HMO, or lactate alone as indicated. Dotted lines labeled “LOQ” indicate the limits of quantitation. FIG. 5A shows propionate quantified using liquid chromatography with tandem mass spectrometry from media collected after 30 hours of culture with media alone or the noted concentration of lactate. FIG 5B shows propionate quantified from media collected after 30 hours using liquid chromatography with tandem mass spectrometry. FIGS. 5C and 5D show abundance of B. infantis (FIG. 5C) or Veillonella spp. (FIG. 5D) quantified using qPCR after 30 hours of culture. Data are geometric mean and standard deviation of 3
independent experiments. Statistics reflect a two-way ANOVA with Tukey’s test for multiple comparisons within each Veillonella condition, using log-transformed data (* indicates adjusted p <0.05, ns indicates p > 0.05). Within each experiment, triplicate culture wells were pooled for propionate analysis and qPCR.
[0033] FIGS. 6A-6D depict in vivo cross-feeding of Veillonella species by B. infantis + HMO. FIG. 6A provides a study schematic. Germ-free mice were divided into three groups, two of which were inoculated with Veillonella parvula and the third with B. infantis as a control. After one week, the two V. parvula-associated groups were given a single gavage of either PBS or B. infantis + HMO; B. infantis-associated animals were gavaged with HMO only. Over the subsequent three days, animals received once-daily gavages of PBS or HMO. FIG. 6B shows quantification of Veillonella in feces. Cage-bottom fecal pellets collected over time were used to quantify the levels of Veillonella in cage-bottom fecal pellets using qPCR. Each pair of connected symbols represents an individual mouse. FIG. 6C shows the levels of propionate quantified in contents of the indicated intestinal segments using LC-MS/MS. FIG. 6D shows the levels of lactate quantified in contents of the indicated intestinal segments using LC-MS/MS. Significance of differences was calculated by Sidak’s multiple comparison test from mixed effects analysis for repeated measures of log- transformed data.
[0034] FIGS. 7A and 7B show relative abundance of B. infantis as determined by qPCR. FIG. 7A shows data from FIG. 2 A replotted to exclude the non-engrafted subjects in the B. infantis + HMO cohort; significances were calculated as in FIG. 2A. FIG. 7B shows B. infantis relative abundance in stool samples determined using qPCR to measure B. infantis (shown in FIG. 2 A) and 16S rRNA and then normalizing B. infantis levels to 16S rRNA levels. Each point represents the median, and error bars represent the interquartile regions. The dotted line represents limit of detection (LOD) calculated across the entire study using the following calculation: [LOD for B. infantis copy number per reaction] / [median 16S rRNA gene copy number per reaction across all samples],
[0035] FIGS. 8A-8H depict microbiome changes after antibiotic perturbation. FIG. 8A shows number of observed reads whole metagenomic sequencing (WMS) data from stool samples over time; significances were calculated as in FIG. 3 A. FIG. 8B shows the geometric mean of 16S rRNA gene copy number per gram of stool for each cohort over time. Each dot represents the geometric mean and error bars represent the 95% confidence intervals. Significances were calculated as in FIG. 3 A except that data were log-transformed
prior to statistical analysis. FIG. 8C provides stacked bar charts of metagenomic sequencing at the family level for subjects receiving B. infantis only (Cohort 2) over time. Each study day is separated into engrafted (E) or not engrafted. FIGS. 8D-I are box and whisker plots of the relative abundance of bacteria highlighted in the ALDEx2 analysis shown in FIG. 3E and Bacteroides over time comparing subjects in the antibiotics only cohort (Cohort 1, left) and the B. Infantis + HMO (engrafted only) cohort (Cohort 3, right).
[0036] FIGS. 9A-9N depict changes in gut metabolites after antibiotic perturbation. FIGS. 9A and 9B show levels of butyrate (FIG. 9A) and propionate (FIG. 9B) measured in stool samples collected on days 1, 5, 9, 11, 14, 17, 28, and 35 from subjects that received antibiotics only (Cohort 1, circles) and engrafted subjects who also received B. infantis and HMO (Cohort 3, squares). Dots represent the geometric mean and error bars represent the 95% confidence intervals. Significances were calculated as in FIG 4A. FIG. 9C shows pH measurements of fecal samples within cohorts over time. Data show average pH with error bars representing standard error. A mixed effects model with Sidak’s multiple comparisons test was used for comparisons. FIGS. 9D-9I show volcano plots of fecal global metabolomic data comparing cohorts (FIG. 9F and FIG. 91) or study days within each cohort (FIGS. 9D, 9E, 9G, and 9H). FIG. 9J shows Bray-Curtis dissimilarity calculated for each sample and displayed by cohort and day in a PcoA plot. FIG. 9K shows HMOs from the stool metabolomics analysis on day 5 represented in batch-normalized imputed peak area values. Each dot represents a subject in the Antibiotics only cohort (Cohort 1, left) or the engrafted subjects in the B. infantis + HMO cohort (Cohort 3, right), with a line at the median. The observed minimum value was imputed for each individual metabolite if a metabolite was undetected in any sample. Significances are derived from two-way repeated measures ANOVA calculated for the entire global metabolomics dataset as described in FIG 4D. FIG. 9L is a volcano plot showing fold change in serum metabolites in the Antibiotics Only cohort (Cohort 1) between Day 5 and Day 1 against significance of the observed change. Significances were calculated with a repeated measures test on log-transformed data and Sidak’s post-test to correct for multiple comparisons. Relevant metabolites are noted in color and labeled. FIG. 9M is a volcano plot showing fold change in serum metabolites on Day 14 between engrafted subjects (Cohort 3) and subjects that only received antibiotics (Cohort 1) plotted against significance of the observed change, calculated as in FIG. 9L. FIG. 9N shows levels of phenol sulfate measured in serum samples over time for each cohort. Data and significances are presented as in FIG. 9A.
[0037] FIGS. 10A-10G show in vitro cross-feeding of Veillonella species by B. infantis + HMO. The indicated species of Veillonella were grown in the absence or presence of B. infantis, HMO, both, or lactate. FIGS. 10A-10C are growth curves of the three different Veillonella strains presented in FIG. 5 A with varying concentrations of lactate. Data are mean and standard deviation of 9 replicates from 3 independent experiments. FIGS. 10D-10G show quantification of acetate (FIGS. 10D and 10F) and lactate (FIGS. 10E and 10G) in cultures presented in FIG. 5. Data represent geometric mean and standard deviation of 3 independent experiments. Within each experiment, triplicate culture wells were pooled for analysis. Dotted lines indicate the limit of quantitation. Statistics reflect a two-way ANOVA with Tukey’s test for multiple comparisons to assess acetate production (FIGS. 10D and 10F) or lactate consumption (FIGS. 10E and 10G) under each set of growth conditions, in all cases using log-transformed data (* indicates adjusted p <0.05, “ns” indicates adjusted p >0.05). Lines in FIG. 10E and 10G indicate the displayed comparisons between levels of lactate in cultures containing Veillonella and the corresponding Veillonella-V culture.
[0038] FIGS. 11A-11C depict in vivo cross-feeding of Veillonella species with B. infantis + HMO in mice described in Example 12. FIGS. HA and 11B show quantification of Veillonella (FIG. 11 A) and B. infantis (FIG. 11B) in mice using qPCR. Data are plotted as the geometric mean and standard deviation. FIG. 11C shows concentration of acetate isolated intestinal sections and feces from mice of the experiments shown in FIG. 6.
[0039] FIG. 12 shows a scatterplot depicting concentrations of propionate (Y axis) and lactate (x axis) of various test strains following a 48 hour incubation with lactate as a carbon source. Starting lactate concentration is indicated by the vertical dotted line.
[0040] FIGS. 13A-13C show quantification of Veillonella sp. (left) and B. longum subsp. infantis (B. infantis,' right) in stool collected at various timepoints from germ free mice inoculated with Veillonella sp., B. infantis, or both and administered a human milk oligosaccharide composition (HMO) or a vehicle control. Veillonella sp. included a strain of V. parvula (FIG. 13A) and two Veillonella strains (FIGS. 13B and 13C) isolated as described in Example 11. Strains were quantified using qPCR. Data are plotted as the geometric mean and standard deviation.
[0041] FIGS. 14A-14C show quantification of propionate levels detected in ileal, cecal, rectal, and fecal samples collected from germ free mice inoculated with Veillonella sp. (V sp.), B. infantis (BI), or both and administered a human milk oligosaccharide composition (HMO) or a vehicle control. Veillonella sp. included a strain of V. parvula (FIG. 14A) and
two Veillonella strains (FIGS. 14B and 14C) isolated as described in Example 11. Data are plotted as the geometric mean and standard deviation. Data are plotted as the geometric mean and standard deviation.
[0042] FIGS. 15A-15C show quantification of lactate levels detected in ileal, cecal, rectal, and fecal samples collected from germ free mice inoculated with Veillonella sp. (V sp.), B. infantis (BI), or both and administered a human milk oligosaccharide composition (HMO) or a vehicle control. Veillonella sp. included a strain of V. parvula (FIG. 15A) and two Veillonella strains (FIGS. 15B and 15C) isolated as described in Example 11. Data are plotted as the geometric mean and standard deviation. Data are plotted as the geometric mean and standard deviation.
[0043] FIGS. 16A-16C show quantification of acetate levels detected in ileal, cecal, rectal, and fecal samples collected from germ free mice inoculated with Veillonella sp. (V sp.), B. infantis (BI), or both and administered a human milk oligosaccharide composition (HMO) or a vehicle control. Veillonella sp. included a strain of V. parvula (FIG. 16A) and two Veillonella strains (FIGS. 16B and 16C) isolated as described in Example 11. Data are plotted as the geometric mean and standard deviation. Data are plotted as the geometric mean and standard deviation.
[0044] FIGS. 17A-17F depict experimental results following inoculation of strains in germ free mice. FIG. 17A show quantification of B. longum subsp. infantis (B. infantis; left) and Megasphaera elsdenii (M. elsdenii; right) in stool collected at various timepoints from germ free mice inoculated with A/, elsdenii, B. infantis, or both and administered a human milk oligosaccharide composition (HMO) or a vehicle control. Strains were quantified using qPCR. FIGS. 17B-17F show quantification of butyrate (FIG. 17B), propionate (FIG. 17C), valerate (FIG. 17D), lactate (FIG. 17E), and acetate (FIG. 17F) levels detected in cecal samples collected from these mice. Data are plotted as the geometric mean and standard deviation.
DETAILED DESCRIPTION
[0045] Provided herein are compositions, kits, and articles of manufacture as well as methods of use thereof. In certain embodiments, the provided compositions, kits, and articles of manufacture are or include prebiotics and at least two probiotic strains of bacteria. In particular embodiments, the prebiotics are or include a mixture, e.g., of oligosaccharides such
as human milk oligosaccharides. In some embodiments, the at least two probiotic strains of bacteria are or include one or more strains of Bifidobacteria, for example a strain of Bifibacterium capable of consuming the prebiotics and/or human milk oligosaccharides, and a strain of bacteria capable of producing the short chain fatty acid propionate, e.g., a strain of Veillonella sp. In some aspects, the provided compositions, kits, and articles of manufacture may be administered to a subject to treat or prevent dysbiosis, e.g., of the intestinal microbiome, as well as conditions, diseases, or disorders that may originate from or cause dysbiosis. In some aspects, the provided compositions, kits, and articles of manufacture may be administered to a subject to treat or prevent conditions, diseases, or disorders that are, include, or contribute to inflammation, infection, allergy, or immune dysfunction.
[0046] In certain aspects, the maintenance of a healthy human metabolism depends on a symbiotic consortium among bacteria, archaea, viruses, fungi, and host eukaryotic cells throughout the human gastrointestinal tract. For example, microbial communities may provide enzymatic machinery and metabolic pathways that contribute to food digestion, xenobiotic metabolism, and production of a variety of bioactive molecules. Disturbances to the microbiome may result in a microbial imbalance (dysbiosis) characterized by phylumlevel changes in the microbiota composition, including a marked decrease in the representation of obligate anaerobic bacteria and an increased relative abundance of facultative anaerobic bacteria. While dysbiosis is associated with numerous diseases and conditions, successfully treating dysbiosis is difficult, particularly in vulnerable or immunocompromised patients.
[0047] The provided compositions, methods, kits, and articles of manufacture address these needs. In particular, the present invention includes specific combinations of prebiotics, such as human milk oligosaccharides, and probiotics, such as Bifidobacterium including B. longum subspecies (subsp.) infantis and bacteria capable of producing propionate, e.g., Veillonella sp., that are particularly safe and effective for treating, ameliorating, or reducing dysbiosis in the gut microbiome as well as effective in treating, ameliorating, or preventing diseases or disorders that may be accompanied by dysbiosis, such including but not limited to diseases associated with immune disorders, inflammatory disorders, or infection. This synbiotic approach effectively allows for the beneficial Bifidobacteria and Veillonella to grow, expand, and engraft within the gut, thereby regulating pH, suppressing growth of opportunistic and/or pathogenic microbes, and producing short chain fatty acids (SCFAs) such as propionate to reduce inflammation in the host.
[0006] All publications, including patent documents, scientific articles, and databases, referred to in this application are incorporated by reference in their entirety for all purposes to the same extent as if each individual publication were individually incorporated by reference. If a definition set forth herein is contrary to or otherwise inconsistent with a definition set forth in the patents, applications, published applications, and other publications that are herein incorporated by reference, the definition set forth herein prevails over the definition that is incorporated herein by reference.
[0007] The section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described.
I. COMPOSITIONS, KITS, AND ARTICLES OF MANUFACTURE
[0008] Provided herein are compositions, kits, and articles of manufacture that are or include i) a prebiotic mixture that are or include one or more oligosaccharides, ii) at least one strain of bacteria from the genus Bifidobacterium that is capable of consuming oligosaccharides, e.g., the oligosaccharides of the mixture and/or human milk oligosaccharides; and iii) one or more strains of bacteria capable of producing propionate. In some embodiments, the prebiotic mixture is or includes human milk oligosaccharides. In certain embodiments, the Bifidobacteria is or includes B. longum subsp. infantis. In particular embodiments, the one or more strains of bacteria capable of producing propionate are or include strains of bacteria from the genus Veillonella.
[0009] In certain embodiments, the Bifidobacteria capable of consuming human milk oligosaccharides, the strain capable of producing propionate, and the prebiotic mixture are or are included in separate compositions, e.g., are administered separately to a subject. Thus, in certain embodiments, provided herein are kits and articles of manufacture that include (i) a composition that is or includes a strain from the genus Bifidobacteria capable of consuming human milk oligosaccharides (ii) a composition that is or includes at least one propionate producing strain of bacterium (also referred to herein and used interchangeably with “strain of bacteria capable of producing propionate” or “propionate producing bacterium” unless otherwise indicated), and (iii) a composition that is or includes the prebiotic mixture, e.g., of human milk oligosaccharides. Also provided are kits and articles of manufacture that are or include two compositions, a probiotic composition that is or includes the Bifidobacteria, e.g.,
B. longum subsp. infantis, and bacteria capable of producing propionate, e.g., Veillonella, and a composition that is or includes the prebiotic mixture, e.g., of human milk oligosaccharides. Thus, in some embodiments, additionally provided are kits and articles of manufacture that include a composition that is or includes all of the Bifidobacteria, e.g., B. longum subsp. infantis, the strain of bacteria capable of producing propionate, e.g., Veillonella, and the prebiotic mixture, e.g., of human milk oligosaccharides.
[0010] In certain embodiments, the provided compositions, kits, and articles of manufacture are or include any of the strains of bacteria capable of producing propionate, (also referred to herein as a “propionate producing strain of bacteria,” “propionate producing strain” or “propionate producing bacterium”) described throughout the application, including in Section I-A. In particular embodiments, the provided compositions, kits, and articles of manufacture are or include any of the one or more strains of the genus Bifidobacterium, e.g., a strain of Bifidobacterium capable of consuming human milk oligosaccharides, described throughout the application including in Section I-B. In some embodiments, the provided compositions, kits, and articles of manufacture are or include any of the prebiotic mixtures, e.g., of human milk oligosaccharides, described throughout the application, including at least at Section I-C. The provided kits and articles of manufacture may also include labels or instructions for use. In some embodiments, such labels or instructions for use may describe any of the uses or methods provided herein, such as those described in Section II.
A. Propionate producing bacteria
[0011] In certain embodiments, the at least one propionate producing bacterium is capable of consuming lactate and/or acetate. In certain embodiments, the at least one propionate producing bacterium is capable of producing, generating, and/or creating propionate in the presence of lactate and/or acetate. In certain embodiments, the at least one propionate producing bacterium is capable of growing or expanding in the presence of a strain of Bifidobacterium, e.g., a strain of Bifidobacterium probiotic strain described herein such as in Section I-B. In some embodiments, the at least one propionate producing strain of bacterium is capable of growing or expanding in the presence of B. longum subsp. infantis.
[0012] In some embodiments, the at least one propionate producing bacterium is or includes at least one, two, three, four, five, six, seven, eight, nine, ten, or more species, subspecies, or strains of bacteria capable of producing propionate. In particular
embodiments, the propionate producing bacterium is or includes at least three strains of bacteria capable of producing propionate. In some embodiments, the propionate producing strain is or includes between one and five species, subspecies, or strains of bacteria capable of producing propionate.
[0013] In certain embodiments, the at least one propionate producing bacterium has or contains one or more functional genes that contribute to the production, generation, synthesis, or making of propionate.
[0014] In particular embodiments, the at least one propionate producing bacterium is or includes a strain of bacterium from the genus Veillonella. In certain embodiments, the at least one propionate producing bacterium is or includes Veillonella sp. In particular embodiments, the at least one strain of bacterium from the genus Veillonella is capable of converting lactate to propionate. In some embodiments, the one or more propionate producing strain of bacteria is or includes Veillonella agrestimuris, Veillonella alcalescens, Veillonella atypica, Veillonella caviae, Veillonella cricetid, Veillonella denticariosi, Veillonella dispar, Veillonella hominis, Veillonella infantium, Veillonella intestinalis, Veillonella magna, Veillonella montpellierensis, Veillonella nakazawae, Veillonella parvula, Veillonella ratti, Veillonella rodentium, Veillonella rogosae, Veillonella seminalis, or Veillonella tobetsuensis.
[0015] In certain embodiments, the propionate producing bacterium is or includes Veillonella atypica (V. atypica), Veillonella dispar (V. dispar), Veillonella infantium (V. infantium), Veillonella nakazawae (V. nakazawae), or Veillonella parvula (V. parvula). In some embodiments, the propionate producing bacterium is or includes V. infantium, V. dispar, or V. nakazawae. In some embodiments, the propionate producing bacterium is or includes V. parvula. In some embodiments, the propionate producing bacterium is or includes V. rogosae.
[0016] In some embodiments, the propionate producing bacterium is or includes a strain of Veillonella sp. that has or includes a nucleic acid of at least 200, 300, 400, 500, 600, 700, 800, 900, 1,000, 1,200, or 1,400 nucleotides in length with at least 85%, 90%, 95%, 97%, 97.5%, 98%, 99% or 100% sequence identity to any of SEQ ID NOS: 40-49. In particular embodiments, the propionate producing bacterium is or includes a strain of Veillonella sp. that has or includes a nucleic acid of at least 200, 300, 400, 500, 600, 700, 800, 900, 1,000, 1,200, or 1,400 nucleotides in length with at least 85%, 90%, 95%, 97%, 97.5%, 98%, 99% or 100% sequence identity to any of SEQ ID NOS: 40-43. In certain
embodiments, the propionate producing bacterium is or includes a strain of Veillonella sp. that has or includes a nucleic acid sequence with at least 85%, 90%, 95%, 97%, 97.5%, 98%, 99% or 100% sequence identity to any of SEQ ID NOS: 40-49. In certain embodiments, the propionate producing bacterium is or includes a strain of Veillonella sp. that has or includes a nucleic acid sequence with at least 85%, 90%, 95%, 97%, 97.5%, 98%, 99% or 100% sequence identity to any of SEQ ID NOS: 40-43.
[0017] In some embodiments, the propionate producing bacterium is or includes a strain of Veillonella sp. that has or includes a nucleic acid sequence encoding an rpoB gene having a sequence of at least 200, 300, 400, 500, 600, 700, 800, 900, 1,000, 1,200, or 1,400 nucleotides in length with at least 95%, 97%, 97.5%, 98%, 99% or 100% sequence identity to SEQ ID NO: 40 or 41. In particular embodiments, the propionate producing bacterium is or includes a strain of Veillonella sp. that has or includes a nucleic acid sequence encoding an rpoB gene with at least 95%, 97%, 97.5%, 98%, 99% or 100% sequence identity to SEQ ID NO: 40 or 41.
[0018] In particular embodiments, the propionate producing bacterium is or includes a strain of Veillonella sp. that has or includes a nucleic acid sequence encoding an 16S gene having a sequence of at least 200, 300, 400, 500, 600, 700, 800, 900, 1,000, 1,200, or 1,400 nucleotides in length with at least 95%, 97%, 97.5%, 98%, 99% or 100% sequence identity to any of SEQ ID NOS: 42-49. In certain embodiments, the propionate producing bacterium is or includes a strain of Veillonella sp. that has or includes a nucleic acid sequence encoding an 16S gene with at least 95%, 97%, 97.5%, 98%, 99% or 100% sequence identity to any of SEQ ID NOS: 42-49. In some embodiments, the propionate producing bacterium is or includes a strain of Veillonella sp. that has or includes a nucleic acid sequence encoding an 16S gene having a sequence of at least 200, 300, 400, 500, 600, 700, 800, 900, 1,000, 1,200, or 1,400 nucleotides in length with at least 95%, 97%, 97.5%, 98%, 99% or 100% sequence identity to SEQ ID NO: 42 or 43. In certain embodiments, the propionate producing bacterium is or includes a strain of Veillonella sp. that has or includes a nucleic acid sequence encoding an 16S gene with at least 95%, 97%, 97.5%, 98%, 99% or 100% sequence identity to SEQ ID NO: 42 or 43.
[0019] In certain embodiments, the propionate producing bacterium is or includes a strain of Veillonella sp. that has or includes a nucleic acid sequence encoding an rpoB gene with at least 97%, 97.5%, 98%, 99% or 100% sequence identity to SEQ ID NO: 40 and a nucleic acid sequence encoding a 16S gene with at least 97%, 97.5%, 98%, 99% or 100%
sequence identity to SEQ ID NO: 42. In some embodiments, the propionate producing bacterium is or includes a strain of Veillonella sp. that has or includes a nucleic acid sequence encoding an rpoB gene with at least 97%, 97.5%, 98%, 99% or 100% sequence identity to SEQ ID NO: 41 and a nucleic acid sequence encoding a 16S gene with at least 97%, 97.5%, 98%, 99% or 100% sequence identity to SEQ ID NO: 43.
[0020] In some embodiments, the propionate producing bacterium is or includes a strain of bacteria that has, includes, or expresses a methylmalonyl-CoA decarboxylase subunit alpha and/or a methylmalonyl-CoA decarboxylase subunit beta protein and/or has or includes one or more genes encoding a methylmalonyl-CoA decarboxylase subunit alpha and/or a methylmalonyl-CoA decarboxylase subunit beta. In particular embodiments, the methylmalonyl-CoA decarboxylase alpha and beta subunits are from and/or are identical to alpha and beta subunits found in and/or expressed by a Veillonella sp. In certain embodiments, the propionate producing bacterium is or includes a strain of bacteria that has, includes, or expresses a protein having an amino acid sequence with at least 85%, 90%, 95%, 98%, or 99% identity to SEQ ID NO: 50 or 51. In particular embodiments, the propionate producing bacterium is or includes a strain of bacteria that has, includes, or expresses a protein having an amino acid sequence with 100% identity to SEQ ID NO: 50 or 51.
[0021] In certain embodiments, the at least one propionate producing bacteria is or includes at least one strain of bacterium from the genus Megasphaera. In particular embodiments, the at least one strain of bacterium from the genus Megasphaera is capable of converting lactate to propionate. In some embodiments, the at least one strain of bacterium from the genus Megasphaera is capable of converting lactate to propionate and butyrate. In particular embodiments, the at least one propionate producing bacterium is or includes at least one strain of Megasphaera sp. In some embodiments, the propionate producing bacterium is or includes Megasphaera elsdenii (M. elsdenii), Megasphaera hominis (M hominis), Megasphaera indica (M indica), Megasphaera massiliensis (M.massiliensis), and/ or Megasphaera micronuciformis (M micromiciformis). In certain embodiments, the propionate producing bacterium is or includes Megasphaera elsdenii and/ or Megasphaera massiliensis.
[0022] In some embodiments, the propionate producing bacterium is or includes a strain of Megasphaera sp. that has or includes a nucleic acid sequence encoding an 16S gene having a sequence of at least 200, 300, 400, 500, 600, 700, 800, 900, 1,000, 1,200, or 1,400 nucleotides in length with at least 95%, 97%, 97.5%, 98%, 99% or 100% sequence identity to
SEQ ID NO: 52 or 53. In particular embodiments, the propionate producing bacterium is or includes a strain of Megasphaera sp. that has or includes a nucleic acid sequence encoding an 16S gene with at least 95%, 97%, 97.5%, 98%, 99% or 100% sequence identity to SEQ ID NO: 52 or 53.
[0023] In some embodiments, the at least one propionate producing bacteria is or includes at least one strain of bacterium from the genus Anaerotignum. In some embodiments, the at least one strain of bacterium from the genus Anaerotignum is capable of converting lactate to propionate and butyrate. In particular embodiments, the at least one propionate producing bacterium is or includes at least one strain of Anaerotignum sp. In some embodiments, the propionate producing bacterium is or includes Anaerotignum lactatifermentans (A. lactatifermentans).
[0024] In some embodiments, the propionate producing bacterium is or includes a strain of Anaerotignum sp. that has or includes a nucleic acid sequence encoding an 16S gene having a sequence of at least 200, 300, 400, 500, 600, 700, 800, 900, 1,000, 1,200, or 1,400 nucleotides in length with at least 95%, 97%, 97.5%, 98%, 99% or 100% sequence identity to SEQ ID NO: 54. In particular embodiments, the propionate producing bacterium is or includes a strain of Anaerotignum sp. that has or includes a nucleic acid sequence encoding an 16S gene with at least 95%, 97%, 97.5%, 98%, 99% or 100% sequence identity to SEQ ID NO: 54.
[0025] In some embodiments, the at least one propionate producing bacteria is or includes at least one strain of bacterium from the genus Bacteroides. In some embodiments, the at least one strain of bacterium from the genus Bacteroides is capable of converting lactate to propionate and butyrate. In particular embodiments, the at least one propionate producing bacterium is or includes at least one strain of Bacteroides sp. In some embodiments, the propionate producing bacterium is or includes Bacteroides fragilis (B. fragilis and/ or Bacteroides caccae (B. caccae).
[0026] In some embodiments, the propionate producing bacterium is or includes a strain of Bacteroides sp. that has or includes a nucleic acid sequence encoding an 16S gene having a sequence of at least 200, 300, 400, 500, 600, 700, 800, 900, 1,000, 1,200, or 1,400 nucleotides in length with at least 95%, 97%, 97.5%, 98%, 99% or 100% sequence identity to SEQ ID NO: 55 and/or 57. In particular embodiments, the propionate producing bacterium is or includes a strain of Bacteroides sp. that has or includes a nucleic acid sequence encoding
an 16S gene with at least 95%, 97%, 97.5%, 98%, 99% or 100% sequence identity to SEQ ID NO: 55 and/or 57.
[0027] In some embodiments, the at least one propionate producing bacteria is or includes at least one strain of bacterium from the genus Coprococcus. In some embodiments, the at least one strain of bacterium from the genus Coprococcus is capable of converting lactate to propionate. In particular embodiments, the at least one propionate producing bacterium is or includes at least one strain of Coprococcus sp. In some embodiments, the propionate producing bacterium is or includes Coprococcus catus (C catus).
[0028] In some embodiments, the propionate producing bacterium is or includes a strain of Coprococcus sp. that has or includes a nucleic acid sequence encoding an 16S gene having a sequence of at least 200, 300, 400, 500, 600, 700, 800, 900, 1,000, 1,200, or 1,400 nucleotides in length with at least 95%, 97%, 97.5%, 98%, 99% or 100% sequence identity to SEQ ID NO: 56. In particular embodiments, the propionate producing bacterium is or includes a strain of Coprococcus sp. that has or includes a nucleic acid sequence encoding an 16S gene with at least 95%, 97%, 97.5%, 98%, 99% or 100% sequence identity to SEQ ID NO: 56.
[0029] In certain embodiments, the at least one propionate producing bacteria is or includes at least one strain of bacterium from the genus Merdimmobilis. In some embodiments, the at least one strain of bacterium from the genus Merdimmobilis is capable of converting lactate to propionate. In particular embodiments, the at least one propionate producing bacterium is or includes at least one strain of Merdimmobilis sp. In some embodiments, the propionate producing bacterium is or includes Merdimmobilis hominis).
[0030] In some embodiments, the propionate producing bacterium is or includes a strain of Merdimmobilis sp. that has or includes a nucleic acid sequence encoding an 16S gene having a sequence of at least 200, 300, 400, 500, 600, 700, 800, 900, 1,000, 1,200, or 1,400 nucleotides in length with at least 95%, 97%, 97.5%, 98%, 99% or 100% sequence identity to SEQ ID NO: 58. In particular embodiments, the propionate producing bacterium is or includes a strain of Merdimmobilis sp. that has or includes a nucleic acid sequence encoding an 16S gene with at least 95%, 97%, 97.5%, 98%, 99% or 100% sequence identity to SEQ ID NO: 58.
[0031] In particular embodiments, the at least one propionate producing bacteria is or includes at least one strain of bacterium from the genus Blautia. In some embodiments, the at least one strain of bacterium from the genus Blautia is capable of converting human milk
oligosaccharides and/or 1,2-propanediol to propionate and/or butyrate. In particular embodiments, the at least one propionate producing bacterium is or includes at least one strain of Blautia sp. In some embodiments, the propionate producing bacterium is or includes Blautia obeum.
[0032] In various embodiments, the at least one propionate producing bacteria is or includes at least one strain of bacterium from the genus Ruminococcus. In certain embodiments, the at least one strain of bacterium from the genus Ruminococcus is capable of converting human milk oligosaccharides and/or 1,2-propanediol to propionate and/or butyrate. In particular embodiments, the at least one propionate producing bacterium is or includes at least one strain of Ruminococcus sp. In some embodiments, the propionate producing bacterium is or includes Ruminococcus torques.
[0033] In particular embodiments, the at least one propionate producing bacteria is or includes at least one strain of bacterium from the genus Roseburia. In certain embodiments, the at least one strain of bacterium from the genus Roseburia is capable of converting human milk oligosaccharides and/or 1,2-propanediol to propionate and/or butyrate. In particular embodiments, the at least one propionate producing bacterium is or includes at least one strain of Roseburia sp. In some embodiments, the propionate producing bacterium is or includes Roseburia inulinivorans .
B. Bifidobacteria
[0048] In particular embodiments, provided herein are compositions that are or include at least one strain of the genus Bifidobacteria, e.g., a strain of Bifidobacteria capable of consuming human milk oligosaccharides such as B. longum subsp. infantis. In certain embodiments, Bifidobacteria is or includes two or more strains or species of Bifidobacteria, such as any two or more of those described herein, e.g., in Section I-B. Some suitable Bifidobacteria strains may be described in PCT App. Pub. No. W02021061991, hereby incorporated by reference in its entirety.
[0049] In particular embodiments, the at least one strain of Bifidobacterium is capable of consuming or metabolizing non-digestible carbohydrates, e.g., oligosaccharides such as human milk oligosaccharides. In particular embodiments, at least one strain of Bifidobacterium is capable of consuming or metabolizing oligosaccharides such as human milk oligosaccharides. In some embodiments, at least one strain of Bifidobacterium is capable of utilizing the prebiotics of the prebiotic mixture, e.g., human milk oligosaccharides,
as a carbon source. In particular embodiments, human milk oligosaccharides are preferentially consumed or metabolized by the at least one strain of Bifidobacterium, e.g., as compared to other microbes or bacteria present in the gut or microbiome. In certain embodiments, the at least one strain of Bifidobacterium is capable of consuming or metabolizing one or more prebiotics of the mixture, including those of any of the prebiotic mixtures described herein, e.g., in Section I-C. In certain embodiments, the at least one strain of Bifidobacterium is capable of consuming or metabolizing all or essentially all of the oligosaccharides of the prebiotic mixture. In certain embodiments, the at least one strain of Bifidobacterium is capable of consuming or metabolizing human milk oligosaccharides.
Particular embodiments contemplate that strains of Bifidobacteria that consume or metabolize human milk oligosaccharides are known and may be identified by routine techniques such as those described in Gotoh et al. Sci Rep. 2018 Sep 18;8(1): 13958, incorporated by reference herein in its entirety.
[0050] In some embodiments, the at least one strain of Bifidobacterium contains one or more enzymes capable of hydrolyzing the prebiotics of the mixture. In certain embodiments, the at least one strain of Bifidobacterium contains one or more enzymes capable of hydrolyzing the human milk oligosaccharides. In particular embodiments, the one or more enzymes hydrolyze external oligosaccharides, e.g., oligosaccharides such as human milk oligosaccharides that are outside of the Bifidobacterial cell. In some embodiments, the one or more enzymes hydrolyze oligosaccharides such as human milk oligosaccharides internally or within the Bifidobacterial cell. In certain embodiments, the one or more enzymes hydrolyze internalized human milk oligosaccharides.
[0051] In particular embodiments, the at least one strain of Bifidobacterium contains one or more enzymes capable of hydrolyzing one or more human milk oligosaccharides. In particular embodiments, the one or more enzymes hydrolyze external human milk oligosaccharides. In some embodiments, the one or more enzymes hydrolyze human milk oligosaccharides that are outside of the Bifidobacterial cell. In some embodiments, the one or more enzymes hydrolyze human milk oligosaccharides internally. In particular embodiments, the one or more enzymes hydrolyze human milk oligosaccharides within the probiotic cell. In certain embodiments, the one or more enzymes hydrolyze internalized human milk oligosaccharides.
[0052] In some embodiments, the at least one strain of Bifidobacterium is capable of internalizing human milk oligosaccharides. In certain embodiments, the at least one strain of
Bifidobacterium internalizes human milk oligosaccharides prior to hydrolyzing the human milk oligosaccharides. In various embodiments, the at least one strain of Bifidobacterium selectively or exclusively utilizes human milk oligosaccharides as a carbon source. Thus, in certain embodiments, if the at least one strain of Bifidobacterium is administered to the subject and/or has engrafted, e.g., within the subject’s microbiome (such as the intestinal microbiome), the at least one strain of Bifidobacterium is present, expands, or increases in amount within the subject’s microbiome when human milk oligosaccharides are administered to and/or ingested by the subject, and, in certain embodiments, the at least one strain of Bifidobacterium is no longer present and/or decreases in amount within the subject’s microbiome when the human milk oligosaccharides are no longer ingested or administered.
