Brain Activity and Oxygenation Inflammatory Bowel Disease (IBD) Patients

April 26, 2021 updated by: University of Calgary

Evaluation of Brain Activity and Oxygenation Using Near-infrared Spectroscopy (NIRS) in Inflammatory Bowel Disease (IBD) Patients

Symptoms such as fatigue, sleep disturbances, anxiety and depression are common in patients with IBD, but the cause is unknown. Understanding how these behaviors occur in IBD and their role in symptoms may help improve management of IBD. How IBD leads to changes in brain function remains unclear. Inflammation and dysfunction of blood flow may occur in patients with IBD, which may be linked to these symptoms. Patients with IBD also have an alteration or imbalance of gut bacteria which may play a role in the development of the disease, but the exact mechanism remains poorly understood;as a result, there are limited therapeutic options available clinically to address this issue. An approved therapy, anti-TNF α, may be useful in improving brain and gut activity as well as quality of life. The purpose of this research study is to better understand brain and gut activity in the context of IBD to possibly improve treatments for the disease. In patients taking anti-TNFα therapy as prescribed clinically as standard of care, the investigators will measure brain activity using NIRS; gut microbiome using stool analysis and quality of life using various questionnaires.

Study Overview

Detailed Description

Hypothesis Conditions with acute systemic inflammation will correlate with changes in gut microbiome signatures, reduced cerebral oxygen saturation (StO2) and altered patterns of microvascular cerebral blood perfusion (as determined by near-infrared spectroscopy NIRS).

Rationale The investigators are investigating the ability of near-infrared spectroscopy (NIRS) to detect altered brain function and oxygenation under a range of clinically relevant medical conditions.

NIRS allows for non-invasive measurement of oxygenated (O2Hb) and deoxygenated hemoglobin (HHb) concentrations in cortical brain tissue. Therefore, NIRS can be used to quantify cortical tissue oxygen saturation (StO2), which reflects changing metabolic rate and perfusion. Furthermore, functional NIRS (fNIRS) can be used to probe brain activity in a similar fashion to functional MRI, by assuming that the measured changes in blood perfusion and oxygenation are due to functional hyperemia (i.e. hemodynamic changes caused by neural activity). Using NIRS in patients, altered patterns of brain blood perfusion and reduced brain oxygenation levels have been shown to correlate with fatigue and impaired cognition, linking reduced cortical perfusion to symptoms that have been reported to occur in patients with IBD.

Tight regulation of cerebral blood flow is critical to ensure normal brain oxygenation and function. Cerebrovascular dysfunction and hypoxia have been implicated in a range of neurodegenerative and neuroinflammatory disorders including Alzheimer's disease and multiple sclerosis (MS). In a recent study using fNIRS, the investigators found altered task associated hemodynamic responses and brain hypoxia in patients with the immune-mediated liver disease, primary biliary cholangitis (PBC). Moreover, the experimental work in mice suggests that these changes may be due to the actions of TNFα on the cerebral vasculature. It also showed that patients with multiple sclerosis had reduced inter-hemispheric coherence and hypoxia. These observations led to the question of whether general systemic inflammation or brain/gut interactions could link to abnormal brain function and hypoxia, as detectable by NIRS.

Supporting this hypothesis is a recent NIRS study by Fujiwara T et al in patients with IBD. They showed that during performance of a task, mean oxygenated hemoglobin concentration was significantly lower in the frontal lobe in IBD patients compared to healthy controls. However, changes in brain oxygenation have not been previously examined during resting state or in the context of markers of peripheral inflammation, quality of life measures and symptom burden, or gut microbiome signatures in IBD patients.

This currently proposed substudy is to examine patients with inflammatory bowel disease (IBD) for their level of hypoxia, fNIRS functional response and fNIRS coherence.

