WO2022171685A1 - Tnf-alpha blocking agent for preventing, suppressing and/or delaying the onset of type 1 diabetes - Google Patents
Tnf-alpha blocking agent for preventing, suppressing and/or delaying the onset of type 1 diabetes Download PDFInfo
- Publication number
- WO2022171685A1 WO2022171685A1 PCT/EP2022/053148 EP2022053148W WO2022171685A1 WO 2022171685 A1 WO2022171685 A1 WO 2022171685A1 EP 2022053148 W EP2022053148 W EP 2022053148W WO 2022171685 A1 WO2022171685 A1 WO 2022171685A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- diabetes
- mice
- sfb
- gut
- subject
- Prior art date
Links
- 206010067584 Type 1 diabetes mellitus Diseases 0.000 title claims abstract description 83
- 239000002981 blocking agent Substances 0.000 title claims abstract description 26
- 210000001035 gastrointestinal tract Anatomy 0.000 claims abstract description 45
- 208000027244 Dysbiosis Diseases 0.000 claims abstract description 13
- 230000007140 dysbiosis Effects 0.000 claims abstract description 13
- 241000227342 Candidatus Arthromitus sp. SFB-mouse Species 0.000 claims description 70
- 238000000034 method Methods 0.000 claims description 37
- 206010018429 Glucose tolerance impaired Diseases 0.000 claims description 13
- 230000002550 fecal effect Effects 0.000 claims description 11
- 108010008165 Etanercept Proteins 0.000 claims description 9
- 108020003175 receptors Proteins 0.000 claims description 7
- 229960000403 etanercept Drugs 0.000 claims description 6
- 230000000750 progressive effect Effects 0.000 claims description 5
- 208000024891 symptom Diseases 0.000 claims description 4
- 229940073621 enbrel Drugs 0.000 claims description 3
- 229960000598 infliximab Drugs 0.000 claims description 3
- 229940116176 remicade Drugs 0.000 claims description 3
- 229960002964 adalimumab Drugs 0.000 claims description 2
- 229960003115 certolizumab pegol Drugs 0.000 claims description 2
- 229940090100 cimzia Drugs 0.000 claims description 2
- 229960001743 golimumab Drugs 0.000 claims description 2
- 229940048921 humira Drugs 0.000 claims description 2
- 229940068638 simponi Drugs 0.000 claims description 2
- 108700012920 TNF Proteins 0.000 claims 8
- 230000004075 alteration Effects 0.000 abstract description 30
- 210000005205 gut mucosa Anatomy 0.000 abstract description 23
- 206010061218 Inflammation Diseases 0.000 abstract description 21
- 230000004054 inflammatory process Effects 0.000 abstract description 21
- 201000001421 hyperglycemia Diseases 0.000 abstract description 18
- 230000000968 intestinal effect Effects 0.000 abstract description 16
- NOESYZHRGYRDHS-UHFFFAOYSA-N insulin Chemical compound N1C(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(NC(=O)CN)C(C)CC)CSSCC(C(NC(CO)C(=O)NC(CC(C)C)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CCC(N)=O)C(=O)NC(CC(C)C)C(=O)NC(CCC(O)=O)C(=O)NC(CC(N)=O)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CSSCC(NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2C=CC(O)=CC=2)NC(=O)C(CC(C)C)NC(=O)C(C)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2NC=NC=2)NC(=O)C(CO)NC(=O)CNC2=O)C(=O)NCC(=O)NC(CCC(O)=O)C(=O)NC(CCCNC(N)=N)C(=O)NCC(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC(O)=CC=3)C(=O)NC(C(C)O)C(=O)N3C(CCC3)C(=O)NC(CCCCN)C(=O)NC(C)C(O)=O)C(=O)NC(CC(N)=O)C(O)=O)=O)NC(=O)C(C(C)CC)NC(=O)C(CO)NC(=O)C(C(C)O)NC(=O)C1CSSCC2NC(=O)C(CC(C)C)NC(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CC(N)=O)NC(=O)C(NC(=O)C(N)CC=1C=CC=CC=1)C(C)C)CC1=CN=CN1 NOESYZHRGYRDHS-UHFFFAOYSA-N 0.000 abstract description 15
- 102000004877 Insulin Human genes 0.000 abstract description 7
- 108090001061 Insulin Proteins 0.000 abstract description 7
- 229940125396 insulin Drugs 0.000 abstract description 7
- 230000003110 anti-inflammatory effect Effects 0.000 abstract description 6
- 239000000090 biomarker Substances 0.000 abstract description 6
- 230000013632 homeostatic process Effects 0.000 abstract description 5
- 230000001681 protective effect Effects 0.000 abstract description 5
- 230000006378 damage Effects 0.000 abstract description 4
- 208000023275 Autoimmune disease Diseases 0.000 abstract description 3
- 241000699670 Mus sp. Species 0.000 description 108
- 206010012601 diabetes mellitus Diseases 0.000 description 68
- 210000003405 ileum Anatomy 0.000 description 44
- 210000001744 T-lymphocyte Anatomy 0.000 description 40
- 108060008682 Tumor Necrosis Factor Proteins 0.000 description 39
- 102000000852 Tumor Necrosis Factor-alpha Human genes 0.000 description 37
- 210000004027 cell Anatomy 0.000 description 24
- 238000011161 development Methods 0.000 description 18
- 230000018109 developmental process Effects 0.000 description 18
- 108020004999 messenger RNA Proteins 0.000 description 17
- 230000007423 decrease Effects 0.000 description 16
- 230000003247 decreasing effect Effects 0.000 description 16
- 108020004414 DNA Proteins 0.000 description 14
- 238000004458 analytical method Methods 0.000 description 14
- 244000005709 gut microbiome Species 0.000 description 13
- 108010074109 interleukin-22 Proteins 0.000 description 13
- 238000004519 manufacturing process Methods 0.000 description 13
- 241000894006 Bacteria Species 0.000 description 12
- 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 12
- 102100030703 Interleukin-22 Human genes 0.000 description 12
- 239000008103 glucose Substances 0.000 description 12
- 230000004048 modification Effects 0.000 description 12
- 238000012986 modification Methods 0.000 description 12
- 239000000523 sample Substances 0.000 description 12
- 102000004127 Cytokines Human genes 0.000 description 11
- 108090000695 Cytokines Proteins 0.000 description 11
- 108050003558 Interleukin-17 Proteins 0.000 description 11
- 102000013691 Interleukin-17 Human genes 0.000 description 11
- 210000004400 mucous membrane Anatomy 0.000 description 11
- 238000011529 RT qPCR Methods 0.000 description 10
- 230000003345 hyperglycaemic effect Effects 0.000 description 10
- 238000012163 sequencing technique Methods 0.000 description 10
- ZSJLQEPLLKMAKR-GKHCUFPYSA-N streptozocin Chemical compound O=NN(C)C(=O)N[C@H]1[C@@H](O)O[C@H](CO)[C@@H](O)[C@@H]1O ZSJLQEPLLKMAKR-GKHCUFPYSA-N 0.000 description 10
- 241000736262 Microbiota Species 0.000 description 9
- 239000011324 bead Substances 0.000 description 9
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 9
- 210000003608 fece Anatomy 0.000 description 9
- 239000000203 mixture Substances 0.000 description 9
- 238000010172 mouse model Methods 0.000 description 9
- 230000009467 reduction Effects 0.000 description 9
- ZSJLQEPLLKMAKR-UHFFFAOYSA-N Streptozotocin Natural products O=NN(C)C(=O)NC1C(O)OC(CO)C(O)C1O ZSJLQEPLLKMAKR-UHFFFAOYSA-N 0.000 description 8
- 230000006698 induction Effects 0.000 description 8
- 239000013615 primer Substances 0.000 description 8
- 230000002829 reductive effect Effects 0.000 description 8
- 229960001052 streptozocin Drugs 0.000 description 8
- 238000011746 C57BL/6J (JAX™ mouse strain) Methods 0.000 description 7
- 101150010169 FUT2 gene Proteins 0.000 description 7
- 210000004369 blood Anatomy 0.000 description 7
- 239000008280 blood Substances 0.000 description 7
- 230000001904 diabetogenic effect Effects 0.000 description 7
- 201000010099 disease Diseases 0.000 description 7
- 210000002919 epithelial cell Anatomy 0.000 description 7
- 210000002865 immune cell Anatomy 0.000 description 7
- 108090000623 proteins and genes Proteins 0.000 description 7
- 230000035945 sensitivity Effects 0.000 description 7
- 210000001519 tissue Anatomy 0.000 description 7
- 108010041865 Ulex europaeus lectins Proteins 0.000 description 6
- 239000012634 fragment Substances 0.000 description 6
- 230000005694 interleukin-22 production Effects 0.000 description 6
- 150000007523 nucleic acids Chemical class 0.000 description 6
- 230000035699 permeability Effects 0.000 description 6
- 230000000770 proinflammatory effect Effects 0.000 description 6
- 235000018102 proteins Nutrition 0.000 description 6
- 102000004169 proteins and genes Human genes 0.000 description 6
- 238000012546 transfer Methods 0.000 description 6
- 108020004465 16S ribosomal RNA Proteins 0.000 description 5
- 229920002307 Dextran Polymers 0.000 description 5
- 230000004913 activation Effects 0.000 description 5
- 230000001363 autoimmune Effects 0.000 description 5
- 230000001580 bacterial effect Effects 0.000 description 5
- 238000006243 chemical reaction Methods 0.000 description 5
- 238000012350 deep sequencing Methods 0.000 description 5
- 210000004443 dendritic cell Anatomy 0.000 description 5
- 230000008595 infiltration Effects 0.000 description 5
- 238000001764 infiltration Methods 0.000 description 5
- 230000005651 interleukin-17A production Effects 0.000 description 5
- 210000004153 islets of langerhan Anatomy 0.000 description 5
- 102000005962 receptors Human genes 0.000 description 5
- 238000012360 testing method Methods 0.000 description 5
- 210000002237 B-cell of pancreatic islet Anatomy 0.000 description 4
- 101100447432 Danio rerio gapdh-2 gene Proteins 0.000 description 4
- 101150112014 Gapdh gene Proteins 0.000 description 4
- 101000852980 Homo sapiens Interleukin-23 subunit alpha Proteins 0.000 description 4
- 102100036705 Interleukin-23 subunit alpha Human genes 0.000 description 4
- 101150058357 Muc2 gene Proteins 0.000 description 4
- 241000699666 Mus <mouse, genus> Species 0.000 description 4
- 108091028043 Nucleic acid sequence Proteins 0.000 description 4
- 101150030755 Ocln gene Proteins 0.000 description 4
- 230000005784 autoimmunity Effects 0.000 description 4
- 150000001875 compounds Chemical class 0.000 description 4
- 230000001351 cycling effect Effects 0.000 description 4
- 230000007547 defect Effects 0.000 description 4
- 238000001514 detection method Methods 0.000 description 4
- 239000003814 drug Substances 0.000 description 4
- 230000000694 effects Effects 0.000 description 4
- 238000002474 experimental method Methods 0.000 description 4
- 238000000684 flow cytometry Methods 0.000 description 4
- 230000033581 fucosylation Effects 0.000 description 4
- 230000006870 function Effects 0.000 description 4
- 210000004185 liver Anatomy 0.000 description 4
- 238000007481 next generation sequencing Methods 0.000 description 4
- 239000002773 nucleotide Substances 0.000 description 4
- 125000003729 nucleotide group Chemical group 0.000 description 4
- 238000001712 DNA sequencing Methods 0.000 description 3
- 206010061818 Disease progression Diseases 0.000 description 3
- 102000004190 Enzymes Human genes 0.000 description 3
- 108090000790 Enzymes Proteins 0.000 description 3
- 102100027581 Forkhead box protein P3 Human genes 0.000 description 3
- 101000861452 Homo sapiens Forkhead box protein P3 Proteins 0.000 description 3
- 101000738771 Homo sapiens Receptor-type tyrosine-protein phosphatase C Proteins 0.000 description 3
- 102100037422 Receptor-type tyrosine-protein phosphatase C Human genes 0.000 description 3
- 239000004480 active ingredient Substances 0.000 description 3
- 238000010171 animal model Methods 0.000 description 3
- 238000000137 annealing Methods 0.000 description 3
- 238000003556 assay Methods 0.000 description 3
- 210000000227 basophil cell of anterior lobe of hypophysis Anatomy 0.000 description 3
- 230000037396 body weight Effects 0.000 description 3
- 230000003915 cell function Effects 0.000 description 3
- 230000001276 controlling effect Effects 0.000 description 3
- 230000005750 disease progression Effects 0.000 description 3
- 230000004064 dysfunction Effects 0.000 description 3
- 238000012165 high-throughput sequencing Methods 0.000 description 3
- 230000002757 inflammatory effect Effects 0.000 description 3
- 230000004609 intestinal homeostasis Effects 0.000 description 3
- 210000004877 mucosa Anatomy 0.000 description 3
- 108020004707 nucleic acids Proteins 0.000 description 3
- 102000039446 nucleic acids Human genes 0.000 description 3
- 210000000056 organ Anatomy 0.000 description 3
- 210000000496 pancreas Anatomy 0.000 description 3
- 230000008569 process Effects 0.000 description 3
- 230000001737 promoting effect Effects 0.000 description 3
- 210000003289 regulatory T cell Anatomy 0.000 description 3
- 230000005945 translocation Effects 0.000 description 3
- MZOFCQQQCNRIBI-VMXHOPILSA-N (3s)-4-[[(2s)-1-[[(2s)-1-[[(1s)-1-carboxy-2-hydroxyethyl]amino]-4-methyl-1-oxopentan-2-yl]amino]-5-(diaminomethylideneamino)-1-oxopentan-2-yl]amino]-3-[[2-[[(2s)-2,6-diaminohexanoyl]amino]acetyl]amino]-4-oxobutanoic acid Chemical compound OC[C@@H](C(O)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CCCN=C(N)N)NC(=O)[C@H](CC(O)=O)NC(=O)CNC(=O)[C@@H](N)CCCCN MZOFCQQQCNRIBI-VMXHOPILSA-N 0.000 description 2
- UEJJHQNACJXSKW-UHFFFAOYSA-N 2-(2,6-dioxopiperidin-3-yl)-1H-isoindole-1,3(2H)-dione Chemical compound O=C1C2=CC=CC=C2C(=O)N1C1CCC(=O)NC1=O UEJJHQNACJXSKW-UHFFFAOYSA-N 0.000 description 2
- MSYGAHOHLUJIKV-UHFFFAOYSA-N 3,5-dimethyl-1-(3-nitrophenyl)-1h-pyrazole-4-carboxylic acid ethyl ester Chemical compound CC1=C(C(=O)OCC)C(C)=NN1C1=CC=CC([N+]([O-])=O)=C1 MSYGAHOHLUJIKV-UHFFFAOYSA-N 0.000 description 2
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 2
- 241000605059 Bacteroidetes Species 0.000 description 2
- 102000004161 Claudin-4 Human genes 0.000 description 2
- 108090000601 Claudin-4 Proteins 0.000 description 2
- 241001464948 Coprococcus Species 0.000 description 2
- 208000011231 Crohn disease Diseases 0.000 description 2
- 238000007400 DNA extraction Methods 0.000 description 2
- 241001143779 Dorea Species 0.000 description 2
- 101150084967 EPCAM gene Proteins 0.000 description 2
- 102100040837 Galactoside alpha-(1,2)-fucosyltransferase 2 Human genes 0.000 description 2
- 102000008214 Glutamate decarboxylase Human genes 0.000 description 2
- 108091022930 Glutamate decarboxylase Proteins 0.000 description 2
- 238000012313 Kruskal-Wallis test Methods 0.000 description 2
- 241001465754 Metazoa Species 0.000 description 2
- 108010008705 Mucin-2 Proteins 0.000 description 2
- 102100034263 Mucin-2 Human genes 0.000 description 2
- 201000010927 Mucositis Diseases 0.000 description 2
- 102000003940 Occludin Human genes 0.000 description 2
- 108090000304 Occludin Proteins 0.000 description 2
- 241000266824 Oscillospira Species 0.000 description 2
- 206010067470 Rotavirus infection Diseases 0.000 description 2
- 108010006785 Taq Polymerase Proteins 0.000 description 2
- 108050001370 Tight junction protein ZO-1 Proteins 0.000 description 2
- 108060008683 Tumor Necrosis Factor Receptor Proteins 0.000 description 2
- 102000004248 Zinc Transporter 8 Human genes 0.000 description 2
- 108090000702 Zinc Transporter 8 Proteins 0.000 description 2
- 102000044820 Zonula Occludens-1 Human genes 0.000 description 2
- 230000002159 abnormal effect Effects 0.000 description 2
- 230000003321 amplification Effects 0.000 description 2
- 244000052616 bacterial pathogen Species 0.000 description 2
- 230000001684 chronic effect Effects 0.000 description 2
- 238000002405 diagnostic procedure Methods 0.000 description 2
- 230000003292 diminished effect Effects 0.000 description 2
- 208000035475 disorder Diseases 0.000 description 2
- 229940079593 drug Drugs 0.000 description 2
- 239000000839 emulsion Substances 0.000 description 2
- 108010079306 galactoside 2-alpha-L-fucosyltransferase Proteins 0.000 description 2
- 210000000987 immune system Anatomy 0.000 description 2
- 238000001727 in vivo Methods 0.000 description 2
- 239000003112 inhibitor Substances 0.000 description 2
- 210000004964 innate lymphoid cell Anatomy 0.000 description 2
- 210000002660 insulin-secreting cell Anatomy 0.000 description 2
- 210000004347 intestinal mucosa Anatomy 0.000 description 2
- 230000000670 limiting effect Effects 0.000 description 2
- 210000001165 lymph node Anatomy 0.000 description 2
- 238000012423 maintenance Methods 0.000 description 2
- 238000005259 measurement Methods 0.000 description 2
- 230000002503 metabolic effect Effects 0.000 description 2
- 238000003199 nucleic acid amplification method Methods 0.000 description 2
- 238000003305 oral gavage Methods 0.000 description 2
- 238000007410 oral glucose tolerance test Methods 0.000 description 2
- 230000001717 pathogenic effect Effects 0.000 description 2
- 238000002360 preparation method Methods 0.000 description 2
- 238000000513 principal component analysis Methods 0.000 description 2
- 238000003762 quantitative reverse transcription PCR Methods 0.000 description 2
- 210000000813 small intestine Anatomy 0.000 description 2
- 239000007787 solid Substances 0.000 description 2
- 230000002269 spontaneous effect Effects 0.000 description 2
- 238000007619 statistical method Methods 0.000 description 2
- 230000008685 targeting Effects 0.000 description 2
- 230000001225 therapeutic effect Effects 0.000 description 2
- 230000000699 topical effect Effects 0.000 description 2
- 102000003298 tumor necrosis factor receptor Human genes 0.000 description 2
- 238000011870 unpaired t-test Methods 0.000 description 2
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 1
