Allowed Publications
Slot System
Featured Buckets
Featured Buckets Admin

Mentorship key to improving GI, hepatology workforce diversity

Article Type
Changed
Wed, 11/23/2022 - 14:51

Increasing mentorship opportunities for gastroenterology and hepatology residents and medical students from populations underrepresented in medicine is essential to increase diversity in the specialty and improve health disparities among patients, according to a special report published simultaneously in Gastroenterology and three other journals.

“This study helps to establish priorities for diversity, equity and inclusion in our field and informs future interventions to improve workforce diversity and eliminate health care disparities among the patients we serve,” Folasade P. May, MD, PhD, MPhil, the study’s corresponding author and an associate professor of medicine at the University of California, Los Angeles, said in a prepared statement.

The report, the result of a partnership between researchers at UCLA and the Intersociety Group on Diversity, reveals the findings of a survey aimed at assessing current perspectives on individuals underrepresented in medicine and health equity within gastroenterology and hepatology. The collaboration involved five gastroenterology professional societies: the American Association for the Study of Liver Disease; American College of Gastroenterology; American Gastroenterological Association; American Society of Gastrointestinal Endoscopy; and North American Society for Pediatric Gastroenterology, Hepatology and Nutrition.

”The current racial and ethnic composition of the GI and hepatology workforce does not reflect the population of patients served or the current matriculants in medicine,” Harman K. Rahal, MD, of UCLA and Cedars-Sinai Medical Center, Los Angeles, and James H. Tabibian, MD, PhD, of UCLA and Olive View–UCLA Medical Center, and colleagues wrote. “As there are several conditions in GI and hepatology with disparities in incidence, treatment, and outcomes, representation of UIM [underrepresented in medicine] individuals is critical to address health disparities.”

The term “underrepresented in medicine” is defined by the Association of American Medical Colleges as “those racial and ethnic populations that are underrepresented in the medical profession relative to their numbers in the general population.” The authors explained that these groups “have traditionally included Latino (i.e., Latino/a/x), Black (or African American), Native American (namely, American Indian, Alaska Native, and Native Hawaiian), Pacific Islander, and mainland Puerto Rican individuals.”

The five gastroenterology and hepatology societies partnered with investigators at UCLA to develop a 33-question electronic survey “to determine perspectives of current racial, ethnic, and gender diversity within GI and hepatology; to assess current views on interventions needed to increase racial, ethnic, and gender diversity in the field; and to collect data on the experiences of UIM individuals and women in our field,” according to the report’s authors. The survey was then distributed to members of those societies, with 1,219 respondents.

The report found that inadequate representation of people from those underrepresented groups in the education and training pipeline was the most frequently reported barrier to improving racial and ethnic diversity in the field (35.4%), followed by insufficient racial and ethnic minority group representation in professional leadership (27.9%) and insufficient racial and ethnic minority group representation among practicing GI and hepatology professionals in the workplace (26.6%). Only 9% of fellows in GI and hepatology are from groups underrepresented in medicine, according to data from the Accreditation Council for Graduate Medical Education. Furthermore, one study has shown that the proportion of UIM in academic faculty has never exceeded 10% at each academic rank; there has even been a decline recently among junior academic faculty positions. That study also found that only 9% of academic gastroenterologists in the United states identify as underrepresented in medicine, with little change over the last decade.

Potential contributors to this low level of representation, the authors wrote, include “lack of racial and ethnic diversity in the medical training pipeline, nondiverse leadership, bias, racial discrimination, and the notion that UIM physicians may be less likely to promote themselves or be promoted.”

Another potential contributor, however, may be complacency within the field about the need to improve diversity and taking actions to do so.

 

 


A majority of White physicians (78%) were very or somewhat satisfied with current levels of workforce diversity, compared with a majority of Black physicians (63%) feeling very or somewhat unsatisfied.

This disconnect was not surprising to Aja McCutchen, MD, a partner at Atlanta Gastroenterology Associates who was not involved in the survey.

“One cannot discount the lived experience of a [person underrepresented in medicine] as it relates to recognizing conscious and unconscious biases, microaggression recognition, and absence of [underrepresented clinicians] in key positions. This is a reality that I do see on a daily basis,” Dr. McCutchen said in an interview.
Dr. Aja McCutchen

Only 35% of respondents felt there is “insufficient racial and ethnic representation in education and training,” and just over a quarter (28%) felt the same about representation in leadership. In fact, most respondents (59.7%) thought that racial and ethnic diversity had increased over the past 5 years even though data show no change, the authors noted.

Although Dr. McCutchen appreciated the broad recognition from respondents, regardless of background, to improve diversity in the pipeline, she noted that “retention of current talent and future talent would also require cultural shifts in understanding the challenges of the [underrepresented] members,” Dr. McCutchen said.

Again, however, the majority of the respondents (64.6%) were themselves not members of underrepresented groups. Nearly half the respondents (48.7%) were non-Hispanic White, and one in five (22.5%) were Asian, Native Hawaiian, or Pacific Islander. The remaining respondents, making up less than a third of the total, were Hispanic (10.6%), Black (9.1%), American Indian or Alaskan Native (0.2%), another race/ethnicity (3.3%), or preferred not to answer (5.7%).

Dr. McCutchen said she had mixed feelings about the survey overall.

“On the one hand, I was eager to read the perceptions of survey respondents as it relates to diversity, equity and inclusion in the GI space as very little cross-organizational data exists,” said Dr. McCutchen. “On the other hand, the responses reminded me that there is a lot of work to be done as I expected more dissatisfaction with the current GI workforce in both academia and private practice respondents.”

She was surprised, for example, that nearly three-quarters of the respondents were somewhat or very satisfied, and that a majority thought racial and ethnic diversity had increased.

Studies on provider-patient concordance have shown that patients feel it’s important to share common ground with their physicians particularly in terms of race, ethnicity and language, the authors noted.

“This patient preference underscores the need to recruit and train a more diverse cohort of trainees into GI and hepatology fellowships if the desired goal is to optimize patient care and combat health disparities,” they wrote. They pointed out that cultural understanding can influence how patients perceive their health, symptoms, and concerns, which can then affect providers’ diagnostic accuracy and treatment recommendations. In turn, patients may have better adherence to treatment recommendations when they share a similar background as their clinician.

“Diversity in medicine also leads to greater diversity in thoughts, better returns on investments, increased scholarly activities related to health equity to name a few,” Dr. McCutchen said.

The top recommendations from respondents for improving representation of currently underrepresented individuals in GI and hepatology were to increase mentorship opportunities for residents (45%) and medical students (43%) from these groups and to increase representation of professionals from these backgrounds in program and professional society leadership (39%). A third of respondents also recommended increasing shadowing opportunities for undergraduate students from these underrepresented populations.

Dr. McCutchen expressed optimism regarding the initiatives to improve diversity, equity and inclusion across the gastroenterology spectrum.

“It is incumbent upon all of us to continue to be the driving force of change, which will be a journey and not a destination,” McCutchen said. “In the future, diversity, equity and inclusion will be the expectation, and we will ultimately move closer to the goal of completely eliminating health care inequities.”

The research was funded by the National Cancer Institute, the UCLA Jonsson Comprehensive Cancer Center, and Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research Ablon Scholars Program. The authors reported no conflicts of interest. Dr. McCutchen disclosed relationships with Bristol-Myers Squibb and Redhill Biopharmaceuticals.
Publications
Topics
Sections

Increasing mentorship opportunities for gastroenterology and hepatology residents and medical students from populations underrepresented in medicine is essential to increase diversity in the specialty and improve health disparities among patients, according to a special report published simultaneously in Gastroenterology and three other journals.

“This study helps to establish priorities for diversity, equity and inclusion in our field and informs future interventions to improve workforce diversity and eliminate health care disparities among the patients we serve,” Folasade P. May, MD, PhD, MPhil, the study’s corresponding author and an associate professor of medicine at the University of California, Los Angeles, said in a prepared statement.

The report, the result of a partnership between researchers at UCLA and the Intersociety Group on Diversity, reveals the findings of a survey aimed at assessing current perspectives on individuals underrepresented in medicine and health equity within gastroenterology and hepatology. The collaboration involved five gastroenterology professional societies: the American Association for the Study of Liver Disease; American College of Gastroenterology; American Gastroenterological Association; American Society of Gastrointestinal Endoscopy; and North American Society for Pediatric Gastroenterology, Hepatology and Nutrition.

”The current racial and ethnic composition of the GI and hepatology workforce does not reflect the population of patients served or the current matriculants in medicine,” Harman K. Rahal, MD, of UCLA and Cedars-Sinai Medical Center, Los Angeles, and James H. Tabibian, MD, PhD, of UCLA and Olive View–UCLA Medical Center, and colleagues wrote. “As there are several conditions in GI and hepatology with disparities in incidence, treatment, and outcomes, representation of UIM [underrepresented in medicine] individuals is critical to address health disparities.”

The term “underrepresented in medicine” is defined by the Association of American Medical Colleges as “those racial and ethnic populations that are underrepresented in the medical profession relative to their numbers in the general population.” The authors explained that these groups “have traditionally included Latino (i.e., Latino/a/x), Black (or African American), Native American (namely, American Indian, Alaska Native, and Native Hawaiian), Pacific Islander, and mainland Puerto Rican individuals.”

The five gastroenterology and hepatology societies partnered with investigators at UCLA to develop a 33-question electronic survey “to determine perspectives of current racial, ethnic, and gender diversity within GI and hepatology; to assess current views on interventions needed to increase racial, ethnic, and gender diversity in the field; and to collect data on the experiences of UIM individuals and women in our field,” according to the report’s authors. The survey was then distributed to members of those societies, with 1,219 respondents.

The report found that inadequate representation of people from those underrepresented groups in the education and training pipeline was the most frequently reported barrier to improving racial and ethnic diversity in the field (35.4%), followed by insufficient racial and ethnic minority group representation in professional leadership (27.9%) and insufficient racial and ethnic minority group representation among practicing GI and hepatology professionals in the workplace (26.6%). Only 9% of fellows in GI and hepatology are from groups underrepresented in medicine, according to data from the Accreditation Council for Graduate Medical Education. Furthermore, one study has shown that the proportion of UIM in academic faculty has never exceeded 10% at each academic rank; there has even been a decline recently among junior academic faculty positions. That study also found that only 9% of academic gastroenterologists in the United states identify as underrepresented in medicine, with little change over the last decade.

Potential contributors to this low level of representation, the authors wrote, include “lack of racial and ethnic diversity in the medical training pipeline, nondiverse leadership, bias, racial discrimination, and the notion that UIM physicians may be less likely to promote themselves or be promoted.”

Another potential contributor, however, may be complacency within the field about the need to improve diversity and taking actions to do so.

 

 


A majority of White physicians (78%) were very or somewhat satisfied with current levels of workforce diversity, compared with a majority of Black physicians (63%) feeling very or somewhat unsatisfied.

This disconnect was not surprising to Aja McCutchen, MD, a partner at Atlanta Gastroenterology Associates who was not involved in the survey.

“One cannot discount the lived experience of a [person underrepresented in medicine] as it relates to recognizing conscious and unconscious biases, microaggression recognition, and absence of [underrepresented clinicians] in key positions. This is a reality that I do see on a daily basis,” Dr. McCutchen said in an interview.
Dr. Aja McCutchen

Only 35% of respondents felt there is “insufficient racial and ethnic representation in education and training,” and just over a quarter (28%) felt the same about representation in leadership. In fact, most respondents (59.7%) thought that racial and ethnic diversity had increased over the past 5 years even though data show no change, the authors noted.

Although Dr. McCutchen appreciated the broad recognition from respondents, regardless of background, to improve diversity in the pipeline, she noted that “retention of current talent and future talent would also require cultural shifts in understanding the challenges of the [underrepresented] members,” Dr. McCutchen said.

Again, however, the majority of the respondents (64.6%) were themselves not members of underrepresented groups. Nearly half the respondents (48.7%) were non-Hispanic White, and one in five (22.5%) were Asian, Native Hawaiian, or Pacific Islander. The remaining respondents, making up less than a third of the total, were Hispanic (10.6%), Black (9.1%), American Indian or Alaskan Native (0.2%), another race/ethnicity (3.3%), or preferred not to answer (5.7%).

Dr. McCutchen said she had mixed feelings about the survey overall.

“On the one hand, I was eager to read the perceptions of survey respondents as it relates to diversity, equity and inclusion in the GI space as very little cross-organizational data exists,” said Dr. McCutchen. “On the other hand, the responses reminded me that there is a lot of work to be done as I expected more dissatisfaction with the current GI workforce in both academia and private practice respondents.”

She was surprised, for example, that nearly three-quarters of the respondents were somewhat or very satisfied, and that a majority thought racial and ethnic diversity had increased.

Studies on provider-patient concordance have shown that patients feel it’s important to share common ground with their physicians particularly in terms of race, ethnicity and language, the authors noted.

“This patient preference underscores the need to recruit and train a more diverse cohort of trainees into GI and hepatology fellowships if the desired goal is to optimize patient care and combat health disparities,” they wrote. They pointed out that cultural understanding can influence how patients perceive their health, symptoms, and concerns, which can then affect providers’ diagnostic accuracy and treatment recommendations. In turn, patients may have better adherence to treatment recommendations when they share a similar background as their clinician.

“Diversity in medicine also leads to greater diversity in thoughts, better returns on investments, increased scholarly activities related to health equity to name a few,” Dr. McCutchen said.

The top recommendations from respondents for improving representation of currently underrepresented individuals in GI and hepatology were to increase mentorship opportunities for residents (45%) and medical students (43%) from these groups and to increase representation of professionals from these backgrounds in program and professional society leadership (39%). A third of respondents also recommended increasing shadowing opportunities for undergraduate students from these underrepresented populations.

Dr. McCutchen expressed optimism regarding the initiatives to improve diversity, equity and inclusion across the gastroenterology spectrum.

“It is incumbent upon all of us to continue to be the driving force of change, which will be a journey and not a destination,” McCutchen said. “In the future, diversity, equity and inclusion will be the expectation, and we will ultimately move closer to the goal of completely eliminating health care inequities.”

The research was funded by the National Cancer Institute, the UCLA Jonsson Comprehensive Cancer Center, and Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research Ablon Scholars Program. The authors reported no conflicts of interest. Dr. McCutchen disclosed relationships with Bristol-Myers Squibb and Redhill Biopharmaceuticals.

Increasing mentorship opportunities for gastroenterology and hepatology residents and medical students from populations underrepresented in medicine is essential to increase diversity in the specialty and improve health disparities among patients, according to a special report published simultaneously in Gastroenterology and three other journals.

“This study helps to establish priorities for diversity, equity and inclusion in our field and informs future interventions to improve workforce diversity and eliminate health care disparities among the patients we serve,” Folasade P. May, MD, PhD, MPhil, the study’s corresponding author and an associate professor of medicine at the University of California, Los Angeles, said in a prepared statement.

The report, the result of a partnership between researchers at UCLA and the Intersociety Group on Diversity, reveals the findings of a survey aimed at assessing current perspectives on individuals underrepresented in medicine and health equity within gastroenterology and hepatology. The collaboration involved five gastroenterology professional societies: the American Association for the Study of Liver Disease; American College of Gastroenterology; American Gastroenterological Association; American Society of Gastrointestinal Endoscopy; and North American Society for Pediatric Gastroenterology, Hepatology and Nutrition.

”The current racial and ethnic composition of the GI and hepatology workforce does not reflect the population of patients served or the current matriculants in medicine,” Harman K. Rahal, MD, of UCLA and Cedars-Sinai Medical Center, Los Angeles, and James H. Tabibian, MD, PhD, of UCLA and Olive View–UCLA Medical Center, and colleagues wrote. “As there are several conditions in GI and hepatology with disparities in incidence, treatment, and outcomes, representation of UIM [underrepresented in medicine] individuals is critical to address health disparities.”

The term “underrepresented in medicine” is defined by the Association of American Medical Colleges as “those racial and ethnic populations that are underrepresented in the medical profession relative to their numbers in the general population.” The authors explained that these groups “have traditionally included Latino (i.e., Latino/a/x), Black (or African American), Native American (namely, American Indian, Alaska Native, and Native Hawaiian), Pacific Islander, and mainland Puerto Rican individuals.”

The five gastroenterology and hepatology societies partnered with investigators at UCLA to develop a 33-question electronic survey “to determine perspectives of current racial, ethnic, and gender diversity within GI and hepatology; to assess current views on interventions needed to increase racial, ethnic, and gender diversity in the field; and to collect data on the experiences of UIM individuals and women in our field,” according to the report’s authors. The survey was then distributed to members of those societies, with 1,219 respondents.

The report found that inadequate representation of people from those underrepresented groups in the education and training pipeline was the most frequently reported barrier to improving racial and ethnic diversity in the field (35.4%), followed by insufficient racial and ethnic minority group representation in professional leadership (27.9%) and insufficient racial and ethnic minority group representation among practicing GI and hepatology professionals in the workplace (26.6%). Only 9% of fellows in GI and hepatology are from groups underrepresented in medicine, according to data from the Accreditation Council for Graduate Medical Education. Furthermore, one study has shown that the proportion of UIM in academic faculty has never exceeded 10% at each academic rank; there has even been a decline recently among junior academic faculty positions. That study also found that only 9% of academic gastroenterologists in the United states identify as underrepresented in medicine, with little change over the last decade.

Potential contributors to this low level of representation, the authors wrote, include “lack of racial and ethnic diversity in the medical training pipeline, nondiverse leadership, bias, racial discrimination, and the notion that UIM physicians may be less likely to promote themselves or be promoted.”

Another potential contributor, however, may be complacency within the field about the need to improve diversity and taking actions to do so.

 

 


A majority of White physicians (78%) were very or somewhat satisfied with current levels of workforce diversity, compared with a majority of Black physicians (63%) feeling very or somewhat unsatisfied.

This disconnect was not surprising to Aja McCutchen, MD, a partner at Atlanta Gastroenterology Associates who was not involved in the survey.

“One cannot discount the lived experience of a [person underrepresented in medicine] as it relates to recognizing conscious and unconscious biases, microaggression recognition, and absence of [underrepresented clinicians] in key positions. This is a reality that I do see on a daily basis,” Dr. McCutchen said in an interview.
Dr. Aja McCutchen

Only 35% of respondents felt there is “insufficient racial and ethnic representation in education and training,” and just over a quarter (28%) felt the same about representation in leadership. In fact, most respondents (59.7%) thought that racial and ethnic diversity had increased over the past 5 years even though data show no change, the authors noted.

Although Dr. McCutchen appreciated the broad recognition from respondents, regardless of background, to improve diversity in the pipeline, she noted that “retention of current talent and future talent would also require cultural shifts in understanding the challenges of the [underrepresented] members,” Dr. McCutchen said.

Again, however, the majority of the respondents (64.6%) were themselves not members of underrepresented groups. Nearly half the respondents (48.7%) were non-Hispanic White, and one in five (22.5%) were Asian, Native Hawaiian, or Pacific Islander. The remaining respondents, making up less than a third of the total, were Hispanic (10.6%), Black (9.1%), American Indian or Alaskan Native (0.2%), another race/ethnicity (3.3%), or preferred not to answer (5.7%).

Dr. McCutchen said she had mixed feelings about the survey overall.

“On the one hand, I was eager to read the perceptions of survey respondents as it relates to diversity, equity and inclusion in the GI space as very little cross-organizational data exists,” said Dr. McCutchen. “On the other hand, the responses reminded me that there is a lot of work to be done as I expected more dissatisfaction with the current GI workforce in both academia and private practice respondents.”

She was surprised, for example, that nearly three-quarters of the respondents were somewhat or very satisfied, and that a majority thought racial and ethnic diversity had increased.

Studies on provider-patient concordance have shown that patients feel it’s important to share common ground with their physicians particularly in terms of race, ethnicity and language, the authors noted.

“This patient preference underscores the need to recruit and train a more diverse cohort of trainees into GI and hepatology fellowships if the desired goal is to optimize patient care and combat health disparities,” they wrote. They pointed out that cultural understanding can influence how patients perceive their health, symptoms, and concerns, which can then affect providers’ diagnostic accuracy and treatment recommendations. In turn, patients may have better adherence to treatment recommendations when they share a similar background as their clinician.

“Diversity in medicine also leads to greater diversity in thoughts, better returns on investments, increased scholarly activities related to health equity to name a few,” Dr. McCutchen said.

The top recommendations from respondents for improving representation of currently underrepresented individuals in GI and hepatology were to increase mentorship opportunities for residents (45%) and medical students (43%) from these groups and to increase representation of professionals from these backgrounds in program and professional society leadership (39%). A third of respondents also recommended increasing shadowing opportunities for undergraduate students from these underrepresented populations.

Dr. McCutchen expressed optimism regarding the initiatives to improve diversity, equity and inclusion across the gastroenterology spectrum.

“It is incumbent upon all of us to continue to be the driving force of change, which will be a journey and not a destination,” McCutchen said. “In the future, diversity, equity and inclusion will be the expectation, and we will ultimately move closer to the goal of completely eliminating health care inequities.”

The research was funded by the National Cancer Institute, the UCLA Jonsson Comprehensive Cancer Center, and Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research Ablon Scholars Program. The authors reported no conflicts of interest. Dr. McCutchen disclosed relationships with Bristol-Myers Squibb and Redhill Biopharmaceuticals.
Publications
Publications
Topics
Article Type
Sections
Article Source

FROM GASTROENTEROLOGY

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Looking for the source of neuroendocrine tumors

Reprogramming cells toward a neuroendocrine fate
Article Type
Changed
Tue, 10/11/2022 - 16:50

The diversity of neuroendocrine tumors (NETs) – which includes variation in location, mutational profile, and response to therapy – may be due to divergent cellular origins in different tissue sites, according to a new study.