[0053] In some embodiments, the at least one strain of Bifidobacterium is capable of internalizing oligosaccharides, such as to consume or metabolize the oligosaccharides. In certain embodiments, the at least one strain of Bifidobacterium is capable of internalizing one or more oligosaccharides of the mixture, including those of any of the oligosaccharides or mixtures described herein, e.g., in Section I-C. In certain embodiments, the probiotic strain is capable of internalizing human milk oligosaccharides.
[0054] In particular embodiments, the at least one strain of Bifidobacterium is or includes a strain of B. longum subsp. infantis, B. adolescentis, B. animalis subsp. animalis, B. animalis subsp. lactis B. bifidum, B. breve, B. catenulatum, B. longum subsp. longum, B. pseudocatanulatum, or B. pseudoIongum.
[0055] In particular embodiments, the species or subspecies of a given strain of Bifidobacteria may be identified by routine techniques. For example, in some embodiments, the species or subspecies is identified by assessing the sequence similarity of one or more genes to corresponding sequences of known members of bacterial species or subspecies. In certain embodiments, a probiotic strain falls within a species or subspecies if all or a portion of its 16S gene has at least 97% sequence identity to all or a portion of a known 16S sequence of a known strain falling within the species. In particular embodiments, a probiotic strain falls within a species or subspecies if all or a portion of its 16S gene has at least 97% sequence identity to all or a portion of a known 16S sequence of a known strain falling within the species. Exemplary full or partial 16S sequences are summarized in Table 1.
[0056] In certain embodiments, the at least one strain of Bifidobacterium has or includes a nucleic acid sequence with at least 97%, at least 98%, at least 99%, or at least 99.5% identity to a nucleic acid sequence set forth in any of SEQ ID NOS: 1-16 or 20-39. In particular embodiments, the at least one strain of Bifidobacterium. In certain embodiments, the at least one strain of Bifidobacterium has or includes a nucleic acid sequence with at least 97%, at least 98%, at least 99%, or at least 99.5% identity to a nucleic acid sequence set forth in any of SEQ ID NOS: 1-7 or 20-39. In some embodiments, the at least one strain of Bifidobacterium has or includes a nucleic acid sequence with at least 97%, at least 98%, at least 99%, or at least 99.5% identity to a nucleic acid sequence set forth in any of SEQ ID NOS: 1-7 or 11. In certain embodiments, the at least one strain of Bifidobacterium has or includes a nucleic acid sequence with at least 97%, at least 98%, at least 99%, or at least 99.5% identity to a nucleic acid sequence set forth in any of SEQ ID NOS: 1-16. In particular embodiments, the at least one strain of Bifidobacterium has or includes a nucleic acid sequence with at least 97%, at least 98%, at least 99%, or at least 99.5% identity to a nucleic acid sequence set forth in any of SEQ ID NOS: 1-7.
[0057] In particular embodiments, the at least one strain of Bifidobacterium is or includes a strain of B. longum subsp. infantis. In particular embodiments, the strain of B. longum subsp. infantis has or includes a nucleic acid sequence with at least 97%, at least
98%, at least 99%, or at least 99.5% identity to a nucleic acid sequence set forth in any of SEQ ID NOS: 1-7 or 20-39. In particular embodiments, the strain of B. longum subsp. infantis has or includes a nucleic acid sequence of at least 200, 300, 400, 500, 600, 700, 800, 900, 1,000, 1,200, or 1,500 nucleotides in length with at least 60%, 70%, 80%, 90%, 95%, 99%, or 99.9% sequence identity to a nucleic acid sequence set forth in SEQ ID NOS: 1-7 or 20-39. In some embodiments, the strain of B. longum subsp. infantis has or includes nucleic acid sequences having at least 90%, 95%, or 99% sequence identity to one or more of the nucleic acid sequences set forth in SEQ ID NOS: 20-39. In some embodiments, the strain of B. longum subsp. infantis has or includes the nucleic acid sequences set forth in one or more of SEQ ID NOS: 20-39. In particular embodiments, the strain of B. longum subsp. infantis has or includes nucleic acid sequences having at least 90%, 95%, or 99% sequence identity to all of the nucleic acid sequences set forth in SEQ ID NOS: 20-39. In various embodiments, the strain of B. longum subsp. infantis has or includes the nucleic acid sequences set forth in SEQ ID NOS: 20-39.
[0058] In some embodiments, at least one strain of Bifidobacterium capable of consuming, metabolizing, and/or internalizing human milk oligosaccharides. In some aspects, HMO cannot be metabolized by the host, e.g., mammals such as humans, or most bacteria, including most bacteria commonly found in the microbiome of adult humans. In particular aspects, some strains, species, or subspecies of Bifidobacterium, such as B. longum subsp. infantis, have enzymatic activity able to degrade specific alpha and beta bonds of human milk oligosaccharides. Five monosaccharides can be found in different HMO structures, glucose, galactose, N-acetyl glucosamine, fucose, and sialic acid (also referred to herein as N-acetyl neuraminic acid). Some strains, species, or subspecies of Bifidobacterium are able to fully degrade HMO intracellularly. Such Bifidobacterium possess genes encoding specific transporters (e.g., ABC transporters such as those described in Sela et al. PNAS (2008) 105 (48) 18964-18969; Schell, et al. PNAS. (2002) 99(22): 14422-14427 and LoCascio et al. Appl Environ Microbiol. (2010) 76(22):7373-81), incorporated by reference herein, that selectively transport or import HMO and enzymes necessary for HMO degradation (alpha-fucosidase, alpha-sialidase, beta-galactosidase, and beta-N- hexosaminidase). Other Bifidobacterium strains, such as for example !?, bifidum, degrades HMO externally or extracellularly, such as for example by lacto-N-biosidase, which cleaves LNB from HMO. The LNB is then internalized by a transporter and degraded by LNB- phosphorylase. In some embodiments, the probiotic strain is at least one strain of bacterium
having one or more genes encoding all or a portion of a transporter, e.g., an ABC transporter, capable of internalizing an oligosaccharide such as an HMO. In particular embodiments, the probiotic strain is a bacterium having one or more genes encoding one or more enzymes, e.g., alpha-fucosidase, alpha-sialidase, beta-galactosidase, and beta-N-hexosaminidase, capable degrading an oligosaccharide such as an HMO. In certain embodiments, the probiotic strain is at least one strain of Bifidobacterium or Bacteroides having one or more genes encoding all or a portion of a transporter, e.g., an ABC transporter, capable of internalizing an oligosaccharide, e.g., an HMO.
[0059] In some embodiments, the at least one strain of Bifidobacterium is B. longum subsp. infantis. Particular embodiments contemplate that B. longum subsp. infantis is known and readily identifiable by those of skill in the art using routine techniques. In some embodiments, B. longum subsp. infantis, including its genome and biology, are known and for example have been described, including in Sela et al. PNAS (2008) 105 (48) 18964- 18969; Underwood et al., Pediatr Res. (2015) 77(0): 229-235, incorporated by reference herein. In certain embodiments, Bifidobacterium, e.g., B. longum subsp. infantis, may be isolated using known selective microbiological media, e.g., De Man, Rogosa and Sharpe agar (MRS), optionally in combination with mupirocin, or those described in O’Sullivan et al., J Appl Microbiol. 2011 Aug;l 11(2):467-73, incorporated by reference herein. In some embodiments, suitable sources for isolating Bifidobacterium, e.g., B. longum subsp. infantis, are known, and include stool samples obtained from breast fed infants. In certain embodiments, bacterial colonies may be identified or characterized by routine biochemical techniques, such as PCR. In some embodiments, B. longum subsp. infantis is identified by taqman qPCR, such as described in Lawley et al., PeerJ. 2017 May 25;5:e3375. e.g., as performed with forward primer sequence ATACAGCAGAACCTTGGCCT (SEQ ID NO: 17), reverse primer sequence GCGATCACATGGACGAGAAC (SEQ ID NO: 18) and probe sequence [FAM dye] -TTTCACGGA - [ZEN quencher] - TCACCGGACCATACG - [3IABkFQ quencher] (SEQ ID NO: 19). In some aspects, a strain may be confirmed as B. longum subsp. infantis by observing growth when human milk oligosaccharides are provided as the sole carbon source, such as with an assay described in Gotoh et al. Sci Rep. 2018 Sep 18;8(1): 13958, incorporated by reference herein.
C. Prebiotic mixtures
[0034] In some embodiments, the prebiotic mixture is a mixture of non-digestible carbohydrates, e.g., oligosaccharides such as human milk oligosaccharides (HMOs), that promotes the growth or expansion of the probiotic strain, e.g., in vivo such as in the human gut and/or within the human gut microbiome. In certain embodiments, the prebiotic mixture, e.g., of non-digestible carbohydrates such as human milk oligosaccharides, promotes, e.g., selectively or exclusively, the colonization, expansion, extension, or increased presence of the probiotic strain within the microbiome. In particular embodiments, the mixture of non- digestible carbohydrates, e.g., human milk oligosaccharides, promotes the growth or expansion of a probiotic strain of Bifidobacterium such as B. longum subsp. infantis, e.g., in vivo such as in the human gut. In certain embodiments, the prebiotic mixture is a mixture of oligosaccharides, e.g., human milk oligosaccharides, that promote, e.g., selectively or exclusively, the colonization, expansion, extension, or increased presence of one or more strains of Bifidobacterium, e.g., B. longum subsp. infanlis, within the microbiome.
[0035] In some embodiments, the prebiotic mixture is or includes a mixture of non- digestible carbohydrates. In various embodiments, the prebiotic mixture is or includes a mixture of oligosaccharides. In particular embodiments, the prebiotic mixture is a mixture of one or more human milk oligosaccharides.
[0036] In some embodiments, the non-digestible carbohydrates are or include oligosaccharides. In particular embodiments, the non-digestible carbohydrates are or include milk oligosaccharides. In certain embodiments, the non-digestible carbohydrates are or include human milk oligosaccharides (human milk oligosaccharides). In some embodiments, the prebiotic mixture is a mixture of non-digestible carbohydrates that are or include human milk oligosaccharides. In particular embodiments, the prebiotic mixture is a mixture of human milk oligosaccharides, such as those that are obtained or derived from permeate, e.g., permeate derived or obtained from pooled human milk.
[0037] In some embodiments, the provided mixture may contain any oligosaccharide that may be internalized by one or more strains of Bifidobacterium such as a strain of B. longum subsp. infantis. In some embodiments, the oligosaccharides of the mixture may include one or more of a fructo-oligosacharide (FOS), galactooligosaccharide (GOS), transgalactooligosaccharide (TOS), gluco-oligosaccharide, xylo-oligosaccharide (XOS), chitosan oligosaccharide (COS), soy oligosaccharide (SOS), isomalto-oligosaccharide (IMOS), or derivatives thereof. In certain embodiments, such derivatives include those with
modifications that may increase the likelihood or probability of consumption, metabolism, and/or internalization (such as by transport or import) of the oligosaccharide by the probiotic strain, e.g., B. longum subsp. infantis. Such modifications may include but are not limited to fucosylation or sialylation. In some embodiments, the oligosaccharides of the mixture may include one or more of a FOS, GOS, TOS, gluco-oligosaccharide, XOS, COS, SOS, IMOS, or derivatives or any or all of the foregoing, that are capable of being metabolized, consumed, and/or internalized by one or more strains, species, or subspecies of Bifidobacterium, e.g., B. longum subsp. infantis. In certain embodiments, the oligosaccharides of the mixture include one or more oligosaccharides that are obtained or derived from a resistant starch, pectin, psyllium, arabinogalactan, glucomannan, galactomannan, xylan, lactosucrose, lactulose, lactitol and various other types of gums such as tara gum, acacia, carob, oat, bamboo, citrus fibers, such as by treatment with enzymes that hydrolyze fiber or polysaccharides. In some embodiments, the one or more oligosaccharides of the mixture that are obtained by these means are capable of being consumed, metabolized, and/or internalized by at least one strain of Bifidobacterium such as B. longum subsp. infantis.
[0038] In certain embodiments, the prebiotic mixture is a mixture that is or includes at least one HMO. In some embodiments, the prebiotic mixture is a mixture of human milk oligosaccharides that is or includes a plurality of human milk oligosaccharides. In some embodiments, the prebiotic mixture is or includes a plurality of, of about, or at least 2, 3, 5, 10, 25, 50, 75, 100, 125, 150 different individual human milk oligosaccharides, e.g., human milk oligosaccharides with different individual chemical formulas or chemical structures. In certain embodiments, the prebiotic mixture is or includes a plurality of, of about, or at least 10, 25, 50, 75, 100, 125, 150 different individual human milk oligosaccharides. In some embodiments, the prebiotic mixture is or includes a plurality of, of about, or at least 25 different individual human milk oligosaccharides. In some embodiments, the prebiotic mixture is or includes a plurality of, of about, or of at least 80 different individual human milk oligosaccharides. Particular embodiments contemplate that one of skill may determine if an oligosaccharide is an HMO, such as if the oligosaccharide has a chemical formula and structure that is identical to an oligosaccharide that is found in human milk, as a matter of routine.
[39] In certain embodiments, the prebiotic mixture contains one or more synthetic human milk oligosaccharides, e.g., human milk oligosaccharides that are obtained, purified, or synthesized from a source other than human milk. In some aspects, synthetic human milk
oligosaccharides, as well as methods for synthesizing oligosaccharides and human milk oligosaccharides, are known, and include but are not limited to those described in PCT Publication Nos.: W02017101958, WO2015197082, WO2015032413, WO2014167538, WO2014167537, WO2014135167, W02013190531, W02013190530, WO2013139344, WO2013182206, WO2013044928, W02019043029, W02019008133, WO2018077892, WO2017042382, WO2015150328, WO2015106943, WO2015049331, WO2015036138, and W02012097950, each of which is incorporated by reference herein in its entirety.
[40] In some embodiments, the prebiotic mixture includes some or all of 2'- fucosyllactose, 3’ -fucosy llactose, 3’-sialyllactose, 6'-sialyllactose, Lacto-N-tetraose, lacto-N- difucohexaose I, lactodifucotetraose, lacto-N-fucopentaose I, sialylacto-N-tetraose c, sialylacto-N-tetraose b, and disialyllacto-N-tetraose. In particular embodiments, the mixture includes all of 2'-fucosyllactose, 3’-fucosyllactose, 3’-sialyllactose, 6'-sialyllactose, lacto-N- tetraose, lacto-N-difucohexaose I, lactodifucotetraose, lacto-N-fucopentaose I, sialylacto-N- tetraose c, sialylacto-N-tetraose b, and disialyllacto-N-tetraose.
[41] In certain embodiments, the prebiotic mixture includes some or all of 2- fucosyllactose, lacto-N-tetraorose, 3-sialyllactose, 3-fucosyllactose, lacto-N-fucopentaose I, lacto-N-fucopentaose II, and 6’sialyllactose. In particular embodiments, the prebiotic mixture includes some or all of 2'-fucosyl-lactose, 3’-fucosyl-lactose, 3’-sialyl-lactose, 6'- sialyl-lactose, lacto-N-tetraose, lacto-N-neo-tetraose, lacto-N-fucopentaose I, lacto-N- fucopentaose II, lacto-N-fucopentaose III, sialyl-lacto-N-tetraose b, sialyl-lacto-N-tetraose c, lacto-N-difuco-hexaose I, lacto-N-difuco-hexaose II, lacto-N-hexaose, para-lacto-N-hexaose, disialyllacto-N-tetraose, Fucosyl-lacto-N-hexaose, difucosyl-lacto-N-hexaose a, and difucosyl-lacto-N-hexaose b.
[0042] In certain embodiments, the prebiotic mixture contains at least 25, 50, 100, 125, or 150 human milk oligosaccharides which include all of 2-fucosyllactose, lacto-N- tetraorose, 3-sialyllactose, 3-fucosyllactose, lacto-N-fucopentaose I, lacto-N-fucopentaose II, and 6’sialyllactose. In particular embodiments, the prebiotic mixture contains at least 25, 50, 100, 125, or 150 human milk oligosaccharides which include all of 2'-fucosyl-lactose, 3’- fucosyl-lactose, 3’-sialyl-lactose, 6'-sialyl-lactose, lacto-N-tetraose, lacto-N-neo-tetraose, lacto-N-fucopentaose I, lacto-N-fucopentaose II, lacto-N-fucopentaose III, sialyl-lacto-N- tetraose b, sialyl-lacto-N-tetraose c, lacto-N-difuco-hexaose I, lacto-N-difuco-hexaose II, lacto-N-hexaose, para-lacto-N-hexaose, disialyllacto-N-tetraose, Fucosyl-lacto-N-hexaose, difucosyl-lacto-N-hexaose a, and difucosyl-lacto-N-hexaose b. In some embodiments, the
prebiotic mixture is a mixture of human milk oligosaccharides that is, includes, or is obtained or derived from human milk or a fraction thereof. In certain embodiments, the prebiotic mixture is or includes a mixture of human milk oligosaccharides that is or is obtained from an ultra-filtered permeate from human skim milk. In various embodiments, the prebiotic mixture is, includes, or is derived or produced from a concentrated human milk permeate. In some embodiments, the mixture of human milk oligosaccharides or the concentrated human milk permeate is or is obtained from a process described herein, e.g., in Section I-C-i. In certain embodiments, the prebiotic mixture is a concentrated human milk permeate, such as those described in U.S. Pat. No. 8,927,027 or in PCT Application No. WO 2018053535, incorporated herein by reference. In some embodiment, the prebiotic mixture is a prebiotic mixture produced or resulting from any of the methods described herein, e.g., in Section I-C- (i).
[0043] In some embodiments, the prebiotic mixture is or includes a concentrated human milk permeate that contains a plurality of human milk oligosaccharides. In particular aspects, the human milk permeate is obtained by filtering human skim milk that was obtained by separating cream from whole human milk. In some embodiments, the permeate was ultrafiltered from the human skim milk. In particular embodiments, the permeate is further concentrated, e.g., by reverse osmosis or nanofiltration, to increase the HMO content within the permeate. In some embodiments, the human milk permeate, e.g., the concentrated human milk permeate, has a concentration of at least 0.5%, 1%, 2.5%, 5%, or 10% w/v HMO. The human milk permeate may undergo additional processing steps, such as to digest or remove sugars, e.g., lactose, prior to its formulation or incorporate as a prebiotic mixture.
[0044] In certain embodiments, the concentrated human milk permeate includes a plurality of, of about, or of at least 1, 2, 3, 5, 10, 25, 50, 75, 100, 125, 150 different individual human milk oligosaccharides, e.g., human milk oligosaccharides with different individual chemical formulas or chemical structures. In certain embodiments, the prebiotic mixture is or includes a plurality of, of about, or of at least 10, 25, 50, 75, 100, 125, 150 different individual human milk oligosaccharides. In some embodiments, the prebiotic mixture is or includes a plurality of, of about, or of at least 25 different individual human milk oligosaccharides. In some embodiments, the prebiotic mixture is or includes a plurality of, of about, or of at least 80 different individual human milk oligosaccharides. Thus, in some embodiments, the prebiotic mixture is or includes a concentrated human milk permeate and contains at least 10, 25, 50, 75, 100, 125, 150 different individual human milk
oligosaccharides. In particular embodiments, the concentrated human milk permeate contains at least 10, 25, 50, 100, 125, or 150 human milk oligosaccharides which include all of 2- fucosyllactose, lacto-N-tetraorose, 3-sialyllactose, 3-fucosyllactose, lacto-N-fucopentaose I, lacto-N-fucopentaose II, and 6’sialyllactose.
[0045] In certain embodiments, the concentrated human milk permeate and/or the prebiotic mixture has an increased amount, level, or concentration of one or more human milk oligosaccharides as compared to what is typically found human milk. In particular embodiments, the prebiotic mixture has an increased amount, level, or concentration of one or more human milk oligosaccharides as compared to what is typically found in untreated human milk permeate, e.g., permeate resulting from ultrafiltration of pooled human skim milk. In particular embodiments, the prebiotic mixture is or includes at least 25, 50, 75, 100, 125, 150, of the different human milk oligosaccharides found, present, or detected in pooled human milk or in permeate (e.g., permeate resulting from ultra-filtering skim) obtained from pooled human milk. In some embodiments, the prebiotic mixture is or includes at least 50%, 60%, 70%, 80%, 85%, 90%, 95%, 97%, 99%, or 99.9% of the different human milk oligosaccharides found, present, or detected in pooled human milk or in permeate (e.g., permeate resulting from ultra-filtering skim) obtained from pooled human milk. In certain embodiments, the prebiotic mixture is or includes at least 50%, 60%, 70%, 80%, 85%, 90%, 95%, 97%, 99%, or 99.9% of the individual human milk oligosaccharides that may be found, present, or detected across samples of human milk, e.g., samples of milk obtained from different individuals. In some embodiments, the prebiotic mixture of human milk oligosaccharides is or includes the same or substantially the same human milk oligosaccharides found, present, or detected in pooled human milk or in permeate (e.g., permeate resulting from ultra-filtering skim) obtained from pooled human milk. In certain embodiments the prebiotic mixture is or includes a human milk permeate resulting from the ultrafiltration of human whole or skim milk pooled from at milk collected from at least 10, 25, 50, or 100 individual human milk donors that is further concentrated, e.g., by nanofiltration or reverse osmosis, to increase the concentration of total HMO (e.g., by w/w).
[0046] In certain embodiments, the prebiotic mixture is free or essentially free of oligosaccharides that are not human milk oligosaccharides. In certain embodiments, the prebiotic mixture contains human milk permeate, e.g., concentrated human milk permeate, and one or more synthetic human milk oligosaccharides, e.g., one or more of synthetically derived 2'-fucosyl-lactose, 3’-fucosyl-lactose, 3’-sialyl-lactose, 6'-sialyl-lactose, lacto-N-
tetraose, lacto-N-neo-tetraose, lacto-N-fucopentaose I, lacto-N-fucopentaose II, lacto-N- fucopentaose III, sialyl-lacto-N-tetraose b, sialyl-lacto-N-tetraose c, lacto-N-difuco-hexaose I, lacto-N-difuco-hexaose II, lacto-N-hexaose, para-lacto-N-hexaose, disialyllacto-N-tetraose, Fucosyl-lacto-N-hexaose, difucosyl-lacto-N-hexaose a, and difucosyl-lacto-N-hexaose b.
[0047] Prebiotic mixtures containing human milk oligosaccharides for use in the compositions and methods disclosed herein may be obtained according to methods known in the art, including, but not limited to, chemical synthesis and purification from human milk. For example, processes to obtain HMO compositions from human milk are described below and are detailed in PCT Pub. Nos. WO/2010/065652 and WO/2018/053535, the contents of which are hereby incorporated in their entirety.
[0048] In some embodiments, the prebiotic mixture is a mixture of human milk oligosaccharides that are or are derived from a concentrated ultra-filtered human milk permeate, e.g., any ultra-filtered human milk permeate described herein such as in Section I- C-(i).
[0049] In some embodiments, the prebiotic mixtures are mixtures of human milk oligosaccharides having an HMO profile that is substantially similar both structurally and functionally to the profile of human milk oligosaccharides observed across the population of whole human milk. That is to say, in some aspects, since the prebiotic mixtures may be obtained from a source of human milk derived from a pool of donors rather than an individual donor, the array of human milk oligosaccharides will be more diverse than in any one typical individual, and will represent or more closely represent the spectrum of human milk oligosaccharides that are found among human milk across a population as opposed to the spectrum of human milk oligosaccharides that are found or typically found in the human milk produced by any particular individual. Thus, in some embodiments, the prebiotic mixture and the concentrated human milk permeate have more individual HMO species than what can be found in human milk obtained from an individual donor.
[0050] In some instances, the ratio of the amount or concentration of individual human milk oligosaccharide species to total human milk oligosaccharides of the prebiotic mixture or the concentrated human milk permeate may be different from what would be observed in whole human milk or pooled human milk.
[0051] In some embodiments, the prebiotic mixture is or includes a greater amount of different individual human milk oligosaccharides than the number of different individual human milk oligosaccharides found in human milk from an individual donor. In certain
embodiments, the prebiotic mixture includes at least 1, 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50 more individual human milk oligosaccharides than the number of different individual human milk oligosaccharides found in human milk from an individual donor. In particular embodiments, a prebiotic mixture is or includes a greater amount of different individual human milk oligosaccharides than the mean or median number of different individual human milk oligosaccharides found in a plurality of human milk samples from individual donors. In certain embodiments, the prebiotic mixture includes at least 1, 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50 more individual human milk oligosaccharides than the number of different individual human milk oligosaccharides found in human milk from an individual donor.
[0052] In some aspects, one of the biggest variables in HMO diversity derives from the mother’s Lewis blood group and specifically whether or not she has an active fucosyltrasferase 2 (FUT2) and/or fucosyltrasferase 3 (FUT3) gene. When there is an active FUT2 gene, an al -2 linked fucose is produced, whereas fucose residues are al -4 linked when the FUT3 gene is active. The result of this “secretor status” is, generally, that “secretors” (i.e. those with an active FUT2 gene) produce a much more diverse profile of human milk oligosaccharides dominated by al-2 linked oligosaccharides, whereas “non-secretors” (i.e. those without an active FUT2 gene) may comprise a more varied array of, for example al, -4 linked oligosaccharides (as compared to secretors), but comprise an overall decrease in diversity since they are unable to synthesize a major component of the secretor’ s HMO repertoire. In some embodiments, the prebiotic mixture includes human milk oligosaccharides that include al-2 linked fucose and human milk oligosaccharides that include al -4 linked fucose.
[0053] In some embodiments, the prebiotic mixture is or includes at least 5% total HMO (w/w). In particular embodiments, the prebiotic mixture is or includes least 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 15%, 20%, 25%, or 50% total HMO (w/w). In certain embodiments, the prebiotic mixture is or includes between 5% and 15%, 7.5% and 12.5%, 8% and 12%, 8.5% and 11%, or 8.4% and 10.6% total HMO (w/w). In certain embodiments, the prebiotic mixture is or includes between 8.5% and 11% total HMO (w/w). In some embodiments, the prebiotic mixture is or includes between 8.4% and 10.6% total HMO (w/w).
[0054] In some embodiments, the prebiotic mixture has a pH of between 4.0 and 5.5. In certain embodiments, the prebiotic mixture has less than 10%, 5%, 1%, or 0.1% lactose
(w/w). In some embodiments, the prebiotic mixture has less than 10%, 5%, 1%, or 0.1% glucose (w/w). In particular embodiments, the prebiotic mixture has less than 10%, 5%, 1%, or 0.1% galactose (w/w). In certain embodiments, the prebiotic mixture has less than 10% galactose, less than 10% glucose, and less than 0.1% lactose.
[0055] In some embodiments, the prebiotic mixture is a liquid formulation. In some embodiments, the prebiotic mixture is in powdered form, e.g., a lyophilized or spray dried composition. In certain embodiments, the prebiotic mixture is incorporated into a dosage form that is a separate composition from the Bifidobacterium strain. In some embodiments, the prebiotic mixture is incorporated into a dosage form that is a separate composition from the propionate producing strain. i) Exemplary methods for manufacturing prebiotic mixtures
[0056] In some embodiments, the prebiotic mixture is or includes human milk oligosaccharides (HMOs) obtained or purified from ultra-filtered permeate from donated human milk. In certain embodiments, the donated human milk is pooled to provide a pool of human milk. In some embodiments, a pool of human milk comprises milk from two or more (e.g., ten or more) donors. In certain embodiments, the pooled human milk contains milk from at least 50, 75, 100, 150, or 200 individual donors. In certain embodiments, the pooled human milk contains human milk from at least 100 individual donors or between 100 and 300 individual donors. In some embodiments, the pooled human milk contains milk from at least ten, at least twenty -five, at least fifty, at least seventy-five, at least one hundred, or at least one hundred fifty individual human milk donors.
[0057] In some embodiments, the human milk oligosaccharides that are contained or included in the prebiotic mixture are synthetic human milk oligosaccharides, such as those derived from non-human milk sources, e.g., derived or obtained as oligosaccharides or precursors from transgenic microorganisms and/or chemically synthesized. In some aspects, synthetic oligosaccharides and human milk oligosaccharides, as well as methods and techniques for synthesizing oligosaccharides and human milk oligosaccharides, are known, and include but are not limited to those described in PCT Publication Nos.: W02017101958, WO2015197082, WO2015032413, WO2014167538, WO2014167537, WO2014135167, WO2013190531, WO2013190530, WO2013139344, WO2013182206, WO2013044928, W02019043029, W02019008133, WO2018077892, WO2017042382, WO2015150328,
WO2015106943, WO2015049331, WO2015036138, and W02012097950, each of which is incorporated by reference herein in its entirety.
[0058] In particular embodiments, one or more synthetically derived human milk oligosaccharides are added to a concentrated human milk permeate to arrive at a prebiotic mixture.
[0059] In certain embodiments, the mixture of oligosaccharides described herein are produced from human milk permeate, e.g., concentrated ultra-filtered permeate from pooled human milk. In some embodiments, the mixture of oligosaccharides described herein contain or are formulated with human milk permeate, e.g., concentrated ultra-filtered permeate from pooled human milk. In some embodiments, the concentrated ultra-filtered permeate may be made according to any suitable method or technique known in the art. In some aspects, suitable methods and techniques include those described in U.S. Pat. No. 8,927,027m PCT Pub. No. WO2018053535, and PCT Pub. No. WO 2021061991, hereby incorporated by reference in their entirety. Exemplary methods and techniques for producing the human milk compositions are briefly summarized herein.
[0060] In some embodiments, the prebiotic mixture is or includes human milk permeate that has been generated or produced by a method described herein. In some embodiments, whole human milk is pooled from multiple donors, and then cream and skim are separated by any suitable technique known in the art, e.g., centrifugation; and then the skim milk is filtered, e.g., ultra-filtered, to obtain human milk permeate and retentate. The human milk permeate may be further processed, such as to remove or digest one or more components, e.g., lactose, or to increase the concentration of human milk oligosaccharides, such as by nanofiltration or reverse osmosis.
[0061] In some embodiments, the donor milk is received frozen, and when desired, is thawed and pooled. In some embodiments, donor milk is then screened, e.g., to identify contaminants, by one or more of the methods discussed herein.
[0062] In some embodiments, the pooled milk is filtered, e.g., through about a 200- micron filter. In some embodiments, the pooled milk is heated, e.g., at about 63°C or greater for about 30 minutes or more. In some embodiments, the milk is transferred to a separator, e.g., a centrifuge, to separate the cream from the skim. In some embodiments, the cream may go through separation once again to yield more skim. In some embodiments, a desired amount of cream is added to the skim prior to ultra-filtration. In certain embodiments,
material that that did not pass through the filter is collected as the retentate fraction, and material that passes through the filter is collected as the permeate fraction.
[0063] In some embodiments, the skim fraction undergoes ultra-filtration. In some embodiments, the ultrafiltration is performed with a filter between 1 kDa and 1000 kDa to obtain a protein rich retentate and the HMO-containing permeate. Details of this process can be found, for example, in US 8,545,920; US 7,914,822; 7,943,315; 8,278,046; 8,628,921; and 9,149,052, each of which is hereby incorporated by reference in its entirety. In some embodiments, the ultra-filtration is performed with a filter that is between 1 kDa and 100 kDa, 5 kDa and 50 kDa, or 10 kDa and 25 kDa. In certain embodiments, the filter is about or at least 5 kDa, 10 kDa, 20 kDa, 25 kDa, 50 kDa, or 100 kDa. In some embodiments, the skim fraction undergoes ultrafiltration with a filter that is about 10 kDa. In certain embodiments, the skim fraction undergoes ultrafiltration with a filter that is about 25 kDa. In particular embodiments, the skim fraction undergoes ultrafiltration with a filter that is about 50 kDa.
[0064] In some embodiments, the ultra-filtered permeate undergoes a process for reducing lactose. In certain embodiments, a process for producing a concentrated human milk permeate composition with substantially reduced levels of lactose is provided. In certain embodiments, the substantial reduction includes or requires the biochemical and/or enzymatic removal of lactose from the lactose-rich human milk permeate fraction, without loss of yield or change in molecular profile of the HMO content of human milk permeate. And, in particular embodiments, without leaving residual inactivated foreign protein, if enzymatic digestion is used to reduce lactose. In certain embodiments, about or at least 50%, 60%, 70%, 80%, 90%, 95%, 99%, 99.9%, or 99.99% of the lactose present in the permeate after ultrafiltration is removed, e.g., enzymatically digested. In certain embodiments, the permeate is free or essentially free of lactose following the enzymatic digestion.
[0065] In certain embodiments, the process for reducing lactose from human milk permeate includes one or more of the steps of a) adjusting the pH of the permeate mixture; b) heating the pH adjusted mixture; c) adding lactase enzyme to the heated permeate mixture to create a permeate/lactase mixture and incubating a period of time; d) removing the lactase from the mixture and filtering the mixture to remove lactase; and e) concentrating human milk oligosaccharides. In some embodiments, the order of when steps (a)-(c) are performed may be varied. Thus, in some aspects, the steps may be performed in the order of (a)-(b)-(c); (a)-(c)-(b); (c)-(b)-(a); (c)-(a)-(b); (b)-(a)-(c); or (b)-(c)-(a), such that, for example, the lactase
enzyme may be added prior to heating the mixture, or, alternatively at any point during the heating process. Similarly, and also by way of example only, the mixture may be heated prior to adjustment of the pH. Furthermore, several steps may be grouped into a single step, for example “enzymatically digesting lactose” or “lactases digestion of lactose” involves steps (a)-(c) as described. These steps may be performed concurrently or consecutive in any order. Therefore, as used herein “lactose digestion” refers to the performance of at least these three steps, in any order, consecutively or concurrently.
[0066] In certain embodiments, the pH of the permeate is adjusted to a pH of about 3 to about 7.5. In some embodiments, the pH is adjusted to a pH of about 3.5 to about 7.0. In particular embodiments, the pH is adjusted to a pH of about 3.0 to about 6.0. In certain embodiments, the pH is adjusted to a pH of about 4 to about 6.5. In some embodiments, pH is adjusted to a pH of about 4.5 to about 6.0. In particular embodiments, the pH is adjusted to a pH of about 5.0 to about 5.5. In certain embodiments, the pH is adjusted to a pH of about 4.3 to about 4.7, preferably 4.5. The pH may be adjusted by adding acid or base. In some embodiments, pH is adjusted by adding acid, for example HC1. In particular embodiments, pH is adjusted by adding IN HC1 and mixing for a period of time, e.g., about 15 minutes.