- 241000731710 Allobaculum Species 0.000 description 1
- 208000035143 Bacterial infection Diseases 0.000 description 1
- 206010070545 Bacterial translocation Diseases 0.000 description 1
- 241000606125 Bacteroides Species 0.000 description 1
- 241000949031 Citrobacter rodentium Species 0.000 description 1
- 241000193403 Clostridium Species 0.000 description 1
- 241001443882 Coprobacillus Species 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
- 230000004544 DNA amplification Effects 0.000 description 1
- 238000007399 DNA isolation Methods 0.000 description 1
- 239000003155 DNA primer Substances 0.000 description 1
- 108010014303 DNA-directed DNA polymerase Proteins 0.000 description 1
- 102000016928 DNA-directed DNA polymerase Human genes 0.000 description 1
- 241000194033 Enterococcus Species 0.000 description 1
- 241000588698 Erwinia Species 0.000 description 1
- 108700039887 Essential Genes Proteins 0.000 description 1
- 241000192125 Firmicutes Species 0.000 description 1
- 208000034826 Genetic Predisposition to Disease Diseases 0.000 description 1
- 101001018097 Homo sapiens L-selectin Proteins 0.000 description 1
- 101000611183 Homo sapiens Tumor necrosis factor Proteins 0.000 description 1
- 101001135589 Homo sapiens Tyrosine-protein phosphatase non-receptor type 22 Proteins 0.000 description 1
- 101150003028 Hprt1 gene Proteins 0.000 description 1
- 102000018251 Hypoxanthine Phosphoribosyltransferase Human genes 0.000 description 1
- 108010091358 Hypoxanthine Phosphoribosyltransferase Proteins 0.000 description 1
- 108010091135 Immunoglobulin Fc Fragments Proteins 0.000 description 1
- 102000018071 Immunoglobulin Fc Fragments Human genes 0.000 description 1
- 102100023915 Insulin Human genes 0.000 description 1
- 102000003746 Insulin Receptor Human genes 0.000 description 1
- 108010001127 Insulin Receptor Proteins 0.000 description 1
- 229940094910 Insulin receptor antagonist Drugs 0.000 description 1
- 102100034343 Integrase Human genes 0.000 description 1
- 102000000589 Interleukin-1 Human genes 0.000 description 1
- 108010002352 Interleukin-1 Proteins 0.000 description 1
- 108010065637 Interleukin-23 Proteins 0.000 description 1
- 102000013264 Interleukin-23 Human genes 0.000 description 1
- 108090001005 Interleukin-6 Proteins 0.000 description 1
- 108090001007 Interleukin-8 Proteins 0.000 description 1
- 206010022678 Intestinal infections Diseases 0.000 description 1
- 241000588748 Klebsiella Species 0.000 description 1
- 102100033467 L-selectin Human genes 0.000 description 1
- OUYCCCASQSFEME-QMMMGPOBSA-N L-tyrosine Chemical compound OC(=O)[C@@H](N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-QMMMGPOBSA-N 0.000 description 1
- 241000186871 Lactobacillus murinus Species 0.000 description 1
- 108090001090 Lectins Proteins 0.000 description 1
- 102000004856 Lectins Human genes 0.000 description 1
- 229930195725 Mannitol Natural products 0.000 description 1
- 102000002274 Matrix Metalloproteinases Human genes 0.000 description 1
- 108010000684 Matrix Metalloproteinases Proteins 0.000 description 1
- 208000008589 Obesity Diseases 0.000 description 1
- 108091034117 Oligonucleotide Proteins 0.000 description 1
- 238000012408 PCR amplification Methods 0.000 description 1
- 239000012807 PCR reagent Substances 0.000 description 1
- 108010027220 PEGylated soluble tumor necrosis factor receptor I Proteins 0.000 description 1
- 241000160321 Parabacteroides Species 0.000 description 1
- 229940123932 Phosphodiesterase 4 inhibitor Drugs 0.000 description 1
- 241000425347 Phyla <beetle> Species 0.000 description 1
- 238000002123 RNA extraction Methods 0.000 description 1
- 108010092799 RNA-directed DNA polymerase Proteins 0.000 description 1
- 238000011530 RNeasy Mini Kit Methods 0.000 description 1
- 102000007056 Recombinant Fusion Proteins Human genes 0.000 description 1
- 108010008281 Recombinant Fusion Proteins Proteins 0.000 description 1
- 238000012311 Shapiro-Wilk normality test Methods 0.000 description 1
- BQCADISMDOOEFD-UHFFFAOYSA-N Silver Chemical compound [Ag] BQCADISMDOOEFD-UHFFFAOYSA-N 0.000 description 1
- 241000194017 Streptococcus Species 0.000 description 1
- 241000495426 Thiobacter Species 0.000 description 1
- 102000000591 Tight Junction Proteins Human genes 0.000 description 1
- 108010002321 Tight Junction Proteins Proteins 0.000 description 1
- 108091023040 Transcription factor Proteins 0.000 description 1
- 102000040945 Transcription factor Human genes 0.000 description 1
- 102100040247 Tumor necrosis factor Human genes 0.000 description 1
- 206010054094 Tumour necrosis Diseases 0.000 description 1
- 102000011017 Type 4 Cyclic Nucleotide Phosphodiesterases Human genes 0.000 description 1
- 108010037584 Type 4 Cyclic Nucleotide Phosphodiesterases Proteins 0.000 description 1
- 102100033138 Tyrosine-protein phosphatase non-receptor type 22 Human genes 0.000 description 1
- 208000036142 Viral infection Diseases 0.000 description 1
- 208000027418 Wounds and injury Diseases 0.000 description 1
- JLCPHMBAVCMARE-UHFFFAOYSA-N [3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-hydroxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methyl [5-(6-aminopurin-9-yl)-2-(hydroxymethyl)oxolan-3-yl] hydrogen phosphate Polymers Cc1cn(C2CC(OP(O)(=O)OCC3OC(CC3OP(O)(=O)OCC3OC(CC3O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c3nc(N)[nH]c4=O)C(COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3CO)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cc(C)c(=O)[nH]c3=O)n3cc(C)c(=O)[nH]c3=O)n3ccc(N)nc3=O)n3cc(C)c(=O)[nH]c3=O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)O2)c(=O)[nH]c1=O JLCPHMBAVCMARE-UHFFFAOYSA-N 0.000 description 1
- 210000005006 adaptive immune system Anatomy 0.000 description 1
- 241001148470 aerobic bacillus Species 0.000 description 1
- 238000012152 algorithmic method Methods 0.000 description 1
- 239000005557 antagonist Substances 0.000 description 1
- 239000003242 anti bacterial agent Substances 0.000 description 1
- 229940088710 antibiotic agent Drugs 0.000 description 1
- 239000000427 antigen Substances 0.000 description 1
- 108091007433 antigens Proteins 0.000 description 1
- 102000036639 antigens Human genes 0.000 description 1
- 238000013459 approach Methods 0.000 description 1
- 230000006470 autoimmune attack Effects 0.000 description 1
- 230000006472 autoimmune response Effects 0.000 description 1
- 208000022362 bacterial infectious disease Diseases 0.000 description 1
- 230000007375 bacterial translocation Effects 0.000 description 1
- 230000008901 benefit Effects 0.000 description 1
- 230000003115 biocidal effect Effects 0.000 description 1
- 239000003124 biologic agent Substances 0.000 description 1
- 239000012472 biological sample Substances 0.000 description 1
- 238000001574 biopsy Methods 0.000 description 1
- 238000009395 breeding Methods 0.000 description 1
- 230000001488 breeding effect Effects 0.000 description 1
- 235000019577 caloric intake Nutrition 0.000 description 1
- 239000002775 capsule Substances 0.000 description 1
- 230000001413 cellular effect Effects 0.000 description 1
- 239000003795 chemical substances by application Substances 0.000 description 1
- 231100000749 chronicity Toxicity 0.000 description 1
- 238000003776 cleavage reaction Methods 0.000 description 1
- 239000002299 complementary DNA Substances 0.000 description 1
- 238000010276 construction Methods 0.000 description 1
- 238000012937 correction Methods 0.000 description 1
- 230000002596 correlated effect Effects 0.000 description 1
- 230000000875 corresponding effect Effects 0.000 description 1
- 230000009260 cross reactivity Effects 0.000 description 1
- 231100000433 cytotoxic Toxicity 0.000 description 1
- 230000001472 cytotoxic effect Effects 0.000 description 1
- 238000007405 data analysis Methods 0.000 description 1
- 230000002950 deficient Effects 0.000 description 1
- 230000002939 deleterious effect Effects 0.000 description 1
- 238000004925 denaturation Methods 0.000 description 1
- 230000036425 denaturation Effects 0.000 description 1
- 238000003745 diagnosis Methods 0.000 description 1
- 238000010586 diagram Methods 0.000 description 1
- 235000005911 diet Nutrition 0.000 description 1
- 230000037213 diet Effects 0.000 description 1
- 238000009792 diffusion process Methods 0.000 description 1
- XPPKVPWEQAFLFU-UHFFFAOYSA-J diphosphate(4-) Chemical compound [O-]P([O-])(=O)OP([O-])([O-])=O XPPKVPWEQAFLFU-UHFFFAOYSA-J 0.000 description 1
- 235000011180 diphosphates Nutrition 0.000 description 1
- 238000009826 distribution Methods 0.000 description 1
- 239000012636 effector Substances 0.000 description 1
- 230000003511 endothelial effect Effects 0.000 description 1
- 230000029142 excretion Effects 0.000 description 1
- 238000013230 female C57BL/6J mice Methods 0.000 description 1
- 230000028564 filamentous growth Effects 0.000 description 1
- 239000000499 gel Substances 0.000 description 1
- 238000007446 glucose tolerance test Methods 0.000 description 1
- 230000036541 health Effects 0.000 description 1
- 210000002443 helper t lymphocyte Anatomy 0.000 description 1
- 210000003958 hematopoietic stem cell Anatomy 0.000 description 1
- 230000009675 homeostatic proliferation Effects 0.000 description 1
- 102000057041 human TNF Human genes 0.000 description 1
- 238000003384 imaging method Methods 0.000 description 1
- 239000012729 immediate-release (IR) formulation Substances 0.000 description 1
- 230000002519 immonomodulatory effect Effects 0.000 description 1
- 230000001900 immune effect Effects 0.000 description 1
- 230000016784 immunoglobulin production Effects 0.000 description 1
- 230000001771 impaired effect Effects 0.000 description 1
- 238000010348 incorporation Methods 0.000 description 1
- 208000015181 infectious disease Diseases 0.000 description 1
- 210000004969 inflammatory cell Anatomy 0.000 description 1
- 230000028709 inflammatory response Effects 0.000 description 1
- 230000000977 initiatory effect Effects 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
- 210000005007 innate immune system Anatomy 0.000 description 1
- 230000006362 insulin response pathway Effects 0.000 description 1
- 210000002490 intestinal epithelial cell Anatomy 0.000 description 1
- 230000003834 intracellular effect Effects 0.000 description 1
- 238000011835 investigation Methods 0.000 description 1
- 238000002955 isolation Methods 0.000 description 1
- 238000002372 labelling Methods 0.000 description 1
- 239000008101 lactose Substances 0.000 description 1
- 239000002523 lectin Substances 0.000 description 1
- 210000000265 leukocyte Anatomy 0.000 description 1
- 239000007788 liquid Substances 0.000 description 1
- 238000001325 log-rank test Methods 0.000 description 1
- 210000004698 lymphocyte Anatomy 0.000 description 1
- 210000002540 macrophage Anatomy 0.000 description 1
- 239000000594 mannitol Substances 0.000 description 1
- 235000010355 mannitol Nutrition 0.000 description 1
- 230000007246 mechanism Effects 0.000 description 1
- 244000005700 microbiome Species 0.000 description 1
- 230000005012 migration Effects 0.000 description 1
- 238000013508 migration Methods 0.000 description 1
- 238000012544 monitoring process Methods 0.000 description 1
- 210000003097 mucus Anatomy 0.000 description 1
- 230000003472 neutralizing effect Effects 0.000 description 1
- 229910052757 nitrogen Inorganic materials 0.000 description 1
- 238000001668 nucleic acid synthesis Methods 0.000 description 1
- 239000002777 nucleoside Substances 0.000 description 1
- -1 nucleoside triphosphates Chemical class 0.000 description 1
- 235000020824 obesity Nutrition 0.000 description 1
- 244000039328 opportunistic pathogen Species 0.000 description 1
- 108090000629 orphan nuclear receptors Proteins 0.000 description 1
- 102000004164 orphan nuclear receptors Human genes 0.000 description 1
- 230000008506 pathogenesis Effects 0.000 description 1
- 230000001575 pathological effect Effects 0.000 description 1
- 230000007170 pathology Effects 0.000 description 1
- 229950000867 pegsunercept Drugs 0.000 description 1
- 230000009984 peri-natal effect Effects 0.000 description 1
- 239000002587 phosphodiesterase IV inhibitor Substances 0.000 description 1
- 239000006187 pill Substances 0.000 description 1
- 229920001184 polypeptide Polymers 0.000 description 1
- 210000003240 portal vein Anatomy 0.000 description 1
- 230000003389 potentiating effect Effects 0.000 description 1
- 239000002243 precursor Substances 0.000 description 1
- 230000002028 premature Effects 0.000 description 1
- 230000009290 primary effect Effects 0.000 description 1
- 108090000765 processed proteins & peptides Proteins 0.000 description 1
- 102000004196 processed proteins & peptides Human genes 0.000 description 1
- 230000035755 proliferation Effects 0.000 description 1
- 125000006239 protecting group Chemical group 0.000 description 1
- 230000009979 protective mechanism Effects 0.000 description 1
- 238000003753 real-time PCR Methods 0.000 description 1
- 238000009256 replacement therapy Methods 0.000 description 1
- 108090000064 retinoic acid receptors Proteins 0.000 description 1
- 102000003702 retinoic acid receptors Human genes 0.000 description 1
- 230000002441 reversible effect Effects 0.000 description 1
- 206010039073 rheumatoid arthritis Diseases 0.000 description 1
- 230000007017 scission Effects 0.000 description 1
- 238000000926 separation method Methods 0.000 description 1
- 210000002966 serum Anatomy 0.000 description 1
- 229910052709 silver Inorganic materials 0.000 description 1
- 239000004332 silver Substances 0.000 description 1
- 239000000243 solution Substances 0.000 description 1
- 238000010561 standard procedure Methods 0.000 description 1
- 210000002784 stomach Anatomy 0.000 description 1
- 239000000829 suppository Substances 0.000 description 1
- 239000000725 suspension Substances 0.000 description 1
- 238000013268 sustained release Methods 0.000 description 1
- 239000012730 sustained-release form Substances 0.000 description 1
- 230000001839 systemic circulation Effects 0.000 description 1
- 230000009885 systemic effect Effects 0.000 description 1
- 239000003826 tablet Substances 0.000 description 1
- 229960003433 thalidomide Drugs 0.000 description 1
- 210000001578 tight junction Anatomy 0.000 description 1
- 238000011200 topical administration Methods 0.000 description 1
- 238000013518 transcription Methods 0.000 description 1
- 230000035897 transcription Effects 0.000 description 1
- 102000035160 transmembrane proteins Human genes 0.000 description 1
- 108091005703 transmembrane proteins Proteins 0.000 description 1
- 239000001226 triphosphate Substances 0.000 description 1
- 235000011178 triphosphate Nutrition 0.000 description 1
- 230000006433 tumor necrosis factor production Effects 0.000 description 1
- 208000001072 type 2 diabetes mellitus Diseases 0.000 description 1
- OUYCCCASQSFEME-UHFFFAOYSA-N tyrosine Natural products OC(=O)C(N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-UHFFFAOYSA-N 0.000 description 1
- 241001148471 unidentified anaerobic bacterium Species 0.000 description 1
- 230000003827 upregulation Effects 0.000 description 1
- 230000009385 viral infection Effects 0.000 description 1
- DGVVWUTYPXICAM-UHFFFAOYSA-N β‐Mercaptoethanol Chemical compound OCCS DGVVWUTYPXICAM-UHFFFAOYSA-N 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/395—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P5/00—Drugs for disorders of the endocrine system
- A61P5/48—Drugs for disorders of the endocrine system of the pancreatic hormones
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P5/00—Drugs for disorders of the endocrine system
- A61P5/48—Drugs for disorders of the endocrine system of the pancreatic hormones
- A61P5/50—Drugs for disorders of the endocrine system of the pancreatic hormones for increasing or potentiating the activity of insulin
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/705—Receptors; Cell surface antigens; Cell surface determinants
- C07K14/715—Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
- C07K14/7151—Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons for tumor necrosis factor [TNF], for lymphotoxin [LT]
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/24—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
- C07K16/241—Tumor Necrosis Factors
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/505—Medicinal preparations containing antigens or antibodies comprising antibodies
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/54—Medicinal preparations containing antigens or antibodies characterised by the route of administration
- A61K2039/541—Mucosal route
- A61K2039/542—Mucosal route oral/gastrointestinal
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2319/00—Fusion polypeptide
- C07K2319/30—Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2319/00—Fusion polypeptide
- C07K2319/32—Fusion polypeptide fusions with soluble part of a cell surface receptor, "decoy receptors"
Definitions
- the present invention is in the field of medicine.