The pathogenesis of gastroenteropancreatic neoplasms (GEP-NENs) is poorly understood, in part because of a lack of modeling systems, according to Suzann Duan, PhD, and colleagues. They are a heterogeneous group of tumors that are increasingly prevalent in the United States. GEP-NENs arise from endocrine-producing cells and include gastric carcinoids, gastrinomas, and pancreatic NETs.

Despite the general mystery surrounding GEP-NENs, there is at least one clue in the form of the MEN1 gene. Both inherited and sporadic mutations of this gene are associated with GEP-NENs. Menin is a tumor suppressor protein, and previous studies have shown that inactivation of MEN1 leads to loss of that protein and is associated with endocrine tumors in the pancreas, pituitary, and upper GI tract.

In new research published in Cellular and Molecular Gastroenterology and Hepatology, researchers investigated the role of MEN1 in neuroendocrine cell development and traced it to a potential role in the development of NETs.

Patients with MEN1 mutations are at increased risk of gastrinomas, which lead to increased production of the peptide hormone gastrin. Gastrin increases acid production and can lead to hyperplasia in parietal and enterochromaffin cells. These generally develop in Brunner’s glands within the submucosa of the duodenum. At time of diagnosis, more than half of such tumors have developed lymph node metastases.

It remains unclear how loss of MEN1 suppresses gastrin production. Previous research showed that homozygous MEN1 deletion in mice is lethal to embryos, while leaving one copy intact leads to heightened risk of endocrine tumors in the pancreas and pituitary gland, but not in the GI tract. The studies did not reveal the tumor’s origin cell.

The researchers developed a novel mouse model in which MEN1 is conditionally deleted from the GI tract epithelium. This led to hyperplasia of gastrin-producing cells (G cells) in the antrum, as well as hypergastrinemia and development of gastric NETs. Exposure to a proton pump inhibitor accelerated gastric NET development, and the researchers identified expansion of enteric glial cells that expressed gastrin and GFAP. Glial cells that differentiated into endocrine phenotype were associated with a reversible loss of menin. “Taken together, these observations suggest that hyperplastic G cells might emerge from reprogrammed neural crest–derived cells in addition to endoderm-derived enteroendocrine cells,” the authors wrote.

That idea is supported by previous research indicating that multipotent glial cells expressing GFAP or SOX10 may play a developmental role in formation of neuroendocrine cells.

With this in mind, the researchers deleted MEN1 in GFAP-expressing cells to see if it would promote neuroendocrine cell development.

The result was hyperplasia in the gastric antrum and NETs in the pituitary and pancreas. To the researchers’ surprise, NET development was associated with loss of GFAP expression as well as activation of neuronal and neuroendocrine-related genes in the stomach, pancreas, and pituitary. There was universal reduction of GFAP protein expression in pituitary and pancreatic NETs, but GFAP transcript levels stayed steady in the gastric antra despite a reduction in GFAP-reporter expression. This could indicate that the menin protein interacts with GFAP. If so, eliminating menin in GFAP-positive cells could change the localization of GFAP, which may in turn lead to changes in glial cell identity.

When the researchers compared transcriptomes of hyperplastic antral tissues to well-differentiated NETs, they found that NETs exhibited a greater loss of glial-restricted progenitor lineage–associated genes as well as more downregulation of gliogenesis-directing factors. “Thus, the transition from a glial-to-neuronal cell phenotype appears to promote the progression from neuroendocrine cell hyperplasia to tumor development,” the authors wrote. They also found that NETs have higher levels of expression of genes associated with neural stem and progenitor cells, as well as upregulation of factors secreted from neural crest cells that promote neurogenesis and restrict the glial cell fate. Many of these factors are part of the Hedgehog signaling pathway, and menin is known to repress Hedgehog signaling.

Intestinal glial cells have a high degree of plasticity. They can become neuronal progenitor cells and yet they can dedifferentiate to differentiate again into other cell lineages.

The research could eventually lead to identification of unique cells-of-origin for these tumors. The authors say that the diversity of the tumors – which includes variation in location, mutational profile, and response to therapy – may be due to divergent cellular origins in different tissue sites. “Defining the cells-of-origin and the events preceding neoplastic transformation will be critical to informing molecular signaling pathways that can then be targeted therapeutically,” the authors wrote.

The authors disclosed no conflicts of interest.

Body

Gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs) share endocrine and neural features but are diverse in terms of their location, behaviors, and response to therapies. One explanation for heterogeneity in GEP-NENs is that they have diverse cellular origins. The study by Duan and colleagues suggests that glia could be a potential cell of origin in GEP-NENs. GEP-NEN development in the pancreas, pituitary, and upper gastrointestinal tract is associated with mutations in the Multiple Endocrine Neoplasia I (MEN1) gene that cause a loss of the tumor suppressor protein menin.

Dr. Brian D. Gulbransen
The authors found that deleting Men1 only in glial fibrillary acidic protein (GFAP)–expressing cells leads to the development of pancreatic and pituitary neuroendocrine tumors and changes to the epithelial lining of the stomach. These observations suggest a role for menin in glial development and/or maturation that, when lost, can contribute to cellular reprogramming toward a neuroendocrine fate. However, it is also possible that deleting Men1 affects the developmental trajectories of GFAP-expressing progenitor cells rather than reprogramming mature glia. Interestingly, tumor development and neuroendocrine reprogramming were only observed in the pituitary, pancreas, and stomach, and did not seem to occur in other organs with large populations of similar GFAP-positive cells such as the brain, spinal cord, or other peripheral organs. This seems to indicate specialized developmental roles of menin in these locations or that glia in the pituitary, pancreas, and stomach exhibit a heightened plastic potential that differs from other populations of glia.

The tumorigenic potential of GFAP-positive cells differs even between the pituitary, pancreas, and stomach since mice lacking Men1 in GFAP-positive cells did not develop gastrinomas while tumors were observed in the pituitary and pancreas. This could indicate that additional drivers are necessary to promote NENs in the intestine which are not required in other locations. These differences could be important when considering treatment strategies given the diverse nature of the cells and mechanisms involved.

Brian D. Gulbransen, PhD, is an associate professor in the department of physiology and an MSU Foundation Professor at Michigan State University, East Lansing. He has no conflicts.

Publications
Topics
Sections
Body

Gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs) share endocrine and neural features but are diverse in terms of their location, behaviors, and response to therapies. One explanation for heterogeneity in GEP-NENs is that they have diverse cellular origins. The study by Duan and colleagues suggests that glia could be a potential cell of origin in GEP-NENs. GEP-NEN development in the pancreas, pituitary, and upper gastrointestinal tract is associated with mutations in the Multiple Endocrine Neoplasia I (MEN1) gene that cause a loss of the tumor suppressor protein menin.

Dr. Brian D. Gulbransen
The authors found that deleting Men1 only in glial fibrillary acidic protein (GFAP)–expressing cells leads to the development of pancreatic and pituitary neuroendocrine tumors and changes to the epithelial lining of the stomach. These observations suggest a role for menin in glial development and/or maturation that, when lost, can contribute to cellular reprogramming toward a neuroendocrine fate. However, it is also possible that deleting Men1 affects the developmental trajectories of GFAP-expressing progenitor cells rather than reprogramming mature glia. Interestingly, tumor development and neuroendocrine reprogramming were only observed in the pituitary, pancreas, and stomach, and did not seem to occur in other organs with large populations of similar GFAP-positive cells such as the brain, spinal cord, or other peripheral organs. This seems to indicate specialized developmental roles of menin in these locations or that glia in the pituitary, pancreas, and stomach exhibit a heightened plastic potential that differs from other populations of glia.

The tumorigenic potential of GFAP-positive cells differs even between the pituitary, pancreas, and stomach since mice lacking Men1 in GFAP-positive cells did not develop gastrinomas while tumors were observed in the pituitary and pancreas. This could indicate that additional drivers are necessary to promote NENs in the intestine which are not required in other locations. These differences could be important when considering treatment strategies given the diverse nature of the cells and mechanisms involved.

Brian D. Gulbransen, PhD, is an associate professor in the department of physiology and an MSU Foundation Professor at Michigan State University, East Lansing. He has no conflicts.

Body

Gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs) share endocrine and neural features but are diverse in terms of their location, behaviors, and response to therapies. One explanation for heterogeneity in GEP-NENs is that they have diverse cellular origins. The study by Duan and colleagues suggests that glia could be a potential cell of origin in GEP-NENs. GEP-NEN development in the pancreas, pituitary, and upper gastrointestinal tract is associated with mutations in the Multiple Endocrine Neoplasia I (MEN1) gene that cause a loss of the tumor suppressor protein menin.

Dr. Brian D. Gulbransen
The authors found that deleting Men1 only in glial fibrillary acidic protein (GFAP)–expressing cells leads to the development of pancreatic and pituitary neuroendocrine tumors and changes to the epithelial lining of the stomach. These observations suggest a role for menin in glial development and/or maturation that, when lost, can contribute to cellular reprogramming toward a neuroendocrine fate. However, it is also possible that deleting Men1 affects the developmental trajectories of GFAP-expressing progenitor cells rather than reprogramming mature glia. Interestingly, tumor development and neuroendocrine reprogramming were only observed in the pituitary, pancreas, and stomach, and did not seem to occur in other organs with large populations of similar GFAP-positive cells such as the brain, spinal cord, or other peripheral organs. This seems to indicate specialized developmental roles of menin in these locations or that glia in the pituitary, pancreas, and stomach exhibit a heightened plastic potential that differs from other populations of glia.

The tumorigenic potential of GFAP-positive cells differs even between the pituitary, pancreas, and stomach since mice lacking Men1 in GFAP-positive cells did not develop gastrinomas while tumors were observed in the pituitary and pancreas. This could indicate that additional drivers are necessary to promote NENs in the intestine which are not required in other locations. These differences could be important when considering treatment strategies given the diverse nature of the cells and mechanisms involved.

Brian D. Gulbransen, PhD, is an associate professor in the department of physiology and an MSU Foundation Professor at Michigan State University, East Lansing. He has no conflicts.

Title
Reprogramming cells toward a neuroendocrine fate
Reprogramming cells toward a neuroendocrine fate

The diversity of neuroendocrine tumors (NETs) – which includes variation in location, mutational profile, and response to therapy – may be due to divergent cellular origins in different tissue sites, according to a new study.

The pathogenesis of gastroenteropancreatic neoplasms (GEP-NENs) is poorly understood, in part because of a lack of modeling systems, according to Suzann Duan, PhD, and colleagues. They are a heterogeneous group of tumors that are increasingly prevalent in the United States. GEP-NENs arise from endocrine-producing cells and include gastric carcinoids, gastrinomas, and pancreatic NETs.

Despite the general mystery surrounding GEP-NENs, there is at least one clue in the form of the MEN1 gene. Both inherited and sporadic mutations of this gene are associated with GEP-NENs. Menin is a tumor suppressor protein, and previous studies have shown that inactivation of MEN1 leads to loss of that protein and is associated with endocrine tumors in the pancreas, pituitary, and upper GI tract.

In new research published in Cellular and Molecular Gastroenterology and Hepatology, researchers investigated the role of MEN1 in neuroendocrine cell development and traced it to a potential role in the development of NETs.

Patients with MEN1 mutations are at increased risk of gastrinomas, which lead to increased production of the peptide hormone gastrin. Gastrin increases acid production and can lead to hyperplasia in parietal and enterochromaffin cells. These generally develop in Brunner’s glands within the submucosa of the duodenum. At time of diagnosis, more than half of such tumors have developed lymph node metastases.

It remains unclear how loss of MEN1 suppresses gastrin production. Previous research showed that homozygous MEN1 deletion in mice is lethal to embryos, while leaving one copy intact leads to heightened risk of endocrine tumors in the pancreas and pituitary gland, but not in the GI tract. The studies did not reveal the tumor’s origin cell.

The researchers developed a novel mouse model in which MEN1 is conditionally deleted from the GI tract epithelium. This led to hyperplasia of gastrin-producing cells (G cells) in the antrum, as well as hypergastrinemia and development of gastric NETs. Exposure to a proton pump inhibitor accelerated gastric NET development, and the researchers identified expansion of enteric glial cells that expressed gastrin and GFAP. Glial cells that differentiated into endocrine phenotype were associated with a reversible loss of menin. “Taken together, these observations suggest that hyperplastic G cells might emerge from reprogrammed neural crest–derived cells in addition to endoderm-derived enteroendocrine cells,” the authors wrote.

That idea is supported by previous research indicating that multipotent glial cells expressing GFAP or SOX10 may play a developmental role in formation of neuroendocrine cells.

With this in mind, the researchers deleted MEN1 in GFAP-expressing cells to see if it would promote neuroendocrine cell development.

The result was hyperplasia in the gastric antrum and NETs in the pituitary and pancreas. To the researchers’ surprise, NET development was associated with loss of GFAP expression as well as activation of neuronal and neuroendocrine-related genes in the stomach, pancreas, and pituitary. There was universal reduction of GFAP protein expression in pituitary and pancreatic NETs, but GFAP transcript levels stayed steady in the gastric antra despite a reduction in GFAP-reporter expression. This could indicate that the menin protein interacts with GFAP. If so, eliminating menin in GFAP-positive cells could change the localization of GFAP, which may in turn lead to changes in glial cell identity.

When the researchers compared transcriptomes of hyperplastic antral tissues to well-differentiated NETs, they found that NETs exhibited a greater loss of glial-restricted progenitor lineage–associated genes as well as more downregulation of gliogenesis-directing factors. “Thus, the transition from a glial-to-neuronal cell phenotype appears to promote the progression from neuroendocrine cell hyperplasia to tumor development,” the authors wrote. They also found that NETs have higher levels of expression of genes associated with neural stem and progenitor cells, as well as upregulation of factors secreted from neural crest cells that promote neurogenesis and restrict the glial cell fate. Many of these factors are part of the Hedgehog signaling pathway, and menin is known to repress Hedgehog signaling.

Intestinal glial cells have a high degree of plasticity. They can become neuronal progenitor cells and yet they can dedifferentiate to differentiate again into other cell lineages.

The research could eventually lead to identification of unique cells-of-origin for these tumors. The authors say that the diversity of the tumors – which includes variation in location, mutational profile, and response to therapy – may be due to divergent cellular origins in different tissue sites. “Defining the cells-of-origin and the events preceding neoplastic transformation will be critical to informing molecular signaling pathways that can then be targeted therapeutically,” the authors wrote.

The authors disclosed no conflicts of interest.

The diversity of neuroendocrine tumors (NETs) – which includes variation in location, mutational profile, and response to therapy – may be due to divergent cellular origins in different tissue sites, according to a new study.

The pathogenesis of gastroenteropancreatic neoplasms (GEP-NENs) is poorly understood, in part because of a lack of modeling systems, according to Suzann Duan, PhD, and colleagues. They are a heterogeneous group of tumors that are increasingly prevalent in the United States. GEP-NENs arise from endocrine-producing cells and include gastric carcinoids, gastrinomas, and pancreatic NETs.

Despite the general mystery surrounding GEP-NENs, there is at least one clue in the form of the MEN1 gene. Both inherited and sporadic mutations of this gene are associated with GEP-NENs. Menin is a tumor suppressor protein, and previous studies have shown that inactivation of MEN1 leads to loss of that protein and is associated with endocrine tumors in the pancreas, pituitary, and upper GI tract.

In new research published in Cellular and Molecular Gastroenterology and Hepatology, researchers investigated the role of MEN1 in neuroendocrine cell development and traced it to a potential role in the development of NETs.

Patients with MEN1 mutations are at increased risk of gastrinomas, which lead to increased production of the peptide hormone gastrin. Gastrin increases acid production and can lead to hyperplasia in parietal and enterochromaffin cells. These generally develop in Brunner’s glands within the submucosa of the duodenum. At time of diagnosis, more than half of such tumors have developed lymph node metastases.

It remains unclear how loss of MEN1 suppresses gastrin production. Previous research showed that homozygous MEN1 deletion in mice is lethal to embryos, while leaving one copy intact leads to heightened risk of endocrine tumors in the pancreas and pituitary gland, but not in the GI tract. The studies did not reveal the tumor’s origin cell.

The researchers developed a novel mouse model in which MEN1 is conditionally deleted from the GI tract epithelium. This led to hyperplasia of gastrin-producing cells (G cells) in the antrum, as well as hypergastrinemia and development of gastric NETs. Exposure to a proton pump inhibitor accelerated gastric NET development, and the researchers identified expansion of enteric glial cells that expressed gastrin and GFAP. Glial cells that differentiated into endocrine phenotype were associated with a reversible loss of menin. “Taken together, these observations suggest that hyperplastic G cells might emerge from reprogrammed neural crest–derived cells in addition to endoderm-derived enteroendocrine cells,” the authors wrote.

That idea is supported by previous research indicating that multipotent glial cells expressing GFAP or SOX10 may play a developmental role in formation of neuroendocrine cells.

With this in mind, the researchers deleted MEN1 in GFAP-expressing cells to see if it would promote neuroendocrine cell development.

The result was hyperplasia in the gastric antrum and NETs in the pituitary and pancreas. To the researchers’ surprise, NET development was associated with loss of GFAP expression as well as activation of neuronal and neuroendocrine-related genes in the stomach, pancreas, and pituitary. There was universal reduction of GFAP protein expression in pituitary and pancreatic NETs, but GFAP transcript levels stayed steady in the gastric antra despite a reduction in GFAP-reporter expression. This could indicate that the menin protein interacts with GFAP. If so, eliminating menin in GFAP-positive cells could change the localization of GFAP, which may in turn lead to changes in glial cell identity.

When the researchers compared transcriptomes of hyperplastic antral tissues to well-differentiated NETs, they found that NETs exhibited a greater loss of glial-restricted progenitor lineage–associated genes as well as more downregulation of gliogenesis-directing factors. “Thus, the transition from a glial-to-neuronal cell phenotype appears to promote the progression from neuroendocrine cell hyperplasia to tumor development,” the authors wrote. They also found that NETs have higher levels of expression of genes associated with neural stem and progenitor cells, as well as upregulation of factors secreted from neural crest cells that promote neurogenesis and restrict the glial cell fate. Many of these factors are part of the Hedgehog signaling pathway, and menin is known to repress Hedgehog signaling.

Intestinal glial cells have a high degree of plasticity. They can become neuronal progenitor cells and yet they can dedifferentiate to differentiate again into other cell lineages.

The research could eventually lead to identification of unique cells-of-origin for these tumors. The authors say that the diversity of the tumors – which includes variation in location, mutational profile, and response to therapy – may be due to divergent cellular origins in different tissue sites. “Defining the cells-of-origin and the events preceding neoplastic transformation will be critical to informing molecular signaling pathways that can then be targeted therapeutically,” the authors wrote.

The authors disclosed no conflicts of interest.

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM CELLULAR AND MOLECULAR GASTROENTEROLOGY AND HEPATOLOGY

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Experts refine nomenclature for eosinophilic GI diseases

Working toward personalized care
Article Type
Changed
Wed, 10/12/2022 - 20:28

A new international consensus paper is recommending that eosinophilic GI diseases (EGIDs) should be named according to more specific criteria. The paper seeks to update nomenclature to improve research and bolster clinical clarity.

The involved part of the GI tract should be specifically named, and the abbreviation “Eo” should be used. Furthermore, the umbrella term should be EGID instead of the currently used “eosinophilic gastroenteritis,” according to the statement published in Clinical Gastroenterology and Hepatology. The statement included 91 authors from five continents who filled out two rounds of surveys. In total, 93% completed the first and 90% completed the second. The paper produced 22 statements, with a consensus reached on all but 2.

EGIDs are chronic, immune-driven disorders that produce gastrointestinal symptoms and are characterized by eosinophil-dominant inflammation in specific GI regions. Although eosinophilic esophagitis (EoE) is the most well known of these conditions, other EGIDs have become more commonly recognized in recent years and are the subject of intense study. Other affected areas include the stomach, small bowel, and colon, where it can occur individually or in combination.

Efforts are underway to develop guidelines for diagnosis and treatment of EGIDs, but there was initial confusion surrounding the term eosinophilic gastroenteritis since its definition varied significantly in different clinical and research settings, according to the authors. That term varyingly referred to stomach alone, small bowel alone, stomach and small bowel, or any region of the GI tract.

“This nonstandardized use of nomenclature highlighted a need for a common language for non-EoE eosinophilic GI disease names, not only for clinical practice, but also for the consistent data collection required for research to continue to advance the field,” co–first authors Evan S. Dellon, MD, MPH, AGAF, from the University of North Carolina at Chapel Hill, and Nirmala Gonsalves, MD, from Northwestern University, Chicago, and colleagues wrote. “This step, while seemingly rudimentary, was essential to inform the guideline efforts that are now underway.”

After responses to the surveys were analyzed, respondents participated in one of two scheduled meetings held on a video conferencing platform in May 2021. Feedback from these meetings was then used to create a second round of 29 statements which were again distributed, and participants were asked to either agree or disagree with each statement. Agreement was set a priori at 70%. In all, 38% of the participants were women, and 91% worked in academic or university settings.

In routine clinical practice, conditions with eosinophil-dominant inflammation in the absence of secondary causes outside of the esophagus can collectively be referred to clinically as non-EoE EGID. Stomach involvement should be called eosinophilic gastritis (EoG), small bowel involvement eosinophilic enteritis (EoN), and colon involvement eosinophilic colitis (EoC).

For research use, and clinical use if desired, the authors called for greater granularity in description of the conditions, with each location named. For example, if the stomach and small bowel are both involved, the condition should be termed eosinophilic gastritis and enteritis. The authors could not reach a consensus for terminology when the esophagus is also involved, leading to the recommendation that it can be included using the phrase “with esophageal involvement” or by using EoE, although they note that this could be confusing since EoE is the current term for eosinophilia isolated in the esophagus.