IBD is increasing in incidence and prevalence worldwide. Several factors have been implicated in the development of IBD, including a dysregulated immune response to altered microbiota in a genetically susceptible host exposed to inciting environmental factors, but the specific cause of IBD remains unknown. Comorbid maladaptive behaviors and pain are prevalent in patients with IBD20-26. These mental health comorbidities complicate management of IBD, adversely impacting patient outcomes and health, and increasing the resource burden to the health care system. However, comorbid mental health issues and symptoms in IBD patients are poorly understood and under-treated, paralleling unmet mental health treatment needs in the general Canadian population. Therefore, an improved understanding of how brain changes occur in association with IBD, and their potential role in symptom development and link to systemic inflammation and gut microbiome signatures is of significant importance to improve management of these patients.

The gastrointestinal tract serves as a dynamic and local ecosystem for gut microbiota. Dysbiosis, which is an alteration of the gut microbial composition that contributes to host disease, occurs in IBD patients. Specifically, IBD patients exhibit a lower microbial α-diversity and are enriched in several groups of bacteria compared with healthy controls. Recently, preclinical, translational, and clinical studies have indicated that alterations in the structural composition or function of the microbiome can contribute to the development of mental illness, including depression-like behavior, and thus, is a vital component linking the gut-brain axis. Consistent with this, strong correlations have been identified between alterations in gut microbiota and the development of chronic inflammatory disorders, such as IBD, suggesting that dysbiosis is an important factor in both gastrointestinal disorders and mental health. However, the precise mechanisms by which gut dysbiosis in IBD may alter behavior remains poorly understood.

How peripheral inflammation, as occurs in IBD, leads to remote changes in brain function remains unclear and, as a result, there are limited therapeutic options available clinically to address this issue. A number of general pathways have been described that link systemic inflammation to changes occurring in the brain, which in turn give rise to altered behavior. These pathways traditionally have included signaling via neural pathways (mainly vagal nerve afferents) and immune signaling (mainly via circulating cytokines like TNFα, which either enter the brain directly or activate cerebral endothelium). Recently, University of Calgary researchers described a novel inflammation signaling pathway which involves increased peripheral TNF-α production driving increased microglial activation, followed by monocyte recruitment into brain vasculature and brain parenchyma, which in turn drives the development of sickness behaviors. TNFα is a multifunctional cytokine that mediates a range of effects that can directly impact brain function (via modulating neurotransmission) and the gut (e.g. affecting intestinal barrier permeability) and modulate tissue inflammation (e.g. activate macrophages to induce cytokine and chemokine expression). Increased circulating levels of TNFα and elevated production by circulating leukocytes are commonly documented in patients with chronic inflammatory conditions associated with a high prevalence of sickness behaviors and mood disorders, including IBD. Hence, blockade of TNFα signaling has been extensively evaluated in clinical studies of IBD. In addition, improvement in symptoms by TNFα signaling inhibition are often evident in treated patients with chronic inflammatory diseases, prior to overt changes in disease activity. In patients with IBD, anti-TNFα therapy was associated with significant improvements in sleep, depression, and anxiety early after initiation of therapy, and these improvements were sustained. Moreover, another study assessed the effect of anti-TNFα therapy on interoceptive signaling in patients with Crohn's disease and demonstrated reductions in visceral sensitivity and improved cognitive-affective processing after anti-TNFα administration, paralleled by improved sense of well being. Changes in cognition were linked to changes in neural activity in prefrontal and limbic brain areas. Moreover, the rapid behavioral and neural changes observed were not associated with significant changes in fecal calprotectin levels (marker of gut inflammation) suggesting they were unlikely related to a simultaneous reduction in intestinal inflammation. Anti-TNFα therapy also alters the gut microbiome in patients with IBD. Specifically, anti-TNFα therapy in IBD patients increased gut microbial species richness and phylodiversity to levels similar to healthy controls. In addition, anti-TNF therapy decreased the relative abundances of proinflammatory bacteria Escherichia and Enterococcus in IBD patients, and increasing genera that produce short-chain fatty acids, which have well-established anti-inflammatory effects in the gut.

