1.02 Common Clinical Diagnoses and Conditions: Acute Gastroenteritis

Article Type
Changed
Thu, 07/02/2020 - 13:52

Introduction

Acute gastroenteritis (AGE) is one of the most common diseases of childhood. Admission to the hospital can be prevented in many cases with appropriate use of oral rehydration. Despite this, annual hospitalization rates in the United States have been reported to be as high 3 to 5 per 1000 US children, and the financial burden of emergency department care and hospitalization accounts for up to $350 million in costs annually. Although uncommon in developed countries, morbidity can be profound, and mortality can occur. Among hospitalized patients, complications including electrolyte abnormalities, sepsis, and malnutrition have been noted. Misdiagnosis of AGE may occur, particularly when vomiting is the predominant symptom, which can lead to inappropriate treatment for potentially life-threatening conditions. Pediatric hospitalists routinely encounter patients with AGE and should provide immediate medical care in an efficient and effective manner.

Knowledge

Pediatric hospitalists should be able to:

  • Describe the signs, symptoms, and common or concerning complications of AGE, including electrolyte disturbances, dehydration, ileus, and hemolytic uremic syndrome.
  • List the common pathogens and related epidemiologic factors for AGE depending upon age, immunization status, geographic location, and exposure and travel history.
  • Discuss the pathophysiology of electrolyte disturbances in AGE.
  • Discuss the indications for hospital admission, including the need for intravenous fluids, correction of fluid, electrolyte and acid base disturbances, close clinical monitoring, and/or further diagnostic evaluation.
  • Discuss essential elements of the history for patients with AGE, including immunization status, water and food sources, method of food preparation, daycare attendance, and recent travel.
  • Describe the elements of the physical examination that aid in the diagnosis of AGE and associated complications.
  • Compare and contrast clinical findings associated with viral, bacterial, and parasitic AGE.
  • Compare and contrast conditions with presentations like that of AGE or its complications, including critical medical and surgical diagnoses such as diabetic ketoacidosis, inborn errors of metabolism, malrotation with midgut volvulus, and bowel obstruction.
  • Compare and contrast the differential diagnoses of isolated emesis, bilious emesis, and emesis with diarrhea.
  • Describe the differences in approach toward diagnosis and treatment for patients with co-morbid conditions or immunosuppression.
  • Discuss the role of infection control in the hospital, as well as public health reporting mandates.
  • Describe the indications for diagnostic laboratory tests, including stool, blood, and urine studies, attending to age groups, predictive value of tests, and cost-effectiveness.
  • Describe the indications and contraindications of the interventions used to manage the symptoms of AGE, including the role of oral rehydration solutions in the treatment of related dehydration.
  • Discuss indications for specialty consultation, such as gastroenterology, nutrition, surgery, and others.
  • Describe criteria for hospital discharge, including specific measures of clinical stability for safe care transition.

Skills

Pediatric hospitalists should be able to:

  • Diagnose gastroenteritis by efficiently performing an accurate history and physical examination, determining if key features of the disease are present.
  • Identify and correctly manage fluid, electrolyte, and acid base derangements.
  • Assess patients efficiently and effectively for complications of gastroenteritis such as sepsis, ileus, and hemolytic uremic syndrome.
  • Identify and appropriately treat patients at risk for AGE secondary to unusual pathogens.
  • Direct a cost-effective and evidence-based evaluation and treatment plan, especially regarding laboratory studies, antibiotics, and oral or intravenous fluid resuscitation.
  • Adhere consistently to infection control practices.
  • Perform careful reassessments daily and as needed, note changes in clinical status, and respond with appropriate actions, taking care to consider alternative conditions as appropriate.
  • Engage consultants efficiently when indicated.
  • Communicate effectively with the family/caregivers and healthcare providers regarding findings and plans.
  • Ensure coordination of care for diagnostic tests and treatment between subspecialists.
  • Create a comprehensive discharge plan that can be expediently activated when appropriate.

Attitudes

Pediatric hospitalists should be able to:

  • Realize responsibility for educating the family/caregivers on the natural course of the disease, identification and management of common complications, and infection control practices to manage expectations and decrease pathogen transmission.
  • Ensure coordination of care for diagnostic tests and treatments between subspecialists.
  • Exemplify and advocate for strict adherence to infection control practices.
  • Exemplify effective communication with patients, the family/caregivers, and healthcare providers regarding findings, care plans, and anticipated health needs after discharge.

Systems Organization and Improvement

In order to improve efficiency and quality within their organizations, pediatric hospitalists should:

  • Lead, coordinate, or participate in the development and implementation of cost-effective, safe, evidence-based care pathways to standardize the evaluation and management for hospitalized children with AGE.
  • Collaborate with hospital administration to create and sustain a process to follow up on laboratory tests pending at discharge.
  • Collaborate with institutional infection control practitioners to improve processes to prevent nosocomial infection related to gastroenteritis.
  • Lead, coordinate, or participate in efforts to assure consistent public health reporting of appropriate infections and response to trends.
References

1. Chow CM, Leung AKC, Hon KL. Acute gastroenteritis: from guidelines to real life. Cli Exp Gastro. 2010; 3:97-112. https://doi.org/10.2147/ceg.s6554.

2. Freedman SB, Gouin S, Bhatt M, et al. Prospective assessment of practice pattern variations in the treatment of pediatric gastroenteritis. Pediatrics.2011;127(2) e287-e295. https://pediatrics.aappublications.org/content/127/2/e287. Accessed August 28, 2019.

Article PDF
Issue
Journal of Hospital Medicine 15(S1)
Publications
Topics
Page Number
e20-e21. DOI: 10.12788/jhm.3397
Sections
Article PDF
Article PDF

Introduction

Acute gastroenteritis (AGE) is one of the most common diseases of childhood. Admission to the hospital can be prevented in many cases with appropriate use of oral rehydration. Despite this, annual hospitalization rates in the United States have been reported to be as high 3 to 5 per 1000 US children, and the financial burden of emergency department care and hospitalization accounts for up to $350 million in costs annually. Although uncommon in developed countries, morbidity can be profound, and mortality can occur. Among hospitalized patients, complications including electrolyte abnormalities, sepsis, and malnutrition have been noted. Misdiagnosis of AGE may occur, particularly when vomiting is the predominant symptom, which can lead to inappropriate treatment for potentially life-threatening conditions. Pediatric hospitalists routinely encounter patients with AGE and should provide immediate medical care in an efficient and effective manner.

Knowledge

Pediatric hospitalists should be able to:

  • Describe the signs, symptoms, and common or concerning complications of AGE, including electrolyte disturbances, dehydration, ileus, and hemolytic uremic syndrome.
  • List the common pathogens and related epidemiologic factors for AGE depending upon age, immunization status, geographic location, and exposure and travel history.
  • Discuss the pathophysiology of electrolyte disturbances in AGE.
  • Discuss the indications for hospital admission, including the need for intravenous fluids, correction of fluid, electrolyte and acid base disturbances, close clinical monitoring, and/or further diagnostic evaluation.
  • Discuss essential elements of the history for patients with AGE, including immunization status, water and food sources, method of food preparation, daycare attendance, and recent travel.
  • Describe the elements of the physical examination that aid in the diagnosis of AGE and associated complications.
  • Compare and contrast clinical findings associated with viral, bacterial, and parasitic AGE.
  • Compare and contrast conditions with presentations like that of AGE or its complications, including critical medical and surgical diagnoses such as diabetic ketoacidosis, inborn errors of metabolism, malrotation with midgut volvulus, and bowel obstruction.
  • Compare and contrast the differential diagnoses of isolated emesis, bilious emesis, and emesis with diarrhea.
  • Describe the differences in approach toward diagnosis and treatment for patients with co-morbid conditions or immunosuppression.
  • Discuss the role of infection control in the hospital, as well as public health reporting mandates.
  • Describe the indications for diagnostic laboratory tests, including stool, blood, and urine studies, attending to age groups, predictive value of tests, and cost-effectiveness.
  • Describe the indications and contraindications of the interventions used to manage the symptoms of AGE, including the role of oral rehydration solutions in the treatment of related dehydration.
  • Discuss indications for specialty consultation, such as gastroenterology, nutrition, surgery, and others.
  • Describe criteria for hospital discharge, including specific measures of clinical stability for safe care transition.

Skills

Pediatric hospitalists should be able to:

  • Diagnose gastroenteritis by efficiently performing an accurate history and physical examination, determining if key features of the disease are present.
  • Identify and correctly manage fluid, electrolyte, and acid base derangements.
  • Assess patients efficiently and effectively for complications of gastroenteritis such as sepsis, ileus, and hemolytic uremic syndrome.
  • Identify and appropriately treat patients at risk for AGE secondary to unusual pathogens.
  • Direct a cost-effective and evidence-based evaluation and treatment plan, especially regarding laboratory studies, antibiotics, and oral or intravenous fluid resuscitation.
  • Adhere consistently to infection control practices.
  • Perform careful reassessments daily and as needed, note changes in clinical status, and respond with appropriate actions, taking care to consider alternative conditions as appropriate.
  • Engage consultants efficiently when indicated.
  • Communicate effectively with the family/caregivers and healthcare providers regarding findings and plans.
  • Ensure coordination of care for diagnostic tests and treatment between subspecialists.
  • Create a comprehensive discharge plan that can be expediently activated when appropriate.

Attitudes

Pediatric hospitalists should be able to:

  • Realize responsibility for educating the family/caregivers on the natural course of the disease, identification and management of common complications, and infection control practices to manage expectations and decrease pathogen transmission.
  • Ensure coordination of care for diagnostic tests and treatments between subspecialists.
  • Exemplify and advocate for strict adherence to infection control practices.
  • Exemplify effective communication with patients, the family/caregivers, and healthcare providers regarding findings, care plans, and anticipated health needs after discharge.

Systems Organization and Improvement

In order to improve efficiency and quality within their organizations, pediatric hospitalists should:

  • Lead, coordinate, or participate in the development and implementation of cost-effective, safe, evidence-based care pathways to standardize the evaluation and management for hospitalized children with AGE.
  • Collaborate with hospital administration to create and sustain a process to follow up on laboratory tests pending at discharge.
  • Collaborate with institutional infection control practitioners to improve processes to prevent nosocomial infection related to gastroenteritis.
  • Lead, coordinate, or participate in efforts to assure consistent public health reporting of appropriate infections and response to trends.

Introduction

Acute gastroenteritis (AGE) is one of the most common diseases of childhood. Admission to the hospital can be prevented in many cases with appropriate use of oral rehydration. Despite this, annual hospitalization rates in the United States have been reported to be as high 3 to 5 per 1000 US children, and the financial burden of emergency department care and hospitalization accounts for up to $350 million in costs annually. Although uncommon in developed countries, morbidity can be profound, and mortality can occur. Among hospitalized patients, complications including electrolyte abnormalities, sepsis, and malnutrition have been noted. Misdiagnosis of AGE may occur, particularly when vomiting is the predominant symptom, which can lead to inappropriate treatment for potentially life-threatening conditions. Pediatric hospitalists routinely encounter patients with AGE and should provide immediate medical care in an efficient and effective manner.

