Recent Advances in Gastroenterology-14 Her Hsin Tsai
INDEX
Page numbers followed by b refer to box, f refer to figure, fc refer to flowchart, and t refer to table.
A
Acetaminophen 240
Acquired hepatocerebral degeneration 248
Acute closed-angle glaucoma 169
Acute hepatitis C 226
infection 226
treatment of 229
Acute respiratory distress syndrome 10, 261
Acute severe colitis 20
management of 20
Adalimumab 1, 9, 41
Adaptive arms 70
Adenoma 177
carcinoma sequence 167
colonic 163f
detection 171, 173, 174
hepatocellular 247
Adhesion molecules 67
Adrenocorticosteroids 60
Akkermansia muciniphila 90
Alanine
amino transferase 198
transaminase 227
Alcohol 238
consumption, substantial 250
Allergic reactions 65
Alzheimer's disease 251
American College of Gastroenterology 132
American Gastroenterology Association 39
American Society of Gastrointestinal Endoscopy 37
Amiselimod 14
Ammonia 250
Amnesia, mild 245
Amoxicillin 108, 112, 113, 119, 120t, 121, 122t, 123125, 127, 128
Amphetamines 240
Anabolic steroids 240
Anaerobic luminal bacteria 89
Anal canal 25
Anastomosis 24
Ancylostoma caninum 57
Anemia 4
Angiotensin receptor blockers 31
Angiotensin-converting enzyme inhibitors 31
Anorexia 229, 244
Anti-adhesion agents 7
Antibiotics 20
side effects 134
Anti-cytotoxic T-lymphocyte-associated protein 4 32
Antidepressants 240
Anti-infliximab antibody 6
Anti-interleukin inhibitors 9
Antimitochondrial antibodies 34
Antineutrophil cytoplasmic antibodies 245
Antinuclear antibodies 34
Anti-programmed cell death
ligand 1 32
protein 1 32
Antiretroviral medications 235
Antithyroid antibodie 34
Anti-TNFα therapy 41
Antiviral therapy 233
Apoptotic cells 73f
Arthralgia 244
Artificial intelligence 151, 152, 153f, 158, 159, 177
application of 155
studies of 160t
terminology 152
use of 177
Artificial neural networks 152, 153, 154f
Ascaris 57
Ascitic tap 52
Aspartate aminotransferase 200, 229
Aspirin 32, 207
Asthma 50
Astrocyte swelling 240
Autoimmune diseases 31
Autoimmune disorders 31, 245
Autoimmune liver disease 239, 252
types of 252
Autonomic neuronal dysfunction 244
Azathioprine 1, 5, 5f, 6, 41, 61
metabolism 5f
B
B lymphocytes 225
Bacterial biofilm 97
Bacterial infectious diseases 107
Bacteroides
fragilis 90
thetaiotaomicron 90
vulgatus 90
Bariatric surgery 199
Barrett's esophagus 159
Barrett's mucosa 159
Basidiobolomycosis 57
Bat-borne virus 261
Beclomethasone dipropionate 39
Beta-blockers 31, 32
Bifidobacterium
animalis 40
breve 90
longum 90
strains 126
Bile acid 86
absorption, abnormal 30
malabsorption 39, 40
Bile duct cannulation 188
Biological therapy, effect of 22
Biopsy 57t, 121, 126
colonic 21, 42
mucosal 52
Bismuth
based regimen 123
levofloxacin-amoxicillin treatment 124
quadruple therapy 115
salicylate 39
Bleeding 21
severe colonic 21
Blood
eosinophilia 74
investigations 34
and stool sampling 34
vessels, superficial 173
Blue laser imaging 173
Body mass index 198
Bone marrow 65
Bowel cancer 265, 266
Brain 58
ammonia 253
cells 240
edema 240
herniation 240
Budesonide 39, 40, 61
Buscopan 169
use of 178
C
Calcinosis 31
Campylobacter 33
jejuni 242
Canagliflozin 203
Cancer 10, 199
cells 76
Capillaria 57
Carbohydrates 59, 197
Carcinogenesis 96
Carcinoma
colonic 21
mucinous 96
Cecal intubation rate 167
Celiac disease, screening for 39
Cells
preservation of 95
sensitive, structure of 154
types of 13
Cenicriviroc 205207
Central nervous system 8, 239, 242
Cerebral edema 240
Cerebrospinal fluid 242
Chemokines 67
Chemotherapeutic drugs 248
Cholangiocarcinoma 247
Cholangiography, percutaneous transhepatic 185
Cholangiopancreatography, endoscopic retrograde 184
Cholangitis, primary sclerosing 21
Cholestatic liver
diseases 248
function 184
Cholesterol 198, 205
Cholestyramine 39, 40
Chromoendoscopy 171, 178
conventional 171
dye-based 171
virtual 171
Chronic hepatitis
C 223, 227, 228
infection 227
treatment of 230
prevention of 196
Churg-Strauss syndrome 58
Cirrhosis 201, 229, 232
compensated 232
primary biliary 252
Clarithromycin 108, 109, 112114, 120t, 121, 123, 125, 127
resistance 109, 125
Closed-angle glaucoma 169
Clostridium difficile 30, 33
Coagulation, abnormal 184
Cocaine 240
Coeliac disease 30
Colectomy 21, 23
risk of 20
subtotal 23
Colitis
collagenous 29, 32, 36
eosinophilic 49, 54f
Colon 24, 87f
cancer 10
removal of 24
Colonic mucosal patterns, visualization of 171
Colonocytes 88
exfoliated 91
Colonoscopes, standard 176
Colonoscopy 167, 172
cap-assisted 174, 174f
high-definition 170
risks of 35
technology 170
water-assisted 170
wide-angle 176
Colorectal cancer 71, 74, 77f, 86, 95, 167
risk of 21
Colorectal carcinoma
cell 75
prevention of 167
Colorectal mucus 73f, 92, 95f, 97f
Colorectal neoplasia 161
Comorbid disease, exacerbation of 186
Concomitant therapy 113
Confocal laser endomicroscopy 36
Consciousness 244
Conventional therapy 248
Convolutional neural networks 152, 153
Coriobacteriaceae 30
Coronavirus 261
disease 261
strain of 261
Corticosteroids 60
Cotton's consensus 185
COVID-19 on
delivery of gastrointestinal services, effect of 265
gastroenterology, impact of 261
gastrointestinal endoscopy, effect of 264
patients with pre-existing gastrointestinal diseases, effect of 265
Crohn's disease 11, 30, 71, 92
Cryoglobulinemia 245
Cryoglobulins 245
Cryptitis 37
Cytokines 10, 12, 67, 72, 253
proinflammatory 61
receptors 11
Cytomegalovirus 241
Cytotoxic cationic proteins 65
Cytotoxic T-cells 74
D
Damage-associated molecular patterns 75, 77
Dapagliflozin 204
Deep learning 154f, 155f
Deep mucosal biopsies 52
Degranulation 77
Delirium 240
Dendritic cells 74
Desulfovibrio desulfuricans 90
Detoxification 238
Device-assisted colonoscopy 174
Diabetes mellitus
treatment of 200
type 1 31
type 2 197, 202, 251
Diarrhea 57, 58, 244, 263
bloody 1
chronic nonbloody watery 33
functional 33
treatment of 30
watery nonbloody 29
Digestive symptoms 264
Diminutive adenomas, diagnosis of 178
Dipeptidyl peptidase-4 inhibitors 202
Diplopia 246
Direct-acting antivirals 228
development of 196
different classes of 230fc
Disease flare, indicator of 3
Double-wire technique 189
Duodenal involvement 54f
Dysbiosis 92
Dysmotility 50
Dyspepsia 131
functional 131
Dysphagia 49, 50
Dysplasia 21, 22
high-grade 21, 22
low-grade 21, 22
Dysreflexia, autonomic 240
Dysregulations, transcriptomic 93
E
Early gastric cancer 131
detection of 159
Eczema 50
Elafibranor 205
Elbasvir 234
Electrolyte solutions 190
Electronic chromoendoscop 35
Elemental diet 59
Encephalomyelitis 244
Encephalopathic syndromes 245
Encephalopathy
acute hepatic 240
chronic hepatic 241
hepatic 227, 238, 239, 241
multi-infarct 245
portal-systemic 239, 241
Endocuff 175, 175f
Endorings 175
Endoscopic bile duct balloon dilatation 189
Endoscopic mucosal resolution 15
Endoscopic ultrasound 188
Endoscopy 52, 109
lower gastrointestinal 161
stack 164
white-light 172
English Bowel Cancer Screening Programme 169
Enteritis 49
Enterocytes 88
Enteroendocrine cells 88
Enzyme 88
activity 5