The authors came to universal agreement in many areas, but there were exceptions that mostly centered on how to name conditions that affect multiple areas of the GI tract. It remains uncertain whether eosinophilia in different regions is caused by the same pathogenesis. Some experts felt that a “primary” location of EGID should be identified using predominant symptoms, endoscopic features, and complications. However, the authors anticipate that this nomenclature will change over time.

The authors noted that the clinical manifestation of the disease should remain the driving factor behind classification. Testing should be driven by clinically relevant questions, and overtesting should be avoided. More details on this are likely to be forthcoming in future guidelines.

The consensus statement is limited by the fact that most participants were from academic settings. These recommendations do not apply to eosinophilic disorders of the gallbladder, liver, or pancreas. Application of the recommendations to the small bowel may be too general or specific, but are meant primarily as a starting point for further refinement.

These limitations should help to drive further research. For example, molecular and pathogenic data could help distinguish EoE from “esophageal involvement” by determining if pathogenic mechanisms are the same or different, which would in turn lead to lumping the conditions into a single term or keeping them separate.

“The iterative and collaborative process led to agreement on nearly all aspects of the proposed nomenclature framework, and has identified future research directions. It is expected that as more data are collected, the nomenclature will again be updated to reflect best practices and the underlying pathogenesis of these disorders,” the authors concluded.

The authors disclosed relationships with various industry companies, including AstraZeneca, Celgene, GlaxoSmithKline, Regeneron, Sanofi, and Takeda.

Body

Eosinophilic conditions of the gastrointestinal tract have risen in incidence, leading to significant patient symptoms and morbidity, but thankfully there have been tremendous innovations in identification, management, treatment, and drug development. In this excellent article, an international consensus was created to reflect this rapidly changing understanding of the phenotypes with updated diagnostic nomenclature.

Dr. Rishi D. Naik
Eosinophilic esophagitis (by far the common eosinophilic GI condition) remains unchanged in its nomenclature, but the prior use of eosinophilic gastroenteritis should no longer be used. Instead, the organ involved – for example, the stomach, small bowel, or colon – should be identified, as eosinophilic gastritis, eosinophilic enteritis, or eosinophilic colitis, respectively. This does reflect clinical and patient practice on where biopsies can be routinely obtained from when patients have symptoms. Debates are still ongoing on how to define overlapping sites (for example, simultaneous esophagus and stomach involvement) or if duodenal, jejunal, and ileum eosinophilic conditions should be separated. This new framework will allow us to begin settling these debates based on patient outcomes.

Redefinition of these conditions will help in many aspects. First, advances in therapy targeted as eosinophilic trafficking have been approved with many biologic therapies in the pipeline, and understanding their treatment effects and targets will help our patients. Second, improved nomenclature will help better understand the genetic, phenotypic, and therapeutic options for these conditions providing our patients with personalized care. As the understanding of eosinophilic conditions expands with the growth of genetic associations and drug therapies, we are matching our inflammatory bowel disease colleagues in their successes to provide our patients with personalized care.

Rishi D. Naik, MD, MSCI, is an assistant professor, department of medicine, section of gastroenterology & hepatology, Esophageal Center at Vanderbilt University Medical Center, Nashville, Tenn.

Publications
Topics
Sections
Body

Eosinophilic conditions of the gastrointestinal tract have risen in incidence, leading to significant patient symptoms and morbidity, but thankfully there have been tremendous innovations in identification, management, treatment, and drug development. In this excellent article, an international consensus was created to reflect this rapidly changing understanding of the phenotypes with updated diagnostic nomenclature.

Dr. Rishi D. Naik
Eosinophilic esophagitis (by far the common eosinophilic GI condition) remains unchanged in its nomenclature, but the prior use of eosinophilic gastroenteritis should no longer be used. Instead, the organ involved – for example, the stomach, small bowel, or colon – should be identified, as eosinophilic gastritis, eosinophilic enteritis, or eosinophilic colitis, respectively. This does reflect clinical and patient practice on where biopsies can be routinely obtained from when patients have symptoms. Debates are still ongoing on how to define overlapping sites (for example, simultaneous esophagus and stomach involvement) or if duodenal, jejunal, and ileum eosinophilic conditions should be separated. This new framework will allow us to begin settling these debates based on patient outcomes.

Redefinition of these conditions will help in many aspects. First, advances in therapy targeted as eosinophilic trafficking have been approved with many biologic therapies in the pipeline, and understanding their treatment effects and targets will help our patients. Second, improved nomenclature will help better understand the genetic, phenotypic, and therapeutic options for these conditions providing our patients with personalized care. As the understanding of eosinophilic conditions expands with the growth of genetic associations and drug therapies, we are matching our inflammatory bowel disease colleagues in their successes to provide our patients with personalized care.

Rishi D. Naik, MD, MSCI, is an assistant professor, department of medicine, section of gastroenterology & hepatology, Esophageal Center at Vanderbilt University Medical Center, Nashville, Tenn.

Body

Eosinophilic conditions of the gastrointestinal tract have risen in incidence, leading to significant patient symptoms and morbidity, but thankfully there have been tremendous innovations in identification, management, treatment, and drug development. In this excellent article, an international consensus was created to reflect this rapidly changing understanding of the phenotypes with updated diagnostic nomenclature.

Dr. Rishi D. Naik
Eosinophilic esophagitis (by far the common eosinophilic GI condition) remains unchanged in its nomenclature, but the prior use of eosinophilic gastroenteritis should no longer be used. Instead, the organ involved – for example, the stomach, small bowel, or colon – should be identified, as eosinophilic gastritis, eosinophilic enteritis, or eosinophilic colitis, respectively. This does reflect clinical and patient practice on where biopsies can be routinely obtained from when patients have symptoms. Debates are still ongoing on how to define overlapping sites (for example, simultaneous esophagus and stomach involvement) or if duodenal, jejunal, and ileum eosinophilic conditions should be separated. This new framework will allow us to begin settling these debates based on patient outcomes.

Redefinition of these conditions will help in many aspects. First, advances in therapy targeted as eosinophilic trafficking have been approved with many biologic therapies in the pipeline, and understanding their treatment effects and targets will help our patients. Second, improved nomenclature will help better understand the genetic, phenotypic, and therapeutic options for these conditions providing our patients with personalized care. As the understanding of eosinophilic conditions expands with the growth of genetic associations and drug therapies, we are matching our inflammatory bowel disease colleagues in their successes to provide our patients with personalized care.

Rishi D. Naik, MD, MSCI, is an assistant professor, department of medicine, section of gastroenterology & hepatology, Esophageal Center at Vanderbilt University Medical Center, Nashville, Tenn.

Title
Working toward personalized care
Working toward personalized care

A new international consensus paper is recommending that eosinophilic GI diseases (EGIDs) should be named according to more specific criteria. The paper seeks to update nomenclature to improve research and bolster clinical clarity.

The involved part of the GI tract should be specifically named, and the abbreviation “Eo” should be used. Furthermore, the umbrella term should be EGID instead of the currently used “eosinophilic gastroenteritis,” according to the statement published in Clinical Gastroenterology and Hepatology. The statement included 91 authors from five continents who filled out two rounds of surveys. In total, 93% completed the first and 90% completed the second. The paper produced 22 statements, with a consensus reached on all but 2.

EGIDs are chronic, immune-driven disorders that produce gastrointestinal symptoms and are characterized by eosinophil-dominant inflammation in specific GI regions. Although eosinophilic esophagitis (EoE) is the most well known of these conditions, other EGIDs have become more commonly recognized in recent years and are the subject of intense study. Other affected areas include the stomach, small bowel, and colon, where it can occur individually or in combination.

Efforts are underway to develop guidelines for diagnosis and treatment of EGIDs, but there was initial confusion surrounding the term eosinophilic gastroenteritis since its definition varied significantly in different clinical and research settings, according to the authors. That term varyingly referred to stomach alone, small bowel alone, stomach and small bowel, or any region of the GI tract.

“This nonstandardized use of nomenclature highlighted a need for a common language for non-EoE eosinophilic GI disease names, not only for clinical practice, but also for the consistent data collection required for research to continue to advance the field,” co–first authors Evan S. Dellon, MD, MPH, AGAF, from the University of North Carolina at Chapel Hill, and Nirmala Gonsalves, MD, from Northwestern University, Chicago, and colleagues wrote. “This step, while seemingly rudimentary, was essential to inform the guideline efforts that are now underway.”

After responses to the surveys were analyzed, respondents participated in one of two scheduled meetings held on a video conferencing platform in May 2021. Feedback from these meetings was then used to create a second round of 29 statements which were again distributed, and participants were asked to either agree or disagree with each statement. Agreement was set a priori at 70%. In all, 38% of the participants were women, and 91% worked in academic or university settings.

In routine clinical practice, conditions with eosinophil-dominant inflammation in the absence of secondary causes outside of the esophagus can collectively be referred to clinically as non-EoE EGID. Stomach involvement should be called eosinophilic gastritis (EoG), small bowel involvement eosinophilic enteritis (EoN), and colon involvement eosinophilic colitis (EoC).

For research use, and clinical use if desired, the authors called for greater granularity in description of the conditions, with each location named. For example, if the stomach and small bowel are both involved, the condition should be termed eosinophilic gastritis and enteritis. The authors could not reach a consensus for terminology when the esophagus is also involved, leading to the recommendation that it can be included using the phrase “with esophageal involvement” or by using EoE, although they note that this could be confusing since EoE is the current term for eosinophilia isolated in the esophagus.

The authors came to universal agreement in many areas, but there were exceptions that mostly centered on how to name conditions that affect multiple areas of the GI tract. It remains uncertain whether eosinophilia in different regions is caused by the same pathogenesis. Some experts felt that a “primary” location of EGID should be identified using predominant symptoms, endoscopic features, and complications. However, the authors anticipate that this nomenclature will change over time.

The authors noted that the clinical manifestation of the disease should remain the driving factor behind classification. Testing should be driven by clinically relevant questions, and overtesting should be avoided. More details on this are likely to be forthcoming in future guidelines.

The consensus statement is limited by the fact that most participants were from academic settings. These recommendations do not apply to eosinophilic disorders of the gallbladder, liver, or pancreas. Application of the recommendations to the small bowel may be too general or specific, but are meant primarily as a starting point for further refinement.

These limitations should help to drive further research. For example, molecular and pathogenic data could help distinguish EoE from “esophageal involvement” by determining if pathogenic mechanisms are the same or different, which would in turn lead to lumping the conditions into a single term or keeping them separate.

“The iterative and collaborative process led to agreement on nearly all aspects of the proposed nomenclature framework, and has identified future research directions. It is expected that as more data are collected, the nomenclature will again be updated to reflect best practices and the underlying pathogenesis of these disorders,” the authors concluded.

The authors disclosed relationships with various industry companies, including AstraZeneca, Celgene, GlaxoSmithKline, Regeneron, Sanofi, and Takeda.

A new international consensus paper is recommending that eosinophilic GI diseases (EGIDs) should be named according to more specific criteria. The paper seeks to update nomenclature to improve research and bolster clinical clarity.

The involved part of the GI tract should be specifically named, and the abbreviation “Eo” should be used. Furthermore, the umbrella term should be EGID instead of the currently used “eosinophilic gastroenteritis,” according to the statement published in Clinical Gastroenterology and Hepatology. The statement included 91 authors from five continents who filled out two rounds of surveys. In total, 93% completed the first and 90% completed the second. The paper produced 22 statements, with a consensus reached on all but 2.

EGIDs are chronic, immune-driven disorders that produce gastrointestinal symptoms and are characterized by eosinophil-dominant inflammation in specific GI regions. Although eosinophilic esophagitis (EoE) is the most well known of these conditions, other EGIDs have become more commonly recognized in recent years and are the subject of intense study. Other affected areas include the stomach, small bowel, and colon, where it can occur individually or in combination.

Efforts are underway to develop guidelines for diagnosis and treatment of EGIDs, but there was initial confusion surrounding the term eosinophilic gastroenteritis since its definition varied significantly in different clinical and research settings, according to the authors. That term varyingly referred to stomach alone, small bowel alone, stomach and small bowel, or any region of the GI tract.

“This nonstandardized use of nomenclature highlighted a need for a common language for non-EoE eosinophilic GI disease names, not only for clinical practice, but also for the consistent data collection required for research to continue to advance the field,” co–first authors Evan S. Dellon, MD, MPH, AGAF, from the University of North Carolina at Chapel Hill, and Nirmala Gonsalves, MD, from Northwestern University, Chicago, and colleagues wrote. “This step, while seemingly rudimentary, was essential to inform the guideline efforts that are now underway.”

After responses to the surveys were analyzed, respondents participated in one of two scheduled meetings held on a video conferencing platform in May 2021. Feedback from these meetings was then used to create a second round of 29 statements which were again distributed, and participants were asked to either agree or disagree with each statement. Agreement was set a priori at 70%. In all, 38% of the participants were women, and 91% worked in academic or university settings.

In routine clinical practice, conditions with eosinophil-dominant inflammation in the absence of secondary causes outside of the esophagus can collectively be referred to clinically as non-EoE EGID. Stomach involvement should be called eosinophilic gastritis (EoG), small bowel involvement eosinophilic enteritis (EoN), and colon involvement eosinophilic colitis (EoC).

For research use, and clinical use if desired, the authors called for greater granularity in description of the conditions, with each location named. For example, if the stomach and small bowel are both involved, the condition should be termed eosinophilic gastritis and enteritis. The authors could not reach a consensus for terminology when the esophagus is also involved, leading to the recommendation that it can be included using the phrase “with esophageal involvement” or by using EoE, although they note that this could be confusing since EoE is the current term for eosinophilia isolated in the esophagus.

The authors came to universal agreement in many areas, but there were exceptions that mostly centered on how to name conditions that affect multiple areas of the GI tract. It remains uncertain whether eosinophilia in different regions is caused by the same pathogenesis. Some experts felt that a “primary” location of EGID should be identified using predominant symptoms, endoscopic features, and complications. However, the authors anticipate that this nomenclature will change over time.

The authors noted that the clinical manifestation of the disease should remain the driving factor behind classification. Testing should be driven by clinically relevant questions, and overtesting should be avoided. More details on this are likely to be forthcoming in future guidelines.

The consensus statement is limited by the fact that most participants were from academic settings. These recommendations do not apply to eosinophilic disorders of the gallbladder, liver, or pancreas. Application of the recommendations to the small bowel may be too general or specific, but are meant primarily as a starting point for further refinement.

These limitations should help to drive further research. For example, molecular and pathogenic data could help distinguish EoE from “esophageal involvement” by determining if pathogenic mechanisms are the same or different, which would in turn lead to lumping the conditions into a single term or keeping them separate.

“The iterative and collaborative process led to agreement on nearly all aspects of the proposed nomenclature framework, and has identified future research directions. It is expected that as more data are collected, the nomenclature will again be updated to reflect best practices and the underlying pathogenesis of these disorders,” the authors concluded.

The authors disclosed relationships with various industry companies, including AstraZeneca, Celgene, GlaxoSmithKline, Regeneron, Sanofi, and Takeda.

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM CLINICAL GASTROENTEROLOGY AND HEPATOLOGY

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Experts refine nomenclature for eosinophilic GI disorders

Working toward personalized care
Article Type
Changed
Mon, 10/10/2022 - 10:55

A new international consensus paper is recommending that eosinophilic GI diseases (EGIDs) should be named according to more specific criteria. The paper seeks to update nomenclature to improve research and bolster clinical clarity.

The involved part of the GI tract should be specifically named, and the abbreviation “Eo” should be used. Furthermore, the umbrella term should be EGID instead of the currently used “eosinophilic gastroenteritis,” according to the statement published in Clinical Gastroenterology and Hepatology. The statement included 91 authors from five continents who filled out two rounds of surveys. In total, 93% completed the first and 90% completed the second. The paper produced 22 statements, with a consensus reached on all but 2.

EGIDs are chronic, immune-driven disorders that produce gastrointestinal symptoms and are characterized by eosinophil-dominant inflammation in specific GI regions. Although eosinophilic esophagitis (EoE) is the most well known of these conditions, other EGIDs have become more commonly recognized in recent years and are the subject of intense study. Other affected areas include the stomach, small bowel, and colon, where it can occur individually or in combination.

Efforts are underway to develop guidelines for diagnosis and treatment of EGIDs, but there was initial confusion surrounding the term eosinophilic gastroenteritis since its definition varied significantly in different clinical and research settings, according to the authors. That term varyingly referred to stomach alone, small bowel alone, stomach and small bowel, or any region of the GI tract.

“This nonstandardized use of nomenclature highlighted a need for a common language for non-EoE eosinophilic GI disease names, not only for clinical practice, but also for the consistent data collection required for research to continue to advance the field,” co–first authors Evan S. Dellon, MD, MPH, AGAF, from the University of North Carolina at Chapel Hill, and Nirmala Gonsalves, MD, from Northwestern University, Chicago, and colleagues wrote. “This step, while seemingly rudimentary, was essential to inform the guideline efforts that are now underway.”

After responses to the surveys were analyzed, respondents participated in one of two scheduled meetings held on a video conferencing platform in May 2021. Feedback from these meetings was then used to create a second round of 29 statements which were again distributed, and participants were asked to either agree or disagree with each statement. Agreement was set a priori at 70%. In all, 38% of the participants were women, and 91% worked in academic or university settings.

In routine clinical practice, conditions with eosinophil-dominant inflammation in the absence of secondary causes outside of the esophagus can collectively be referred to clinically as non-EoE EGID. Stomach involvement should be called eosinophilic gastritis (EoG), small bowel involvement eosinophilic enteritis (EoN), and colon involvement eosinophilic colitis (EoC).

For research use, and clinical use if desired, the authors called for greater granularity in description of the conditions, with each location named. For example, if the stomach and small bowel are both involved, the condition should be termed eosinophilic gastritis and enteritis. The authors could not reach a consensus for terminology when the esophagus is also involved, leading to the recommendation that it can be included using the phrase “with esophageal involvement” or by using EoE, although they note that this could be confusing since EoE is the current term for eosinophilia isolated in the esophagus.

The authors came to universal agreement in many areas, but there were exceptions that mostly centered on how to name conditions that affect multiple areas of the GI tract. It remains uncertain whether eosinophilia in different regions is caused by the same pathogenesis. Some experts felt that a “primary” location of EGID should be identified using predominant symptoms, endoscopic features, and complications. However, the authors anticipate that this nomenclature will change over time.

The authors noted that the clinical manifestation of the disease should remain the driving factor behind classification. Testing should be driven by clinically relevant questions and overtesting should be avoided. More details on this are likely to be forthcoming in future guidelines.

The consensus statement is limited by the fact that most participants were from academic settings. These recommendations do not apply to eosinophilic disorders of gallbladder, liver, or pancreas. Application of the recommendations to the small bowel may be too general or specific, but are meant primarily as a starting point for further refinement.

These limitations should help to drive further research. For example, molecular and pathogenic data could help distinguish EoE from “esophageal involvement” by determining if pathogenic mechanisms are the same or different, which would in turn lead to lumping the conditions into a single term or keeping them separate.

“The iterative and collaborative process led to agreement on nearly all aspects of the proposed nomenclature framework, and has identified future research directions. It is expected that as more data are collected, the nomenclature will again be updated to reflect best practices and the underlying pathogenesis of these disorders,” the authors concluded.

The authors disclosed relationships with various industry companies, include AstraZeneca, Celgene, GlaxoSmithKline, Regeneron, Sanofi, and Takeda.

Body

Eosinophilic conditions of the gastrointestinal tract have risen in incidence leading to significant patient symptom and morbidity, but thankfully there have been tremendous innovations in identification, management, treatment, and drug development. In this excellent article, an international consensus was created to reflect these rapidly changing understanding of the phenotypes with updated diagnostic nomenclature.

Dr. Rishi D. Naik
Eosinophilic esophagitis (by far the common eosinophilic GI condition) remains unchanged in its nomenclature, but the prior use of eosinophilic gastroenteritis should no longer be used. Instead, the organ involved – for example, stomach, small bowel, or colon – should be identified, as eosinophilic gastritis, eosinophilic enteritis, or eosinophilic colitis, respectively. This does reflect clinical and patient practice on where biopsies can be routine obtained from when patients have symptoms. Debates are still ongoing on how to define overlapping sites (for example, simultaneous esophagus and stomach involvement) or if duodenal, jejunal, and ileum eosinophilic conditions should be separated. This new framework will allow us to begin settling these debates based on patient outcomes.

Redefinition of these conditions will help in many aspects. First, advances in therapy targeted as eosinophilic trafficking have been approved with many biologic therapies in the pipeline and understanding their treatment effects and targets will help our patients. Second, improved nomenclature will help better understand the genetic, phenotypic, and therapeutic options for these conditions providing our patients with personalized care. As the understanding of eosinophilic conditions expands with the growth of genetic associations and drug therapies, we are matching our inflammatory bowel disease colleagues in their successes to provide our patients with personalized care.

Rishi D. Naik, MD, MSCI, is an assistant professor, department of medicine, section of gastroenterology & hepatology, Esophageal Center at Vanderbilt University Medical Center, Nashville, Tenn.

Publications
Topics
Sections
Body

Eosinophilic conditions of the gastrointestinal tract have risen in incidence leading to significant patient symptom and morbidity, but thankfully there have been tremendous innovations in identification, management, treatment, and drug development. In this excellent article, an international consensus was created to reflect these rapidly changing understanding of the phenotypes with updated diagnostic nomenclature.