Knowledge

Pediatric hospitalists should be able to:

  • Describe the signs, symptoms, and common or concerning complications of AGE, including electrolyte disturbances, dehydration, ileus, and hemolytic uremic syndrome.
  • List the common pathogens and related epidemiologic factors for AGE depending upon age, immunization status, geographic location, and exposure and travel history.
  • Discuss the pathophysiology of electrolyte disturbances in AGE.
  • Discuss the indications for hospital admission, including the need for intravenous fluids, correction of fluid, electrolyte and acid base disturbances, close clinical monitoring, and/or further diagnostic evaluation.
  • Discuss essential elements of the history for patients with AGE, including immunization status, water and food sources, method of food preparation, daycare attendance, and recent travel.
  • Describe the elements of the physical examination that aid in the diagnosis of AGE and associated complications.
  • Compare and contrast clinical findings associated with viral, bacterial, and parasitic AGE.
  • Compare and contrast conditions with presentations like that of AGE or its complications, including critical medical and surgical diagnoses such as diabetic ketoacidosis, inborn errors of metabolism, malrotation with midgut volvulus, and bowel obstruction.
  • Compare and contrast the differential diagnoses of isolated emesis, bilious emesis, and emesis with diarrhea.
  • Describe the differences in approach toward diagnosis and treatment for patients with co-morbid conditions or immunosuppression.
  • Discuss the role of infection control in the hospital, as well as public health reporting mandates.
  • Describe the indications for diagnostic laboratory tests, including stool, blood, and urine studies, attending to age groups, predictive value of tests, and cost-effectiveness.
  • Describe the indications and contraindications of the interventions used to manage the symptoms of AGE, including the role of oral rehydration solutions in the treatment of related dehydration.
  • Discuss indications for specialty consultation, such as gastroenterology, nutrition, surgery, and others.
  • Describe criteria for hospital discharge, including specific measures of clinical stability for safe care transition.

Skills

Pediatric hospitalists should be able to:

  • Diagnose gastroenteritis by efficiently performing an accurate history and physical examination, determining if key features of the disease are present.
  • Identify and correctly manage fluid, electrolyte, and acid base derangements.
  • Assess patients efficiently and effectively for complications of gastroenteritis such as sepsis, ileus, and hemolytic uremic syndrome.
  • Identify and appropriately treat patients at risk for AGE secondary to unusual pathogens.
  • Direct a cost-effective and evidence-based evaluation and treatment plan, especially regarding laboratory studies, antibiotics, and oral or intravenous fluid resuscitation.
  • Adhere consistently to infection control practices.
  • Perform careful reassessments daily and as needed, note changes in clinical status, and respond with appropriate actions, taking care to consider alternative conditions as appropriate.
  • Engage consultants efficiently when indicated.
  • Communicate effectively with the family/caregivers and healthcare providers regarding findings and plans.
  • Ensure coordination of care for diagnostic tests and treatment between subspecialists.
  • Create a comprehensive discharge plan that can be expediently activated when appropriate.

Attitudes

Pediatric hospitalists should be able to:

  • Realize responsibility for educating the family/caregivers on the natural course of the disease, identification and management of common complications, and infection control practices to manage expectations and decrease pathogen transmission.
  • Ensure coordination of care for diagnostic tests and treatments between subspecialists.
  • Exemplify and advocate for strict adherence to infection control practices.
  • Exemplify effective communication with patients, the family/caregivers, and healthcare providers regarding findings, care plans, and anticipated health needs after discharge.

Systems Organization and Improvement

In order to improve efficiency and quality within their organizations, pediatric hospitalists should:

  • Lead, coordinate, or participate in the development and implementation of cost-effective, safe, evidence-based care pathways to standardize the evaluation and management for hospitalized children with AGE.
  • Collaborate with hospital administration to create and sustain a process to follow up on laboratory tests pending at discharge.
  • Collaborate with institutional infection control practitioners to improve processes to prevent nosocomial infection related to gastroenteritis.
  • Lead, coordinate, or participate in efforts to assure consistent public health reporting of appropriate infections and response to trends.
References

1. Chow CM, Leung AKC, Hon KL. Acute gastroenteritis: from guidelines to real life. Cli Exp Gastro. 2010; 3:97-112. https://doi.org/10.2147/ceg.s6554.

2. Freedman SB, Gouin S, Bhatt M, et al. Prospective assessment of practice pattern variations in the treatment of pediatric gastroenteritis. Pediatrics.2011;127(2) e287-e295. https://pediatrics.aappublications.org/content/127/2/e287. Accessed August 28, 2019.

References

1. Chow CM, Leung AKC, Hon KL. Acute gastroenteritis: from guidelines to real life. Cli Exp Gastro. 2010; 3:97-112. https://doi.org/10.2147/ceg.s6554.

2. Freedman SB, Gouin S, Bhatt M, et al. Prospective assessment of practice pattern variations in the treatment of pediatric gastroenteritis. Pediatrics.2011;127(2) e287-e295. https://pediatrics.aappublications.org/content/127/2/e287. Accessed August 28, 2019.

Issue
Journal of Hospital Medicine 15(S1)
Issue
Journal of Hospital Medicine 15(S1)
Page Number
e20-e21. DOI: 10.12788/jhm.3397
Page Number
e20-e21. DOI: 10.12788/jhm.3397
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
Open Access (article Unlocked/Open Access)
Alternative CME
Disqus Comments
Default
Gate On Date
Tue, 05/26/2020 - 12:15
Un-Gate On Date
Tue, 05/26/2020 - 12:15
Use ProPublica
CFC Schedule Remove Status
Tue, 05/26/2020 - 12:15
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Article PDF Media

1.01 Common Clinical Diagnoses and Conditions: Acute Abdominal Pain and Acute Abdomen

Article Type
Changed
Thu, 07/02/2020 - 13:47

Introduction

Acute abdominal pain is a common presenting symptom of children and adolescents and prompts the consideration of an extensive differential diagnosis. Pediatric hospitalists frequently encounter children with acute abdominal pain. Acute abdominal pain may be due to common and self-limited medical conditions such as gastroenteritis or constipation, but it may also herald life threatening surgical conditions or systemic illness. The differential diagnosis of acute abdominal pain is broad, making a careful, skilled, and thorough history and physical examination essential in the evaluation of children presenting with this symptom. Identifying children with a true medical or surgical emergency is critical. Early diagnosis and treatment reduce morbidity, mortality, and length of hospital stay.

Knowledge

Pediatric hospitalists should be able to:

  • Describe features of the medical history and physical examination that prompt specific and expedient diagnostic evaluation.
  • Compare and contrast the differential diagnoses of acute abdominal pain and acute abdomen for children of varying ages.
  • Describe and differentiate the clinical presentation consistent with intestinal obstruction from other causes of acute abdominal pain such as appendicitis, acute cholecystitis, and others.
  • Differentiate etiologies of acute abdominal pain related to biological sex, such as testicular torsion, ovarian cyst rupture, ectopic pregnancy, and others.
  • Discuss the presenting symptoms associated with abdominal emergencies (such as mid-gut volvulus and intussusception), including bilious emesis, bloody diarrhea, and severe pain.
  • List the appropriate radiological studies for evaluation of various abdominal emergencies.
  • Identify how the presentation of abdominal emergencies may differ in neonates and infants by including nonspecific symptoms, such as vomiting or lethargy.
  • List conditions that may mimic the acute abdomen, including lower lobe pneumonia, diabetic ketoacidosis, and others.
  • Discuss the benefits, risks, and limitations of commonly performed diagnostic studies, including abdominal radiography, ultrasonography, computed tomography, magnetic resonance imaging, and nuclear medicine scans, noting the benefits of and barriers to use of contrast enhancement for these studies.
  • Describe the laboratory tests indicated to evaluate acute abdominal pain and acute abdomen.
  • Discuss the importance of and indications for early surgical consultation in the child with an acute abdomen.
  • Describe the principles of stabilization of the child with an acute abdomen, including volume resuscitation, antibiotics, and bowel decompression.
  • Discuss the approach toward pain management in patients presenting with acute abdominal pain, including the impact of medication on serial exams.
  • Describe indications for patient placement in various locations in the hospital system, such as an observation unit, surgical or medical ward, step-down, or intensive care unit.
  • Discuss indications for patient transfer to a referral center, such as need for pediatric-specific services not available at the local facility.

Skills

Pediatric hospitalists should be able to:

  • Obtain an accurate and thorough history to identify symptoms, triggers, and clinical course of acute abdominal pain and acute abdomen.
  • Perform a physical exam to elicit signs of abdominal pain, differentiate findings of acute abdomen, and assess illness severity.
  • Formulate a targeted differential diagnosis based on elements from the history and physical examination.
  • Identify the child with an acute abdomen who requires emergent surgical consultation.
  • Identify and manage the child with concomitant hypovolemia or sepsis requiring immediate medical stabilization.
  • Direct an appropriate and cost-effective evaluation for acute abdominal pain and acute abdomen.
  • Create and implement a treatment plan for non-surgical causes of abdominal pain.
  • Order and correctly interpret basic diagnostic imaging and laboratory studies.
  • Consult surgeons and other subspecialists effectively and efficiently when indicated.
  • Provide pre- and post-operative general pediatric care for the child requiring surgery, including pain management, according to local practice parameters.
  • Coordinate care with the primary care provider and subspecialists to arrange an appropriate transition plan for hospital discharge.

Attitudes

Pediatric hospitalists should be able to:

  • Exemplify collaborative practice with subspecialists, including surgical teams and primary care providers, to ensure efficient care within the hospital setting and coordinated longitudinal care.
  • Realize responsibility for promoting effective communication with patients, family/caregivers, and healthcare providers regarding findings and care plans.

Systems Organization and Improvement

In order to improve efficiency and quality within their organizations, pediatric hospitalists should:

  • Lead, coordinate, or participate in education of healthcare providers, trainees, the family/caregivers regarding the signs and symptoms of the acute abdomen to encourage early detection and prompt evaluation.
  • Lead, coordinate, or participate in a multidisciplinary team to provide optimal care for children with acute abdominal pain with and without acute abdomen.
  • Lead, coordinate, or participate in institutional efforts to improve the expediency of diagnostic laboratory and radiographic studies, availability of specialty care, and other resources for children with acute abdominal pain and acute abdomen.
  • Lead, coordinate, or participate in institutional efforts to develop surgical consultation or co-management models, clearly defining roles to ensure timely, high quality, and comprehensive care for pediatric patients requiring surgical care.

 

References

1. Baker RD. Acute Abdominal Pain. Peds Rev. 2018;39(3):130-139.

2. Tsao K. and Anderson K.T. Evaluation of abdominal pain in children. BMJ Best Practice. https://bestpractice.bmj.com/topics/en-us/787/diagnosis-approach. Accessed August 27, 2019.

Article PDF
Issue
Journal of Hospital Medicine 15(S1)
Publications
Topics
Page Number
e18-e19
Sections
Article PDF
Article PDF

Introduction

Acute abdominal pain is a common presenting symptom of children and adolescents and prompts the consideration of an extensive differential diagnosis. Pediatric hospitalists frequently encounter children with acute abdominal pain. Acute abdominal pain may be due to common and self-limited medical conditions such as gastroenteritis or constipation, but it may also herald life threatening surgical conditions or systemic illness. The differential diagnosis of acute abdominal pain is broad, making a careful, skilled, and thorough history and physical examination essential in the evaluation of children presenting with this symptom. Identifying children with a true medical or surgical emergency is critical. Early diagnosis and treatment reduce morbidity, mortality, and length of hospital stay.