Healthy cerebral endothelial cells (CECs) are critically important for brain blood flow regulation, as well as the normal function of the blood-brain barrier (BBB). Cytokines, including TNFα, originating from within the central nervous system or present in the peripheral blood circulation, can induce CEC dysfunction and increase BBB permeability, which is known to be associated with many neurological disorders including Alzheimer's and MS, as well as peripherally induced neuroinflammation. Indeed, CECs exposed to TNFα express fewer interendothelial proteins and have greater reactive oxygen species generation, leading to increased BBB permeability. Furthermore, CECs can be activated by cytokines that are either present in the blood or released by circulating immune cells in intimate contact with CECs, to produce secondary signaling molecules (e.g., additional cytokines, prostaglandin E2, nitric oxide) which interact with cells within the brain to alter NVC responses. Consistent with this suggestion, in a mouse model of peripheral inflammation, it was shown that activated monocytes within the cerebral circulation adhere to CECs and induce CEC activation through TNFα-TNF receptor-1 interactions. This immune cell and TNFα-driven CEC activation up-regulates inducible nitric oxide synthase expression within CECs and was directly linked to activation of microglia within the brain (especially those cells in close proximity to blood vessels), and ultimately to altered behavior. These previous findings from this group provide a mechanism by which peripheral inflammation can impact the function of CECs, thereby altering cerebrovascular function. Blocking activated immune cells within the circulation from adhering to CECs increases task-induced cortical blood flow (i.e., NVC)66, suggesting that immune cell-CEC adhesive interactions can lead to neurovascular uncoupling. CECs can also play an essential role in the regulation of cerebral blood flow by passively allowing vessel smooth muscle to relax and contract. Interestingly, activated leukocytes have been shown to promote endothelial cell-dependent vasospasticity and reduce arterial relaxation in vitro and in vivo. Further, endothelial cell-dependent vasospasticity is heightened in the presence of protein aggregates in the blood, which would effectively reduce the dynamic range of vasodilation. This is particularly interesting considering previous findings that peripheral inflammation leads to increased CEC presentation of the adhesion molecule P-selectin, which in turn promotes leukocyte and platelet adhesion to CECs. Taken together, these findings suggest that increased leukocyte-CEC interactions associated with peripheral inflammation may significantly contribute to altered cerebrovascular dynamics and potential neurovascular uncoupling in the setting of peripheral inflammatory disease, which is manifest as altered cerebral oxygenation.

Considered in the context of neuroinflammation associated with chronic peripheral inflammation, alterations in cortical perfusion and oxygenation may contribute, at least in part, to the behavioral symptoms associated with IBD and disease-associated reductions in health related quality of life (QoL).

The IBD clinic is routinely testing new treatments for IBD. They have agreed to integrate NIRS into a pilot protocol to determine if there is sufficient evidence to warrant a longer trial of NIRS as a biomarker of neurological involvement in IBD.

The investigators propose the following series of experiments, using NIRS, to determine (A) whether patients with IBD exhibit reduced cortical oxygen saturation and altered patterns of microvascular cerebral blood perfusion; (B) whether NIRS findings of oxygen saturation and perfusion correlate with clinical disease activity markers, a disease-specific gut microbiome signature, and/or symptom and quality of life scores in patients with IBD; (C) the impact of anti-TNF therapy (administered as clinically indicated, as standard of practice) on NIRS changes, the fecal microbiome, markers of systemic inflammation, and symptom severity.

Study Type

Observational

Enrollment (Anticipated)

40

Contacts and Locations

This section provides the contact details for those conducting the study, and information on where this study is being conducted.

Study Contact

Study Contact Backup

Study Locations

    • Alberta
      • Calgary, Alberta, Canada, T2N 4Z6
        • Recruiting
        • University of Calgary
        • Contact:
          • Blessing Odia
          • Phone Number: 403-220-5782

Participation Criteria

Researchers look for people who fit a certain description, called eligibility criteria. Some examples of these criteria are a person's general health condition or prior treatments.