Dr. Rishi D. Naik
Eosinophilic esophagitis (by far the common eosinophilic GI condition) remains unchanged in its nomenclature, but the prior use of eosinophilic gastroenteritis should no longer be used. Instead, the organ involved – for example, stomach, small bowel, or colon – should be identified, as eosinophilic gastritis, eosinophilic enteritis, or eosinophilic colitis, respectively. This does reflect clinical and patient practice on where biopsies can be routine obtained from when patients have symptoms. Debates are still ongoing on how to define overlapping sites (for example, simultaneous esophagus and stomach involvement) or if duodenal, jejunal, and ileum eosinophilic conditions should be separated. This new framework will allow us to begin settling these debates based on patient outcomes.

Redefinition of these conditions will help in many aspects. First, advances in therapy targeted as eosinophilic trafficking have been approved with many biologic therapies in the pipeline and understanding their treatment effects and targets will help our patients. Second, improved nomenclature will help better understand the genetic, phenotypic, and therapeutic options for these conditions providing our patients with personalized care. As the understanding of eosinophilic conditions expands with the growth of genetic associations and drug therapies, we are matching our inflammatory bowel disease colleagues in their successes to provide our patients with personalized care.

Rishi D. Naik, MD, MSCI, is an assistant professor, department of medicine, section of gastroenterology & hepatology, Esophageal Center at Vanderbilt University Medical Center, Nashville, Tenn.

Body

Eosinophilic conditions of the gastrointestinal tract have risen in incidence leading to significant patient symptom and morbidity, but thankfully there have been tremendous innovations in identification, management, treatment, and drug development. In this excellent article, an international consensus was created to reflect these rapidly changing understanding of the phenotypes with updated diagnostic nomenclature.

Dr. Rishi D. Naik
Eosinophilic esophagitis (by far the common eosinophilic GI condition) remains unchanged in its nomenclature, but the prior use of eosinophilic gastroenteritis should no longer be used. Instead, the organ involved – for example, stomach, small bowel, or colon – should be identified, as eosinophilic gastritis, eosinophilic enteritis, or eosinophilic colitis, respectively. This does reflect clinical and patient practice on where biopsies can be routine obtained from when patients have symptoms. Debates are still ongoing on how to define overlapping sites (for example, simultaneous esophagus and stomach involvement) or if duodenal, jejunal, and ileum eosinophilic conditions should be separated. This new framework will allow us to begin settling these debates based on patient outcomes.

Redefinition of these conditions will help in many aspects. First, advances in therapy targeted as eosinophilic trafficking have been approved with many biologic therapies in the pipeline and understanding their treatment effects and targets will help our patients. Second, improved nomenclature will help better understand the genetic, phenotypic, and therapeutic options for these conditions providing our patients with personalized care. As the understanding of eosinophilic conditions expands with the growth of genetic associations and drug therapies, we are matching our inflammatory bowel disease colleagues in their successes to provide our patients with personalized care.

Rishi D. Naik, MD, MSCI, is an assistant professor, department of medicine, section of gastroenterology & hepatology, Esophageal Center at Vanderbilt University Medical Center, Nashville, Tenn.

Title
Working toward personalized care
Working toward personalized care

A new international consensus paper is recommending that eosinophilic GI diseases (EGIDs) should be named according to more specific criteria. The paper seeks to update nomenclature to improve research and bolster clinical clarity.

The involved part of the GI tract should be specifically named, and the abbreviation “Eo” should be used. Furthermore, the umbrella term should be EGID instead of the currently used “eosinophilic gastroenteritis,” according to the statement published in Clinical Gastroenterology and Hepatology. The statement included 91 authors from five continents who filled out two rounds of surveys. In total, 93% completed the first and 90% completed the second. The paper produced 22 statements, with a consensus reached on all but 2.

EGIDs are chronic, immune-driven disorders that produce gastrointestinal symptoms and are characterized by eosinophil-dominant inflammation in specific GI regions. Although eosinophilic esophagitis (EoE) is the most well known of these conditions, other EGIDs have become more commonly recognized in recent years and are the subject of intense study. Other affected areas include the stomach, small bowel, and colon, where it can occur individually or in combination.

Efforts are underway to develop guidelines for diagnosis and treatment of EGIDs, but there was initial confusion surrounding the term eosinophilic gastroenteritis since its definition varied significantly in different clinical and research settings, according to the authors. That term varyingly referred to stomach alone, small bowel alone, stomach and small bowel, or any region of the GI tract.

“This nonstandardized use of nomenclature highlighted a need for a common language for non-EoE eosinophilic GI disease names, not only for clinical practice, but also for the consistent data collection required for research to continue to advance the field,” co–first authors Evan S. Dellon, MD, MPH, AGAF, from the University of North Carolina at Chapel Hill, and Nirmala Gonsalves, MD, from Northwestern University, Chicago, and colleagues wrote. “This step, while seemingly rudimentary, was essential to inform the guideline efforts that are now underway.”

After responses to the surveys were analyzed, respondents participated in one of two scheduled meetings held on a video conferencing platform in May 2021. Feedback from these meetings was then used to create a second round of 29 statements which were again distributed, and participants were asked to either agree or disagree with each statement. Agreement was set a priori at 70%. In all, 38% of the participants were women, and 91% worked in academic or university settings.

In routine clinical practice, conditions with eosinophil-dominant inflammation in the absence of secondary causes outside of the esophagus can collectively be referred to clinically as non-EoE EGID. Stomach involvement should be called eosinophilic gastritis (EoG), small bowel involvement eosinophilic enteritis (EoN), and colon involvement eosinophilic colitis (EoC).

For research use, and clinical use if desired, the authors called for greater granularity in description of the conditions, with each location named. For example, if the stomach and small bowel are both involved, the condition should be termed eosinophilic gastritis and enteritis. The authors could not reach a consensus for terminology when the esophagus is also involved, leading to the recommendation that it can be included using the phrase “with esophageal involvement” or by using EoE, although they note that this could be confusing since EoE is the current term for eosinophilia isolated in the esophagus.

The authors came to universal agreement in many areas, but there were exceptions that mostly centered on how to name conditions that affect multiple areas of the GI tract. It remains uncertain whether eosinophilia in different regions is caused by the same pathogenesis. Some experts felt that a “primary” location of EGID should be identified using predominant symptoms, endoscopic features, and complications. However, the authors anticipate that this nomenclature will change over time.

The authors noted that the clinical manifestation of the disease should remain the driving factor behind classification. Testing should be driven by clinically relevant questions and overtesting should be avoided. More details on this are likely to be forthcoming in future guidelines.

The consensus statement is limited by the fact that most participants were from academic settings. These recommendations do not apply to eosinophilic disorders of gallbladder, liver, or pancreas. Application of the recommendations to the small bowel may be too general or specific, but are meant primarily as a starting point for further refinement.

These limitations should help to drive further research. For example, molecular and pathogenic data could help distinguish EoE from “esophageal involvement” by determining if pathogenic mechanisms are the same or different, which would in turn lead to lumping the conditions into a single term or keeping them separate.

“The iterative and collaborative process led to agreement on nearly all aspects of the proposed nomenclature framework, and has identified future research directions. It is expected that as more data are collected, the nomenclature will again be updated to reflect best practices and the underlying pathogenesis of these disorders,” the authors concluded.

The authors disclosed relationships with various industry companies, include AstraZeneca, Celgene, GlaxoSmithKline, Regeneron, Sanofi, and Takeda.

A new international consensus paper is recommending that eosinophilic GI diseases (EGIDs) should be named according to more specific criteria. The paper seeks to update nomenclature to improve research and bolster clinical clarity.

The involved part of the GI tract should be specifically named, and the abbreviation “Eo” should be used. Furthermore, the umbrella term should be EGID instead of the currently used “eosinophilic gastroenteritis,” according to the statement published in Clinical Gastroenterology and Hepatology. The statement included 91 authors from five continents who filled out two rounds of surveys. In total, 93% completed the first and 90% completed the second. The paper produced 22 statements, with a consensus reached on all but 2.

EGIDs are chronic, immune-driven disorders that produce gastrointestinal symptoms and are characterized by eosinophil-dominant inflammation in specific GI regions. Although eosinophilic esophagitis (EoE) is the most well known of these conditions, other EGIDs have become more commonly recognized in recent years and are the subject of intense study. Other affected areas include the stomach, small bowel, and colon, where it can occur individually or in combination.

Efforts are underway to develop guidelines for diagnosis and treatment of EGIDs, but there was initial confusion surrounding the term eosinophilic gastroenteritis since its definition varied significantly in different clinical and research settings, according to the authors. That term varyingly referred to stomach alone, small bowel alone, stomach and small bowel, or any region of the GI tract.

“This nonstandardized use of nomenclature highlighted a need for a common language for non-EoE eosinophilic GI disease names, not only for clinical practice, but also for the consistent data collection required for research to continue to advance the field,” co–first authors Evan S. Dellon, MD, MPH, AGAF, from the University of North Carolina at Chapel Hill, and Nirmala Gonsalves, MD, from Northwestern University, Chicago, and colleagues wrote. “This step, while seemingly rudimentary, was essential to inform the guideline efforts that are now underway.”

After responses to the surveys were analyzed, respondents participated in one of two scheduled meetings held on a video conferencing platform in May 2021. Feedback from these meetings was then used to create a second round of 29 statements which were again distributed, and participants were asked to either agree or disagree with each statement. Agreement was set a priori at 70%. In all, 38% of the participants were women, and 91% worked in academic or university settings.

In routine clinical practice, conditions with eosinophil-dominant inflammation in the absence of secondary causes outside of the esophagus can collectively be referred to clinically as non-EoE EGID. Stomach involvement should be called eosinophilic gastritis (EoG), small bowel involvement eosinophilic enteritis (EoN), and colon involvement eosinophilic colitis (EoC).

For research use, and clinical use if desired, the authors called for greater granularity in description of the conditions, with each location named. For example, if the stomach and small bowel are both involved, the condition should be termed eosinophilic gastritis and enteritis. The authors could not reach a consensus for terminology when the esophagus is also involved, leading to the recommendation that it can be included using the phrase “with esophageal involvement” or by using EoE, although they note that this could be confusing since EoE is the current term for eosinophilia isolated in the esophagus.

The authors came to universal agreement in many areas, but there were exceptions that mostly centered on how to name conditions that affect multiple areas of the GI tract. It remains uncertain whether eosinophilia in different regions is caused by the same pathogenesis. Some experts felt that a “primary” location of EGID should be identified using predominant symptoms, endoscopic features, and complications. However, the authors anticipate that this nomenclature will change over time.

The authors noted that the clinical manifestation of the disease should remain the driving factor behind classification. Testing should be driven by clinically relevant questions and overtesting should be avoided. More details on this are likely to be forthcoming in future guidelines.

The consensus statement is limited by the fact that most participants were from academic settings. These recommendations do not apply to eosinophilic disorders of gallbladder, liver, or pancreas. Application of the recommendations to the small bowel may be too general or specific, but are meant primarily as a starting point for further refinement.

These limitations should help to drive further research. For example, molecular and pathogenic data could help distinguish EoE from “esophageal involvement” by determining if pathogenic mechanisms are the same or different, which would in turn lead to lumping the conditions into a single term or keeping them separate.

“The iterative and collaborative process led to agreement on nearly all aspects of the proposed nomenclature framework, and has identified future research directions. It is expected that as more data are collected, the nomenclature will again be updated to reflect best practices and the underlying pathogenesis of these disorders,” the authors concluded.

The authors disclosed relationships with various industry companies, include AstraZeneca, Celgene, GlaxoSmithKline, Regeneron, Sanofi, and Takeda.

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM CLINICAL GASTROENTEROLOGY AND HEPATOLOGY

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

AGA Clinical Practice Update: Expert review of management of subepithelial lesions

Article Type
Changed
Tue, 10/11/2022 - 16:30

 

The proper management of subepithelial lesions (SELs) depends on the size, histopathology, malignant potential, and presence of symptoms, according to a new American Gastroenterological Association clinical practice update published in Clinical Gastroenterology and Hepatology.

“SELs are found in 1 in every 300 endoscopies, and two-thirds of these lesions are located in the stomach,” explained Kaveh Sharzehi, MD, an associate professor of medicine in the division of gastroenterology and hepatology at Oregon Health & Science University, Portland, and colleagues. “They represent a heterogeneous group of lesions including nonneoplastic lesions such as ectopic pancreatic tissue and neoplastic lesions. The neoplastic SELs can vary from lesions with no malignant potential such as lipomas to those with malignant potential such as gastrointestinal stromal tumors (GISTs). The majority of SELs are small and found incidentally.”

Dr. Kaveh Sharzehi

The authors developed 10 clinical practice advice statements on the diagnosis and management of subepithelial lesions based on a review of the published literature and expert opinion.

First, standard mucosal biopsies often don’t reach deep enough to obtain a pathologic diagnosis for SELs because the lesions have normal overlying mucosa. Forceps bite-on-bite/deep-well biopsies or tunnel biopsies may help to establish a pathologic diagnosis.

Used as an adjunct to standard endoscopy, endoscopic ultrasound (EUS) has become the primary method for determining diagnostic and prognostic characteristics of SELs – such as the layer of origin, echogenicity, and presence of blood vessels within the lesion. It can also help with tissue acquisition.

For SELs arising from the submucosa, EUS-guided fine-needle aspiration and fine-needle biopsy have evolved as widely used methods for obtaining tissue. For SELs arising from muscularis propria, fine-needle aspiration and fine-needle biopsy should be used to determine whether the lesion is a GIST or leiomyoma. Using structural assessment and staining will allow for the differentiation of mesenchymal tumors and assessment of their malignant potential.

To remove SELs, multiple endoscopic resection techniques may be appropriate, depending on the layer of origin, size, and location, with the goal of complete, en bloc resection with no disruption to the wall or capsule of the lesion. These techniques should be limited to endoscopists skilled in advanced tissue resection.

SELs without malignant potential, such as lipoma or pancreatic rest, don’t need further evaluation or surveillance.

SELs that are ulcerated, bleeding, or causing symptoms should be considered for resection.

Other lesions are managed with resection or surveillance based on pathology. For example, leiomyomas, which are benign and most often found in the esophagus, generally don’t require surveillance or resection. On the other hand, all GISTs have some malignant potential, and management varies by size, location, and presence of symptoms. GISTs larger than 2 cm, should be considered for resection. Some GISTs between 2 cm and 4 cm without high-risk features can be removed by using advanced endoscopic resection techniques.

The determination for resection in all cases should include a multidisciplinary approach, with confirmation of a low mitotic index and lack of metastatic disease on cross-sectional imaging.

“The ultimate goal of endoscopic resection is to have a complete resection,” the authors wrote. “Determining the layer of involvement by EUS is critical in planning resection techniques.”

The authors reported no grant support or funding sources for this report. One author serves as a consultant for Boston Scientific, Fujifilm, Intuitive Surgical, Medtronic, and Olympus. The remaining authors disclosed no conflicts.

Publications
Topics
Sections

 

The proper management of subepithelial lesions (SELs) depends on the size, histopathology, malignant potential, and presence of symptoms, according to a new American Gastroenterological Association clinical practice update published in Clinical Gastroenterology and Hepatology.

“SELs are found in 1 in every 300 endoscopies, and two-thirds of these lesions are located in the stomach,” explained Kaveh Sharzehi, MD, an associate professor of medicine in the division of gastroenterology and hepatology at Oregon Health & Science University, Portland, and colleagues. “They represent a heterogeneous group of lesions including nonneoplastic lesions such as ectopic pancreatic tissue and neoplastic lesions. The neoplastic SELs can vary from lesions with no malignant potential such as lipomas to those with malignant potential such as gastrointestinal stromal tumors (GISTs). The majority of SELs are small and found incidentally.”

Dr. Kaveh Sharzehi

The authors developed 10 clinical practice advice statements on the diagnosis and management of subepithelial lesions based on a review of the published literature and expert opinion.

First, standard mucosal biopsies often don’t reach deep enough to obtain a pathologic diagnosis for SELs because the lesions have normal overlying mucosa. Forceps bite-on-bite/deep-well biopsies or tunnel biopsies may help to establish a pathologic diagnosis.

Used as an adjunct to standard endoscopy, endoscopic ultrasound (EUS) has become the primary method for determining diagnostic and prognostic characteristics of SELs – such as the layer of origin, echogenicity, and presence of blood vessels within the lesion. It can also help with tissue acquisition.

For SELs arising from the submucosa, EUS-guided fine-needle aspiration and fine-needle biopsy have evolved as widely used methods for obtaining tissue. For SELs arising from muscularis propria, fine-needle aspiration and fine-needle biopsy should be used to determine whether the lesion is a GIST or leiomyoma. Using structural assessment and staining will allow for the differentiation of mesenchymal tumors and assessment of their malignant potential.

To remove SELs, multiple endoscopic resection techniques may be appropriate, depending on the layer of origin, size, and location, with the goal of complete, en bloc resection with no disruption to the wall or capsule of the lesion. These techniques should be limited to endoscopists skilled in advanced tissue resection.

SELs without malignant potential, such as lipoma or pancreatic rest, don’t need further evaluation or surveillance.

SELs that are ulcerated, bleeding, or causing symptoms should be considered for resection.

Other lesions are managed with resection or surveillance based on pathology. For example, leiomyomas, which are benign and most often found in the esophagus, generally don’t require surveillance or resection. On the other hand, all GISTs have some malignant potential, and management varies by size, location, and presence of symptoms. GISTs larger than 2 cm, should be considered for resection. Some GISTs between 2 cm and 4 cm without high-risk features can be removed by using advanced endoscopic resection techniques.

The determination for resection in all cases should include a multidisciplinary approach, with confirmation of a low mitotic index and lack of metastatic disease on cross-sectional imaging.

“The ultimate goal of endoscopic resection is to have a complete resection,” the authors wrote. “Determining the layer of involvement by EUS is critical in planning resection techniques.”

The authors reported no grant support or funding sources for this report. One author serves as a consultant for Boston Scientific, Fujifilm, Intuitive Surgical, Medtronic, and Olympus. The remaining authors disclosed no conflicts.

 

The proper management of subepithelial lesions (SELs) depends on the size, histopathology, malignant potential, and presence of symptoms, according to a new American Gastroenterological Association clinical practice update published in Clinical Gastroenterology and Hepatology.

“SELs are found in 1 in every 300 endoscopies, and two-thirds of these lesions are located in the stomach,” explained Kaveh Sharzehi, MD, an associate professor of medicine in the division of gastroenterology and hepatology at Oregon Health & Science University, Portland, and colleagues. “They represent a heterogeneous group of lesions including nonneoplastic lesions such as ectopic pancreatic tissue and neoplastic lesions. The neoplastic SELs can vary from lesions with no malignant potential such as lipomas to those with malignant potential such as gastrointestinal stromal tumors (GISTs). The majority of SELs are small and found incidentally.”

Dr. Kaveh Sharzehi

The authors developed 10 clinical practice advice statements on the diagnosis and management of subepithelial lesions based on a review of the published literature and expert opinion.

First, standard mucosal biopsies often don’t reach deep enough to obtain a pathologic diagnosis for SELs because the lesions have normal overlying mucosa. Forceps bite-on-bite/deep-well biopsies or tunnel biopsies may help to establish a pathologic diagnosis.

Used as an adjunct to standard endoscopy, endoscopic ultrasound (EUS) has become the primary method for determining diagnostic and prognostic characteristics of SELs – such as the layer of origin, echogenicity, and presence of blood vessels within the lesion. It can also help with tissue acquisition.

For SELs arising from the submucosa, EUS-guided fine-needle aspiration and fine-needle biopsy have evolved as widely used methods for obtaining tissue. For SELs arising from muscularis propria, fine-needle aspiration and fine-needle biopsy should be used to determine whether the lesion is a GIST or leiomyoma. Using structural assessment and staining will allow for the differentiation of mesenchymal tumors and assessment of their malignant potential.

To remove SELs, multiple endoscopic resection techniques may be appropriate, depending on the layer of origin, size, and location, with the goal of complete, en bloc resection with no disruption to the wall or capsule of the lesion. These techniques should be limited to endoscopists skilled in advanced tissue resection.

SELs without malignant potential, such as lipoma or pancreatic rest, don’t need further evaluation or surveillance.

SELs that are ulcerated, bleeding, or causing symptoms should be considered for resection.

Other lesions are managed with resection or surveillance based on pathology. For example, leiomyomas, which are benign and most often found in the esophagus, generally don’t require surveillance or resection. On the other hand, all GISTs have some malignant potential, and management varies by size, location, and presence of symptoms. GISTs larger than 2 cm, should be considered for resection. Some GISTs between 2 cm and 4 cm without high-risk features can be removed by using advanced endoscopic resection techniques.

The determination for resection in all cases should include a multidisciplinary approach, with confirmation of a low mitotic index and lack of metastatic disease on cross-sectional imaging.

“The ultimate goal of endoscopic resection is to have a complete resection,” the authors wrote. “Determining the layer of involvement by EUS is critical in planning resection techniques.”

The authors reported no grant support or funding sources for this report. One author serves as a consultant for Boston Scientific, Fujifilm, Intuitive Surgical, Medtronic, and Olympus. The remaining authors disclosed no conflicts.

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM CLINICAL GASTROENTEROLOGY AND HEPATOLOGY

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

AGA Clinical Practice Update: Expert review of management of refractory celiac disease

Article Type
Changed
Wed, 11/09/2022 - 11:44

The diagnosis and management of refractory celiac disease remains challenging, but ongoing studies can provide the proper diagnostic criteria and identify the optimal management strategies, according to a new American Gastroenterological Association expert review published in Gastroenterology.

Celiac disease is present in about 1% of the U.S. population and can cause various symptoms, wrote Peter H. R. Green, MD, director of the Celiac Disease Center at Columbia University, New York, and colleagues. Adhering to a strict gluten-free diet can improve symptoms, normalize serum antibody levels, and reverse small bowel villous atrophy. However, persistent or recurrent symptoms and elevated celiac antibodies can persist in some patients after a year of trying a gluten-free diet, a condition called nonresponsive celiac disease. In some patients, this raises concern for refractory celiac disease, or RCD.