Knowledge

Pediatric hospitalists should be able to:

  • Describe features of the medical history and physical examination that prompt specific and expedient diagnostic evaluation.
  • Compare and contrast the differential diagnoses of acute abdominal pain and acute abdomen for children of varying ages.
  • Describe and differentiate the clinical presentation consistent with intestinal obstruction from other causes of acute abdominal pain such as appendicitis, acute cholecystitis, and others.
  • Differentiate etiologies of acute abdominal pain related to biological sex, such as testicular torsion, ovarian cyst rupture, ectopic pregnancy, and others.
  • Discuss the presenting symptoms associated with abdominal emergencies (such as mid-gut volvulus and intussusception), including bilious emesis, bloody diarrhea, and severe pain.
  • List the appropriate radiological studies for evaluation of various abdominal emergencies.
  • Identify how the presentation of abdominal emergencies may differ in neonates and infants by including nonspecific symptoms, such as vomiting or lethargy.
  • List conditions that may mimic the acute abdomen, including lower lobe pneumonia, diabetic ketoacidosis, and others.
  • Discuss the benefits, risks, and limitations of commonly performed diagnostic studies, including abdominal radiography, ultrasonography, computed tomography, magnetic resonance imaging, and nuclear medicine scans, noting the benefits of and barriers to use of contrast enhancement for these studies.
  • Describe the laboratory tests indicated to evaluate acute abdominal pain and acute abdomen.
  • Discuss the importance of and indications for early surgical consultation in the child with an acute abdomen.
  • Describe the principles of stabilization of the child with an acute abdomen, including volume resuscitation, antibiotics, and bowel decompression.
  • Discuss the approach toward pain management in patients presenting with acute abdominal pain, including the impact of medication on serial exams.
  • Describe indications for patient placement in various locations in the hospital system, such as an observation unit, surgical or medical ward, step-down, or intensive care unit.
  • Discuss indications for patient transfer to a referral center, such as need for pediatric-specific services not available at the local facility.

Skills

Pediatric hospitalists should be able to:

  • Obtain an accurate and thorough history to identify symptoms, triggers, and clinical course of acute abdominal pain and acute abdomen.
  • Perform a physical exam to elicit signs of abdominal pain, differentiate findings of acute abdomen, and assess illness severity.
  • Formulate a targeted differential diagnosis based on elements from the history and physical examination.
  • Identify the child with an acute abdomen who requires emergent surgical consultation.
  • Identify and manage the child with concomitant hypovolemia or sepsis requiring immediate medical stabilization.
  • Direct an appropriate and cost-effective evaluation for acute abdominal pain and acute abdomen.
  • Create and implement a treatment plan for non-surgical causes of abdominal pain.
  • Order and correctly interpret basic diagnostic imaging and laboratory studies.
  • Consult surgeons and other subspecialists effectively and efficiently when indicated.
  • Provide pre- and post-operative general pediatric care for the child requiring surgery, including pain management, according to local practice parameters.
  • Coordinate care with the primary care provider and subspecialists to arrange an appropriate transition plan for hospital discharge.

Attitudes

Pediatric hospitalists should be able to:

  • Exemplify collaborative practice with subspecialists, including surgical teams and primary care providers, to ensure efficient care within the hospital setting and coordinated longitudinal care.
  • Realize responsibility for promoting effective communication with patients, family/caregivers, and healthcare providers regarding findings and care plans.

Systems Organization and Improvement

In order to improve efficiency and quality within their organizations, pediatric hospitalists should:

  • Lead, coordinate, or participate in education of healthcare providers, trainees, the family/caregivers regarding the signs and symptoms of the acute abdomen to encourage early detection and prompt evaluation.
  • Lead, coordinate, or participate in a multidisciplinary team to provide optimal care for children with acute abdominal pain with and without acute abdomen.
  • Lead, coordinate, or participate in institutional efforts to improve the expediency of diagnostic laboratory and radiographic studies, availability of specialty care, and other resources for children with acute abdominal pain and acute abdomen.
  • Lead, coordinate, or participate in institutional efforts to develop surgical consultation or co-management models, clearly defining roles to ensure timely, high quality, and comprehensive care for pediatric patients requiring surgical care.

 

Introduction

Acute abdominal pain is a common presenting symptom of children and adolescents and prompts the consideration of an extensive differential diagnosis. Pediatric hospitalists frequently encounter children with acute abdominal pain. Acute abdominal pain may be due to common and self-limited medical conditions such as gastroenteritis or constipation, but it may also herald life threatening surgical conditions or systemic illness. The differential diagnosis of acute abdominal pain is broad, making a careful, skilled, and thorough history and physical examination essential in the evaluation of children presenting with this symptom. Identifying children with a true medical or surgical emergency is critical. Early diagnosis and treatment reduce morbidity, mortality, and length of hospital stay.

Knowledge

Pediatric hospitalists should be able to:

  • Describe features of the medical history and physical examination that prompt specific and expedient diagnostic evaluation.
  • Compare and contrast the differential diagnoses of acute abdominal pain and acute abdomen for children of varying ages.
  • Describe and differentiate the clinical presentation consistent with intestinal obstruction from other causes of acute abdominal pain such as appendicitis, acute cholecystitis, and others.
  • Differentiate etiologies of acute abdominal pain related to biological sex, such as testicular torsion, ovarian cyst rupture, ectopic pregnancy, and others.
  • Discuss the presenting symptoms associated with abdominal emergencies (such as mid-gut volvulus and intussusception), including bilious emesis, bloody diarrhea, and severe pain.
  • List the appropriate radiological studies for evaluation of various abdominal emergencies.
  • Identify how the presentation of abdominal emergencies may differ in neonates and infants by including nonspecific symptoms, such as vomiting or lethargy.
  • List conditions that may mimic the acute abdomen, including lower lobe pneumonia, diabetic ketoacidosis, and others.
  • Discuss the benefits, risks, and limitations of commonly performed diagnostic studies, including abdominal radiography, ultrasonography, computed tomography, magnetic resonance imaging, and nuclear medicine scans, noting the benefits of and barriers to use of contrast enhancement for these studies.
  • Describe the laboratory tests indicated to evaluate acute abdominal pain and acute abdomen.
  • Discuss the importance of and indications for early surgical consultation in the child with an acute abdomen.
  • Describe the principles of stabilization of the child with an acute abdomen, including volume resuscitation, antibiotics, and bowel decompression.
  • Discuss the approach toward pain management in patients presenting with acute abdominal pain, including the impact of medication on serial exams.
  • Describe indications for patient placement in various locations in the hospital system, such as an observation unit, surgical or medical ward, step-down, or intensive care unit.
  • Discuss indications for patient transfer to a referral center, such as need for pediatric-specific services not available at the local facility.

Skills

Pediatric hospitalists should be able to:

  • Obtain an accurate and thorough history to identify symptoms, triggers, and clinical course of acute abdominal pain and acute abdomen.
  • Perform a physical exam to elicit signs of abdominal pain, differentiate findings of acute abdomen, and assess illness severity.
  • Formulate a targeted differential diagnosis based on elements from the history and physical examination.
  • Identify the child with an acute abdomen who requires emergent surgical consultation.
  • Identify and manage the child with concomitant hypovolemia or sepsis requiring immediate medical stabilization.
  • Direct an appropriate and cost-effective evaluation for acute abdominal pain and acute abdomen.
  • Create and implement a treatment plan for non-surgical causes of abdominal pain.
  • Order and correctly interpret basic diagnostic imaging and laboratory studies.
  • Consult surgeons and other subspecialists effectively and efficiently when indicated.
  • Provide pre- and post-operative general pediatric care for the child requiring surgery, including pain management, according to local practice parameters.
  • Coordinate care with the primary care provider and subspecialists to arrange an appropriate transition plan for hospital discharge.

Attitudes

Pediatric hospitalists should be able to:

  • Exemplify collaborative practice with subspecialists, including surgical teams and primary care providers, to ensure efficient care within the hospital setting and coordinated longitudinal care.
  • Realize responsibility for promoting effective communication with patients, family/caregivers, and healthcare providers regarding findings and care plans.

Systems Organization and Improvement

In order to improve efficiency and quality within their organizations, pediatric hospitalists should:

  • Lead, coordinate, or participate in education of healthcare providers, trainees, the family/caregivers regarding the signs and symptoms of the acute abdomen to encourage early detection and prompt evaluation.
  • Lead, coordinate, or participate in a multidisciplinary team to provide optimal care for children with acute abdominal pain with and without acute abdomen.
  • Lead, coordinate, or participate in institutional efforts to improve the expediency of diagnostic laboratory and radiographic studies, availability of specialty care, and other resources for children with acute abdominal pain and acute abdomen.
  • Lead, coordinate, or participate in institutional efforts to develop surgical consultation or co-management models, clearly defining roles to ensure timely, high quality, and comprehensive care for pediatric patients requiring surgical care.

 

References

1. Baker RD. Acute Abdominal Pain. Peds Rev. 2018;39(3):130-139.

2. Tsao K. and Anderson K.T. Evaluation of abdominal pain in children. BMJ Best Practice. https://bestpractice.bmj.com/topics/en-us/787/diagnosis-approach. Accessed August 27, 2019.

References

1. Baker RD. Acute Abdominal Pain. Peds Rev. 2018;39(3):130-139.

2. Tsao K. and Anderson K.T. Evaluation of abdominal pain in children. BMJ Best Practice. https://bestpractice.bmj.com/topics/en-us/787/diagnosis-approach. Accessed August 27, 2019.