- Type I diabetes is an autoimmune disease characterized by the destruction of pancreatic b-cells that produce insulin [1]
- the pathological activation of the immune system results in pancreatic inflammation, anti-islet antibody production, and islet infiltration by cytotoxic cells [2,3]
- Both innate and adaptive immune systems are inappropriately activated and involved in disease progression, in the pancreas and associated pancreatic lymph nodes (PLN) [2,3]
- PPN pancreatic lymph nodes
- T1D Another organ severely affected in T1D is the gastrointestinal tract.
- Several studies have described structural alterations of the gut mucosa, such as a reduction in crypt length and tight junction proteins expression in both T1D patients and animal models [4-9]
- Functional studies using mannitol -lactose clearance in patients with T1D and FITC-dextran in animal models of T1D demonstrated that gut integrity is weakened, allowing an increased crossing of bacterial components from the gut lumen to systemic circulation [10-13]
- Several factors are suspected to be involved in gut permeability rise in the context of diabetes, including alimentation, microbiota alteration, and enteric infection [8,14-17]
- the present invention is defined by the claims.
- the present invention relates to the use of TNFa blocking agent for preventing, suppressing and/or delaying the onset of type 1 diabetes.
- Type 1 diabetes is an autoimmune disease caused by the destruction of pancreatic b-cells producing insulin. Both T1D patients and animal models exhibit gut microbiota and mucosa alterations, although the exact cause for these remains poorly understood.
- the inventors investigated the production of key cytokines controlling gut integrity, the abundance of Segmented Filamentous Bacteria (SFB) involved in the production of these cytokines, and the respective role of auto-immune inflammation and hyperglycemia.
- the inventors used several mouse models of autoimmune T1D as well as mice rendered hyperglycemic without inflammation to study gut mucosa and microbiota dysbiosis.
- Anti-inflammatory treatment with TNF-blocking agent preserves gut homeostasis and protective commensal flora reducing T1D incidence. More particularly, the results reveal SFB as a potential biomarker of T1D progression in at-risk individuals and suggest that an anti-inflammatory treatment with TNF blocking agents might be of interest to reduce intestinal alteration associated with T1D onset.
- the present invention relates to methods for treating, preventing, suppressing and/or delaying the onset, or reducing the risk of developing type 1 diabetes, or the symptoms associated with, or related to, type 1 diabetes, in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a TNFa blocking agent.
- Type 1 diabetes has its general meaning in the art and refers to the condition in which a subject has, in the presence of autoimmunity towards the pancreatic beta cell as detected through pancreatic biopsy or imaging), a fasting (i.e., no caloric intake for 8 hours) blood glucose or serum glucose concentration greater than 125 mg/dL (6.94 mmol/L).
- HbAlc should be performed using a method certified by the National Glycohemoglobin Standardization Program (NGSP) and standardized or traceable to the Diabetes Control and Complications Trial (DCCT) reference assay.
- NGSP National Glycohemoglobin Standardization Program
- DCCT Diabetes Control and Complications Trial
- the blood sugar level of a diabetic will be in excess of 200 mg of glucose per dL (11.1 mmol/I) of plasma 2 hours after 75 g of glucose have been taken on an empty stomach, in the presence of autoimmunity towards the pancreatic beta cell.
- 75 g of glucose are administered orally to the patient being tested after a minimum of 8 hours, typically, 10-12 hours, of fasting and the blood sugar level is recorded immediately before taking the glucose and 1 and 2 hours after taking it.
- a genetic predisposition is present (e.g. HLA, INS VNTR and PTPN22), but this is not always the case.
- overt diabetes or “diabetes onset” is intended to mean a disease state in which the pancreatic islet cells are destroyed and which is manifested clinically by overt hyperglycemia (i.e., fasting blood glucose levels 3 250 mg/dL).
- overt hyperglycemia i.e., fasting blood glucose levels 3 250 mg/dL.
- time of progression from presymptomatic to clinical type 1 diabetes varies from weeks to decades.
- the subject is a child. In some embodiments, the subject is a premature human infant. In some embodiments the child is 3 to 5 years old. In some embodiments, the subject is an adult. In some embodiments, the adult is more than 18 years old. In some embodiments, the subject is a pre-diabetic subject.
- pre-diabetic subject is a subject at risk for the development of diabetes (e.g., genetically predisposed) at any stage in the disease process prior to overt diabetes or diabetes onset. In particular, the pre-diabetic subject exhibits one or more biomarkers that indicate that the subject is at risk of having type 1 diabetes and/or abnormal first phase insulin test (insulin response to an i.v.
- the autoantibodies currently used as biomarkers of type 1 diabetes in the clinic are mainly those targeting insulin, glutamic acid decarboxylase (GAD), a tyrosine phosphatase-like protein (islet antigen-2 [IA-2]) and zinc transporter-8 (ZnT8).
- GAD glutamic acid decarboxylase
- IA-2 islet antigen-2
- ZnT8 zinc transporter-8
- the development of autoantibodies against multiple beta cell antigens is recognized as a critical step in the disease pathogenesis and is associated with a significantly higher type 1 diabetes risk than the presence of just a single autoantibody.
- multiple beta cell autoantibody positive individuals are at high risk of type 1 diabetes regardless of family history, and children who develop two or more beta cell autoantibody types almost inevitably progress to clinically symptomatic diabetes.
- biomarkers are typically discussed in Mathieu, C., Lahesmaa, R., Bonifacio, E. et al. Immunological biomarkers for the development and progression of type 1 diabetes. Diabetologia 61, 2252 2258 (2016).
- the subject exhibits a dysbiosis.
- dysbiosis has its general meaning in the art and refers to an altered state of the microbiota occurring during diseases compared to the compositional and functional homeostasis in healthy individuals.
- Dysbiosis is currently viewed as a sign of an altered microbe-host crosstalk and is a primary target of strategies aiming at restoring or maintaining intestinal functional homeostasis.
- the dysbiosis includes progressive loss of Segmented Filamentous Bacteria B (SFB).
- SFB Segmented Filamentous Bacteria B
- the dysbiosis is progressive loss of Segmented Filamentous Bacteria B (SFB).
- SFB Segmented Filamentous Bacteria B
- the method of the present invention comprises the steps of measuring the abundance of SFB in a fecal sample obtained from the subject.
- the method herein described comprises the steps consisting of i) measuring the abundance of SFB in a fecal sample obtained from the subject ii) comparing the determined abundance with a predetermined reference value wherein differential between said determined abundance and said predetermined reference value indicates whether or not the subject is administered with a therapeutically effective amount of a TNFa blocking agent.
- a therapeutically effective amount of a TNFa blocking agent is administered to the subject when the determined abundance is lower than said predetermined reference value.
- the term “abundance” refers to the quantity or the concentration of said bacteria in a location/sample. In some embodiments, the abundance is absolute abundance. As used herein, the term “absolute abundance” refers to the concentration of said bacteria in a location/sample expressed for instance in number of UFC per mL or genome equivalent per mL. In some embodiments, the abundance is relative abundance. As used herein, the term “relative abundance” refers to the percent composition of a bacterium genus relative to the total number of bacteria genus in a given location/sample.
- SFB Segmented Filamentous Bacteria B
- SFB has its general meaning in the art and refers to a group of bacteria that are unique immune modulatory bacteria colonizing the small intestine of a variety of animals in a host-specific manner. SFB exhibit filamentous growth and attach to the host’s intestinal epithelium. The SFB was classified as Candidatus savagella.
- the abundance of SFB bacteria is measuring by any routine method well known in the art and typically by using molecular methods. In some embodiments, the abundance of SFB is measuring using 16S rRNA deep-sequencing. In some embodiments, the abundance of SFB is measuring using the abundance table generated by the next-generation sequencing of 16S rRNA genes of all bacteria within a given biological sample using qPCR technique. Nucleic acids may be extracted from a sample by routine techniques such as those described in Diagnostic Molecular Microbiology: Principles and Applications (Persing et al. (eds), 1993, American Society for Microbiology, Washington D.C.). U.S. Pat. Nos.
- PCR typically employs two oligonucleotide primers that bind to a selected target nucleic acid sequence.
- Primers useful in the present invention include oligonucleotides capable of acting as a point of initiation of nucleic acid synthesis within the target nucleic acid sequence.
- qPCR involves use of a thermostable polymerase.
- the polymerase is a Taq polymerase (i.e. Thermus aquaticus polymerase).
- the primers are combined with PCR reagents under reaction conditions that induce primer extension.
- the newly synthesized strands form a double-stranded molecule that can be used in the succeeding steps of the reaction.
- the steps of strand separation, annealing, and elongation can be repeated as often as needed to produce the desired quantity of amplification products corresponding to the target nucleic acid sequence molecule.
- the limiting factors in the reaction are the amounts of primers, thermostable enzyme, and nucleoside triphosphates present in the reaction.
- the cycling steps i.e., denaturation, annealing, and extension
- the 16S deep-sequencing technique is well-described in the state of the art for instance, Shendure and Ji. "Next-generation DNA sequencing", Nature Biotechnology, 26(10): 1135-1145 (2008)).
- the 16S deep-sequencing technique also known as “next-generation DNA sequencing” (“NGS”), “high-throughput sequencing”, “massively parallel sequencing” and “deep sequencing” refers to a method of sequencing a plurality of nucleic acids in parallel.
- the DNA to be sequenced is either fractionated and supplied with adaptors or segments of DNA can be PCR-amplified using primers containing the adaptors.
- the adaptors are nucleotide 25- mers required for binding to the DNA Capture Beads and for annealing the emulsion PCR Amplification Primers and the Sequencing Primer.
- the DNA fragments are made single stranded and are attached to DNA capture beads in a manner that allows only one DNA fragment to be attached to one bead.
- the DNA containing beads are emulsified in a water- in-oil mixture resulting in microreactors containing just one bead. Within the microreactor, the fragment is PCR-amplified, resulting in a copy number of several million per bead. After PCR, the emulsion is broken and the beads are loaded onto a pico titer plate.
- Each well of the pico- titer plate can contain only one bead. Sequencing enzymes are added to the wells and nucleotides are flowed across the wells in a fixed order. The incorporation of a nucleotide results in the release of a pyrophosphate, which catalyzes a reaction leading to a chemiluminescent signal. This signal is recorded by a CCD camera and a software is used to translate the signals into a DNA sequence.
- a pyrophosphate which catalyzes a reaction leading to a chemiluminescent signal.
- This signal is recorded by a CCD camera and a software is used to translate the signals into a DNA sequence.
- lllumina method (Bentley (2008))
- single stranded, adaptor-supplied fragments are attached to an optically transparent surface and subjected to "bridge amplification". This procedure results in several million clusters, each containing copies of a unique DNA fragment.
- the method of the present invention comprises the steps of i) determining the abundance of SFB in a fecal sample obtained from the subject, ii) comparing the abundance determined at step i) with a predetermined reference value and iii) administering to the subject a therapeutically effective amount of a TNFa blocking agent when the abundance is lower than the predetermined reference value.
- the predetermined reference value is a threshold value or a cut-off value.
- a “threshold value” or “cut-off value” can be determined experimentally, empirically, or theoretically.
- a threshold value can also be arbitrarily selected based upon the existing experimental and/or clinical conditions, as would be recognized by a person of ordinary skilled in the art. For example, retrospective measurement in properly banked historical subject samples may be used in establishing the predetermined reference value. The threshold value has to be determined in order to obtain the optimal sensitivity and specificity according to the function of the test and the benefit/risk balance (clinical consequences of false positive and false negative).
- a series of different cut-off values are set as continuous variables to calculate a series of sensitivity and specificity values. Then sensitivity is used as the vertical coordinate and specificity is used as the horizontal coordinate to draw a curve. The higher the area under the curve (AUC), the higher the accuracy of diagnosis.
- AUC area under the curve
- the point closest to the far upper left of the coordinate diagram is a critical point having both high sensitivity and high specificity values.
- the AUC value of the ROC curve is between 1.0 and 0.5. When AUC>0.5, the diagnostic result gets better and better as AUC approaches 1. When AUC is between 0.5 and 0.7, the accuracy is low. When AUC is between 0.7 and 0.9, the accuracy is moderate.
- the specific therapeutically effective dose level for any particular subject will depend upon a variety of factors including the disorder being treated and the severity of the disorder; activity of the specific compound employed; the specific composition employed, the age, body weight, general health, sex and diet of the subject; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidential with the specific polypeptide employed; and like factors well known in the medical arts.
- the daily dosage of the products may be varied over a wide range from 0.01 to 1,000 mg per adult per day.
- Anti-TNFa treatment prevents gut mucosa alterations, restores SFB levels, and prevents T1D onset.
- anti-TNFa reduced the frequency and absolute numbers of BDC2.5 T cells in ileum but not in PLN (data not shown). This was associated with a significant increase of FOXP3 + BDC2.5 T cell frequency in ileum and not in PLN (data not shown). Further analysis of autoreactive T cells showed a significant reduction of their IFN-g production in both ileum and PLN from mice treated with anti-TNFa compared to control mice whereas TNFa production by BDC2.5 T cells remained unchanged (data not shown). Finally, the decreased IFN-g production by BDC2.5 T cells in anti-TNFa treated mice was associated with a lower incidence of T1D and less severe insulitis (Figure 2F).
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Organic Chemistry (AREA)
- Medicinal Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Diabetes (AREA)
- Veterinary Medicine (AREA)
- Engineering & Computer Science (AREA)
- Pharmacology & Pharmacy (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Animal Behavior & Ethology (AREA)
- Immunology (AREA)
- Public Health (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Biophysics (AREA)
- Genetics & Genomics (AREA)
- Molecular Biology (AREA)
- Biochemistry (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- General Chemical & Material Sciences (AREA)
- Endocrinology (AREA)
- Microbiology (AREA)
- Mycology (AREA)
- Epidemiology (AREA)
- Cell Biology (AREA)
- Toxicology (AREA)
- Zoology (AREA)
- Gastroenterology & Hepatology (AREA)
- Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
Abstract
Type 1 diabetes (T1D) is an autoimmune disease caused by the destruction of pancreatic cells producing insulin. The present studies demonstrate that gut mucosa alterations and dysbiosis in T1D are primarily linked to inflammation rather than hyperglycemia. Anti- inflammatory treatment with TNF-blocking agent preserves gut homeostasis and protective commensal flora reducing T1D incidence. More particularly, the results reveal SFB as a potential biomarker of T1D progression in at-risk individuals and suggest that an anti- inflammatory treatment with TNF blocking agents might be of interest to reduce intestinal alteration associated with T1D onset.
Description
TNF-ALPHA BLOCKING AGENT FOR PREVENTING, SUPPRESSING AND/OR DELAYING THE ONSET OF TYPE 1 DIABETES
FIELD OF THE INVENTION:
The present invention is in the field of medicine.