“RCD is believed to occur in only approximately 1% of patients with celiac disease, although this may be an overestimate, as data are obtained from referral centers,” the authors wrote.

RCD can be classified into two subtypes with different diagnostic criteria, prognoses, and therapy responses. The first, called RCD1, is characterized by villous atrophy but has intraepithelial lymphocytes similar to conventional celiac disease. The other, called RCD2, is characterized by aberrant clonal T-cell expansion in the intestinal tract and other organs, has a poorer prognosis than RCD1, and has a risk of developing ulcerative jejunoileitis or enteropathy-associated T-cell lymphoma.

The experts developed 10 clinical practice advice statements based on a review of the published literature and expert opinion.

First, in patients who have persistent or recurring symptoms, an initial celiac disease diagnosis should be confirmed through review of prior diagnostic testing, including serologies, endoscopies, and histologic findings. Celiac disease can overlap with other gastrointestinal conditions, and some pathologic findings aren’t specific to celiac disease. Results of serologic testing with tissue transglutaminase immunoglobulin A, deamidated gliadin peptide IgA and IgG, and endomysial antibodies should be reviewed or obtained if not previously performed.

Next, in those with confirmed but nonresponsive celiac disease, ongoing gluten ingestion should be excluded as a cause of symptoms with serologic testing, dietitian review, and potentially detection of immunogenic peptides in stool or urine samples. The authors noted that persistent gluten ingestion, whether intentional or inadvertent, accounts for 40%-50% of patients with nonresponsive celiac disease. In these cases, esophagogastroduodenoscopy and small bowel biopsies should be performed to look for persistent villous atrophy, which can also be caused by common variable immunodeficiency, autoimmune enteropathy, tropical sprue, and medication-induced enteropathy. Patients with villous atrophy due to other causes won’t respond to a gluten-free diet.

After excluding gluten, clinicians should perform a systematic evaluation for other potential causes of symptoms, including functional bowel disorders, lactose or fructose intolerance, microscopic colitis, pancreatic insufficiency, inflammatory bowel disease, and small intestinal bacterial growth. Irritable bowel syndrome, for instance, may contribute to persistent symptoms and respond to fermentable oligo-, di-, and monosaccharides and polyols (FODMAP) restriction. RCD should be strongly considered in patients with persistent symptoms or signs of malabsorption after the exclusion of the other causes.

To distinguish between the two subtypes of RCD and exclude enteropathy-associated T-cell lymphoma, clinicians should use flow cytometry, immunohistochemistry, and T-cell receptor rearrangement studies. RCD1 has a normal intraepithelial lymphocyte population, and RCD2 has an aberrant, clonal intraepithelial lymphocyte population. Consulting with a hematopathologist may be necessary to interpret these studies.

After RCD2 is diagnosed, complications such as enteropathy-associated T-cell lymphoma and ulcerative jejunoileitis should be excluded through small bowel imaging with capsule endoscopy and either computed tomography (CT) or magnetic resonance enterography. In general, the extent and severity of villous atrophy is greater in patients with RCD2, compared with RCD1.

In patients diagnosed with RCD, clinicians should complete a detailed nutritional assessment with investigation of micronutrient and macronutrient deficiencies. Check albumin as an independent prognostic factor. Then, try to correct deficiencies with oral supplements. Malnourished patients may need enteral support, and those with severe malnutrition due to malabsorption may need parenteral support.

So far, RCD management suggestions are based on small retrospective studies and expert opinion, with minimal prospective data and no Food and Drug Administration–approved therapies. The goals should be to improve symptoms and duodenal mucosal abnormalities, manage malnutrition, and prevent lymphoma. Glucocorticoids are considered first-line therapy, typically open-capsule budesonide given as 3 mg three times daily. Prednisone serves as an alternative with proven efficacy but a higher risk for adverse effects.

The optimal choice for second-line therapy is unknown, but the addition of an immunosuppressant agent to steroids appears to be effective in RCD1, including azathioprine, mercaptopurine, and tioguanine. The best treatment for RCD2 is unknown, though clinical response has been reported with steroids, and cladribine has been well tolerated in some patients.

Patients with RCD who don’t respond to steroids may benefit from referral to a center with expertise for management or evaluation for inclusion in clinical trials. Frequent medical visits are advised until the disease is well controlled, with regular follow-up after that. 


Ultimately, “patients with RCD benefit from evaluation and regular follow-up by a multidisciplinary team, including gastroenterologists and dietitians, to assess clinical and histologic response to therapy,” the authors wrote. “Identify local experts with expertise in celiac disease to assist with management.”

The authors reported no grant support or funding sources for this study. One author has received research report from Freenome, and another is on the celiac disease advisory board for Takeda. The remaining authors disclosed no conflicts.

Publications
Topics
Sections

The diagnosis and management of refractory celiac disease remains challenging, but ongoing studies can provide the proper diagnostic criteria and identify the optimal management strategies, according to a new American Gastroenterological Association expert review published in Gastroenterology.

Celiac disease is present in about 1% of the U.S. population and can cause various symptoms, wrote Peter H. R. Green, MD, director of the Celiac Disease Center at Columbia University, New York, and colleagues. Adhering to a strict gluten-free diet can improve symptoms, normalize serum antibody levels, and reverse small bowel villous atrophy. However, persistent or recurrent symptoms and elevated celiac antibodies can persist in some patients after a year of trying a gluten-free diet, a condition called nonresponsive celiac disease. In some patients, this raises concern for refractory celiac disease, or RCD.

“RCD is believed to occur in only approximately 1% of patients with celiac disease, although this may be an overestimate, as data are obtained from referral centers,” the authors wrote.

RCD can be classified into two subtypes with different diagnostic criteria, prognoses, and therapy responses. The first, called RCD1, is characterized by villous atrophy but has intraepithelial lymphocytes similar to conventional celiac disease. The other, called RCD2, is characterized by aberrant clonal T-cell expansion in the intestinal tract and other organs, has a poorer prognosis than RCD1, and has a risk of developing ulcerative jejunoileitis or enteropathy-associated T-cell lymphoma.

The experts developed 10 clinical practice advice statements based on a review of the published literature and expert opinion.

First, in patients who have persistent or recurring symptoms, an initial celiac disease diagnosis should be confirmed through review of prior diagnostic testing, including serologies, endoscopies, and histologic findings. Celiac disease can overlap with other gastrointestinal conditions, and some pathologic findings aren’t specific to celiac disease. Results of serologic testing with tissue transglutaminase immunoglobulin A, deamidated gliadin peptide IgA and IgG, and endomysial antibodies should be reviewed or obtained if not previously performed.

Next, in those with confirmed but nonresponsive celiac disease, ongoing gluten ingestion should be excluded as a cause of symptoms with serologic testing, dietitian review, and potentially detection of immunogenic peptides in stool or urine samples. The authors noted that persistent gluten ingestion, whether intentional or inadvertent, accounts for 40%-50% of patients with nonresponsive celiac disease. In these cases, esophagogastroduodenoscopy and small bowel biopsies should be performed to look for persistent villous atrophy, which can also be caused by common variable immunodeficiency, autoimmune enteropathy, tropical sprue, and medication-induced enteropathy. Patients with villous atrophy due to other causes won’t respond to a gluten-free diet.

After excluding gluten, clinicians should perform a systematic evaluation for other potential causes of symptoms, including functional bowel disorders, lactose or fructose intolerance, microscopic colitis, pancreatic insufficiency, inflammatory bowel disease, and small intestinal bacterial growth. Irritable bowel syndrome, for instance, may contribute to persistent symptoms and respond to fermentable oligo-, di-, and monosaccharides and polyols (FODMAP) restriction. RCD should be strongly considered in patients with persistent symptoms or signs of malabsorption after the exclusion of the other causes.

To distinguish between the two subtypes of RCD and exclude enteropathy-associated T-cell lymphoma, clinicians should use flow cytometry, immunohistochemistry, and T-cell receptor rearrangement studies. RCD1 has a normal intraepithelial lymphocyte population, and RCD2 has an aberrant, clonal intraepithelial lymphocyte population. Consulting with a hematopathologist may be necessary to interpret these studies.

After RCD2 is diagnosed, complications such as enteropathy-associated T-cell lymphoma and ulcerative jejunoileitis should be excluded through small bowel imaging with capsule endoscopy and either computed tomography (CT) or magnetic resonance enterography. In general, the extent and severity of villous atrophy is greater in patients with RCD2, compared with RCD1.

In patients diagnosed with RCD, clinicians should complete a detailed nutritional assessment with investigation of micronutrient and macronutrient deficiencies. Check albumin as an independent prognostic factor. Then, try to correct deficiencies with oral supplements. Malnourished patients may need enteral support, and those with severe malnutrition due to malabsorption may need parenteral support.

So far, RCD management suggestions are based on small retrospective studies and expert opinion, with minimal prospective data and no Food and Drug Administration–approved therapies. The goals should be to improve symptoms and duodenal mucosal abnormalities, manage malnutrition, and prevent lymphoma. Glucocorticoids are considered first-line therapy, typically open-capsule budesonide given as 3 mg three times daily. Prednisone serves as an alternative with proven efficacy but a higher risk for adverse effects.

The optimal choice for second-line therapy is unknown, but the addition of an immunosuppressant agent to steroids appears to be effective in RCD1, including azathioprine, mercaptopurine, and tioguanine. The best treatment for RCD2 is unknown, though clinical response has been reported with steroids, and cladribine has been well tolerated in some patients.

Patients with RCD who don’t respond to steroids may benefit from referral to a center with expertise for management or evaluation for inclusion in clinical trials. Frequent medical visits are advised until the disease is well controlled, with regular follow-up after that. 


Ultimately, “patients with RCD benefit from evaluation and regular follow-up by a multidisciplinary team, including gastroenterologists and dietitians, to assess clinical and histologic response to therapy,” the authors wrote. “Identify local experts with expertise in celiac disease to assist with management.”

The authors reported no grant support or funding sources for this study. One author has received research report from Freenome, and another is on the celiac disease advisory board for Takeda. The remaining authors disclosed no conflicts.

The diagnosis and management of refractory celiac disease remains challenging, but ongoing studies can provide the proper diagnostic criteria and identify the optimal management strategies, according to a new American Gastroenterological Association expert review published in Gastroenterology.

Celiac disease is present in about 1% of the U.S. population and can cause various symptoms, wrote Peter H. R. Green, MD, director of the Celiac Disease Center at Columbia University, New York, and colleagues. Adhering to a strict gluten-free diet can improve symptoms, normalize serum antibody levels, and reverse small bowel villous atrophy. However, persistent or recurrent symptoms and elevated celiac antibodies can persist in some patients after a year of trying a gluten-free diet, a condition called nonresponsive celiac disease. In some patients, this raises concern for refractory celiac disease, or RCD.

“RCD is believed to occur in only approximately 1% of patients with celiac disease, although this may be an overestimate, as data are obtained from referral centers,” the authors wrote.

RCD can be classified into two subtypes with different diagnostic criteria, prognoses, and therapy responses. The first, called RCD1, is characterized by villous atrophy but has intraepithelial lymphocytes similar to conventional celiac disease. The other, called RCD2, is characterized by aberrant clonal T-cell expansion in the intestinal tract and other organs, has a poorer prognosis than RCD1, and has a risk of developing ulcerative jejunoileitis or enteropathy-associated T-cell lymphoma.

The experts developed 10 clinical practice advice statements based on a review of the published literature and expert opinion.

First, in patients who have persistent or recurring symptoms, an initial celiac disease diagnosis should be confirmed through review of prior diagnostic testing, including serologies, endoscopies, and histologic findings. Celiac disease can overlap with other gastrointestinal conditions, and some pathologic findings aren’t specific to celiac disease. Results of serologic testing with tissue transglutaminase immunoglobulin A, deamidated gliadin peptide IgA and IgG, and endomysial antibodies should be reviewed or obtained if not previously performed.

Next, in those with confirmed but nonresponsive celiac disease, ongoing gluten ingestion should be excluded as a cause of symptoms with serologic testing, dietitian review, and potentially detection of immunogenic peptides in stool or urine samples. The authors noted that persistent gluten ingestion, whether intentional or inadvertent, accounts for 40%-50% of patients with nonresponsive celiac disease. In these cases, esophagogastroduodenoscopy and small bowel biopsies should be performed to look for persistent villous atrophy, which can also be caused by common variable immunodeficiency, autoimmune enteropathy, tropical sprue, and medication-induced enteropathy. Patients with villous atrophy due to other causes won’t respond to a gluten-free diet.

After excluding gluten, clinicians should perform a systematic evaluation for other potential causes of symptoms, including functional bowel disorders, lactose or fructose intolerance, microscopic colitis, pancreatic insufficiency, inflammatory bowel disease, and small intestinal bacterial growth. Irritable bowel syndrome, for instance, may contribute to persistent symptoms and respond to fermentable oligo-, di-, and monosaccharides and polyols (FODMAP) restriction. RCD should be strongly considered in patients with persistent symptoms or signs of malabsorption after the exclusion of the other causes.

To distinguish between the two subtypes of RCD and exclude enteropathy-associated T-cell lymphoma, clinicians should use flow cytometry, immunohistochemistry, and T-cell receptor rearrangement studies. RCD1 has a normal intraepithelial lymphocyte population, and RCD2 has an aberrant, clonal intraepithelial lymphocyte population. Consulting with a hematopathologist may be necessary to interpret these studies.

After RCD2 is diagnosed, complications such as enteropathy-associated T-cell lymphoma and ulcerative jejunoileitis should be excluded through small bowel imaging with capsule endoscopy and either computed tomography (CT) or magnetic resonance enterography. In general, the extent and severity of villous atrophy is greater in patients with RCD2, compared with RCD1.

In patients diagnosed with RCD, clinicians should complete a detailed nutritional assessment with investigation of micronutrient and macronutrient deficiencies. Check albumin as an independent prognostic factor. Then, try to correct deficiencies with oral supplements. Malnourished patients may need enteral support, and those with severe malnutrition due to malabsorption may need parenteral support.

So far, RCD management suggestions are based on small retrospective studies and expert opinion, with minimal prospective data and no Food and Drug Administration–approved therapies. The goals should be to improve symptoms and duodenal mucosal abnormalities, manage malnutrition, and prevent lymphoma. Glucocorticoids are considered first-line therapy, typically open-capsule budesonide given as 3 mg three times daily. Prednisone serves as an alternative with proven efficacy but a higher risk for adverse effects.

The optimal choice for second-line therapy is unknown, but the addition of an immunosuppressant agent to steroids appears to be effective in RCD1, including azathioprine, mercaptopurine, and tioguanine. The best treatment for RCD2 is unknown, though clinical response has been reported with steroids, and cladribine has been well tolerated in some patients.

Patients with RCD who don’t respond to steroids may benefit from referral to a center with expertise for management or evaluation for inclusion in clinical trials. Frequent medical visits are advised until the disease is well controlled, with regular follow-up after that. 


Ultimately, “patients with RCD benefit from evaluation and regular follow-up by a multidisciplinary team, including gastroenterologists and dietitians, to assess clinical and histologic response to therapy,” the authors wrote. “Identify local experts with expertise in celiac disease to assist with management.”

The authors reported no grant support or funding sources for this study. One author has received research report from Freenome, and another is on the celiac disease advisory board for Takeda. The remaining authors disclosed no conflicts.

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM GASTROENTEROLOGY

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Complete endoscopic healing key when stopping anti-TNFs in IBD

Article Type
Changed
Wed, 09/28/2022 - 17:02

 

The level of remission in patients with remitting inflammatory bowel disease (IBD) appears to play a major role in whether they will relapse after treatment when biologic therapies are discontinued, according to a new prospective study.

Patients with complete endoscopic healing have half the rate of relapse after withdrawal of anti-tumor necrosis factor alpha (anti-TNF) treatment than those with only partial healing, according to a study published online in Clinical Gastroenterology and Hepatology.

“Applying strict criteria for endoscopic healing and mesalamine treatment ... may lower the risk of relapse after withdrawal of anti-TNF treatment,” write Bas Oldenburg, MD, PhD, a professor at University Medical Center Utrecht, the Netherlands, and colleagues in their analysis of 81 patients.

De-escalation of anti-TNF treatment in IBD patients in remission has the potential to “reduce side effects, including risks of serious infections and malignancies, decrease health care expenditures, and meet patients’ preferences,” they note.

However, withdrawal of the drugs increases the risk of relapse by 30%-45% at 12 months. When patients relapse, reintroduction of anti-TNF therapy returns over 80% to remission.

Although no consensus exists on how to select patients for therapy de-escalation, evidence suggests that persistent inflammation affects outcomes and that the “depth” of endoscopic healing is a key indicator, the authors note.
 

Study details

To further the knowledge base, they conducted a prospective study of patients in remission to determine the relapse rate following de-escalation of anti-TNF therapy; evaluate relapse factors, including degree of endoscopic healing; and assess outcomes after reintroduction of anti-TNF therapy.

The study was limited to adult patients with IBD with at least 6 months of corticosteroid-free clinical remission, confirmed baseline clinical remission and endoscopic healing, no current hospitalization, and no pregnancy.

The patients underwent elective discontinuation of anti-TNF therapy between 2018 and 2020. The recommended protocol was to measure C-reactive protein (CRP) and fecal calprotectin at 3, 6, 12, and 24 months and to perform endoscopy at 12 months.

Patients also completed questionnaires at baseline and at 3, 6, 12, and 24 months. The authors selected the patient–Harvey-Bradshaw Index for patients with Crohn’s disease and the patient–Simple Clinical Colitis Activity Index for patients with ulcerative colitis and unclassified IBD, as well as the short IBD Quality of Life measure.

Of the 81 patients from 13 centers who took part, 51% had Crohn’s disease. The median duration of remission at baseline was 3.5 years, and the median disease duration was 9.1 years.

All patients had evidence of endoscopic healing, and 88% met the strict criteria for complete endoscopic healing. In 34%, trough levels of anti-TNF treatments were judged to be subtherapeutic.

After withdrawal of the drugs, 25.9% of patients continued on immunomodulators.

Over a median follow-up of 2 years, 49% of patients relapsed, which was confirmed via endoscopy, fecal calprotectin, or CRP in 83% of cases and inferred from treatment escalation for clinical flare in 17%. Rates of relapse were comparable between patients with Crohn’s disease and ulcerative colitis or unclassified IBD and between those discontinuing adalimumab and those stopping infliximab.
 

Better healing, better outcomes

However, analysis showed that partial endoscopic healing was independently associated with a higher risk of relapse, at an adjusted hazard ratio versus complete endoscopic healing of 3.28.

 

 

At 12 months, 70% of patients with partial endoscopic healing had relapsed versus 35% of those with complete endoscopic healing.

Treatment with the anti-inflammatory agent mesalamine (multiple brands) was independently associated with a reduced risk of relapse, at an adjusted hazard ratio of 0.08. No other potential predictors of relapse were identified.

Of the patients who relapsed, 75% restarted anti-TNF treatment, and the majority (87%) were restarted on the same agent at a median of 0.9 years since its withdrawal and a median of 24 days since the onset of relapse.

Clinical remission was achieved at 3 months in 73% of patients who restarted anti-TNF therapy, which was found to restore quality of life and well-being in relapsed patients, the authors report.
 

Reluctance remains

Stephen B. Hanauer, MD, professor of medicine (gastroenterology and hepatology) at Northwestern University Feinberg School of Medicine, Chicago, said the findings “reinforce the benefits of the maintenance versus the withdrawal of therapy” and “the deeper the remission” the more likely it is to be sustained.

The 35% relapse rate at 12 months, even in patients with compete endoscopic healing, indicates that treatment should be maintained, Dr. Hanauer said.

“What is also relevant, but was not evaluated, is the additional endpoint of histologic healing, which is likely to sustain remissions even longer,” he added.

Nevertheless, Dr. Hanauer said, the “observed relapse rate is important to discuss in shared decision-making with patients.”

The findings are interesting, but the study didn’t follow the patients for long enough to understand why 35% of those with complete endoscopic healing relapsed, Miguel Regueiro, MD, chair of the Cleveland Clinic’s Digestive Disease & Surgery Institute, told this news organization.

“Are there predictors, factors, or other treatments that could be used to reduce that 35% risk of relapse further?” he questioned.

Although the study didn’t clear up that question for Dr. Regueiro, he found it compelling that mesalamine continuation resulted in higher rates of sustained remission after anti-TNF withdrawal among patients with ulcerative colitis.

Dr. Regueiro said that he will not begin recommending withdrawal of advanced therapies, including anti-TNF drugs, in patients who have achieved a stable remission.

“We have not yet found the cure for IBD, and my concern is that patients may relapse with more severe disease than previously and that recurrence of inflammation could have potential risks for complications,” he said.

“Nonetheless, this study is intriguing and important, and at least prompts the discussion of withdrawing therapy in those who have achieved a deep endoscopic remission for a sustained period of time,” Dr. Regueiro added.

The study received support from the Dutch Health Insurance Innovation Fund.

Dr. Oldenburg declares relationships with AbbVie, Celltrion, Ferring, Takeda, Galapagos, Pfizer, Cablon, PBMS, Janssen, and MSD. Other authors also declare numerous relationships. The full list can be found with the original article. Dr. Hanauer declares relationships with AbbVie, Janssen, Pfizer, and Boehringer Ingelheim. Dr. Regueiro declares relationships with AbbVie, Janssen, UCB, Takeda, Pfizer, Miraca Labs, Amgen, Celgene, Seres, Allergan, Genentech, Gilead, Salix, Prometheus, Lilly, TARGET Pharma Solutions, ALFASIGMA, S.p.A., BMS, CME Outfitters, Imedex, GI Health Foundation, Cornerstones, Remedy, MJH Life Sciences, Medscape, MDEducation, WebMD, and HMPGlobal.