Issue
Journal of Hospital Medicine 15(S1)
Issue
Journal of Hospital Medicine 15(S1)
Page Number
e18-e19
Page Number
e18-e19
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
Open Access (article Unlocked/Open Access)
Alternative CME
Disqus Comments
Default
Gate On Date
Tue, 05/26/2020 - 12:00
Un-Gate On Date
Tue, 05/26/2020 - 12:00
Use ProPublica
CFC Schedule Remove Status
Tue, 05/26/2020 - 12:00
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Article PDF Media

Pediatric Hospital Medicine Core Competencies: 2020 Revision. Table of Contents

Article Type
Changed
Thu, 04/01/2021 - 11:19


Authors and Editors.............................................................3
External Reviewers.............................................................11

ORIGINAL RESEARCH

The Pediatric Hospital Medicine Core Competencies: 
2020 Revision—Introduction and Methodology.............................................................12
Jennifer Maniscalco, MD, MPH, MAcM, FAAP; Sandra Gage, MD, PhD, SFHM, FAAP; Sofia Teferi, MD, SFHM, FAAP; Erin Stucky Fisher, MD, MHM, FAAP

SECTION 1: COMMON CLINICAL DIAGNOSES AND CONDITIONS

1.01 Acute Abdominal Pain and Acute Abdomen.............................................................18
1.02 Acute Gastroenteritis.............................................................20
1.03 Acute Respiratory Failure.............................................................22
1.04 Altered Mental Status.............................................................24
1.05 Asthma.............................................................26
1.06 Bone and Joint Infections.............................................................28
1.07 Brief Resolved Unexplained Event.............................................................30
1.08 Bronchiolitis.............................................................32
1.09 Central Nervous System Infections.............................................................34
1.10 Constipation.............................................................36
1.11 Diabetes Mellitus.............................................................37
1.12 Failure to Thrive.............................................................39
1.13 Fever of Unknown Origin.............................................................41
1.14 Fluid and Electrolyte Management.............................................................43
1.15 Gastrointestinal and Digestive Disorders.............................................................45
1.16 Head and Neck Disorders.............................................................47
1.17 Kawasaki Disease.............................................................49
1.18 Neonatal Abstinence Syndrome/Neonatal Opioid Withdrawal Syndrome.............................................................50
1.19 Neonatal Fever.............................................................52
1.20 Neonatal Jaundice.............................................................54
1.21 Pneumonia.............................................................56
1.22 Seizures.............................................................57
1.23 Sepsis and Shock.............................................................59
1.24 Sickle Cell Disease.............................................................61
1.25 Skin and Soft Tissue Infections.............................................................63
1.26 Toxin Ingestion and Exposure.............................................................65
1.27 Urinary Tract Infections.............................................................67

SECTION 2: CORE SKILLS

2.01 Bladder Catheterization and Interpretation 
of Urinalysis.............................................................68
2.02 Communication.............................................................70
2.03 Diagnostic Imaging.............................................................72
2.04 Electrocardiogram Interpretation.............................................................74
2.05 Feeding Tubes.............................................................75
2.06 Intravenous Access and Phlebotomy.............................................................77
2.07 Lumbar Puncture.............................................................79
2.08 Non-invasive Monitoring.............................................................81
2.09 Nutrition.............................................................82
2.10 Oxygen Delivery and Airway Management.............................................................84
2.11 Pain Management.............................................................86
2.12 Pediatric Advanced Life Support.............................................................88
2.13 Peri-procedural Care.............................................................90
2.14 Preventive Care Services.............................................................92
2.15 Procedural Sedation.............................................................94

SECTION 3: SPECIALIZED SERVICES

3.01 Acute Behavioral and Psychiatric Conditions.............................................................96
3.02 Adolescent and Young Adult Medicine.............................................................98
3.03 Child Abuse and Neglect.............................................................100
3.04 Child with Medical Complexity.............................................................102
3.05 Chronic Behavioral and Psychiatric Conditions.............................................................104
3.06 Newborn Care and Delivery Room Management.............................................................106
3.07 Palliative Care and Hospice.............................................................108
3.08 Pediatric Interfacility Transport.............................................................110

SECTION 4: HEALTHCARE SYSTEMS: SUPPORTING AND ADVANCING CHILD HEALTH

4.01 Advocacy.............................................................112
4.02 Business Practices.............................................................114
4.03 Consultation and Co-management.............................................................116
4.04 Education.............................................................118
4.05 Ethics.............................................................120
4.06 Evidence-based Medicine.............................................................122
4.07 Family Centered Care.............................................................123
4.08 Handoffs and Transitions of Care.............................................................125
4.09 Health Information Technology.............................................................127
4.10 High Value Care.............................................................129
4.11 Infection Control and Antimicrobial Stewardship.............................................................131
4.12 Leadership in Healthcare.............................................................133
4.13 Legal Issues and Risk Management.............................................................134
4.14 Patient Safety.............................................................136
4.15 Quality Improvement.............................................................138
4.16 Research.............................................................140

APPENDIX

Chapter Links.............................................................142
These chapter links are guides to assist the reader in identifying chapters where some key relationships across knowledge, skills, attitudes, and systems organization and improvement may overlap. Chapter links are limited to 5 per chapter, are not comprehensive, and are intended as a general guide for the reader.
Figure: Needs Assessment Survey.............................................................145
Dedication.............................................................153
To Michael Burke, our friend and colleague

Article PDF
Issue
Journal of Hospital Medicine 15(S1)
Publications
Topics
Page Number
e1-e2
Sections
Article PDF
Article PDF


Authors and Editors.............................................................3
External Reviewers.............................................................11

ORIGINAL RESEARCH

The Pediatric Hospital Medicine Core Competencies: 
2020 Revision—Introduction and Methodology.............................................................12
Jennifer Maniscalco, MD, MPH, MAcM, FAAP; Sandra Gage, MD, PhD, SFHM, FAAP; Sofia Teferi, MD, SFHM, FAAP; Erin Stucky Fisher, MD, MHM, FAAP

SECTION 1: COMMON CLINICAL DIAGNOSES AND CONDITIONS

1.01 Acute Abdominal Pain and Acute Abdomen.............................................................18
1.02 Acute Gastroenteritis.............................................................20
1.03 Acute Respiratory Failure.............................................................22
1.04 Altered Mental Status.............................................................24
1.05 Asthma.............................................................26
1.06 Bone and Joint Infections.............................................................28
1.07 Brief Resolved Unexplained Event.............................................................30
1.08 Bronchiolitis.............................................................32
1.09 Central Nervous System Infections.............................................................34
1.10 Constipation.............................................................36
1.11 Diabetes Mellitus.............................................................37
1.12 Failure to Thrive.............................................................39
1.13 Fever of Unknown Origin.............................................................41
1.14 Fluid and Electrolyte Management.............................................................43
1.15 Gastrointestinal and Digestive Disorders.............................................................45
1.16 Head and Neck Disorders.............................................................47
1.17 Kawasaki Disease.............................................................49
1.18 Neonatal Abstinence Syndrome/Neonatal Opioid Withdrawal Syndrome.............................................................50
1.19 Neonatal Fever.............................................................52
1.20 Neonatal Jaundice.............................................................54
1.21 Pneumonia.............................................................56
1.22 Seizures.............................................................57
1.23 Sepsis and Shock.............................................................59
1.24 Sickle Cell Disease.............................................................61
1.25 Skin and Soft Tissue Infections.............................................................63
1.26 Toxin Ingestion and Exposure.............................................................65
1.27 Urinary Tract Infections.............................................................67

SECTION 2: CORE SKILLS

2.01 Bladder Catheterization and Interpretation 
of Urinalysis.............................................................68
2.02 Communication.............................................................70
2.03 Diagnostic Imaging.............................................................72
2.04 Electrocardiogram Interpretation.............................................................74
2.05 Feeding Tubes.............................................................75
2.06 Intravenous Access and Phlebotomy.............................................................77
2.07 Lumbar Puncture.............................................................79
2.08 Non-invasive Monitoring.............................................................81
2.09 Nutrition.............................................................82
2.10 Oxygen Delivery and Airway Management.............................................................84
2.11 Pain Management.............................................................86
2.12 Pediatric Advanced Life Support.............................................................88
2.13 Peri-procedural Care.............................................................90
2.14 Preventive Care Services.............................................................92
2.15 Procedural Sedation.............................................................94

SECTION 3: SPECIALIZED SERVICES

3.01 Acute Behavioral and Psychiatric Conditions.............................................................96
3.02 Adolescent and Young Adult Medicine.............................................................98
3.03 Child Abuse and Neglect.............................................................100
3.04 Child with Medical Complexity.............................................................102
3.05 Chronic Behavioral and Psychiatric Conditions.............................................................104
3.06 Newborn Care and Delivery Room Management.............................................................106
3.07 Palliative Care and Hospice.............................................................108
3.08 Pediatric Interfacility Transport.............................................................110

SECTION 4: HEALTHCARE SYSTEMS: SUPPORTING AND ADVANCING CHILD HEALTH

4.01 Advocacy.............................................................112
4.02 Business Practices.............................................................114
4.03 Consultation and Co-management.............................................................116
4.04 Education.............................................................118
4.05 Ethics.............................................................120
4.06 Evidence-based Medicine.............................................................122
4.07 Family Centered Care.............................................................123
4.08 Handoffs and Transitions of Care.............................................................125
4.09 Health Information Technology.............................................................127
4.10 High Value Care.............................................................129
4.11 Infection Control and Antimicrobial Stewardship.............................................................131
4.12 Leadership in Healthcare.............................................................133
4.13 Legal Issues and Risk Management.............................................................134
4.14 Patient Safety.............................................................136
4.15 Quality Improvement.............................................................138
4.16 Research.............................................................140

APPENDIX

Chapter Links.............................................................142
These chapter links are guides to assist the reader in identifying chapters where some key relationships across knowledge, skills, attitudes, and systems organization and improvement may overlap. Chapter links are limited to 5 per chapter, are not comprehensive, and are intended as a general guide for the reader.
Figure: Needs Assessment Survey.............................................................145
Dedication.............................................................153
To Michael Burke, our friend and colleague


Authors and Editors.............................................................3
External Reviewers.............................................................11

ORIGINAL RESEARCH

The Pediatric Hospital Medicine Core Competencies: 
2020 Revision—Introduction and Methodology.............................................................12
Jennifer Maniscalco, MD, MPH, MAcM, FAAP; Sandra Gage, MD, PhD, SFHM, FAAP; Sofia Teferi, MD, SFHM, FAAP; Erin Stucky Fisher, MD, MHM, FAAP

SECTION 1: COMMON CLINICAL DIAGNOSES AND CONDITIONS

1.01 Acute Abdominal Pain and Acute Abdomen.............................................................18
1.02 Acute Gastroenteritis.............................................................20
1.03 Acute Respiratory Failure.............................................................22
1.04 Altered Mental Status.............................................................24
1.05 Asthma.............................................................26
1.06 Bone and Joint Infections.............................................................28
1.07 Brief Resolved Unexplained Event.............................................................30
1.08 Bronchiolitis.............................................................32
1.09 Central Nervous System Infections.............................................................34
1.10 Constipation.............................................................36
1.11 Diabetes Mellitus.............................................................37
1.12 Failure to Thrive.............................................................39
1.13 Fever of Unknown Origin.............................................................41
1.14 Fluid and Electrolyte Management.............................................................43
1.15 Gastrointestinal and Digestive Disorders.............................................................45
1.16 Head and Neck Disorders.............................................................47
1.17 Kawasaki Disease.............................................................49
1.18 Neonatal Abstinence Syndrome/Neonatal Opioid Withdrawal Syndrome.............................................................50
1.19 Neonatal Fever.............................................................52
1.20 Neonatal Jaundice.............................................................54
1.21 Pneumonia.............................................................56
1.22 Seizures.............................................................57
1.23 Sepsis and Shock.............................................................59
1.24 Sickle Cell Disease.............................................................61
1.25 Skin and Soft Tissue Infections.............................................................63
1.26 Toxin Ingestion and Exposure.............................................................65
1.27 Urinary Tract Infections.............................................................67

SECTION 2: CORE SKILLS

2.01 Bladder Catheterization and Interpretation 
of Urinalysis.............................................................68
2.02 Communication.............................................................70
2.03 Diagnostic Imaging.............................................................72
2.04 Electrocardiogram Interpretation.............................................................74
2.05 Feeding Tubes.............................................................75
2.06 Intravenous Access and Phlebotomy.............................................................77
2.07 Lumbar Puncture.............................................................79
2.08 Non-invasive Monitoring.............................................................81
2.09 Nutrition.............................................................82
2.10 Oxygen Delivery and Airway Management.............................................................84
2.11 Pain Management.............................................................86
2.12 Pediatric Advanced Life Support.............................................................88
2.13 Peri-procedural Care.............................................................90
2.14 Preventive Care Services.............................................................92
2.15 Procedural Sedation.............................................................94