BACKGROUND OF THE INVENTION:
Type I diabetes (T1D) is an autoimmune disease characterized by the destruction of pancreatic b-cells that produce insulin [1] The pathological activation of the immune system results in pancreatic inflammation, anti-islet antibody production, and islet infiltration by cytotoxic cells [2,3] Both innate and adaptive immune systems are inappropriately activated and involved in disease progression, in the pancreas and associated pancreatic lymph nodes (PLN) [2,3] The resulting loss of insulin production leads to chronic hyperglycemia and forces patients to rely on an insulin replacement therapy for life [1]
Another organ severely affected in T1D is the gastrointestinal tract. Several studies have described structural alterations of the gut mucosa, such as a reduction in crypt length and tight junction proteins expression in both T1D patients and animal models [4-9] Functional studies using mannitol -lactose clearance in patients with T1D and FITC-dextran in animal models of T1D demonstrated that gut integrity is weakened, allowing an increased crossing of bacterial components from the gut lumen to systemic circulation [10-13] Several factors are suspected to be involved in gut permeability rise in the context of diabetes, including alimentation, microbiota alteration, and enteric infection [8,14-17]
In parallel, the gut immune system, which resides in close contact with microbiota, displays pro-inflammatory features upon T1D development [18-20] The incidence of chronic intestinal inflammation is higher in T1D patients compared to the general population [21,22] Infiltration of pro-inflammatory cells is increased in the gut mucosa of T1D patients compared to healthy controls while Forkhead box P3+ (FOXP3+) regulatory T cells frequencies are reduced [18,19,23] We previously reported that Mucosal-Associated Invariant T (MAIT) cells exert a key role in maintaining gut integrity that is lost during T1D progression in Non-obese diabetic (NOD) mice [24] Indeed, MAIT-deficient NOD mice develop an exacerbated disease associated with increased gut alterations and bacteria DNA translocation into PLN [24] During disease progression in NOD females, MAIT cells lose IL-17A and IL-22 production, two critical cytokines required to maintain gut integrity [25-27]
SUMMARY OF THE INVENTION:
The present invention is defined by the claims. In particular, the present invention relates to the use of TNFa blocking agent for preventing, suppressing and/or delaying the onset of type 1 diabetes.
PET ATT /ED DESCRIPTION OF THE INVENTION:
Type 1 diabetes (T1D) is an autoimmune disease caused by the destruction of pancreatic b-cells producing insulin. Both T1D patients and animal models exhibit gut microbiota and mucosa alterations, although the exact cause for these remains poorly understood. The inventors investigated the production of key cytokines controlling gut integrity, the abundance of Segmented Filamentous Bacteria (SFB) involved in the production of these cytokines, and the respective role of auto-immune inflammation and hyperglycemia. The inventors used several mouse models of autoimmune T1D as well as mice rendered hyperglycemic without inflammation to study gut mucosa and microbiota dysbiosis. They analyzed cytokine expression by immune cells, epithelial cell function, SFB abundance and microbiota composition by 16S sequencing. They also assessed the role of anti-TNFa on gut mucosa inflammation and T1D onset. The inventors show in models of autoimmune T1D a conserved loss of IL-17A, IL-22, and IL-23A in gut mucosa. Intestinal epithelial cell function was altered and gut integrity was impaired. These defects were associated with dysbiosis including progressive loss of SFB. Transfer of diabetogenic T-cells recapitulated these gut alterations, whereas induction of hyperglycemia with no inflammation failed to do so. Moreover, anti inflammatory treatment restored gut mucosa and immune cell function and dampened diabetes incidence. The results demonstrate that gut mucosa alterations and dysbiosis in T1D are primarily linked to inflammation rather than hyperglycemia. Anti-inflammatory treatment with TNF-blocking agent preserves gut homeostasis and protective commensal flora reducing T1D incidence. More particularly, the results reveal SFB as a potential biomarker of T1D progression in at-risk individuals and suggest that an anti-inflammatory treatment with TNF blocking agents might be of interest to reduce intestinal alteration associated with T1D onset.
The present invention relates to methods for treating, preventing, suppressing and/or delaying the onset, or reducing the risk of developing type 1 diabetes, or the symptoms associated with,
or related to, type 1 diabetes, in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a TNFa blocking agent.
As used herein, the term “Type 1 diabetes” has its general meaning in the art and refers to the condition in which a subject has, in the presence of autoimmunity towards the pancreatic beta cell as detected through pancreatic biopsy or imaging), a fasting (i.e., no caloric intake for 8 hours) blood glucose or serum glucose concentration greater than 125 mg/dL (6.94 mmol/L). Type 1 diabetes is also defined as the condition in which a subject has, in the presence of autoimmunity towards the pancreatic beta-cell, HbAlc equal to, or greater than 6.5%, a two hour plasma glucose equal to, or greater than 200 mg/dL (11.1 mmol/L) during an oral glucose tolerance test (OGTT) or a random glucose equal to, or greater than 200 mg/dL (11.1 mmol/L) in conjunction with classic symptoms of hyperglycaemia or hyperglycaemic crisis. The measurement of blood glucose values is a standard procedure in routine medical analysis. The assessment of HbAlc should be performed using a method certified by the National Glycohemoglobin Standardization Program (NGSP) and standardized or traceable to the Diabetes Control and Complications Trial (DCCT) reference assay. If an OGTT is carried out, the blood sugar level of a diabetic will be in excess of 200 mg of glucose per dL (11.1 mmol/I) of plasma 2 hours after 75 g of glucose have been taken on an empty stomach, in the presence of autoimmunity towards the pancreatic beta cell. In a glucose tolerance test 75 g of glucose are administered orally to the patient being tested after a minimum of 8 hours, typically, 10-12 hours, of fasting and the blood sugar level is recorded immediately before taking the glucose and 1 and 2 hours after taking it. Typically a genetic predisposition is present (e.g. HLA, INS VNTR and PTPN22), but this is not always the case.
As used herein, the term “overt diabetes” or “diabetes onset” is intended to mean a disease state in which the pancreatic islet cells are destroyed and which is manifested clinically by overt hyperglycemia (i.e., fasting blood glucose levels ³ 250 mg/dL). Typically, the time of progression from presymptomatic to clinical type 1 diabetes varies from weeks to decades.
In some embodiments, the subject is a child. In some embodiments, the subject is a premature human infant. In some embodiments the child is 3 to 5 years old. In some embodiments, the subject is an adult. In some embodiments, the adult is more than 18 years old.
In some embodiments, the subject is a pre-diabetic subject. As used herein the term “prediabetic subject” is a subject at risk for the development of diabetes (e.g., genetically predisposed) at any stage in the disease process prior to overt diabetes or diabetes onset. In particular, the pre-diabetic subject exhibits one or more biomarkers that indicate that the subject is at risk of having type 1 diabetes and/or abnormal first phase insulin test (insulin response to an i.v. glucose bolus). The autoantibodies currently used as biomarkers of type 1 diabetes in the clinic are mainly those targeting insulin, glutamic acid decarboxylase (GAD), a tyrosine phosphatase-like protein (islet antigen-2 [IA-2]) and zinc transporter-8 (ZnT8). The development of autoantibodies against multiple beta cell antigens is recognized as a critical step in the disease pathogenesis and is associated with a significantly higher type 1 diabetes risk than the presence of just a single autoantibody. Moreover, multiple beta cell autoantibody positive individuals are at high risk of type 1 diabetes regardless of family history, and children who develop two or more beta cell autoantibody types almost inevitably progress to clinically symptomatic diabetes. Typically, biomarkers are typically discussed in Mathieu, C., Lahesmaa, R., Bonifacio, E. et al. Immunological biomarkers for the development and progression of type 1 diabetes. Diabetologia 61, 2252 2258 (2018).
In some embodiments, the subject exhibits a dysbiosis. As used herein, the term “dysbiosis” has its general meaning in the art and refers to an altered state of the microbiota occurring during diseases compared to the compositional and functional homeostasis in healthy individuals. Dysbiosis is currently viewed as a sign of an altered microbe-host crosstalk and is a primary target of strategies aiming at restoring or maintaining intestinal functional homeostasis.
In some embodiments, the dysbiosis includes progressive loss of Segmented Filamentous Bacteria B (SFB).
In some embodiments, the dysbiosis is progressive loss of Segmented Filamentous Bacteria B (SFB).
Thus, in some embodiments, the method of the present invention comprises the steps of measuring the abundance of SFB in a fecal sample obtained from the subject. In some embodiments, the method herein described comprises the steps consisting of i) measuring the abundance of SFB in a fecal sample obtained from the subject ii) comparing the determined abundance with a predetermined reference value wherein differential between said determined
abundance and said predetermined reference value indicates whether or not the subject is administered with a therapeutically effective amount of a TNFa blocking agent.
In some embodiment, a therapeutically effective amount of a TNFa blocking agent is administered to the subject when the determined abundance is lower than said predetermined reference value.
As used herein, the term “abundance” refers to the quantity or the concentration of said bacteria in a location/sample. In some embodiments, the abundance is absolute abundance. As used herein, the term “absolute abundance” refers to the concentration of said bacteria in a location/sample expressed for instance in number of UFC per mL or genome equivalent per mL. In some embodiments, the abundance is relative abundance. As used herein, the term “relative abundance” refers to the percent composition of a bacterium genus relative to the total number of bacteria genus in a given location/sample.
As used herein, the term “Segmented Filamentous Bacteria B” or “SFB” has its general meaning in the art and refers to a group of bacteria that are unique immune modulatory bacteria colonizing the small intestine of a variety of animals in a host-specific manner. SFB exhibit filamentous growth and attach to the host’s intestinal epithelium. The SFB was classified as Candidatus savagella.
In some embodiments, the abundance of SFB bacteria is measuring by any routine method well known in the art and typically by using molecular methods. In some embodiments, the abundance of SFB is measuring using 16S rRNA deep-sequencing. In some embodiments, the abundance of SFB is measuring using the abundance table generated by the next-generation sequencing of 16S rRNA genes of all bacteria within a given biological sample using qPCR technique. Nucleic acids may be extracted from a sample by routine techniques such as those described in Diagnostic Molecular Microbiology: Principles and Applications (Persing et al. (eds), 1993, American Society for Microbiology, Washington D.C.). U.S. Pat. Nos. 4,683,202, 4,683,195, 4,800,159, and 4,965,188 disclose conventional PCR techniques. PCR typically employs two oligonucleotide primers that bind to a selected target nucleic acid sequence. Primers useful in the present invention include oligonucleotides capable of acting as a point of initiation of nucleic acid synthesis within the target nucleic acid sequence. qPCR involves use of a thermostable polymerase. Typically, the polymerase is a Taq polymerase (i.e. Thermus aquaticus polymerase). The primers are combined with PCR reagents under reaction conditions
that induce primer extension. The newly synthesized strands form a double-stranded molecule that can be used in the succeeding steps of the reaction. The steps of strand separation, annealing, and elongation can be repeated as often as needed to produce the desired quantity of amplification products corresponding to the target nucleic acid sequence molecule. The limiting factors in the reaction are the amounts of primers, thermostable enzyme, and nucleoside triphosphates present in the reaction. The cycling steps (i.e., denaturation, annealing, and extension) are preferably repeated at least once. For use in detection, the number of cycling steps will depend, e.g., on the nature of the sample. If the sample is a complex mixture of nucleic acids, more cycling steps will be required to amplify the target sequence sufficient for detection. Generally, the cycling steps are repeated at least about 20 times, but may be repeated as many as 40, 60, or even 100 times. The 16S deep-sequencing technique is well-described in the state of the art for instance, Shendure and Ji. "Next-generation DNA sequencing", Nature Biotechnology, 26(10): 1135-1145 (2008)). The 16S deep-sequencing technique also known as “next-generation DNA sequencing” ("NGS"), "high-throughput sequencing", "massively parallel sequencing” and "deep sequencing" refers to a method of sequencing a plurality of nucleic acids in parallel. See e.g., Bentley et al, Nature 2008, 456:53-59. The leading commercially available platforms produced by Roche/454 (Margulies et al, 2005a), Illumina/Solexa (Bentley et al, 2008), Life/APG (SOLiD) (McKernan et al, 2009) and Pacific Biosciences (Eid et al, 2009) may be used for deep sequencing. For example, in the 454 method, the DNA to be sequenced is either fractionated and supplied with adaptors or segments of DNA can be PCR-amplified using primers containing the adaptors. The adaptors are nucleotide 25- mers required for binding to the DNA Capture Beads and for annealing the emulsion PCR Amplification Primers and the Sequencing Primer. The DNA fragments are made single stranded and are attached to DNA capture beads in a manner that allows only one DNA fragment to be attached to one bead. Next, the DNA containing beads are emulsified in a water- in-oil mixture resulting in microreactors containing just one bead. Within the microreactor, the fragment is PCR-amplified, resulting in a copy number of several million per bead. After PCR, the emulsion is broken and the beads are loaded onto a pico titer plate. Each well of the pico- titer plate can contain only one bead. Sequencing enzymes are added to the wells and nucleotides are flowed across the wells in a fixed order. The incorporation of a nucleotide results in the release of a pyrophosphate, which catalyzes a reaction leading to a chemiluminescent signal. This signal is recorded by a CCD camera and a software is used to translate the signals into a DNA sequence. In the lllumina method (Bentley (2008)), single stranded, adaptor-supplied fragments are attached to an optically transparent surface and
subjected to "bridge amplification". This procedure results in several million clusters, each containing copies of a unique DNA fragment. DNA polymerase, primers and four labeled reversible terminator nucleotides are added and the surface is imaged by laser fluorescence to determine the location and nature of the labels. Protecting groups are then removed and the process is repeated for several cycles. The SOLiD process (Shendure (2005)) is similar to 454 sequencing, DNA fragments are amplified on the surface of beads. Sequencing involves cycles of ligation and detection of labeled probes. Several other techniques for high-throughput sequencing are currently being developed. Examples of such are The Helicos system (Harris (2008)), Complete Genomics (Drmanac (2010)) and Pacific Biosciences (Lundquist (2008)). As this is an extremely rapidly developing technical field, the applicability to the present invention of high throughput sequencing methods will be obvious to a person skilled in the art.
In some embodiments, the method of the present invention comprises the steps of i) determining the abundance of SFB in a fecal sample obtained from the subject, ii) comparing the abundance determined at step i) with a predetermined reference value and iii) administering to the subject a therapeutically effective amount of a TNFa blocking agent when the abundance is lower than the predetermined reference value.
In other words, the invention refers to a methods for treating, preventing, suppressing and/or delaying the onset, or reducing the risk of developing type 1 diabetes, or the symptoms associated with, or related to, type 1 diabetes, in a subject in need thereof comprising the steps of i) measuring the abundance of SFB in a fecal sample obtained from the subject, ii) comparing the abundance determined at step i) with a predetermined reference value and iii) administering to the subject a therapeutically effective amount of a TNFa blocking agent when the abundance determined at step i) is lower than the predetermined reference value.
Typically, the predetermined reference value is a threshold value or a cut-off value. Typically, a "threshold value" or "cut-off value" can be determined experimentally, empirically, or theoretically. A threshold value can also be arbitrarily selected based upon the existing experimental and/or clinical conditions, as would be recognized by a person of ordinary skilled in the art. For example, retrospective measurement in properly banked historical subject samples may be used in establishing the predetermined reference value. The threshold value has to be determined in order to obtain the optimal sensitivity and specificity according to the function of the test and the benefit/risk balance (clinical consequences of false positive and false negative). Typically, the optimal sensitivity and specificity (and so the threshold value)
can be determined using a Receiver Operating Characteristic (ROC) curve based on experimental data. For example, after determining the abundance of SFB in a group of reference, one can use algorithmic analysis for the statistic treatment of the abundances determined in samples to be tested, and thus obtain a classification standard having significance for sample classification. The full name of ROC curve is receiver operator characteristic curve, which is also known as receiver operation characteristic curve. It is mainly used for clinical biochemical diagnostic tests. ROC curve is a comprehensive indicator that reflects the continuous variables of true positive rate (sensitivity) and false positive rate (1 -specificity). It reveals the relationship between sensitivity and specificity with the image composition method. A series of different cut-off values (thresholds or critical values, boundary values between normal and abnormal results of diagnostic test) are set as continuous variables to calculate a series of sensitivity and specificity values. Then sensitivity is used as the vertical coordinate and specificity is used as the horizontal coordinate to draw a curve. The higher the area under the curve (AUC), the higher the accuracy of diagnosis. On the ROC curve, the point closest to the far upper left of the coordinate diagram is a critical point having both high sensitivity and high specificity values. The AUC value of the ROC curve is between 1.0 and 0.5. When AUC>0.5, the diagnostic result gets better and better as AUC approaches 1. When AUC is between 0.5 and 0.7, the accuracy is low. When AUC is between 0.7 and 0.9, the accuracy is moderate. When AUC is higher than 0.9, the accuracy is high. This algorithmic method is preferably done with a computer. Existing software or systems in the art may be used for the drawing of the ROC curve, such as: MedCalc 9.2.0.1 medical statistical software, SPSS 9.0, ROCPOWER.SAS, DESIGNROC.FOR, MULTIREADER POWER S AS, CREATE-ROC.SAS, GB STAT VIO.O (Dynamic Microsystems, Inc. Silver Spring, Md., USA), etc.
As used herein, the term "TNFa" or “TNF-alpha” denotes the tumor necrosis factor - alpha. The human TNF-alpha is a human cytokine encoded by the TNF-alpha gene. TNF-alpha, a naturally occurring cytokine, plays a central role in the inflammatory response and in immune injury. It is formed by the cleavage of a precursor transmembrane protein, forming soluble molecules, which aggregate to form trimolecular complexes. These complexes then bind to receptors found on a variety of cells. Binding produces an array of pro-inflammatory effects, including release of other pro-inflammatory cytokines, including IL-6, IL-8, and IL-1; release of matrix metalloproteinases; and up regulation of the expression of endothelial adhesion molecules, further amplifying the inflammatory and immune cascade by attracting leukocytes into extravascular tissues.