[0067] In some embodiments, the pH-adjusted permeate is heated to a temperature of about of about 25°C to about 60°C. In certain embodiments, the permeate is heated to a temperature of about 30°C to about 55°C. In some embodiments, the permeate is heated to a temperature of about 40°C to about 50°C. In certain embodiments, the permeate is heated to a temperature of about 48°C to about 50°C. In some embodiments, the permeate is heated to a temperature about 50°C. In some embodiments, the permeate is heated to a temperature less than or equal to about 40°C.
[0068] In particular embodiments, lactase enzyme is added to the pH-adjusted, heated permeate to create a permeate/lactase mixture. In certain embodiments, lactose within the permeate/lactase mixture is broken down into monosaccharides. In certain embodiments, lactase enzyme is added at about 0.1% w/w to about 0.5% w/w concentration. In certain embodiments, lactase enzyme is added at about 0.1% w/w, or 0.2% or 0.3% or 0.4% or 0.5% w/w. There are many commercially available lactase enzymes that may be used. As such, the lactase enzyme may be derived from any origin (e.g., fungal or bacterial in origin).
[0069] In some embodiments, the pH-adjusted, heated permeate is incubated with the lactase enzyme for about 5 to about 225 minutes. In certain embodiments, the incubation time is about 15 min to about 90 min. In some embodiments, the incubation time is about 30
minutes to about 90 minutes. In particular embodiments, the incubation time is about 60 minutes. Some aspects contemplate that that incubation time is dependent upon myriad of factors including, but not limited to, the source of the enzyme used, the temperature and pH of the mixture and the concentration of enzyme used. Thus, in some embodiments, incubation time with the lactase enzyme may be adjusted to factor in such variables as a matter of routine. While the pH, temperature, and enzyme incubation conditions provided here are what work optimally for the process described herein, one of skill in the art would understand that modifications may be made to one or more of these variables to achieve similar results. For example, if less enzyme is used than the about 0.1% w/w to about 0.5% w/w described herein, the incubation time may need to be extended to achieve the same level of lactose digestion. Similar adjustments may be made to both the temperature and pH variables as well.
[0070] In certain embodiments, after incubation the permeate/lactase mixture is cooled to a temperature of about 20°C to about 30°C. In a particular embodiment, the permeate/lactase mixture is cooled to a temperature of about 25°C.
[0071] In some embodiments, the permeate/lactase mixture is clarified to remove insoluble constituents. In certain embodiments, insoluble material may form throughout the change in pH and temperature. Thus, in some embodiments, it may be necessary or beneficial to clarify the mixture to remove these insoluble constituents, for example, through a depth filter. The filters may be 0.1 to 10 micron filters. In some embodiments, the filters are about 1 to about 5 micron filters. Alternatively, removal of insoluble constituents can be achieved through a centrifugation process or a combination of centrifugation and membrane filtration. The clarification step is not essential for the preparation of a diverse HMO composition, as described herein, rather, this optional step aids in obtaining a more purified permeate composition. Furthermore, the clarification step is important in the reusability of the filtration membranes and thus to the scalability of the process. Some aspects contemplate that, without adequate clarification, substantially more filter material is required, increasing the difficulty and expense to produce permeate compositions at clinical scale. However, one will understand that more or less stringent clarification may be performed at this stage in order to produce more or less purified permeate compositions, depending on formulation and application. For example, precipitated minerals may be less of a problem for a formulation destined for lyophilization.
[0072] In certain embodiments the spent and excess lactase enzyme is removed from the clarified permeate/lactase mixture. There may, however, be some instances where the inactivated foreign protein will carry no biological risk and therefore the added steps of lactase removal or even inactivation may not be necessary. In some embodiments, the spent and excess lactase is inactivated, for example by high temperature, pressure, or both. In some embodiments, the inactivated lactase is not removed from the composition.
[0073] In other embodiments, however, a further purification to remove foreign proteins will be called for. In such embodiments lactase enzyme removal may be accomplished by ultrafiltration. In some embodiments, ultrafiltration is accomplished using an ultrafiltration membrane, for example using a membrane with molecular weight cut-off of < 50,000 Dalton, e.g., a BIOMAX-50K. In some embodiments, the molecular weight cut-off less than or equal to about 10 kDa. In certain embodiments, the molecular weight cut-off less than or equal to about 25 kDa. In particular embodiments, the molecular weight cut-off less than or equal to about 50 kDa.
[0074] In certain embodiments, an additional ultrafiltration is performed through a smaller membrane than the initial membrane with molecular weight cut-off of < 50,000 Dalton. In some embodiments, the additional ultrafiltration is performed with a membrane with a molecular weight cut-off of between 10 kDa and 50 kDa, 1 kDa and 10 kDa, 1 kDa and 5 kDa, or 2 kDa and 3 kDa. In certain embodiments, the additional ultrafiltration is performed with a membrane with a molecular weight cut-off of between 2 kDa and 3 kDa. In certain embodiments, an additional ultrafiltration is not performed. In some embodiments, the additional filtration step is performed, such as to aid in the overall purity of the permeate product, such as by assisting in the removal of smaller potentially bioactive and/or immunogenic factors such as microRNAs and exosomes.
[0075] In some embodiments, the clarified mixture that has undergone at least one, and in some cases two or more rounds of ultrafiltration (or alternative lactase removal means) is further filtered to purify and concentrate human milk oligosaccharides and to reduce the mineral and monosaccharides content.
[0076] In some embodiments, filtration can be accomplished using a nanofiltration membrane. In some embodiments, the membrane has a molecular weight cut-off of < 1,000 Dalton. In certain embodiments, the membrane has a molecular weight cut-off of between 1 kDa and 1,000 kDa. In certain embodiments, the membrane has a molecular weight cut-off of less than 600 Da. In certain embodiments, the membrane has a molecular weight cut-off
between 400 Da and 500 Da. In some aspects, the additional nanofiltration removes monosaccharides, minerals, particularly calcium, and smaller molecules to produce the final purified HMO composition.
[0077] In some embodiments, additional or alternative steps may be taken for the removal of minerals. Such an additional step may include, for example, centrifugation, membrane clarification (< 0.6 micron), or combination of centrifugation and membrane filtration of heated (> 40°C) or refrigerated/frozen and thawing of HMO Concentrate. The collected supernatant or filtrate of these additional or alternative steps, in some embodiments, is concentrated further using a nanofiltration membrane. In some embodiments, the nanofiltration comprises filtration through a membrane with a molecular cut-off of < 600 Dalton. In some embodiments, these additional steps may be performed at any stage of the process, including but not limited to prior to or after pasteurization.
[0078] In some embodiments, the physical property of nanofiltration membranes can be modified, such as chemical modification, to selectively concentrate sialylated human milk oligosaccharides, for example, allowing greater efficiency of neutral human milk oligosaccharides removal from HMO concentrate, in instances where concentrated sialylated human milk oligosaccharides are preferred.
[0079] In certain embodiments, the permeate may be further processed, e.g., concentrated or diluted. In some embodiments, the permeate may be concentrated by a suitable process such as nanofiltration, reverse osmosis, or dried, e.g., lyophilized.
[0080] In some embodiments, the permeate is treated to reduce bioburden, such as by any means known in the art. In some embodiments, the purified HMO composition is pasteurized. In some aspects, pasteurization is accomplished at > 63°C for a minimum of 30 minutes. Following pasteurization, the composition is cooled to about 25°C to about 30°C and clarified through a 0.2 micron filter to remove any residual precipitated material.
[0081] In certain embodiments, the mixture of oligosaccharides is formulated with an ultra-filtered permeate obtained from human milk. In some embodiments, the mixture of oligosaccharides is formulated with permeate that has been ultra filtered from the skim fraction of pooled human milk. In certain embodiments, lactose is removed, e.g., enzymatically degraded, prior to formulation into the mixture of oligosaccharides.
D. Butyrate producing bacteria
[0082] In certain embodiments, a butyrate producing strain of bacteria is administered along with or in addition to the provided prebiotic mixture, e.g., of human milk oligosaccharides, at least one Bifidobacterium and at least one propionate producing bacterium. In some embodiments, the at least one butyrate producing strain is capable of consuming lactate and/or acetate. In particular embodiments, the at least one butyrate producing strain is capable of producing, generating, and/or creating butyrate in the presence of lactate and/or acetate. In certain embodiments, the at least one butyrate producing strain is capable of producing, generating, and/or creating butyrate in the presence of the at least one Bifidobacterium, e.g., any Bifidobacterium described herein such as in Section I-B, and the prebiotic mixture, e.g., a prebiotic mixture described herein such as in Section I-C. Suitable butyrate producing strains are described in PCT App. Pub. No. WO2022036225, hereby incorporated by reference in its entirety.
[0083] In some embodiments, the at least one butyrate producing strain is or includes at least one, two, three, four, five, six, seven, eight, nine, ten, or more species, subspecies, or strains of bacteria capable of producing butyrate. In particular embodiments, the butyrate producing strain is or includes at least three strains of bacteria capable of producing butyrate. In some embodiments, the butyrate producing strain is or includes between one and five species, subspecies, or strains of bacteria capable of producing butyrate.
[0084] In certain embodiments, the at least one butyrate producing strain has one or more genes that contribute to the production, generation, or making of butyrate. In particular embodiments, the at least one butyrate producing strain has a functional butyryl-Co A: acetate CoA-transferase (but) gene. In certain embodiments, the at least one butyrate producing strain has a functional butyrate kinase (buk) gene. In some embodiments, the at least one butyrate producing strain has a functional butyryl-CoA:4-hydroxybutyrate CoA transferase (4Hbf) gene. In particular embodiments, the at least one butyrate producing strain has a functional butyryl-Co A: acetoacetate CoA transferase (Ato) gene.
[0085] In some embodiments, the at least one butyrate producing strain is or includes one or more species, subspecies, or strains of Clostridium Cluster IV bacteria. In certain embodiments, the at least one butyrate producing strain is or includes one or more species, subspecies, or strains of Clostridium Cluster XlVa bacteria. In certain embodiments, the at least one butyrate producing strain is or includes one or more species, subspecies, or strains of bacteria belonging to the Clostridium, Eubacterium, Ruminococcus, Coprococcus, Dorea,
Lachnospira, Roseburia, Butyrivibrio, or Anaerofilum genera. In some embodiments, the at least one butyrate producing strain is or includes one or more species, subspecies, or strains of bacteria belonging to the Clostridium, Eubacterium, Ruminococcus, Coprococcus, Dorea, Lachnospira, Roseburia or Butyrivibrio genera. In particular embodiments, the at least one butyrate producing strain is or includes one or more species, subspecies, or strains of bacteria belonging to the Clostridium, Eubacterium, Ruminococcus ov Anaerofilum genera.
[0086] In some embodiments, the at least one butyrate producing strain is or includes one or more species, subspecies, or strains of Agathobacter rectalis (also referred to as Eubacterium rectale), Anaerobutyricum hallii (also referred to as Eubacterium hallii), Anaerobutyricum soehngenii, Anaerocolumna aminovalerica (also referred to as Clostridium aminovalericum), Anaerostipes butyraticus, Anaerostipes caccae, Anaerostipes hadrus (also referred to as Eubacterium hadrum), Anaerostipes rhamnosivorans, Anaerotruncus colihominis, Blautia argi, Blautia caecimuris, Blautia coccoides (also referred to as Clostridium coccoides), Blautia faecicola, Blautia faecis, Blautia glucerasea, Blautia hansenii (also referred to as Streptococcus hansenii), Blautia schinkii, Blautia ster coris, Blautia wexlerae, Blautia hominis, Blautia hydrogenotrophica (also referred to as Ruminococcus hydrogenotrophicus), Blautia luti (also referred to as Ruminococcus luti), Blautia obeum (also referred to as Ruminococcus obeum), Blautia producta (also referred to as Ruminococcus productus; Streptococcus productus; Peptostreptococcus productus), Blautia schinkii (also referred to as Ruminococcus schinkii), Butyrivibrio crossotus, Clostridium cellulosi, Clostridium hylemonae, Clostridium innocuum, Clostridium leptum, Clostridium nexile, Clostridium orbiscidens, Clostridium scindens, Clostridium sporosphaeroides, Clostridium symbiosum, Clostridium viride, Coprococcus catus, Coprococcus comes, Coprococcus eutactus, Coprococcus phoceensis, Eisenbergiella tayi, Emergencia timonensis, Enterocloster aldenensis (also referred to as Clostridium aldenense), Enterocloster asparagiformis (also referred to as Clostridium asparagiforme), Enterocloster bolteae (also referred to as Clostridium bolteae), Enterocloster citroniae (Clostridium citroniae), Enterocloster clostridioformis (also referred to as Clostridium clostridioforme), Enterocloster lavalensis (Clostridium lavalense), Erysipelatoclostridium ramosum (also referred to as Thomasclavelia ramosa), Eubacterium siraeum, Eubacterium ventriosum, Dorea formicigenerans (also referred to as Eubacterium formicigenerans), Dorea longicatena, Faecalibacterium prausnitzii (also referred to as Fusobacterium prausnitzii),
Faecalicatena contorta (also referred to as Eubacterium contortum), Faecalicatena fissicatena (also referred to as Eubacterium fissicatena), Faecalicatena orotica (also referred to as Clostridium oroticum), Faecalimonas umbilicate, Flavonifractor plautii (also referred to as Clostridium orbiscindens), Hungatella effluvii, Hungatella hathewayi (also referred to as Clostridium hathewayi), Hungatella xylanolytica, Lachnospira eligens (also referred to as Eubacterium eligens), Lachnospira multipara, Lachnospira pectinoschiza, Lacrimispora aerotolerans (also referred to as Clostridium aerotolerans), Lacrimispora algidixylanolytica (also referred to as Clostridium algidixylanolyticum), Lacrimispora amygdalina (also referred to as Clostridium amygdalinum), Lacrimispora celerecrescens (also referred to as Clostridium celerecrescens), Lacrimispora indolis (also referred to as Clostridium indolis), Lacrimispora saccharolytica (also referred to as Clostridium saccharolyticum), Lacrimispora sphenoides (also referred to as Bacillus sphenoides), Lacrimispora xylanolytica (also referred to as Clostridium xylanolyticum), Lactonifactor longoviformis, Marvinbryantia formatexigens (also referred to as Bryantella formatexigens), Mediterraneibacter butyricigenes, Mediterraneibacter faecis (also referred to as Ruminococcus faecis), Mediterraneibacter glycyrrhizinilyticus (also referred to as Clostridium glycyrrhizinilyticum), Mediterraneibacter gnavus (also referred to as Ruminococcus gnavus), Mediterraneibacter lactaris (also referred to as Ruminococcus lactaris), Mediterraneibacter massiliensis, Mediterraneibacter torques (also referred to as Ruminococcus torques), Merdimonas faecis, Murimonas intestini, Oscillibacter massiliensis, Papillibacter cinnamivorans, Pseudoflavonifractor capillosus, Robinsoniella peoriensis, Roseburia faecis, Roseburia hominis, Roseburia intestinalis, Roseburia inulinivorans, Ruminococcus albus, Ruminococcus bromii, Subdoligranulum variabile, or Syntrophococcus sucromutans.
[0087] In certain embodiments, the at least one butyrate producing strain is or includes one or more species, subspecies, or strains of Agathobacter rectalis, Anaerobutyricum hallii, Anaerostipes caccae, Blautia producta, Clostridium leptum, Faecalibacterium prausnitzii, Anaerobutyricum soehngenii, Roseburia hominis, or Roseburia intestinalis. In some embodiments, the at least one butyrate producing strain is or includes one or more species, subspecies, or strains of Anaerostipes caccae, Clostridium innocuum, Roseburia hominis, or Roseburia intestinalis.
E. Exemplary compositions, kits, and articles of manufacture
[0060] In some embodiments, provided herein are compositions, kits, or articles of manufacture that are or include a combination of prebiotics, e.g., prebiotic mixtures of non- digestible carbohydrates such as human milk oligosaccharides, and probiotics that include one or more strains of Bifidobacteria such as B. longum subsp. infantis and one or more strains of propionate producing bacteria, e.g., Veillonella sp. In certain aspects, the prebiotic mixture and probiotics may be formulated as a pharmaceutical composition or a nutritional composition. In some embodiments, the provided composition includes or incorporates the prebiotic mixture and the probiotic strains. In particular embodiments, the probiotic strains may be formulated as a pharmaceutical composition or a nutritional composition. In certain embodiments, the prebiotic mixture and the probiotic are formulated or manufactured as separate compositions. In particular embodiments, provided herein are kits or articles of manufacture that are or include separate prebiotic and probiotic compositions.
[0088] In particular embodiments, provided herein are kits or articles of manufacture that are or include prebiotic mixtures that are or include one or more human milk oligosaccharides, and probiotics that include one or more strains of Bifidobacteria and one or more strains of propionate producing bacteria. In some embodiments, the prebiotic mixture is or includes any of the prebiotic mixtures described herein, e.g., in Section I-C. In particular embodiments, the prebiotic mixture contains at least 2, at least 5, at least 10, at least 25, at least 50, at least 100, or at least 150 human milk oligosaccharides, e.g., at least two, three, four, five, or more than five synthetic human milk oligosaccharides or at least 10, at least 25, at least 50, or at least 100 human milk oligosaccharides from a human milk source such as human milk permeate. In some embodiments, the one or more strains of Bifidobacteria are or include one or more strains of Bifidobacteria described herein, e.g., in Section I-B. In certain embodiments, the one or more strains of Bifidobacteria are capable of consuming, internalizing, and/or hydrolyzing human milk oligosaccharides. In some embodiments, the one or more strains of propionate producing bacteria are or include any of the propionate producing bacteria described herein, e.g., in Section I-A. In some embodiments, the one or more strains of propionate producing bacteria are or include at least one strain from the genus Veillonella or Megasphaera.
[0089] In particular embodiments, the kits or articles of manufacture are or include i) prebiotic mixtures that are or include at least one or more of 2'-fucosyllactose, 3’- fucosyllactose, 3’-sialyllactose, 6'-sialyllactose, lacto-N-tetraose, lacto-N-difucohexaose I, lactodifucotetraose, lacto-N-fucopentaose I, sialylacto-N-tetraose c, sialylacto-N-tetraose b,
and/or disialyllacto-N-tetraose; ii) at least one Bifidobacterium that is or includes B. longum subsp. infantis, and iii) at least one propionate producing bacterium that is or includes one or more strains of the genus of the genus Veillonella or Megasphaera. In certain embodiments, the kits or articles of manufacture are or include i) prebiotic mixtures that are or include at least one, some, or all of 2'-fucosyllactose, 3’-fucosyllactose, 3’-sialyllactose, 6'-sialyllactose, lacto-N-tetraose, lacto-N-difucohexaose I, lactodifucotetraose, lacto-N-fucopentaose I, sialylacto-N-tetraose c, sialylacto-N-tetraose b, and/or disialyllacto-N-tetraose; ii) at least one Bifidobacterium that is or includes B. longum subsp. infanlis. iii) at least one propionate producing bacterium that is or includes one or more of Megaspaera elsdenii, Veillonella atypica (V. atypica), Veillonella dispar (V. dispar), Veillonella infantium (V. infantium), Veillonella nakazawae (V. nakazawae), or Veillonella parvula (V. parvula), and/or Veillonella rogosae (V. rogosae). In some embodiments, the kits or articles of manufacture include one or more strains of butyrate producing bacteria, e.g., any of the butyrate producing bacteria described herein such as in Section I-D. In certain embodiments, the butyrate producing bacteria is or includes one or more of Agathobacter rectalis, Anaerobutyricum hallii, Anaerostipes caccae, Blautia producta, Clostridium leptum, Faecalibacterium prausnitzii, Anaerobutyricum soehngenii, Roseburia hominis, or Roseburia intestinalis.
[0090] In some embodiments, provided herein are articles of manufacture that are or include prebiotic mixtures that are or include one or more human milk oligosaccharides, probiotics that include one or more strains of Bifidobacteria and one or more strains of propionate producing bacteria, and instructions for use, e.g., describing any method herein such as the methods described in Section II. In certain embodiments, the articles of manufacture are or include i) prebiotic mixtures that are or include at least one or more of 2'- fucosyllactose, 3’ -fucosy llactose, 3’-sialyllactose, 6'-sialyllactose, lacto-N-tetraose, lacto-N- difucohexaose I, lactodifucotetraose, lacto-N-fucopentaose I, sialylacto-N-tetraose c, sialylacto-N-tetraose b, and/or disialyllacto-N-tetraose; ii) at least one Bifidobacterium that is or includes B. longum subsp. infantis, iii) at least one propionate producing bacterium that is or includes one or more strains of the genus of the genus Veillonella or Megasphaera, and iv) instructions for use describing one or more methods included herein in Section II. In particular embodiments, the articles of manufacture are or include i) prebiotic mixtures that are or include at least one, some, or all of 2'-fucosyllactose, 3’-fucosyllactose, 3’- sialyllactose, 6'-sialyllactose, lacto-N-tetraose, lacto-N-difucohexaose I, lactodifucotetraose, lacto-N-fucopentaose I, sialylacto-N-tetraose c, sialylacto-N-tetraose b,
and/or disialyllacto-N-tetraose; ii) at least one Bifidobacterium that is or includes B. longum subsp. infantis, iii) at least one propionate producing bacterium that is or includes one or more oi Megaspaera elsdenii, Veillonella atypica (V. atypica), Veillonella dispar (V. dispar), Veillonella infantium (V. infantium), Veillonella nakazawae (V. nakazawae), or Veillonella parvula (V. parvula), and/or Veillonella rogosae (V. rogosae), and iv) instructions for use describing one or more methods included herein in Section II. In some embodiments, the articles of manufacture include one or more strains of butyrate producing bacteria described herein in Section I-D, such as one or more of Agathobacter rectalis, Anaerobutyricum hallii, Anaerostipes caccae, Blautia producta, Clostridium leptum, Faecalibacterium prausnitzii, Anaerobutyricum soehngenii, Roseburia hominis, or Roseburia intestinalis.
II. USES OF THE PROVIDED COMPOSITONS AND FORMULATIONS
[0091] In particular embodiments, provided herein are methods for treating, preventing, or ameliorating one or more diseases, disorders, or conditions in a subject in need thereof. In certain embodiments, provided herein are methods for generating propionate, and/or increasing the level or amount of propionate, within the gut and/or intestine of a subject in need thereof. In some embodiments, the method is or includes steps for administering to the subject a combination of probiotic bacteria that is or includes at least one strain of Bifidobacterium, e.g., such as any described herein, e.g., in Section I-B, and a strain of bacterium capable of producing propionate, such as any of the propionate producing bacterium described herein, e.g., in Section I-A, and a prebiotic mixture, such as any described herein, e.g., in Section I-C. In some embodiments, the prebiotic mixture, the Bifidobacterium, and the propionate producing bacterium are administered together, such as at the same time, on the same treatment days, and/or within the same dosage formula. In certain embodiments, the prebiotic mixture, the Bifidobacterium, and the propionate producing bacterium are administered separately, such as at different times, on different treatment days, and/or within different dosage formulations. In some embodiments, the treatment may include administering the prebiotic mixture, the Bifidobacterium, and the propionate producing bacterium together during certain days or phases of the treatment and then separately during other certain days or phases of the treatment.
[0092] In certain embodiments, provided herein are methods for treating, preventing, or ameliorating one or more diseases, disorders, or conditions that are or may be associated with dysbiosis, e.g., of the intestinal microbiome, in a subject in need thereof. In some aspects, the intestinal microbiome is involved in or associated with a number of physiological functions including digestion, metabolism, extraction of nutrients, synthesis of vitamins, prevention of pathogen colonization, and immune modulation. In some such aspects, alterations or changes in composition and biodiversity of the intestinal microbiome may be associated with or exacerbate various metabolic states, gastrointestinal disorders, and other pathophysiological conditions. In some aspects, conditions, diseases, or disorders with inflammatory components or components relating to infection, allergy, or immune dysfunction may be exacerbated by dysbiosis or may have an underlying contribution of dysbiosis to the pathology. Thus, in certain aspects, targeting the microbiome with the provided prebiotic and probiotic compositions may successfully treat, alleviate, or prevent a wide range of conditions, diseases, and disorders.
[0093] In some embodiments, provided herein is a method for treating, reducing, ameliorating, or preventing dysbiosis. In particular embodiments, the method is or includes steps for administering to the subject at least one strain of Bifidobacterium, e.g., such as any described herein, e.g., in Section I-B, a strain of bacterium capable of producing propionate, such as any of the propionate producing bacterium described herein, e.g., in Section I-A, and a prebiotic mixture, such as any described herein, e.g., in Section I-C. In some embodiments, the one or more diseases, disorders, or conditions is, includes, or is associated with dysbiosis, e.g., of the intestinal microbiome. In certain embodiments, the microbiome is an intestinal microbiome of a human. In various embodiments, the microbiome is the intestinal microbiome of an infant or young child. In certain embodiments, the microbiome is an intestinal microbiome of an adult human.
[0094] In some embodiments, the method is or includes steps for administering to the subject at least one strain of Bifidobacterium, e.g., such as any described herein, e.g., in Section I-B, a strain of bacterium capable of producing propionate, such as any of the propionate producing bacterium described herein, e.g., in Section I-A, and a prebiotic mixture, such as any described herein, e.g., in Section I-C for the treatment of one or more diseases, disorders, or conditions associated with inflammation, infection, allergy, immune dysfunction, or dysbiosis of the intestinal microbiome. In certain embodiments, the one or more conditions, diseases, or disorders is, includes, or is associated with dysbiosis. In certain
embodiments, the one or more conditions, diseases, or disorders is, includes, or is associated with inflammation. In particular embodiments, the one or more condition, disease, or disorder is, includes, or is associated with an autoimmune disease. In particular embodiments, the one or more conditions, diseases, or disorders is or is associated with an allergy. In certain embodiments, the prebiotic mixture, e.g., of human milk oligosaccharides, the Bifidobacterium, e.g., B. longum subsp. infantis, and the propionate producing bacteria, e.g., Veillonella sp., are administered to prevent a disease, disorder, or condition. In some embodiments, the prebiotic mixture and the probiotic strain prevent a condition described herein, e.g, in Section II-B. In particular embodiments, the prebiotic mixture and the probiotic strain reduce the risk, likelihood, or probability of the disease, disorder, or condition, and/or of experiencing one or more symptoms associated with the disease, disorder, or condition. In some embodiments, the risk, likelihood, or probability is reduced by at least 10%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 90%, 99%, or 99.9% as compared to alternative treatments or no treatments, or as compared to administration of the probiotic strain or prebiotic mixture alone.
[0095] As used herein, “subject” and “subject in need thereof’ are used interchangeably. In particular embodiments, the subject is a human. In some embodiments, the subject is an infant, a child, a juvenile, or an adult. In certain embodiments, the subject is at least 1 month, 3 months, 6 months, 12 months, 18 months, or 24 months of age. In certain embodiments, the subject is at least 1 year, 2 years, 5 years, 10 years, 12 years, 16 years, or at least 18 years of age. In some embodiments, the subject is at least 12 years old. In certain embodiments, the subject is at least 18 years old. In some embodiments, the subject is an adult. In certain embodiments, the subject is elderly, e.g, at least 65 or 75 years of age.
A. Administering Provided Compositions to the Subject
[0096] In certain embodiments, the provided methods include one or more treatment phases that are or include administration of one or more of the Bifidobacterium, e.g., B. longum subsp. infantis, the propionate producing bacterium, e.g., Veillonella sp. and the prebiotic mixture, e.g., of human milk oligosaccharides. In some embodiments, the method is or includes a treatment where all three of the propionate producing bacterium, the Bifidobacterium, and the prebiotic mixture are administered. In certain embodiments, the method is or includes a treatment phase where the Bifidobacterium and the prebiotic mixture
are administered, e.g., in the absence of the propionate producing bacterium. In certain embodiments, the method is or includes a treatment phase where the prebiotic mixture is administered, e.g., in the absence of the propionate producing bacterium and the Bifidobacterium. In some embodiments, the method includes two or more treatment phases.
[0097] In certain embodiments, the prebiotic mixture is administered daily for at least 2, 3, 4, 5, 7, 10, 14, 21, or 28 days, e.g., consecutive days. In certain embodiments, the prebiotic mixture is administered in an amount of at least 0.001 g, 0.01 g, 0.1 g, 1 g, 2 g, 3 g, 4 g, 5 g, 6 g, 7.5 g, 8 g, 9 g, 10 g, 12 g, 16 g, 18 g, 20 g, 25 g, or 50 g per day, e.g., total weight of the prebiotics such as non-digestible carbohydrates such as human milk oligosaccharides. In particular embodiments, the prebiotic mixture in an amount of at least 0.001 g, 0.01 g, 0.1 g, 1 g, 2 g, 3 g, 4 g, 5 g, 6 g, 7.5 g, 8 g, 9 g, 10 g, 12 g, 16 g, 18 g, 20 g, 25 g, or 50 g total human milk oligosaccharides per day. In some embodiments, the prebiotic mixture is administered in an amount of between 0.1 g and 50 g; 0.5 g and 25 g, 1 g and 20 g, 2 g and 18 g, 1 g and 5 g, 2 g and 3 g, 3 g and 6 g, 4 g and 5 g, 5 g and 10 g, 8 g and 10 g, 10 g and 20 g, 15 g and 20 g, or 17 g and 19 g total human milk oligosaccharides per day. In some embodiments, the prebiotic mixture is administered in an amount of, of about, or of at least 2 g, 4.5 g, 6 g, 9 g, 12 g, 16 g, or 18 g total human milk oligosaccharides per day. In some embodiments, the prebiotic mixture is administered in a single dose per day. In various embodiments, the prebiotic mixture is administered in multiple doses per day, e.g., at or at least two, three, four, five, or six or more doses per day. In some embodiments, a daily amount of human milk oligosaccharides, e.g., 2 g, 4.5 g, 6 g, 9 g, 12 g, 16 g, or 18 g total human milk oligosaccharides per day, are administered over two, three, four, five, six, or more than six doses per day to achieve the total amount. In certain embodiments, doses of 9 g of total human milk oligosaccharides are administered twice daily for a total of 18 g/day.
[0098] In certain embodiments, the at least one Bifidobacterium is administered daily for at least 2, 3, 4, 5, 7, 10, 14, 21, or 28 days, e.g., consecutive days. In some embodiments, the at least one Bifidobacterium is administered in an amount of at least 1 x 101, 5 x 101,! x 102, 1 x 103, 1 x 104, 1 x 105, 1 x 106, 5 x 106, 1 x 107, 1 x 107, 5 x 107, 1 x 108, or 5 x 108 colony forming units (CFU) per day. In various embodiments, the at least one Bifidobacterium is administered in an amount of at least 1 x 101, 1 x 102, 1 x 103, 1 x 104, 1 x 105, 1 x 106, 5 x 106, 1 x 107, 5 x 107, l x 108, or 5 x 108 colony forming units (CFU) per dose. In certain embodiments, the at least one Bifidobacterium is administered in an amount of between 1 x 106 and 1 x 1012, 5 x 106 and 1 x 1010, 1 x 107 and 1 x 109, or 1 x 107 and 1 x 108
CFU per day. In some embodiments, the Bifidobacterium strain is administered in an amount of, of about, or at least 5 x 106 colony forming units (CFU) per dose or per day. In some embodiments, the Bifidobacterium strain is administered in an amount of, of about, or at least 8 x 107 colony forming units (CFU) per dose or per day.
[0099] In certain embodiments, the at least one propionate producing bacterium is administered daily for at least 2, 3, 4, 5, 7, 10, 14, 21, or 28 days, e.g., consecutive days. In some embodiments, the at least one propionate producing bacterium is administered in an amount of at least 1 x 101, 5 x 101,! x 102, 1 x 103, 1 x 104, 1 x 105,l x 106, 5 x 106, 1 x 107, 5 x 107, 1 x 108, or 5 x 108 colony forming units (CFU) per day. In various embodiments, the at least one propionate producing bacterium is administered in an amount of at least 1 x 101, 1 x 102, 1 x 103, 1 x 104, 1 x 105, 1 x 106, 5 x 106, 1 x 107, 5 x 107, 1 x 108, or 5 x 108 colony forming units (CFU) per dose. In certain embodiments, the at least one propionate producing bacterium is administered in an amount of between 1 x 106 and 1 x 1012, 5 x 106 and 1 x IO10, 1 x 107 and 1 x 109, or 1 x 107 and 1 x 108 CFU per day. In some embodiments, the at least one propionate producing bacterium is administered in an amount of, of about, or at least 5 x 106 colony forming units (CFU) per dose or per day. In some embodiments, the probiotic strain is administered in an amount of, of about, or at least 8 x 107 colony forming units (CFU) per dose or per day.
[0100] In some embodiments, the prebiotic mixture is administered at least once within 3 months, 2 months, 1 month, 60 days, 45 days, 30 days, 6 weeks, 5 weeks, 4 weeks, 3 weeks, 28 days, 21 days, 14 days, 10 days, 7 days, 5 days, 3 days, or 1 day prior to administration of the at least one Bifidobacterium. In certain embodiments, the prebiotic mixture is administered at least once daily for at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 21, or 28 consecutive days prior to the at least one Bifidobacterium. In certain embodiments, the prebiotic mixture is administered at least once within 3 months, 2 months, 1 month, 60 days, 45 days, 30 days, 6 weeks, 5 weeks, 4 weeks, 3 weeks, 28 days, 21 days, 14 days, 10 days, 7 days, 5 days, 3 days, or 1 day after administration of the at least one Bifidobacterium. In certain embodiments, the prebiotic mixture is administered at least once daily for at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 21, or 28 consecutive days after the at least one Bifidobacterium. In various embodiments, the prebiotic mixture is administered prior to and after the at least one Bifidobacterium.
[0101] In certain embodiments, the prebiotic mixture, the at least one Bifidobacterium, and the at least one propionate producing bacterium are administered
together or separately during the same treatment regimen. In certain embodiments, the treatment regimen has separate treatment phases. The treatment regimen may include separate treatment phases with different combinations, doses, or timing of doses for some or all of the prebiotic mixture, the at least one Bifidobacterium, and the at least one propionate producing bacterium are administered to the subject. In some embodiments, all of the prebiotic mixture, the at least one Bifidobacterium, and the at least one propionate producing bacterium are administered during a treatment phase that occurs during the treatment regimen, and, in a different treatment phase, only one or two of the prebiotic mixture, the at least one Bifidobacterium, and the at least one propionate producing bacterium are administered in a different treatment phase that occurs during the same treatment regimen.
[0102] In certain embodiments, a treatment phase has a duration of at least one, two, three, four, five, six, seven, eight, nine, or ten days, or for at least one, two, three, four, five, or six weeks, or for at least one, two, three, four, five, or six months. In some embodiments, a treatment phase is or is at least seven days. In particular embodiments, a treatment phase is or is at least fourteen days.