Help your patients better understand their IBD treatment options by sharing AGA’s patient education, “Living with IBD,” in the AGA GI Patient Center at www.gastro.org/IBD. 

A version of this article first appeared on Medscape.com.

Publications
Topics
Sections

 

The level of remission in patients with remitting inflammatory bowel disease (IBD) appears to play a major role in whether they will relapse after treatment when biologic therapies are discontinued, according to a new prospective study.

Patients with complete endoscopic healing have half the rate of relapse after withdrawal of anti-tumor necrosis factor alpha (anti-TNF) treatment than those with only partial healing, according to a study published online in Clinical Gastroenterology and Hepatology.

“Applying strict criteria for endoscopic healing and mesalamine treatment ... may lower the risk of relapse after withdrawal of anti-TNF treatment,” write Bas Oldenburg, MD, PhD, a professor at University Medical Center Utrecht, the Netherlands, and colleagues in their analysis of 81 patients.

De-escalation of anti-TNF treatment in IBD patients in remission has the potential to “reduce side effects, including risks of serious infections and malignancies, decrease health care expenditures, and meet patients’ preferences,” they note.

However, withdrawal of the drugs increases the risk of relapse by 30%-45% at 12 months. When patients relapse, reintroduction of anti-TNF therapy returns over 80% to remission.

Although no consensus exists on how to select patients for therapy de-escalation, evidence suggests that persistent inflammation affects outcomes and that the “depth” of endoscopic healing is a key indicator, the authors note.
 

Study details

To further the knowledge base, they conducted a prospective study of patients in remission to determine the relapse rate following de-escalation of anti-TNF therapy; evaluate relapse factors, including degree of endoscopic healing; and assess outcomes after reintroduction of anti-TNF therapy.

The study was limited to adult patients with IBD with at least 6 months of corticosteroid-free clinical remission, confirmed baseline clinical remission and endoscopic healing, no current hospitalization, and no pregnancy.

The patients underwent elective discontinuation of anti-TNF therapy between 2018 and 2020. The recommended protocol was to measure C-reactive protein (CRP) and fecal calprotectin at 3, 6, 12, and 24 months and to perform endoscopy at 12 months.

Patients also completed questionnaires at baseline and at 3, 6, 12, and 24 months. The authors selected the patient–Harvey-Bradshaw Index for patients with Crohn’s disease and the patient–Simple Clinical Colitis Activity Index for patients with ulcerative colitis and unclassified IBD, as well as the short IBD Quality of Life measure.

Of the 81 patients from 13 centers who took part, 51% had Crohn’s disease. The median duration of remission at baseline was 3.5 years, and the median disease duration was 9.1 years.

All patients had evidence of endoscopic healing, and 88% met the strict criteria for complete endoscopic healing. In 34%, trough levels of anti-TNF treatments were judged to be subtherapeutic.

After withdrawal of the drugs, 25.9% of patients continued on immunomodulators.

Over a median follow-up of 2 years, 49% of patients relapsed, which was confirmed via endoscopy, fecal calprotectin, or CRP in 83% of cases and inferred from treatment escalation for clinical flare in 17%. Rates of relapse were comparable between patients with Crohn’s disease and ulcerative colitis or unclassified IBD and between those discontinuing adalimumab and those stopping infliximab.
 

Better healing, better outcomes

However, analysis showed that partial endoscopic healing was independently associated with a higher risk of relapse, at an adjusted hazard ratio versus complete endoscopic healing of 3.28.

 

 

At 12 months, 70% of patients with partial endoscopic healing had relapsed versus 35% of those with complete endoscopic healing.

Treatment with the anti-inflammatory agent mesalamine (multiple brands) was independently associated with a reduced risk of relapse, at an adjusted hazard ratio of 0.08. No other potential predictors of relapse were identified.

Of the patients who relapsed, 75% restarted anti-TNF treatment, and the majority (87%) were restarted on the same agent at a median of 0.9 years since its withdrawal and a median of 24 days since the onset of relapse.

Clinical remission was achieved at 3 months in 73% of patients who restarted anti-TNF therapy, which was found to restore quality of life and well-being in relapsed patients, the authors report.
 

Reluctance remains

Stephen B. Hanauer, MD, professor of medicine (gastroenterology and hepatology) at Northwestern University Feinberg School of Medicine, Chicago, said the findings “reinforce the benefits of the maintenance versus the withdrawal of therapy” and “the deeper the remission” the more likely it is to be sustained.

The 35% relapse rate at 12 months, even in patients with compete endoscopic healing, indicates that treatment should be maintained, Dr. Hanauer said.

“What is also relevant, but was not evaluated, is the additional endpoint of histologic healing, which is likely to sustain remissions even longer,” he added.

Nevertheless, Dr. Hanauer said, the “observed relapse rate is important to discuss in shared decision-making with patients.”

The findings are interesting, but the study didn’t follow the patients for long enough to understand why 35% of those with complete endoscopic healing relapsed, Miguel Regueiro, MD, chair of the Cleveland Clinic’s Digestive Disease & Surgery Institute, told this news organization.

“Are there predictors, factors, or other treatments that could be used to reduce that 35% risk of relapse further?” he questioned.

Although the study didn’t clear up that question for Dr. Regueiro, he found it compelling that mesalamine continuation resulted in higher rates of sustained remission after anti-TNF withdrawal among patients with ulcerative colitis.

Dr. Regueiro said that he will not begin recommending withdrawal of advanced therapies, including anti-TNF drugs, in patients who have achieved a stable remission.

“We have not yet found the cure for IBD, and my concern is that patients may relapse with more severe disease than previously and that recurrence of inflammation could have potential risks for complications,” he said.

“Nonetheless, this study is intriguing and important, and at least prompts the discussion of withdrawing therapy in those who have achieved a deep endoscopic remission for a sustained period of time,” Dr. Regueiro added.

The study received support from the Dutch Health Insurance Innovation Fund.

Dr. Oldenburg declares relationships with AbbVie, Celltrion, Ferring, Takeda, Galapagos, Pfizer, Cablon, PBMS, Janssen, and MSD. Other authors also declare numerous relationships. The full list can be found with the original article. Dr. Hanauer declares relationships with AbbVie, Janssen, Pfizer, and Boehringer Ingelheim. Dr. Regueiro declares relationships with AbbVie, Janssen, UCB, Takeda, Pfizer, Miraca Labs, Amgen, Celgene, Seres, Allergan, Genentech, Gilead, Salix, Prometheus, Lilly, TARGET Pharma Solutions, ALFASIGMA, S.p.A., BMS, CME Outfitters, Imedex, GI Health Foundation, Cornerstones, Remedy, MJH Life Sciences, Medscape, MDEducation, WebMD, and HMPGlobal.

Help your patients better understand their IBD treatment options by sharing AGA’s patient education, “Living with IBD,” in the AGA GI Patient Center at www.gastro.org/IBD. 

A version of this article first appeared on Medscape.com.

 

The level of remission in patients with remitting inflammatory bowel disease (IBD) appears to play a major role in whether they will relapse after treatment when biologic therapies are discontinued, according to a new prospective study.

Patients with complete endoscopic healing have half the rate of relapse after withdrawal of anti-tumor necrosis factor alpha (anti-TNF) treatment than those with only partial healing, according to a study published online in Clinical Gastroenterology and Hepatology.

“Applying strict criteria for endoscopic healing and mesalamine treatment ... may lower the risk of relapse after withdrawal of anti-TNF treatment,” write Bas Oldenburg, MD, PhD, a professor at University Medical Center Utrecht, the Netherlands, and colleagues in their analysis of 81 patients.

De-escalation of anti-TNF treatment in IBD patients in remission has the potential to “reduce side effects, including risks of serious infections and malignancies, decrease health care expenditures, and meet patients’ preferences,” they note.

However, withdrawal of the drugs increases the risk of relapse by 30%-45% at 12 months. When patients relapse, reintroduction of anti-TNF therapy returns over 80% to remission.

Although no consensus exists on how to select patients for therapy de-escalation, evidence suggests that persistent inflammation affects outcomes and that the “depth” of endoscopic healing is a key indicator, the authors note.
 

Study details

To further the knowledge base, they conducted a prospective study of patients in remission to determine the relapse rate following de-escalation of anti-TNF therapy; evaluate relapse factors, including degree of endoscopic healing; and assess outcomes after reintroduction of anti-TNF therapy.

The study was limited to adult patients with IBD with at least 6 months of corticosteroid-free clinical remission, confirmed baseline clinical remission and endoscopic healing, no current hospitalization, and no pregnancy.

The patients underwent elective discontinuation of anti-TNF therapy between 2018 and 2020. The recommended protocol was to measure C-reactive protein (CRP) and fecal calprotectin at 3, 6, 12, and 24 months and to perform endoscopy at 12 months.

Patients also completed questionnaires at baseline and at 3, 6, 12, and 24 months. The authors selected the patient–Harvey-Bradshaw Index for patients with Crohn’s disease and the patient–Simple Clinical Colitis Activity Index for patients with ulcerative colitis and unclassified IBD, as well as the short IBD Quality of Life measure.

Of the 81 patients from 13 centers who took part, 51% had Crohn’s disease. The median duration of remission at baseline was 3.5 years, and the median disease duration was 9.1 years.

All patients had evidence of endoscopic healing, and 88% met the strict criteria for complete endoscopic healing. In 34%, trough levels of anti-TNF treatments were judged to be subtherapeutic.

After withdrawal of the drugs, 25.9% of patients continued on immunomodulators.

Over a median follow-up of 2 years, 49% of patients relapsed, which was confirmed via endoscopy, fecal calprotectin, or CRP in 83% of cases and inferred from treatment escalation for clinical flare in 17%. Rates of relapse were comparable between patients with Crohn’s disease and ulcerative colitis or unclassified IBD and between those discontinuing adalimumab and those stopping infliximab.
 

Better healing, better outcomes

However, analysis showed that partial endoscopic healing was independently associated with a higher risk of relapse, at an adjusted hazard ratio versus complete endoscopic healing of 3.28.

 

 

At 12 months, 70% of patients with partial endoscopic healing had relapsed versus 35% of those with complete endoscopic healing.

Treatment with the anti-inflammatory agent mesalamine (multiple brands) was independently associated with a reduced risk of relapse, at an adjusted hazard ratio of 0.08. No other potential predictors of relapse were identified.

Of the patients who relapsed, 75% restarted anti-TNF treatment, and the majority (87%) were restarted on the same agent at a median of 0.9 years since its withdrawal and a median of 24 days since the onset of relapse.

Clinical remission was achieved at 3 months in 73% of patients who restarted anti-TNF therapy, which was found to restore quality of life and well-being in relapsed patients, the authors report.
 

Reluctance remains

Stephen B. Hanauer, MD, professor of medicine (gastroenterology and hepatology) at Northwestern University Feinberg School of Medicine, Chicago, said the findings “reinforce the benefits of the maintenance versus the withdrawal of therapy” and “the deeper the remission” the more likely it is to be sustained.

The 35% relapse rate at 12 months, even in patients with compete endoscopic healing, indicates that treatment should be maintained, Dr. Hanauer said.

“What is also relevant, but was not evaluated, is the additional endpoint of histologic healing, which is likely to sustain remissions even longer,” he added.

Nevertheless, Dr. Hanauer said, the “observed relapse rate is important to discuss in shared decision-making with patients.”

The findings are interesting, but the study didn’t follow the patients for long enough to understand why 35% of those with complete endoscopic healing relapsed, Miguel Regueiro, MD, chair of the Cleveland Clinic’s Digestive Disease & Surgery Institute, told this news organization.

“Are there predictors, factors, or other treatments that could be used to reduce that 35% risk of relapse further?” he questioned.

Although the study didn’t clear up that question for Dr. Regueiro, he found it compelling that mesalamine continuation resulted in higher rates of sustained remission after anti-TNF withdrawal among patients with ulcerative colitis.

Dr. Regueiro said that he will not begin recommending withdrawal of advanced therapies, including anti-TNF drugs, in patients who have achieved a stable remission.

“We have not yet found the cure for IBD, and my concern is that patients may relapse with more severe disease than previously and that recurrence of inflammation could have potential risks for complications,” he said.

“Nonetheless, this study is intriguing and important, and at least prompts the discussion of withdrawing therapy in those who have achieved a deep endoscopic remission for a sustained period of time,” Dr. Regueiro added.

The study received support from the Dutch Health Insurance Innovation Fund.

Dr. Oldenburg declares relationships with AbbVie, Celltrion, Ferring, Takeda, Galapagos, Pfizer, Cablon, PBMS, Janssen, and MSD. Other authors also declare numerous relationships. The full list can be found with the original article. Dr. Hanauer declares relationships with AbbVie, Janssen, Pfizer, and Boehringer Ingelheim. Dr. Regueiro declares relationships with AbbVie, Janssen, UCB, Takeda, Pfizer, Miraca Labs, Amgen, Celgene, Seres, Allergan, Genentech, Gilead, Salix, Prometheus, Lilly, TARGET Pharma Solutions, ALFASIGMA, S.p.A., BMS, CME Outfitters, Imedex, GI Health Foundation, Cornerstones, Remedy, MJH Life Sciences, Medscape, MDEducation, WebMD, and HMPGlobal.

Help your patients better understand their IBD treatment options by sharing AGA’s patient education, “Living with IBD,” in the AGA GI Patient Center at www.gastro.org/IBD. 

A version of this article first appeared on Medscape.com.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Ustekinumab has comparatively low infection rate in IBD

Discuss these with greater confidence
Article Type
Changed
Tue, 09/20/2022 - 16:49

Patients with inflammatory bowel disease (IBD) taking ustekinumab (Stelara) are less likely to have infections as a side effect than those taking tofacitinib (Xeljanz) or anti–tumor necrosis factor-alpha (anti-TNF), researchers say.

Although the risk of infections is not large for any of these drugs, clinicians should nonetheless take it into account, given their similar efficacy, said Ashwin Ananthakrishnan, MD, associate professor of medicine at Massachusetts General Hospital, Boston.

“These findings may not dramatically change prescribing patterns, but in those who may be particularly vulnerable for infection, they could point toward the safer biologic being Stelara,” he told this news organization.

The study by Dr. Ananthakrishnan and his colleagues was published in Clinical Gastroenterology and Hepatology.

Biologic and small-molecule immunosuppressive therapies have emerged as effective treatments for Crohn’s disease and ulcerative colitis, the two conditions that comprise IBD.

But the recent proliferation of drugs in these classes with different mechanisms of action has made prescribing decisions complicated. And because the drugs suppress the immune system, they can make patients vulnerable to infection.

Randomized controlled trials, even those comparing the drugs head to head, have not been large enough to compare safety outcomes with statistical accuracy, Dr. Ananthakrishnan and his colleagues found.
 

Accessing a large, real-world cohort

To help fill this gap, they analyzed data on 89,972,617 people enrolled in Aetna, the nationwide U.S. health insurance plan, from 2008-2019.

They identified 19,096 patients whose IBD was treated with anti-TNF agents, 2,420 with ustekinumab, and 305 with tofacitinib. The number of patients taking tofacitinib is small because it is a newer drug, Dr. Ananthakrishnan said.

They found a higher rate of infection and rate of infection-related hospitalization for the patients taking anti-TNF agents (44% and 7%, respectively), compared with those taking ustekinumab (32% and 4%, respectively) over the course of up to a year.

The researchers adjusted for age, sex, IBD duration, prior corticosteroid use, prior immunomodulator use, prior IBD hospitalization, and comorbidities, after which they then compared the risks of infection.

They found that patients taking ustekinumab were 7% less likely to have any infection than patients taking anti-TNF agents (hazard ratio, 0.93), a statistically significant difference (95% confidence interval, 0.86-0.99; P = .041). The reduction in the risk of infection-related hospitalizations was similar but not statistically significant.

The advantage with ustekinumab over anti-TNF agents was larger for patients with comorbidities. There was a 25% reduction in the risk of infections for patients taking ustekinumab who had a Charlson comorbidity index score of at least 2 and who were younger than 65 years (HRm 0.71; 95% CI, 0.58-0.87; P < .001).

For patients taking tofacitinib, there were no significant differences in the rate of infection (HR, 0.97; 95% CI, 0.75-1.24) or infection-related hospitalizations (HR, 0.59; 95% CI, 0.27-1.05), compared with TNF-antagonists.

The respiratory system and the urinary tract were the most common sites of infections. Bacterial, viral, and fungal infections were similarly distributed in the three groups.
 

Remaining questions

Further research is needed, said Dr. Ananthakrishnan, as “to understand the comparative safety may be particularly important for vulnerable populations, like those who are older or have other underlying comorbidity, where safety is increasingly an important issue.”

The findings are similar to those of other studies comparing infections in association with ustekinumab to anti-TNF medications for related conditions, such as psoriatic arthritis and psoriasis, he said.

Clinicians have seen similar differences among the drugs in clinical practice, said Miguel Regueiro, MD, chair of the Digestive Disease and Surgery Institute at Cleveland Clinic, Ohio, who was not involved in the study.

“It aligns well with what we’ve thought, but it’s nice to see in a publication,” he said in an interview.

The implications of the study are limited, because it was not a prospective randomized trial and because the number of patients taking tofacitinib was so small, Dr. Regueiro added.

Another limitation is that the patients who were admitted to the hospital in this database were not necessarily admitted because of their infections, said Stephen Hanauer, MD, medical director of the Digestive Health Center at Northwestern University, Chicago, who also was not involved in the study.

Dr. Hanauer told this news organization that comparisons with other agents would be helpful.

“They didn’t look at vedolizumab (Entyvio) in this database,” he said. “Entyvio is generally considered to be safer than TNF inhibitors or tofacitinib with fairly comparable safety to ustekinumab.”

Dr. Ananthakrishnan reported financial relationships with Gilead, Ikena Therapeutics, and Sun Pharma. Dr. Regueiro reported financial relationships with AbbVie, BMS, Janssen, UCB, Pfizer, Takeda, Celgene, Genentech, Gilead, UCB, Miraca Labs, Amgen, Celgene, Seres, Allergan, Salix, Prometheus, Lilly, TARGET Pharma Solutions, Alfasigigma, and BMS. Dr. Hanauer reported financial relationships with Janssen Pharmaceuticals, AbbVie, Takeda Pharmaceutical, and Pfizer.

A version of this article first appeared on Medscape.com.

Body

With our growing armamentarium of effective medical therapies for Crohn’s disease (CD) and ulcerative colitis (UC) come increasing decisions for patients and providers for treatments based on comparative effectiveness and safety. One of the most frequent concerns by both patients and providers is risk of infection. While we have partial data on safety from clinical trials, trials are underpowered to compare safety outcomes and also typically compare placebo rather than other active treatments.

This study by Cheng et al. provides further reassurance that ustekinumab and tofacitinib are at least as safe from an infection standpoint as TNF inhibitors, with UST having a small, but statistically significant lower risk of infection overall compared to TNF inhibitors. While there have been signals that ustekinumab may have lower infection risk compared to TNF inhibitors from clinical trials and real-world analyses of ustekinumab in other disease states, this study is remarkable in that it studies CD and UC specifically. Ustekinumab dosing for CD and UC are higher than that used for other indications, so it’s highly relevant to study ustekinumab in CD and UC, specifically for safety. This study is also notable in that no statistically significant difference in infection, particularly herpes zoster, was observed in tofacitinib vs. TNF inhibitors.

This study has limitations as a retrospective administrative dataset, including its inability to determine indication of prescription for CD or UC vs. rheumatologic or dermatologic condition, lack of adjustment for concomitant immunomodulator use, and inability to determine primary indication for hospitalization. However, this study should allow providers to discuss with patients with greater confidence that infection risks of ustekinumab and tofacitinib were overall low and that ustekinumab has lower risks of infections than TNF inhibitors.
 

Jason K. Hou, MD, MS, is an investigator in the clinical epidemiology and outcomes program in the Center for Innovations in Quality, Effectiveness and Safety at the Michael E. DeBakey VA Medical Center and an associate professor at the Baylor College of Medicine, both in Houston. He has no relevant conflicts of interest.

Publications
Topics
Sections
Body

With our growing armamentarium of effective medical therapies for Crohn’s disease (CD) and ulcerative colitis (UC) come increasing decisions for patients and providers for treatments based on comparative effectiveness and safety. One of the most frequent concerns by both patients and providers is risk of infection. While we have partial data on safety from clinical trials, trials are underpowered to compare safety outcomes and also typically compare placebo rather than other active treatments.

This study by Cheng et al. provides further reassurance that ustekinumab and tofacitinib are at least as safe from an infection standpoint as TNF inhibitors, with UST having a small, but statistically significant lower risk of infection overall compared to TNF inhibitors. While there have been signals that ustekinumab may have lower infection risk compared to TNF inhibitors from clinical trials and real-world analyses of ustekinumab in other disease states, this study is remarkable in that it studies CD and UC specifically. Ustekinumab dosing for CD and UC are higher than that used for other indications, so it’s highly relevant to study ustekinumab in CD and UC, specifically for safety. This study is also notable in that no statistically significant difference in infection, particularly herpes zoster, was observed in tofacitinib vs. TNF inhibitors.

This study has limitations as a retrospective administrative dataset, including its inability to determine indication of prescription for CD or UC vs. rheumatologic or dermatologic condition, lack of adjustment for concomitant immunomodulator use, and inability to determine primary indication for hospitalization. However, this study should allow providers to discuss with patients with greater confidence that infection risks of ustekinumab and tofacitinib were overall low and that ustekinumab has lower risks of infections than TNF inhibitors.
 