immunoassay 226
Eosin stain staining 55
Eosinophilic gastroenteritis 49, 59
classification 49
clinical
assessment 50
course 58
features 50
diagnosis 55
differential diagnosis 55
epidemiology 50
natural history 58
pathogenesis 50
small bowel involvement of 55f
Eosinophilic gastrointestinal diseases 59, 62
researcher 58
Eosinophilic gastrointestinal disorders 49, 50
Eosinophilopoiesis 66f
Eosinophils 55, 60, 6567, 7072, 73f, 74, 94, 97
constitute 65, 70
cut-off levels of 57t
functions of 68
human 65
maturation 65
migration of 67
secretory functions of 67
Epithelial barriers 65
Epithelial cell 68
colonic 88
Epithelial surface 91
Epithelium 72
intestinal 91
Epstein-Barr virus 241
Erythema 35
Erythrocytes 73f
Escherichia coli 40, 97
Esomeprazole 32, 121
Esophageal cancer, diagnosis of 159
Esophageal disease 49
Esophageal dysmotility 31
Esophageal tissue 263
Esophagitis, eosinophilic 49
Esophagogastroduodenoscopy 36
Esophagus 49
Estimated glomerular filtration rate 234
Etrasimod 14
efficacy of 14
Etrolizumab 9
European Association for Study of Liver 207
European Helicobacter and Microbiota Study Group 109
European Microscopic Colitis Group 40
Exogenous hormone therapy 30
Extracellular DNA trap formation 77, 97
Eyes 51
F
Fatigue 229
central 252
peripheral 252
Fatty acids 250
Fatty liver disease, nonalcoholic 196, 206, 207, 215, 216, 238, 239, 250, 251, 265
Fecal immunochemical test 176
Fecal transplant 42
Fetal scalp monitors 233
Fever 4
low-grade 229
Fibrosis 229, 238
advanced 215f
regression 198
Flexible spectral imaging color enhancement 173
Fluoroquinolones 127
quadruple therapy 123
Focal liver lesion detection 157
Focal nodular hyperplasia 247
Food allergies 50
Fuji intelligent chromoendoscopy 35
Fujifilm endoscopes 173
Full-spectrum endoscopy 177
Fusobacterium nucleatum 97
G
Galactosamine 240
Gamma
activated sequence 11
activation factor 11
glutamyl transferase 203
Gastric
antral mass 58
cancer
endoscopic detection of 160t
infiltrative 57
epithelium 123
hydrochloric acid 86
malignancy 164
diagnosis of 159
obstruction 51
Gastritis, eosinophilic 49, 53f
Gastroenterology 151
Gastrointestinal
bleeding 58, 238
conditions, group of 49
diseases 33
endoscopy 159
mucus structure 87f
physicians 151
physiology 98
radiology 155
symptoms 263
tract 86
Generic biosimilars 2
G-eye colonoscopy 177
Glucagon-like peptide-1 200, 207
receptor agonists 202
Goblet cell 88, 89, 97
adjacent 89
depletion 96
development 69
mucus-producing 97
Granulocyte
leukocytes 65
macrophage colony-stimulating factor 66
Grasp reflexes 249
Grazoprevir 234
Guanidine monophosphate synthetase 5
Guanosine monophosphate synthetase 5
Guillain-Barré syndrome 242
Gut bacteria 88
Gut immune system, maintenance of 70
Gut inflammation, mediators of 1
Gut microbiota 94
Gut mucus 86, 8990, 98
H
H2 receptor antagonist 32
Hallucinogens 240
Healthy liver 238
Heart 58
disease 199
failure, worsening 204
Helicobacter pylori 106, 108, 111, 116, 134
antibiotic resistance 127
clarithromycin resistance 127
cure rate 107
eradication 108, 124, 126, 128131, 132t, 133
failure 123, 127
regimens 115, 117, 129
therapy 108, 126, 131, 134
treatment 116, 126
gastritis 107
growth of 124
infection 106110, 112, 113, 119, 120t, 122125, 127, 133, 134
after treatment 133
cure of 130
eradication of 122t
risks of 129
treatment of 106b, 106t, 107, 123, 125, 133
isolates 113
regimens 110
related disease 131
resistance of 123
strains 114
therapy 107, 129, 130
treatment 110, 111, 129, 131, 132
failure 134
testing after 132
Hemangioma, hepatic 247
Hemianopsia 244
Hemiparesis 244
Hemodialysis 226
Hemorrhage 10
Hepatic coma 240
Hepatic failure, fulminant 240
Hepatic fat accumulation 197
Hepatic tissue 238
Hepatitis 238, 240, 241
A 239, 241, 242
virus 241
autoimmune 252, 265
B 239, 243
chronic 244
complications of 243
surface antigen 243
virus 243
C 227, 234, 239, 244
diagnosis 227
during pregnancy, treatment of 232
epidemiology 223
management of 223
pathogenesis 224
therapeutics 231
treatment 229, 233, 234
viremia 227
virus infection 196, 223, 224f, 225, 225f, 226fc, 227, 229, 230, 234, 244, 246
virus infection 223
Hepatobiliary-pancreatic disorders 184
Hepatocellular carcinoma 196, 226
development of 204
Hepatopathy, congestive 238
Hepatorenal syndrome 227
Heroin 240
Herpes simplex virus 241
Homeostasis, immunological 238
Hormonal balance 238
Host cell exfoliation 91
Human immunodeficiency virus 234, 244
Human intestinal tract 65
Human leukocyte antigen 30
Hybrid therapy 115
Hydration, intravenous 190
Hypereosinophilic syndrome 57
Hyperplastic polyps 167, 177
types of 167
Hypertension 198
I
Idiopathic noncirrhotic portal 247
Ileal pouch 24
anal anastomosis 22, 24
formation 24
procedures 23
Ileostomy 23, 24
defunctioning 23, 24
permanent 22, 25
Immune
cells 97
transepithelial migration of 74
checkpoint inhibitor 31, 32, 36
complexes 243
system, activation of 29
Immunomodulators 41
Infertility, risk of 24
Inflammatory bowel disease 6, 19, 31, 57, 71, 86, 92, 266
prevalence of 1
surgery for 23
Inflammatory bowel disorder 29
Inflammatory cells 37, 247
abundance of 95f
Inflammatory conditions, treatment of 12
Infliximab 1, 15, 41
monotherapy 6
plus azathioprine 6
serum 6
Innate arms 70
Inner mucus layer 87, 94
Insulin 198
glucose-dependent 200
Interferon 11
regulatory factor 11
sensitivity determining region 225
stimulated response element 11
Intestinal microbiome, role of 206
Intestinal mucosal barrier, functionality of 68
Intestinal protective barrier, component of 88
Intestine, small 87f
Intracellular accumulation 240
Intracellular tight junctions 88
Ipragliflozin 203
group 204
Irritable bowel syndrome 2
J
Jak-stat pathway 11f
Jaundice 184, 242
John Cunningham virus 8
K
Kidney 58
disease 234
L
Lactobacillus
acidophilus 40
johnsonii 90
plantarum 90
reuteri 90
rhamnosus 90
strains 126
Lamina propria 30, 37, 6770, 75, 89
cellularity 35
intestinal 88
Langerhans cell histiocytosis 58
Lansoprazole 32, 121
Laparoscopy 52
Leaving scar tissue 238
Leukoencephalitis 244
Leukoencephalopathy, progressive multifocal 8
Levofloxacin 122t, 123
bismuth quadruple 124
resistant strains 123
therapy 122
Linitis plastica 52, 57
Lipolytic digestive enzymes 86
Lipoprotein, low-density 205
Liraglutide 200, 207
Liver
biopsy 198, 203
cancer, primary 223
cirrhosis, alcoholic 239, 265
conditions, chronic 266
disease 202, 228, 238, 239, 239t, 249, 253
end-stage 201, 238
neurological complications in 238
stage of 238
enzyme tests 264
failure 238
acute 239
chronic 239, 253
fibrosis 228
stages 205
function 264
abnormal 248
lesions 156
diffuse 158
toxins 240
transplantation 238
Low-birth weight infants 232
Lungs 58
Lymph glands 51
Lymph nodes 13
Lymphocytes 8, 30
intraepithelial 37
Lymphocytic colitis 29, 32, 36
Lymphoid cells 68
innate 70
Lymphoid organs 13
Lymphoid tissue
gut-associated 69
mucosa-associated 131
Lymphoma 131
M
Maastricht consensus report 132
Machine learning 152, 153, 154f, 155f