[0103] In some embodiments, the treatment regimen includes more than one treatment phase. In particular embodiments, in one treatment phase, e.g., a first treatment phase, the prebiotic mixture and the at least one Bifidobacterium strain are administered. In some embodiments, the prebiotic mixture and the at least one Bifidobacterium strain are administered within the same composition during the treatment phase, e.g., the first treatment phase. In some embodiments, the prebiotic mixture and the at least one Bifidobacterium strain are administered as separate compositions during the treatment phase, e.g., the first treatment phase. In certain embodiments, one or both of the at least one Bifidobacterium strain and the prebiotic mixture is administered at least once, at least twice, at least three times, at least once per week, at least twice per week, at least three time per week, every other day, and/or every day during the treatment phase, e.g., the first treatment phase. In some embodiments, the prebiotic mixture and the at least one Bifidobacterium strain are administered on the same day for at least one, some, or all of the days of the treatment phase, e.g., the first treatment phase. In particular embodiments, the at least one propionate producing strain is also administered during the treatment phase, e.g., the first treatment phase. In some embodiments, the at least one propionate producing strain is administered at least once, at least twice, at least three times, at least once per week, at least twice per week, at least three time per week, every other day, and/or every day during the treatment phase,
e.g., the first treatment phase. In particular embodiments, the at least one propionate producing strain is administered on the same days as the at least one Bifidobacterium strain during the treatment phase, e.g., the first treatment phase. In some embodiments, the treatment phase, e.g., the first treatment phase, has a duration of at least one, two, three, four, five, six, seven, ten, fourteen days, or at least one, two, three, four, five, or six weeks, or from one day to fourteen days or from three days to seven days.
[0104] In some embodiments, the treatment regimen includes a treatment phase, e.g., a second treatment phase, where the prebiotic mixture and not the at least one Bifidobacterium strain is administered. In certain embodiments, the prebiotic mixture is administered at least once, at least twice, at least three times, at least once per week, at least twice per week, at least three time per week, every other day, and/or every day during the treatment phase, e.g., the second treatment phase. In particular embodiments, the at least one propionate producing strain is also administered during the second treatment phase. In some embodiments, the at least one propionate producing strain is administered at least once, at least twice, at least three times, at least once per week, at least twice per week, at least three time per week, every other day, and/or every day during the second treatment phase. In certain embodiments, the at least one propionate producing strain is not administered during the second treatment phase. In particular embodiments, the second treatment phase has a duration of at least one, two, three, four, five, six, seven, ten, fourteen days, or at least one, two, three, four, five, or six weeks, or from one day to fourteen days or from three days to seven days.
[0105] In some embodiments, the first and second treatment phases occur once during a treatment regimen. In particular embodiments, the first and second treatment phases occur more than once during a treatment regimen, such as cycling or repeating throughout the duration of the treatment regimen. In some embodiments, there may be a gap or duration, e.g., for at least one, two, three, five, seven, ten, or fourteen, days, with no treatments between cycles, e.g., after the end of the second treatment phase and before the beginning of a subsequent first treatment phase.
[0106] In some embodiments, the prebiotic mixture and the at least one Bifidobacterium strain are administered together or separately for a period of time, such as in a treatment regimen. In some embodiments, the administration of the prebiotic mixture, e.g., of HMOs, allows for the engraftment and expansion of the Bifidobacterium strain, e.g., B. longum subsp. infantis. In certain embodiments, the Bifidobacterium strain is exogenous to
the subject’s microbiome, e.g., intestinal microbiome. In particular embodiments, the Bifidobacterium strain is not present within the subject’s microbiome prior to administration. In certain embodiments, the prebiotic mixture is administered concurrently with and/or subsequently to administration of the at least one Bifidobacterium strain. In some embodiments, the at least one Bifidobacterium strain is present and/or expands within the subject’s microbiome during a time period in which the prebiotic mixture is administered. In certain embodiments, the presence or amount of the at least one Bifidobacterium strain within the microbiome is reduced when administration of the prebiotic mixture ends, is ceased or is terminated. In particular embodiments, the Bifidobacterium strain is absent and/or undetectable following the termination or end of administration of the prebiotic mixture. In certain embodiments, the presence of the Bifidobacterium strain, e.g., B. infantis, is transient and is regulated by administration of the prebiotic mixture.
[0107] In certain embodiments, at least one Bifidobacterium strain is capable of consuming or metabolizing some or all of the oligosaccharides, e.g., HMOs, of the prebiotic mixture. In some embodiments, the timing or dosing for administering the at least one Bifidobacterium strain and the prebiotic mixture achieves a growth or expansion of the Bifidobacterium strain in vivo, e.g., within the microbiome of the subject. In certain embodiments, the administered oligosaccharides, e.g., HMOs, selectively or exclusively serve as a carbon source for the at least one Bifidobacterium strain, e.g., as opposed to other bacterial strains present in the gut or microbiome. In some embodiments, the oligosaccharides of the mixture selectively or exclusively serve as an energy source for the at least one Bifidobacterium strain e.g., as opposed to other bacterial strains present in the gut or microbiome.
[0108] In certain embodiments, subjects are administered (e.g., at least once daily) all of the at least one Bifidobacterium strain, e.g., B. longum subsp. infantis, the at least one propionate producer, and the prebiotic mixture, e.g., of human milk oligosaccharides, during a first or initial treatment phase, e.g., for at least 1, 3, 7, or 14 days, and then are administered the prebiotic mixture alone during a subsequent treatment phase, e.g., such that occurs immediately after the first or initial treatment phase. In some embodiments, administration of the prebiotic mixture extends the duration of the colonization of the Bifidobacterium strain within the subject’s gut and/or microbiome.
[0109] In certain embodiments, administering the prebiotic mixture regulates the expansion, level, or amount of the Bifidobacterium strain, e.g., B. longum subsp. infantis,
and/or the production or generation of metabolites, e.g., lactate and/or acetate, by the Bifidobacterium strain. In some aspects, expansion of the Bifidobacterium strain and/or generation or production of metabolites by the Bifidobacterium strain promotes the engraftment and/or the expansion of the propionate producing strain and/or promotes the production or generation of propionate by the propionate producing strain. Thus, in some aspects, the Bifidobacterium and propionate producing strains are administered to the subject, and the concurrent or subsequent administration of the prebiotic mixture may be adjusted to provide a therapeutic response, e.g., to promote growth or expansion of beneficial microbiota and/or to promote the generation or production of propionate within the subject’s gut or microbiome. In some embodiments, the dosage and/or duration of treatment with the prebiotic mixture, e.g., of HMOs, can depend on several factors, including severity and responsiveness of the disease, route of administration, time course of treatment (days to months to years), and time to amelioration of the disease.
[0110] As used herein, “subject” and “subject in need thereof’ are used interchangeably. In some embodiments, the subject is an infant, a child, a juvenile, or an adult. In certain embodiments, the subject is at least 1 month, 3 months, 6 months, 12 months, 18 months, or 24 months of age. In certain embodiments, the subject is at least 1 year, 2 years, 5 years, 10 years, 12 years, 16 years, or at least 18 years of age. In some embodiments, the subject is at least 12 years old. In certain embodiments, the subject is at least 18 years old. In some embodiments, the subject is an adult.
[OHl] In some embodiments, provided herein is a method for treating, reducing, ameliorating, or preventing dysbiosis. In particular embodiments, a method is or includes steps for administering to the subject a prebiotic mixture, such as any described herein e.g., in Section I-C, at least one Bifidobacterium strain, such as a Bifidobacterium strain described herein, e.g., in Section I-B or listed in Table 1, and a propionate producing bacterium, e.g., as described in such as in Section I-A. In some embodiments, the method treats or prevents one or more diseases or conditions that include or are associated with dysbiosis, e.g., of the intestinal microbiome. In certain embodiments, the microbiome is an intestinal microbiome of a human.
B. Conditions, Diseases, and Disorders
[0061] In certain embodiments, provided herein are methods for treating, preventing, or ameliorating one or more diseases, disorders, or conditions that are or may be associated
with dysbiosis, e.g., of the intestinal microbiome, in a subject in need thereof. In certain embodiments, administration of the at least one strain of Bifidobacterium, e.g., such as any described herein, e.g., in Section I-B, the strain of bacterium capable of producing propionate, such as any of the propionate producing bacterium described herein, e.g., in Section I- A, and the prebiotic mixture, such as any described herein, e.g., in Section I-C, are useful to treat, ameliorate, remedy, or prevent diseases, disorders, or conditions such as obesity, inflammatory bowel disease (IBD), celiac disease, irritable bowel syndrome (IBS), colon cancer, diabetes, liver disorders, cystic fibrosis, and allergies.
[0062] In certain embodiments, the at least one strain of Bifidobacterium, e.g., B. longum subsp. infantis, the strain of bacterium capable of producing propionate, e.g., Veillonella sp., and the prebiotic mixture, e.g., of human milk oligosaccharides, are administered to a subject to treat, ameliorate, remedy, or prevent a gastrointestinal condition, disease, or disorder associated with, related to, or caused by dysbiosis, e.g., of the intestinal microbiome. In certain embodiments, the gastrointestinal condition, disease, or disorder is or includes one or more of a chronic inflammatory disease, an autoimmune disease, an infection, bowel resection, and/or a condition associated with chronic diarrhea. In certain embodiments, the gastrointestinal condition, disease, or disorder is or includes one or more of irritable bowel syndrome (IBS), inflammatory bowel disease (IBD) including Crohn's Disease and colitis, short bowel syndrome (SBS), celiac disease, small intestinal bacterial overgrowth (SIBO), gastroenteritis, leaky gut syndrome, and gastric lymphoma. In certain embodiments, the gastrointestinal condition, disease, or disorder is associated with a bacterial, viral or parasitic infection or overgrowth. In a particular embodiment, the disease or disorder is associated with infection by drug-resistant bacteria, e.g., vancomycin-resistant enterococcus (VRE). In particular embodiments, administration of the at least one prebiotic mixture, e.g., of human milk oligosaccharides, the strain of bacterium capable of producing propionate, e.g., Veillonella sp., and the at least one probiotic strain, e.g., B. longum subsp. infantis, prevents, reduces, or ameliorates one or more symptoms of the gastrointestinal condition.
[0063] In some embodiments, the at least one strain of Bifidobacterium, e.g., B. longum subsp. infantis, the strain of bacterium capable of producing propionate, e.g., Veillonella sp., and the prebiotic mixture, e.g., of human milk oligosaccharides, are administered to a subject with an immune dysfunction. In some embodiments, the subject is immunocompromised. In certain embodiments, the administration prevents, reduces, treats, or ameliorates an infection in the immunocompromised subject. In some embodiments, the
administration prevents, reduces, treats, or ameliorates overgrowth or domination of pathogenic bacteria. In some embodiments, the immunocompromised subject has undergone one or more treatments for cancer. In some embodiments, the treatments are or include chemotherapy. In certain embodiments, the treatment is or includes an allogenic transplant, e.g, a hematopoietic stem cell transplant or bone marrow transplant. In certain embodiments, the immunocompromised subject is in an ICU, has received an organ transplant, is elderly (e.g., at least 65 or 75 years old) and/or has been on prolonged antibiotic treatment (e.g., for at least 2, 3, 4, 6, 8, 10, or 12 weeks, or at least 1, 2, 3, 6, 12, 18, or 24 months). In certain embodiments, the administration prevents or reduces the probability or likelihood of a systemic infection by, by about, or by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 90%, or 95%, e.g., as compared to a subject administered an alternative treatment and/or not administered the probiotic strains (e.g., the Bifidobacterium and the propionate producing bacterium) and/or the prebiotic mixture.
[0064] In particular embodiments, the at least one strain of Bifidobacterium, e.g., B. longum subsp. infantis, the strain of bacterium capable of producing propionate, e.g., Veillonella sp., and the prebiotic mixture, e.g., of human milk oligosaccharides, are administered to treat or prevent overgrowth or domination of pathogenic bacteria (also referred to herein as gut domination). In some aspects, domination of pathogenic bacteria refers to the presence of a species of bacteria (e.g, a pathogenic species), of at least 1%, 5%, 10%, 20%, or 30%, relative to the bacteria present in the subject’s gut or intestinal microbiome. Particular embodiments contemplate that overgrowth or domination may be determined by routine techniques in the art, such as including but not limited to PCR or high throughput sequencing.
[0065] In certain embodiments, the at least one strain of Bifidobacterium, e.g., B. longum subsp. infantis, the strain of bacterium capable of producing propionate, e.g., Veillonella sp., and the prebiotic mixture, e.g., of human milk oligosaccharides, are administered to a subject having, suspected of having, or at risk of having dysbiosis, e.g., of the intestinal microbiome. In certain embodiments, the transient presence, engraftment, or expansion of the Bifidobacterium strain, e.g., B. longum subsp. infantis, and the propionate producing strain, e.g., Veilloinella sp. reduces, decreases, or ameliorates the dysbiosis. Particular embodiments contemplate that the presence, engraftment, or expansion of the Bifidobacterium strain, e.g., B. longum subsp. infantis, and the propionate producing strain, e.g., Veilloinella sp., creates, promotes, or generates an environment and/or one or more
conditions that (i) promotes the presence, growth, or expansion of beneficial microbiota; (ii) decreases the presence, growth, or expansion of pathogenic microbiota; (iii) promotes diversity of microbiota present within the microbiome; or (iv) any or all of (i) through (iii).
[0066] In certain embodiments, administration of the Bifidobacterium strain, e.g., B. longum subsp. infantis, and the propionate producing strain, e.g., Veilloinella sp., reduces the presence or abundance of pathogenic bacteria in the subject’s gut. In certain embodiments, administration of the Bifidobacterium strain, e.g., B. longum subsp. infantis, and the propionate producing strain, e.g., Veilloinella sp., reduces gut domination by pathogenic taxa (e.g., Enterob acteriaceae, Enterococcus, Staphylococcus). In particular embodiments, the growth of the Bifidobacterium strain, e.g., B. longum subsp. infantis, and the propionate producing strain, e.g., Veilloinella sp., within the gut or microbiome reduces the abundance, level, activity, or presence of pathogenic taxa. In certain embodiments, administration of the Bifidobacterium strain, e.g., B. longum subsp. infantis, and the propionate producing strain, e.g., Veilloinella sp. and the prebiotic mixture, e.g., of human milk oligosaccharides, reduces the abundance, level, activity, or presence of pathogenic bacteria and/or taxa by, by about, or by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 90%, 95%, or 100%, e.g., as compared to prior to the administration or as compared to the gut or microbiome of a subject not administered the Bifidobacterium strain, the propionate producing strain, and/or the prebiotic mixture. In particular embodiments, the growth of the Bifidobaclerium ivVm, e.g., B. longum subsp. infantis, within the gut or microbiome increases the amount, level, presence, or concentration of at least one short chain fatty acid, e.g., acetate, propionate, or butyrate, within the gut.
[0067] In certain embodiments, the prebiotic mixture, e.g., of human milk oligosaccharides, the propionate producing strain, e.g., Veilloinella sp., and the Bifidobacterium strain, e.g., B. longum subsp. infantis, are administered to a subject who is at risk of an infection or gut domination, e.g., by pathogenic bacteria. In some embodiments, the subject has an increased risk of infection or gut domination, e.g., as compared to the general population. In certain embodiments the subject is immunocompromised, undergoing an extended antibiotic treatment regimen (e.g., lasting at least 2, 3, 4, 5, 6, 8 10, or 12 weeks or 2, 3, 6, 12, 18, or 24 months), is elderly, is hospitalized e.g., in an intensive care unit (ICU), has received an organ transplant, and/or is immunosuppressed. In certain aspects, the subject will undergo or has received a medical procedure such as a surgery or a chemotherapy that may increase the risk, likelihood, or probability of infection.
[0068] In certain embodiments, administration of the prebiotic mixture, the propionate producing strain, and the Bifidobacterium strain reduces the risk, likelihood, or probability of infection, e.g, by pathogenic bacteria, is reduced by at least 10%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 90%, 99%, or 99.9% as compared to alternative treatments or no treatments, or as compared to administration of the probiotic strains or prebiotic mixture alone. In some embodiments, the prebiotic mixture and the probiotic strains are administered at least once at least 1 hour, 2 hours, 3 hours, 4 hours, 6 hours, 8 hours, 12 hours, 18 hours, 24 hours, 1 day, 2 days, 3 days, 4 days, 5 days, 7 days, 10 days, 1 week, 2 weeks, 4 weeks, 6 weeks, one month, or two months prior to the medical procedure, e.g, surgery or chemotherapy. In particular embodiments, the prebiotic mixture and the probiotic strains are administered at least once during the medical procedure, e.g., surgery or chemotherapy. In certain embodiments, the prebiotic mixture and the probiotic strains are administered at least once at least 1 hour, 2 hours, 3 hours, 4 hours, 6 hours, 8 hours, 12 hours, 18 hours, 24 hours, 1 day, 2 days, 3 days, 4 days, 5 days, 7 days, 10 days, 1 week, 2 weeks, 4 weeks, 6 weeks, one month, or two months after to the medical procedure, e.g., surgery or chemotherapy.
[0069] Pathogenic bacteria may include known microbes with pathogenicity for the gastrointestinal tract, e.g., from esophagus down to rectum. In some embodiments, pathogenic bacteria are or include one or more species, subspecies, or strains of Proteobacteria. In certain embodiments, the pathogenic bacteria may include, but are not limited to strains, species, subspecies, or strains of one or more of Firmicutes, Clostridium, Enter obacteriaceae, Enterococcus, Staphylococcus, Corynebacteria, Salmonella, Shigella, Staphylococcus, Campylobacter (e.g., Campylobacter jejuni), Clostridia, Escherichia coli, Yersinia, Vibrio cholerae, Mycobacterium avium subspecies paratuberculosis, Brachyspira hyodysenteriae, or Law sonia intracellularis . In particular embodiments, administration of the prebiotic mixture and the probiotic strains reduces or decreases the presence, growth, or abundance of pathogenic bacteria within the gut.
[0070] In some embodiments, administration of the prebiotic mixture the propionate producing strain, and the Bifidobacterium strain impairs the growth of one or more pathogens. Such pathogens treated by the provided methods include, but are not limited to, Aeromonas hydr ophila, Bacillus, e.g., Bacillus cereus, Bifidobacterium, Bordetella, Borrelia, Brucella, Burkholderia, C. difficile, Campylobacter, e.g., Campylobacter fetus and Campylobacter jejuni, Chlamydia, Chlamydophila, Clostridium, e.g., Clostridium botulinum,
Clostridium difficile, and Clostridium perfringens, Corynebacterium, Coxiella, Ehrlichia, Enterob acteriaceae, e.g., Carbapenem-resistant Enterobacteriaceae (CRE) and Extended Spectrum Beta-Lactamase producing Enterobacterales (ESBL-E), fluoroquinolone-resistant Enterobacteriaceae, Enterococcus, e.g., vancomycin-resistant enterococcus spp., extended spectrum beta-lactam resistant Enterococci (ESBL), and vancomycin-resistant Enterococci (VRE), Escherichia, e.g., enteroaggregative Escherichia coli, enterohemorrhagic Escherichia coli, enteroinvasive Escherichia coli, enterop athogenic E. coli, enterotoxigenic Escherichia coli (such as but not limited to LT and/or ST), Escherichia coli 0157:H7, and multi-drug resistant bacteria E. coli, Francisella, Haemophilus, Helicobacter, e.g., Helicobacter pylori, Klebsiella, e.g., Klebsiellia pneumonia and multi-drug resistant bacteria Klebsiella, Legionella, Leptospira, Listeria, e.g., Lysteria monocytogenes, Morganella, Mycobacterium, Mycoplasma, Neisseria, Orientia, Plesiomonas shigelloides, Antibiotic-resistant Proteobacteria, Proteus, Pseudomonas, Rickettsia, Salmonella, e.g., Salmonella paratyphi, Salmonella spp., and Salmonella typhi, Shigella, e.g., Shigella spp., Staphylococcus, e.g., Staphylococcus aureus and Staphylococcus spp., Streptococcus, Treponema, Vibrio, e.g., Vibrio cholerae, Vibrio parahaemolyticus, Vibrio spp., and Vibrio vulnificus, and Yersinia, e.g., Yersinia enter ocolitica. At least one of the one or more pathogens can be an antibioticresistant bacterium (ARB), e.g., Antibiotic-resistant Proteobacteria, Vancomycin Resistant Enterococcus (VRE), Carbapenem Resistant Enterobacteriaceae (CRE), fluoroquinoloneresistant Enterobacteriaceae, or Extended Spectrum Beta-Lactamase producing Enterobacterales (ESBL-E).
[0071] In some embodiments, administration of the prebiotic mixture the propionate producing strain, and the Bifidobacterium strain impairs the growth of of antibiotic-resistant bacterium (ARB), Antibiotic-resistant Proteobacteria, Carbapenem-resistant Enterobacteriaceae (CRE), Extended Spectrum Beta-Lactamase producing Enterobacterales (ESBL-E), fluoroquinolone-resistant Enterobacteriaceae, vancomycin-resistant Enterococci (VRE), multi-drug resistant A. coli, or multi-drug resistant Klebsiella.
[0072] In some embodiments, administration of the prebiotic mixture, the propionate producing strain, and the Bifidobacterium strain treat, prevent, or ameliorate an infection or gut domination by one or more of Caproiciproducens, Citrobacter freundii, Citrobacter koseri, Enterobacter aerogenes, Enterobacter cloacae, Enterococcus faecalis, Enterococcus faecium, Escherichia coli, Klebsiella oxytoca, Klebsiella pneumoniae, Morganella morganii, Papillibacter cinnamivorans, Papillibacter, Proteus mirabilis, Serratia marcescens,
Sporobacter, Staphylococcus aureus, Staphylococcus epidermidis, Streptococcus anginosus, Streptococcus australis, Streptococcus constellatus, Streptococcus cristatus, Streptococcus gordonii, Streptococcus infantis, Streptococcus intermedius, Streptococcus mitis, Streptococcus mutans, Streptococcus oligofermentans, Streptococcus oralis, Streptococcus parasanguinis, Streptococcus peroris, Streptococcus pneumoniae, Streptococcus pseudopneumoniae, Streptococcus salivarius, Streptococcus sanguinis, Streptococcus sobrinus, Streptococcus tigurinus, Streptococcus vestibularis, or Syntrophococcus sp. In certain embodiments, administration of the prebiotic mixture, the propionate producing strain, and the Bifidobacterium strain treat, prevent, or ameliorate an infection or gut domination by one or more of Citrobacter freundii, Citrobacter koseri, Enterobacter aerogenes, Enterobacter cloacae, Enterococcus faecalis, Enterococcus faecium, Escherichia coli, Klebsiella oxytoca, Klebsiella pneumoniae, Morganella morganii, Proteus mirabilis, Serratia marcescens, Staphylococcus aureus, Staphylococcus epidermidis, Streptococcus anginosus, Streptococcus australis, Streptococcus constellatus, Streptococcus cristatus, Streptococcus gordonii, Streptococcus infantis, Streptococcus intermedius, Streptococcus mitis, Streptococcus mutans, Streptococcus oligofermentans, Streptococcus oralis, Streptococcus parasanguinis, Streptococcus peroris, Streptococcus pneumoniae, Streptococcus pseudopneumoniae, Streptococcus salivarius, Streptococcus sanguinis, Streptococcus sobrinus, Streptococcus tigurinus, or Streptococcus vestibularis.
[0073] In some embodiments, the condition, disease, or disorder is an immune dysfunction that is an autoimmune disorder. In some embodiments, the autoimmune disorder includes, but is not limited to, acute disseminated encephalomyelitis (ADEM), acute necrotizing hemorrhagic leukoencephalitis, Addison's disease, agammaglobulinemia, alopecia areata, amyloidosis, ankylosing spondylitis, anti-GBM/anti-TBM nephritis, antiphospholipid syndrome (APS), autoimmune angioedema, autoimmune aplastic anemia, autoimmune dysautonomia, autoimmune hemolytic anemia, autoimmune hepatitis, autoimmune hyperlipidemia, autoimmune immunodeficiency, autoimmune inner ear disease (AIED), autoimmune myocarditis, autoimmune oophoritis, autoimmune pancreatitis, autoimmune retinopathy, autoimmune thrombocytopenic purpura (ATP), autoimmune thyroid disease, autoimmune urticarial, axonal & neuronal neuropathies, Balo disease, Behcet's disease, bullous pemphigoid, cardiomyopathy, Castleman disease, celiac disease, Chagas disease, chronic inflammatory demyelinating polyneuropathy (CIDP), chronic recurrent multifocal ostomyelitis (CRMO), Churg-Strauss syndrome, cicatricial
pemphigoid/benign mucosal pemphigoid, Crohn's disease, Cogan's syndrome, cold agglutinin disease, congenital heart block, Coxsackie myocarditis, CREST disease, essential mixed cryoglobulinemia, demyelinating neuropathies, dermatitis herpetiformis, dermatomyositis, Devic's disease (neuromyelitis optica), discoid lupus, Dressier's syndrome, endometriosis, eosinophilic esophagitis, eosinophilic fasciitis, erythema nodosum, experimental allergic encephalomyelitis, Evans syndrome, fibrosing alveolitis, giant cell arteritis (temporal arteritis), giant cell myocarditis, glomerulonephritis, Goodpasture's syndrome, granulomatosis with polyangiitis (GPA), Graves' disease, Guillain-Barre syndrome, Hashimoto's encephalitis, Hashimoto's thyroiditis, hemolytic anemia, Henoch-Schonlein purpura, herpes gestationis, hypogammaglobulinemia, idiopathic thrombocytopenic purpura (ITP), IgA nephropathy, IgG4-related sclerosing disease, immunoregulatory lipoproteins, inclusion body myositis, interstitial cystitis, juvenile arthritis, juvenile idiopathic arthritis, juvenile myositis, Kawasaki syndrome, Lambert-Eaton syndrome, leukocytoclastic vasculitis, lichen planus, lichen sclerosus, ligneous conjunctivitis, linear IgA disease (LAD), lupus (systemic lupus erythematosus), chronic Lyme disease, Meniere's disease, microscopic polyangiitis, mixed connective tissue disease (MCTD), Mooren's ulcer, Mucha-Habermann disease, multiple sclerosis, myasthenia gravis, myositis, narcolepsy, neuromyelitis optica (Devic's), neutropenia, ocular cicatricial pemphigoid, optic neuritis, palindromic rheumatism, PANDAS (Pediatric Autoimmune Neuropsychiatric Disorders Associated with Streptococcus), paraneoplastic cerebellar degeneration, paroxysmal nocturnal hemoglobinuria (PNH), Parry Romberg syndrome, Parsonnage-Tumer syndrome, pars planitis (peripheral uveitis), pemphigus, peripheral neuropathy, perivenous encephalomyelitis, pernicious anemia, POEMS syndrome, polyarteritis nodosa, type I, II, & Ill autoimmune polyglandular syndromes, polymyalgia rheumatic, polymyositis, postmyocardial infarction syndrome, postpericardiotomy syndrome, progesterone dermatitis, primary biliary cirrhosis, primary sclerosing cholangitis, psoriasis, psoriatic arthritis, idiopathic pulmonary fibrosis, pyoderma gangrenosum, pure red cell aplasia, Raynaud's phenomenon, reactive arthritis, reflex sympathetic dystrophy, Reiter's syndrome, relapsing polychondritis, restless legs syndrome, retroperitoneal fibrosis, rheumatic fever, rheumatoid arthritis, sarcoidosis, Schmidt syndrome, scleritis, scleroderma, Sjogren's syndrome, sperm and testicular autoimmunity, stiff person syndrome, subacute bacterial endocarditis (SBE), Susac's syndrome, sympathetic ophthalmia, Takayasu's arteritis, temporal arteritis/giant cell arteritis, thrombocytopenic purpura (TTP), Tolosa-Hunt syndrome, transverse myelitis, type 1 diabetes, asthma, ulcerative colitis,
undifferentiated connective tissue disease (UCTD), uveitis, vasculitis, vesiculobullous dermatosis, vitiligo, and Wegener's granulomatosis.
[0074] In some embodiments, the condition, disease, or disorder is a diarrheal disease including, but not limited to, acute bloody diarrhea (e.g., dysentery), acute watery diarrhea (e.g., cholera), checkpoint inhibitor-associated colitis, diarrhea due to food poisoning, persistent diarrhea, and traveler's diarrhea.
[0075] In some embodiments, administration of the prebiotic mixture, the propionate producing strain, and the Bifidobacterium strain treats or prevents various GI disorders known to result from or be associated or accompanied with dysbiosis of the intestinal microbiome. In certain embodiments, administration of the prebiotic mixture, the propionate producing strain, and the Bifidobacterium strain reduces GI immunoactivation and/or inflammation. In some embodiments, GI immunoactivation and inflammation may be assessed by known methods that are routine in the art. In some embodiments, the condition, disease, or disorder is an inflammatory bowel disease (IBD) or related disease including, but not limited to, Behcet's disease, collagenous colitis, Crohn's disease, diversion colitis, fulminant colitis, intermediate colitis, left-sided colitis, lymphocytic colitis, pancolitis, pouchitis, proctosigmoiditis, short bowel syndrome, ulcerative colitis, and ulcerative proctitis.
[0076] In various embodiments, administration of the prebiotic mixture, the propionate producing strain, and the Bifidobacterium strain treats or prevents various bloodstream infections (BSI). In certain embodiments, administration of the probiotic strains and the prebiotic mixture treats or prevents catheter or intravascular-line infections (e.g., central-line infections). In some embodiments, administration of the probiotic strains and the prebiotic mixture treats or prevents chronic inflammatory diseases.
[0077] In particular embodiments, administration of the prebiotic mixture, the propionate producing strain, and the Bifidobacterium strain treats or prevents meningitis; pneumonia, e.g., ventilator-associated pneumonia; skin and soft tissue infections; surgical- site infections; urinary tract infections (e.g., antibiotic-resistant urinary tract infections and catheter-associated urinary tract infections); wound infections; and/or antibiotic-resistant infections and antibiotic-sensitive infections.
[0078] In certain embodiments, administration of the prebiotic mixture, the propionate producing strain, and the Bifidobacterium strain treats or prevents diseases or disorders relating to the "gut-brain axis", including neurodegenerative, neurodevelopmental, and neurocognitive disorders, such as anorexia, anxiety, autism-spectrum disorder,
depression, Parkinson's, and Schizophrenia. In certain embodiments, administration of the probiotic strains and the prebiotic mixture reduces one or more symptoms associated with anorexia, anxiety, autism-spectrum disorder, depression, Parkinson's, and/or Schizophrenia.
[0079] In some embodiments, administration of the prebiotic mixture, the propionate producing strain, and the Bifidobacterium strain treats or prevents a side effect of an anticancer therapy and/or increases efficacy of an anti -cancer therapeutic agent and/or anticancer therapy. In some embodiments, the anti -cancer therapy is surgery, radiation therapy, chemotherapy (including hormonal therapy) and/or targeted therapy (including an immunotherapy). Illustrative chemotherapeutics agents are provided elsewhere herein. In particular embodiments, the immunotherapy binds to and/or recognizes a tumor-cell antigen and/or a cancer-cell antigen, e.g., CTLA-4, PD-1, PD-L1, or PD-L2. In some embodiments, the immunotherapy comprises administration of Keytruda (Pembrolizumab), Opdivo (Nivolumab), Yervoy (Ipilimumab), Tecentriq (atezolizumab), Bavencio (avelumab), and Imfinzi (durvalumab).
[0080] In some embodiments, the subject is refractory and/or non-responsive to an anti-cancer therapy. In certain embodiments, the prebiotic mixture, the propionate producing strain, and the Bifidobacterium strain treats a subject that presents a non-curative response, a limited response, or no response to the anti-cancer therapy, or even progress, after 12 weeks or so of receiving the anti -cancer therapy. Thus, in some aspects, the provided probiotic strains and prebiotic mixture of the present invention can rescue subjects that are refractory and/or non-responsive to the anti-cancer therapy. In certain embodiments, the subject is refractory and/or non-responsive to a treatment directed to a checkpoint molecule, e.g., CTLA-4, PD-1, PD-L1, and/or PD-L2. In particular embodiments, the treatment directed to a checkpoint molecule comprises administration of Keytruda (Pembrolizumab), Opdivo (Nivolumab), Yervoy (Ipilimumab), Tecentriq (atezolizumab), Bavencio (avelumab), or Imfinzi (durvalumab).
[0081] In some embodiments, the prebiotic mixture, c.g, of human milk oligosaccharides, the propionate producing strain, e.g., Veilloinella sp., and the at least one Bifidobacterium strain, e.g, B. longum subsp. infantis, are administered to an immunocompromised subject. In certain embodiments, the administration prevents, reduces, treats, or ameliorates an infection in the immunocompromised subject. In some embodiments, the administration prevents, reduces, treats, or ameliorates overgrowth or domination of pathogenic bacteria. In some embodiments, the immunocompromised subject
has undergone one or more treatments for cancer. In some embodiments, the treatments are or include chemotherapy. In certain embodiments, the treatment is or includes an allogenic transplant, e.g., a hematopoietic stem cell transplant or bone marrow transplant. In certain embodiments, the administration prevents or reduces the probability or likelihood of a systemic infection by, by about, or by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 90%, or 95%, e.g., as compared to an alternative treatment or treatment with either the probiotic strains or prebiotic mixture alone.
[0082] In certain embodiments, the prebiotic mixture, e.g., of human milk oligosaccharides, the propionate producing strain, e.g., Veilloinella sp., and the at least one Bifidobacterium strain, e.g, B. longum subsp. infantis, are administered to a subject who has or is at risk of sepsis. In some embodiments, the probability or likelihood of sepsis is reduced or decreased by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 99%, e.g, as compared to a subject (e.g., who has or is at risk for sepsis) not administered the prebiotic mixture or the probiotics. In certain embodiments, the administration of the prebiotic mixture and the probiotics improves or increases the survival of the subject over 6 months, 12 months, 18 months, 1 year, 2 years, 5 years, 10 years, and/or 20 years or more by, by about, or by at least 5%, 10%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 90%, 95%, 100%, or 1-fold, 2-fold, 3-fold, 4-fold, or 5-fold greater than in subjects (e.g., who have or are at risk for sepsis) not administered the prebiotic mixture and the probiotic strains.