As used herein, the term “TNFa blocking agent" or "TBA", it is herein meant a biological agent which is capable of neutralizing the effects of TNFa. Said agent is a preferentially a protein such as a soluble TNFa receptor, e.g. Pegsunercept, or an antibody. In some embodiments, the TBA is a monoclonal antibody having specificity for TNFa or for TNFa receptor. In some embodiments, the TBA is selected in the group consisting of Etanercept (Enbrel®), Infliximab (Remicade®), Adalimumab (Humira®), Certolizumab pegol (Cimzia®), and golimumab (Simponi®). Recombinant TNF-receptor based proteins have also been developed (e.g. etanercept, a recombinant fusion protein consisting of two extracellular parts of soluble TNFa receptor 2 (p75) joined by the Fc fragment of a human IgGl molecule). A pegylated soluble TNF type 1 receptor can also be used as a TNF blocking agent. Additionally, thalidomide has been demonstrated to be a potent inhibitor of TNF production. TNFa blocking agents thus further include phosphodiesterase 4 (IV) inhibitor thalidomide analogues and other phosphodiesterase IV inhibitors. As used herein, the term “etanercept” or “ETA” denotes the tumor necrosis factor - alpha (TNFa) antagonist used for the treatment of rheumatoid arthritis. The term “etanercept” (ETA, ETN, Enbrel) is a recombinant TNF-receptor IgG-Fc-fusion protein composed of the p75 TNF receptor genetically fused to the Fc domain of IgGl. Etanercept neutralizes the proinflammatory cytokine tumor necrosis factor-a (TNFa) and lymphotoxin-a (Batycka-Baran et ah, 2012).
As demonstrated in EXAMPLE, the TNFa blocking agent would be suitable for restoring intestinal homeostasis in pre-diabetic subjects and more particular would be suitable for reducing gut inflammation. Even more particularly, the TNFa blocking agent would also be suitable for reducing the promotion of the anti-islet T cell and thus would be suitable for reducing to pancreatic infiltration. Reducing inflammation the TNFa blocking agent would also be suitable for correcting T ID-associated SFB dysbiosis and mucosal dysfunctions.
As used herein, the term "therapeutically effective amount" as used herein refers to an amount or dose of the TNFa blocking agent that is sufficient to prevent the onset of diabetes. The "therapeutically effective amount" is determined using procedures routinely employed by those of skill in the art such that an "improved therapeutic outcome" results. It will be understood, however, that the total daily usage of the compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment. The specific therapeutically effective dose level for any particular subject will depend upon a variety of factors including the disorder being treated and the severity of the disorder; activity of the
specific compound employed; the specific composition employed, the age, body weight, general health, sex and diet of the subject; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidential with the specific polypeptide employed; and like factors well known in the medical arts. For example, it is well within the skill of the art to start doses of the compound at levels lower than those required to achieve the desired therapeutic effect and to gradually increase the dosage until the desired effect is achieved. However, the daily dosage of the products may be varied over a wide range from 0.01 to 1,000 mg per adult per day. Typically, the compositions contain 0.01, 0.05, 0.1, 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 25.0, 50.0, 100, 250 and 500 mg of the active ingredient for the symptomatic adjustment of the dosage to the subject to be treated. A medicament typically contains from about 0.01 mg to about 500 mg of the active ingredient, preferably from 1 mg to about 100 mg of the active ingredient. An effective amount of the drug is ordinarily supplied at a dosage level from 0.0002 mg/kg to about 20 mg/kg of body weight per day, especially from about 0.001 mg/kg to 7 mg/kg of body weight per day.
In some embodiments, the TNFa blocking agent is locally administered to the patient (i.e. in the subject’s gut). Typically the present invention thus comprises preparations of TNFa blocking agent which are suitable for topical delivery. Such topical preparations may be prepared in the form of aqueous-based solutions, gels, or patches for topical administration, and suspensions for administration, as pills, tablets, capsules or suppositories for immediate or sustained release to the gastrointestinal tract.
The invention will be further illustrated by the following figures and examples. However, these examples and figures should not be interpreted in any way as limiting the scope of the present invention.
FIGURES:
Figure 1. T1D development in NOD mice is associated with decreased SFB level. LP cells from small intestine (ileum) of prediabetic (8-10 weeks) and diabetic females NOD mice were analyzed. (A, B) Ileal tissues and fecal content were collected and extracted DNA was analyzed by qPCR for the presence of intestinal SFB (n=15 mice per group). (C) SFB level was assessed by qPCR in feces from non-diabetic NOD mice from 4 to 13 weeks of age (n=4-12 mice per
group). (D) SFB level was quantified by qPCR and followed from 7 weeks to 13 weeks of age in feces from female C57BL/6J mice, female NOD mice that became, or not, diabetic at 13 weeks of age (n=4-15 mice per group). Significant differences between C57BL/6J and diabetic NOD mice are represented by a $ symbol. (E) Spearman’s correlation between SFB level and Fut2 relative expression in ileal tissues (n=16 diabetic mice). (F) Intestinal gut permeability measured by in vivo FITC-dextran assay in NOD mice at 6, 10, 12 weeks of age and at diabetes onset (n=3-10 mice per group). Data are representative from at least two independent experiments and horizontal lines indicate the median, each data point represents an individual mouse. */$P<0.05, **P<0.01, ***P<0.001 and ****p<0.0001 (two-tailed Mann-Whitney test).
Figure 2. Anti-TNFa treatment prevents gut mucosa alteration and diabetes onset.
Recipient CD45.1+ Ir' ac NOD mice were transferred with 0.5 x 106 CD45.2+ CD4+ CD62L+ BDC2.5 T cells and treated with anti-TNFa (lOmg/kg) or human control IgGl antibodies on day 0, 3, and 6. (A) RT-qPCR analysis of mRNA encoding for 1117a, 1122, 1122a and Foxp3 from LP cells (n=9-ll mice per group). Results were normalized to the expression of Gapdh. (B) Flow cytometric analysis of intestinal ECs (CD45 EpCam+) was performed with UEA-1- FITC to detect fucosylated-ECs. Representative dot-plots and percentages of fucosylated-ECs are shown (n=3-9 mice per group) according to cell size (FSC-A). (C) RT-qPCR analysis of mRNA encoding for Cldn4 , Tjpl, Ocln , and Muc2 in sorted ECs from ileum (n=10 mice per group). Results were normalized to the expression of Gapdh. (D) Intestinal gut permeability was evaluated with in vivo FITC-dextran assay (n=6-10 mice per group). (E) Relative SFB abundance and level determined by 16S-sequencing and qPCR of fecal contents and ileum tissue on day 13 post-transfer (n=6-10 mice per group). qPCR results were normalized to the respective abundance of total bacteria. (F) Analysis of pancreatic islet infiltration and incidence of diabetes of recipient Trac/~ NOD mice on day 15 after transfer with BDC2.5 T cells and treatment with either anti-TNFa or human control IgGl antibodies (n=26-27 mice per group). All data are representative of at least three independent experiments and horizontal lines indicate the median, each data point represents an individual mouse. *P<0.05, **P<0.01 and ***P<0.001 (unpaired t-test (in panel A, for Foxp3 mRNA) and two-tailed Mann-Whitney test).
EXAMPLE:
Methods
Mice. Male and female C57BL/6J, male and female NOD, female RORyt-GFP NOD, female BDC2.5 CD45.2+/+ Trac NOD, and female recipient CD45.1+/+ Trac NOD mice were previously described [24,59] All mice were bred under specific pathogen-free conditions. This study was approved by the Ethics Committee on Animal Experimentation (APAFIS#3474- 2015102016444419v3 and APAFIS# 17515-201811091441828v4).
Isolation of DNA, RNA, and RT. For DNA extraction, fresh stool or small pieces of ileum tissue were weighted, recovered, immediately frozen in liquid nitrogen upon collection, and stored at -80°C until DNA isolation. DNA extraction was performed using NucleoSpin Tissue (Macherey -Nagel) following the manufacturer’s instructions. Sections of liver near the portal vein were recovered from mice at different ages and DNA was extracted as previously described [24] For RNA isolation, cells were lysed in RLT buffer with 1% b-mercaptoethanol, and mRNA was purified from lysed cells using RNeasy Mini Kit (Qiagen). cDNA was synthesized using the Superscript III reverse transcriptase (Invitrogen). qPCR Quantitative-PCR analysis was performed with SYBR Green (Roche) and analyzed using a LightCycler 480 (Roche). The relative expression was calculated using the 2 DDa method and normalized to the expression of the housekeeping gene Gapdh or the expression of total bacteria, as appropriate. The stability of Gapdh expression was confirmed by comparison with hypoxanthine phosphoribosyltransferase ( Hprt ) mRNA expression.
Fecal microbiota analysis. 16S rRNA gene amplification and library constructions were performed according to Illumina recommendations. Sequencing was performed on an Illumina MiSeq. Taxonomy and data analyses were conducted using QIIME.
Statistical analysis. Statistical analyses were performed with Prism software (Graph Pad V8.3). All datasets were tested for normal distribution using the Shapiro-Wilk normality test. Datasets were compared using either parametric two-tailed unpaired t-test, nonparametric two-tailed Mann-Whitney or Kruskal -Wallis tests with multiple comparisons Dunn’s post-test, as appropriate. Significant differences in taxonomic abundances were assessed with nonparametric Kruskal-Wallis test with the false-discovery rate correction implemented in Qiime. Diabetes incidence was plotted according to the Kaplan-Meier method and statistical differences were analyzed using the Mantel-Cox (log-rank test). Differences were considered significant at P < 0.05 (*/$P<0.05, **P<0.01, ***P<0.001, ****P<0.0001).
Results
T1D development in NOD mice is associated with decreased production of IL-17A, IL-22, and IL-23A by intestinal immune cells.
We first began our investigation of gut alterations by analyzing the production of cytokines in the lamina propria (LP) of ileum from female NOD mice at pre-diabetic stage (8-10 weeks) and at diabetes onset. A significant decrease in transcripts for //-77a, 11-22 and 11-23a was observed in diabetic compared to younger pre-diabetic NOD mice (data not shown). To confirm this decrease at the protein level, flow cytometry analyses of LP cells were performed. Intracellular labeling of IL-17A and IL-22 cytokines of hematopoietic cells (CD45+), TCR-b- cells and TCR-b lymphoid cells also revealed a significant decrease in the frequency of IL- 17A and IL-22-producing cells in diabetic mice (data not shown). Similar reductions were observed in absolute cell numbers (data not shown).
Since the Retinoic-acid-receptor-related Orphan nuclear Receptor gamma (RORyt) is a key transcription factor controlling IL-17A [28] and IL-22 production [27], we used a RORyt-GFP reporter NOD mouse line that we had generated to evaluate RORyt cellular expression [24] At the diabetic stage, an overall decrease of RORyt-GFP expression was observed in CD45+, TCRb+, and TCRb cells (data not shown). Moreover, we observed the same decrease in CD4 T, invariant Natural Killer T (iNKT), MAIT, gdT, and innate lymphoid cells (ILCs) (data not shown). These data show that T1D development is associated with a general defect in IL-17A and IL-22 production by gut mucosa immune cells.
T1D development is associated with alteration of gut mucosa functions and decreased segmented filamentous bacteria abundance.
The fucosyltransferase 2 (FUT2) enzyme expressed by gut epithelial cells (EC) is representative of a protective mechanism promoting the maintenance of the commensal gut microbiota, protecting the host against opportunistic pathogens [29] Since IL-22 can induce Fut2 expression by ileum ECs, we analyzed Fut2 expression in the ileum of female NOD mice at different disease stages [29,30] mRNA expression of Fut2 in ileum of diabetic mice was significantly lower than in younger prediabetic mice (data not shown). In addition, analysis of a(l,2)-fucose levels on EC surface by the binding of fluorescent Ulex europaeus agglutinin-1 (UEA-1) lectin showed a significant decrease in the frequency and intensity of EC fucosylation in diabetic mice (data not shown).
Since SFB are intimately attached to ECs in ileum and induce IL-17A and IL-22 production by gut immune cells [31-36], we investigated whether ECs alterations were associated with a loss of SFB. The relative abundance of SFB in ileum was significantly reduced in diabetic mice compared to prediabetic mice (Figure 1A). The decrease of SFB relative quantities in diabetic mice was also observed in feces (Figure IB). Analysis revealed that SFB levels progressively decreased from 4-week up to 13-week old female NOD mice free from diabetes (Figure 1C). Progressive loss of SFB was similar between age-matched female C57BL/6J and female NOD mice that remained diabetes-free until up to 13 weeks of age. However, SFB loss was significantly increased as soon as 9 weeks of age in female NOD mice that became subsequently diabetic at 13 weeks of age (Figure ID). Of note, SFB relative quantity stabilized at diabetes onset. The diminished amount of SFB in ileum positively correlated with the decreased expression of Fut2 mRNA in diabetic mice (data not shown).
After oral gavage, blood FITC-dextran concentration was significantly higher in diabetic NOD mice than in 10-week old NOD mice (data not shown). We also observed a reduction of claudin-4 ( Cldn4 ), tight junction protein 1 ( Tjpl ), occludin ( Ocln ) and mucin-2 (Muc2) mRNA in sorted ECadhesion molecule (EpCam)+ ECs of ileum from diabetic compared to younger mice (data not shown).
We next analyzed whether loss of gut integrity in diabetic mice was associated with bacterial translocation. Increased detection of 16S DNA was observed in liver of diabetic NOD mice compared to prediabetic NOD and C57BL/6J mice (data not shown). There was also an increase of CFU of anaerobic and aerobic bacteria in liver from 17-week old and diabetic mice compared to younger NOD and control C57BL/6J mice (data not shown). Bacteria analysis highlighted that bacteria of anaerobic and aerobic culture from the liver are mostly Lactobacillus murinus (data not shown). Together these data show that progression toward diabetes in NOD mice is associated with several gut mucosa alterations and a dramatic decrease of SFB.
Gut mucosa alterations in streptozotocin-induced diabetes in NOD and C57BL/6J mice We next investigated whether the observed gut mucosa alterations and SFB loss were restricted to T1D development in NOD mice or could be observed in other T1D mouse models. A commonly used mouse model of T1D is based on treatment with multiple low doses of streptozotocin (STZ) [37] (data not shown). NOD males became diabetic shortly after STZ treatment and harbored the same mucosa alterations as spontaneous diabetic NOD females: notably reduced //-77a, 11-22 and Il-23a mRNA expression in LP cells of ileum (data not
shown), a significant decrease of UEA-1+ ECs frequencies (figure 3C) as well as a reduction of Fut2 mRNA transcription (data not shown) and diminished levels of SFB in feces associated with treatment duration (data not shown). To exclude the possibility of NOD mouse-specific defects we also analyzed C57BL/6J mice treated with STZ (data not shown). As in diabetic NOD mice, diabetes in STZ-treated C57BL/6J mice was associated with reduced transcripts for II- 17 a, 11-22 , and Il-23a in LP cells of ileum (data not shown), decreased frequencies of UEA- 1+ ileum ECs, as well as a slight decrease of mean fluorescence intensity (MFI) of UEA-1 (p=0.0519) (data not shown). We observed decreased levels of SFB in ileum and feces as in NOD mice (data not shown). Therefore, these gut microbiota and mucosa alterations seem to represent common features associated with T1D onset.
Hyperglycemia does not replicate intestinal alterations observed in T1D
Next, we investigated whether these alterations observed in diabetic mice could occur upon induction of hyperglycemia in absence of autoimmune/inflammatory reaction. To this end, C57BL/6J mice were grafted with a pump allowing continuous diffusion of S961, an insulin receptor antagonist [38] (data not shown). In contrast to what was observed in prediabetic NOD and STZ-treated mice, hyperglycemia in S961 -treated mice induced a significant increase of SFB abundance in feces and ileum (data not shown). Moreover, there was no difference in UEA-1 expression on intestinal ECs and no modification of //-77a and 11-22 mRNA expression in cells from ileum LP (data not shown). However, hyperglycemia induced a significant increase of Il-23a mRNA expression in LP cells from ileum (data not shown). These data show that hyperglycemia alone does not induce SFB, IL-17A, IL-22 and IL-23A loss that we observed in NOD and STZ-treated mice.
Diabetes induced by diabetogenic T-cells alters intestinal homeostasis and SFB level.
We transferred i. v. diabetogenic T cells (BDC2.5) in Trac NOD recipient mice. This model allows synchronization of T1D induction and monitoring of diabetogenic CD4+ T cells migration within different tissues. Onset of diabetes in recipient mice (data not shown) was associated with the same gut alterations as observed in NOD mice such as a significant decrease of SFB in ileum and feces (data not shown), an enhanced gut permeability evaluated by FITC- dextran in blood after oral gavage (data not shown), and a decrease in mRNA transcripts coding for 7/-/7a, 11-22 and 11-23a in cells from ileum (data not shown). Increased gut permeability in diabetic recipient mice was associated with a significant decrease of Tjpl and
Muc2 mRNA levels and a slight reduction of Cldn4 (p=0.07) and Ocln (p=0.10) mRNA levels in intestinal epithelium cells (data not shown).