Jason K. Hou, MD, MS, is an investigator in the clinical epidemiology and outcomes program in the Center for Innovations in Quality, Effectiveness and Safety at the Michael E. DeBakey VA Medical Center and an associate professor at the Baylor College of Medicine, both in Houston. He has no relevant conflicts of interest.

Body

With our growing armamentarium of effective medical therapies for Crohn’s disease (CD) and ulcerative colitis (UC) come increasing decisions for patients and providers for treatments based on comparative effectiveness and safety. One of the most frequent concerns by both patients and providers is risk of infection. While we have partial data on safety from clinical trials, trials are underpowered to compare safety outcomes and also typically compare placebo rather than other active treatments.

This study by Cheng et al. provides further reassurance that ustekinumab and tofacitinib are at least as safe from an infection standpoint as TNF inhibitors, with UST having a small, but statistically significant lower risk of infection overall compared to TNF inhibitors. While there have been signals that ustekinumab may have lower infection risk compared to TNF inhibitors from clinical trials and real-world analyses of ustekinumab in other disease states, this study is remarkable in that it studies CD and UC specifically. Ustekinumab dosing for CD and UC are higher than that used for other indications, so it’s highly relevant to study ustekinumab in CD and UC, specifically for safety. This study is also notable in that no statistically significant difference in infection, particularly herpes zoster, was observed in tofacitinib vs. TNF inhibitors.

This study has limitations as a retrospective administrative dataset, including its inability to determine indication of prescription for CD or UC vs. rheumatologic or dermatologic condition, lack of adjustment for concomitant immunomodulator use, and inability to determine primary indication for hospitalization. However, this study should allow providers to discuss with patients with greater confidence that infection risks of ustekinumab and tofacitinib were overall low and that ustekinumab has lower risks of infections than TNF inhibitors.
 

Jason K. Hou, MD, MS, is an investigator in the clinical epidemiology and outcomes program in the Center for Innovations in Quality, Effectiveness and Safety at the Michael E. DeBakey VA Medical Center and an associate professor at the Baylor College of Medicine, both in Houston. He has no relevant conflicts of interest.

Title
Discuss these with greater confidence
Discuss these with greater confidence

Patients with inflammatory bowel disease (IBD) taking ustekinumab (Stelara) are less likely to have infections as a side effect than those taking tofacitinib (Xeljanz) or anti–tumor necrosis factor-alpha (anti-TNF), researchers say.

Although the risk of infections is not large for any of these drugs, clinicians should nonetheless take it into account, given their similar efficacy, said Ashwin Ananthakrishnan, MD, associate professor of medicine at Massachusetts General Hospital, Boston.

“These findings may not dramatically change prescribing patterns, but in those who may be particularly vulnerable for infection, they could point toward the safer biologic being Stelara,” he told this news organization.

The study by Dr. Ananthakrishnan and his colleagues was published in Clinical Gastroenterology and Hepatology.

Biologic and small-molecule immunosuppressive therapies have emerged as effective treatments for Crohn’s disease and ulcerative colitis, the two conditions that comprise IBD.

But the recent proliferation of drugs in these classes with different mechanisms of action has made prescribing decisions complicated. And because the drugs suppress the immune system, they can make patients vulnerable to infection.

Randomized controlled trials, even those comparing the drugs head to head, have not been large enough to compare safety outcomes with statistical accuracy, Dr. Ananthakrishnan and his colleagues found.
 

Accessing a large, real-world cohort

To help fill this gap, they analyzed data on 89,972,617 people enrolled in Aetna, the nationwide U.S. health insurance plan, from 2008-2019.

They identified 19,096 patients whose IBD was treated with anti-TNF agents, 2,420 with ustekinumab, and 305 with tofacitinib. The number of patients taking tofacitinib is small because it is a newer drug, Dr. Ananthakrishnan said.

They found a higher rate of infection and rate of infection-related hospitalization for the patients taking anti-TNF agents (44% and 7%, respectively), compared with those taking ustekinumab (32% and 4%, respectively) over the course of up to a year.

The researchers adjusted for age, sex, IBD duration, prior corticosteroid use, prior immunomodulator use, prior IBD hospitalization, and comorbidities, after which they then compared the risks of infection.

They found that patients taking ustekinumab were 7% less likely to have any infection than patients taking anti-TNF agents (hazard ratio, 0.93), a statistically significant difference (95% confidence interval, 0.86-0.99; P = .041). The reduction in the risk of infection-related hospitalizations was similar but not statistically significant.

The advantage with ustekinumab over anti-TNF agents was larger for patients with comorbidities. There was a 25% reduction in the risk of infections for patients taking ustekinumab who had a Charlson comorbidity index score of at least 2 and who were younger than 65 years (HRm 0.71; 95% CI, 0.58-0.87; P < .001).

For patients taking tofacitinib, there were no significant differences in the rate of infection (HR, 0.97; 95% CI, 0.75-1.24) or infection-related hospitalizations (HR, 0.59; 95% CI, 0.27-1.05), compared with TNF-antagonists.

The respiratory system and the urinary tract were the most common sites of infections. Bacterial, viral, and fungal infections were similarly distributed in the three groups.
 

Remaining questions

Further research is needed, said Dr. Ananthakrishnan, as “to understand the comparative safety may be particularly important for vulnerable populations, like those who are older or have other underlying comorbidity, where safety is increasingly an important issue.”

The findings are similar to those of other studies comparing infections in association with ustekinumab to anti-TNF medications for related conditions, such as psoriatic arthritis and psoriasis, he said.

Clinicians have seen similar differences among the drugs in clinical practice, said Miguel Regueiro, MD, chair of the Digestive Disease and Surgery Institute at Cleveland Clinic, Ohio, who was not involved in the study.

“It aligns well with what we’ve thought, but it’s nice to see in a publication,” he said in an interview.

The implications of the study are limited, because it was not a prospective randomized trial and because the number of patients taking tofacitinib was so small, Dr. Regueiro added.

Another limitation is that the patients who were admitted to the hospital in this database were not necessarily admitted because of their infections, said Stephen Hanauer, MD, medical director of the Digestive Health Center at Northwestern University, Chicago, who also was not involved in the study.

Dr. Hanauer told this news organization that comparisons with other agents would be helpful.

“They didn’t look at vedolizumab (Entyvio) in this database,” he said. “Entyvio is generally considered to be safer than TNF inhibitors or tofacitinib with fairly comparable safety to ustekinumab.”

Dr. Ananthakrishnan reported financial relationships with Gilead, Ikena Therapeutics, and Sun Pharma. Dr. Regueiro reported financial relationships with AbbVie, BMS, Janssen, UCB, Pfizer, Takeda, Celgene, Genentech, Gilead, UCB, Miraca Labs, Amgen, Celgene, Seres, Allergan, Salix, Prometheus, Lilly, TARGET Pharma Solutions, Alfasigigma, and BMS. Dr. Hanauer reported financial relationships with Janssen Pharmaceuticals, AbbVie, Takeda Pharmaceutical, and Pfizer.

A version of this article first appeared on Medscape.com.

Patients with inflammatory bowel disease (IBD) taking ustekinumab (Stelara) are less likely to have infections as a side effect than those taking tofacitinib (Xeljanz) or anti–tumor necrosis factor-alpha (anti-TNF), researchers say.

Although the risk of infections is not large for any of these drugs, clinicians should nonetheless take it into account, given their similar efficacy, said Ashwin Ananthakrishnan, MD, associate professor of medicine at Massachusetts General Hospital, Boston.

“These findings may not dramatically change prescribing patterns, but in those who may be particularly vulnerable for infection, they could point toward the safer biologic being Stelara,” he told this news organization.

The study by Dr. Ananthakrishnan and his colleagues was published in Clinical Gastroenterology and Hepatology.

Biologic and small-molecule immunosuppressive therapies have emerged as effective treatments for Crohn’s disease and ulcerative colitis, the two conditions that comprise IBD.

But the recent proliferation of drugs in these classes with different mechanisms of action has made prescribing decisions complicated. And because the drugs suppress the immune system, they can make patients vulnerable to infection.

Randomized controlled trials, even those comparing the drugs head to head, have not been large enough to compare safety outcomes with statistical accuracy, Dr. Ananthakrishnan and his colleagues found.
 

Accessing a large, real-world cohort

To help fill this gap, they analyzed data on 89,972,617 people enrolled in Aetna, the nationwide U.S. health insurance plan, from 2008-2019.

They identified 19,096 patients whose IBD was treated with anti-TNF agents, 2,420 with ustekinumab, and 305 with tofacitinib. The number of patients taking tofacitinib is small because it is a newer drug, Dr. Ananthakrishnan said.

They found a higher rate of infection and rate of infection-related hospitalization for the patients taking anti-TNF agents (44% and 7%, respectively), compared with those taking ustekinumab (32% and 4%, respectively) over the course of up to a year.

The researchers adjusted for age, sex, IBD duration, prior corticosteroid use, prior immunomodulator use, prior IBD hospitalization, and comorbidities, after which they then compared the risks of infection.

They found that patients taking ustekinumab were 7% less likely to have any infection than patients taking anti-TNF agents (hazard ratio, 0.93), a statistically significant difference (95% confidence interval, 0.86-0.99; P = .041). The reduction in the risk of infection-related hospitalizations was similar but not statistically significant.

The advantage with ustekinumab over anti-TNF agents was larger for patients with comorbidities. There was a 25% reduction in the risk of infections for patients taking ustekinumab who had a Charlson comorbidity index score of at least 2 and who were younger than 65 years (HRm 0.71; 95% CI, 0.58-0.87; P < .001).

For patients taking tofacitinib, there were no significant differences in the rate of infection (HR, 0.97; 95% CI, 0.75-1.24) or infection-related hospitalizations (HR, 0.59; 95% CI, 0.27-1.05), compared with TNF-antagonists.

The respiratory system and the urinary tract were the most common sites of infections. Bacterial, viral, and fungal infections were similarly distributed in the three groups.
 

Remaining questions

Further research is needed, said Dr. Ananthakrishnan, as “to understand the comparative safety may be particularly important for vulnerable populations, like those who are older or have other underlying comorbidity, where safety is increasingly an important issue.”

The findings are similar to those of other studies comparing infections in association with ustekinumab to anti-TNF medications for related conditions, such as psoriatic arthritis and psoriasis, he said.

Clinicians have seen similar differences among the drugs in clinical practice, said Miguel Regueiro, MD, chair of the Digestive Disease and Surgery Institute at Cleveland Clinic, Ohio, who was not involved in the study.

“It aligns well with what we’ve thought, but it’s nice to see in a publication,” he said in an interview.

The implications of the study are limited, because it was not a prospective randomized trial and because the number of patients taking tofacitinib was so small, Dr. Regueiro added.

Another limitation is that the patients who were admitted to the hospital in this database were not necessarily admitted because of their infections, said Stephen Hanauer, MD, medical director of the Digestive Health Center at Northwestern University, Chicago, who also was not involved in the study.

Dr. Hanauer told this news organization that comparisons with other agents would be helpful.

“They didn’t look at vedolizumab (Entyvio) in this database,” he said. “Entyvio is generally considered to be safer than TNF inhibitors or tofacitinib with fairly comparable safety to ustekinumab.”

Dr. Ananthakrishnan reported financial relationships with Gilead, Ikena Therapeutics, and Sun Pharma. Dr. Regueiro reported financial relationships with AbbVie, BMS, Janssen, UCB, Pfizer, Takeda, Celgene, Genentech, Gilead, UCB, Miraca Labs, Amgen, Celgene, Seres, Allergan, Salix, Prometheus, Lilly, TARGET Pharma Solutions, Alfasigigma, and BMS. Dr. Hanauer reported financial relationships with Janssen Pharmaceuticals, AbbVie, Takeda Pharmaceutical, and Pfizer.

A version of this article first appeared on Medscape.com.

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM CLINICAL GASTROENTEROLOGY AND HEPATOLOGY

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Usefulness of IBD biomarker may vary based on microbiome

Looking beyond endoscopic evaluation
Article Type
Changed
Tue, 09/20/2022 - 16:48

Levels of calprotectin, a biomarker used to detect intestinal inflammation, may vary in fecal samples based on an individual’s microbiome composition, according to researchers. The results, if confirmed, might help refine its use in monitoring inflammatory bowel disease (IBD).

Researchers used a new ex vivo functional assay to identify specific bacteria that degrade calprotectin and may play a role in variations found in vivo. “Microbiome-based calibration could improve sensitivity and specificity of fecal calprotectin readouts, thereby facilitating more reliable real-time monitoring and ultimately enabling more timely interventions,” the authors wrote in their research letter, which was published in Gastro Hep Advances.

The standard for diagnosing ulcerative colitis (UC) and Crohn’s disease (CD) is endoscopy and biopsy because it allows both visual and histological examination of the severity and extent of intestinal inflammation, but this cannot be used to monitor patients on an ongoing basis.

Calprotectin is a promising biomarker for intestinal inflammation, but a meta-analysis found that it has a pooled sensitivity of 85% and a pooled specificity of 75% for the diagnosis of endoscopically active inflammatory bowel disease.

The researchers investigated whether an individual’s microbiome can metabolize calprotectin, which would complicate measurement of fecal calprotectin. They recruited 22 individuals with IBD (64% female, 73% with colonic disease), who provided stool samples. They completed a symptom questionnaire in advance of a colonoscopy. Overall, 64% had endoscopically inactive disease, and 82% had clinically inactive disease.

At a cutoff of 50 mcg/g, 9 patients had normal fecal calprotectin levels, and 13 had elevated levels. Those with clinically or endoscopically active disease had higher levels of fecal calprotectin (P < .0001).

There was a significant but poor correlation between disease activity measures and fecal calprotectin levels in CD (r = 0.62; P = .008), but there was no statistically significant association for UC (r = –0.29; P = .6). Endoscopic disease activity was also significantly correlated with fecal calprotectin in CD (r = 0.83; P < .001), but not UC (r = 0.50; P = .4).

The researchers created an ex vivo functional assay to measure calprotectin metabolism by the microbiome. They anaerobically cultured fecal samples in the presence of calprotectin and measured levels of calprotectin after 24 hours. Control samples were grown without microbes, without calprotectin, or lacking both. The researchers tested samples in both standard media and in media with low levels of amino acids, reasoning that the latter condition might encourage catabolism of calprotectin. The cultures with low amino acid content had lower calprotectin levels than those with normal amino acid content (P < .0007).

The researchers found greater calprotectin degradation in the low amino acid media, and the difference was more pronounced among samples taken from individuals with UC than CD (P < .02).

They used metagenomic sequencing data from fecal samples to identify bacterial species associated with calprotectin metabolism. Similarly to previous reports, the researchers found that Firmicutes was dominant, while Subdoligranulum correlated with calprotectin degradation in low amino acid media (P = .04).

For 5 days, they cultured Subdoligranulum variabile in low amino acid media that also contained calprotectin. Calprotectin levels were lower than a control sample with cultured Akkermansia muciniphila, which was previously shown to not be associated with calprotectin degradation in low amino acid media (P = .03). Because Subdoligranulum species were not detectable in 5 of 22 fecal samples, the authors say they are unlikely to be the only species capable of metabolizing calprotectin.

Among IBD patients, only one had both endoscopically active colitis and a low fecal calprotectin level. The patient’s micobiome had Subdoligranulum present, and their fecal sample was able to metabolize calprotectin in the functional assay.

The study was limited by its small sample size, which prevented development of a calibration model for fecal calprotectin, and the researchers called for additional studies among individuals with active colitis.

Body

The search continues for reliable, noninvasive methods for monitoring disease activity in inflammatory bowel disease (IBD). Noninvasive disease activity measures improve quality of care by facilitating more frequent assessment of therapeutic efficacy, which for IBD otherwise depends on periodic endoscopic evaluation and biopsy. Available tools such as fecal calprotectin are valuable and widely used but are imperfect.

Dr. Deborah C. Rubin
In this report by Kamp et al., the authors provide a potential explanation for variation in fecal calprotectin levels. They make the novel observation that calprotectin can be metabolized by components of the patient’s fecal microbiome. The authors generated an ex-vivo functional assay in which they anaerobically cultured fecal samples with or without calprotectin, and showed that Subdoligranulum species (Subdoligranulum variabile) when cultured in low–amino acid media, degraded calprotectin, yet another microbiome component, Akkermansia muciniphilia, did not. Subdoligranulum species were isolated from 17 of 22 patient microbiomes.

Of note, a patient with endoscopically active colitis and a relatively low fecal calprotectin level harbored Subdoligranulum species, which – when isolated and assayed ex vivo – degraded calprotectin. These studies suggest that individualized, patient microbiome–based calibration assays might help improve the sensitivity and specificity of fecal calprotectin levels for monitoring disease activity. As the authors note, more patients need to be studied, especially focusing on those with active disease and paradoxically low calprotectin levels.
 

Deborah C. Rubin, MD, AGAF, is the William B. Kountz Professor of Medicine and professor of developmental biology in the division of gastroenterology at Washington University, St. Louis. She had no conflicts of interest to disclose.

Publications
Topics
Sections
Body

The search continues for reliable, noninvasive methods for monitoring disease activity in inflammatory bowel disease (IBD). Noninvasive disease activity measures improve quality of care by facilitating more frequent assessment of therapeutic efficacy, which for IBD otherwise depends on periodic endoscopic evaluation and biopsy. Available tools such as fecal calprotectin are valuable and widely used but are imperfect.

Dr. Deborah C. Rubin
In this report by Kamp et al., the authors provide a potential explanation for variation in fecal calprotectin levels. They make the novel observation that calprotectin can be metabolized by components of the patient’s fecal microbiome. The authors generated an ex-vivo functional assay in which they anaerobically cultured fecal samples with or without calprotectin, and showed that Subdoligranulum species (Subdoligranulum variabile) when cultured in low–amino acid media, degraded calprotectin, yet another microbiome component, Akkermansia muciniphilia, did not. Subdoligranulum species were isolated from 17 of 22 patient microbiomes.

Of note, a patient with endoscopically active colitis and a relatively low fecal calprotectin level harbored Subdoligranulum species, which – when isolated and assayed ex vivo – degraded calprotectin. These studies suggest that individualized, patient microbiome–based calibration assays might help improve the sensitivity and specificity of fecal calprotectin levels for monitoring disease activity. As the authors note, more patients need to be studied, especially focusing on those with active disease and paradoxically low calprotectin levels.
 

Deborah C. Rubin, MD, AGAF, is the William B. Kountz Professor of Medicine and professor of developmental biology in the division of gastroenterology at Washington University, St. Louis. She had no conflicts of interest to disclose.

Body

The search continues for reliable, noninvasive methods for monitoring disease activity in inflammatory bowel disease (IBD). Noninvasive disease activity measures improve quality of care by facilitating more frequent assessment of therapeutic efficacy, which for IBD otherwise depends on periodic endoscopic evaluation and biopsy. Available tools such as fecal calprotectin are valuable and widely used but are imperfect.

Dr. Deborah C. Rubin
In this report by Kamp et al., the authors provide a potential explanation for variation in fecal calprotectin levels. They make the novel observation that calprotectin can be metabolized by components of the patient’s fecal microbiome. The authors generated an ex-vivo functional assay in which they anaerobically cultured fecal samples with or without calprotectin, and showed that Subdoligranulum species (Subdoligranulum variabile) when cultured in low–amino acid media, degraded calprotectin, yet another microbiome component, Akkermansia muciniphilia, did not. Subdoligranulum species were isolated from 17 of 22 patient microbiomes.

Of note, a patient with endoscopically active colitis and a relatively low fecal calprotectin level harbored Subdoligranulum species, which – when isolated and assayed ex vivo – degraded calprotectin. These studies suggest that individualized, patient microbiome–based calibration assays might help improve the sensitivity and specificity of fecal calprotectin levels for monitoring disease activity. As the authors note, more patients need to be studied, especially focusing on those with active disease and paradoxically low calprotectin levels.
 

Deborah C. Rubin, MD, AGAF, is the William B. Kountz Professor of Medicine and professor of developmental biology in the division of gastroenterology at Washington University, St. Louis. She had no conflicts of interest to disclose.

Title
Looking beyond endoscopic evaluation
Looking beyond endoscopic evaluation

Levels of calprotectin, a biomarker used to detect intestinal inflammation, may vary in fecal samples based on an individual’s microbiome composition, according to researchers. The results, if confirmed, might help refine its use in monitoring inflammatory bowel disease (IBD).

Researchers used a new ex vivo functional assay to identify specific bacteria that degrade calprotectin and may play a role in variations found in vivo. “Microbiome-based calibration could improve sensitivity and specificity of fecal calprotectin readouts, thereby facilitating more reliable real-time monitoring and ultimately enabling more timely interventions,” the authors wrote in their research letter, which was published in Gastro Hep Advances.

The standard for diagnosing ulcerative colitis (UC) and Crohn’s disease (CD) is endoscopy and biopsy because it allows both visual and histological examination of the severity and extent of intestinal inflammation, but this cannot be used to monitor patients on an ongoing basis.

Calprotectin is a promising biomarker for intestinal inflammation, but a meta-analysis found that it has a pooled sensitivity of 85% and a pooled specificity of 75% for the diagnosis of endoscopically active inflammatory bowel disease.

The researchers investigated whether an individual’s microbiome can metabolize calprotectin, which would complicate measurement of fecal calprotectin. They recruited 22 individuals with IBD (64% female, 73% with colonic disease), who provided stool samples. They completed a symptom questionnaire in advance of a colonoscopy. Overall, 64% had endoscopically inactive disease, and 82% had clinically inactive disease.

At a cutoff of 50 mcg/g, 9 patients had normal fecal calprotectin levels, and 13 had elevated levels. Those with clinically or endoscopically active disease had higher levels of fecal calprotectin (P < .0001).