Eligibility Criteria

Ages Eligible for Study

18 years to 85 years (ADULT, OLDER_ADULT)

Accepts Healthy Volunteers

No

Genders Eligible for Study

All

Sampling Method

Non-Probability Sample

Study Population

Participants will have either Ulcerative Colitis or Crohn's Disease

Description

Inclusion Criteria:

Eligible patients will be:

  • > 18 years of age with moderate-to-severe UC or (partial Mayo score [excluding endoscopy] ≥5 with rectal bleeding subscore ≥1; or total Mayo subscore 6-12 with RBS ≥1) or CD (Harvey-Bradshaw index [HBI] of 7 or greater, and active CD confirmed on POC bowel ultrasound (defined by bowel wall thickness >3mm and positive Color doppler signal and a fecal calprotectin > 50 μg/g ).
  • based on their active disease status patients cannot be taking > 20 mg prednisone daily and
  • must be eligible for anti-TNF therapy as per standard of care (clinical decision made by IBD specialist caring for the patient).

Exclusion Criteria:

  • patients with severely active UC (clinical signs of fulminant colitis or toxic megacolon) or CD (HBI > 16), requiring > 20 mg of prednisone daily at induction, suicidal ideation or psychosis.

Study Plan

This section provides details of the study plan, including how the study is designed and what the study is measuring.

How is the study designed?

Design Details

Cohorts and Interventions

Group / Cohort
Intervention / Treatment
IBD Patients (UC and CD)

Participants will complete a Mayo Clinic Score (UC) or HBI score (CD), short IBDQ73, *EQ5D-5L, *GAD-774, *PHQ-975, *PROMIS (Gastrointestinal Belly Pain), Multidimensional Assessment of Interoceptive Awareness (MAIA)77, *Pain Catastrophizing Scale, Pittsburg Sleep Quality Index (PSQI) and Fatigue Severity Scale (FSS) at baseline and 16 weeks after the start of anti-TNF therapy (*questionnaires available through the CIHR IMAGINE grant).

Stool will be collected at baseline and after 16 weeks for assessment of the known biomarker fecal calprotectin, in addition to fecal bacterial and fungal microbiome (through the International Microbiome Center [IMC], U of C). Blood will be drawn and urine collected at baseline and after 16 weeks for inflammatory markers and metabolomic [IMC] analysis

Mayo Clinic Score (UC) or HBI score (CD), short IBDQ73, *EQ5D-5L, *GAD-774, *PHQ-975, *PROMIS (Gastrointestinal Belly Pain), Multidimensional Assessment of Interoceptive Awareness (MAIA)77, *Pain Catastrophizing Scale, Pittsburg Sleep Quality Index (PSQI) and Fatigue Severity Scale (FSS) at baseline and 16 weeks after the start of anti-TNF therapy
A TechEn NIRSOptix continuous-wave fNIRS system will be used to record changes in cerebral oxygenation, at a sampling rate of 25 Hz
For assessment of the known biomarker fecal calprotectin, in addition to fecal bacterial and fungal microbiome
for inflammatory markers and metabolomic [IMC] analysis

What is the study measuring?

Primary Outcome Measures

Outcome Measure
Measure Description
Time Frame
a) Clinically: CD (decrease of >3 points in the modified Harvey-Bradshaw Index); UC (decrease of >2 points in the partial Mayo Score)
Time Frame: 16 weeks

Defining differences in alterations of resting state brain NIRS findings occurring in IBD patients, between baseline and at 16 weeks after starting anti-TNF therapy, in anti-TNF responders vs. non-responders, defined:

a) Clinically: CD (decrease of >3 points in the modified Harvey-Bradshaw Index); UC (decrease of >2 points in the partial Mayo Score)

16 weeks
b) Radiographically: decrease in bowel wall thickness ≥25% in most affected segment with 2 point decrease in color Doppler signal score or normalization
Time Frame: 16 weeks

Defining differences in alterations of resting state brain NIRS findings occurring in IBD patients, between baseline and at 16 weeks after starting anti-TNF therapy, in anti-TNF responders vs. non-responders, defined:

b) Radiographically: decrease in bowel wall thickness ≥25% in most affected segment with 2 point decrease in color Doppler signal score or normalization

16 weeks
c) Biomarker: decrease in fecal calprotectin >50% from baseline or ≤250 ug/g
Time Frame: 16 weeks