Macrophages 73f, 74, 94, 97
Major colorectal diseases 65, 70
Malaise 245
Malignant cells, exfoliated 97
Malignant diseases, treatment of 157
Malignant lesions, chemotherapy of 157
Mature eosinophils
functional characteristics of 65
migration of 66f
structural characteristics of 65
Mayo score 2
Medical management, failure of 21
Menopausal hormone therapy 30
Mepolizumab 62
Mercaptans 250
Mercaptopurine 1, 5
Mesalamine 39, 40
Metabolic syndrome 196, 200
Metalloproteinase 30, 88
Metastatic disease 247
Metformin 200, 207
Methotrexate 41
Methylmercaptopurine 5
Methylthioinosine monophosphate 5
Metronidazole 110, 111, 114, 116, 123, 125128
Microbiome 90
manipulation 206
Microscopic colitis 29, 30, 33, 34t, 36t, 42
antibiotics 40
clinical features 33
development of 32t
diagnosis 34
disease activity index 38, 38t
environmental factors 31
epidemiology 29
lifestyle modification 38
management 38
medications 39
pathophysiology 29
prescribed medications 31
probiotics 40
risk factors 33
surgery 42
Migration 91
Modern health delivery system 158
Mononeuropathy multiplex 243
Motor axonal neuropathy, acute 242
Mucosa 52, 95, 171, 173
colonic 29
Mucosal Addressin-cell adhesion molecule 8
Multidisciplinary team 19
part of 19, 158
Multidrug-resistant transporter genes 118
Multifactorial disease 74
Multi-light technology 173
Multiorgan failure 238
Myelopathy, hepatic 239, 249, 250
Myoclonus 248, 249
Myopathies
inflammatory 246
noninflammatory 246
N
N-acetyl-aspartate 246
N-acetyl-aspartyl-glutamate concentrations 246
Narrow-band imaging 160162, 163f, 172
Natalizumab 8
Nausea 229, 244
Neoplasm 238, 239, 247, 253
Neoplastic lesions 159
Neurotoxicity, risk of 40
Neutrophils 67, 73f, 94, 95f, 97
derived etosis 97
extracellular traps 76
Nitroglycerine, transdermal 190
Nitroimidazole 112, 113, 123
Nonbismuth quadruple sequential and concomitant regimens 123
Non-nucleoside inhibitor 230
Nonsteroidal anti-inflammatory drugs 31, 32, 34, 190
Non-Wilsonian acquired hepatocerebral degeneration 239, 248
Nucleoside inhibitor 230
Nucleotide inhibitor 230
Nystagmus 249
O
Obesity, complications of 216
Obeticholic acid 205, 207
Oddi dysfunction, sphincter of 184
Oddi pressure 190
Olympus medical systems 176
Omalizumab 62
Omeprazole 32, 118, 121, 125
Optimized 5-ASA therapies 4
Optimized immunosuppressive therapies 4
Optimizing anti-TNF treatments 5
Optimizing conventional therapies 2
Oral contraceptive pill 30
Organ failure 186
Orlistat 200
Ozanimod 14
P
Pain, abdominal 57, 58, 229, 244
Painful red eye 169
Pancreatic duct 188
stenting 189
Pancreatic inflammation 186
Pancreatitis Across Nations Clinical Research and Education Alliance 186
Pancreatogram, endoscopic 184
Paneth cells 88
Panproctocolectomy 23
Pantoprazole 32, 121
Parasitic infestations 51, 57
Parvimonas micra 97
Pediatric obesity 215
Pelvic
dissection 24
sepsis 25
Penicillin allergy 125
Pentoxifylline 41
Peptic ulcer 130
Peptostreptococcus stomatis 97
Peroxisome proliferator-activated receptor 201, 207
Pioglitazone 201
Polyarteritis nodosa 58, 243
Polymerase chain reaction 228
Polyneuropathy, chronic relapsing 243
Polypeptide, glucose-dependent insulinotropic 202
Polyps
adenomatous 167
colonic 162t
detection of 161, 167, 174
Portal hypertension, presence of 228
Positive predictive value 159, 160
Positron emission tomography 158
Post-endoscopic retrograde cholangiopancreatography pancreatitis 184, 185
Pouch
function 24
procedure 23
vaginal fistula 24
Precut sphincterotomies 189
Proctectomy, completion 23, 25
Proctocolectomy 23, 25
Prophylactic pancreatic stent 189
Prostate cancer 10
Protein
nonstructural 225, 230
tyrosine kinase family 11
Proteolytic granzyme 76
Proton-pump inhibitor 31, 32, 107, 120t, 230
combination of 122t
dose of 117
Proximal colon retroflexion 169
Pruritus 242
Pseudotumor, inflammatory 247
Psychomotor, dysfunctional 244
Ptosis, severe 246
Pylorus, thickened 53f
Pyramidal tract signs 248
Q
Quadruple regimen 125
Quinolone 117, 127
R
Rabeprazole 121
Rapid cell death 67
Rapid mucus renewal 88
Raynaud's phenomenon 31
Rectal cancer 10
Rectal malignancy, risk of 23
Rectal stump
dehiscence, risk of 23
management of 23
Rectal tumors, low 24
Rectum 24, 25
removal of 24
Red blood cells 37
Reflux 50
Renal papillary cancer 10
Reslizumab 62
Retroflexion 169
Rheumatoid
arthritis 31
factor 34
Rhinitis 50
Ribonucleic acid 223, 226, 244
Rifabutin 119
therapy 119, 124
Rosiglitazone 201
Ruminococcus
gnavus 90
torques 90
S
Saccharomyces boulardii 126
Sanger sequencing 228
SARS-CoV-2
on gastrointestinal tract, effect of 263
structure of 261
to host cell, attachment of 262f
Saturated fats 198
Scar tissue 238
Sclerodactyly 31
Seizures 240
Selective serotonin reuptake inhibitors 31, 32, 34
Selonsertib 205, 207
Sensory axonal neuropathy 242
Sentinel cells 89
Serotonin-norepinephrine reuptake inhibitors 34
Sertraline 32
Serum fasting glucose 203
Sessile serrated polyps 167
Sigmoid eosinophilic colitis, colonoscopic appearance of 54f
Sjogren's syndrome 31
Skin 51
cancers, nonmelanoma 10
Small bowel obstruction 24
Sodium
cromoglycate 61
glucose cotransporter 203
Somatostatin 190
Spectrum bias 152
Sphincteroplasty 189
pancreatic 189
Sphincters 24
Sphingosine 1-phosphate 12, 13
receptor modulators 12, 13f
Sphingosine kinase 13
Sphingosine lyase 13
Standard six-food elimination diet 59
Standard triple therapy, efficacy of 120t
Statins 31, 204, 207
Steatohepatitis 251
nonalcoholic 196, 207, 251
Steroids 20, 25, 39, 42
dependency 21
long-term 21
Stiff nonpliable mucosa 35
Stoma
management of 19
permanent 24
Stomach 87f, 159
marked antral thickening of 53f
Stone diseases, surgery for 184
Stool urgency 33
Strongyloides 51, 57
Subepithelial collagen band 35
Sufferer's quality of life 1
Surgery
acute indications 19
indications for 19
outcomes of 22
T
Tachycardia 4
T-cell trafficking 8f
Telangiectasia 31, 161
Tetracycline 111, 125, 126
quadruple therapy 110
Thiazolidinediones 201
Thioacetamide 240
Thioguanine nucleotide 5
Thioinosine
monophosphate 5
triphosphate 5
Thiopurine 1, 41
analogs 1
methyltransferase 5
Thiouric acid 5
Thioxanthine monophosphate 5
Thumb, rules of 133b
Tissue remodeling 65
Tofacitinib 12, 15
Total colectomy 21, 22, 23
Toxic megacolon 20
Toxic metabolite 6-methylmercaptopurine 5
Toxin resistance 238
Toxocara 51, 57
Transverse colon 54f
Trichinella 57
Trichiura 57
Trigger gene expression 11
Tumor necrosis factor-alpha 1, 32
U
Ulcerative colitis 1, 2, 11, 19, 30, 71, 92
active 73f, 95f
acute severe 3f
endoscopic index of severity 2
moderate-to-severe 1
severe 3f
surgical management of 19
Upper gastrointestinal endoscopy 159
Urinary retention 244
Ustekinumab 9, 10, 15
efficacy of 10
V
Varicella zoster virus 241
Vedolizumab 8, 9, 15, 42
Velpatasvir 231
Viremic infections 223
Vitamin E 201, 207
Vomiting 49
W
Water
exchange 170
immersion 170
Weight loss 244
World Health Organization 261
Wound infection 24
X
Xanthine oxidase 5
Y
Yersinia 30, 33
×
Chapter Notes