SECTION 3: SPECIALIZED SERVICES

3.01 Acute Behavioral and Psychiatric Conditions.............................................................96
3.02 Adolescent and Young Adult Medicine.............................................................98
3.03 Child Abuse and Neglect.............................................................100
3.04 Child with Medical Complexity.............................................................102
3.05 Chronic Behavioral and Psychiatric Conditions.............................................................104
3.06 Newborn Care and Delivery Room Management.............................................................106
3.07 Palliative Care and Hospice.............................................................108
3.08 Pediatric Interfacility Transport.............................................................110

SECTION 4: HEALTHCARE SYSTEMS: SUPPORTING AND ADVANCING CHILD HEALTH

4.01 Advocacy.............................................................112
4.02 Business Practices.............................................................114
4.03 Consultation and Co-management.............................................................116
4.04 Education.............................................................118
4.05 Ethics.............................................................120
4.06 Evidence-based Medicine.............................................................122
4.07 Family Centered Care.............................................................123
4.08 Handoffs and Transitions of Care.............................................................125
4.09 Health Information Technology.............................................................127
4.10 High Value Care.............................................................129
4.11 Infection Control and Antimicrobial Stewardship.............................................................131
4.12 Leadership in Healthcare.............................................................133
4.13 Legal Issues and Risk Management.............................................................134
4.14 Patient Safety.............................................................136
4.15 Quality Improvement.............................................................138
4.16 Research.............................................................140

APPENDIX

Chapter Links.............................................................142
These chapter links are guides to assist the reader in identifying chapters where some key relationships across knowledge, skills, attitudes, and systems organization and improvement may overlap. Chapter links are limited to 5 per chapter, are not comprehensive, and are intended as a general guide for the reader.
Figure: Needs Assessment Survey.............................................................145
Dedication.............................................................153
To Michael Burke, our friend and colleague

Issue
Journal of Hospital Medicine 15(S1)
Issue
Journal of Hospital Medicine 15(S1)
Page Number
e1-e2
Page Number
e1-e2
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
Open Access (article Unlocked/Open Access)
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
Article PDF Media

The Pediatric Hospital Medicine Core Competencies: 2020 Revision Dedication

Article Type
Changed
Thu, 07/23/2020 - 16:03

We dedicate this publication of The Pediatric Hospital Medicine Core Competencies: 2020 Revision to our esteemed colleague, friend, and mentor, Dr. Michael Burke who died unexpectedly on July 10, 2019. Michael was the physician we all aspire to be: thoughtful, kind, gentle, intelligent, and dedicated to the science and the art of medicine. A number of those reading these pages will have known Michael’s warm smile and ability to make you feel as though you were the only one in the room, the only one who mattered just then. To those of you who did not have the luck to know him personally, you have not lost an opportunity to learn from him. His touch is on these pages, encouraging us to never stop learning, to always listen to patients and the family/caregivers, and to never fail to believe in yourself.

 

Thank you, Michael, for making us a stronger and more compassionate PHM Community.

The Editors and Associate Editors of The Pediatric Hospital Medicine Core Competencies: 2020 Revision:

Francisco Alvarez; Weijen Chang; Erin Fisher; Sandra Gage; Jennifer Maniscalco; Vineeta Mittal; Anand Sekaran; Amit Singh; Sofia Teferi

Article PDF
Issue
Journal of Hospital Medicine 15(S1)
Publications
Topics
Page Number
e153.
Sections
Article PDF
Article PDF

We dedicate this publication of The Pediatric Hospital Medicine Core Competencies: 2020 Revision to our esteemed colleague, friend, and mentor, Dr. Michael Burke who died unexpectedly on July 10, 2019. Michael was the physician we all aspire to be: thoughtful, kind, gentle, intelligent, and dedicated to the science and the art of medicine. A number of those reading these pages will have known Michael’s warm smile and ability to make you feel as though you were the only one in the room, the only one who mattered just then. To those of you who did not have the luck to know him personally, you have not lost an opportunity to learn from him. His touch is on these pages, encouraging us to never stop learning, to always listen to patients and the family/caregivers, and to never fail to believe in yourself.

 

Thank you, Michael, for making us a stronger and more compassionate PHM Community.

The Editors and Associate Editors of The Pediatric Hospital Medicine Core Competencies: 2020 Revision:

Francisco Alvarez; Weijen Chang; Erin Fisher; Sandra Gage; Jennifer Maniscalco; Vineeta Mittal; Anand Sekaran; Amit Singh; Sofia Teferi

We dedicate this publication of The Pediatric Hospital Medicine Core Competencies: 2020 Revision to our esteemed colleague, friend, and mentor, Dr. Michael Burke who died unexpectedly on July 10, 2019. Michael was the physician we all aspire to be: thoughtful, kind, gentle, intelligent, and dedicated to the science and the art of medicine. A number of those reading these pages will have known Michael’s warm smile and ability to make you feel as though you were the only one in the room, the only one who mattered just then. To those of you who did not have the luck to know him personally, you have not lost an opportunity to learn from him. His touch is on these pages, encouraging us to never stop learning, to always listen to patients and the family/caregivers, and to never fail to believe in yourself.

 

Thank you, Michael, for making us a stronger and more compassionate PHM Community.

The Editors and Associate Editors of The Pediatric Hospital Medicine Core Competencies: 2020 Revision:

Francisco Alvarez; Weijen Chang; Erin Fisher; Sandra Gage; Jennifer Maniscalco; Vineeta Mittal; Anand Sekaran; Amit Singh; Sofia Teferi

Issue
Journal of Hospital Medicine 15(S1)
Issue
Journal of Hospital Medicine 15(S1)
Page Number
e153.
Page Number
e153.
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
Article PDF Media

APPENDIX

Article Type
Changed
Mon, 06/29/2020 - 14:53

NEEDS ASSESSMENT SURVEY

The editors conducted a needs assessment survey with several stakeholder groups, including SHM’s Pediatrics and Medicine-Pediatrics SIGs, AAP Section on Hospital Medicine and its subcommittees, APA Hospital Medicine SIG, PHM Fellowship Directors Council,and PHM Division Directors, with encouragement to pass the survey link to others in the PHM community interested in providing input (Appendix Figure). The solicitation asked for comment  on existing chapters and suggestions for new chapters. For any new chapter, respondents were asked to note the intended purpose of the chapter and the anticipated value that chapter would bring to our profession and the children and the caregivers served by pediatric hospitalists.

CHAPTER LINKS

These chapter links are guides to assist the reader in identifying chapters where some key relationships across knowledge, skills, attitudes, and systems organization and improvement may overlap. Chapter links are limited to 5 per chapter, are not comprehensive, and are intended as a general guide for the reader.

Files
Article PDF
Issue
Journal of Hospital Medicine 15(S1)
Publications
Topics
Page Number
e142-e152
Sections
Files
Files
Article PDF
Article PDF

NEEDS ASSESSMENT SURVEY

The editors conducted a needs assessment survey with several stakeholder groups, including SHM’s Pediatrics and Medicine-Pediatrics SIGs, AAP Section on Hospital Medicine and its subcommittees, APA Hospital Medicine SIG, PHM Fellowship Directors Council,and PHM Division Directors, with encouragement to pass the survey link to others in the PHM community interested in providing input (Appendix Figure). The solicitation asked for comment  on existing chapters and suggestions for new chapters. For any new chapter, respondents were asked to note the intended purpose of the chapter and the anticipated value that chapter would bring to our profession and the children and the caregivers served by pediatric hospitalists.

CHAPTER LINKS

These chapter links are guides to assist the reader in identifying chapters where some key relationships across knowledge, skills, attitudes, and systems organization and improvement may overlap. Chapter links are limited to 5 per chapter, are not comprehensive, and are intended as a general guide for the reader.

NEEDS ASSESSMENT SURVEY

The editors conducted a needs assessment survey with several stakeholder groups, including SHM’s Pediatrics and Medicine-Pediatrics SIGs, AAP Section on Hospital Medicine and its subcommittees, APA Hospital Medicine SIG, PHM Fellowship Directors Council,and PHM Division Directors, with encouragement to pass the survey link to others in the PHM community interested in providing input (Appendix Figure). The solicitation asked for comment  on existing chapters and suggestions for new chapters. For any new chapter, respondents were asked to note the intended purpose of the chapter and the anticipated value that chapter would bring to our profession and the children and the caregivers served by pediatric hospitalists.

CHAPTER LINKS

These chapter links are guides to assist the reader in identifying chapters where some key relationships across knowledge, skills, attitudes, and systems organization and improvement may overlap. Chapter links are limited to 5 per chapter, are not comprehensive, and are intended as a general guide for the reader.

Issue
Journal of Hospital Medicine 15(S1)
Issue
Journal of Hospital Medicine 15(S1)
Page Number
e142-e152
Page Number
e142-e152
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
Article PDF Media
Media Files

Research News: Neurologic Disorders (FULL)

Article Type
Changed
Mon, 06/01/2020 - 15:23
Display Headline
Research News: Neurologic Disorders

While several dozen studies have been conducted into cannabis-based treatments for symptoms of multiple sclerosis (MS), a new systematic review deems most to be of fair to poor quality. Reviewers found modest evidence of benefit and plenty of room for more research.

“Cannabis-based medicine may be useful for refractory MS symptoms, especially spasticity and pain, and side effects are usually well tolerated,” study lead author Natasha Breward, a graduate student at the College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, said in an interview. Breward spoke prior to the presentation of the study findings at the 2019 meeting of the Consortium of Multiple Sclerosis Centers.

For the review, Breward and colleagues focused on 60 studies—26 randomized controlled trials and 34 trials with other designs. Forty of the studies used nabiximols, an oromucosal spray that is derived from the cannabis sativa plant and approved for use in multiple countries but not yet in the US.

According to Breward, some of the other treatments included dried cannabis that is smoked or eaten and cannabidiol that’s typically delivered with tetrahydrocannabinol (THC) either oromucosally or as an oral capsule.

MS symptoms treated in the studies included spasticity (n = 29), pain (n = 8), and cognition (n = 6). The researchers considered 22 studies to be poor quality, 14 to be fair quality, and 24 to be good/excellent quality.

The researchers found that the cannabis-based medicine “significantly reduced spasticity and pain in several individual good-quality studies,” Breward said. The drugs seem to work by inhibiting neurotransmitter release via cannabinoids. “However, the variability in study quality—and in the products and regimens studied—make it hard to draw any conclusions about specific products and doses that may have the most potential benefit,” she added.

“Further research should focus on the use of different products and formulations of cannabis-based medicine such as cannabis oil and cannabidiol-prominent products, as no studies have focused on this area,” she said. “Research should also look at the potential of cannabis-based medicine for the treatment of disease progression, as cannabinoids are anti-inflammatory and immunomodulatory. Finally, more research regarding the potentially synergistic effects of cannabis-based medicine administered with current MS medications would also be useful.”