[0083] In particular embodiments, administration of the prebiotic mixture, the propionate producing strain, and the Bifidobacterium strain prevents, reduces, decreases, remedies, or ameliorates one or more symptoms associated with a gastrointestinal condition, disease, or disorder. In certain embodiments, the one or more symptoms associated with gastrointestinal condition, disease, or disorder may include, but are not limited to, diarrhea, fever, fatigue, abdominal pain and cramping, blood in stool, mouth sores, weight loss, fistula, inflammation (of skin, eyes, or joints), inflamed liver or bile ducts, delayed growth (in children). In particular embodiments, administration of the prebiotic mixture, the propionate producing strain, and the Bifidobacterium strain reduces the risk or probability for the subject of experiencing one or more symptoms associated with the gastrointestinal condition, disease, or disorder by, by about, or by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 90%, or 95%, e.g., as compared to a subject not administered the probiotic strains and/or the prebiotic mixture. In certain embodiments, administration of the prebiotic mixture and the probiotic strains increases probability or likelihood for remission by, by about, or by at least
10%, 20%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 90%, 100%, or 1-fold, 2-fold, 5-fold, 10- fold, 20-fold, 50-fold, or 100-fold e.g., as compared to a subject not administered the probiotic strains and/or the prebiotic mixture. In some embodiments, administration of the prebiotic mixture, the propionate producing strain, and the Bifidobacterium strain increases probability or likelihood for remission within 12 weeks, 10 weeks, 8 weeks, 6 weeks, 4 weeks, or less than 4 weeks, e.g., from the initiation or termination of the administration.
[0084] In various embodiments, the prebiotic mixture, e.g., of human milk oligosaccharides, the propionate producing strain, e.g., Veilloinella sp., and the at least one Bifidobacterium strain, e.g, B. longum subsp. infantis, are administered to a subject to treat, ameliorate, remedy, or prevent a chronic inflammatory disease, an autoimmune disease, an infection, bowel resection, and/or a condition associated with chronic diarrhea. According to particular embodiments, the pathology is selected from the group consisting of: irritable bowel syndrome (IBS), inflammatory bowel disease (IBD), short bowel syndrome (SBS), celiac disease, small intestinal bacterial overgrowth (SIBO), gastroenteritis, leaky gut syndrome, and gastric lymphoma. In another embodiment the disease or disorder is associated with a bacterial, viral, or parasitic infection or overgrowth, e.g., by drug-resistant bacteria. In some embodiments, administration of the prebiotic mixture the propionate producing strain, and the Bifidobacterium strain increases probability or likelihood for cure or remission of the chronic inflammatory disease, autoimmune disease, infection, bowel resection, and/or chronic diarrhea for by, by about, or by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 90%, 100%, or 1-fold, 2-fold, 5-fold, 10-fold, 20-fold, 50-fold, or 100-fold e.g, as compared to a subject not administered the probiotic strains and/or the prebiotic mixture. In some embodiments, administration of the prebiotic mixture the propionate producing strain, and the Bifidobacterium strain increases probability or likelihood for the cure or remission within 12 weeks, 10 weeks, 8 weeks, 6 weeks, 4 weeks, or less than 4 weeks, e.g., from the initiation or termination of the administration.
[0085] In certain embodiments, the prebiotic mixture, the propionate producing strain, and the Bifidobacterium strain are administered to a subject to treat, prevent, or ameliorate an allergy. In some embodiments, the allergy is a food allergy. In certain embodiments, the food allergy is or includes a chronic or acute immunological hypersensitivity reaction (e.g., a type I hypersensitivity reaction) elicited in a mammal in response to an ingested material or food antigen (also referred to in the art as a “food allergen”). Identification and diagnosis of food allergy is routine among persons of ordinary skill in the art. Food allergies may include,
but are not limited to, allergies to nuts, peanuts, shellfish, fish, milk, eggs, wheat, or soybeans.
[0086] In some embodiments, the prebiotic mixture the propionate producing strain, and the Bifidobacterium strain are administered to treat or ameliorate an allergy, e.g., a food allergy. In certain embodiments, the prebiotic mixture, the propionate producing strain, and the Bifidobacterium strain reduce or decrease the severity of the allergic response to the allergen, e.g., as compared to the allergic response prior to any treatment with the probiotic strains and prebiotic mixture. In certain embodiments, the prebiotic mixture the propionate producing strain, and the Bifidobacterium strain attenuates or reduces the severity or intensity of one or more symptoms or clinical manifestations of the allergy, e.g., food allergy, to subsequent exposures to the allergen, e.g., as compared to symptoms or clinical manifestations observed prior to treatment with the probiotic strains and prebiotic mixture. In some embodiments, the symptoms or clinical manifestations of the allergy may include, but are not limited to rash, eczema, atopic dermatitis, hives, urticaria, angiodema, asthma, rhinitis, wheezing, sneezing, dyspnea, swelling of the airways, shortness of breath, other respiratory symptoms, abdominal pain, cramping, nausea, vomiting, diarrhea, melena, tachycardia, hypotension, syncope, seizures, and anaphylactic shock.
[0087] In particular embodiments, the prebiotic mixture, the propionate producing strain, and the Bifidobacterium strain are administered to a subject, e.g., a subject at risk of having or developing an allergy, to prevent or reduce or decrease the probability or likelihood experiencing an allergic response. In certain embodiments, administration of the probiotic strains and prebiotic mixture reduce the likelihood or probability of having an allergic response within the next month, 3 months, 6 months, 12 months, 18 months, year, 2 years, 3 years, 5 years, 10 years, or 20 years. In some embodiments, the probability or likelihood of developing the allergy is reduced by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 90%, or 99% as compared to a subject with a similar risk profile who is not administered the probiotic strains and the prebiotic mixture. In some embodiments, administration of the probiotic strains and prebiotic mixture reduces the severity of one or more symptoms or clinical manifestations of an allergic response following exposure to the allergen over the next month, 3 months, 6 months, 12 months, 18 months, year, 2 years, 3 years, 5 years, 10 years, or 20 years, e.g., as compared to exposure of the allergen to a subject with the same or similar allergy who was not administered the probiotic strains and the prebiotic mixture.
[0088] In some embodiments, the prebiotic mixture, e.g., of human milk oligosaccharides, the propionate producing strain, e.g., Veilloinella sp., and the at least one Bifidobacterium strain, e.g., B. longum subsp. infantis, are administered to a subject to treat, ameliorate, remedy, or prevent pouchitis. In certain aspects, pouchitis is inflammation that occurs in the lining of a pouch created during surgery to treat ulcerative colitis or certain other diseases. In some embodiments, the surgery is or includes removal of a diseased colon or portion thereof. In certain embodiments, the surgery is a J pouch surgery (ileoanal anastomosis — IPAA).
[0089] In some embodiments, the prebiotic mixture, e.g., of human milk oligosaccharides, the propionate producing strain, e.g., Veilloinella sp., and the at least one Bifidobacterium strain, e.g., B. longum subsp. infantis, are administered to a subject to treat, ameliorate, remedy, or prevent pouchitis in a subject in need thereof, e.g., a subject who has undergone an IPAA surgery. In particular embodiments, administration of the prebiotic mixture and the probiotic strains prevents, reduces, decreases, remedies, or ameliorates one or more symptoms associated with pouchitis. In certain embodiments, the one or more symptoms associated with pouchitis may include, but are not limited to, increased stool frequency, tenesmus, straining during defecation, blood in the stool, incontinence, seepage of waste matter during sleep, abdominal cramps, pelvic or abdominal discomfort, or tail bone pain. In certain embodiments, symptoms associated with more severe pouchitis include, but are not limited to, fever, dehydration, malnutrition, fatigue, iron-deficiency anemia, or joint pain. In particular embodiments, administration of the prebiotic mixture and the at least one probiotic strain reduces the risk or probability for the subject of experiencing pouchitis by, by about, or by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 90%, or 95%, e.g., as compared to a subject not administered the probiotic strains and/or the prebiotic mixture.
[0090] In various embodiments, the prebiotic mixture, e.g., of human milk oligosaccharides, the propionate producing strain, e.g., Veilloinella sp., and the at least one Bifidobacterium strain, e.g., B. longum subsp. infantis, are administered to a subject to treat, ameliorate, remedy, or prevent a chronic inflammatory disease, an autoimmune disease, an infection, bowel resection, and/or a condition associated with chronic diarrhea. Such pathology includes, but is not limited to: irritable bowel syndrome (IBS), inflammatory bowel disease (IBD), short bowel syndrome (SBS), celiac disease, small intestinal bacterial overgrowth (SIBO), gastroenteritis, leaky gut syndrome, and gastric lymphoma. In some embodiments the disease or disorder is associated with a bacterial, viral, or parasitic infection
or overgrowth, e.g., by drug-resistant bacteria. In some embodiments, administration of the prebiotic mixture and the probiotic strains increases probability or likelihood for cure or remission of the chronic inflammatory disease, autoimmune disease, infection, bowel resection, and/or chronic diarrhea for by, by about, or by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 90%, 100%, or 1-fold, 2-fold, 5-fold, 10-fold, 20-fold, 50-fold, or 100-fold e.g., as compared to a subject not administered the probiotic strains and prebiotic mixture and/or a subject administered an alternative therapy. In some embodiments, administration of the prebiotic mixture and the probiotic strains increases probability or likelihood for the cure or remission within 12 weeks, 10 weeks, 8 weeks, 6 weeks, 4 weeks, or less than 4 weeks, e.g., from the initiation or termination of the administration e.g., as compared to a subject not administered the probiotic strains and prebiotic mixture and/or a subject administered an alternative therapy.
[0091] In some embodiments, the prebiotic mixture, e.g., of human milk oligosaccharides, the propionate producing strain, e.g., Veilloinella sp., the Bifidobacterium strain, e.g., B. longum subsp. infantis, are administered to a subject to treat, ameliorate, remedy, or prevent pouchitis. In certain aspects, pouchitis is inflammation that occurs in the lining of a pouch created during surgery to treat ulcerative colitis or certain other diseases. In some embodiments, the surgery is or includes removal of a diseased colon or portion thereof. In certain embodiments, the surgery is a J pouch surgery (ileoanal anastomosis — IPAA).
[0092] In some embodiments, the prebiotic mixture, e.g., of human milk oligosaccharides, the propionate producing strain, e.g., Veilloinella sp., and the Bifidobacterium strain, e.g., B. longum subsp., are administered to a subject to treat, ameliorate, remedy, or prevent pouchitis in a subject in need thereof, e.g., a subject who has undergone an IPAA surgery. In particular embodiments, administration of the prebiotic mixture and the probiotic strains prevents, reduces, decreases, remedies, or ameliorates one or more symptoms associated with pouchitis. In certain embodiments, the one or more symptoms associated with pouchitis may include, but are not limited to, increased stool frequency, tenesmus, straining during defecation, blood in the stool, incontinence, seepage of waste matter during sleep, abdominal cramps, pelvic or abdominal discomfort, or tail bone pain. In certain embodiments, symptoms associated with more severe pouchitis include, but are not limited to, fever, dehydration, malnutrition, fatigue, iron-deficiency anemia, or joint pain. In particular embodiments, administration of the prebiotic mixture, the Bifidobacterium strain, and the propionate producing strain reduces the risk or probability for the subject of
experiencing pouchitis by, by about, or by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 90%, or 95%, e.g, as compared to a subject not administered the probiotic strains and/or the prebiotic mixture.
[0093] In some embodiments, the subject is a patient in an intensive care unit (ICU). In some embodiments, the subject is an organ transplant recipient. In some embodiments, the subject is a geriatric patient (e.g., at least 65, 70, 75, 80, or 85 years old). In some embodiments, the subject has received prolonged antibiotic treatment (e.g., at least 2, 3, 4, 5, 6, 8, 10, or 12 weeks, or at least 1, 2, 3, 6, 12, 18, or 24 months). In some embodiments, the subject is a recipient of a broad-spectrum antibiotic treatment. In some embodiments, the subject is a recipient, or recent recipient (e.g., within at least 1, 2, 3, 4, 5, 6, or 7 days, or within at least 1, 2, 3, or 4 weeks), of parenteral nutrition (e.g., total parenteral nutrition or partial parenteral nutrition). In some embodiments, the subject is a recipient of enteral nutrition.
[0094] In particular embodiments, provided herein are methods of preventing or reducing the incidence or severity of graft versus host disease (GVHD) in a subject in need thereof. In certain embodiments, the provided methods prevent or reduce incidence or severity of GVHD in a subject that has received or will receive an allogenic stem cell transplant. In some embodiments, the provided mixtures of HMOs are formulated to be administered to subjects who have, are, or will undergo an allogenic transplant, e.g., BMT or HSCT. In some embodiments, the at least one strain of Bifidobacterium and the at least one propionate producing strain are formulated to be administered to subject who has underwent, is undergoing, or will undergo an allogenic transplant. In particular embodiments, at least one strain of Bifidobacterium, e.g., such as any described herein, e.g., in Section I-B, the strain of bacterium capable of producing propionate, such as any of the propionate producing bacterium described herein, e.g., in Section I-A, and the prebiotic mixture, such as any described herein, e.g, in Section I-C, are administered to a subject who will undergo or who has undergone an allogenic transplant.
[0095] In certain embodiments, the subject is a human infant, child, adolescent, or adult. In particular embodiments, the subject is at risk or suspected of being at risk of having GVHD. In some embodiments, the GHVD is associated with, or accompanied by, an allogenic transplant, such as an allogenic bone marrow transplant (BMT) or an allogenic hematopoietic stem cell transplant (HSCT).
[0096] In some embodiments, at least one strain of the Bifidobacterium, e.g., B. longum subsp. infantis, a strain of bacterium capable of producing propionate, e.g., a Veillonella sp. and a prebiotic mixture, e.g., of human milk oligosaccharides, is administered to a subject has undergone or will undergo an allogenic stem cell transplant. In certain embodiments, the allogenic transplant is a bone marrow transplant (BMT). In particular embodiments, the allogenic transplant is a hematopoietic stem cell transplantation (HSCT). In particular embodiments, the subject has undergone the allogenic stem cell transplant within 12 weeks, 8 weeks, 6 weeks, 4 weeks, 3 weeks, 2 weeks, 14 days, 12 days 10 days, 7 days, 5 days, 4 days, 3 days, 2 days, or 1 day prior to administration of a first dose of the prebiotic mixture or the at least one probiotic strain. In certain embodiments, the first dose of the prebiotic mixture or the at least one probiotic strain is administered within 12 weeks, 8 weeks, 6 weeks, 4 weeks, 3 weeks, 2 weeks, 14 days, 12 days 10 days, 7 days, 5 days, 4 days, 3 days, 2 days, or 1 day prior to receiving the allogenic stem cell transplant.
[0097] In some embodiments, provided herein are methods for treating, preventing, or ameliorating GVHD in a subject in need thereof. In certain embodiments, provided herein are methods for treating, preventing, or ameliorating a condition or disease associated or accompanied with GVHD in a subject in need thereof. In certain embodiments, provided herein are methods for treating, preventing, reducing, decreasing, or ameliorating the severity or presence of one or more symptoms associated with GVHD or a disease or condition associated or accompanied with GVHD in a subject in need thereof.
[0098] In particular embodiments, administration of the Bifidobacterium, e.g., B. longum subsp. infantis, a propionate producing strain, e.g., a Veillonella sp., and a prebiotic mixture, e.g., of human milk oligosaccharides, reduces or decreases the probability or likelihood of experiencing GVHD. In certain embodiments, the probability or likelihood is reduced or decreased by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 99%, e.g., as compared to a subject not administered the prebiotic mixture or the probiotics. In certain embodiments, the probability or likelihood of experiencing GVHD within 20 years, 10 years, 7 years, 5 years, 2 years or 1 year, or within the subject’s lifetime, is reduced or decreased, e.g., as compared to a subject not administered the prebiotic mixture or the probiotics.
[0099] In certain embodiments, the Bifidobacterium, e.g., B. longum subsp. infantis, a propionate producing strain, e.g., a Veillonella sp. and a prebiotic mixture, e.g., of human milk oligosaccharides, are administered to decrease or reduce mortality associated with an
allogenic transplant, e.g., BMT or HSCT, or with GVHD. In some embodiments, the Bifidobacterium, e.g., B. longum subsp. infantis, the propionate producing strain, e.g., a Veillonella sp. and the prebiotic mixture, e.g., of human milk oligosaccharides are administered to increase survival of subjects who undergo an allogenic transplant, e.g., BMT or HSCT. In particular embodiments, administration improves or increases the survival of the subject over 6 months, 12 months, 18 months, 1 year, 2 years, 5 years, 10 years, and/or 20 years or more by, by about, or by at least 5%, 10%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 90%, 95%, 100%, or 1-fold, 2-fold, 3-fold, 4-fold, or 5-fold greater than in subjects (e.g., subjects who received an allogenic transplant, e.g., BMT or HSCT) not administered the prebiotic mixture and the probiotic strains.
[0100] In some embodiments, the Bifidobacterium, e.g., B. longum subsp. infantis, a propionate producing strain, e.g., the Veillonella sp. And the prebiotic mixture, e.g., of human milk oligosaccharides, are administered to treat, prevent, ameliorate, reduce, or decrease the severity, occurrence, or likelihood of experiencing one or more symptoms, e.g., symptoms associated with or accompanying GHVD. In particular embodiments, the GVHD is acute GVHD. In some embodiments, the GVHD is chronic GVHD. In particular embodiments, the symptoms of GVHD are or include, but are not limited to, a rash, such as with burning or itching sensation; blistering, e.g., of the skin; flaking of the skin; nausea; vomiting; abdominal cramps; loss of appetite; diarrhea; and jaundice. In some embodiments, the symptoms of GVHD are or include, but are not limited to dry mouth, mouth ulcers, difficulty eating, gum disease, tooth decay, rash, itchy sensation, thickening and tightening of the skin, jaundice, changes in skin coloration, hair loss, premature gray hair, loss of body hair, loss of appetite, unexplained weight loss, nausea, vomiting diarrhea, stomach pain, shortness of breath, difficulty breathing, persistent or chronic cough, wheezing, impaired liver function, abdominal swelling, muscle weakness, muscle cramps, and joint stiffness. In particular embodiments, administration of the Bifidobacterium, e.g., B. longum subsp. infantis, the propionate producing strain, e.g., a Veillonella sp., and the prebiotic mixture, e.g., of human milk oligosaccharides treats, prevents, ameliorates, reduces, or decreases the severity, occurrence, or likelihood of the one or more symptoms as compared to what is observed in subjects (e.g., subjects who have had or will undergo an allogeneic transplant) that are not administered the Bifidobacterium, the propionate producing strain, and the prebiotic mixture. In some aspects, the presence, occurrence, and severity of a symptom may be recognized, identified, or scored by skilled person (e.g., a healthcare practitioner) as a matter of routine.
[0101] In certain embodiments, the Bifidobacterium, e.g., B. longum subsp. infantis, and the prebiotic mixture, e.g., of human milk oligosaccharides, are administered to treat, prevent, ameliorate, reduce, or decrease the severity, occurrence, or likelihood of experiencing one or more symptoms, e.g., symptoms associated with or accompanying acute radiation syndrome (ARS). In particular embodiments, a propionate producing strain, e.g., the Veillonella sp., is administered with or concurrent to the administration of the Bifidobacterium and the prebiotic mixture.
[0102] In some embodiments, the subject has or is suspected of having ARS. In certain embodiments, the subject has been exposed or is suspected of having been exposed to high doses of ionizing radiation. Subjects may be exposed to high doses of ionizing radiation under various circumstances that may include but are not limited to accidental, medical, or terrorist incidents. In humans, ARS resulting from such exposure may present as hematopoietic dysfunction, gastrointestinal damage, and/or neurovascular injury. Particular embodiments contemplate that while medical countermeasures exist to treat hematopoietic dysfunctions, there are currently no approved therapies to treat or prevent gastrointestinal damage associated with ARS.
[0103] In particular embodiments, the Bifidobacterium, e.g., B. longum subsp. infantis, and the prebiotic mixture, e.g., of human milk oligosaccharides, are administered to prevent, treat, and/or reduce the severity of one or more symptoms associated with ARS. In some embodiments, the at least one propionate producing bacterium, e.g., Veillonella sp., is also administered. In certain embodiments, the symptoms are or include symptoms associated with hematopoietic or bone marrow syndrome and/or damage, gastrointestinal syndrome and/or damage, and/or cardiovascular/central nervous system (CNS) syndrome and/or damage. In some embodiments, the symptoms of hematopoietic syndrome and/or hematopoietic damage are or include anorexia, nausea and vomiting, fever, malaise, and/or drops in blood cell counts (e.g., red, white, or both). In certain embodiments, the symptoms of gastrointestinal syndrome and/or gastrointestinal damage are or include anorexia, severe nausea, vomiting, cramps, diarrhea, malaise, fever, dehydration, and/or electrolyte imbalance. In particular embodiments, the symptoms of cardiovasculature/CNS syndrome and/or cardiovasculature/CNS damage are or include extreme nervousness and confusion, severe nausea, vomiting, and diarrhea, loss of consciousness, burning sensations on the skin, convulsions, and/or coma. In certain embodiments, the administration prevents the onset of one or more symptoms associated with ARS. In particular embodiments, the administration
reduces the severity or intensity of one or more symptoms associated with ARS, e.g., as compared to an unadministered subject exposed to a similar dose of ionizing radiation.
[0104] In certain embodiments, the Bifidobacterium, e.g., B. longum subsp. inf amis. and the prebiotic mixture, e.g., of human milk oligosaccharides, are administered to prevent, treat, and/or reduce the severity of gastrointestinal damage associated with ARS and/or associated with exposure to a high dose of ionizing radiation. In some embodiments, the at least one propionate producing bacterium, e.g., Veillonella sp., is also administered. In certain embodiments, the administration preserves gut barrier integrity and development of the gastrointestinal tract resulting from or associated with ARS or a high dose of ionizing radiation, e.g., as compared to an untreated subject exposed to the same or similar ionizing radiation. In particular embodiments, administration prevents or reduces the likelihood of infection due to pathogen outgrowth and/or translocation associated with ARS or a high dose of ionizing radiation, e.g., as compared to an untreated subject exposed to the same or similar ionizing radiation.
[0105] In some embodiments, the subject has been exposed to a high dose of ionizing radiation. For example, exposure to ionizing radiation doses of 0.3 Gray (Gy) or 30 rads may be sufficient to cause or induce symptoms associated with ARS, and doses of at least 0.7 Gray (Gy) or 70 rads may generally be considered as sufficient to cause or induce ARS. In certain embodiments, the dose is at least 0.3 Gy, 0.5 Gy, 0.7 Gy, 1.0 Gy, 2.0 Gy, 3.0 Gy, 4.0 Gy, 5.0 Gy, 6.0 Gy, 7.0 Gy, 8.0 Gy, 9.0 Gy, 10 Gy, 15 Gy, 20 Gy, 25 Gy, 30 Gy, 40 Gy, or at least 50 Gy. In particular embodiments, the dose is at least 30 rad, 50 rad, 70 rad, 100 rad, 150 rad, 200 rad, 250 rad, 300 rad, 500 rad, 600 rad, 700 rad, 750 rad, 800 rad, 900 rad, 1,000 rad, 2,000 rad, 3,000 rad, 4,000 rad, or at least 5,000 rad. In some embodiments, the dose is associated with an external source of radiation, e.g., a source outside of the subject’s body. In particular embodiments, the ionizing radiation dose may be penetrating and/or able to reach the subject’s internal organs, and may include, but are not limited to, high energy X-rays, gamma rays, and/or neutrons. In some embodiments, the subject may be exposed to the ionizing radiation dose over of relatively short amount of time, e.g., within minutes, such as over an amount of time less than one hundred eighty minutes, one hundred twenty minutes, ninety minutes, sixty minutes, thirty minutes, fifteen minutes, ten minutes, five minutes, three minutes, two minutes, or less than one minute.
[0106] In some embodiments, the Bifidobacterium, e.g., B. longum subsp. inf antis. and the prebiotic mixture, e.g., of human milk oligosaccharides, are administered to prevent
or reduce mortality associated with and/or resulting from ARS. In certain embodiments, the at least one propionate producing bacterium, e.g., Veillonella sp., is also administered. In some embodiments, at least one butyrate producing bacterium, such as those described herein, e.g., in Section I-D such as Anaerostipes caccae, Clostridium innocuum, Roseburia hominis, o Roseburia inleslinalis. is also administered. In certain embodiments, B. longum subsp. infantis, the prebiotic mixture of human milk oligosaccharides, and the propionate producing bacterium are administered to prevent or reduce mortality associated with and/or resulting from ARS. In some embodiments, B. longum subsp. infantis, the prebiotic mixture of human milk oligosaccharides, and the butyrate producing bacterium are administered to prevent or reduce mortality associated with and/or resulting from ARS. In various embodiments, B. longum subsp. infanlis. the prebiotic mixture of human milk oligosaccharides, the propionate producing bacterium, and the butyrate producing bacterium are administered to prevent or reduce mortality associated with and/or resulting from ARS.
[0107] In some embodiments, the administration improves survival in subjects that have or are suspected of having ARS and/or that have been exposed or are suspected of having been exposed to a high dose of ionizing radiation. In particular embodiments, the subjects’ survival rate is about or at least 50%, 60%, 75%, 80%, 90%, or at least 95% or more after about, at least, or at least about two weeks, four weeks, six weeks, eight weeks, ten weeks, twelve weeks, two months, three months, four months, six months, nine months, twelve months, sixteen months, eighteen months, one year, two years, or three years from the exposure to the dose of ionizing radiation and/or the initial onset of one or more ARS symptoms. In certain embodiments, the subjects who are administered have a survival rate that at least 5%, 10%, 20%, 25%, 30%, 40%, 50%, 60%, 75%, 80%, 90%, or at least 95%, or at least 1-fold, 1.5 fold, 2 fold, 3-fold, 4-fold, or 5-fold or more after about, at least, or at least about two weeks, four weeks, six weeks, eight weeks, ten weeks, twelve weeks, two months, three months, four months, six months, nine months, twelve months, sixteen months, eighteen months, one year, two years, or three years from the exposure to the dose of ionizing radiation and/or the initial onset of one or more ARS symptoms than is observed in unadmini stered subj ects .
III. PHARMACEUTICAL FORMULATIONS
[0108] In certain embodiments, the provided at least one Bifidobacteria, e.g., B. longum subsp. infantis, the provided propionate producing bacterium, e.g., Veillonella sp., and the provided prebiotic mixture, e.g, of human milk oligosaccharides, are formulated together or separately, e.g, for administering to a human subject. In some embodiments, the at least one Bifidobacteria, the provided propionate producing bacterium, and the provided prebiotic mixture, are formulated together in the same pharmaceutical composition. In certain embodiments, the at least one Bifidobacterium and the prebiotic mixture are formulated together into the same pharmaceutical composition. In particular embodiments, the at least one Bifidobacterium and the at least one propionate producing bacterium are formulated together into the same pharmaceutical composition. In certain embodiments, the at least one Bifidobacteria, the provided propionate producing bacterium, and the provided prebiotic mixture are formulated separately into separate pharmaceutical compositions.
[0109] The compositions described herein, e.g., any or all of the at least one Bifidobacterium, the at least one propionate producing bacterium, and/or the prebiotic mixture, may be formulated in a conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries, which facilitate processing of the active ingredients into compositions for pharmaceutical use. Methods of formulating pharmaceutical compositions are known in the art (see, e.g., “Remington's Pharmaceutical Sciences,” Mack Publishing Co., Easton, Pa.). In some embodiments, the compositions described herein are subjected to tableting, lyophilizing, direct compression, conventional mixing, dissolving, granulating, levigating, emulsifying, encapsulating, entrapping, or spray drying to form tablets, granulates, nanoparticles, nanocapsules, microcapsules, microtablets, pellets, or powders, which may be enterically coated or uncoated. Appropriate formulation depends on the route of administration.
[0110] The probiotic strains (e.g., the provided Bifidobacterium and/or the provided propionate producing bacterium) and prebiotic mixture described herein may be formulated into pharmaceutical compositions in any suitable dosage form (e.g., liquids, capsules, sachet, hard capsules, soft capsules, tablets, enteric coated tablets, suspension powders, granules, or matrix sustained release formations for oral administration) and for any suitable type of administration (e.g., oral, topical, injectable, immediate-release, pulsatile-release, delay ed- release, or sustained release). Suitable dosage amounts for the provided probiotics may range
from about 105 to 1012 bacteria, e.g., at, at about, or at least 105 bacteria, 106 bacteria, 107 bacteria, 108 bacteria, 109 bacteria, IO10 bacteria, IO11 bacteria, or 1012 bacteria, or more.
[OHl] In some embodiments, the prebiotic mixture, e.g., prebiotic mixture of human milk oligosaccharides, is administered to the subject generally in the range of about 20 mg to about 20 g, e.g., total prebiotic weight (such as weight of total human milk oligosaccharides) per dose. In certain embodiments, between or between about 50 mg and 10 g, 100 mg and 7.5 g, 500 mg to 5 g, 1 g and 2.5 g, 50 mg and 20 g, 100 mg and 15 g, 500 mg and 10 g, or 1 g and 7.5 g prebiotic weight per dose. In some embodiments, during an initial treatment phase, the dosing can be higher; for example, 100 mg to 20 g, 100 mg to 30 g, 500 mg to 15 g, 1 g to 10 g, or 2.5 g to 7.5 g per dose. During a secondary treatment phase, the dosing can be reduced; for example, in certain embodiments, to 20 mg to 10 g per dose, 100 mg to 7.5 g per dose, 500 mg to 2.5 g per dose, 750 mg to 1.5 g per dose, 20 mg to 20 g per dose, 100 mg to 10 g per dose, 500 mg to 7.5 g per dose, or 750 mg to 5 g per dose. In certain embodiments, the prebiotic mixture is administered to the subject in an amount of or about 5 g per day. In some embodiments, a dose of the prebiotic mixture is administered at least once per month, once per week, or once per day. In some embodiments, a dose of the prebiotic mixture is administered at least once, twice, three times, four times, five times, six times, eight times, ten times, or twelve times daily.
[0112] In some embodiments, the pharmaceutical compositions may be administered once or more daily, weekly, or monthly. The probiotics (e.g., the at least one Bifidobacterium and/or the at least one propionate producing bacteria) may be formulated, together or separately, into pharmaceutical compositions comprising one or more pharmaceutically acceptable carriers, thickeners, diluents, buffers, buffering agents, surface active agents, neutral or cationic lipids, lipid complexes, liposomes, penetration enhancers, carrier compounds, and other pharmaceutically acceptable carriers or agents. For example, the pharmaceutical composition may include, but is not limited to, the addition of calcium bicarbonate, sodium bicarbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils, polyethylene glycols, and surfactants, including, for example, polysorbate 20. In some embodiments, the probiotic strain may be formulated in a solution of sodium bicarbonate, e.g., 1 molar solution of sodium bicarbonate (to buffer an acidic cellular environment, such as the stomach, for example). The prebiotic mixture may be administered and formulated as neutral or salt forms. Pharmaceutically acceptable salts include those formed with anions such as those derived from hydrochloric, phosphoric,
acetic, oxalic, tartaric acids, etc., and those formed with cations such as those derived from sodium, potassium, ammonium, calcium, ferric hydroxides, isopropylamine, tri ethyl amine, 2- ethylamino ethanol, histidine, procaine, etc.
[0113] In some embodiments, the pharmaceutical compositions containing the provided at least one Bifidobacterium, the at least one propionate producing bacterium, and the prebiotic mixture, e.g., together or as separate compositions, may be administered orally and formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions, etc. Pharmacological compositions for oral use can be made using a solid excipient, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries if desired, to obtain tablets or dragee cores. Suitable excipients include, but are not limited to, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose compositions such as maize starch, wheat starch, rice starch, potato starch, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carbomethylcellulose; and/or physiologically acceptable polymers such as) polyethylene glycol (PEG). Disintegrating agents may also be added, such as cross-linked agar, alginic acid or a salt thereof such as sodium alginate.
[0114] Tablets or capsules can be prepared by conventional means with pharmaceutically acceptable excipients such as binding agents (e.g, pregelatinized maize starch, hydroxypropyl methylcellulose, carboxymethyl cellulose, polyethylene glycol, sucrose, glucose, sorbitol, starch, gum, and tragacanth); fillers (e.g, lactose, microcrystalline cellulose, or calcium hydrogen phosphate); lubricants (e.g., calcium, aluminum, zinc, stearic acid, polyethylene glycol, sodium lauryl sulfate, starch, sodium benzoate, magnesium stearate, talc, or silica); disintegrants (e.g., starch, potato starch, sodium starch glycolate, sugars, cellulose derivatives, silica powders); or wetting agents (e.g., sodium lauryl sulphate). The tablets may be coated by methods well known in the art. A coating shell may be present, such as with membrane selected from, but not limited to, polylactide, polyglycolic acid, polyanhydride, other biodegradable polymers, hydroymethylacrylate-methyl methacrylate (HEMA-MMA), multilayered HEMA-MMA-MAA, polyethylene glycol/poly pentamethylcyclopentasiloxane/ polydimethylsiloxane (PEG/PD5/PDMS), siliceous encapsulates, cellulose acetate phthalate, calcium alginate, k-carrageenan-locust bean gum gel beads,, poly(lactide-co-glycolides), carrageenan, starch polyanhydrides, starch polymethacrylates, and enteric coating polymers.
[0115] In some embodiments, the at least one Bifidobacterium, the at least one propionate producing bacterium, and/or the prebiotic mixture are enterically coated, such as in order to remain viable during transit through the stomach, reduce contact with bile acids in the small intestine, or for release into the gut or a particular region of the gut, for example, the large intestine. The typical pH profile from the stomach to the colon is about 1-4 (stomach), 5.5-6 (duodenum), 7.3-8.0 (ileum), and 5.5-6.5 (colon). In some diseases, the pH profile may be modified. In some embodiments, the coating is degraded in specific pH environments in order to specify the site of release. In some embodiments, at least two coatings are used. In some embodiments, the outside coating and the inside coating are degraded at different pH levels.
[0116] In certain embodiments, the pharmaceutical compositions are formulated as liquid preparations. Liquid preparations for oral administration may take the form of solutions, syrups, suspensions, or a dry product for constitution with water or other suitable vehicle before use. Such liquid preparations may be prepared by conventional means with pharmaceutically acceptable agents such as suspending agents (e.g., sorbitol syrup, cellulose derivatives, or hydrogenated edible fats); emulsifying agents (e.g., lecithin or acacia); nonaqueous vehicles (e.g., almond oil, oily esters, ethyl alcohol, or fractionated vegetable oils); and preservatives (e.g., methyl or propyl-p-hydroxybenzoates or sorbic acid). The preparations may also contain buffer salts, flavoring, coloring, and sweetening agents as appropriate. Preparations for oral administration may be suitably formulated for slow release, controlled release, or sustained release of the bacteria described herein.
[0117] In some embodiments, the at least one Bifidobacterium, the at least one propionate producing bacterium, and prebiotic mixture may be formulated in a composition suitable for administration to pediatric subjects. As is well known in the art, children differ from adults in many aspects, including different rates of gastric emptying, pH, gastrointestinal permeability, etc. Moreover, pediatric formulation acceptability and preferences, such as route of administration and taste attributes, are critical for achieving acceptable pediatric compliance. Thus, in one embodiment, the composition suitable for administration to pediatric subjects may include easy-to-swallow or dissolvable dosage forms, or more palatable compositions, such as compositions with added flavors, sweeteners, taste blockers, or suitable to be mixed in a foodstuff, e.g., applesauce. In one embodiment, a composition suitable for administration to pediatric subjects may also be suitable for administration to adults.