Save Clear


Advances in the Treatment of Moderate-to-severe Ulcerative Colitis: Anti-TNFs and BeyondCHAPTER 1

Her Hsin Tsai
 
INTRODUCTION
The incidence and prevalence of inflammatory bowel disease (IBD) have been increasing globally, with the highest incidence in Europe and North America. Ulcerative colitis (UC) is the most common form of IBD with the annual incidence of about 5–20 per 100,000 person years and prevalence of up to 500 cases per 100,000 in some parts of the world. UC affects a variable extent of the colon from the rectum extending proximally with primarily mucosal inflammation. Clinically, it presents as bloody diarrhea, urgency, and abdominal pain, and runs a relapsing and remitting course. It is associated with significant morbidity, with an estimated 30–60% of patients experiencing at least one relapse per year, and approximately 20% of patients suffering from severe form of the disease. These symptoms have a major impact on sufferer's quality of life.
Treatment depends on severity and extent of the disease. The aim is to induce symptom-free remission of the disease and maintaining it. For mild-to-moderate disease, 5-aminosalicylate (orally and rectally) remains useful in both induction and maintenance therapy for UC. For moderate-to-severe disease, corticosteroids remain the primary therapy of UC but are limited by serious side effects. Should induction be successful the steroids are withdrawn and replaced by immunosuppressants such as thiopurine analogs, e.g., azathioprine (AZA) and 6-mercaptopurine (6MP).
More targeted therapies have been developed that specifically inhibit the mediators of gut inflammation. Infliximab is the first, an intravenously administered chimeric monoclonal antibody targeting tumor necrosis factor-alpha (TNF-α), a key proinflammatory cytokine involved in gut inflammation. The ACT 1 trial showed that patients with moderate-to-severe UC had a clinical response rate to infliximab of 65.5% at week 8, and almost 50% maintained response at week 30.1 Adalimumab, a subcutaneously administered humanized anti-TNF antibody, was subsequently developed, with the ULTRA 2 trial demonstrating a clinical response rate of nearly 50% at week 8.2 These biologic therapies are well established and most physicians caring for these patients are comfortable with their use. They are 2also demonstrably cost effective, as prices have tumbled with the advent of generic biosimilars.3
In this chapter, we shall discuss recent advances in the treatment of moderate-to-severe UC. It is not meant to be a comprehensive discussion on the clinical management of UC. It will focus on evolving clinical concepts in optimizing of treatment and new and emerging drug therapy that will likely impact on the clinical management of UC either already licensed or likely to be in the very near future. The number of treatment choices are growing at a remarkable rate and is by understanding the mode of action of the drugs and trial data that the optimal choices can be made for the specific clinical problem.
 
OPTIMIZING CONVENTIONAL THERAPIES
It is a common tendency to move onto a new treatment before optimizing current and often cheaper or less toxic treatments. This may be due to pressures put onto the physician either from patient or carer or from pharmaceutical sales pitch. There are several questions the physician should ask before escalation of treatment. This is particularly true of IBDs.
1. Are you treating the right disease?
It might seem an obvious question but there are a number of differential diagnoses that may ensnare the physician. The diagnosis is reached by a combination of clinical, radiological, endoscopic, and histological features. This is why a multidisciplinary approach is desirable, even essential if mistakes are to be not made.
2. Has the severity and extent of ulcerative colitis been properly assessed?
It is a common mistake to equate subjective patient symptom reporting with disease activity. While all reported symptoms need to be addressed with a treatment plan, only mucosal inflammation will respond to immunological modulation. There is understandably often a large functional element in the patient's symptoms. Irritable bowel syndrome (IBS) often coexists with IBD and the physician will need to be cautious in equating symptoms with disease severity. Hence before escalation of treatment, the patient needs to be reassessed. The best tool is colonoscopy. It will demonstrate extent and severity of inflammation. A form of mucosal inflammation scoring should be adopted [e.g., Mayo score or Ulcerative Colitis Endoscopic Index of Severity (UCEIS)] and extent recorded. Biopsies may be helpful. In acute severe disease, it should be done cautiously and once it is clear that the disease is severe (Mayo 3; UCEIS > 7) then the scope may be withdrawn and a plain abdominal X-ray taken. This is not only safer but remarkably helpful in demonstrating extent of disease, presence of mucosal thickening, and serious complication of a dilated colon. CT scans (or MRI) can also be very helpful in the severe cases (Figs. 1A and B). In the non-urgent cases, colonoscopy is by far the most useful assessment tool.3
zoom view
Fig. 1A: Sigmoid inflammation in patient with severe ulcerative colitis.
zoom view
Fig. 1B: Colonic dilatation in acute severe ulcerative colitis.
In recent years, fecal calprotectin has emerged as a useful tool in monitoring disease activity of UC. If done regularly, serial fecal calprotectin may be indicator of disease flare. However, the test in not reliable in predicting severity or extent of disease and should not be used as a justification for treatment escalation but should be used only as supporting evidence of mucosal inflammation.4
3. Is the disease acute severe?
Acute severe UC is a potentially life-threatening condition and patients are at risk for progressing to toxic megacolon (Fig. 1B) or bowel perforation. Hence recognition of this clinical condition is vital. In the original Truelove and Witts’5 criteria, it is characterized by:
  • Bloody stool frequency ≥6 per day4
PLUS at least one of the following evidence of systemic toxicity:
  • Fever (temperature ≥ 37.8°C)
  • Tachycardia (heart rate ≥ 90 beats/min)
  • Anemia (hemoglobin < 105 g/L)
  • Elevated inflammatory marker [C-reactive protein (CRP) > 100 g/L, erythrocyte sedimentation rate)
A subgroup with abdominal distension and pain is described as acute fulminant colitis.5
Patients with acute severe disease have a high risk of requiring colectomy and need to be recognized and admitted as an acute emergency and treated with fluids and intravenous steroids. They should be assessed frequently and if they do not respond within 3–5 days or if day 3 CRP remains very elevated, then they should be considered for treatment with cyclosporine or infliximab or surgery.6 Comparative trials have shown no difference between cyclosporine and infliximab.7,8 Furthermore, there is no evidence that medical therapy results in lower long-term colectomy rates but it is generally accepted that elective surgery carries a lower morbidity.9,10 The surgical management is discussed in the following chapter.
4. Have you optimized 5-ASA therapies?
5-aminosalicylic acid (5-ASA) treatments are the mainstay in the management of mild-to-moderate UC. This is particularly true of distal disease such as proctitis where the patient may experience a lot of urgency and frequency but may have very limited overall inflammatory load. Formal assessment may classify these patients as having moderate-to-severe disease even though the extent of inflammation may be very limited because of the dependence of scoring systems on bowel frequency counts. In this scenario, immunosuppression may not be the best treatment. Local treatment with 5-ASA products is often the key, with high dose oral and rectal 5-ASA preparations having demonstrable efficacy. This approach is often underused.11
5. Have you optimized immunosuppressive therapies?
Immunosuppressive therapy, mainly the use of AZA and 6MP, is the cornerstone of management of moderate-to-severe UC. Patience is required as the drug is slow acting, taking at least 3 months and sometimes up to 6 months to act. It may be used alongside 5-ASA or as monotherapy in patients intolerant of 5-ASA. It should also be used with biologic therapy where it acts synergistically by suppressing antibody formation.
These drugs have many toxic effects and as many as 25% of patients are intolerant of the drug but only 1–2% develop serious toxicity. Liver toxicity and myelosuppression are the principal severe adverse reactions. Thus physicians may adopt a taciturn approach to the drug use for fear of toxicity and is a reason for suboptimal dosing in UC treatment. Enzyme testing and drug metabolite testing is helpful to in optimizing treatment.5
zoom view
Fig. 2: Azathioprine metabolism: Azathioprine is serially converted to 6-TGNs and metabolized in a rate-limiting fashion into 6-MMP by TPMT enzyme.
(Aza: Azathioprine; Gmps: Guanosine Monophosphate Synthetase; 6-Mp: 6-Mercaptopurine; 6-Mmp: 6-Methylmercaptopurine; Tgn: Thioguanine Nucleotide; Tpmt: Thiopurine Methyltransferase; Xo: Xanthine Oxidase; Timp: Thioinosine Monophosphate; Txmp: Thioxanthine Monophosphate; Titp: Thioinosine Triphosphate; Tu: Thiouric Acid; Gmps: Guanidine Monophosphate Synthetase; Mtimp: Methylthioinosine Monophosphate)
Thiopurine methyltransferase (TPMT) is the critical (Fig. 2) enzyme in AZA and 6-MP metabolism and determines the levels of active molecule 6-thioguanine (6-TG) and toxic metabolite 6-methylmercaptopurine (6-MMP) levels. Approximately 89% of the population has wild type TPMT, which is associated with normal or “high” TPMT enzyme activity, while 11% are heterozygous and have corresponding low TPMT enzyme activity. A small number (1 in 300) of the population is homozygous for mutations of TPMT and thus have negligible activity, which causes 6-MP to be preferentially metabolized to produce high levels of 6-TG, which then leads to bone marrow suppression. Very high levels of TPMT may result in increased accumulation of 6-MMP increasing risk of liver toxicity. Thus determining the activity of TPMT may help optimize treatment.
Direct measurements of drug metabolite levels are available. Levels of 6-MMP can also be helpful to predict liver toxicity. The active moiety 6-TG can be a good measure of optimal dose of the drug. A recent meta-analysis of studies looking at 6-TG levels also demonstrated that clinical remission was significantly more likely among patients with 6-TG levels over a cut-off value between 230 and 260 (odds ratio 3.2, 95% CI 2.4–4.1).12
 