Randy Dotinga, MDedge.com/neurology

 

 

Brain volumes of specific regions of interest can be used to classify traumatic brain injury subjects that fall into predetermined symptom categories, according to a study presented at the annual meeting of the American Academy of Neurology.

Traumatic brain injury (TBI) damages brain tissue and causes subsequent volume loss, which may result in clinical symptoms. It is a prevalent worldwide health problem caused by a mechanical insult to the head, resulting in transient or permanent alteration to brain tissue and/or function. Standard neuroimaging with computed cranial tomography (CT) and structural magnetic resonance imaging (MRI) is often unrevealing during the evaluation of patients with TBI, particularly those classified as mild TBI.

In this study, James Rock, MD, of Penn Presbyterian Medical Center and the University of Pennsylvania, and colleagues sought to examine the value of quantitative analysis of regional brain volumes in the evaluation of TBI. The investigators reviewed the medical records and MRI imaging from 44 patients with TBI evaluated at a Level I trauma center. They also read clinical notes to assess reported symptoms and physical findings.

Regional volumes from TBI subjects were derived using the software package Freesurfer image analysis suite (surfer.nmr.mgh.harvard.edu), which utilizes a T1-weighted structural scan to calculate volumetric information. A machine learning algorithm, random forests, was employed across volume measurements from 25 regions of interest to determine the most important regions for classifying subjects based on clinical outcome and symptomology.

Basal ganglia volume showed the highest variable importance with regards to classifying subjects who exhibited symptoms of cognitive dysfunction in quantitative analysis. Left lateral ventricle volume was important in classifying subjects with motor and vestibular alterations. Left choroid plexus volume was the most important region for classifying subjects with sensation and somatic dysfunction.

In an abstract, the researchers noted that their study is ongoing. “It will be extended to a larger cohort to determine whether volume changes in specific [regions of interest] can act as useful clinical biomarkers for chronic symptoms,” they said.

Dr. Diaz-Arrastia received personal compensation from Neural Analytics, Inc; BrainBox Solutions, Inc; and Bioscience Pharma Partners. Dr. Diaz-Arrastia holds stock and/or stock options in Neural Analytics, Inc and has received research support from BrainBox Solutions. The other authors reported no other disclosures.

Glenn S. Williams, MDedge.com/neurology

 

 

Concomitant medication use is common when patients with multiple sclerosis (MS) start disease-modifying drugs (DMDs), according to research presented at the 2019 meeting of the Consortium of Multiple Sclerosis Centers. The likelihood of particular comorbidities and concomitant medications varies by age and sex, researchers reported.

“This may have implications for MS treatment,” said study author Jacqueline Nicholas, MD, MPH, of Ohio Multiple Sclerosis Center in Columbus and colleagues. “A better understanding of the effects of comorbidities and concomitant medications on the effectiveness and safety of DMDs is needed to support clinical decision making.”

Researchers have examined comorbidities in patients with MS, but concomitant medication use among patients starting DMDs is poorly understood, the authors said.

To study this question, Dr. Nicholas and colleagues analyzed retrospective administrative claims data from IQVIA Real-World Data Adjudicated Claims–US database from Jan. 1, 2010, to June 30, 2017. Their analysis included patients with ≥ 2 MS diagnosis claims and at least 1 DMD claim between Jan. 1, 2011, and June 30, 2015. Eligible patients were aged 18 to 63 years and had continuous eligibility with commercial insurance 1 year before and 2 years after DMD initiation. In addition, patients had no evidence of DMD use during the 1-year baseline period.

The investigators used International Classification of Diseases, 9th and 10th revisions, Clinical Modification codes and claims to evaluate patients’ comorbidities and concomitant medications during the study period.

The researchers identified 8,251 eligible patients. Patients had a mean age of 43.2 years, and 75.5% were female. Average baseline Charlson Comorbidity Index was 0.41. In the 2 years after DMD initiation, common comorbid diagnoses were hyperlipidemia (30.0%), hypertension (28.2%), gastrointestinal disorders (26.2%), depression (25.5%), and anxiety (20.1%).

Common concomitant medications included antibiotics (70.6%); analgesics (57.0%); corticosteroids (52.0%); antidepressants (47.7%); anticonvulsants (46.7%); anxiolytics, sedatives, or hypnotics (43.2%); spasticity medications (36.2%); and muscle relaxants (35.4%).

Most comorbidities and many medications, including bladder and antifatigue medications, were more common among patients aged ≥ 55 years. Hyperlipidemia, hypertension, and diabetes mellitus were more likely in males than in females. Females were more likely to have gastrointestinal disease, depression, thyroid disease, anxiety, lung disease, and arthritis. In addition, females were more likely than males to use many of the concomitant medications.

Dr. Nicholas disclosed grant support from EMD Serono. A coauthor is an employee of Health Services Consulting Corporation and received funding from EMD Serono to conduct the study. Other coauthors are employees of EMD Serono.

Jake Remaly, MDedge.com/neurology

 

 

In the 12 months after diagnosis, pain, fatigue, depression, and anxiety are common among patients with multiple sclerosis (MS), researchers reported at the 2019 meeting of the Consortium of Multiple Sclerosis Centers. In a novel study, about half of patients with MS reported clinically significant symptoms of depression or pain, and about 60% reported fatigue during that time.

Pain, fatigue, depression, and anxiety are common in MS, but their prevalence in the first year after diagnosis is not well understood. To examine the rates of these conditions and how often they co-occur during that period, Anna L. Kratz, PhD, associate professor of physical medicine and rehabilitation at the University of Michigan in Ann Arbor, and her research colleagues had 231 adults with MS complete validated surveys at 1, 2, 3, 6, 9, and 12 months after diagnosis to assess symptoms of these conditions.

Overall, 47.2% of patients reported clinically significant levels of depression, 38.5% reported clinically significant levels of anxiety, 50.4% reported clinically significant pain, and 62.2% reported clinically significant fatigue at any point during the year after diagnosis. “Of those who did not have clinically significant symptoms at time of diagnosis, 21.3% went on to develop clinically significant depression, 17.0% anxiety, 30.9% pain, and 34.1% fatigue,” the authors reported.

About 23% of patients did not have clinically significant symptoms for any condition, while 20% had clinically significant symptoms for 1 condition, 21% for 2, 19% for 3, and 17% for all 4. Depression and fatigue had the highest rate of comorbidity, whereas pain and anxiety had the lowest rate of comorbidity.

“Important clinical symptoms associated with MS are present at high levels in the first year post diagnosis,” Dr. Kratz and colleagues concluded. “While the rates and severity are marginally lower than have been identified in studies of individuals farther into the MS disease course, this study is a reminder that early MS intervention should incorporate interventions for these symptoms that are known to have strong associations with quality of life.”

The researchers had no disclosures.

Jake Remaly, MDedge.com/neurology

 

 

An international group of experts has proposed a new name, staging criteria, and recommendations for a recently recognized brain disorder that mimics Alzheimer disease and is marked by a proteinopathy caused by malformed transactive response DNA-binding protein of 43 kDa (TDP-43).

The term limbic-predominant age-related TDP-43 encephalopathy (LATE) was coined in an effort to raise awareness and kick-start research into this “pathway to dementia,” the experts wrote in a report appearing in Brain. “As there is currently no universally agreed-upon terminology or staging system for common age-related TDP-43 proteinopathy, this condition is understudied and not well recognized, even among investigators in the field of dementia research,” wrote the authors of the report, led by Peter T. Nelson, MD, PhD, of the University of Kentucky, Lexington.

LATE neuropathologic changes, associated with a progressive amnesia syndrome that mimics Alzheimer, are seen in > 20% of individuals aged > 80 years, according to large, community-based autopsy series. It coexists with Alzheimer disease in many patients, lowering the threshold for developing dementia, authors said.

The term LATE is designed to encompass several other terms related to TDP-43 pathology, including hippocampal sclerosis and cerebral age-related TDP-43 with sclerosis, Dr. Nelson and colleagues noted.

The TDP-43 protein is encoded by the TARDBP gene and provides several functions related to the regulation of gene expression, the authors wrote.

Misfolded TDP-43 was known to play a causative role in amyotrophic lateral sclerosis and frontotemporal lobar degeneration, the authors noted, and then was also identified in the brains of older individuals with hippocampal sclerosis or Alzheimer disease neuropathologic changes.

The authors proposed a 3-stage classification system for LATE neuropathologic change based on TDP-43 immunohistochemistry performed during routine autopsy evaluation of the amygdala, hippocampus, and middle frontal gyrus. The amygdala is an area affected early in the course of the disease (Stage 1), whereas involvement of the hippocampus represents a more intermediate stage (Stage 2), and the middle frontal gyrus is more affected in advanced stages of the disease (Stage 3), according to the schema.

Five genes have been identified with risk alleles for LATE neuropathologic changes, authors said. Of note, several groups have found that the apolipoprotein E ∑ 4 allele, known to be a risk factor for Alzheimer disease neuropathologic changes and Lewy body disease, is also linked to increased risk of TDP-43 proteinopathy.

There are no established biomarkers specific to TDP-43 proteinopathy yet, which hampers development of clinical trials designed to test interventions to treat or prevent LATE, Dr. Nelson and colleagues said in their report. LATE also could obscure the effects of potentially disease-modifying agents being tested in Alzheimer disease clinical trials, which can complicate the interpretation of study results, they added.

“Until there are biomarkers for LATE, clinical trials should be powered to account for TDP-43 proteinopathy,” they wrote. Dr. Nelson and coauthors reported no author disclosures.

Source: Nelson PT, Dickson DW, Trojanowski JQ, et al. Limbic-predominant age-related TDP-43 encephalopathy (LATE): consensus working group report. Brain. 2019;142(6):1503-1527.

Andrew D. Bowser, MDedge.com/neurology

Article PDF
Issue
Federal Practitioner - 36(4)s
Publications
Topics
Page Number
S8-S10
Sections
Article PDF
Article PDF

While several dozen studies have been conducted into cannabis-based treatments for symptoms of multiple sclerosis (MS), a new systematic review deems most to be of fair to poor quality. Reviewers found modest evidence of benefit and plenty of room for more research.

“Cannabis-based medicine may be useful for refractory MS symptoms, especially spasticity and pain, and side effects are usually well tolerated,” study lead author Natasha Breward, a graduate student at the College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, said in an interview. Breward spoke prior to the presentation of the study findings at the 2019 meeting of the Consortium of Multiple Sclerosis Centers.

For the review, Breward and colleagues focused on 60 studies—26 randomized controlled trials and 34 trials with other designs. Forty of the studies used nabiximols, an oromucosal spray that is derived from the cannabis sativa plant and approved for use in multiple countries but not yet in the US.

According to Breward, some of the other treatments included dried cannabis that is smoked or eaten and cannabidiol that’s typically delivered with tetrahydrocannabinol (THC) either oromucosally or as an oral capsule.

MS symptoms treated in the studies included spasticity (n = 29), pain (n = 8), and cognition (n = 6). The researchers considered 22 studies to be poor quality, 14 to be fair quality, and 24 to be good/excellent quality.