We next assessed whether diabetogenic BDC2.5 T cells could home into the ileum of recipient mice. Flow cytometry analysis showed presence of BDC2.5 T cells from day 2 to day 13 in the ileum, PLN and mesenteric lymph nodes (MLN) (data not shown). BDC2.5 T cell frequency in ileum was higher at day 13 compared to MLN and PLN (data not shown). Furthermore, an elevated frequency of BDC2.5 T cells in ileum produced IFN-g and TNF-a compared to MLN and PLN (data not shown). We also transferred BDC2.5 T cells in wild-type NOD recipient mice, in which they do not induce diabetes [39], allowing analysis of BDC2.5 T cell behavior without potential contribution of homeostatic proliferation that could occur in Trac A NOD recipient mice. Flow cytometry analysis confirmed the presence of BDC2.5 T cells at day 8 in ileum, MLN, PLN and pancreatic islets (data not shown). Increased frequency of BDC2.5 T cells in these organs resulted from their higher proliferation rate compared to host CD45.2 CD4+ T cells (data not shown). Moreover, BDC2.5 T cells produced high levels of IFN-g and TNFa in ileum, MLN and PLN compared to host CD45.2 CD4+ T cells, and the highest TNFa level in BDC2.5 T cells was observed in ileum even when compared to all organs, including pancreatic islets (data not shown). These data suggest that gut mucosa alterations in recipient mice are caused by diabetogenic T cell production of inflammatory cytokines in ileum.
Gut microbiota alteration in NOD mice matches the observations in BDC2.5 model.
As our data revealed the impact of gut inflammation on SFB levels, we next performed a more extensive analysis of gut microbiota alterations in mouse models of T1D versus our non- autoimmune hyperglycemic mouse model. 16S ribosomal DNA sequencing confirmed SFB reduction with T1D progression in NOD mice at different ages as well as in BDC2.5 T cell recipient mice (data not shown). 16S DNA sequencing also confirmed the increase of SFB in hyperglycemic S961 -treated mice (figure 6A). Principal Component Analysis (PCA) of 16S ribosomal DNA sequencing showed an unsupervised overview of gut microbiota modification in these mice (data not shown). In NOD mice there was a shift of the gut microbiome from 6/10-week old mice to diabetic mice with the PC2 (9.85%). In BDC2.5 T cell transfer experiments, two clusters separated control from BDC2.5 T cell-induced diabetic mice mostly by PC2 (20.23%) (data not shown). Lastly, in S961 experiments we observed two clusters that separated control from hyperglycemic mice mostly by PCI (34.05%) (data not shown).
We next explored more precisely gut microbiota modifications during T1D progression in NOD mice. 16S sequencing of fecal content from NOD mice from 6 weeks of age to diabetes onset
showed a total of 24 phyla and genera modifications (data not shown). As previously shown, relative abundance of Bacteroidetes and Firmicutes were respectively decreased and increased in diabetic NOD mice as compared to pre-diabetic mice [40] We further observed other modifications during T1D progression in NOD mice and several were common with modifications occurring in BDC2.5 T cell recipient mice that became diabetic. A significant increase of Coprococcus , Dorea, Oscillospira , Thiobacter was associated with disease progression (data not shown). However, other gut microbiota modifications were similar during the development of diabetes in NOD mice and induction of hyperglycemia in S961- treated mice. In both mouse models, there was a significant decrease of Bacteroidetes phylum and an increase of Bacteroides genus. Conversely, Parabacteroides was reduced in hyperglycemic mice compared to diabetic NOD mice (data not shown). We did not observe any similarities in microbiome alteration between BDC2.5 T cells transferred mice and hyperglycemic S961 -treated mice (data not shown). Some modifications only occurred in S961 -treated mice such as an increase of Enterococcus, Erwinia, and Klebsiella and a decrease of Allobaculum genera (data not shown). While Coprococcus increased in diabetic NOD mice and BDC2.5 T cell transferred mice, this genus was rather reduced (p=0.0642) in hyperglycemic S961 -treated mice compared to their controls (data not shown). Similarly, the increase of Dorea and Oscillospira genera in both diabetic NOD mice and BDC2.5 T cell transferred mice was not observed in hyperglycemic S961 -treated mice. Overall, these data highlight several gut microbiota modifications occurring in two mouse models of autoimmune diabetes but not upon induction of hyperglycemia.
Anti-TNFa treatment prevents gut mucosa alterations, restores SFB levels, and prevents T1D onset.
Since transferred BDC2.5 diabetogenic T cells migrated and expanded to the ileum where they produced a large amount of TNFa, we assessed the impact of Remicade®, an anti-TNFa antibody (Infliximab), on gut inflammation and diabetes development. Anti-TNFa antibody treatment had a major impact on gut mucosa as it increased II- 17 a and 11-22 mRNA level as well as Foxp3 transcript abundance in LP cells of ileum from treated mice compared to isotype- injected control mice (Figure 2A). Ileal EC function was improved by anti-TNFa treatment as UEA-1 expression on intestinal ECs increased (Figure 2B). In addition, transcript levels coding for Cldn4 , Ocln were significantly elevated, with a slight increase of Muc2 mRNA level (p=0.07), and gut permeability was significantly decreased (Figure 2C,D) This was associated with an increase of SFB frequencies and relative quantity in fecal content, although it remained
similar in ileum (Figure 2E). Changes were also observed in other bacterial genera with an increase mAF12, Parracoccus, Acinobacter, and a decrease in Clostridium , Coprobacillus , and Streptococcus (data not shown). Of note, macrophages, which are able to produce TNFa in the LP, were not affected by anti-TNFa treatment (data not shown). We next analyzed dendritic cells (DCs) since we had previously shown that loss of gut integrity in NOD mice could lead to bacterial components translocation and increased CDl lc+CDl lb+CD103+ DCs frequencies and activation [24] Indeed, anti-TNFa antibodies induced a significant reduction of this DCs subset in ileum of BCD2.5 T cell-transferred mice, however, such reduction was not observed in PLN (data not shown). Moreover, anti-TNFa treatment also decreased ileum CDl lc+CDl lb+CD103+ DCs activation as shown by their lower surface levels of MHC-II compared to non-treated control mice (data not shown). Interestingly anti-TNFa reduced the frequency and absolute numbers of BDC2.5 T cells in ileum but not in PLN (data not shown). This was associated with a significant increase of FOXP3+ BDC2.5 T cell frequency in ileum and not in PLN (data not shown). Further analysis of autoreactive T cells showed a significant reduction of their IFN-g production in both ileum and PLN from mice treated with anti-TNFa compared to control mice whereas TNFa production by BDC2.5 T cells remained unchanged (data not shown). Finally, the decreased IFN-g production by BDC2.5 T cells in anti-TNFa treated mice was associated with a lower incidence of T1D and less severe insulitis (Figure 2F). Altogether these data support again a key role for gut inflammation in promoting anti-islet T cell effector function leading to pancreatic infiltration and T1D onset. Reducing inflammation (by anti-TNFa treatment) helps correcting TID-associated SFB dysbiosis and mucosal dysfunctions (data not shown).
Discussion:
Our study brings new knowledge on the nature and the origin of gut mucosa and microbiota alterations associated with T1D development. Using several mouse models of T1D, we showed that with disease development i) gut immune cells produced lower levels of IL-17A, IL-22 and IL-23 A, ii) intestinal ECs displayed decreased expression markers of gut integrity (Claudin 4, Occludin, Tight junction protein 1, and Mucin 2), and decreased fucosylation levels and iii) gut microbiota changes were associated with reduced abundance of SFB in ileum. Moreover, our results revealed that these modifications were not observed upon metabolic induction of hyperglycemia. Several reports had described gut alterations in T1D, obesity, and T2D as well as after induction of hyperglycemia by insulin receptor blockade [12,13,41] While NOD mice develop gut alterations in several weeks, the other diabetic models (STZ multi -low-dose
injections, S961 -treatment, and BDC2.5 T cell transfer) induced diabetes within a week. Therefore, chronicity of gut inflammation is not required for gut alterations observed in T1D. Modifications of gut microbiota composition were very similar between spontaneously diabetic NOD, and BDC2.5 T cells transferred Trac^ recipient mice in contrast to gut microbiota modification observed in metabolically induced hyperglycemic mice. Taken together, this suggests that targeting only hyperglycemia in T1D would not resolve mucosal dysfunctions whereas treating inflammation could restore intestinal homeostasis.
T1D is characterized by the development of autoimmune responses associated with inappropriate activation of immune cells that infiltrate the pancreas [1-3] We and others have described gut inflammation associated with T1D suggesting that gut inflammation could lead to decreased mucosal IL-17A, IL-22 and IL-23A production, EC defect and loss of ileum SFB [24,42] It was striking that BDC2.5 T cell frequency was higher in the gut mucosa than in MLN and PLN, and they produced the highest level of TNFa in the gut. TNFa production by low-proliferating BDC2.5 T cells in ileum is probably due to the pro-inflammatory environment rather than potential microbiota cross-reactivity. These results prompted us to assess the efficiency of anti-TNFa treatment, which is a common treatment in Crohn’s disease [43] This treatment had a major effect on the gut mucosa as it increased IL-22 and IL-17A production, fucosylation of ECs and SFB abundance. Moreover, lower DC activation and increased T regulatory cell frequency were observed in the ileum but not in PLN supporting a primary effect on the gut mucosa. Furthermore, it has been reported that gut IL-10+ T regulatory cells could migrate to PLN [44] Systemic anti-TNFa treatment does not exclude that anti-inflammatory mAb could directly act on pathogenic cells in PLN and pancreas. TNFa can exert an ambivalent role in the development of T1D in mouse models [45-48] and a previous report has shown that anti-TNFa treatment at the perinatal period in NOD mice prevents T1D onset [47] Of note, gut mucosa was not analyzed in this pioneer study. Interestingly, anti-TNFa treatment in a patient with Crohn’s disease and T1D not only improved intestinal pathology but also T1D [49] Our present data further highlights the relevance of controlling gut inflammatory status to dampen pathogenic anti -islet T cells in PLN, severe insulitis and T1D.
We showed for the first time that SFB levels in ileum and feces were decreased during the establishment of spontaneous diabetes in NOD mice, in T1D induced by STZ treatment in NOD and C57BL/6J mice as well as upon transfer of autoreactive BDC2.5 T cells. In contrast, metabolic induction of hyperglycemia enhanced SFB levels in ileum and feces and is reminiscent with SFB requiring high glucose levels to grow on ECs [50] Thus, low SFB level in T1D might be a consequence of local inflammation, despite increased glycemia. This finding
is interesting in light to previous studies asking whether SFB could exert a protective role against T1D. Comparison of two NOD colonies from different breeding companies showed that the colony harboring SFB had a lower incidence of T1D than the other colony [42] In another study, germ-free NOD mice were colonized with SFB however incidence of T1D was only slightly decreased in recipient males, not females [51] Our study established a strong parallel between SFB abundance in ileum and intestinal IL-17A and IL-22 production. Both cytokines can be induced by SFB [32-36] and maintain gut homeostasis thereby preventing bacterial component translocation and inflammation. Several mechanisms could be at play. IL-22 can upregulate Fut2 expression and ECs fucosylation favoring the maintenance of commensal flora [29,30] IL-22 can also maintain gut integrity by promoting expression of tight junctions proteins and production of mucus preventing pathogenic bacteria invasiveness [52] IL-22 could also alleviate pancreatic b cell stress, preventing the generation of islet neoantigens playing a critical role in the autoimmune attack of b cells [52-54]
The role of SFB in the dampening of gut mucosa inflammation and diabetes development can also be associated with increased diabetes incidence after antibiotic treatment [55] Indeed, SFB is extremely sensitive to antibiotics [56] Another protective role of SFB could be related to their ability to prevent bacterial and viral infections. SFB can induce the production of IL-22 that controls C. rodentium infection [57], an intestinal pathogenic bacteria that is known to accelerate T1D development in NOD mice [8] Similarly, SFB prevents rotavirus infection in mice that might be involved in T1D development [56,58] Thus, our study provides new insights in the deleterious role of gut inflammation against protective commensal microbiota in the physiopathology of T1D.
REFERENCES:
Throughout this application, various references describe the state of the art to which this invention pertains. The disclosures of these references are hereby incorporated by reference into the present disclosure.
1 Atkinson MA, Eisenbarth GS, Michels AW. Type 1 diabetes. Lancet 2014;383:69-82. doi:10.1016/S0140-6736(13)60591-7
2 Lehuen A, Diana J, Zaccone P, et al. Immune cell crosstalk in type 1 diabetes. Nat Rev Immunol 2010;10:501-13. doi:10.1038/nri2787
3 Diana J, Simoni Y, Furio L, et al. Crosstalk between neutrophils, B-la cells and plasmacytoid dendritic cells initiates autoimmune diabetes. Nat Med 2013;19:65-73. doi: 10.1038/nm.3042
4 Meddings JB, Jarand J, Urbanski SJ, et al. Increased gastrointestinal permeability is an early lesion in the spontaneously diabetic BB rat. Am J Physiol 1999;276:G951-957. doklO.l 152/ajpgi.1999.276.4. G951
5 Alam C, Valkonen S, Palagani V, et al. Inflammatory tendencies and overproduction of IL-17 in the colon of young NOD mice are counteracted with diet change. Diabetes 2010;59:2237-46. doi:10.2337/dbl0-0147
6 Sapone A, de Magistris L, Pietzak M, et al. Zonulin upregulation is associated with increased gut permeability in subjects with type 1 diabetes and their relatives. Diabetes 2006;55:1443-9. doi:10.2337/db05-1593
7 Graham S, Courtois P, Malaisse WJ, et al. Enteropathy precedes type 1 diabetes in the BB rat. Gut 2004;53:1437-44. doi:10.1136/gut.2004.042481
8 Lee AS, Gibson DL, Zhang Y, et al. Gut barrier disruption by an enteric bacterial pathogen accelerates insulitis in NOD mice. Diabetologia 2010;53:741-8. doi:10.1007/s00125- 009-1626-y
9 Miranda MCG, Oliveira RP, Torres L, et al. Frontline Science: Abnormalities in the gut mucosa of non-obese diabetic mice precede the onset of type 1 diabetes. J Leukoc Biol 2019;106:513-29. doi:10.1002/JLB.3HI0119-024RR
10 Carratu R, Secondulfo M, de Magistris L, et al. Altered intestinal permeability to mannitol in diabetes mellitus type I. J Pediatr Gastroenterol Nutr 1999;28:264-9. doi: 10.1097/00005176-199903000-00010
11 Kuitunen M, Saukkonen T, Ilonen J, et al. Intestinal permeability to mannitol and lactulose in children with type 1 diabetes with the HLA-DQB1*02 allele. Autoimmunity 2002;35:365-8. doi:10.1080/0891693021000008526
12 Vaarala O. Leaking gut in type 1 diabetes. Curr Opin Gastroenterol 2008;24:701-6. doi : 10.1097/MOG.ObO 13 e32830e6d98
13 Thaiss CA, Levy M, Grosheva I, et al. Hyperglycemia drives intestinal barrier dysfunction and risk for enteric infection. Science 2018;359:1376-83. doi:10.1126/science.aar3318
14 Hansen CHF, Krych L, Buschard K, et al. A maternal gluten-free diet reduces inflammation and diabetes incidence in the offspring of NOD mice. Diabetes 2014;63:2821- 32. doi:10.2337/dbl3-1612
15 Dunne JL, Triplett EW, Gevers D, et al. The intestinal microbiome in type 1 diabetes. Clin Exp Immunol 2014;177:30-7. doi: 10.1111/cei.12321
16 Kostic AD, Gevers D, Siljander H, et al. The dynamics of the human infant gut microbiome in development and in progression toward type 1 diabetes. Cell Host Microbe 2015;17:260-73. doi:10.1016/j.chom.2015.01.001
17 Alkanani AK, Hara N, Gottlieb PA, et al. Alterations in Intestinal Microbiota Correlate With Susceptibility to Type 1 Diabetes. Diabetes 2015;64:3510-20. doi:10.2337/dbl4-1847
18 Westerholm-Ormio M, Vaarala O, Pihkala P, et al. Immunologic activity in the small intestinal mucosa of pediatric patients with type 1 diabetes. Diabetes 2003;52:2287-95. doi: 10.2337/diabetes.52.9.2287
19 Pellegrini S, Sordi V, Bolla AM, et al. Duodenal Mucosa of Patients With Type 1 Diabetes Shows Distinctive Inflammatory Profile and Microbiota. J Clin Endocrinol Metab 2017;102:1468-77. doi:10.1210/jc.2016-3222
20 Hardin JA, Donegan L, Woodman RC, et al. Mucosal inflammation in a genetic model of spontaneous type I diabetes mellitus. Can J Physiol Pharmacol 2002;80:1064-70. doi:10.1139/y02-138
21 Leeds JS, Hopper AD, Hadjivassiliou M, et al. Inflammatory bowel disease is more common in type 1 diabetes mellitus. Gut 2011;60:A208-A208. doi:10.1136/gut.2011.239301.438
22 Rewers M, Eisenbarth GS. Celiac disease in T1DM — the need to look long term. Nat Rev Endocrinol 2011;8:5-8. doi : 10.1038/nrendo.2011.193
23 Badami E, Sorini C, Coccia M, et al. Defective differentiation of regulatory FoxP3+ T cells by small-intestinal dendritic cells in patients with type 1 diabetes. Diabetes 2011;60:2120- 4. doi :10.2337/db 10-1201
24 Rouxel O, Da Silva J, Beaudoin L, et al. Cytotoxic and regulatory roles of mucosal- associated invariant T cells in type 1 diabetes. Nat Immunol 2017;18:1321-31. doi:10.1038/ni.3854
25 Colonna M. Interleukin-22-producing natural killer cells and lymphoid tissue inducer like cells in mucosal immunity. Immunity 2009;31:15-23. doi:10.1016/j.immuni.2009.06.008
26 Lee JS, Tato CM, Joyce-Shaikh B, et al. Interleukin-23 -Independent IL-17 Production Regulates Intestinal Epithelial Permeability. Immunity 2015;43:727-38. doi: 10.1016/j.immuni.2015.09.003
27 Dudakov JA, Hanash AM, van den Brink MRM. Interleukin-22: immunobiology and pathology. Annu Rev Immunol 2015;33:747-85. doi: 10.1146/annurev-immunol-032414- 112123
28 Ivanov II, McKenzie BS, Zhou L, et al. The orphan nuclear receptor RORgammat directs the differentiation program of proinflammatory IL-17+ T helper cells. Cell 2006;126:1121-33. doi:10.1016/j.cell.2006.07.035
29 Pickard JM, Maurice CF, Kinnebrew MA, et al. Rapid fucosylation of intestinal epithelium sustains host-commensal symbiosis in sickness. Nature 2014;514:638-41. doi : 10.1038/nature 13823
30 Goto Y, Obata T, Kunisawa J, et al. Innate lymphoid cells regulate intestinal epithelial cell glycosylation. Science 2014;345:1254009. doi: 10.1126/science.1254009
31 Chase DG, Erlandsen SL. Evidence for a complex life cycle and endospore formation in the attached, filamentous, segmented bacterium from murine ileum. J Bacterid 1976;127:572-83.