Defining differences in alterations of resting state brain NIRS findings occurring in IBD patients, between baseline and at 16 weeks after starting anti-TNF therapy, in anti-TNF responders vs. non-responders, defined:

c) Biomarker: decrease in fecal calprotectin >50% from baseline or ≤250 ug/g

16 weeks
d) Endoscopically (where available): CD (decrease in Simple Endoscopic Score for CD ≥50% from baseline); UC (decrease in Mayo endoscopic subscore ≥1 point from baseline)
Time Frame: 16 weeks

Defining differences in alterations of resting state brain NIRS findings occurring in IBD patients, between baseline and at 16 weeks after starting anti-TNF therapy, in anti-TNF responders vs. non-responders, defined:

d) Endoscopically (where available): CD (decrease in Simple Endoscopic Score for CD ≥50% from baseline); UC (decrease in Mayo endoscopic subscore ≥1 point from baseline)

16 weeks

Secondary Outcome Measures

Outcome Measure
Measure Description
Time Frame
(i) alterations in markers of bowel (defined by bowel ultrasound) and systemic inflammation.
Time Frame: 16 weeks

Delineating whether baseline brain NIRS findings, and changes in NIRS findings from baseline to 16 weeks after starting anti-TNF therapy, correlate with:

(i) alterations in markers of bowel (defined by bowel ultrasound) and systemic inflammation.

16 weeks
(ii) reduction from baseline measurement of fecal calprotectin level.
Time Frame: 16 weeks

Delineating whether baseline brain NIRS findings, and changes in NIRS findings from baseline to 16 weeks after starting anti-TNF therapy, correlate with:

(ii) reduction from baseline measurement of fecal calprotectin level.

16 weeks
(iii) specific gut microbiome and metabolomics signatures.
Time Frame: 16 weeks

Delineating whether baseline brain NIRS findings, and changes in NIRS findings from baseline to 16 weeks after starting anti-TNF therapy, correlate with:

(iii) specific gut microbiome and metabolomics signatures.

16 weeks
(iv) changes in symptom severity in IBD patients that respond (defined as above), or do not respond to anti-TNF therapy.
Time Frame: 16 weeks

Delineating whether baseline brain NIRS findings, and changes in NIRS findings from baseline to 16 weeks after starting anti-TNF therapy, correlate with:

(iv) changes in symptom severity in IBD patients that respond (defined as above), or do not respond to anti-TNF therapy.

16 weeks

Collaborators and Investigators

This is where you will find people and organizations involved with this study.

Investigators

  • Principal Investigator: Mark Swain, MD, University of Calgary

Publications and helpful links

The person responsible for entering information about the study voluntarily provides these publications. These may be about anything related to the study.

General Publications

Study record dates

These dates track the progress of study record and summary results submissions to ClinicalTrials.gov. Study records and reported results are reviewed by the National Library of Medicine (NLM) to make sure they meet specific quality control standards before being posted on the public website.

Study Major Dates

Study Start (ANTICIPATED)

April 26, 2021

Primary Completion (ANTICIPATED)

January 5, 2025

Study Completion (ANTICIPATED)

January 5, 2026

Study Registration Dates

First Submitted

November 23, 2020

First Submitted That Met QC Criteria

January 28, 2021

First Posted (ACTUAL)

February 2, 2021

Study Record Updates

Last Update Posted (ACTUAL)

April 28, 2021

Last Update Submitted That Met QC Criteria

April 26, 2021

Last Verified

November 1, 2020

More Information

Terms related to this study

Plan for Individual participant data (IPD)

Plan to Share Individual Participant Data (IPD)?

NO

Drug and device information, study documents

Studies a U.S. FDA-regulated drug product

No

Studies a U.S. FDA-regulated device product

No

This information was retrieved directly from the website clinicaltrials.gov without any changes. If you have any requests to change, remove or update your study details, please contact register@clinicaltrials.gov. As soon as a change is implemented on clinicaltrials.gov, this will be updated automatically on our website as well.

Clinical Trials on IBD

Clinical Trials on Quality of life questionnaires

3
Subscribe