The researchers found that the cannabis-based medicine “significantly reduced spasticity and pain in several individual good-quality studies,” Breward said. The drugs seem to work by inhibiting neurotransmitter release via cannabinoids. “However, the variability in study quality—and in the products and regimens studied—make it hard to draw any conclusions about specific products and doses that may have the most potential benefit,” she added.

“Further research should focus on the use of different products and formulations of cannabis-based medicine such as cannabis oil and cannabidiol-prominent products, as no studies have focused on this area,” she said. “Research should also look at the potential of cannabis-based medicine for the treatment of disease progression, as cannabinoids are anti-inflammatory and immunomodulatory. Finally, more research regarding the potentially synergistic effects of cannabis-based medicine administered with current MS medications would also be useful.”

Randy Dotinga, MDedge.com/neurology

 

 

Brain volumes of specific regions of interest can be used to classify traumatic brain injury subjects that fall into predetermined symptom categories, according to a study presented at the annual meeting of the American Academy of Neurology.

Traumatic brain injury (TBI) damages brain tissue and causes subsequent volume loss, which may result in clinical symptoms. It is a prevalent worldwide health problem caused by a mechanical insult to the head, resulting in transient or permanent alteration to brain tissue and/or function. Standard neuroimaging with computed cranial tomography (CT) and structural magnetic resonance imaging (MRI) is often unrevealing during the evaluation of patients with TBI, particularly those classified as mild TBI.

In this study, James Rock, MD, of Penn Presbyterian Medical Center and the University of Pennsylvania, and colleagues sought to examine the value of quantitative analysis of regional brain volumes in the evaluation of TBI. The investigators reviewed the medical records and MRI imaging from 44 patients with TBI evaluated at a Level I trauma center. They also read clinical notes to assess reported symptoms and physical findings.

Regional volumes from TBI subjects were derived using the software package Freesurfer image analysis suite (surfer.nmr.mgh.harvard.edu), which utilizes a T1-weighted structural scan to calculate volumetric information. A machine learning algorithm, random forests, was employed across volume measurements from 25 regions of interest to determine the most important regions for classifying subjects based on clinical outcome and symptomology.

Basal ganglia volume showed the highest variable importance with regards to classifying subjects who exhibited symptoms of cognitive dysfunction in quantitative analysis. Left lateral ventricle volume was important in classifying subjects with motor and vestibular alterations. Left choroid plexus volume was the most important region for classifying subjects with sensation and somatic dysfunction.

In an abstract, the researchers noted that their study is ongoing. “It will be extended to a larger cohort to determine whether volume changes in specific [regions of interest] can act as useful clinical biomarkers for chronic symptoms,” they said.

Dr. Diaz-Arrastia received personal compensation from Neural Analytics, Inc; BrainBox Solutions, Inc; and Bioscience Pharma Partners. Dr. Diaz-Arrastia holds stock and/or stock options in Neural Analytics, Inc and has received research support from BrainBox Solutions. The other authors reported no other disclosures.

Glenn S. Williams, MDedge.com/neurology

 

 

Concomitant medication use is common when patients with multiple sclerosis (MS) start disease-modifying drugs (DMDs), according to research presented at the 2019 meeting of the Consortium of Multiple Sclerosis Centers. The likelihood of particular comorbidities and concomitant medications varies by age and sex, researchers reported.

“This may have implications for MS treatment,” said study author Jacqueline Nicholas, MD, MPH, of Ohio Multiple Sclerosis Center in Columbus and colleagues. “A better understanding of the effects of comorbidities and concomitant medications on the effectiveness and safety of DMDs is needed to support clinical decision making.”

Researchers have examined comorbidities in patients with MS, but concomitant medication use among patients starting DMDs is poorly understood, the authors said.

To study this question, Dr. Nicholas and colleagues analyzed retrospective administrative claims data from IQVIA Real-World Data Adjudicated Claims–US database from Jan. 1, 2010, to June 30, 2017. Their analysis included patients with ≥ 2 MS diagnosis claims and at least 1 DMD claim between Jan. 1, 2011, and June 30, 2015. Eligible patients were aged 18 to 63 years and had continuous eligibility with commercial insurance 1 year before and 2 years after DMD initiation. In addition, patients had no evidence of DMD use during the 1-year baseline period.

The investigators used International Classification of Diseases, 9th and 10th revisions, Clinical Modification codes and claims to evaluate patients’ comorbidities and concomitant medications during the study period.

The researchers identified 8,251 eligible patients. Patients had a mean age of 43.2 years, and 75.5% were female. Average baseline Charlson Comorbidity Index was 0.41. In the 2 years after DMD initiation, common comorbid diagnoses were hyperlipidemia (30.0%), hypertension (28.2%), gastrointestinal disorders (26.2%), depression (25.5%), and anxiety (20.1%).

Common concomitant medications included antibiotics (70.6%); analgesics (57.0%); corticosteroids (52.0%); antidepressants (47.7%); anticonvulsants (46.7%); anxiolytics, sedatives, or hypnotics (43.2%); spasticity medications (36.2%); and muscle relaxants (35.4%).

Most comorbidities and many medications, including bladder and antifatigue medications, were more common among patients aged ≥ 55 years. Hyperlipidemia, hypertension, and diabetes mellitus were more likely in males than in females. Females were more likely to have gastrointestinal disease, depression, thyroid disease, anxiety, lung disease, and arthritis. In addition, females were more likely than males to use many of the concomitant medications.

Dr. Nicholas disclosed grant support from EMD Serono. A coauthor is an employee of Health Services Consulting Corporation and received funding from EMD Serono to conduct the study. Other coauthors are employees of EMD Serono.

Jake Remaly, MDedge.com/neurology

 

 

In the 12 months after diagnosis, pain, fatigue, depression, and anxiety are common among patients with multiple sclerosis (MS), researchers reported at the 2019 meeting of the Consortium of Multiple Sclerosis Centers. In a novel study, about half of patients with MS reported clinically significant symptoms of depression or pain, and about 60% reported fatigue during that time.

Pain, fatigue, depression, and anxiety are common in MS, but their prevalence in the first year after diagnosis is not well understood. To examine the rates of these conditions and how often they co-occur during that period, Anna L. Kratz, PhD, associate professor of physical medicine and rehabilitation at the University of Michigan in Ann Arbor, and her research colleagues had 231 adults with MS complete validated surveys at 1, 2, 3, 6, 9, and 12 months after diagnosis to assess symptoms of these conditions.

Overall, 47.2% of patients reported clinically significant levels of depression, 38.5% reported clinically significant levels of anxiety, 50.4% reported clinically significant pain, and 62.2% reported clinically significant fatigue at any point during the year after diagnosis. “Of those who did not have clinically significant symptoms at time of diagnosis, 21.3% went on to develop clinically significant depression, 17.0% anxiety, 30.9% pain, and 34.1% fatigue,” the authors reported.

About 23% of patients did not have clinically significant symptoms for any condition, while 20% had clinically significant symptoms for 1 condition, 21% for 2, 19% for 3, and 17% for all 4. Depression and fatigue had the highest rate of comorbidity, whereas pain and anxiety had the lowest rate of comorbidity.

“Important clinical symptoms associated with MS are present at high levels in the first year post diagnosis,” Dr. Kratz and colleagues concluded. “While the rates and severity are marginally lower than have been identified in studies of individuals farther into the MS disease course, this study is a reminder that early MS intervention should incorporate interventions for these symptoms that are known to have strong associations with quality of life.”

The researchers had no disclosures.

Jake Remaly, MDedge.com/neurology

 

 

An international group of experts has proposed a new name, staging criteria, and recommendations for a recently recognized brain disorder that mimics Alzheimer disease and is marked by a proteinopathy caused by malformed transactive response DNA-binding protein of 43 kDa (TDP-43).

The term limbic-predominant age-related TDP-43 encephalopathy (LATE) was coined in an effort to raise awareness and kick-start research into this “pathway to dementia,” the experts wrote in a report appearing in Brain. “As there is currently no universally agreed-upon terminology or staging system for common age-related TDP-43 proteinopathy, this condition is understudied and not well recognized, even among investigators in the field of dementia research,” wrote the authors of the report, led by Peter T. Nelson, MD, PhD, of the University of Kentucky, Lexington.

LATE neuropathologic changes, associated with a progressive amnesia syndrome that mimics Alzheimer, are seen in > 20% of individuals aged > 80 years, according to large, community-based autopsy series. It coexists with Alzheimer disease in many patients, lowering the threshold for developing dementia, authors said.

The term LATE is designed to encompass several other terms related to TDP-43 pathology, including hippocampal sclerosis and cerebral age-related TDP-43 with sclerosis, Dr. Nelson and colleagues noted.

The TDP-43 protein is encoded by the TARDBP gene and provides several functions related to the regulation of gene expression, the authors wrote.

Misfolded TDP-43 was known to play a causative role in amyotrophic lateral sclerosis and frontotemporal lobar degeneration, the authors noted, and then was also identified in the brains of older individuals with hippocampal sclerosis or Alzheimer disease neuropathologic changes.

The authors proposed a 3-stage classification system for LATE neuropathologic change based on TDP-43 immunohistochemistry performed during routine autopsy evaluation of the amygdala, hippocampus, and middle frontal gyrus. The amygdala is an area affected early in the course of the disease (Stage 1), whereas involvement of the hippocampus represents a more intermediate stage (Stage 2), and the middle frontal gyrus is more affected in advanced stages of the disease (Stage 3), according to the schema.

Five genes have been identified with risk alleles for LATE neuropathologic changes, authors said. Of note, several groups have found that the apolipoprotein E ∑ 4 allele, known to be a risk factor for Alzheimer disease neuropathologic changes and Lewy body disease, is also linked to increased risk of TDP-43 proteinopathy.

There are no established biomarkers specific to TDP-43 proteinopathy yet, which hampers development of clinical trials designed to test interventions to treat or prevent LATE, Dr. Nelson and colleagues said in their report. LATE also could obscure the effects of potentially disease-modifying agents being tested in Alzheimer disease clinical trials, which can complicate the interpretation of study results, they added.

“Until there are biomarkers for LATE, clinical trials should be powered to account for TDP-43 proteinopathy,” they wrote. Dr. Nelson and coauthors reported no author disclosures.

Source: Nelson PT, Dickson DW, Trojanowski JQ, et al. Limbic-predominant age-related TDP-43 encephalopathy (LATE): consensus working group report. Brain. 2019;142(6):1503-1527.

Andrew D. Bowser, MDedge.com/neurology

While several dozen studies have been conducted into cannabis-based treatments for symptoms of multiple sclerosis (MS), a new systematic review deems most to be of fair to poor quality. Reviewers found modest evidence of benefit and plenty of room for more research.

“Cannabis-based medicine may be useful for refractory MS symptoms, especially spasticity and pain, and side effects are usually well tolerated,” study lead author Natasha Breward, a graduate student at the College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, said in an interview. Breward spoke prior to the presentation of the study findings at the 2019 meeting of the Consortium of Multiple Sclerosis Centers.

For the review, Breward and colleagues focused on 60 studies—26 randomized controlled trials and 34 trials with other designs. Forty of the studies used nabiximols, an oromucosal spray that is derived from the cannabis sativa plant and approved for use in multiple countries but not yet in the US.