32 Ivanov II, Atarashi K, Manel N, et al. Induction of intestinal Thl7 cells by segmented filamentous bacteria. Cell 2009;139:485-98. doi:10.1016/j.cell.2009.09.033
33 Gaboriau-Routhiau V, Rakotobe S, Lecuyer E, et al. The key role of segmented filamentous bacteria in the coordinated maturation of gut helper T cell responses. Immunity 2009;31 :677-89. doi: 10.1016/j .immuni.2009.08.020
34 Goto Y, Panea C, Nakato G, et al. Segmented filamentous bacteria antigens presented by intestinal dendritic cells drive mucosal Thl7 cell differentiation. Immunity 2014;40:594- 607. doi: 10.1016/j .immuni.2014.03.005
35 Lecuyer E, Rakotobe S, Lengline-Garnier H, et al. Segmented filamentous bacterium uses secondary and tertiary lymphoid tissues to induce gut IgA and specific T helper 17 cell responses. Immunity 2014;40:608-20. doi:10.1016/j. immuni.2014.03.009
36 Omenetti S, Bussi C, Metidji A, et al. The Intestine Harbors Functionally Distinct Homeostatic Tissue-Resident and Inflammatory Thl7 Cells. Immunity Published Online First: June 2019. doi: 10.1016/j .immuni.2019.05.004
37 Leiter EH. Multiple low-dose streptozotocin-induced hyperglycemia and insulitis in C57BL mice: influence of inbred background, sex, and thymus. Proc Natl Acad Sci U S A 1982;79:630-4. doi: 10.1073/pnas.79.2.630
38 Schaffer L, Brand CL, Hansen BF, et al. A novel high-affinity peptide antagonist to the insulin receptor. Biochem Biophys Res Commun 2008;376:380-3. doi: 10.1016/j .bbrc.2008.08.151
39 Beaudoin L, Diana J, Ghazarian L, et al. Plasmacytoid dendritic cells license regulatory T cells, upon iNKT-cell stimulation, to prevent autoimmune diabetes. Eur J Immunol 2014;44:1454-66. doi:10.1002/eji.201343910
40 Hu Y, Peng J, Li F, et al. Evaluation of different mucosal microbiota leads to gut microbiota-based prediction of type 1 diabetes in NOD mice. Sci Rep 2018;8: 15451. doi : 10.1038/s41598-018-33571-z
41 Winer DA, Winer S, Dranse HJ, et al. Immunologic impact of the intestine in metabolic disease. J Clin Invest 2017;127:33-42. doi:10.1172/JCI88879
42 Kriegel MA, Sefik E, Hill JA, et al. Naturally transmitted segmented filamentous bacteria segregate with diabetes protection in nonobese diabetic mice. Proc Natl Acad Sci 2011 ; 108 : 11548-53. doi : 10.1073/pnas.1108924108
43 Colombel JF, Sandborn WJ, Reinisch W, et al. Infliximab, azathioprine, or combination therapy for Crohn’s disease. N Engl J Med 2010;362:1383-95. doi:10.1056/NEJMoa0904492
44 Yu H, Gagliani N, Ishigame H, et al. Intestinal type 1 regulatory T cells migrate to periphery to suppress diabetogenic T cells and prevent diabetes development. Proc Natl Acad Sci U S A 2017; 114: 10443-8. doi: 10.1073/pnas.1705599114
45 Jacob CO, Aiso S, Michie SA, et al. Prevention of diabetes in nonobese diabetic mice by tumor necrosis factor (TNF): similarities between TNF-alpha and interleukin 1. Proc Natl Acad Sci U S A 1990;87:968-72. doi:10.1073/pnas.87.3.968
46 Jacob CO, Aiso S, Schreiber RD, et al. Monoclonal anti-tumor necrosis factor antibody renders non-obese diabetic mice hypersensitive to irradiation and enhances insulitis development. Int Immunol 1992;4:611-4. doi:10.1093/intimm/4.5.611
47 Yang XD, Tisch R, Singer SM, et al. Effect of tumor necrosis factor alpha on insulin- dependent diabetes mellitus in NOD mice. I. The early development of autoimmunity and the diabetogenic process. J Exp Med 1994;180:995-1004. doi:10.1084/jem.l80.3.995
48 Green EA, Gorelik L, McGregor CM, et al. CD4+CD25+ T regulatory cells control anti islet CD8+ T cells through TGF-beta-TGF-beta receptor interactions in type 1 diabetes. Proc Natl Acad Sci U S A 2003;100:10878-83. doi: 10.1073/pnas.1834400100
49 Timper K, Hruz P, Beglinger C, et al. Infliximab in the treatment of Crohn disease and type 1 diabetes. Diabetes Care 2013;36:e90-91. doi:10.2337/dcl3-0199
50 Schnupf P, Gaboriau-Routhiau V, Gros M, et al. Growth and host interaction of mouse segmented filamentous bacteria in vitro. Nature 2015;520:99-103. doi:10.1038/naturel4027
51 Yurkovetskiy L, Burrows M, Khan AA, et al. Gender bias in autoimmunity is influenced by microbiota. Immunity 2013;39:400-12. doi:10.1016/j.immuni.2013.08.013
52 Rutz S, Wang X, Ouyang W. The IL-20 subfamily of cytokines— from host defence to tissue homeostasis. Nat Rev Immunol 2014;14:783-95. doi:10.1038/nri3766
53 Hasnain SZ, Borg DJ, Harcourt BE, et al. Glycemic control in diabetes is restored by therapeutic manipulation of cytokines that regulate beta cell stress. Nat Med 2014;20:1417-26. doi: 10.1038/nm.3705
54 Delong T, Wiles TA, Baker RL, et al. Pathogenic CD4 T cells in type 1 diabetes recognize epitopes formed by peptide fusion. Science 2016;351:711-4. doi : 10.1126/ science. aad2791
55 Livanos AE, Greiner TU, Vangay P, et al. Antibiotic-mediated gut microbiome perturbation accelerates development of type 1 diabetes in mice. Nat Microbiol 2016; 1:16140. doi : 10.1038/nmicrobiol .2016.140 56 Shi Z, Zou J, Zhang Z, et al. Segmented Filamentous Bacteria Prevent and Cure
Rotavirus Infection. Cell 2019;179:644-658. el3. doi:10.1016/j.cell.2019.09.028 57 Zheng Y, Valdez PA, Danilenko DM, et al. Interleukin-22 mediates early host defense against attaching and effacing bacterial pathogens. Nat Med 2008;14:282-9. doi: 10.1038/nm 1720 58 Honeyman MC, Coulson BS, Stone NL, et al. Association between rotavirus infection and pancreatic islet autoimmunity in children at risk of developing type 1 diabetes. Diabetes 2000;49:1319-24. doi:10.2337/diabetes.49.8.1319
59 Katz JD, Benoist C, Mathis D. T helper cell subsets in insulin-dependent diabetes. Science 1995;268:1185-8. doi:10.1126/science.7761837
Claims
1. A method for treating, preventing, suppressing and/or delaying the onset, or reducing the risk of developing type 1 diabetes, or the symptoms associated with, or related to, type 1 diabetes, in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a TNFa blocking agent.
2. The method of the claim 1 wherein the subject is a child.
3. The method of claim 1 wherein the subject is a pre-diabetic subject.
4. The method of claim 1 wherein the subject exhibits a dysbiosis.
5. The method of claim 4 wherein the dysbiosis includes progressive loss of Segmented Filamentous Bacteria B (SFB).
6. The method of claim 1 that comprises the steps of measuring the abundance of SFB in a fecal sample obtained from the subject.
7. The method of claim 1 that comprises the steps of i) determining the abundance of SFB in a fecal sample obtained from the subject, ii) comparing the abundance determined at step i) with a predetermined reference value and iii) administering to the subject a therapeutically effective amount of a TNFa blocking agent when the abundance is lower than the predetermined reference value.
8. The method of claim 1 wherein wherein the TNFa blocking agent is a soluble TNFa receptor or an antibody having specificity for TNFa or for TNFa receptor 1.
9. The method of claim 8 wherein the TNFa blocking agent is selected from the group consisting of Etanercept (Enbrel®), Infliximab (Remicade®), Adalimumab (Humira®), Certolizumab pegol (Cimzia®), and golimumab (Simponi®).
10. The method of claim 1 wherein the TNFa blocking agent is locally administered to the gastrointestinal tract of the subject.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
EP21305178.2 | 2021-02-10 | ||
EP21305178 | 2021-02-10 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2022171685A1 true WO2022171685A1 (en) | 2022-08-18 |
Family
ID=74701444
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/EP2022/053148 WO2022171685A1 (en) | 2021-02-10 | 2022-02-09 | Tnf-alpha blocking agent for preventing, suppressing and/or delaying the onset of type 1 diabetes |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2022171685A1 (en) |
Citations (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US4683195A (en) | 1986-01-30 | 1987-07-28 | Cetus Corporation | Process for amplifying, detecting, and/or-cloning nucleic acid sequences |
US4683202A (en) | 1985-03-28 | 1987-07-28 | Cetus Corporation | Process for amplifying nucleic acid sequences |
US4800159A (en) | 1986-02-07 | 1989-01-24 | Cetus Corporation | Process for amplifying, detecting, and/or cloning nucleic acid sequences |
US4965188A (en) | 1986-08-22 | 1990-10-23 | Cetus Corporation | Process for amplifying, detecting, and/or cloning nucleic acid sequences using a thermostable enzyme |
WO2017136524A2 (en) * | 2016-02-05 | 2017-08-10 | Janssen Biotech, Inc. | Anti-tnf antibodies, compositions, methods and use for the treatment or prevention of type 1 diabetes |
-
2022
- 2022-02-09 WO PCT/EP2022/053148 patent/WO2022171685A1/en active Application Filing
Patent Citations (7)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US4683202A (en) | 1985-03-28 | 1987-07-28 | Cetus Corporation | Process for amplifying nucleic acid sequences |
US4683202B1 (en) | 1985-03-28 | 1990-11-27 | Cetus Corp | |
US4683195A (en) | 1986-01-30 | 1987-07-28 | Cetus Corporation | Process for amplifying, detecting, and/or-cloning nucleic acid sequences |
US4683195B1 (en) | 1986-01-30 | 1990-11-27 | Cetus Corp | |
US4800159A (en) | 1986-02-07 | 1989-01-24 | Cetus Corporation | Process for amplifying, detecting, and/or cloning nucleic acid sequences |
US4965188A (en) | 1986-08-22 | 1990-10-23 | Cetus Corporation | Process for amplifying, detecting, and/or cloning nucleic acid sequences using a thermostable enzyme |
WO2017136524A2 (en) * | 2016-02-05 | 2017-08-10 | Janssen Biotech, Inc. | Anti-tnf antibodies, compositions, methods and use for the treatment or prevention of type 1 diabetes |
Non-Patent Citations (72)
Title |
---|
"Diagnostic Molecular Microbiology: Principles and Applications", 1993, AMERICAN SOCIETY FOR MICROBIOLOGY |
ALAM CVALKONEN SPALAGANI V ET AL.: "Inflammatory tendencies and overproduction of IL-17 in the colon of young NOD mice are counteracted with diet change", DIABETES, vol. 59, 2010, pages 2237 - 46 |
ALKANANI AKHARA NGOTTLIEB PA ET AL.: "Alterations in Intestinal Microbiota Correlate With Susceptibility to Type 1 Diabetes", DIABETES, vol. 64, 2015, pages 3510 - 20 |
ATKINSON MAEISENBARTH GSMICHELS AW: "Type 1 diabetes", LANCET, vol. 383, 2014, pages 69 - 82 |
BADAMI ESORINI CCOCCIA M ET AL.: "Defective differentiation of regulatory FoxP3+ T cells by small-intestinal dendritic cells in patients with type 1 diabetes", DIABETES, vol. 60, 2011, pages 2120 - 4 |
BEAUDOIN LDIANA JGHAZARIAN L ET AL.: "Plasmacytoid dendritic cells license regulatory T cells, upon iNKT-cell stimulation, to prevent autoimmune diabetes", EUR J IMMUNOL, vol. 44, 2014, pages 1454 - 66 |
BENTLEY ET AL., NATURE, vol. 456, 2008, pages 53 - 59 |
BIBBIO; STEFANO ET AL: "Is there a role for gut microbiota in type 1 diabetes pathogenesis?", ANNALS OF MEDICINE, vol. 49, no. 1, 2 January 2017 (2017-01-02), SE, pages 11 - 22, XP055819273, ISSN: 0785-3890, DOI: 10.1080/07853890.2016.1222449 * |
CARRATU RSECONDULFO MDE MAGISTRIS L ET AL.: "Altered intestinal permeability to mannitol in diabetes mellitus type I", J PEDIATR GASTROENTEROL NUTR, vol. 28, 1999, pages 264 - 9, XP001562288, DOI: 10.1097/00005176-199903000-00010 |
CHASE DGERLANDSEN SL: "Evidence for a complex life cycle and endospore formation in the attached, filamentous, segmented bacterium from murine ileum", J BACTERIOL, vol. 127, 1976, pages 572 - 83 |
CHEN BO ET AL: "Presence of Segmented Filamentous Bacteria in Human Children and Its Potential Role in the Modulation of Human Gut Immunity", FRONTIERS IN MICROBIOLOGY, vol. 9, 29 January 2018 (2018-01-29), Lausanne, XP055819278, ISSN: 1664-302X, DOI: 10.3389/fmicb.2018.01403 * |
COLOMBEL JFSANDBORN WJREINISCH W ET AL.: "Infliximab, azathioprine, or combination therapy for Crohn's disease", N ENGL J MED, vol. 362, 2010, pages 1383 - 95 |
COLONNA M: "Interleukin-22-producing natural killer cells and lymphoid tissue inducer-like cells in mucosal immunity", IMMUNITY, vol. 31, 2009, pages 15 - 23, XP002674617, DOI: 10.1016/j.immuni.2009.06.008 |
DELONG TWILES TABAKER RL ET AL.: "Pathogenic CD4 T cells in type 1 diabetes recognize epitopes formed by peptide fusion", SCIENCE, vol. 351, 2016, pages 711 - 4, XP055476372, DOI: 10.1126/science.aad2791 |
DIANA JSIMONI YFURIO L ET AL.: "Crosstalk between neutrophils, B-la cells and plasmacytoid dendritic cells initiates autoimmune diabetes", NAT MED, vol. 19, 2013, pages 65 - 73 |
DUDAKOV JAHANASH AMVAN DEN BRINK MRM: "Interleukin-22: immunobiology and pathology", ANNU REV IMMUNOL, vol. 33, 2015, pages 747 - 85, XP055383956, DOI: 10.1146/annurev-immunol-032414-112123 |
DUNNE JL, TRIPLETT EW, GEVERS D: "The intestinal microbiome in type 1 diabetes", CLIN EXP IMMUNOL, vol. 177, 2014, pages 30 - 7, XP055515776, DOI: 10.1111/cei.12321 |
GABORIAU-ROUTHIAU VRAKOTOBE SLECUYER E ET AL.: "The key role of segmented filamentous bacteria in the coordinated maturation of gut helper T cell responses", IMMUNITY, vol. 31, 2009, pages 677 - 89, XP055005245, DOI: 10.1016/j.immuni.2009.08.020 |
GOTO YOBATA TKUNISAWA J ET AL.: "Innate lymphoid cells regulate intestinal epithelial cell glycosylation", SCIENCE, vol. 345, 2014, pages 1254009 |
GOTO YPANEA CNAKATO G ET AL.: "Segmented filamentous bacteria antigens presented by intestinal dendritic cells drive mucosal Thl7 cell differentiation", IMMUNITY, vol. 40, 2014, pages 594 - 607 |
GRAHAM SCOURTOIS PMALAISSE WJ ET AL.: "Enteropathy precedes type 1 diabetes in the BB rat", GUT, vol. 53, 2004, pages 1437 - 44 |
GREEN EAGORELIK LMCGREGOR CM ET AL.: "CD4+CD25+ T regulatory cells control anti-islet CD8+ T cells through TGF-beta-TGF-beta receptor interactions in type 1 diabetes", PROC NATL ACAD SCI U S A, vol. 100, 2003, pages 10878 - 83 |
HANSEN CHFKRYCH LBUSCHARD K ET AL.: "A maternal gluten-free diet reduces inflammation and diabetes incidence in the offspring of NOD mice", DIABETES, vol. 63, 2014, pages 2821 - 32 |