[0118] In certain embodiments, the pharmaceutical composition that is suitable for administration to pediatric subjects may include a solution, syrup, suspension, elixir, powder for reconstitution as suspension or solution, dispersible/effervescent tablet, chewable tablet, gummy candy, lollipop, freezer pop, troche, chewing gum, oral thin strip, orally disintegrating tablet, sachet, soft gelatin capsule, sprinkle oral powder, or granules. In one embodiment, the composition is a gummy candy, which is made from a gelatin base, giving the candy elasticity, desired chewy consistency, and longer shelf-life. In some embodiments, the gummy candy may also comprise sweeteners or flavors.
[0119] In some embodiments, the pharmaceutical composition, e.g., composition suitable for administration to pediatric subjects, may include a flavor. As used herein, “flavor” is a substance (liquid or solid) that provides a distinct taste and aroma to the formulation. Flavors also help to improve the palatability of the formulation. Flavors include, but are not limited to, strawberry, vanilla, lemon, grape, bubble gum, cherry, and chocolate.
[0120] In particular embodiments, the at least one Bifidobacterium, the at least one propionate producing bacterium, and the prebiotic mixture may, together or separately, be orally administered, such as with an inert diluent or an assimilable edible carrier. In some aspects, the pharmaceutical composition may also be enclosed in a hard or soft-shell gelatin capsule, a hydroxypropylmethyl cellulose (HPMC) capsule, compressed into tablets, or incorporated directly into the subject's diet. For oral therapeutic administration, the probiotic strain and prebiotic mixture may, together or separately, be incorporated with excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like. In some aspects, it may be necessary to coat or co-administer the pharmaceutical composition with a material to prevent inactivation of the probiotic strain and/or the prebiotic mixture.
[0121] In some embodiments, the composition containing the at least one Bifidobacterium, the at least one propionate producing bacterium, and/or the prebiotic mixture may be a nutritional or a comestible product, e.g., a food product or nutritional composition. In some embodiments, the composition is a nutritional composition such as food product. In certain embodiments, the food product or nutritional composition is or includes milk, concentrated milk, fermented milk (yogurt, sour milk, frozen yogurt, lactic acid bacteria-fermented beverages), milk powder, ice cream, cream cheeses, dry cheeses, soybean milk, fermented soybean milk, vegetable-fruit juices, fruit juices, sports drinks, confectionery, candies, infant foods (such as infant cakes), nutritional food products, animal
feeds, or dietary supplements. In some embodiments, the nutritional composition or food product is a fermented food, such as a fermented dairy product. In particular embodiments, the fermented dairy product is yogurt. In certain embodiments, the fermented dairy product is cheese, milk, cream, ice cream, milk shake, or kefir. In some embodiments, the probiotic strain of the invention, e.g., B. longum subsp. infantis strain, is combined in a preparation containing other live bacterial cells intended to serve as probiotics. In some embodiments, the food product is a beverage. In one embodiment, the beverage is a fruit juice-based beverage or a beverage containing plant or herbal extracts. In certain embodiments, the food product or nutritional composition is a jelly or a pudding. Other food products suitable for administration of the probiotic strain and prebiotic mixtures provided herein are known, such as those described in U.S. Application Nos. 2015/0359894 and 2015/0238545. In yet another embodiment, the pharmaceutical composition of the invention is injected into, sprayed onto, or sprinkled onto a food product, such as bread, yogurt, or cheese.
[0122] In some embodiments, the composition, e.g, pharmaceutical composition, that includes the at least one Bifidobacterium, the at least one propionate producing bacterium, and/or the prebiotic mixture is formulated for intraintestinal administration, intrajejunal administration, intraduodenal administration, intraileal administration, gastric shunt administration, or intracolic administration, via nanoparticles, nanocapsules, microcapsules, or microtablets, which are enterically coated or uncoated. The compositions may also be formulated in rectal compositions such as suppositories or retention enemas, using, e.g., conventional suppository bases such as cocoa butter or other glycerides. The compositions may be suspensions, solutions, or emulsions in oily or aqueous vehicles, and may contain suspending, stabilizing and/or dispersing agents.
[0123] In some embodiments, disclosed herein are pharmaceutically acceptable compositions containing the at least one Bifidobacterium, the at least one propionate producing bacterium, and/or the prebiotic mixture in single dosage forms. Single dosage forms may be in a liquid or a solid form. Single dosage forms may be administered directly to a subject without modification or may be diluted or reconstituted prior to administration. In certain embodiments, a single dosage form may be administered in bolus form, e.g., single injection, single oral dose, including an oral dose that comprises multiple tablets, capsule, pills, etc. In alternate embodiments, a single dosage form may be administered over a period of time, e.g., by infusion.
[0124] Single dosage forms of the pharmaceutical composition containing the at least one Bifidobacterium, the at least one propionate producing bacterium, and/or the prebiotic mixture may be prepared by portioning the pharmaceutical composition into smaller aliquots, single dose containers, single dose liquid forms, or single dose solid forms, such as tablets, granulates, nanoparticles, nanocapsules, microcapsules, microtablets, pellets, or powders, which may be enterically coated or uncoated. A single dose in a solid form may be reconstituted by adding liquid, typically sterile water or saline solution, prior to administration to a subject.
[0125] In certain embodiments, the composition can be delivered in a controlled release or sustained release system. In one embodiment, a pump may be used to achieve controlled or sustained release. In another embodiment, polymeric materials can be used to achieve controlled or sustained release of the therapies of the present disclosure (see, e.g., U.S. Pat. No. 5,989,463). Examples of polymers used in sustained release formulations include, but are not limited to, poly((2 -hydroxy ethyl methacrylate), poly(methyl methacrylate), poly(acrylic acid), poly(ethylene-co-vinyl acetate), poly(methacrylic acid), polyglycolides (PLG), polyanhydrides, poly(N-vinyl pyrrolidone), poly(vinyl alcohol), poly(ethylene glycol), polylactides (PLA), poly(lactide-co-glycolides) (PLGA), and polyorthoesters. The polymer used in a sustained release formulation may be inert, free of leachable impurities, stable on storage, sterile, and biodegradable. In some embodiments, a controlled or sustained release system can be placed in proximity of the prophylactic or therapeutic target, thus requiring only a fraction of the systemic dose. Any suitable technique known to one of skill in the art may be used.
[0126] Dosage regimens of the at least one Bifidobacterium, the at least one propionate producing bacterium, and/or the prebiotic mixture may be adjusted to provide a therapeutic response, e.g., to improve or maintain propionate production. Dosing can depend on several factors, including severity and responsiveness of the disease, route of administration, time course of treatment (days to months to years), and time to amelioration of the disease. For example, a single bolus of one or both of the mixture and the probiotic strain may be administered at one time, several divided doses may be administered over a predetermined period of time, or the dose may be reduced or increased as indicated by the therapeutic situation. The specification for the dosage is dictated by the unique characteristics of the active compound and the particular therapeutic effect to be achieved. Dosage values may vary with the type and severity of the condition to be alleviated. For any particular
subject, specific dosage regimens may be adjusted over time according to the individual need and the professional judgment of the treating clinician. Toxicity and therapeutic efficacy of compounds provided herein can be determined by standard pharmaceutical procedures in cell culture or animal models. For example, LD50, ED50, EC50, and IC50 may be determined, and the dose ratio between toxic and therapeutic effects (LD50/ED50) may be calculated as the therapeutic index. Compositions that exhibit toxic side effects may be used, with careful modifications to minimize potential damage to reduce side effects. Dosing may be estimated initially from cell culture assays and animal models. The data obtained from in vitro and in vivo assays and animal studies can be used in formulating a range of dosage for use in humans.
[0127] In some embodiments, ingredients are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water-free concentrate in a hermetically sealed container such as an ampoule or sachet indicating the quantity of active agent.
[0128] The pharmaceutical compositions, e.g., containing the at least one Bifidobacterium, the at least one propionate producing bacterium, and/or the prebiotic mixture, may be packaged in a hermetically sealed container such as an ampoule or sachet indicating the quantity of the agent. In one embodiment, one or more of the pharmaceutical compositions is supplied as a dry sterilized lyophilized powder or water-free concentrate in a hermetically sealed container and can be reconstituted (e.g., with water or saline) to the appropriate concentration for administration to a subject. In an embodiment, one or more of the prophylactic or therapeutic agents or pharmaceutical compositions is supplied as a dry sterile lyophilized powder in a hermetically sealed container stored between 2° C. and 8° C. and administered within 1 hour, within 3 hours, within 5 hours, within 6 hours, within 12 hours, within 24 hours, within 48 hours, within 72 hours, or within one week after being reconstituted. Cryoprotectants can be included for a lyophilized dosage form, principally 0- 10% sucrose (optimally 0.5- 1.0%). Other suitable cryoprotectants include trehalose and lactose. Other suitable bulking agents include poly dextrose, dextrins (e.g., maltodextrin (e.g., a native maltodextrin or a resistant maltodextrin)), inulin, P-glucan, resistant starches (e.g., resistant maltodextrin), hydrocolloids (e.g., one or more of gum Arabic, pectin, guar gum, alginate, carrageenan, xanthan gum and cellulose gum), corn syrup solids and the like and polysorbate 80.-. Additional surfactants include but are not limited to polysorbate 20 and BRIJ surfactants. The pharmaceutical composition may be prepared as an injectable solution
and can further comprise an agent useful as an adjuvant, such as those used to increase absorption or dispersion, e.g., hyaluronidase.
[0129] In some embodiments, the pharmaceutical compositions, e.g., containing one or both of the probiotic strain and prebiotic mixtures are administered with food. In alternate embodiments, the pharmaceutical composition is administered before or after eating food. The pharmaceutical compositions may be administered in combination with one or more dietary modifications, e.g., low-protein diet and amino acid supplementation. The dosage of the pharmaceutical compositions and the frequency of administration may be selected based on the severity of the symptoms and the progression of the disorder. The appropriate therapeutically effective dose and/or frequency of administration can be selected by a treating clinician.
IV. DEFINITIONS
[0112] Unless defined otherwise, all terms of art, notations and other technical and scientific terms or terminology used herein are intended to have the same meaning as is commonly understood by one of ordinary skill in the art to which the claimed subject matter pertains. In some cases, terms with commonly understood meanings are defined herein for clarity and/or for ready reference, and the inclusion of such definitions herein should not necessarily be construed to represent a substantial difference over what is generally understood in the art.
[0113] As used herein, the singular forms "a," "an," and "the" include plural referents unless the context clearly dictates otherwise. For example, "a" or "an" means "at least one" or "one or more." It is understood that aspects and variations described herein include "consisting" and/or "consisting essentially of’ aspects and variations.
[0114] Throughout this disclosure, various aspects of the claimed subject matter are presented in a range format. It should be understood that the description in range format is merely for convenience and brevity and should not be construed as an inflexible limitation on the scope of the claimed subject matter. Accordingly, the description of a range should be considered to have specifically disclosed all the possible sub-ranges as well as individual numerical values within that range. For example, where a range of values is provided, it is understood that each intervening value, between the upper and lower limit of that range and any other stated or intervening value in that stated range is encompassed within the claimed
subject matter. The upper and lower limits of these smaller ranges may independently be included in the smaller ranges, and are also encompassed within the claimed subject matter, subject to any specifically excluded limit in the stated range. Where the stated range includes one or both of the limits, ranges excluding either or both of those included limits are also included in the described or claimed subject matter. This applies regardless of the breadth of the range.
[0115] Throughout this disclosure, ranges that are presented or expressed as “between” two endpoints, e.g., “between A and B” are understood to include the endpoints, e.g. “A” and “B”, unless otherwise indicated.
[0116] The term "about" as used herein refers to the usual error range for the respective value readily known to the skilled person in this technical field. Reference to "about" a value or parameter herein includes (and describes) embodiments that are directed to that value or parameter per se. For example, description referring to "about X" includes description of "X". In some embodiments, “about” a value means within a range of ±25%, ±10%, ±5%, ±1%, ±0.1%, or ±0.01% of the value.
[0117] As used herein the term "pharmaceutical composition" means, for example, a mixture or formulation containing a specified amount, e.g., a therapeutically effective amount, of an active ingredient such as a human milk fraction, in a pharmaceutically acceptable carrier to be administered to a mammal, e.g., a human.
[0118] As used herein the term "pharmaceutically acceptable" refers to those compounds, materials, compositions and/or dosage forms, which are, within the scope of sound medical judgment, suitable for contact with the tissues of mammals, especially humans, without excessive toxicity, irritation, allergic response, and other problem complications commensurate with a reasonable benefit/risk ratio. Such reasonable benefit/risk ratios may be determined by one of skill as a matter of routine.
[0119] By “human milk oligosaccharide(s)” (also referred to herein as “HMO(s)”) is meant a family of structurally diverse unconjugated glycans that are found in human breast milk. As used herein human milk oligosaccharides include oligosaccharides found in human milk that contain lactose at the reducing end and, typically, fucose, sialic acid or N- acetylglucosamine at the non-reducing end (Morrow et al., J. Nutri. 2005 135: 1304-1307). Unless otherwise indicated, human milk oligosaccharides also encompass 3'-sialyllactose (3'- SL) and 6'-sialyllactose (6'-SL) oligosaccharides that are found in human milk.
[0120] Unless otherwise noted, a number of human milk oligosaccharides, e.g., “at least 5 human milk oligosaccharides,” refers to the number of unique species of human milk oligosaccharides, e.g., human milk oligosaccharides having different chemical structures or formulas.
[0121] Glycans in milk are found as oligosaccharides or conjugated to milk proteins as glycoproteins, or lipid as glycolipids etc. HMO are free glycans that constitute the third most abundant component of human milk, after lactose and lipid (Morrow, 2005). The majority of HMO, however, are not metabolized by the infant and can be found in infant feces largely intact.
[0122] By “consisting essentially” of, as used herein refers to compositions containing particular recited components while excluding other major bioactive factors.
[0123] “Probiotic” as used herein, refers to any live, non-pathogenic microorganisms, e.g., bacteria, which can confer health benefits to a host organism, e.g., a mammal such as a human, that contains an appropriate amount of the microorganism. In some aspects, those of skill in the art may readily identify species, strains, and/or subtypes of non-pathogenic bacteria that are recognized as probiotic bacteria. Examples of probiotic bacteria may include, but are not limited to, Bifidobacteria, Escherichia coli, Lactobacillus, and Saccharomyces, e.g., Bifidobacterium bifidum, Enterococcus faecium, Escherichia coli strain Nissle, Lactobacillus acidophilus, Lactobacillus bulgaricus, Lactobacillus paracasei, Lactobacillus plantarum, and Saccharomyces boulardii (Dinleyici et al., 2014; U.S. Pat. Nos. 5,589,168; 6,203,797; 6,835,376). The probiotic may be a variant or a mutant strain of bacterium (Arthur et al., 2012; Cuevas-Ramos et al., 2010; Olier et al., 2012; Nougayrede et al., 2006).
[0124] “Bifidobacterium” or “Bifidobacteria” as used herein, refers to a genus of gram-positive, nonmotile, anaerobic bacteria. In some aspects, Bifidobacterium are ubiquitous inhabitants of the gastrointestinal tract, vagina, and mouth of mammals, including humans. In certain aspects, Bifidobacteria are one of the major genera of bacteria that make up the gastrointestinal tract microbiota in mammals. In certain aspects, some or all species, subspecies, or strains of Bifidobacterium are probiotics.
[0125] The term "dysbiosis" as used herein refers to a state of the microbiota of the gut or other body area in a subject, in which the normal diversity and/or function of the microbial populations is disrupted. This unhealthy state can be due to a decrease in diversity, the overgrowth of one or more pathogens or pathobionts, symbiotic organisms able to cause
disease only when certain genetic and/or environmental conditions are present in a subject, or the shift to an ecological microbial network that no longer provides an essential function to the host subject, and therefore no longer promotes health. According to non-limitative examples, essential functions may include enhancement of the gut mucosal barrier, direct or indirect reduction and elimination of invading pathogens, enhancement of the absorption of specific substances, and suppression of GI inflammation.
[0126] As used herein, the terms “gut microbiome” and “intestinal microbiome” are used interchangeably unless otherwise noted.
[0127] As used herein, “non-digestible” as used in the term “non-digestible carbohydrate” refers to the fact that the carbohydrate is not digested by the host or human subject.
[0128] The term “essentially” such as when used in the phrase “essentially all” of a given substance may be used to infer that the substance, e.g., oligosaccharides, includes unavoidable impurities, e.g., no more impurities than what might be unavoidable with standard techniques for manufacture, formulation, transporting, and storage. Likewise, when used in the phrase “essentially free” of a given substance (or “essentially no” or “essentially none of’ a given substance) may mean no more of the given substance than is unavoidable, e.g., as an impurity.
[0129] The term "internalization" such as in reference to an internalization of an oligosaccharide by a bacterial cell refers to the transfer of the oligosaccharide from the outside of the bacterial cell to the inside of the bacterial cell. Unless otherwise indicated, “internalization of an oligosaccharide” refers to the internalization of the intact oligosaccharide.
EXAMPLES
[0130] The following examples are included for illustrative purposes only and are not intended to limit the scope of the invention.
EXAMPLE 1: MANUFACTURE OF A CONCENTRATED HMO MIXTURE
[0131] Human milk from previously screened and approved donors was tested to verify donor identity and then mixed together to generate a pool of donor milk. In a clean room environment, the pool of donor milk was further tested, including for specific pathogens and bovine proteins. After testing, the pooled donor milk was filtered through a 200 pm filter, heated to a temperature of approximately 63 °C for 30 minutes, and then cooled to between 2°C and 8°C. The pooled human milk was then transferred to a centrifuge to separate the cream from the skim. The resulting skim milk was ultra-filtered with a 10 kDa membrane, and the material that passed through the filter was collected as the permeate fraction. The permeate was frozen and stored at approximately -20°C.
[0132] Multiple samples of the permeate fractions were thawed and pooled. The pH of the pooled permeate was adjusted to a target pH of 4.5 ± 0.2. The permeate was then heated to approximately 50°C. Lactase enzyme was added to the permeate at a 0.1% weight/weight (w/w) concentration and incubated at approximately 50°C for 60 minutes. The permeate and lactase enzyme mixture then was cooled to between 20°C and 30°C and clarified by depth filtration (Filtrox CHI 13P). The resulting depth filter filtrate was then ultra-filtered (Biomax- 1 OK membrane) to remove the lactase. The ultra-filtered permeate was then concentrated by nanofiltration using membranes with estimated 400 to 500 Dalton molecular weight cut-off (GE G-5 UF). The concentrated HMO composition was then pasteurized and clarified though 0.2 pm sterile filters. This final HMO composition was then filled into containers and stored at < -20°C. The final concentrations of HMO were quantified using high performance anion exchange chromatography with pulsed amperometry detection (HPAEC-PAD) with commercially available standards.
EXAMPLE 2: ADMINISTRATION OF B. LONGUM SUBSP. INFANTIS AND A CONCENTRATED HUMAN MILK OLIGOSACCHARIDE MIXTURE TO SUBJECTS TREATED WITH ANTIBIOTICS
[0133] A clinical study assessing the administration of B. longum subsp. infantis and human milk oligosaccharides in healthy adult subjects who were administered antibiotics was performed as summarized in FIG. 1 and Table El. A total of 56 subjects were enrolled in three different cohorts: those who received antibiotics alone for days 1-5 (Cohort 1); those who received antibiotics on days 1-5 and B. longum subsp. infantis on days 1-14 (Cohort 2); and those who received antibiotics on days 1-5, B. longum subsp. infantis on days 1-14, and a human milk oligosaccharide mixture (HMO mixture) generated as described in Example 1 on days 1-28 (Cohort 3). For antibiotic treatment, subjects were administered 250 mg vancomycin three times daily, which preferentially affects gram positive bacteria, and 500 mg metronidazole three times daily, which affects anaerobes including gram negative bacteria. Used together, these antibiotics broadly impact the gut microbial community. Subjects administered B. longum subsp. infantis received 8xl09 CFU per oral dose of a commercially available probiotic once a day from days 1-14. Subjects receiving the HMO mixture were administered HMO orally in liquid form twice a day from days 1-28 for a total of 18 g/day of total human milk oligosaccharides.
Table El. Study Design Summary
included in analyses.
[0134] All subjects were followed to day 35, which was one week after completion of HMO dosing in the corresponding cohort. Stool samples were collected during eligibility screening and on days 1, 3, 4, 5, 7, 9, 11, 14, 17, 21, 28, and 35, where day 1 was the first day of the protocol. Some subjects were asked to return to the study site and provide a stool sample on or after day 90. Blood samples were also drawn on days 1, 5, 14, 28 and 35, processed to serum, and frozen. On study days 1, 5, 9, 14, 28, and 35, stool was refrigerated at 4°C after production and frozen at -80°C within 24 hours. On days 3, 4, 7, 11, 17, and 21 of the study, a portion of each stool was preserved in ethanol immediately after production and frozen at -80°C within one week.
[0135] No serious adverse effects were observed for any of the subjects.
EXAMPLE 3: B. LONGUM SUBSP. INFANTIS DETECTION BY QPCR IN HEALTHY SUBJECTS AFTER VANCOMYCIN AND METRONIDAZOLE EXPOSURE
[0136] Genomic DNA was extracted from stool samples collected from subjects as described in Example 2. Extracted DNA was assessed for B. longum subsp. infantis by qPCR performed similar to as described in Lawley et al., PeerJ. 2017 May 25;5:e3375 with forward and reverse primers to the sialidase gene identical to SEQ ID NOS: 17 and 18 and a probe sequence identical to SEQ ID NO: 19.
[0137] Results are displayed in FIGS. 2A-2C. As shown in FIG. 2A, subjects from Cohort 1 (antibiotics alone) had B. longum subsp. infantis levels that remained at or below the limit of detection (27 copies/ng DNA) or barely detectable (< 2% of samples at or below 93 copies/ng DNA). During the antibiotic dosing period (Days 1-5), low levels of B. longum subsp. infantis were detected in the DNA from stool of subjects in both cohorts that received B. longum subsp. infantis. These levels were consistent with a “pass through” reflecting the daily consumption of the bacteria, as B. longum subsp. infantis is sensitive to both antibiotics used in this study and was unlikely to remain viable in the gut. With continued dosing beyond the antibiotic treatment, B. longum subsp. infantis levels rose in both A longum subsp. infantis-treated cohorts above the “pass-through” levels by day 11. On days 21 and 28, a significant difference was observed between cohorts. Subjects from Cohort 2 (B. longum subsp. infantis alone) exhibited a steady decline in B. longum subsp. infantis levels after the end of dosing and with presumed recovery of the microbiome, while most Cohort 3 subjects (B. longum subsp. infantis and HMO) displayed stable, high levels of B. longum subsp.
infantis through day 28. After HMO dosing ceased, levels of B. longum subsp. infantis declined rapidly. Together, these data demonstrate that while antibiotic treatment may temporarily reduce colonization resistance to enable transient expansion of B. longum subsp. infantis, co-dosing with HMO is essential for durable, high-level engraftment.
[0138] In both cohorts dosed with B. longum subsp. infantis (Cohorts 2 and 3), there was evidence of two subpopulations: an “engrafted” population for which B. longum subsp. infantis was consistently maintained during the HMO dosing period, defined as exhibiting B. longum subsp. infantis signal more than two standard deviations above the “pass through” level during antibiotic treatment for at least two consecutive time points, and a “not engrafted” population, where B. longum subsp. infantis was not consistently maintained (FIGS 2B and 2C). Of the Cohort 3 subjects (B. longum subsp. infantis and HMO), 76% (13/17) were deemed engrafted (FIG. 2B). Among the four subjects defined as not engrafted, each produced at least one stool sample without detectable levels of B. longum subsp. infantis during the 14-day dosing period, which may suggest issues with compliance that could have influenced engraftment success. The mean level of B. longum subsp. infantis at day 14 in engrafted subjects was approximately 5 -fold higher than the overall mean when all subjects were included (1.1 x 105 vs 2.4 x 104 copies/ng DNA). Moreover, when only engrafted subjects within the Cohort 3 were compared to Cohort 2, B. longum subsp. infantis levels were significantly higher as early as day 11 (p = 0.0004; Sidak’s post-hoc test; Figure 7A).
[0139] Engraftment was also observed in 4 of the Cohort 2 subjects (B. longum subsp. infantis alone), representing -24% (4/17) of the cohort (FIG. 2C). However, B. longum subsp. infantis levels in these subjects were maintained at -103 copies/ng DNA through day 35, which is -100-fold lower than for engrafted subjects who also received HMO. The rate of engraftment was also significantly different between the two cohorts (13/17 engrafted vs 4/17 engrafted; p = 0.0053 by Fisher’s exact test). Here, engraftment in the absence of HMO treatment may be due to loss of colonization resistance to B. longum subsp. infantis following antibiotic exposure, although this was observed in only a subset of subjects.
[0140] Some engrafted subjects from both cohorts retained B. longum subsp. infantis signal at day 35. Additional stool samples were from a subset of engrafted subjects on or after day 90 (Table E2). B. longum subsp. infantis was below the limit of detection for the nine subjects analyzed from Cohort 3 (B. longum subsp. infantis and HMO). Of three engrafted subjects evaluated from Cohort 2 (B. longum subsp. infantis only), one displayed levels of B. longum subsp. infantis below the limit of detection and the other two had detectable but low
levels (7.7 x 101 and 5.79 x 102 copies/ng DNA; Table E2). These data suggest that for the majority of subjects, administration of B. longum subsp. infantis did not lead to durable engraftment after antibiotic treatment in the absence of treatment with HMO.
EXAMPLE 4: B. LONGUM SUBSP. INFANTIS DETECTION BY WHOLE META GENOMIC SEQUENCING IN HEALTHY SUBJECTS AFTER VANCOMYCIN AND METRONIDAZOLE EXPOSURE
[0141] Whole metagenomic sequencing (WMS) on stool samples was used with a strain-specific tracking algorithm to quantify the relative abundance of B. longum subsp. infantis in the microbiome for each cohort.
[0142] DNA extracted from human stool samples collected at days 1, 5, 9, 14, 28, and 35 as described in Example 2 was used to prepare libraries for shotgun metagenomic sequencing. Paired-end sequencing (2 x 150 bp) was performed on an Illumina NovaSeq instrument to generate a target of ~2 million reads per sample (BoosterShot, Diversigen Inc.).
[0143] Sequence analyses followed an established pipeline (Diversigen Inc.). Briefly, sequences were aligned to a curated database containing all representative genomes in RefSeq for bacteria with additional manually curated strains. Alignments were made at 97% identity against all reference genomes. Every input sequence was compared to every reference sequence in the Diversigen Venti database using fully gapped alignment with BURST 123. Ties were broken by minimizing the overall number of unique Operational Taxonomic Units (OTUs). For taxonomy assignment, each input sequence was assigned the lowest common ancestor that was consistent across at least 80% of all reference sequences tied for best hit. Samples with fewer than 10,000 sequences were discarded. OTUs accounting for less than one millionth of all strain-level markers and those with less than 0.01% of their unique genome regions covered (and < 0.1% of the whole genome) at the species level were discarded. The counts for each OTU in this filtered table were normalized to the OTU's genome length, and filtered tables for the normalized counts and relative abundance of each OTU were generated. Strain-level tracking, including for B. longum subsp. infantis, used the “capitalist” algorithm in BURST to assign reads to genomes in the Diversigen Venti database. Rather than applying a lowest common ancestor approach, the “capitalist” algorithm returns a minimal set of genomes that can explain all the reads in a sample. From that output, an OTU table was calculated with the read counts per genome per sample. Alpha diversity metrics (observed reads and Shannon entropy) were calculated using the filtered OTU table rarefied to a read depth of 76,000 using the R package phyloseq (v.1.41.0). Bray-Curtis dissimilarity was calculated on the same filtered data, aggregated at the genus taxonomic level and rarefied to 76,000 reads using the R package phyloseq (vl.41.0124). The filtered OTU table aggregated at the Family taxonomic level was used to calculate the top Families among cohorts. All taxa >1% abundance in any cohort were kept and all taxa <1% among taxa were grouped into an “Other” category. The count table of aggregated genera was then center log-ratio (CLR) transformed by taking the median CLR value for each OTU sampled 150 times from a Dirichlet model using the R package ALDEx2 (v.1.29.1125).
[0144] As shown in FIG. 2D, engrafted Cohort 3 subjects (B. longum subsp. infantis and HMO) displayed a median B. longum subsp. infantis relative abundance of 45.9%, with a maximum of 81% observed in one subject at day 9. Abundance was much lower in engrafted Cohort 2 subjects (B. longum subsp. infantis alone), ranging from 0.7-7.8% with a median of 3.4%. Use of an orthogonal method with different sequence biases determined B. longum subsp. infantis relative abundance by calculating the ratio of B. longum subsp. infantis to total bacterial (16S) qPCR signals (FIG. 7B). This method similarly confirmed the high abundances observed by WMS with median relative abundance of engrafted subjects in the B. longum subsp. infantis and HMO cohort at 8.6% with a range of 0.05-48.4% during days 14- 28.
[0145] These data are consistent with promotion of B. longum subsp. infantis engraftment by human milk oligosaccharides in a higher frequency of subjects and at high abundance in subjects where dysbiosis has been induced by antibiotics. This durable, high- level engraftment is consistent with a high likelihood of positively impacting the gut microbiome.
EXAMPLE 5: HUMAN MILK OLIGOSACCHARIDE-MEDIATED ENGRAFTMENT WITH B. LONGUM SUBSP. INFANTIS ALTERS MlCROBIOME COMMUNITIES AFTER TRANSIENT, ANTIBIOTIC-INDUCED DYSBIOSIS
[0146] WMS data generated as described in Example 4 from genomic DNA collected from stool samples from Cohort 1 (antibiotics only control) and Cohort 3 (HMO and B. longum subsp. infantis) subjects described in Example 2 was assessed. Antibiotic treatment induced a transient, acute dysbiosis in all subjects of both Cohorts. Comparison of samples from days 1 (baseline) to samples from day 5 (end of antibiotic dosing) displayed robust decreases in microbial diversity and a consistent shift in the microbial community composition (FIGS. 3 and 8A). Shannon Entropy, which quantifies diversity and evenness in community structure, was significantly decreased in both cohorts at day 5 relative to day 1 (FIG. 3 A); similar results were observed when alpha diversity was quantified using observed reads (FIG.8A). As expected, there was also a significant decrease in total bacterial abundance, quantified using qPCR of the 16S rRNA gene (FIG. 8B). Overall microbiome community structure, as measured by Bray-Curtis dissimilarity, was also significantly different on day 5 relative to day 1 (FIG. 3B and Table E3). Levels of Lactobacillaceae and
Enterob acteriaceae increased from <0.2% of the population to >76% and >16% respectively (FIG. 3C).
Bray-Curtis dissimilarity was calculated for each sample with rarefied sequences aggregated to the genus taxonomic level, and PERMANOVA analysis was run with the pairwise Adonis package (v0.4).
[0147] From day 5 to 35, all measures of microbiome diversity rebounded in both cohorts. Shannon Entropy partially recovered, as values at day 35 increased from day 5, but also remained significantly different from day 1 baseline levels, suggesting recovery was incomplete at day 35 (FIG. 3 A). Incomplete recovery was also observed for measures of bacterial abundance (FIGS. 8A and 8B). Similarly, microbial community composition became progressively more similar to pre-treatment samples, although it remained distinct from baseline in all cohorts at all post-antibiotic timepoints by PERMANOVA analysis, consistent with incomplete recovery or recovery to an alternate stable state (FIG. 3B, Table E3). Levels of Lactobacillaceae and Enterobacteriaceae, which were at high abundance at the end of antibiotic treatment, decreased during the post-antibiotic period, and by day 28, bacterial families characteristic of a healthy human microbiome such as Lachnospiraceae and Bacteroidaceae were observed in both cohorts (FIG. 3C), suggesting that a high abundance of B. longum subsp. infantis does not preclude recovery of these taxa.
[0148] Cohort 3 (B. longum subsp. infantis and HMO) was compared to Cohort 1 (antibiotics only control). Both Shannon Entropy and observed reads suggested a slower recovery toward baseline levels for Cohort 3 subjects (FIGS. 3A and 8A), although total bacterial abundance did not differ between cohorts (FIG. 8B). There were also significant differences in microbiome composition between the cohorts when days 14 and 28 were compared (p < 0.001, Table E4).
Bray-Curtis dissimilarity was calculated for each sample with rarefied sequences aggregated to the genus taxonomic level, and PERMANOVA analysis was run with pairwise Adonis package (v0.4). “With Bifidobacterium" comparisons included all reads, while comparisons “Without Bifidobacterium" were run on samples with reads assigned to the genus Bifidobacterium removed prior to analysis.
[0149] Possible subtle changes in the underlying community structure of engrafted subjects were assessed by subtracting reads corresponding to the genus Bifidobacterium using an approach similar to the American Gut Project to exclude data resulting from microbial blooms during sample storage at room temperature (Amir et al., (2017). mSystems 2, e00199- 16). Even with Bifidobacterium excluded, there were statistically significant differences in microbial community structure between engrafted subjects in Cohort 3 compared to Cohort 1 at day 14 and day 28 (FIG. 3D, Table E4). Further, measuring the distance between each point and the centroid for its respective cohort indicated that there is significantly less Bray- Curtis dissimilarity among individual subjects within the B. longum subsp. infantis + HMO cohort compared to the antibiotic cohort at day 14, regardless of the inclusion of Bifidobacterium reads (FIG. 3D, p<0.05 by ANOVA). This result is consistent with the engraftment of B. longum subsp. infantis after antibiotic treatment may steer microbiome recovery toward a more consistent community structure.