There was a significant but poor correlation between disease activity measures and fecal calprotectin levels in CD (r = 0.62; P = .008), but there was no statistically significant association for UC (r = –0.29; P = .6). Endoscopic disease activity was also significantly correlated with fecal calprotectin in CD (r = 0.83; P < .001), but not UC (r = 0.50; P = .4).

The researchers created an ex vivo functional assay to measure calprotectin metabolism by the microbiome. They anaerobically cultured fecal samples in the presence of calprotectin and measured levels of calprotectin after 24 hours. Control samples were grown without microbes, without calprotectin, or lacking both. The researchers tested samples in both standard media and in media with low levels of amino acids, reasoning that the latter condition might encourage catabolism of calprotectin. The cultures with low amino acid content had lower calprotectin levels than those with normal amino acid content (P < .0007).

The researchers found greater calprotectin degradation in the low amino acid media, and the difference was more pronounced among samples taken from individuals with UC than CD (P < .02).

They used metagenomic sequencing data from fecal samples to identify bacterial species associated with calprotectin metabolism. Similarly to previous reports, the researchers found that Firmicutes was dominant, while Subdoligranulum correlated with calprotectin degradation in low amino acid media (P = .04).

For 5 days, they cultured Subdoligranulum variabile in low amino acid media that also contained calprotectin. Calprotectin levels were lower than a control sample with cultured Akkermansia muciniphila, which was previously shown to not be associated with calprotectin degradation in low amino acid media (P = .03). Because Subdoligranulum species were not detectable in 5 of 22 fecal samples, the authors say they are unlikely to be the only species capable of metabolizing calprotectin.

Among IBD patients, only one had both endoscopically active colitis and a low fecal calprotectin level. The patient’s micobiome had Subdoligranulum present, and their fecal sample was able to metabolize calprotectin in the functional assay.

The study was limited by its small sample size, which prevented development of a calibration model for fecal calprotectin, and the researchers called for additional studies among individuals with active colitis.

Levels of calprotectin, a biomarker used to detect intestinal inflammation, may vary in fecal samples based on an individual’s microbiome composition, according to researchers. The results, if confirmed, might help refine its use in monitoring inflammatory bowel disease (IBD).

Researchers used a new ex vivo functional assay to identify specific bacteria that degrade calprotectin and may play a role in variations found in vivo. “Microbiome-based calibration could improve sensitivity and specificity of fecal calprotectin readouts, thereby facilitating more reliable real-time monitoring and ultimately enabling more timely interventions,” the authors wrote in their research letter, which was published in Gastro Hep Advances.

The standard for diagnosing ulcerative colitis (UC) and Crohn’s disease (CD) is endoscopy and biopsy because it allows both visual and histological examination of the severity and extent of intestinal inflammation, but this cannot be used to monitor patients on an ongoing basis.

Calprotectin is a promising biomarker for intestinal inflammation, but a meta-analysis found that it has a pooled sensitivity of 85% and a pooled specificity of 75% for the diagnosis of endoscopically active inflammatory bowel disease.

The researchers investigated whether an individual’s microbiome can metabolize calprotectin, which would complicate measurement of fecal calprotectin. They recruited 22 individuals with IBD (64% female, 73% with colonic disease), who provided stool samples. They completed a symptom questionnaire in advance of a colonoscopy. Overall, 64% had endoscopically inactive disease, and 82% had clinically inactive disease.

At a cutoff of 50 mcg/g, 9 patients had normal fecal calprotectin levels, and 13 had elevated levels. Those with clinically or endoscopically active disease had higher levels of fecal calprotectin (P < .0001).

There was a significant but poor correlation between disease activity measures and fecal calprotectin levels in CD (r = 0.62; P = .008), but there was no statistically significant association for UC (r = –0.29; P = .6). Endoscopic disease activity was also significantly correlated with fecal calprotectin in CD (r = 0.83; P < .001), but not UC (r = 0.50; P = .4).

The researchers created an ex vivo functional assay to measure calprotectin metabolism by the microbiome. They anaerobically cultured fecal samples in the presence of calprotectin and measured levels of calprotectin after 24 hours. Control samples were grown without microbes, without calprotectin, or lacking both. The researchers tested samples in both standard media and in media with low levels of amino acids, reasoning that the latter condition might encourage catabolism of calprotectin. The cultures with low amino acid content had lower calprotectin levels than those with normal amino acid content (P < .0007).

The researchers found greater calprotectin degradation in the low amino acid media, and the difference was more pronounced among samples taken from individuals with UC than CD (P < .02).

They used metagenomic sequencing data from fecal samples to identify bacterial species associated with calprotectin metabolism. Similarly to previous reports, the researchers found that Firmicutes was dominant, while Subdoligranulum correlated with calprotectin degradation in low amino acid media (P = .04).

For 5 days, they cultured Subdoligranulum variabile in low amino acid media that also contained calprotectin. Calprotectin levels were lower than a control sample with cultured Akkermansia muciniphila, which was previously shown to not be associated with calprotectin degradation in low amino acid media (P = .03). Because Subdoligranulum species were not detectable in 5 of 22 fecal samples, the authors say they are unlikely to be the only species capable of metabolizing calprotectin.

Among IBD patients, only one had both endoscopically active colitis and a low fecal calprotectin level. The patient’s micobiome had Subdoligranulum present, and their fecal sample was able to metabolize calprotectin in the functional assay.

The study was limited by its small sample size, which prevented development of a calibration model for fecal calprotectin, and the researchers called for additional studies among individuals with active colitis.

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM GASTRO HEP ADVANCES

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Inhibiting adenosine pathways may be key to CRC treatment

​​​​​​​A conceptual advance toward combinatorial therapy
Article Type
Changed
Mon, 09/12/2022 - 14:56

Research that could set the stage for future clinical trials explored the potential role of adenosine signaling in altering the immune microenvironment of colorectal cancer (CRC), according to a study appearing in Cellular and Molecular Gastroenterology and Hepatology.

The study in human-derived cells and CRC mouse models suggests that addition of a CD73 inhibitor to the CDK4/6 inhibitor palbociclib may hold promise in the treatment of CRC because CD73 can produce extracellular adenosine. The results hold promise for future research since anticancer therapies often prompt increased expression of PD-L1, and CDK4/6 inhibitor monotherapy can fail because of accumulation of PD-L1 after such treatment.

Previous research has suggested that extracellular adenosine-mediated signaling can lead to accumulation of tumor-associated macrophages (TAMs) and an immunosuppressive tumor microenvironment. TAMs play an important role in the intestinal mucosal immune system in CRC, which mediates tumor-promoting metabolites in the intestine and inflammatory pathways that can lead to and progress CRC. TAMs are also linked to increases in extracellular enzymes like CD39 and CD73 as well as resistance to chemotherapy and anti-PD-1/PD-L1 therapy in CRC. Furthermore, they express PD-L1 and they promote other immunosuppressive molecules.

In normal tissues, CD73 produces adenosine to tamp down excessive immune responses. Some tumors express CD73 or even induce expression in normal cells, leading to immunosuppression in the tumor microenvironment. Previous studies have shown that CD73 expression is a biomarker for poor outcomes in gastric, liver, pancreatic, and colorectal cancer, the authors of this study noted.

To better understand the impact of adenosine, the researchers exposed human macrophages derived from peripheral blood to adenosine, and then analyzed the results using flow cytometry and Western blot. They used RNA sequencing and proteomics to discern changes in the cells that resulted from the exposure.

Adenosine treatment led to changes in the expression levels of genes involved in the cell cycle, cell division, cell cycle phase transition, and DNA repair. The researchers emphasized that extracellular treatment with adenosine led to a reduction in expression of the cell cycle–related gene CCND1, which encodes cyclin D1. Among three genes in the cyclin D family tested, CCND1 was the only one affected by adenosine. Cyclin D1 protein levels also went down.

Cyclin D1 is a known actor in regulating the cell cycle and tumorigenesis, among other roles. Previous reports indicated that cyclin D1 participates in posttranslational regulation of PD-L1, and the current study suggests it plays a similar role in TAMs after exposure to adenosine in the tumor microenvironment. Myeloid cells high in cyclin D1 expression had low levels or even an absence of the immunosuppressive molecule CD39. “Taken together, cyclin D1 may be one of the major orchestrators that trigger the differentiation of pro-tumorigenic TAMs. Our findings suggest a novel immune checkpoint regulatory mechanism of extracellular adenosine signaling, which might be related to the cell cycle of macrophages,” the authors wrote.

The researchers then introduced a short hairpin RNA directed against CCND1. This led to increases in PD-L1 protein levels. Simultaneous treatment with adenosine led to a slight increase in levels of the PD-L1 protein, which suggests that reduction of CCND1 levels is the primary cause of increased PD-L1, according to the authors.

The CD73 inhibitor AB680, currently in phase 1 clinical trials for castration-resistant prostate cancer and advanced pancreatic cancer and developed by Arcus Biosciences, led to reduced PD-L1 levels in both human and mouse macrophages, and AB680 combined with the CDK4/6 inhibitor palbociclib led to greater inhibition of tumor growth than palbociclib alone in CRC mouse models.

“Thus, the promising effects of CD73 inhibitors might breathe new life for those old drugs and provide potent therapeutic strategies. Given that the therapeutic effects of PD-1/PD-L1 immunotherapy have not been conclusively demonstrated in patients with CRC, our observations should support clinical trials of new combinational therapies for CRC,” the authors wrote.

The authors disclose no conflicts.
 

Body

Although cancer immunotherapy has emerged as a powerful treatment modality, its application to colorectal cancer (CRC) is presently restricted to the minor subclass of tumors exhibiting deficient mismatch repair and high microsatellite instability. Programmed death 1 and its ligand, PD-L1, are molecules that typically suppress tumor-killing lymphocytes, yet immune checkpoint inhibitors targeting PD-1 or PD-L1 have limited to no effectiveness in the major form of proficient mismatch repair CRC. Better definition of the immune microenvironment of the different forms of CRC could lead to new treatment regimens.
 

Copyright Bogdan Condor
Dr. Cambrian Y. Liu
Recent work published by Chang Hoon Lee’s group in Cellular and Molecular Gastroenterology and Hepatology illustrates the central role of tumor-associated macrophages in suppressing the immune response to CRC. These macrophages possess biosynthetic enzymes that produce high levels of adenosine. The newly generated adenosine, in turn, alters the cellular protein processing machinery and enhances the expression of PD-L1. Importantly, as treatment of CRC with cell cycle inhibitors such as palbociclib increases expression of PD-L1 and implicates a mechanism of tumor resistance, the new work shows that combined therapy to block adenosine production (with AB680) during palbociclib treatment improves tumor outcomes in mouse models.

This study offers a conceptual advance towards combinatorial therapy that can simultaneously inhibit tumor cell proliferation and activate immune surveillance, providing clear and testable hypotheses for the clinic.
 

Cambrian Y. Liu, PhD, is research assistant professor in the section of gastroenterology, hepatology, and nutrition in the department of medicine, University of Chicago. He has no relevant conflicts of interest.

Publications
Topics
Sections
Body

Although cancer immunotherapy has emerged as a powerful treatment modality, its application to colorectal cancer (CRC) is presently restricted to the minor subclass of tumors exhibiting deficient mismatch repair and high microsatellite instability. Programmed death 1 and its ligand, PD-L1, are molecules that typically suppress tumor-killing lymphocytes, yet immune checkpoint inhibitors targeting PD-1 or PD-L1 have limited to no effectiveness in the major form of proficient mismatch repair CRC. Better definition of the immune microenvironment of the different forms of CRC could lead to new treatment regimens.
 

Copyright Bogdan Condor
Dr. Cambrian Y. Liu
Recent work published by Chang Hoon Lee’s group in Cellular and Molecular Gastroenterology and Hepatology illustrates the central role of tumor-associated macrophages in suppressing the immune response to CRC. These macrophages possess biosynthetic enzymes that produce high levels of adenosine. The newly generated adenosine, in turn, alters the cellular protein processing machinery and enhances the expression of PD-L1. Importantly, as treatment of CRC with cell cycle inhibitors such as palbociclib increases expression of PD-L1 and implicates a mechanism of tumor resistance, the new work shows that combined therapy to block adenosine production (with AB680) during palbociclib treatment improves tumor outcomes in mouse models.

This study offers a conceptual advance towards combinatorial therapy that can simultaneously inhibit tumor cell proliferation and activate immune surveillance, providing clear and testable hypotheses for the clinic.
 

Cambrian Y. Liu, PhD, is research assistant professor in the section of gastroenterology, hepatology, and nutrition in the department of medicine, University of Chicago. He has no relevant conflicts of interest.

Body

Although cancer immunotherapy has emerged as a powerful treatment modality, its application to colorectal cancer (CRC) is presently restricted to the minor subclass of tumors exhibiting deficient mismatch repair and high microsatellite instability. Programmed death 1 and its ligand, PD-L1, are molecules that typically suppress tumor-killing lymphocytes, yet immune checkpoint inhibitors targeting PD-1 or PD-L1 have limited to no effectiveness in the major form of proficient mismatch repair CRC. Better definition of the immune microenvironment of the different forms of CRC could lead to new treatment regimens.
 

Copyright Bogdan Condor
Dr. Cambrian Y. Liu
Recent work published by Chang Hoon Lee’s group in Cellular and Molecular Gastroenterology and Hepatology illustrates the central role of tumor-associated macrophages in suppressing the immune response to CRC. These macrophages possess biosynthetic enzymes that produce high levels of adenosine. The newly generated adenosine, in turn, alters the cellular protein processing machinery and enhances the expression of PD-L1. Importantly, as treatment of CRC with cell cycle inhibitors such as palbociclib increases expression of PD-L1 and implicates a mechanism of tumor resistance, the new work shows that combined therapy to block adenosine production (with AB680) during palbociclib treatment improves tumor outcomes in mouse models.

This study offers a conceptual advance towards combinatorial therapy that can simultaneously inhibit tumor cell proliferation and activate immune surveillance, providing clear and testable hypotheses for the clinic.
 

Cambrian Y. Liu, PhD, is research assistant professor in the section of gastroenterology, hepatology, and nutrition in the department of medicine, University of Chicago. He has no relevant conflicts of interest.

Title
​​​​​​​A conceptual advance toward combinatorial therapy
​​​​​​​A conceptual advance toward combinatorial therapy

Research that could set the stage for future clinical trials explored the potential role of adenosine signaling in altering the immune microenvironment of colorectal cancer (CRC), according to a study appearing in Cellular and Molecular Gastroenterology and Hepatology.

The study in human-derived cells and CRC mouse models suggests that addition of a CD73 inhibitor to the CDK4/6 inhibitor palbociclib may hold promise in the treatment of CRC because CD73 can produce extracellular adenosine. The results hold promise for future research since anticancer therapies often prompt increased expression of PD-L1, and CDK4/6 inhibitor monotherapy can fail because of accumulation of PD-L1 after such treatment.

Previous research has suggested that extracellular adenosine-mediated signaling can lead to accumulation of tumor-associated macrophages (TAMs) and an immunosuppressive tumor microenvironment. TAMs play an important role in the intestinal mucosal immune system in CRC, which mediates tumor-promoting metabolites in the intestine and inflammatory pathways that can lead to and progress CRC. TAMs are also linked to increases in extracellular enzymes like CD39 and CD73 as well as resistance to chemotherapy and anti-PD-1/PD-L1 therapy in CRC. Furthermore, they express PD-L1 and they promote other immunosuppressive molecules.

In normal tissues, CD73 produces adenosine to tamp down excessive immune responses. Some tumors express CD73 or even induce expression in normal cells, leading to immunosuppression in the tumor microenvironment. Previous studies have shown that CD73 expression is a biomarker for poor outcomes in gastric, liver, pancreatic, and colorectal cancer, the authors of this study noted.

To better understand the impact of adenosine, the researchers exposed human macrophages derived from peripheral blood to adenosine, and then analyzed the results using flow cytometry and Western blot. They used RNA sequencing and proteomics to discern changes in the cells that resulted from the exposure.

Adenosine treatment led to changes in the expression levels of genes involved in the cell cycle, cell division, cell cycle phase transition, and DNA repair. The researchers emphasized that extracellular treatment with adenosine led to a reduction in expression of the cell cycle–related gene CCND1, which encodes cyclin D1. Among three genes in the cyclin D family tested, CCND1 was the only one affected by adenosine. Cyclin D1 protein levels also went down.

Cyclin D1 is a known actor in regulating the cell cycle and tumorigenesis, among other roles. Previous reports indicated that cyclin D1 participates in posttranslational regulation of PD-L1, and the current study suggests it plays a similar role in TAMs after exposure to adenosine in the tumor microenvironment. Myeloid cells high in cyclin D1 expression had low levels or even an absence of the immunosuppressive molecule CD39. “Taken together, cyclin D1 may be one of the major orchestrators that trigger the differentiation of pro-tumorigenic TAMs. Our findings suggest a novel immune checkpoint regulatory mechanism of extracellular adenosine signaling, which might be related to the cell cycle of macrophages,” the authors wrote.

The researchers then introduced a short hairpin RNA directed against CCND1. This led to increases in PD-L1 protein levels. Simultaneous treatment with adenosine led to a slight increase in levels of the PD-L1 protein, which suggests that reduction of CCND1 levels is the primary cause of increased PD-L1, according to the authors.

The CD73 inhibitor AB680, currently in phase 1 clinical trials for castration-resistant prostate cancer and advanced pancreatic cancer and developed by Arcus Biosciences, led to reduced PD-L1 levels in both human and mouse macrophages, and AB680 combined with the CDK4/6 inhibitor palbociclib led to greater inhibition of tumor growth than palbociclib alone in CRC mouse models.

“Thus, the promising effects of CD73 inhibitors might breathe new life for those old drugs and provide potent therapeutic strategies. Given that the therapeutic effects of PD-1/PD-L1 immunotherapy have not been conclusively demonstrated in patients with CRC, our observations should support clinical trials of new combinational therapies for CRC,” the authors wrote.

The authors disclose no conflicts.
 

Research that could set the stage for future clinical trials explored the potential role of adenosine signaling in altering the immune microenvironment of colorectal cancer (CRC), according to a study appearing in Cellular and Molecular Gastroenterology and Hepatology.

The study in human-derived cells and CRC mouse models suggests that addition of a CD73 inhibitor to the CDK4/6 inhibitor palbociclib may hold promise in the treatment of CRC because CD73 can produce extracellular adenosine. The results hold promise for future research since anticancer therapies often prompt increased expression of PD-L1, and CDK4/6 inhibitor monotherapy can fail because of accumulation of PD-L1 after such treatment.

Previous research has suggested that extracellular adenosine-mediated signaling can lead to accumulation of tumor-associated macrophages (TAMs) and an immunosuppressive tumor microenvironment. TAMs play an important role in the intestinal mucosal immune system in CRC, which mediates tumor-promoting metabolites in the intestine and inflammatory pathways that can lead to and progress CRC. TAMs are also linked to increases in extracellular enzymes like CD39 and CD73 as well as resistance to chemotherapy and anti-PD-1/PD-L1 therapy in CRC. Furthermore, they express PD-L1 and they promote other immunosuppressive molecules.

In normal tissues, CD73 produces adenosine to tamp down excessive immune responses. Some tumors express CD73 or even induce expression in normal cells, leading to immunosuppression in the tumor microenvironment. Previous studies have shown that CD73 expression is a biomarker for poor outcomes in gastric, liver, pancreatic, and colorectal cancer, the authors of this study noted.

To better understand the impact of adenosine, the researchers exposed human macrophages derived from peripheral blood to adenosine, and then analyzed the results using flow cytometry and Western blot. They used RNA sequencing and proteomics to discern changes in the cells that resulted from the exposure.

Adenosine treatment led to changes in the expression levels of genes involved in the cell cycle, cell division, cell cycle phase transition, and DNA repair. The researchers emphasized that extracellular treatment with adenosine led to a reduction in expression of the cell cycle–related gene CCND1, which encodes cyclin D1. Among three genes in the cyclin D family tested, CCND1 was the only one affected by adenosine. Cyclin D1 protein levels also went down.

Cyclin D1 is a known actor in regulating the cell cycle and tumorigenesis, among other roles. Previous reports indicated that cyclin D1 participates in posttranslational regulation of PD-L1, and the current study suggests it plays a similar role in TAMs after exposure to adenosine in the tumor microenvironment. Myeloid cells high in cyclin D1 expression had low levels or even an absence of the immunosuppressive molecule CD39. “Taken together, cyclin D1 may be one of the major orchestrators that trigger the differentiation of pro-tumorigenic TAMs. Our findings suggest a novel immune checkpoint regulatory mechanism of extracellular adenosine signaling, which might be related to the cell cycle of macrophages,” the authors wrote.

The researchers then introduced a short hairpin RNA directed against CCND1. This led to increases in PD-L1 protein levels. Simultaneous treatment with adenosine led to a slight increase in levels of the PD-L1 protein, which suggests that reduction of CCND1 levels is the primary cause of increased PD-L1, according to the authors.

The CD73 inhibitor AB680, currently in phase 1 clinical trials for castration-resistant prostate cancer and advanced pancreatic cancer and developed by Arcus Biosciences, led to reduced PD-L1 levels in both human and mouse macrophages, and AB680 combined with the CDK4/6 inhibitor palbociclib led to greater inhibition of tumor growth than palbociclib alone in CRC mouse models.

“Thus, the promising effects of CD73 inhibitors might breathe new life for those old drugs and provide potent therapeutic strategies. Given that the therapeutic effects of PD-1/PD-L1 immunotherapy have not been conclusively demonstrated in patients with CRC, our observations should support clinical trials of new combinational therapies for CRC,” the authors wrote.

The authors disclose no conflicts.
 

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM CELLULAR AND MOLECULAR GASTROENTEROLOGY AND HEPATOLOGY

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article