HARDIN JADONEGAN LWOODMAN RC ET AL.: "Mucosal inflammation in a genetic model of spontaneous type I diabetes mellitus", CAN J PHYSIOL PHARMACOL, vol. 80, 2002, pages 1064 - 70 |
HASNAIN SZBORG DJHARCOURT BE ET AL.: "Glycemic control in diabetes is restored by therapeutic manipulation of cytokines that regulate beta cell stress", NAT MED, vol. 20, 2014, pages 1417 - 26, XP055561932, DOI: 10.1038/nm.3705 |
HIGUCHI BRUNA STEVANATO ET AL: "Intestinal Dysbiosis in Autoimmune Diabetes Is Correlated With Poor Glycemic Control and Increased Interleukin-6: A Pilot Study", FRONTIERS IN IMMUNOLOGY, vol. 9, 1 January 2018 (2018-01-01), pages 1689, XP055819143, Retrieved from the Internet <URL:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6068285/pdf/fimmu-09-01689.pdf> DOI: 10.3389/fimmu.2018.01689 * |
HONEYMAN MCCOULSON BSSTONE NL ET AL.: "Association between rotavirus infection and pancreatic islet autoimmunity in children at risk of developing type 1 diabetes", DIABETES, vol. 49, 2000, pages 1319 - 24, XP002961465, DOI: 10.2337/diabetes.49.8.1319 |
HU YPENG JLI F ET AL.: "Evaluation of different mucosal microbiota leads to gut microbiota-based prediction of type 1 diabetes in NOD mice", SCI REP, vol. 8, 2018, pages 15451 |
IVANOV IIATARASHI KMANEL N ET AL.: "Induction of intestinal Th17 cells by segmented filamentous bacteria", CELL, vol. 139, 2009, pages 485 - 98, XP055005112, DOI: 10.1016/j.cell.2009.09.033 |
IVANOV IIMCKENZIE BSZHOU L ET AL.: "The orphan nuclear receptor RORgammat directs the differentiation program of proinflammatory IL-17+ T helper cells", CELL, vol. 126, 2006, pages 1121 - 33, XP008091324, DOI: 10.1016/j.cell.2006.07.035 |
JACOB COAISO SMICHIE SA ET AL.: "Prevention of diabetes in nonobese diabetic mice by tumor necrosis factor (TNF): similarities between TNF-alpha and interleukin 1", PROC NATL ACAD SCI U S A, vol. 87, 1990, pages 968 - 72 |
JACOB COAISO SSCHREIBER RD ET AL.: "Monoclonal anti-tumor necrosis factor antibody renders non-obese diabetic mice hypersensitive to irradiation and enhances insulitis development", INT IMMUNOL, vol. 4, 1992, pages 611 - 4 |
KATZ JD, BENOIST C, MATHIS D: "T helper cell subsets in insulin-dependent diabetes", SCIENCE, vol. 268, 1995, pages 1185 - 8, XP002676462, DOI: 10.1126/science.7761837 |
KOSTIC ADGEVERS DSILJANDER H ET AL.: "The dynamics of the human infant gut microbiome in development and in progression toward type 1 diabetes", CELL HOST MICROBE, vol. 17, 2015, pages 260 - 73, XP029139280, DOI: 10.1016/j.chom.2015.01.001 |
KRIEGEL M. A. ET AL: "Naturally transmitted segmented filamentous bacteria segregate with diabetes protection in nonobese diabetic mice", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, vol. 108, no. 28, 12 July 2011 (2011-07-12), pages 11548 - 11553, XP055819125, ISSN: 0027-8424, Retrieved from the Internet <URL:http://dx.doi.org/10.1073/pnas.1108924108> DOI: 10.1073/pnas.1108924108 * |
KRIEGEL MASEFIK EHILL JA ET AL.: "Naturally transmitted segmented filamentous bacteria segregate with diabetes protection in nonobese diabetic mice", PROC NATL ACAD SCI, vol. 108, 2011, pages 11548 - 53, XP055819125, DOI: 10.1073/pnas.1108924108 |
KUITUNEN MSAUKKONEN TILONEN J ET AL.: "Intestinal permeability to mannitol and lactulose in children with type 1 diabetes with the HLA-DQB 1 02 allele", AUTOIMMUNITY, vol. 35, 2002, pages 365 - 8 |
L. MASTRANDREA ET AL: "Etanercept Treatment in Children With New-Onset Type 1 Diabetes: Pilot randomized, placebo-controlled, double-blind study", DIABETES CARE, vol. 32, no. 7, 1 July 2009 (2009-07-01), US, pages 1244 - 1249, XP055609403, ISSN: 0149-5992, DOI: 10.2337/dc09-0054 * |
LECUYER ERAKOTOBE SLENGLINE-GARNIER H ET AL.: "Segmented filamentous bacterium uses secondary and tertiary lymphoid tissues to induce gut IgA and specific T helper 17 cell responses", IMMUNITY, vol. 40, 2014, pages 608 - 20 |
LEE ASGIBSON DLZHANG Y ET AL.: "Gut barrier disruption by an enteric bacterial pathogen accelerates insulitis in NOD mice", DIABETOLOGIA, vol. 53, 2010, pages 741 - 8, XP019797099 |
LEE JSTATO CMJOYCE-SHAIKH B ET AL.: "Interleukin-23-Independent IL-17 Production Regulates Intestinal Epithelial Permeability", IMMUNITY, vol. 43, 2015, pages 727 - 38 |
LEEDS JSHOPPER ADHADJIVASSILIOU M ET AL.: "Inflammatory bowel disease is more common in type 1 diabetes mellitus", GUT, vol. 60, 2011, pages A208 - A208 |
LEHUEN ADIANA JZACCONE P ET AL.: "Immune cell crosstalk in type 1 diabetes", NAT REV IMMUNOL, vol. 10, 2010, pages 501 - 13, XP055102677, DOI: 10.1038/nri2787 |
LEITER EH: "Multiple low-dose streptozotocin-induced hyperglycemia and insulitis in C57BL mice: influence of inbred background, sex, and thymus", PROC NATL ACAD SCI U S A, vol. 79, 1982, pages 630 - 4 |
LIVANOS AEGREINER TUVANGAY P ET AL.: "Antibiotic-mediated gut microbiome perturbation accelerates development of type 1 diabetes in mice", NAT MICROBIOL, vol. 1, 2016, pages 16140 |
MATHIEU, C.LAHESMAA, R.BONIFACIO, E. ET AL.: "Immunological biomarkers for the development and progression of type 1 diabetes", DIABETOLOGIA, vol. 61, 2018, pages 2252 - 2258, XP036883239, DOI: 10.1007/s00125-018-4726-8 |
MEDDINGS JBJARAND JURBANSKI SJ ET AL.: "Increased gastrointestinal permeability is an early lesion in the spontaneously diabetic BB rat", AM J PHYSIOL, vol. 276, 1999, pages G951 - 957 |
MIRANDA MCGOLIVEIRA RPTORRES L ET AL.: "Frontline Science: Abnormalities in the gut mucosa of non-obese diabetic mice precede the onset of type 1 diabetes", J LEUKOC BIOL, vol. 106, 2019, pages 513 - 29 |
OMENETTI SBUSSI CMETIDJI A ET AL.: "The Intestine Harbors Functionally Distinct Homeostatic Tissue-Resident and Inflammatory Thl7 Cells", IMMUNITY, June 2019 (2019-06-01) |
PELLEGRINI SSORDI VBOLLA AM ET AL.: "Duodenal Mucosa of Patients With Type 1 Diabetes Shows Distinctive Inflammatory Profile and Microbiota", J CLIN ENDOCRINOL METAB, vol. 102, 2017, pages 1468 - 77 |
PICKARD JMMAURICE CFKINNEBREW MA ET AL.: "Rapid fucosylation of intestinal epithelium sustains host-commensal symbiosis in sickness", NATURE, vol. 514, 2014, pages 638 - 41, XP037437643, DOI: 10.1038/nature13823 |
QUATTRIN TERESA ET AL: "Golimumab and Beta-Cell Function in Youth with New-Onset Type 1 Diabetes", THE NEW ENGLAND JOURNAL OF MEDICINE, vol. 383, no. 21, 19 November 2020 (2020-11-19), US, pages 2007 - 2017, XP055819133, ISSN: 0028-4793, DOI: 10.1056/NEJMoa2006136 * |
REWERS MEISENBARTH GS: "Celiac disease in TIDM-the need to look long term", NAT REV ENDOCRINOL, vol. 8, 2011, pages 5 - 8 |
ROUXEL ODA SILVA JBEAUDOIN L ET AL.: "Cytotoxic and regulatory roles of mucosal-associated invariant T cells in type 1 diabetes", NAT IMMUNOL, vol. 18, 2017, pages 1321 - 31 |
RUTZ SWANG XOUYANG W: "The IL-20 subfamily of cytokines--from host defence to tissue homeostasis", NAT REV IMMUNOL, vol. 14, 2014, pages 783 - 95, XP055288533, DOI: 10.1038/nri3766 |
SAPONE ADE MAGISTRIS LPIETZAK M ET AL.: "Zonulin upregulation is associated with increased gut permeability in subjects with type 1 diabetes and their relatives", DIABETES, vol. 55, 2006, pages 1443 - 9, XP055257188, DOI: 10.2337/db05-1593 |
SCHAFFER LBRAND CLHANSEN BF ET AL.: "A novel high-affinity peptide antagonist to the insulin receptor", BIOCHEM BIOPHYS RES COMMUN, vol. 376, 2008, pages 380 - 3, XP025495743, DOI: 10.1016/j.bbrc.2008.08.151 |
SCHNUPF PGABORIAU-ROUTHIAU VGROS M ET AL.: "Growth and host interaction of mouse segmented filamentous bacteria in vitro", NATURE, vol. 520, 2015, pages 99 - 103, XP037439790, DOI: 10.1038/nature14027 |
SHENDUREJI: "Next-generation DNA sequencing", NATURE BIOTECHNOLOGY, vol. 26, no. 10, 2008, pages 1135 - 1145 |
SHI ZZOU JZHANG Z ET AL.: "Segmented Filamentous Bacteria Prevent and Cure Rotavirus Infection", CELL, vol. 179, 2019, pages 644 - 658 |
TAI NINGWEN ET AL: "The role of gut microbiota in the development of type 1, type 2 diabetes mellitus and obesity", REVIEWS IN ENDOCRINE & METABOLIC DISORDERS, NEW YORK, NY : SPRINGER, US, vol. 16, no. 1, 27 January 2015 (2015-01-27), pages 55 - 65, XP035460199, ISSN: 1389-9155, [retrieved on 20150127], DOI: 10.1007/S11154-015-9309-0 * |
THAISS CALEVY MGROSHEVA I ET AL.: "Hyperglycemia drives intestinal barrier dysfunction and risk for enteric infection", SCIENCE, vol. 359, 2018, pages 1376 - 83 |
TIMPER KHRUZ PBEGLINGER C ET AL.: "Infliximab in the treatment of Crohn disease and type 1 diabetes", DIABETES CARE, vol. 36, 2013, pages e90 - 91, XP002793259, DOI: 10.2337/dc13-0199 |
VAARALA O: "Leaking gut in type 1 diabetes", CURR OPIN GASTROENTEROL, vol. 24, 2008, pages 701 - 6 |
WESTERHOLM-ORMIO MVAARALA OPIHKALA P ET AL.: "Immunologic activity in the small intestinal mucosa of pediatric patients with type 1 diabetes", DIABETES, vol. 52, 2003, pages 2287 - 95 |
WINER DAWINER SDRANSE HJ ET AL.: "Immunologic impact of the intestine in metabolic disease", J CLIN INVEST, vol. 127, 2017, pages 33 - 42 |
YANG XDTISCH RSINGER SM ET AL.: "Effect of tumor necrosis factor alpha on insulin-dependent diabetes mellitus in NOD mice. I. The early development of autoimmunity and the diabetogenic process", J EXP MED, vol. 180, 1994, pages 995 - 1004, XP002651733 |
YU HGAGLIANI NISHIGAME H ET AL.: "Intestinal type 1 regulatory T cells migrate to periphery to suppress diabetogenic T cells and prevent diabetes development", PROC NATL ACAD SCI U S A, vol. 114, 2017, pages 10443 - 8 |
YURKOVETSKIY LBURROWS MKHAN AA ET AL.: "Gender bias in autoimmunity is influenced by microbiota", IMMUNITY, vol. 39, 2013, pages 400 - 12, XP055209575, DOI: 10.1016/j.immuni.2013.08.013 |
ZHENG YVALDEZ PADANILENKO DM ET AL.: "Interleukin-22 mediates early host defense against attaching and effacing bacterial pathogens", NAT MED, vol. 14, 2008, pages 282 - 9 |
ZHOU HE ET AL: "Evaluating the Causal Role of Gut Microbiota in Type 1 Diabetes and Its Possible Pathogenic Mechanisms", FRONTIERS IN ENDOCRINOLOGY, vol. 11, 24 March 2020 (2020-03-24), CH, XP055819255, ISSN: 1664-2392, Retrieved from the Internet <URL:https://www.ncbi.nlm.nih.gov/pmc/ivip/1664-2392> DOI: 10.3389/fendo.2020.00125 * |
ZHOU HE ET AL: "Gut Microbiota Profile in Patients with Type 1 Diabetes Based on 16S rRNA Gene Sequencing: A Systematic Review", DISEASE MARKERS, vol. 2020, 28 August 2020 (2020-08-28), GB, pages 1 - 11, XP055819263, ISSN: 0278-0240, Retrieved from the Internet <URL:http://downloads.hindawi.com/journals/dm/2020/3936247.xml> DOI: 10.1155/2020/3936247 * |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US11033588B2 (en) | Compositions for treating inflammation and uses thereof | |
Fei et al. | Endotoxin producers overgrowing in human gut microbiota as the causative agents for nonalcoholic fatty liver disease | |
JP6201982B2 (en) | Diabetes-inducing bacteria | |
Matricon et al. | Immunopathogenesis of inflammatory bowel disease | |
US20160120915A1 (en) | Methods for manipulating immune responses by altering microbiota | |
Jamwal et al. | Intestinal epithelial expression of MHCII determines severity of chemical, T-cell–induced, and infectious colitis in mice | |
Braun et al. | Body traffic: ecology, genetics, and immunity in inflammatory bowel disease | |
Gu et al. | Fusobacterium nucleatum causes microbial dysbiosis and exacerbates visceral hypersensitivity in a colonization-independent manner | |
US10736927B2 (en) | Pharmaceutical compositions for preventing or treating inflammatory bowel diseases | |
Rouland et al. | Gut mucosa alterations and loss of segmented filamentous bacteria in type 1 diabetes are associated with inflammation rather than hyperglycaemia | |
Anderson et al. | NOD2 influences trajectories of intestinal microbiota recovery after antibiotic perturbation | |
Chen et al. | The intracellular innate immune sensor NLRP12 attenuates colon inflammation by maintaining colonic microbial diversity and promoting protective commensal bacterial growth | |
Qiu et al. | Latent autoimmune diabetes in adults in China | |
WO2016002827A1 (en) | Therapeutic agent for progressive demyelinating immune disease | |
EP4011394A1 (en) | Method for treating alzheimer's disease by inhibiting uptake of amino acids by t cells | |
AU2016361499B2 (en) | Methods and compositions for identifying and treating subjects at risk for checkpoint blockade therapy associated colitis | |
WO2022171685A1 (en) | Tnf-alpha blocking agent for preventing, suppressing and/or delaying the onset of type 1 diabetes | |
WO2020227420A1 (en) | Monitoring and altering the gut microbiome in disease | |
EP3167892A1 (en) | Methods and pharmaceutical compositions for preventing or treating imflammatory bowel diseases | |
Lin et al. | Akkermansia muciniphila-mediated degradation of host mucin expands the tryptophan utilizer alistipes and exacerbates autoimmunity by promoting Th17 immune responses | |
Lin et al. | Influence of TRAIL deficiency on Th17 cells and colonic microbiota in experimental colitis mouse model | |
US9885089B2 (en) | Flagellum and anti-flagellum antibody levels in samples as a diagnostic biomarker and therapeutic target for metabolic syndrome | |
US20230127277A1 (en) | Microbiome-based therapeutics | |
WO2021221110A1 (en) | Small intestinal bacterium inducing proliferation or activation of th1 cells and/or th17 cells | |
McDaniel et al. | Maternal Influence and Murine Housing Confound Impact of NLRP1 Inflammasome on Microbiome Composition |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 22705387 Country of ref document: EP Kind code of ref document: A1 |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
122 | Ep: pct application non-entry in european phase |
Ref document number: 22705387 Country of ref document: EP Kind code of ref document: A1 |