OPTIMIZING ANTI-TNF TREATMENTS
When it is clear that in the patient who has failed or is intolerant of the conventional treatments for UC, and that the patient has disease of at least moderate severity, then a biologic drug is the next step. It is usual to choose an anti-TNF as it is well established and availability of biosimilars has made it more affordable. There are some recent evolving concepts that may help optimize therapy.6
 
Addition of Immunosuppressives to Anti-TNF Treatment Improves Efficacy
In a clinical trial of infliximab monotherapy versus infliximab plus azathioprine versus azathioprine alone (UC-SUCCESS), corticosteroid-free remission at week 16 was achieved by 39.7% of patients receiving infliximab/azathioprine, compared with 22.1% receiving infliximab alone (p = 0.017) and 23.7% receiving azathioprine alone (p = 0.032).13 The effect is likely to be due to improved infliximab serum concentrations due to reduced antibody development in patients on combination therapy. Sub-analyses of a similar Crohn's study (SONIC) showing higher week 30 infliximab trough levels with combination versus infliximab monotherapy, 3.5 μg/mL versus 1.6 μg/mL (p < 0.001), and lower incidence of anti-infliximab antibody, 0.9% versus 14.6%. Interestingly, serious adverse events were actually lower with combination versus infliximab monotherapy (15.1% vs. 23.9%, p = 0.04).14
The dose and length of AZA treatment required remain uncertain but antibody suppression requires only a small dose of AZA, hence it is sensible to keep the patient on AZA for at least 3 months.
 
Treatment Goals and Targets
It has been noted in many trials involving infliximab that patients who had achieved mucosal healing had a more durable remission than those with symptom resolution without complete mucosal healing.15 In 2015, the Selecting Therapeutic Targets in Inflammatory Bowel Disease (STRIDE) committee defined the treat-to-target approach for IBD, which shifted the goal of UC treatment from short-term symptom resolution to long-term prevention of disease complications (dysplasia/cancer, hospitalizations, and colectomy).16 The three most promising composite targets for UC were symptom resolution (normalization of bowel habit and absence of rectal bleeding), endoscopic mucosal healing (Mayo or UCIES inflammation score of 0), and fecal calprotectin <100 µg/g. Although this area is still subject to ongoing long-term clinical investigation, early indication is that target achievement with aggressive medical therapy is associated with better outcome, lower colectomy rates, and hospitalizations.17
 
Personalizing Treatment
The ultimate refinement in the management of UC is to accurately personalize treatments for the individual patient. This depends on the ability of the clinician to accurately predict the likely clinical course and prognosis of the disease. This can be difficult to achieve. Currently we depend on clinical features, endoscopic appearance, and response to therapy to help us predict those who are likely to relapse or end up with colectomies. These measures are not adequate. Genetic studies have not been very helpful. Current best 7practice of personalizing treatment is based on optimizing treatments and selecting the right targets as already discussed above. The future may be the identification of biomarkers that can accurately predict the patients who are likely to have more aggressive disease. One concept is to identify T-cell transcriptional signatures using machine learning and a promising biomarker may soon be available.18
 
Prospective and Scheduled Therapeutic Drug Monitoring
Therapeutic drug monitoring is now widely available both for thiopurines and anti-TNFs. With anti-TNFs both trough drug levels taken just before a scheduled dose and antibody levels are available. Currently, the majority of clinicians perform trough and antibody levels reactively in response to suboptimal treatment response. Prospective scheduled monitoring is regarded as expensive but with costs of therapeutic drug monitoring falling due to the economies of scale, scheduled testing may become increasingly the norm. There is no doubt that it improves patient outcome and gastroenterological associations are recommending its more widespread use.19 Recent studies suggest that prospective therapeutic drug monitoring may be highly cost effective.20
 
NEW DRUGS
Although anti-TNFs have revolutionized treatment of UC, some 40–60% still fail to respond. If the patient initially responded but then lose response (secondary failure to respond) or if antibody formation is identified as the reason for failure, then switching to a different drug within this class may be appropriate. But when the patient never responded to anti-TNFs (primary failure to respond) or when anti-TNFs have been optimized by scheduled therapeutic drug monitoring and patient still has inadequate response to therapy then a change in class of drug is indicated. Fortunately, there are several such drugs to choose from, some already licensed and others in various stages of development and certification.
 
Anti-adhesion Agents
One of the key components of the inflammatory response is the ability of the body's immune system to recruit immune modulator cells to the part of the body where they are needed. This is through trafficking of inflammatory cells via the circulatory system. To facilitate this, the vascular endothelium of vessels neighboring the areas where inflammatory cells are required express on its surface the mucosal addressin-cell adhesion molecule (usually abbreviated, MadCAM-1) which binds to integrins expressed on circulatory T-cells (Fig. 3).8
zoom view
Fig. 3: T-cell trafficking.
(MAdCAM: mucosal addressin-cell adhesion molecule)
These integrins are heterodimeric receptors composed of an α and β subunit, that is expressed on the surface of circulating lymphocytes when they are activated. This process leads to the binding of circulating lymphocytes onto the endothelium and migration into the lamina propria and tissue, contributing to the inflammatory process in IBD.21
These integrins and MAdCAM-1 are thus potential therapeutic targets for IBD therapy. Integrin antagonists are a class of monoclonal antibodies that can block the trafficking of lymphocytes to the intestinal endothelium. The first integrin antagonist to emerge is natalizumab, a humanized IgG4 monoclonal antibody that leads to inhibition of the α4 integrin. Unfortunately, the use of natalizumab was limited by associated increased incidence of progressive multifocal leukoencephalopathy (PML), a fatal demyelinating disease of the central nervous system (CNS) caused by the opportunistic human John Cunningham (JC) virus. Vedolizumab (also known as LDP-02 and MLN02, MLN002), a humanized monoclonal IgG1 antibody, was subsequently developed as a gut selective anti-integrin specifically targeting α4β7 integrins in the gut and importantly not the integrins found in the CNS.21,22
The efficacy of vedolizumab was demonstrated in two integrated regulatory trials (GEMINI 1). Response rates at week 6 were found to be 47.1% and 25.5% among patients in the vedolizumab group and placebo group, respectively (95% CI, 11.6–31.7; p < 0.001). At week 52, 41.8% of patients who continued to receive vedolizumab every 8 weeks were in clinical remission [Mayo Clinic Score (MCS) ≤2 and no subscore >1], as compared with 15.9% of patients who switched to placebo (95% CI, 14.9–37.2; p < 0.001).23,24
There may be advantages of using vedolizumab as a second line biologic therapy in UC. Its mode of action is very different from anti-TNF and represents a change in class. In theory, changing of one anti-TNF to another would only be effective if there is drug tolerance due to antibody formation. Primary infliximab failures, that is, those who have never responded to anti-TNF, are not likely to respond to another anti-TNF. The mode of action of vedolizumab also appears more benign compared to anti-TNFs as it does not interfere with critical antibacterial, especially anti-mycobacterial, 9pathways which make anti-TNFs so hazardous in TB positive individuals. In this respect, there are compelling reasons to suggest that vedolizumab is less harmful than anti-TNFs.25
Treatment is given as an IV infusion with an induction of 300 mg at weeks 0, 2, and 6 followed by 8 weekly dosing of 300 mg. There are indications from studies that vedolizumab may take longer than anti-TNFs to achieve therapeutic results. Clinical response may not be apparent until week 10 and hence a little patience from the clinician and forewarning to the patient would be judicious. Some would advocate an additional dose at week 10 if there is a lack of adequate response at week 8.
Using vedolizumab as a first line biologic therapy also can be considered. At present, the cost differential to generic infliximab is too high to make it a cost effective strategy. This is based on equal efficacy of both biologic treatments. However recent data may suggest otherwise. A network meta-analysis has suggested that vedolizumab is superior to adalimumab.26 This finding is now confirmed by a head-to-head study between adalimumab and vedolizumab. In a randomized trial comparing vedolizumab with adalimumab in over 700 patients with moderately-to-severely active UC, vedolizumab resulted in a significantly higher rate of clinical remission (31% vs. 22%).27 The rate of serious infections was similar in both groups (<2%). The therapeutic gain may well be sufficiently large to make vedolizumab a cost-effective first line therapy for UC.
Etrolizumab is very similar, being a humanized monoclonal antibody against the β7 subunit of integrins α4β7 and αEβ7. Studies on the induction and maintenance treatment of UC are very promising.28,29 At the time of writing, the drug has not yet been approved by the FDA.
An antibody to the adhesion MadCAM-1 is also under development.30
 