[0150] The abundance of specific taxa in engrafted Cohort 3 (B. longum subsp. infantis and HMO) subjects was compared to Cohort 1 subjects (antibiotics only control). Besides the expected increase in Bifidobacterium signal at various timepoints, the genera Lactobacillus and Veillonella were present at significantly higher levels in Cohort 3 at days 14 and 28, as was Pediococcus at day 14 (FIGS. 3E and 3F and 8E and 8F). Taxa that differed when Bifidobacterium reads were subtracted from the dataset displayed similarly significant increases of the same three genera. Lactobacillus became dominant in the microbiome of nearly all subjects during the antibiotic treatment period, and limited consumption of HMO has been reported for some strains, so it is possible that persistence of this genus was associated with either B. longum subsp. infantis abundance or the presence of HMO. Median Veillonella abundance increased from <0.05% at day 1 in both cohorts to a maximum of 9.8% and average of ~2% after antibiotic treatment at day 9 and appeared to be maintained at higher levels (average of ~2%) in the presence of B. longum subsp. infantis and HMO by day 14, whereas in the antibiotics only cohort had dropped below 1% by this point (FIG. 3F). Veillonella is known to consume lactate, one of the organic acids produced by B. longum subsp. infantis during fermentation of human milk oligosaccharides, so it may be enriched due to increased availability of lactate in the gastrointestinal environment when B. longum subsp. infantis is present. Enrichment of Lactobacillus could also lead to increased lactate availability, and this occurring during antibiotic treatment may explain early (day 9) increases in Veillonella in both cohorts, even though by day 14 it was significantly more abundant in subjects engrafted with B. longum subsp. infantis. Additionally, there was significantly less Clostridiales at the end of the antibiotic treatment period (day 5) in the B. longum subsp. infantis + HMO cohort (FIGS 3E and 8G), and Ruminococcus was also significantly lower but only at day 14 (FIGS. 3E and 8H)
EXAMPLE 6: QUANTIFICATION OF SHORT-CHAIN FATTY ACIDS AND LACTATE IN HUMAN STOOL
[0151] Stool samples collected as described in Example 2 were sent to Precion, Ltd. (Morrisville, NC) for extraction and analysis. A portion of a neat sample was transferred into a 2 mL cryotube containing three stainless steel 1/8” cone balls, and the exact weight of the sample was recorded. A solution of deuterium-labelled internal standards in water (50 pL) and 1.5 mL of methanol was added to the cryotube and the sample was homogenized by vortex-mixing for 2 minutes. Alternatively, ethanol-preserved stool samples were centrifuged
and 50 pL of the supernatant was transferred to a 2 mL cryotube together with 50 pL standards and 1.0 mL methanol, and vortex-mixed for 2 minutes. The resulting suspensions were centrifuged at 2,000 x g at 20°C for 10 minutes, and 50 pL of the supernatant was transferred to a 96-well plate and derivatized using a modified version of the published protocol. An aliquot of the reaction mixture was analyzed by liquid-chromatography mass spectrometry (LC-MS/MS) using an Exion UHPLC (Sciex) coupled to a 5500+ Triple Quadrupole Mass Spectrometer (Sciex) in ESI negative mode using a C18 column (Zorbax Eclipse plus C18 1.8 micron, 2.1x50 mm, Agilent). The peak areas of the respective parent to product ion transitions were measured against the peak area of the parent to product ion transitions of the corresponding labelled internal standards for the quantitated metabolites. Quantitation was performed with Sciex OS-MQ software (Sciex) based on fortified calibration standards prepared immediately prior to each run.
[0152] For neat stool samples, raw data were weight corrected to account for the individual wet weight of each sample providing the analyte content in pg/g wet weight. Additionally, using a separate sample aliquot, the dry weight/wet weight ratio was determined for each sample and each dry weight/wet weight ratio was used to calculate the analyte content of each sample in pg/g dry weight. For ethanol-preserved samples, raw data were weight corrected to account for the individual dry weight of each sample. Using a separate sample aliquot (750 pL sample suspension), the dry weight/volume ratio was determined for each sample. Each dry weight/volume ratio was used to calculate the analyte content of each sample in pg/g dry weight. Data were then transformed to mmol/kg by dividing by the molecular weight of the analyte.
EXAMPLE 7: QUANTIFICATION OF METABOLITES IN HUMAN SERUM
[0153] Serum samples collected as described in Example 2 were quantitatively analyzed by LC-MS/MS for 70 analytes at Precion Ltd., Morrisville, NC, USA. This analysis consisted of two extractions of the serum samples. To the first extraction (100 pL), a solution of stable labelled internal standards was added followed by protein precipitation. After centrifugation, one portion of the supernatant was removed, evaporated to dryness, reconstituted and an aliquot analyzed on a Sciex Exion LC/Sciex 5500+ Triple Quadrupole Mass Spectrometer LC-MS/MS system in ESI negative mode using Cl 8 reversed phase chromatography. This assay covered organic acids, phenolic compounds, sulfates, and other analytes that are negatively charged under negative ESI mass spectrometer conditions. A
second portion of the supernatant was removed, evaporated to dryness, reconstituted, and an aliquot analyzed on a Sciex Exion LC/Sciex 5500+ Triple Quadrupole Mass Spectrometer LC-MS/MS system in ESI positive mode using Cl 8 reversed phase chromatography. This analysis covered amino acids, amines, and other analytes that were positively charged under positive ESI mass spectrometer conditions.
[0154] A second extraction was performed from 50.0 pL of the serum sample. A solution of stable labelled internal standards was added to serum samples followed by protein precipitation. After centrifugation, a portion of the supernatant was removed and derivatized with a substituted hydrazine to form the corresponding acid hydrazides of short chain fatty acids. An aliquot of the resulting mixture was analyzed on a Sciex Exion LC/Sciex 5500+ Triple Quadrupole Mass Spectrometer LC-MS/MS system in ESI positive mode using Cl 8 reversed phase chromatography.
[0155] For all methods, the peak areas of the respective parent to product ion transitions were measured against the peak areas of the parent to product ion transitions of the corresponding labelled internal standards. Stable labelled versions of each of the 70 analytes were used as internal standards. Quantitation was performed using a weighted linear least squares regression analysis generated from fortified calibration standards (6 to 10 concentration levels, depending on analyte) prepared concurrently with study samples and quality control samples in each analytical run.
EXAMPLE 8: GLOBAL METABOLOMICS ANALYSIS OF HUMAN STOOL
[0156] Untargeted metabolomic profiling was performed on stool samples collected as described in Example 2 from days 1, 5, and 14 at Metabolon, Inc (Morrisville, NC, USA) using a combination of LC-MS methods (Evans et al., (2014). Metabolomics 4, 1000132). All methods utilized a Waters ACQUITY UPLC and a Thermo Scientific Q-Exactive high resolution/accurate mass spectrometer interfaced with a heated electrospray ionization (HESI-II) source and Orbitrap mass analyzer operated at 35,000 mass resolution. Briefly, the sample extract was dried then reconstituted in solvents compatible to each of the four methods. Each reconstitution solvent contained a series of standards at fixed concentrations to ensure injection and chromatographic consistency. Based on Metabolon, Inc protocols and previously published methods, the first aliquot was analyzed using acidic positive ion conditions, chromatographically optimized for more hydrophilic compounds. The second aliquot was also analyzed using acidic positive ion conditions; however, it was
chromatographically optimized for more hydrophobic compounds. The third aliquot was analyzed using basic negative ion optimized conditions and a separate dedicated Cl 8 column. The fourth aliquot was analyzed via negative ionization following elution from a HILIC column (Waters UPLC BEH Amide 2.1 x 150 mm, 1.7 pm) using a gradient consisting of water and acetonitrile with 10 mM Ammonium Formate, pH 10.8. The MS analysis alternated between MS and data-dependent MSn scans using dynamic exclusion. The scan range varied slighted between methods but covered 70-1000 m/z.
[0157] Raw data were extracted, peak-identified, and QC processed using Metabolon’s hardware and software. These systems are built on a web-service platform utilizing Microsoft’s .NET technologies, which run on high-performance application servers and fiber-channel storage arrays in clusters to provide active failover and load-balancing. Compounds are identified by comparison to library entries of purified standards or recurrent unknown entities. Metabolon maintains a library based on authenticated standards that contains the retention time/index (RI), mass to charge ratio (m/z), and chromatographic data (including MS/MS spectral data) on all molecules present in the library. Furthermore, biochemical identifications are based on three criteria: retention index within a narrow RI window of the proposed identification, accurate mass match to the library +/- 10 ppm, and the MS/MS forward and reverse scores. MS/MS scores are based on a comparison of the ions present in the experimental spectrum to ions present in the library entry spectrum. While there may be similarities between these molecules based on one of these factors, the use of all three data points can be utilized to distinguish and differentiate biochemicals. More than 4500 commercially available purified standard compounds have been acquired and registered into a database for analysis on all platforms for determination of their analytical characteristics. Additional mass spectral entries have been created for structurally unnamed biochemicals, which have been identified by virtue of their recurrent nature (both chromatographic and mass spectral). These compounds have the potential to be identified by future acquisition of a matching purified standard or by classical structural analysis.
[0158] Metabolites were identified by comparison to a referenced library of chemical standards, and area-under-the-curve analysis was performed for peak quantification and normalized to day median value. To ensure high quality of the dataset, control and curation processes were subsequently used to ensure true chemical assignment and remove artifacts and background noise. Metabolites were scaled by run-day medians and log-transformed before statistical analysis. Two-way repeated measures ANOVA contrasts used to analyze the
data. For all analyses, missing values, if any, were imputed with the observed minimum for that compound. The statistical analyses were performed on natural log-transformed data using Array Studio.
EXAMPLE 9: ADMINISTRATION OF B. LONGUM SUBSP. INFANTIS AND HMO SIGNIFICANTLY ALTERS GUT METABOLITES IN SUBJECTS TREATED WITH ANTIBIOTICS
[0159] The impacts of B. longum subsp. infantis and HMO treatment on fecal and serum metabolite profiles were evaluated using a quantitative fecal assay for short-chain fatty acids and lactate as described in Examples 6, an untargeted global fecal metabolomics method as described in Example 7, and a targeted serum assay for microbiome-associated metabolites as described in Example 8. Only data from the engrafted subjects within Cohort 3 were included in these analyses.
[0160] Antibiotic treatment radically impacted metabolites detectable in both fecal and serum samples, with decreases between day 1 and day 5 in microbially-produced metabolites such as short-chain fatty acids, secondary bile acids, and certain tryptophan and tyrosine metabolites (FIGS. 4 and 9). A significant increase in lactate was observed in all subjects during this time frame as well (FIG. 4C), likely attributable to the blooms of Lactobacillaceae in all subjects (FIG. 3).
[0161] The high abundance of B. longum subsp. infantis in engrafted subjects, coupled with the availability of human milk oligosaccharides, suggested that the organic acids produced during the fermentation of human milk oligosaccharides should be detectable. Consistent with recovery of the microbiome after antibiotic treatment, levels of fecal acetate began to increase in all subjects in both cohorts after day 5 (FIG. 4 A). Although there were no significant differences in acetate at individual timepoints between cohorts, levels of acetate significantly differed between the cohorts when the data were analyzed over time (FIG. 4A). Recovery of fecal acetate levels was assessed across subjects in both cohorts using a Kaplan-Meier curve. All subjects in Cohort 3 (B. longum subsp. infantis + HMO) recovered back to the day 1 average and recovered significantly faster than those in Cohort 1 (antibiotics only control); moreover, 2 subjects in Cohort 1 had not returned to this baseline level by day 35 (FIG. 4B). This observation is consistent with promotion of acetate recovery by treatment with B. longum subsp. infantis and HMO. A correlation between acetate levels and B. longum subsp. infantis abundance was observed across days 5-28 in Cohort 3 (Pearson r=0.73; r2 0.53). Cohort 3 stool lactate levels were increased on days 11, 14, 17, and 28 (FIG
4C). Consistent with an increase in organic acids, stool from engrafted subjects within the B. longum subsp. infantis + HMO cohort had a significantly lower pH (5.8) on day 9 than stool from the control cohort (6.6) (FIG. 9C).
[0162] No difference in recovery of butyrate or propionate levels was observed between the two cohorts (FIGS. 9A and 9B). However, cross-sample analysis of SCFAs in stool samples may be confounded by differences in absorption through the distal gastrointestinal tract.
[0163] To look more broadly at the impact of B. longum subsp. infantis engraftment on the metabolome and the host, two complementary approaches were employed: global metabolomics analysis at days 1, 5, and 14 to identify differences in diverse gut metabolites, and a quantitative targeted panel for microbial metabolites in serum over time to highlight changes that translate to a systemic impact. Little difference was observed in fecal global metabolite profiles between cohorts on day 1 prior to antibiotic treatment (FIG. 9D). Within each cohort, significant changes were observed from days 1 to 5 and again from days 5 to 14, reflective of the changes in bacterial diversity caused by antibiotic treatment (FIGS. 9E-9I). However, on day 14, when B. longum subsp. infantis was at high levels in engrafted subjects, 231 metabolites were significantly different between the cohorts; 77 metabolites were decreased and 154 were increased (FIG. 4D). A subset of these (56/231) are unidentified compounds, and others could be linked directly to treatment with B. longum subsp. infantis and HMO (lactate, 3-fucosyllactose). Increased levels of indolelactate, a potentially beneficial immunomodulatory tryptophan metabolite known to be produced by B. longum subsp. infantis and other gut microbes, were observed in both serum and stool samples (FIG. 4E).
[0164] The uremic toxin p-cresol sulfate (pCS) was decreased in both fecal and serum samples of Cohort 3 subjects (FIG. 4F). pCS is a host-generated conjugate of p-cresol known to contribute to inflammatory conditions and is produced from tyrosine or phenylalanine by intestinal bacteria. Serum levels of other tyrosine and phenylalanine metabolites were also shifted, including decreases in the uremic toxins phenol sulfate (FIGS. 9M and 9N) and p- cresol-glucuronide (pCG), and in other pathway intermediates 4-hydroxyphenylpropionate, 3- hydroxyhippurate, and 3 -hydroxybenzoate. Conversely, there were increases in 4- hydroxyphenylacetate, the precursor of p-cresol, and phenyllactate, an alternative microbial metabolic pathway for tyrosine and phenylalanine. Together, these changes suggest /?.
longum subsp. infantis + HMO treatment may redirect tyrosine and phenylalanine flux away from production of inflammatory toxins.
[0165] Several bile acids were differentially detected in fecal samples. Two hostgenerated glucuronide conjugates of the primary bile acid cholate and its associated secondary bile acid deoxycholate were increased in the B. longum subsp. infantis + HMO cohort (Figure 4D). Additionally, there were significant decreases in the secondary bile acids ursocholate, ursodeoxycholate, and isoursodeoxycholate, which are all 7-P-hydroxy epimers and associated derivatives of the primary bile acids cholate and chenodeoxycholate (FIG. 4D). Ursodeoxycholate was also included in the serum analysis, but no changes were observed between cohorts for that or two other bile acids: chenodeoxycholate and cholate. Interestingly, serum levels of the secondary bile acid deoxycholate were similar between the cohorts at day 14 but began to decrease in the B. longum subsp. infantis + HMO cohort at day 28.
[0166] Engraftment of B. longum subsp. infantis influenced the levels of a number of other metabolites indicative of changes in microbial metabolism (FIG. 4D). Over a quarter of the identified metabolites (47) are considered xenobiotics, or chemical compounds not naturally produced in the gut. Other identified metabolites are associated with carbohydrate metabolism (12), amino acids and derivatives (45), and lipid synthesis and metabolism (39), suggesting general shifts in microbial metabolism, gut mucosal properties, and/or enterocyte energetics. For example, increases in carbon sources, such as sugar alcohols and plant-based molecules like feruloylquinates, suggest either decreased degradation of these molecules or increased absorption, and increases in eight gamma-glutamyl amino acids suggest altered protein degradation, reflecting a change seen in infants that consume HMO that is linked to lower rates of respiratory illness.
EXAMPLE 10: ISOLATION OF VEILLONELLA SP. FROM STOOL SAMPLES
[0167] Bacterial strains of Veillonella sp. were isolated from fecal samples of Cohort 3 subjects (B. longum subsp. infantis and HMO) described in Example 2. The fecal samples had been previously preserved at -80°C, and an aliquot was thawed and serially diluted into anoxic filter-sterilized PBS (pH 7.0). Aliquots of serial dilutions were plated onto prereduced Brucella Blood Agar (Anaerobe Systems). Plates were incubated anaerobically at 37°C for 2-5 days. Individual colonies were picked, arrayed onto a fresh plate of the same agar, and regrown for 2-5 days. DNA was extracted from the arrayed isolates using an
alkaline lysis buffer technique (Vyhlidalova et al. (2020). IJMS 21, 2614) and the bacterial 16S rRNA gene was amplified by PCR using the established universal 16S primers 27F and 1492R (Weisburg et al. (1991) J Bacteriol 173, 697-703). The PCR products were cleaned using Exo-SapIT (Applied Biosystems) and two-directional Sanger sequencing was performed using the same primers to assign taxonomic identity to each strain (Azenta Life Sciences).
[0168] Under anaerobic conditions, pure cultures of arrayed isolates identified as Veillonella were prepared by picking and streaking onto fresh media three times. A single colony was inoculated into 30 mL of liquid Reinforced Clostridial Media (RCM) with 60% sodium lactate, and the culture was grown anaerobically at 37°C for 1-3 days. The cell culture was mixed with pre-reduced glycerol and PBS to a final concentration of 15% glycerol, aliquots were distributed to cryovials, and the cryovials were frozen and stored at - 80°C. The same culturing conditions were used to propagate and bank V. parvula ATCC 10790, after initial revival from a lyophilized stock by streaking onto BRU.
[0169] Pure banks of isolates were subjected to PCR and Sanger sequencing using the 16S rRNA primers described above and primers specific to the Veillonella rpoB gene, with resulting sequences aligned to known Veillonella sequences to assign specific species identity. One strain was identified as highly similar to V. infantium and a second strain was identified as V. rogosae.
EXAMPLE 11: ENHANCED IN VITRO GROWTH OF AND PRODUCTION OF PROPIONATE BY VEILLONELLA SP. CO-CULTURED WITH B. LONGUM SUBSP. INFANTIS AND HMO
[0170] The two Veillonella strains isolated as described in Example 11 were evaluated for in vitro growth by optical density and species-specific qPCR. A commercially available strain of V. parvula sourced from the human gut was used for comparison. Levels of propionate, lactate, and acetate were measured during growth and, while kinetics varied between the strains, all Veillonella sp. grew and produced propionate and acetate in a lactatedependent manner (FIGS. 5A and 10A-10E). Veillonella sp. were cultured alone, with human milk oligosaccharides prepared from the mixture described in Example 1 (HMO), cocultured with B. longum subsp. infantis, or cocultured with B. longum subsp. infantis in the presence of human milk oligosaccharides. Neither human milk oligosaccharides alone nor B. longum subsp. infantis alone supported Veillonella growth or propionate production, however, when both B. longum subsp. infantis and HMO were included, all Veillonella sp. grew and
produced more propionate than with an equivalent level of lactate alone (FIGS. 5B-5D). These data are consistent with B. longum subsp. infantis production of lactate during growth on HMO cross-feeding with Veillonella. resulting in promotion of Veillonella growth and propionate production.
EXAMPLE 12: ENHANCED IN VIVO GROWTH OF AND PRODUCTION OF PROPIONATE BY VEILLONELLA SP. CO-CULTURED WITH B. LONGUM SUBSP. INFANTIS AND HMO
[0171] Germ-free mice were inoculated with V. parvula or B. longum subsp. infantis as a control and, after a one-week stabilization, mice were gavaged with either PBS or B. longum subsp. infantis with human milk oligosaccharides prepared as described in Example 1 (HMO; FIG. 6A). Over the following three days, mice were gavaged once per day with either PBS or HMO. V. parvula levels increased over time only in mice treated with B. longum subsp. infantis + HMO but not in the control mice (FIGS. 6B and 11 A). B. longum subsp. infantis levels were predictably only detected after mice were dosed with B. longum subsp. infantis (FIG. 1 IB). Three hours after the final gavage, all animals were euthanized and levels of propionate, lactate, and acetate in different intestinal segments were quantified (FIGS. 6C-6D, 11C). Mice colonized with V. parvula and subsequently treated with B. longum subsp. infantis and HMO had an increase in Veillonella abundance and an increase in cecal propionate relative to PBS-treated mice (FIG. 6C), indicating that cross-feeding occurred. The differences in propionate levels were not significant in freshly collected rectal samples or cage-bottom fecal samples, suggesting absorption or other factors may introduce variability in samples from these compartments as has been previously observed in both rodents and humans. No propionate was detected in mice monocolonized with B. longum subsp. infantis (FIG. 6C). Mice monocolonized with A longum subsp. infantis and treated with HMO had high levels of lactate in cecal and rectal samples. In contrast, levels were lower in V. parvula-associated mice treated with B. longum subsp. infantis + HMO (FIG. 6D) even though similar levels of B. longum subsp. infantis were measured in both groups (FIG. 1 IB). Lower lactate levels are consistent with lactate consumption by Veillonella.
EXAMPLE 13: EVALUATING ADMINISTRATION OF B. LONGUM SUBSP. INFANTIS AND HMO IN A MOUSE MODEL OF ACUTE RADIATION SYNDROME (ARS)
[0172] Mice are exposed to a total body dose of ionizing radiation that is sufficient to induce gastrointestinal damage. The irradiated mice are orally administered human milk
oligosaccharides and B. longum subsp. infantis or a control. Controls may include vehicle (e.g., saline or PBS) and/or B. longum subsp. infantis alone or human milk oligosaccharides alone. The mice are assessed for survival, clinical scores, and/or incidence of diarrhea.
[0173] Irradiated mice administered human milk oligosaccharides and B. longum subsp. infantis may have improved survival as compared to control mice. Irradiated mice administered human milk oligosaccharides and B. longum subsp. infantis may have clinical scores indicating reduced severity of gastrointestinal damage or disease in mice administered human milk oligosaccharides and B. longum subsp. infantis as compared to controls. These mice may also have less instances of diarrhea or runny stool than control mice.
[0174] At the study endpoint, mice are sacrificed and plasma, cecal contents, and tissue samples are harvested. Stool samples and/or cecal contents may be evaluated to quantify microbial compositions of intestinal microbiota. Samples from mice administered human milk oligosaccharides and B. longum subsp. infantis may have greater levels, amounts, or portions of B. longum subsp. infantis than samples from control mice. These samples may also have greater diversity or less levels, amounts, or portions or percentage (e.g., relative to total intestinal microbiota) of pathogenic bacteria.
[0175] Histological analysis of intestinal tissue samples may indicate a higher degree or more instances of tissue regeneration, e.g., intestinal crypt regeneration, in mice administered human milk oligosaccharides and B. longum subsp. infantis than are observed in control mice. Histological analyses may also reveal more cellular proliferation and/or more cells positive for proliferation markers (e.g., Ki-67) in intestinal tissues collected from mice administered human milk oligosaccharides and B. longum subsp. infantis than observed tissues from control mice. Tests may also indicate reduced_intestinal barrier permeability and/or reduced pathogen translocation in mice administered human milk oligosaccharides and B. longum subsp. infantis as compared to mice in control groups.
[0176] Tissues and/or plasma samples collected from irradiated mice may also be tested for markers of inflammation. Tissues and/or plasma samples collected from mice administered human milk oligosaccharides and B. longum subsp. infantis may have lower detectable levels or amounts of one or more inflammatory markers compared to plasma and/or tissues collected from mice in control groups.
[0177] Intestinal contents and plasma samples from irradiated mice may also be tested for the presence, amount, and/or concentration of metabolites, e.g., short chain fatty acids such as including acetate, lactate, butyrate, and/or propionate. Irradiated mice
administered B. longum subsp. infantis may have altered levels of metabolites as compared to controls, including metabolites such as short chain fatty acids and/or metabolites with known or suspected anti-inflammatory properties.
[0178] Follow-on experiments may include experimental groups of irradiated mice administered human milk oligosaccharides, B. longum subsp. infantis, and Veillonella sp. Observations of increased survival, reduced gastrointestinal damage severity, increased regeneration, reduced intestinal permeability, and/or reduced inflammation with respect to irradiated mice administered with B. longum subsp. infantis and human milk oligosaccharides and/or mice treated with controls may be observed.
[0179] Similar experiments may be performed in other animal models, such as in rats, cats, dogs, or non-human primates. In these experiments, irradiated animals orally administered B. longum subsp. infantis and human milk oligosaccharides may have increased survival, reduced gastrointestinal damage severity, increased regeneration, reduced intestinal permeability, and/or reduced inflammation with respect to animals receiving control treatments.
EXAMPLE 14: VEILLONELLA SP. CULTURED IN MEDIA CONTAINING LACTATE PRODUCE PROPIONATE
[0180] Various strains of bacteria including strains from the genus Veillonella were cultured in a yeast casitone fatty acid (YCFA) broth similar to as described in Duncan et al., (International Journal of Systematic and Evolutionary Microbiology, 52(6): 2141-2146 (2002)), but lacking propionate and valerate, and with lactate added as the main fermentable carbohydrate source (1.25% weight/volume). After incubation for 48 hours, propionate and lactate levels were measured. As shown in FIG. 12, propionate was detected above background levels (approximately 0 pg/mL) in the incubated media accompanied by decreased levels of lactate as compared to initial levels (dotted vertical line) for all Veillonella sp. strains tested. Similar levels of propionate and lactate were observed in cultures of Anaerotignum lactatifermentans and Megasphera massiliensis . Detectable levels of propionate were also detected in media from Bacteroides fragilis (B. fragilis) cultures. While propionate detection was coupled with decreased lactate in media from some B. fragilis test strains, other media from other B. fragilis test strains had detectable propionate levels and increased lactate concentration relative to initial lactate levels. Notably, while
lactate was provided as the main carbon source, the YCFA media also contained other potential carbon sources such as acetate and amino acids. Taken together, these results are consistent with the capability for Veillonella sp., as well as species of the genera Anaerotignum and Megasphaera and some strains of B. fragilis, to produce propionate in the presence of lactate.
EXAMPLE 15: ENHANCED IN VIVO PRODUCTION OF PROPIONATE BY VEILLONELLA SP. COCULTURED WITH B. LONGUM SUBSP. INFANTIS AND HMO
[0181] In vivo propionate production was assessed in various strains of Veillonella in mice initially inoculated with Veillonella sp. or B. longum subsp. infantis, and then subsequently treated with oral gavage of phosphate buffered saline (PBS) or B. longum subsp. infantis with human milk oligosaccharides (HMO) similar to as described in Example 12. The Veillonella test strains included V. parvula, V. atypica, and the two Veillonella strains isolated as described in Example 11.
[0182] As shown in FIGS. 13A-C, mice inoculated with V. parvula or the isolated Veillonella strains had detectable levels of Veillonella in stool at study days 8-11 regardless of whether they were treated with B. longum subsp. infantis and HMO. Similarly, mice inoculated with ZZ. longum subsp. infantis had detectable levels of B. longum subsp. infantis in stool at study days 8-11 regardless of whether they were treated with HMO. In mice initially inoculated with V. parvula or the Veillonella strains and later administered B. longum subsp. infantis and HMO, stool levels of B. longum subsp. infantis increased from the level of detection at day 8 to detectable levels at days 10 and 11. Of the eighteen mice inoculated with the strain of V. atypica prepared for these experiments, fifteen mice across experimental conditions did not display V. atypica engraftment (data not shown).
[0183] Three hours after the final gavage, mice were euthanized and levels of propionate, lactate, and acetate in different intestinal segments were quantified. Increased levels of propionate were observed in cecal samples collected from FeZZZcweZZa-inoculated test mice administered B. longum subsp. infantis and HMO with respect to cecal samples collected from control mice (FIG. 14A-14C). In contrast, lower levels of lactate were detected in cecal samples collected from Veillonella- -inoculated test mice administered B. longum subsp. infantis and HMO than in samples from control mice that received B. longum subsp. infantis and HMO (FIG. 15A-15C). Acetate levels did not appear to be reduced in cecal samples collected from Veillonella-inoculated test mice administered B. longum subsp.
I l l
infantis and HMO as compared to samples from control mice that received B. longum subsp. infantis and HMO (FIG. 16A-16C). Taken together, these data are consistent with in vivo cross-feeding whereby Veillonella sp. consumes lactate produced by B. longum subsp. infantis through HMO consumption.
EXAMPLE 16: ENHANCED IN VIVO PRODUCTION OF PROPIONATE BY MEGASPHAERA SP. COCULTURED WITH B. LONGUM SUBSP. INFANTIS AND HMO
[0184] In vivo propionate production was assessed in Megasphaera elsdenii (M. elsdenii) in mice initially inoculated with M. elsdenii or B. longum subsp. infantis, and then subsequently treated with oral gavage of phosphate buffered saline (PBS) or B. longum subsp. infantis with human milk oligosaccharides (HMO) similar to as described in Example 12. Germ-free mice were initially inoculated with a A-/. elsdenii or B. longum subsp. infantis as a control. After a one-week stabilization, mice were gavaged with B. longum subsp. infantis and HMO followed by daily gavage with HMO for three days or with PBS.
[0185] As shown in FIG. 17A, mice inoculated with M. elsdenii had detectable levels of M. elsdenii in stool at study days 8-11 regardless of whether they were treated with PBS or B. longum subsp. infantis and HMO. Similarly, mice inoculated with B. longum subsp. infantis had detectable levels of B. longum subsp. infantis in stool at study days 8-11 regardless of whether they were treated with HMO. In mice initially inoculated with M. elsdenii and later administered B. longum subsp. infantis and HMO, stool levels of B. longum subsp. infantis increased from the level of detection at day 8 to detectable levels at days 10 and 11.
[0186] Three hours after the final gavage, mice were euthanized and levels of short chain fatty acids were assed in cecal samples collected from the mice. Levels of butyrate, propionate, valerate, lactate, and acetate in cecal contents were quantified (FIGS. 17B-17F). Increased levels of butyrate, propionate, and valerate were observed in samples from mice inoculated with
elsdenii that were administered B. longum subsp. infantis and HMO as compared to samples collected from control mice (FIGS. 17B-17D). In contrast, lower levels of lactate were detected in AL elsdenii inoculated mice administered B. longum subsp. infantis and HMO than in samples from control mice that received B. longum subsp. infantis and HMO (FIG. 17E). Acetate levels did not appear to be reduced in cecal samples collected from Veillonella-inoculated test mice administered B. longum subsp. infantis and HMO as
compared to samples from control mice that received B. longum subsp. infantis and HMO (FIG. 17F).
Claims
1. A method of treating or preventing a disease, disorder, or condition associated with one or more of inflammation, immune dysfunction, cancer, allergy, or dysbiosis of the intestinal microbiome in a subject in need thereof, the method comprising administering to the subject i) a prebiotic mixture comprising one or more human milk oligosaccharides, ii) at least one Bifidobacterium capable of consuming the one or more human milk oligosaccharides; and iii) at least one propionate producing bacterium.
2. A method of ameliorating a symptom of a disease, disorder, or condition associated with one or more of inflammation, immune dysfunction, cancer, allergy, or dysbiosis of the intestinal microbiome in a subject in need thereof, the method comprising administering to the subject i) a prebiotic mixture comprising one or more human milk oligosaccharides, ii) at least one Bifidobacterium capable of consuming the one or more human milk oligosaccharides; and iii) at least one propionate producing bacterium.
3. The method of claim 1 or 2, wherein the subject has received or will receive an allogenic hematopoietic stem cell transplant, and wherein the disease, condition, or disorder comprises graft versus host disease.
4. The method of claim 1 or 2, wherein the disease, disorder, or condition comprises one or more of obesity, type II diabetes, a chronic inflammatory disease, an autoimmune disease, an infection, an infectious disease domination, bowel resection, or a condition associated with chronic diarrhea.
5. The method of claim 1 or 2, wherein the disease, disorder, or condition comprises one or more of irritable bowel syndrome (IBS), inflammatory bowel disease (IBD), short bowel syndrome (SBS), celiac disease, small intestinal bacterial overgrowth (SIBO), gastroenteritis, leaky gut syndrome, pouchitis, or gastric lymphoma.
6. The method of claim 1 or 2, wherein the disease, disorder, or condition comprises an atopic disease.
7. The method of claim 6, wherein the atopic disease comprises atopic dermatitis, food allergy, and/or asthma.
8. The method of any of claim 1 or 2, wherein the disease, condition, or disorder is associated with an infection.
9. The method of claim 8, wherein the infection comprises a bacterial infection or gut domination.
10. The method of claim 9, wherein the bacterial infection or gut domination comprises an infection or gut domination by one or more species, subspecies, or strains of Aeromonas, Bacillus, Bordetella, Brucella, Burkholderia, Campylobacter, Chlamydia, Chlamydophila, Citrobacter, Clostridium, Corynebacterium, Coxiella, Ehrlichia, Enterobacter, Enterobacteriaceae, Enterococcus, Escherichia, Francisella, Haemophilus, Klebsiella, Legionella, Leptospira, Listeria, Morganella, Mycobacterium, Mycoplasma, Neisseria, Orientia, Plesiomonas, Proteus, Pseudomonas, Rickettsia, Salmonella, Shigella, Staphylococcus, Streptococcus, Treponema, Vibrio, or Yersinia, optionally one or more of Aeromonas hydrophila, Bacillus cereus, Campylobacter fetus, Campylobacter jejuni, Clostridium botulinum, Clostridium difficile, Clostridium perfringens, enteroaggregative Escherichia coli, enterohemorrhagic Escherichia coli, enteroinvasive Escherichia coli, enteropathogenic E. coli, enterotoxigenic Escherichia coli, Escherichia coli 0157:H7, Helicobacter pylori, Klebsiella pneumoniae, Listeria monocytogenes, Salmonella paratyphi, Salmonella typhi, Staphylococcus aureus, Vibrio cholerae, Vibrio parahaemolyticus, Vibrio vulnificus, or Yersinia enterocolitica.
11. The method of claim 9 or 10, wherein the bacterial infection or gut domination comprises an infection or gut domination by one or more of Citrobacter freundii, Citrobacter koseri, Enterobacter aerogenes, Enterobacter cloacae, Enterococcus faecalis, Enterococcus faecium, Escherichia coli, Klebsiella oxytoca, Klebsiella pneumoniae, Morganella morganii, Proteus mirabilis, Serratia marcescens, Staphylococcus aureus, Staphylococcus epidermidis, Streptococcus anginosus, Streptococcus australis, Streptococcus constellatus, Streptococcus cristatus, Streptococcus gordonii, Streptococcus infantis, Streptococcus intermedins, Streptococcus mitis, Streptococcus mutans, Streptococcus oligofermentans, Streptococcus
oralis, Streptococcus parasanguinis, Streptococcus peroris, Streptococcus pneumoniae, Streptococcus pseudopneumoniae, Streptococcus salivarius, Streptococcus sanguinis, Streptococcus sobrinus, Streptococcus tigurinus, or Streptococcus vestibularis.
12. The method of any of claims 9-11, wherein the bacterial infection or gut domination comprises an infection or gut domination by drug-resistant bacteria.
13. The method of claim 12, wherein the drug-resistant bacteria comprises one or more of antibiotic-resistant bacterium (ARB), Antibiotic-resistant Proteobacteria, Carbapenem-resistantEwtero zcterzaceae (CRE), Extended Spectrum Beta-Lactamase producing Enterobacterales (ESBL-E), fluoroquinolone-resistant Enterobacteriaceae, vancomycin-resistant Enterococci (VRE), multi-drug resistant A. coli, or multi-drug resistant Klebsiella.
14. The method of claim 1 or 2, wherein the subject has undergone or will undergo an ileal pouch-anal anastomosis (IPAA) surgery, and wherein the disease, condition, or disorder comprises pouchitis.