Anti-interleukin Inhibitors
The proinflammatory cytokines interleukin 12 (IL-12) and interleukin 23 (IL-23) play an important role in the pathophysiology of Crohn's disease (CD) and possibly UC.31 These cytokines bind to receptors of CD4+ T-cells and lead to their differentiation into activated Th1 and Th17 cells (Fig. 4). There are multiple lines of evidence suggesting that UC is mediated by Th1 and CD by a combination of Th1 and Th17 cells.32 Furthermore, the IL-23 receptor mutation has been identified as a possible IBD gene.33 Ustekinumab, a fully human immunoglobulin G1 kappa monoclonal antibody that binds with high affinity to the p40 subunit of human IL-12 and IL-23, has recently been approved for the treatment of moderately to severely active CD in adults. Ustekinumab prevents IL-12 and IL-23 bioactivity by preventing their interaction with their cell surface receptor protein IL-12Rβ1. Through this mechanism of action, ustekinumab effectively neutralizes IL-12 (Th1)- and IL-23 (Th17)-mediated cellular responses.10
zoom view
Fig. 4: The role of IL-23 and IL-12.
Evidence for the efficacy of ustekinumab in UC had been investigated and encouraging results published recently.34 In this study, the percentage of patients who had clinical remission at week 8 among patients who received intravenous ustekinumab at a dose of 130 mg (15.6%) or 6 mg/kg (15.5%) was significantly higher than that among patients who received placebo (5.3%) (p < 0.001 for both comparisons). Among patients who had a response to induction therapy with ustekinumab and underwent a second randomization, the percentage of patients who had clinical remission at week 44 was significantly higher among patients assigned to 90 mg of subcutaneous ustekinumab every 12 weeks (38.4%) or every 8 weeks (43.8%) than among those assigned to placebo (24.0%) (p = 0.002 and p < 0.001, respectively). The incidence of serious adverse events with ustekinumab was similar to that with placebo. However, there were two deaths (one each from acute respiratory distress syndrome and hemorrhage from esophageal varices) and seven cases of cancer (one each of prostate, colon, renal papillary, and rectal cancer and three nonmelanoma skin cancers) among 825 patients who received ustekinumab and no deaths and one case of cancer (testicular cancer) among 319 patients who received placebo. Although these do not constitute a clear hazard signal, some caution must be taken until more data becomes available.
Risankizumab, an IL-23 antibody, is currently being assessed in UC after encouraging results in CD.35
 
JAK/STAT Pathway Inhibitors
Cytokines are released by the immune system in response to a signal principally by gut stellate cells or macrophages in response to a signal. They bind to specific receptors on the T-cell, triggering activation and synthesis of specific proteins that initiate the immune response. Cytokines can take the form of many structurally unrelated proteins that are typed according to their binding to distinct receptor families, which include type I cytokine receptors, type II cytokine receptors, the TNF receptor family, and the IL-1 receptor family receptors.11
zoom view
Fig. 5: The JAK-STAT pathway.
(ISRE: interferon-stimulated response element; GAF: gamma activation factor; GAS: gamma activated sequence; IFN: interferon; IRF: interferon regulatory factor)
The cytokines bind to the relevant receptor and trigger intracellular changes, resulting in signal transduction and trigger gene expression. The signal transduction is via various protein kinases. The Janus kinase (JAK) is a family of receptor-associated tyrosine kinases that are essential for the cytokine signaling cascade (Fig. 5), downstream of type I and type II cytokine receptors.36 The JAK-signal transducers and activators of transcription (STAT) pathway plays an important role in innate immunity, adaptive immunity, and hematopoiesis, participating in cellular processes such as cell growth, survival, differentiation, and migration. There are four members of the JAK family (JAK1, JAK2, JAK3, and TYK2) and seven signal transducers and transcription activators called signal transducer and activator of transcription, or STAT (STAT 1–4, 5a, 5b, and 6). The unique structure of each JAK clearly distinguishes them from other members of the protein tyrosine kinase family.37
Ulcerative colitis and Crohn's disease involve many cytokine signaling which in turn depend on JAK-STAT pathway for immune cell gene 12transcription. The key cytokines in the pathogenesis of IBD belong to type I and type II cytokines receptors. These are the receptors of certain key cytokines namely IL-6, IL-5, IL-9, IL-10, IL-13, IL-12/23, IL-22, granulocyte–macrophage colony-stimulating factor (GM-CSF), and IFN-γ. All these cytokines signal through the JAK/STAT pathway. In contrast, the cytokines TNF, IL-1, and IL-17, which are the other major drivers of IBD, do not use the JAK-STAT pathway in their signaling pathways but they do however induce the expression of a wide range of downstream proinflammatory cytokines that, in turn, depend on JAK/STAT signaling.38
Thus specific inhibitors of these kinases are a logical target as a treatment of inflammatory conditions. Several of these molecules are in development and are small, orally active molecules. Tofacitinib is the first JAK inhibitor to be approved for the treatment of moderate to severely active UC by the FDA (US) and EMA (European) agencies. Several other molecules are undergoing trials and are in advanced stages of development. Tofacitinib (Xeljanz, Pfizer) is a pan-JAK inhibitor that preferentially inhibits JAK1 and JAK3, in a dose-dependent fashion. Oral tofacitinib is well absorbed and is cleared principally by the liver. It has a short half-life of 3 hours.39
Tofacitinib was found to be effective for induction of remission for patients with UC. In two randomized trials (OCTAVE Induction 1 and 2 trials of 598 and 541 patients, respectively), patients with moderate-to-severe UC who received tofacitinib 10 mg twice daily experienced remission at 8 weeks more frequently compared with those receiving placebo; for OCTAVE 1, 18% versus 8% (absolute difference 10, 95% CI: 4–16) and for OCTAVE 2, 17% versus 4% (absolute difference 13, 95% CI: 8–18).40 In the OCTAVE Sustain trial, 593 patients who had a clinical response to induction therapy were randomly assigned to receive maintenance therapy with tofacitinib (either 5 mg or 10 mg twice daily) or placebo for 52 weeks. The primary end point was remission at 52 weeks which occurred in 34.3% of the patients in the 5-mg tofacitinib group and 40.6% in the 10-mg tofacitinib group versus 11.1% in the placebo group (p < 0.001 for both comparisons with placebo).40
The onset of action of tofacitinib varies greatly with some patients have responding rapidly, while for other patients, response may take up to 8 weeks. Caution should be used when prescribing tofacitinib for patients with a history of thromboembolic disease, cardiovascular disease, or those ≥50 years old with at least one cardiovascular risk factor because of an increased risk of thromboembolic events and mortality in patients who were treated with tofacitinib.
 
Sphingosine-1-phosphate Receptor Modulators
Sphingosine-1-phosphate (S1P) is a signaling sphingolipid becoming the active moiety by phosphorylation of sphingosine which is catalyzed by sphingosine kinase, an enzyme found in the cytosol and endoplasmic reticulum of various types of cells and S1P can be dephosphorylated to sphingosine by sphingosine phosphatases.13
zoom view
Fig. 6: Sphingosine-1-phosphate (S1P) signaling and receptor modulation.
(SL: sphingosine lyase; SK: sphingosine kinase; PINK1: PTEN-induced kinase 1)
S1P produced in endovascular or lymphatic endothelial cells is secreted into the vasculature through the specific transporter SPNS2 (Fig. 6).
Sphingosine-1-phosphate is of importance in the entire human body; it is a major regulator of vascular and immune systems. In the vascular system, S1P regulates angiogenesis, vascular stability, and permeability. In the immune system, it is now recognized as a major regulator of trafficking of T- and B-cells. S1P interacts with its specific receptors S1PR of which there are five subtypes 1–5. S1P1, S1P4, and S1P5 are involved in regulation of the immune system, while S1P2 and S1P3 may be associated with cardiovascular and pulmonary system, and lead to cell proliferation and theoretical cancer-related risks. S1PR1 is of particular importance in immune regulation as it is found on T-cells. It appears to function as an elixir of life to the T-cells ensuring cell survival while a deficiency of S1P results in apoptosis and cell death. There is a concentration gradient between the lymphoid tissue in thymus or nodes and the lymphatic vessels and as the T-cells are attracted to the higher concentration in the vessels, it leads to egress of immune cells from the lymphoid organs (such as thymus and lymph nodes) into the lymphatic vessels.41
Sphingosine-1-phosphate is thus a promising target for pharmacologic intervention. When a synthetic agonist targets the S1PR receptor, the receptor–drug complex is internalized and voided and the T-cell loses its ability to respond to the S1P concentration gradient and thus remains the lymphoid tissue rather than egress into the lymphatic vessels and thus into the wider circulation and to the target organ like the gut in IBDs.42 Several 14small molecule agonists of S1PR have been developed.43 Selective targeting S1P receptors for inflammatory conditions is important to avoid unwanted vascular and proliferative effects. Three S1P modulators with differing selectivity for S1P receptors were in clinical development for IBD—ozanimod, etrasimod, and amiselimod.
In the ozanimod study, the primary outcome of remission occurred in 16% of the patients who received 1 mg of ozanimod and in 14% of those who received 0.5 mg of ozanimod, as compared with 6% of those who received placebo (p = 0.048 and p = 0.14, respectively, for the comparison of the two doses of ozanimod with placebo). Clinical response (decrease in MCS of ≥3 points and ≥30% and decrease in rectal bleeding subscore of ≥1 point or a subscore ≤1) at 8 weeks occurred in 57% of those receiving 1 mg of ozanimod and 54% of those receiving 0.5 mg, as compared with 37% of those receiving placebo. At week 32, the rate of clinical remission was 21% in the group that received 1 mg of ozanimod, 26% in the group that received 0.5 mg of ozanimod, and 6% in the group that received placebo; the rate of clinical response was 51%, 35%, and 20%, respectively. At week 8, absolute lymphocyte counts declined 49% from baseline (BL) in the group that received 1 mg of ozanimod and 32% from BL in the group that received 0.5 mg. There were no safety concerns.44
In the OASIS study, etrasimod 2 mg improved change from BL in 3-component MCS versus placebo (difference, 0.99 points; 90% CI, 0.30–1.68; p = 0.009). More patients receiving etrasimod 2 mg achieved endoscopic improvement (41.8% vs. 17.8% for PBO; p = 0.003). At week 12, there was a significant decrease in circulating lymphocyte counts from BL with etrasimod 1 mg and 2 mg relative to PBO (37.2% and 57.3%, respectively; p < 0.001 for both).45
Etrasimod demonstrated durable clinical remission in the treatment of moderate to severely active UC in a phase 2 open-label extension study. The 34-week open-label extension study explored the long-term safety, tolerability, and efficacy of etrasimod in 118 patients who completed the 12-week phase 2 OASIS randomized controlled trial. Researchers analyzed the data to determine clinical response, clinical remission, and endoscopic improvement at the end of 46 weeks of treatment. Of the patients who completed 2 mg of etrasimod treatment during the extension study (n = 84), 79% achieved clinical response, 39% achieved clinical remission, and 51% had endoscopic improvement by the end of the study. Among patients who also received 2 mg of etrasimod during the OASIS trial (n = 22), 82% experienced clinical response, 50% were in clinical remission, and 55% had endoscopic improvement. For patients who achieved clinical response or clinical remission after the OASIS trial, 93% experienced sustained response and 75% experienced sustained remission at both 12 and 46 weeks. Investigators found that adverse events in the extension study were mild-to-moderate and did not discover any new safety concerns.4515
 
Choosing a Second (or Third)-line Biologic Therapy
As these newer biologic therapies undergo regulatory trials to achieve certification, the best drug for each clinical scenario is still to be worked out. Until further studies are available, a sensible strategy based on currently available data can be adopted. After optimizing conventional therapies of 5-ASA and immunosuppressives, anti-TNFs in combination with thiopurines can be used a sensible first-line biologic. There may be a case for the use of vedolizumab as a first-line biologic but based on cost and physician familiarity, most experts would regard anti-TNFs to be the logical choice. There may also be a case for using infliximab in preference to adalimumab or golimumab based on network meta-analyses.46
In terms of treatment targets, the best single marker is still endoscopic mucosal resolution.47 Hence an assessment colonoscopic examination at 6 months after commencement of the drug is sensible. If treatment target is not achieved and if trough drug levels are adequate, then a change in class of drug is indicated. Currently, there are three licensed drugs that may be used; vedolizumab, ustekinumab, and tofacitinib. Vedolizumab is probably the best second line drug after anti-TNF failure. If an oral agent is to be preferred, then a case for tofacitinib can be made. Ustekinumab can be held for use as a third line drug.
The pace of change in the landscape of UC therapy continues unabated and it is challenging for the gastroenterologist to keep in step. However, with a sound grounding in scientific basis of the biology of the disease and with a critical eye over trial data, the IBD physician can optimize and personalize treatment of this disabling disease.
REFERENCES
  1. Rutgeerts P, Sandborn WJ, Feagan BG, Reinisch W, Olson A, Johanns J, et al. Infliximab for induction and maintenance therapy for ulcerative colitis. N Engl J Med. 2005;353(23):2462–76.
  1. Sandborn WJ, van Assche G, Reinisch W, Colombel JF, D'Haens G, Wolf DC, et al. Adalimumab induces and maintains clinical remission in patients with moderate-to-severe ulcerative colitis. Gastroenterology. 2012;142(2):257–65.
  1. Tsai HH, Punekar YS, Morris J, Fortun P. A model of the long-term cost-effectiveness of scheduled maintenance treatment with infliximab for moderate-to-severe ulcerative colitis. Aliment Pharmacol Ther. 2008;28(10):1230–9.
  1. Reenaers C, Bossuyt P, Hindryckx P, Vanpoucke H, Cremer A, Baert F. Expert opinion for use of faecal calprotectin in diagnosis and monitoring of inflammatory bowel disease in daily clinical practice. United European Gastroenterol J. 2018;6(8):1117–25.
  1. Truelove SC, Witts LJ. Cortisone in ulcerative colitis; final report on a therapeutic trial. Br Med J. 1955;2(4947):1041–8.
  1. Travis S, Satsangi J, Lémann M. Predicting the need for colectomy in severe ulcerative colitis: a critical appraisal of clinical parameters and currently available biomarkers. Gut. 2011;60(1):3–9.