15. A method for increasing propionate concentration and/or propionate production in the gut of a subject in need thereof, the method comprising administering to the subject i) a prebiotic mixture comprising one or more human milk oligosaccharides, ii) at least one Bifidobacterium capable of consuming the one or more human milk oligosaccharides; and iii) at least one propionate producing bacterium.
16. The method of any of claims 1-15, wherein the at least one propionate producing bacterium comprises a strain that is isolated from a human intestinal microbiome and/or is capable of engrafting within the human intestinal microbiome.
17. The method of any of claims 1-16, wherein the at least one propionate producing bacterium comprises one or more strains of the genus Veillonella.
18. The method of claim 1-17, wherein the at least one propionate producing bacterium comprises one or more of Veillonella atypica (V. atypica), Veillonella dispar (V.
dispar), Veillonella infantium (V. infantium), Veillonella nakazawae (V. nakazawae), Veillonella parvula (V. parvula), Veillonella ratti (V. ratti), Veillonella rogosae (V. rogosae), Veillonella seminalis (V. seminalis), and/or Veillonella tobetsuensis (V. tobetsuensis).
19. The method of any of claims 1-17, wherein the at least one propionate producing bacterium comprises V atypica, V dispar, V. infantium, V. nakazawae, V. parvula, and/or V. rogosae.
20. The method of any of claims 1-17, wherein the at least one propionate producing bacterium comprises V. infantium, V. nakazawae, V. parvula, and/or V. rogosae.
21. The method of any of claims 1-17, wherein the at least one propionate producing bacterium comprises V. infantium, V. dispar, and/or V. nakazawae.
22. The method of any of claims 1-17, wherein the at least one propionate producing bacterium comprises V. parvula.
23. The method of any of claims 1-17, wherein the at least one propionate producing bacterium comprises V. rogosae.
24. The method of any of claims 1-23, wherein the at least one propionate producing bacterium comprises one or more strains of the genus Megasphaera.
25. The method of claim 24, wherein the at least one propionate producing bacterium comprises one or more of Megasphaera elsdenii (M. elsdenii), Megasphaera hominis (M. hominis), Megasphaera indica (M. indica), Megasphaera massiliensis (M.massiliensis), and/ or Megasphaera micronuciformis (M. micronuciformis).
26. The method of claim 25, wherein the at least one propionate producing bacterium comprises one or more oiM. elsdenii ndJ M. massiliensis.
27. The method of any of claims 1-26, wherein the at least one propionate producing bacterium comprises one or more strains of the genus Anaerotignum.
28. The method of claim 27, wherein the at least one propionate producing bacterium comprises Anaerotignum lactatifermentans (A. lactatifermentans).
29. The method of any of claims 1-28, wherein the at least one propionate producing bacterium comprises one or more strains of the genus Bacteroides.
30. The method of claim 29, wherein the at least one propionate producing bacterium comprises Bacteroides fragilis (B. fragilis) o Bacteroides caccae (B. caccae).
31. The method of any of claims 1-30, wherein the at least one propionate producing bacterium comprises one or more strains of the genus Coprococcus.
32. The method of claim 31, wherein the at least one propionate producing bacterium comprises Coprococcus catus (C. catus).
33. The method of any of claims 1-32, wherein the at least one propionate producing bacterium comprises one or more strains of the genus Merdimmobilis.
34. The method of claim 33, wherein the at least one propionate producing bacterium comprises Merdimmobilis hominis.
35. The method of any of claims 1-34, wherein the propionate producing bacterium comprises a nucleotide sequence with at least 97%, 98%, or 99% sequence identity to any of SEQ ID NOS: 40-49 or 52-58 and/or an amino acid sequence with at least 97%, 98%, or 99% sequence identity to SEQ ID NO: 50 or 51.
36. The method of any of claims 1-35, wherein the Bifidobacterium comprises B. breve, B. bifidum, or B. longum subsp. infantis.
37. The method of any of claims 1-36, wherein the Bifidobacterium comprises B. longum subsp. infantis.
38. The method of any of claims 1-37, further comprising at least one butyrate producing strain of bacteria.
39. The method of claim 38, wherein the at least one butyrate producing strain comprises a strain of Clostridium Cluster IV or Clostridium Cluster XlVa bacteria.
40. The method of claim 38 or 39, wherein the at least one butyrate producing strain comprises one or more of Agathobacter rectalis, Anaerobutyricum hallii, Anaerostipes caccae, Blautia producta, Clostridium leptum, Faecalibacterium prausnitzii, Anaerobutyricum soehngenii, Roseburia hominis, or Roseburia intestinalis.
41. The method of any of claims 38-40, wherein the at least one butyrate producing strain comprises one or more of Anaerostipes caccae, Clostridium innocuum, Roseburia hominis, or Roseburia intestinalis.
42. The method of any of claims 1-41, wherein the prebiotic mixture comprises one or more of 2'-fucosyllactose, 3-fucosyllactose, difucosyllactose, 3’-sialyllactose, 6'- sialyllactose, lacto-N-tetraose, lacto-N-neo-tetraose, lacto-N-fucopentaose I, lacto-N- fucopentaose II, lacto-N-fucopentaose III, sialyl-lacto-N-tetraose a, sialyl-lacto-N-tetraose b, sialyl-lacto-N-tetraose c, lacto-N-difuco-hexaose I, lacto-N-difuco-hexaose II, lacto-N- hexaose, para-1 acto-N-hexaose, disialyllacto-N-tetraose, Fucosyl-lacto-N-hexaose, difucosyl- lacto-N-hexaose a, difucosyl-lacto-N-hexaose b, lactodifucotetraose, 6’galactosyllactose,
3 ’galactosyllactose, 3-sialyl -3-fucosyllactose, sialylfucosyllacto-N-tetraose, sialyllacto-N- fucopentaose V, disialyl-lacto-n-fucopentaose II, disialyl-lacto-n-fucopentaose V, lacto-N- neo-difucohexaose II, 3-Fucosyl-sialylacto-N-tetraose c, para-lacto-N-neohexose, lacto-N- octaose, lacto-N-neooctaose, lacto-N-neohexaose, lacto-N-fucopentaose V, iso-lacto-N- octaose, para-lacto-N-octaose, lacto-decaose, or sialyl-lacto-N-fucopentaose I.
43. The method of any of claims 1-42, wherein the prebiotic mixture comprises one or more of 2'-fucosyllactose, 3-fucosyllactose, 3'-sialyllactose, 6'-sialyllactose, lacto-N- tetraose, lacto-N-difucohexaose I, lactodifucotetraose, lacto-N-fucopentaose I, sialylacto-N- tetraose c, sialylacto-N-tetraose b, or disialyllacto-N-tetraose.
44. The method of any of claims 1-42, wherein the prebiotic mixture comprises one or more of 2'-fucosyl-lactose, 3-fucosyllactose, 3’-sialyllactose, 6'-sialyllactose, lacto-N- tetraose, lacto-N-neotetraose, or difucosyllactose.
45. The method of any of claims 1-42, wherein the prebiotic mixture comprises one or more of 2'-fucosyllactose, 3-fucosyllactose, lacto-N-tetraose, or lacto-N-neotetraose.
46. The method of any of claims 1-42, wherein the prebiotic mixture comprises one or both of 2'-fucosyllactose and lacto-N-neotetraose.
47. The method of any of claims 1-46, wherein the prebiotic mixture comprises at least 10, at least 25, at least 50, at least 100, or at least 150 human milk oligosaccharides.
48. The method of any of claims 1-47, wherein the prebiotic mixture comprises 2'- fucosyllactose, 3-fucosyllactose, 3’-sialyllactose, 6'-sialyllactose, lacto-N-tetraose, lacto-N- difucohexaose I, lactodifucotetraose, lacto-N-fucopentaose I, sialylacto-N-tetraose c, sialylacto-N-tetraose b, and disialyllacto-N-tetraose.
49. The method of any of claims 1-48, wherein the prebiotic mixture is, is derived from, or comprises a concentrated human milk permeate, wherein the concentrated human milk permeate is obtained by a process comprising the steps of ultra-filtering human skim milk to obtain human milk permeate and concentrating the human milk oligosaccharide content of the human milk permeate.
50. The method of claim 49, wherein the human skim milk is obtained from human milk pooled from at least 25, 50, or 100 individual donors.
51. A kit comprising i) a prebiotic mixture comprising one or more human milk oligosaccharides, ii) at least one Bifidobacterium capable of consuming the one or more human milk oligosaccharides; and iii) at least one propionate producing bacterium.
52. The kit of claim 51, wherein the at least one propionate producing bacterium comprises a strain that is isolated from a human intestinal microbiome and/or is capable of engrafting within the human intestinal microbiome.
53. The kit of claim 51 or 52, wherein the at least one propionate producing bacterium comprises one or more strains of the genus Veillonella.
54. The kit of any of claims 51-53, wherein the at least one propionate producing bacterium comprises one or more of Veillonella atypica (V. atypica), Veillonella dispar (V. dispar), Veillonella infantium (V. infantium), Veillonella nakazawae (V. nakazawae), Veillonella parvula (V. parvula), Veillonella ratti (V. ratti), Veillonella rogosae (V. rogosae), Veillonella seminalis (V. seminalis), and/or Veillonella tobetsuensis (V. tobetsuensis).
55. The kit of any of claims 51-54, wherein the at least one propionate producing bacterium comprises V atypica, V dispar, V. infantium, V. nakazawae, V. parvula, and/or V. rogosae.
56. The kit of any of claims 51-54, wherein the at least one propionate producing bacterium comprises V. infantium, V. nakazawae, V. parvula, and/or V. rogosae.
57. The kit of any of claims 51-54, wherein the at least one propionate producing bacterium comprises V. infantium, V. dispar, and/or V. nakazawae.
58. The kit of any of claims 51-54, wherein the at least one propionate producing bacterium comprises V. parvula.
59. The kit of any of claims 51-54, wherein the at least one propionate producing bacterium comprises V. rogosae.
60. The kit of any of claims 51-59, wherein the at least one propionate producing bacterium comprises one or more strains of the genus Megasphaera.
61. The kit of claim60, wherein the at least one propionate producing bacterium comprises one or more of Megasphaera elsdenii (M elsdenii), Megasphaera hominis (M hominis), Megasphaera indica (M indica), Megasphaera massiliensis (M massiliensis), and/ or Megasphaera micronuciformis (M micronuciformis).
62. The kit of claim 60 or 61, wherein the at least one propionate producing bacterium comprises one or more of A7. elsdenii a Mo M. massiliensis.
63. The kit of any of claims 51-62, wherein the at least one propionate producing bacterium comprises one or more strains of the genus Anaerotignum.
64. The kit of claim 63, wherein the at least one propionate producing bacterium comprises Anaerotignum lactatifermentans (A. lactatifermentans).
65. The kit of any of claims 51-64, wherein the at least one propionate producing bacterium comprises one or more strains of the genus Bacteroides.
66. The kit of claim 65, wherein the at least one propionate producing bacterium comprises Bacteroides fragilis (B. fragilis) o Bacteroides caccae (B. caccae).
67. The kit of any of claims 51-66, wherein the at least one propionate producing bacterium comprises one or more strains of the genus Coprococcus.
68. The kit of claim 67, wherein the at least one propionate producing bacterium comprise Coprococcus catus (C. catus).
69. The kit of any of claims 51-68, wherein the at least one propionate producing bacterium comprises one or more strains of the genus Merdimmobilis .
70. The kit of claim 69, wherein the at least one propionate producing bacterium comprises Merdimmobilis hominis.
71. The kit of any of claims 51-70, wherein the propionate producing bacterium comprises a nucleotide sequence with at least 97%, 98%, or 99% sequence identity to any of SEQ ID NOS: 40-49 or 52-58 and/or an amino acid sequence with at least 97%, 98%, or 99% sequence identity to SEQ ID NO: 50 or 51.
72. The kit of any of claims 51-71, wherein the Bifidobacterium comprises B. breve, B. bifidum, or B. longum subsp. infantis.
73. The kit of any of claims 51 -72, wherein the Bifidobacterium comprises B. longum subsp. infantis.
74. The kit of any of claims 51-73, further comprising at least one butyrate producing strain of bacteria.
75. The kit of claim 74, wherein the at least one butyrate producing strain comprises a strain of Clostridium Cluster IV or Clostridium Cluster XlVa bacteria.
76. The kit of claim 74 or 75, wherein the at least one butyrate producing strain comprises one or more of Agathobacter rectalis, Anaerobutyricum hallii, Anaerostipes caccae, Blautia producta, Clostridium leptum, Faecalibacterium prausnitzii, Anaerobutyricum soehngenii, Roseburia hominis, or Roseburia intestinalis.
77. The kit of claim 74 or 75, wherein the at least one butyrate producing strain comprises one or more of Anaerostipes caccae, Clostridium innocuum, Roseburia hominis, or Roseburia intestinalis.
78. The kit of any of claims 51-77, wherein the prebiotic mixture comprises one or more of 2'-fucosyllactose, 3-fucosyllactose, difucosyllactose, 3’-sialyllactose, 6'-sialyllactose, lacto-N-tetraose, lacto-N-neo-tetraose, lacto-N-fucopentaose I, lacto-N-fucopentaose II, lacto-N-fucopentaose III, sialyl-lacto-N-tetraose a, sialyl-lacto-N-tetraose b, si alyl -lacto-N- tetraose c, lacto-N-difuco-hexaose I, lacto-N-difuco-hexaose II, lacto-N-hexaose, para-lacto- N-hexaose, disialyllacto-N-tetraose, Fucosyl-lacto-N-hexaose, difucosyl-lacto-N-hexaose a, difucosyl-lacto-N-hexaose b, lactodifucotetraose, 6’galactosyllactose, 3 ’galactosyllactose, 3-
sialyl-3 -fucosyllactose, sialylfucosyllacto-N-tetraose, sialyllacto-N-fucopentaose V, disialyl- lacto-n-fucopentaose II, disialyl-lacto-n-fucopentaose V, lacto-N-neo-difucohexaose II, 3- Fucosyl-sialylacto-N-tetraose c, para-lacto-N-neohexose, lacto-N-octaose, lacto-N- neooctaose, lacto-N-neohexaose, lacto-N-fucopentaose V, iso-lacto-N-octaose, para-lacto-N- octaose, lacto-decaose, or sialyl-lacto-N-fucopentaose I.
79. The kit of any of claims 51-78, wherein the prebiotic mixture comprises one or more of 2'-fucosyllactose, 3 -fucosyllactose, 3'-sialyllactose, 6'-sialyllactose, lacto-N-tetraose, lacto-N-difucohexaose I, lactodifucotetraose, lacto-N-fucopentaose I, sialylacto-N-tetraose c, sialylacto-N-tetraose b, or disialyllacto-N-tetraose.
80. The kit of any of claims 51-78, wherein the prebiotic mixture comprises one or more of 2'-fucosyl-lactose, 3 -fucosyllactose, 3’-sialyllactose, 6'-sialyllactose, lacto-N- tetraose, lacto-N-neotetraose, or difucosyllactose.
81. The kit of any of claims 51-78, wherein the prebiotic mixture comprises one or more of 2'-fucosyllactose, 3 -fucosyllactose, lacto-N-tetraose, or lacto-N-neotetraose.
82. The kit of any of claims 51-78, wherein the prebiotic mixture comprises one or both of 2'-fucosyllactose and lacto-N-neotetraose.
83. The kit of any of claims 51-82, wherein the prebiotic mixture comprises at least 10, at least 25, at least 50, at least 100, or at least 150 human milk oligosaccharides.
84. The kit of any of claims 51-83, wherein the prebiotic mixture comprises 2'- fucosyllactose, 3 -fucosyllactose, 3’-sialyllactose, 6'-sialyllactose, lacto-N-tetraose, lacto-N- difucohexaose I, lactodifucotetraose, lacto-N-fucopentaose I, sialylacto-N-tetraose c, sialylacto-N-tetraose b, and disialyllacto-N-tetraose.
85. The kit of any of claims 51-84, wherein the prebiotic mixture is, is derived from, or comprises a concentrated human milk permeate, wherein the concentrated human milk permeate is obtained by a process comprising the steps of ultra-filtering human skim
milk to obtain human milk permeate and concentrating the human milk oligosaccharide content of the human milk permeate.
86. The kit of claim 85, wherein the human skim milk is obtained from human milk pooled from at least 25, 50, or 100 individual donors.
87. A method for increasing propionate concentration and/or propionate production in the gut of a subject in need thereof, the method comprising administering to the subject the prebiotic mixture, the at least one Bifidobacterium, and the at least one propionate producing bacterium of the kit of any of claims 51-86.
88. A method of treating or preventing a disease, disorder, or condition associated with one or more of inflammation, immune dysfunction, cancer, allergy, or dysbiosis of the intestinal microbiome in a subject in need thereof, the method comprising administering to the subject the prebiotic mixture, the at least one Bifidobacterium, and the at least one propionate producing bacterium of the kit of any of claims 51-86.
89. A method of ameliorating a symptom of a disease, disorder, or condition associated with one or more of inflammation, immune dysfunction, cancer, allergy, or dysbiosis of the intestinal microbiome in a subject in need thereof, the method comprising administering to the subject the prebiotic mixture, the at least one Bifidobacterium, and the at least one propionate producing bacterium of the kit of any of claims 51-86.
90. A pharmaceutical composition comprising i) a prebiotic mixture comprising one or more human milk oligosaccharides, ii) at least one Bifidobacterium capable of consuming the one or more human milk oligosaccharides; and iii) at least one propionate producing bacterium.
91. The pharmaceutical composition of claim 90, wherein the at least one Bifidobacterium comprises B. longum subsp. inf antis and the at least one propionate producing bacterium comprises one or more strains of the genus Veillonella.
92. The pharmaceutical composition of claim 90 or claim 91, further comprising at least one butyrate producing strain of bacteria, optionally wherein the at least one butyrate producing strain comprises one or more of Agathobacter rectalis, Anaerobutyricum hallii, Anaerostipes caccae, Blautia producta, Clostridium leptum, Faecalibacterium prausnitzii, Anaerobutyricum soehngenii, Roseburia hominis, or Roseburia intestinalis.
93. A method for treating one or more symptoms of acute radiation syndrome in a subject in need thereof, the method comprising administering to the subject i) a prebiotic mixture comprising one or more human milk oligosaccharides and ii) at least one Bifidobacterium capable of consuming the one or more human milk oligosaccharides, thereby treating the symptoms of acute radiation syndrome.
94. The method of claim 93, wherein the at least one Bifidobacterium comprises B. longum subsp. infantis.
95. The method of claim 93 or 94, further comprising administering to the subject in need thereof iii) at least one propionate producing bacterium.
96. The method of claim 95, wherein the at least one propionate producing bacterium comprises one or more strains of the genus Veillonella.
97. The method of any of claims 93-96, wherein the one or more symptoms of acute radiation syndrome comprise gastrointestinal damage.
98. The method of any of claims 93-97, wherein the subject was exposed to an ionizing radiation dose of at least 0.3 Gray (Gy), optionally at least 0.7 Gy, 1 Gy, 2 Gy, 5 Gy, 6 Gy, or 10 Gy over a period of time lasting under 60 minutes, optionally under 30 minutes, 15 minutes, 10 minutes, 5 minutes, 3 minutes, 2 minutes, or 1 minute.
99. An article of manufacture, comprising the kit any one of claims 51-86 and instructions for use, wherein the instructions for use describe the method of any of claims 1- 50, 87-89, and 93-98.
Applications Claiming Priority (4)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US202263387807P | 2022-12-16 | 2022-12-16 | |
US63/387,807 | 2022-12-16 | ||
US202363490283P | 2023-03-15 | 2023-03-15 | |
US63/490,283 | 2023-03-15 |
Publications (2)
Publication Number | Publication Date |
---|---|
WO2024130119A2 true WO2024130119A2 (en) | 2024-06-20 |
WO2024130119A3 WO2024130119A3 (en) | 2024-07-18 |
Family
ID=89663231
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2023/084290 WO2024130119A2 (en) | 2022-12-16 | 2023-12-15 | Synbiotic compositions for short chain fatty acid production |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2024130119A2 (en) |
Citations (34)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US5589168A (en) | 1991-04-08 | 1996-12-31 | Unilever Patent Holdings B.V. | Probiotic |
US5989463A (en) | 1997-09-24 | 1999-11-23 | Alkermes Controlled Therapeutics, Inc. | Methods for fabricating polymer-based controlled release devices |
US6203797B1 (en) | 1998-01-06 | 2001-03-20 | Stephen C. Perry | Dietary supplement and method for use as a probiotic, for alleviating the symptons associated with irritable bowel syndrome |
US6835376B1 (en) | 1999-03-11 | 2004-12-28 | Nestec S.A. | Lactobacillus paracasei strain for preventing diarrhea caused by pathogenic bacteria |
WO2010065652A1 (en) | 2008-12-02 | 2010-06-10 | Prolacta Bioscience, Inc. | Human milk permeate compositions and methods of making and using same |
US7914822B2 (en) | 2001-05-14 | 2011-03-29 | Prolacta Bioscience, Inc. | Method of producing nutritional products from human milk tissue and compositions thereof |
US7943315B2 (en) | 2005-09-20 | 2011-05-17 | Prolacta Bioscience, Inc. | Methods for testing milk |
WO2012097950A1 (en) | 2011-01-20 | 2012-07-26 | Jennewein Biotechnologie Gmbh | Novel fucosyltransferases and their applications |
WO2013044928A1 (en) | 2011-09-30 | 2013-04-04 | Glycom A/S | Synthesis of hmo core structures |
WO2013139344A1 (en) | 2012-03-20 | 2013-09-26 | Glycom A/S | Synthesis of the trisaccharide 3-o-fucosyllactose and intermediates thereof |
US8545920B2 (en) | 2006-11-29 | 2013-10-01 | Prolacta Bioscience Inc. | Human milk compositions and methods of making and using same |
WO2013182206A1 (en) | 2012-06-08 | 2013-12-12 | Glycom A/S | Method for producing oligosaccharides and oligosaccharide glycosides by fermentation |
WO2013190531A1 (en) | 2012-06-22 | 2013-12-27 | Glycom A/S | Glycosylated galactosyl disaccharides, methods for their production and their use in consumable products |
WO2013190530A1 (en) | 2012-06-22 | 2013-12-27 | Glycom A/S | Modified galactooligosaccharides |
WO2014135167A1 (en) | 2013-03-08 | 2014-09-12 | Glycom A/S | Purification of oligosaccaharides by reversible derivatization |
WO2014167538A1 (en) | 2013-04-12 | 2014-10-16 | Glycom A/S | Synthesis of sialylated/fucosylated human milk oligosaccharides |
WO2014167537A1 (en) | 2013-04-12 | 2014-10-16 | Glycom A/S | Synthesis of sialylated/fucosylated oligosaccharides |
WO2015032413A1 (en) | 2013-09-06 | 2015-03-12 | Glycom A/S | Fermentative production of oligosaccharides |
WO2015036138A1 (en) | 2013-09-10 | 2015-03-19 | Jennewein Biotechnologie Gmbh | Production of oligosaccharides |
WO2015049331A1 (en) | 2013-10-04 | 2015-04-09 | Jennewein Biotechnologie Gmbh | Process for purification of a neutral human milk oligosaccharide using simulated moving bed chromatography |
WO2015106943A1 (en) | 2014-01-20 | 2015-07-23 | Jennewein Biotechnologie Gmbh | PROCESS FOR EFFICIENT PURIFICATION OF NEUTRAL HUMAN MILK OLIGOSACCHARIDES (HMOs) FROM MICROBIAL FERMENTATION |
US20150238545A1 (en) | 2000-07-25 | 2015-08-27 | Thomas Julius Borody | Probiotic recolonisation therapy |
US9149052B2 (en) | 2006-08-30 | 2015-10-06 | Prolacta Bioscience, Inc. | Methods of obtaining sterile milk and compositions thereof |
WO2015150328A1 (en) | 2014-03-31 | 2015-10-08 | Jennewein Biotechnologie Gmbh | Total fermentation of oligosaccharides |
US20150359894A1 (en) | 2006-09-27 | 2015-12-17 | Little Calumet Holdings, Llc | Probiotic oral dosage forms |
WO2015197082A1 (en) | 2014-06-27 | 2015-12-30 | Glycom A/S | Oligosaccharide production |
WO2017042382A1 (en) | 2015-09-12 | 2017-03-16 | Jennewein Biotechnologie Gmbh | Production of human milk oligosaccharides in microbial hosts with engineered import / export |
WO2017101958A1 (en) | 2015-12-18 | 2017-06-22 | Glycom A/S | Fermentative production of oligosaccharides |
WO2018053535A1 (en) | 2016-09-19 | 2018-03-22 | Prolacta Bioscience, Inc. | Purified human milk oligosaccharides compositions |
WO2018077892A1 (en) | 2016-10-29 | 2018-05-03 | Jennewein Biotechnologie Gmbh | Improved process for the production of fucosylated oligosaccharides |
WO2019008133A1 (en) | 2017-07-07 | 2019-01-10 | Jennewein Biotechnologie Gmbh | Fucosyltransferases and their use in producing fucosylated oligosaccharides |
WO2019043029A1 (en) | 2017-08-29 | 2019-03-07 | Jennewein Biotechnologie Gmbh | Process for purifying sialylated oligosaccharides |
WO2021061991A1 (en) | 2019-09-24 | 2021-04-01 | Prolacta Bioscience, Inc. | Compositions and methods for treatment of inflammatory and immune diseases |
WO2022036225A1 (en) | 2020-08-14 | 2022-02-17 | Prolacta Bioscience, Inc. | Human milk oligosaccharide compositions for use with bacteriotherapies |
Family Cites Families (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2010057022A1 (en) * | 2008-11-14 | 2010-05-20 | Genomatica, Inc. | Microorganisms for the production of methyl ethyl ketone and 2-butanol |
WO2016049932A1 (en) * | 2014-09-30 | 2016-04-07 | Bgi Shenzhen Co., Limited | Biomarkers for obesity related diseases |
BR112022010411A2 (en) * | 2019-11-27 | 2022-08-23 | Seres Therapeutics Inc | DESIGNED BACTERIAL COMPOSITIONS AND USES THEREOF |
-
2023
- 2023-12-15 WO PCT/US2023/084290 patent/WO2024130119A2/en unknown
Patent Citations (37)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US5589168A (en) | 1991-04-08 | 1996-12-31 | Unilever Patent Holdings B.V. | Probiotic |
US5989463A (en) | 1997-09-24 | 1999-11-23 | Alkermes Controlled Therapeutics, Inc. | Methods for fabricating polymer-based controlled release devices |
US6203797B1 (en) | 1998-01-06 | 2001-03-20 | Stephen C. Perry | Dietary supplement and method for use as a probiotic, for alleviating the symptons associated with irritable bowel syndrome |
US6835376B1 (en) | 1999-03-11 | 2004-12-28 | Nestec S.A. | Lactobacillus paracasei strain for preventing diarrhea caused by pathogenic bacteria |
US20150238545A1 (en) | 2000-07-25 | 2015-08-27 | Thomas Julius Borody | Probiotic recolonisation therapy |
US7914822B2 (en) | 2001-05-14 | 2011-03-29 | Prolacta Bioscience, Inc. | Method of producing nutritional products from human milk tissue and compositions thereof |
US7943315B2 (en) | 2005-09-20 | 2011-05-17 | Prolacta Bioscience, Inc. | Methods for testing milk |
US8278046B2 (en) | 2005-09-20 | 2012-10-02 | Prolacta Bioscience | Methods for testing milk |
US8628921B2 (en) | 2005-09-20 | 2014-01-14 | Prolacta Bioscience Inc. | Methods for testing milk |
US9149052B2 (en) | 2006-08-30 | 2015-10-06 | Prolacta Bioscience, Inc. | Methods of obtaining sterile milk and compositions thereof |
US20150359894A1 (en) | 2006-09-27 | 2015-12-17 | Little Calumet Holdings, Llc | Probiotic oral dosage forms |
US8545920B2 (en) | 2006-11-29 | 2013-10-01 | Prolacta Bioscience Inc. | Human milk compositions and methods of making and using same |
WO2010065652A1 (en) | 2008-12-02 | 2010-06-10 | Prolacta Bioscience, Inc. | Human milk permeate compositions and methods of making and using same |
US8927027B2 (en) | 2008-12-02 | 2015-01-06 | Prolacta Bioscience | Human milk permeate compositions and methods of making and using same |
WO2012097950A1 (en) | 2011-01-20 | 2012-07-26 | Jennewein Biotechnologie Gmbh | Novel fucosyltransferases and their applications |
WO2013044928A1 (en) | 2011-09-30 | 2013-04-04 | Glycom A/S | Synthesis of hmo core structures |
WO2013139344A1 (en) | 2012-03-20 | 2013-09-26 | Glycom A/S | Synthesis of the trisaccharide 3-o-fucosyllactose and intermediates thereof |
WO2013182206A1 (en) | 2012-06-08 | 2013-12-12 | Glycom A/S | Method for producing oligosaccharides and oligosaccharide glycosides by fermentation |
WO2013190530A1 (en) | 2012-06-22 | 2013-12-27 | Glycom A/S | Modified galactooligosaccharides |
WO2013190531A1 (en) | 2012-06-22 | 2013-12-27 | Glycom A/S | Glycosylated galactosyl disaccharides, methods for their production and their use in consumable products |
WO2014135167A1 (en) | 2013-03-08 | 2014-09-12 | Glycom A/S | Purification of oligosaccaharides by reversible derivatization |
WO2014167538A1 (en) | 2013-04-12 | 2014-10-16 | Glycom A/S | Synthesis of sialylated/fucosylated human milk oligosaccharides |
WO2014167537A1 (en) | 2013-04-12 | 2014-10-16 | Glycom A/S | Synthesis of sialylated/fucosylated oligosaccharides |
WO2015032413A1 (en) | 2013-09-06 | 2015-03-12 | Glycom A/S | Fermentative production of oligosaccharides |
WO2015036138A1 (en) | 2013-09-10 | 2015-03-19 | Jennewein Biotechnologie Gmbh | Production of oligosaccharides |
WO2015049331A1 (en) | 2013-10-04 | 2015-04-09 | Jennewein Biotechnologie Gmbh | Process for purification of a neutral human milk oligosaccharide using simulated moving bed chromatography |
WO2015106943A1 (en) | 2014-01-20 | 2015-07-23 | Jennewein Biotechnologie Gmbh | PROCESS FOR EFFICIENT PURIFICATION OF NEUTRAL HUMAN MILK OLIGOSACCHARIDES (HMOs) FROM MICROBIAL FERMENTATION |
WO2015150328A1 (en) | 2014-03-31 | 2015-10-08 | Jennewein Biotechnologie Gmbh | Total fermentation of oligosaccharides |
WO2015197082A1 (en) | 2014-06-27 | 2015-12-30 | Glycom A/S | Oligosaccharide production |
WO2017042382A1 (en) | 2015-09-12 | 2017-03-16 | Jennewein Biotechnologie Gmbh | Production of human milk oligosaccharides in microbial hosts with engineered import / export |
WO2017101958A1 (en) | 2015-12-18 | 2017-06-22 | Glycom A/S | Fermentative production of oligosaccharides |
WO2018053535A1 (en) | 2016-09-19 | 2018-03-22 | Prolacta Bioscience, Inc. | Purified human milk oligosaccharides compositions |
WO2018077892A1 (en) | 2016-10-29 | 2018-05-03 | Jennewein Biotechnologie Gmbh | Improved process for the production of fucosylated oligosaccharides |
WO2019008133A1 (en) | 2017-07-07 | 2019-01-10 | Jennewein Biotechnologie Gmbh | Fucosyltransferases and their use in producing fucosylated oligosaccharides |
WO2019043029A1 (en) | 2017-08-29 | 2019-03-07 | Jennewein Biotechnologie Gmbh | Process for purifying sialylated oligosaccharides |
WO2021061991A1 (en) | 2019-09-24 | 2021-04-01 | Prolacta Bioscience, Inc. | Compositions and methods for treatment of inflammatory and immune diseases |
WO2022036225A1 (en) | 2020-08-14 | 2022-02-17 | Prolacta Bioscience, Inc. | Human milk oligosaccharide compositions for use with bacteriotherapies |
Non-Patent Citations (11)
Title |
---|
EVANS ET AL., METABOLOMICS, vol. 4, 2014, pages 1000132 |
GOTOH ET AL., SCI REP, vol. 8, no. 1, 18 September 2018 (2018-09-18), pages 13958 |
GOTOH ET AL., SCI REP., vol. 8, no. 1, 18 September 2018 (2018-09-18), pages 13958 |
LAWLEY ET AL., PEERJ., vol. 5, 25 May 2017 (2017-05-25), pages e3375 |
LOCASCIO ET AL., APPL ENVIRON MICROBIOL, vol. 76, no. 22, 2010, pages 7373 - 81 |
MORROW ET AL., J. NUTRI., vol. 135, 2005, pages 1304 - 1307 |
O'SULLIVAN ET AL., J APPL MICROBIOL, vol. 111, no. 2, August 2011 (2011-08-01), pages 467 - 73 |
SCHELL ET AL., PNAS, vol. 99, no. 22, 2002, pages 14422 - 14427 |
SELA ET AL., PNAS, vol. 105, no. 48, 2008, pages 18964 - 18969 |
UNDERWOOD ET AL., PEDIATR RES, vol. 77, no. 0, 2015, pages 229 - 235 |
VYHLIDALOVA ET AL., IJMS, vol. 21, 2020, pages 2614 |
Also Published As
Publication number | Publication date |
---|---|
WO2024130119A3 (en) | 2024-07-18 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US20230068960A1 (en) | Compositions and methods for treatment of inflammatory and immune diseases | |
US8425930B2 (en) | Prebiotic oligosaccharides | |
AU2016102461A6 (en) | Nutritional compositions and infant formulas comprising Bifidobacterium animalis ssp. lactis and optionally a mix of oligosaccharides for inducing a gut microbiota close to the one of breast fed infants | |
CA2973223A1 (en) | Probiotic and prebiotic compositions, and methods of use thereof for modulation of the microbiome | |
US20240139222A1 (en) | Human milk oligosaccharide compositions for use with bacteriotherapies | |
US20230181653A1 (en) | Compositions for metabolic health | |
US20110158951A1 (en) | Use of blood group status ii | |
US20210069266A1 (en) | Methods for the isolation of microbes with enhanced persistance and compositions with such microbes | |
JP2024502629A (en) | Synbiotic treatment regimen | |
US20110158950A1 (en) | Use of blood group status i | |
WO2024130119A2 (en) | Synbiotic compositions for short chain fatty acid production | |
WO2024201041A2 (en) | Probiotic and postbiotic compositions, products and uses thereof | |
US20240066072A1 (en) | Clostrodioides difficile treatment | |
US20120315250A1 (en) | Probiotic bifidobacterial composition in accordance with secretor blood group status | |
CN116997341A (en) | Synbiotic treatment regimen |