  1. 16 Williams JG, Alam MF, Alrubaiy L, Arnott I, Clement C, Cohen D, et al. Infliximab versus ciclosporin for steroid-resistant acute severe ulcerative colitis (CONSTRUCT): a mixed methods, open-label, pragmatic randomised trial. Lancet Gastroenterol Hepatol. 2016;1(1):15–24.
  1. Laharie D, Bourreille A, Branche J, Allez M, Bouhnik Y, Filippi J, et al. Ciclosporin versus infliximab in patients with severe ulcerative colitis refractory to intravenous steroids: a parallel, open-label randomised controlled trial. Lancet. 2012;380(9857):1909–15.
  1. Laharie D, Bourreille A, Branche J, Allez M, Bouhnik Y, Filippi J, et al. Long-term outcome of patients with steroid-refractory acute severe UC treated with ciclosporin or infliximab. Gut. 2018;67(2):237–43.
  1. Assche GV, Vermeire S, Rutgeerts P. Management of acute severe ulcerative colitis. Gut. 2011;60(1):130–3.
  1. Seibold F, Fournier N, Beglinger C, Mottet C, Pittet V, Rogler G, et al. Topical therapy is underused in patients with ulcerative colitis. J Crohns Colitis. 2014;8(1):56–63.
  1. Moreau AC, Paul S, Del Tedesco E, Rinaudo-Gaujous M, Boukhadra N, Genin C, et al. Association between 6-thioguanine nucleotides levels and clinical remission in inflammatory disease: a meta-analysis. Inflamm Bowel Dis. 2014;20(3):464.
  1. Panaccione R, Ghosh S, Middleton S, Márquez JR, Scott BB, Flint L, et al. Combination therapy with infliximab and azathioprine is superior to monotherapy with either agent in ulcerative colitis. Gastroenterology. 2014;146(2):392–400.
  1. Colombel JF, Sandborn WJ, Reinisch W, Mantzaris GJ, Kornbluth A, Rachmilewitz D, et al. Infliximab, azathioprine, or combination therapy for Crohn's disease. N Engl J Med. 2010;362(15):1383–95.
  1. Colombel JF, Rutgeerts P, Reinisch W, Esser D, Wang Y, Lang Y, et al. Early mucosal healing with infliximab is associated with improved long-term clinical outcomes in ulcerative colitis. Gastroenterology. 2011;141(4):1194–201.
  1. Colombel JF, Narula N, Peyrin-Biroulet L. Management strategies to improve outcomes of patients with inflammatory bowel diseases. Gastroenterology. 2017;152(2):351–61.
  1. Ungaro R, Colombel JF, Lissoos T, Peyrin-Biroulet L. A treat-to-target update in ulcerative colitis: a systematic review. Am J Gastroenterol. 2019;114(6):874–83.
  1. Biasci D, Lee JC, Noor NM, Pombal DR, Hou M, Lewis N, et al. A blood-based prognostic biomarker in inflammatory bowel disease. Gut. 2019;68(8):1386–95.
  1. Feuerstein JD, Nguyen GC, Kupfer SS, Falck-Ytter Y, Singh S; American Gastroenterological Association Institute Clinical Guidelines Committee. American Gastroenterological Association Institute Guideline on Therapeutic Drug Monitoring in Inflammatory Bowel Disease. Gastroenterology. 2017;153(3):827–34.
  1. Gettigan NM, Keogh A, McCarthy O, McNally M, Deane C, Slattery E. The effects of proactive therapeutic drug monitoring versus reactive therapeutic drug monitoring in a virtual biologic clinic, a retrospective cohort study. GastroHep. 2019;1(6):274–83.
  1. Lau MS, Tsai HH. Review of vedolizumab for the treatment of ulcerative colitis. World J Gastrointest Pharmacol Ther. 2016;7(1):107–11.
  1. Tsai HH, Black C. A review of the cost-effectiveness of vedolizumab for treating moderate-to severely active ulcerative colitis. Expert Rev Pharmacoecon Outcomes Res. 2016;16(6):679–83.

  1. 17 Feagan BG, Rutgeerts P, Sands BE, Hanauer S, Colombel JF, Sandborn WJ, et al. Vedolizumab as induction and maintenance therapy for ulcerative colitis. N Engl J Med. 2013;369(8):699–710.
  1. Sandborn WJ, Feagan BG, Rutgeerts P, Hanauer S, Colombel JF, Sands BE, et al. Vedolizumab as induction and maintenance therapy for Crohn's disease. N Engl J Med. 2013;369(23):2183–96.
  1. Bickston SJ, Behm BW, Tsoulis DJ, Cheng J, MacDonald JK, Khanna R, et al. Vedolizumab for induction and maintenance of remission in ulcerative colitis. Cochrane Database Syst Rev. 2014;(8):CD007571.
  1. Tsai HH. Editorial: tofacitinib and biologics for moderate-to-severe ulcerative colitis–what is best in class? Aliment Pharmacol Ther. 2018;47(4):539–40.
  1. Sands BE, Peyrin-Biroulet L, Loftus EV Jr, Danese S, Colombel JF, Törüner M, et al. Vedolizumab versus adalimumab for moderate-to-severe ulcerative colitis. N Engl J Med. 2019;381(13):1215–26.
  1. Vermeire S, O'Byrne S, Keir M, Williams M, Lu TT, Mansfield JC, et al. Etrolizumab as induction therapy for ulcerative colitis: a randomised, controlled, phase 2 trial. Lancet. 2014;384(9940):309–18.
  1. Rosenfeld G, Parker CE, MacDonald JK, Bressler B. Etrolizumab for induction of remission in ulcerative colitis. Cochrane Database Syst Rev. 2015;(12):CD011661.
  1. Vermeire S, Sandborn WJ, Danese S, Hébuterne X, Salzberg BA, Klopocka M, et al. Anti-MAdCAM antibody (PF-00547659) for ulcerative colitis (TURANDOT): a phase 2, randomised, double-blind, placebo-controlled trial. Lancet. 2017;390(10090):135–44.
  1. Uhlig HH, McKenzie BS, Hue S, Thompson C, Joyce-Shaikh B, Stepankova R, et al. Differential activity of IL-12 and IL-23 in mucosal and systemic innate immune pathology. Immunity. 2006;25(2):309–18.
  1. Geremia A, Arancibia-Cárcamo CV, Fleming MP, Rust N, Singh B, Mortensen NJ, et al. IL-23-responsive innate lymphoid cells are increased in inflammatory bowel disease. J Exp Med. 2011;208(6):1127–33.
  1. Duerr RH, Taylor KD, Brant SR, Rioux JD, Silverberg MS, Daly MJ, et al. A genome-wide association study identifies IL23R as an inflammatory bowel disease gene. Science. 2006;314(5804):1461–3.
  1. Sands BE, Sandborn WJ, Panaccione R, O'Brien CD, Zhang H, Johanns J, et al. Ustekinumab as induction and maintenance therapy for ulcerative colitis. N Engl J Med. 2019;381(13):1201–14.
  1. Feagan BG, Sandborn WJ, D'Haens G, Panés J, Kaser A, Ferrante M, et al. Induction therapy with the selective interleukin-23 inhibitor risankizumab in patients with moderate-to-severe Crohn's disease: a randomised, double-blind, placebo-controlled phase 2 study. Lancet. 2017;389(10080):1699–709.
  1. Schwartz DM, Kanno Y, Villarino A, Ward M, Gadina M, O'Shea JJ. JAK inhibition as a therapeutic strategy for immune and inflammatory diseases. Nat Rev Drug Discov. 2017;16(12):843–62.
  1. Banerjee S, Biehl A, Gadina M, Hasni S, Schwartz DM. JAK–STAT signaling as a target for inflammatory and autoimmune diseases: current and future prospects. Drugs. 2017;77(5):521–46.

  1. 18 Olivera P, Danese S, Peyrin-Biroulet L. JAK inhibition in inflammatory bowel disease. Expert Rev Clin Immunol. 2017;13(7):693–703.
  1. Hemperly A, Sandborn WJ, Vande Casteele N. Clinical pharmacology in adult and pediatric inflammatory bowel disease. Inflamm Bowel Dis. 2018;24(12):2527–42.
  1. Sandborn WJ, Su C, Sands BE, D'Haens GR, Vermeire S, Schreiber S, et al. Tofacitinib as induction and maintenance therapy for ulcerative colitis. N Engl J Med. 2017;376(18):1723–36.
  1. Pérès M, Montfort A, Andrieu-Abadie N, Colacios C, Ségui B. S1P: the elixir of life for naive T cells. Cell Mol Immunol. 2018;15(7):657–9.
  1. Wollny T, Wątek M, Durnaś B, Niemirowicz K, Piktel E, Żendzian-Piotrowska M, et al. Sphingosine-1-phosphate metabolism and its role in the development of inflammatory bowel disease. Int J Mol Sci. 2017;18(4):741.
  1. Peyrin-Biroulet L, Christopher R, Behan D, Lassen C. Modulation of sphingosine-1-phosphate in inflammatory bowel disease. Autoimmun Rev. 2017;16(5):495–503.
  1. Sandborn WJ, Feagan BG, Wolf DC, D'Haens G, Vermeire S, Hanauer SB, et al. Ozanimod induction and maintenance treatment for ulcerative colitis. N Engl J Med. 2016;374(18):1754–62.
  1. Sandborn W, Peyrin-Biroulet L, Trokan L, Zhang J, Kűhbacher T, Chiorean M, et al. A randomized, double-blind, placebo-controlled trial of a selective, oral sphingosine 1-phosphate (S1P) receptor modulator, Etrasimod (A PD334), in moderate to severe ulcerative colitis (UC): Results from the OASIS study. Am J Gastroenterol. 2018;113:S327–8.
  1. Bonovas S, Lytras T, Nikolopoulos G, Peyrin-Biroulet L, Danese S. Systematic review with network meta-analysis: comparative assessment of tofacitinib and biological therapies for moderate-to-severe ulcerative colitis. Aliment Pharmacol Ther. 2018;47(4):454–65.
  1. Lichtenstein GR, Rutgeerts P. Importance of mucosal healing in ulcerative colitis. Inflamm Bowel Dis. 2010;16(2):338–46.