According to Breward, some of the other treatments included dried cannabis that is smoked or eaten and cannabidiol that’s typically delivered with tetrahydrocannabinol (THC) either oromucosally or as an oral capsule.

MS symptoms treated in the studies included spasticity (n = 29), pain (n = 8), and cognition (n = 6). The researchers considered 22 studies to be poor quality, 14 to be fair quality, and 24 to be good/excellent quality.

The researchers found that the cannabis-based medicine “significantly reduced spasticity and pain in several individual good-quality studies,” Breward said. The drugs seem to work by inhibiting neurotransmitter release via cannabinoids. “However, the variability in study quality—and in the products and regimens studied—make it hard to draw any conclusions about specific products and doses that may have the most potential benefit,” she added.

“Further research should focus on the use of different products and formulations of cannabis-based medicine such as cannabis oil and cannabidiol-prominent products, as no studies have focused on this area,” she said. “Research should also look at the potential of cannabis-based medicine for the treatment of disease progression, as cannabinoids are anti-inflammatory and immunomodulatory. Finally, more research regarding the potentially synergistic effects of cannabis-based medicine administered with current MS medications would also be useful.”

Randy Dotinga, MDedge.com/neurology

 

 

Brain volumes of specific regions of interest can be used to classify traumatic brain injury subjects that fall into predetermined symptom categories, according to a study presented at the annual meeting of the American Academy of Neurology.

Traumatic brain injury (TBI) damages brain tissue and causes subsequent volume loss, which may result in clinical symptoms. It is a prevalent worldwide health problem caused by a mechanical insult to the head, resulting in transient or permanent alteration to brain tissue and/or function. Standard neuroimaging with computed cranial tomography (CT) and structural magnetic resonance imaging (MRI) is often unrevealing during the evaluation of patients with TBI, particularly those classified as mild TBI.

In this study, James Rock, MD, of Penn Presbyterian Medical Center and the University of Pennsylvania, and colleagues sought to examine the value of quantitative analysis of regional brain volumes in the evaluation of TBI. The investigators reviewed the medical records and MRI imaging from 44 patients with TBI evaluated at a Level I trauma center. They also read clinical notes to assess reported symptoms and physical findings.

Regional volumes from TBI subjects were derived using the software package Freesurfer image analysis suite (surfer.nmr.mgh.harvard.edu), which utilizes a T1-weighted structural scan to calculate volumetric information. A machine learning algorithm, random forests, was employed across volume measurements from 25 regions of interest to determine the most important regions for classifying subjects based on clinical outcome and symptomology.

Basal ganglia volume showed the highest variable importance with regards to classifying subjects who exhibited symptoms of cognitive dysfunction in quantitative analysis. Left lateral ventricle volume was important in classifying subjects with motor and vestibular alterations. Left choroid plexus volume was the most important region for classifying subjects with sensation and somatic dysfunction.

In an abstract, the researchers noted that their study is ongoing. “It will be extended to a larger cohort to determine whether volume changes in specific [regions of interest] can act as useful clinical biomarkers for chronic symptoms,” they said.

Dr. Diaz-Arrastia received personal compensation from Neural Analytics, Inc; BrainBox Solutions, Inc; and Bioscience Pharma Partners. Dr. Diaz-Arrastia holds stock and/or stock options in Neural Analytics, Inc and has received research support from BrainBox Solutions. The other authors reported no other disclosures.

Glenn S. Williams, MDedge.com/neurology

 

 

Concomitant medication use is common when patients with multiple sclerosis (MS) start disease-modifying drugs (DMDs), according to research presented at the 2019 meeting of the Consortium of Multiple Sclerosis Centers. The likelihood of particular comorbidities and concomitant medications varies by age and sex, researchers reported.

“This may have implications for MS treatment,” said study author Jacqueline Nicholas, MD, MPH, of Ohio Multiple Sclerosis Center in Columbus and colleagues. “A better understanding of the effects of comorbidities and concomitant medications on the effectiveness and safety of DMDs is needed to support clinical decision making.”

Researchers have examined comorbidities in patients with MS, but concomitant medication use among patients starting DMDs is poorly understood, the authors said.

To study this question, Dr. Nicholas and colleagues analyzed retrospective administrative claims data from IQVIA Real-World Data Adjudicated Claims–US database from Jan. 1, 2010, to June 30, 2017. Their analysis included patients with ≥ 2 MS diagnosis claims and at least 1 DMD claim between Jan. 1, 2011, and June 30, 2015. Eligible patients were aged 18 to 63 years and had continuous eligibility with commercial insurance 1 year before and 2 years after DMD initiation. In addition, patients had no evidence of DMD use during the 1-year baseline period.

The investigators used International Classification of Diseases, 9th and 10th revisions, Clinical Modification codes and claims to evaluate patients’ comorbidities and concomitant medications during the study period.

The researchers identified 8,251 eligible patients. Patients had a mean age of 43.2 years, and 75.5% were female. Average baseline Charlson Comorbidity Index was 0.41. In the 2 years after DMD initiation, common comorbid diagnoses were hyperlipidemia (30.0%), hypertension (28.2%), gastrointestinal disorders (26.2%), depression (25.5%), and anxiety (20.1%).

Common concomitant medications included antibiotics (70.6%); analgesics (57.0%); corticosteroids (52.0%); antidepressants (47.7%); anticonvulsants (46.7%); anxiolytics, sedatives, or hypnotics (43.2%); spasticity medications (36.2%); and muscle relaxants (35.4%).

Most comorbidities and many medications, including bladder and antifatigue medications, were more common among patients aged ≥ 55 years. Hyperlipidemia, hypertension, and diabetes mellitus were more likely in males than in females. Females were more likely to have gastrointestinal disease, depression, thyroid disease, anxiety, lung disease, and arthritis. In addition, females were more likely than males to use many of the concomitant medications.

Dr. Nicholas disclosed grant support from EMD Serono. A coauthor is an employee of Health Services Consulting Corporation and received funding from EMD Serono to conduct the study. Other coauthors are employees of EMD Serono.

Jake Remaly, MDedge.com/neurology

 

 

In the 12 months after diagnosis, pain, fatigue, depression, and anxiety are common among patients with multiple sclerosis (MS), researchers reported at the 2019 meeting of the Consortium of Multiple Sclerosis Centers. In a novel study, about half of patients with MS reported clinically significant symptoms of depression or pain, and about 60% reported fatigue during that time.

Pain, fatigue, depression, and anxiety are common in MS, but their prevalence in the first year after diagnosis is not well understood. To examine the rates of these conditions and how often they co-occur during that period, Anna L. Kratz, PhD, associate professor of physical medicine and rehabilitation at the University of Michigan in Ann Arbor, and her research colleagues had 231 adults with MS complete validated surveys at 1, 2, 3, 6, 9, and 12 months after diagnosis to assess symptoms of these conditions.

Overall, 47.2% of patients reported clinically significant levels of depression, 38.5% reported clinically significant levels of anxiety, 50.4% reported clinically significant pain, and 62.2% reported clinically significant fatigue at any point during the year after diagnosis. “Of those who did not have clinically significant symptoms at time of diagnosis, 21.3% went on to develop clinically significant depression, 17.0% anxiety, 30.9% pain, and 34.1% fatigue,” the authors reported.

About 23% of patients did not have clinically significant symptoms for any condition, while 20% had clinically significant symptoms for 1 condition, 21% for 2, 19% for 3, and 17% for all 4. Depression and fatigue had the highest rate of comorbidity, whereas pain and anxiety had the lowest rate of comorbidity.

“Important clinical symptoms associated with MS are present at high levels in the first year post diagnosis,” Dr. Kratz and colleagues concluded. “While the rates and severity are marginally lower than have been identified in studies of individuals farther into the MS disease course, this study is a reminder that early MS intervention should incorporate interventions for these symptoms that are known to have strong associations with quality of life.”

The researchers had no disclosures.

Jake Remaly, MDedge.com/neurology

 

 

An international group of experts has proposed a new name, staging criteria, and recommendations for a recently recognized brain disorder that mimics Alzheimer disease and is marked by a proteinopathy caused by malformed transactive response DNA-binding protein of 43 kDa (TDP-43).

The term limbic-predominant age-related TDP-43 encephalopathy (LATE) was coined in an effort to raise awareness and kick-start research into this “pathway to dementia,” the experts wrote in a report appearing in Brain. “As there is currently no universally agreed-upon terminology or staging system for common age-related TDP-43 proteinopathy, this condition is understudied and not well recognized, even among investigators in the field of dementia research,” wrote the authors of the report, led by Peter T. Nelson, MD, PhD, of the University of Kentucky, Lexington.

LATE neuropathologic changes, associated with a progressive amnesia syndrome that mimics Alzheimer, are seen in > 20% of individuals aged > 80 years, according to large, community-based autopsy series. It coexists with Alzheimer disease in many patients, lowering the threshold for developing dementia, authors said.

The term LATE is designed to encompass several other terms related to TDP-43 pathology, including hippocampal sclerosis and cerebral age-related TDP-43 with sclerosis, Dr. Nelson and colleagues noted.

The TDP-43 protein is encoded by the TARDBP gene and provides several functions related to the regulation of gene expression, the authors wrote.

Misfolded TDP-43 was known to play a causative role in amyotrophic lateral sclerosis and frontotemporal lobar degeneration, the authors noted, and then was also identified in the brains of older individuals with hippocampal sclerosis or Alzheimer disease neuropathologic changes.

The authors proposed a 3-stage classification system for LATE neuropathologic change based on TDP-43 immunohistochemistry performed during routine autopsy evaluation of the amygdala, hippocampus, and middle frontal gyrus. The amygdala is an area affected early in the course of the disease (Stage 1), whereas involvement of the hippocampus represents a more intermediate stage (Stage 2), and the middle frontal gyrus is more affected in advanced stages of the disease (Stage 3), according to the schema.

Five genes have been identified with risk alleles for LATE neuropathologic changes, authors said. Of note, several groups have found that the apolipoprotein E ∑ 4 allele, known to be a risk factor for Alzheimer disease neuropathologic changes and Lewy body disease, is also linked to increased risk of TDP-43 proteinopathy.

There are no established biomarkers specific to TDP-43 proteinopathy yet, which hampers development of clinical trials designed to test interventions to treat or prevent LATE, Dr. Nelson and colleagues said in their report. LATE also could obscure the effects of potentially disease-modifying agents being tested in Alzheimer disease clinical trials, which can complicate the interpretation of study results, they added.

“Until there are biomarkers for LATE, clinical trials should be powered to account for TDP-43 proteinopathy,” they wrote. Dr. Nelson and coauthors reported no author disclosures.

Source: Nelson PT, Dickson DW, Trojanowski JQ, et al. Limbic-predominant age-related TDP-43 encephalopathy (LATE): consensus working group report. Brain. 2019;142(6):1503-1527.

Andrew D. Bowser, MDedge.com/neurology

Issue
Federal Practitioner - 36(4)s
Issue
Federal Practitioner - 36(4)s
Page Number
S8-S10
Page Number
S8-S10
Publications
Publications
Topics
Article Type
Display Headline
Research News: Neurologic Disorders
Display Headline
Research News: Neurologic Disorders
Sections
Citation Override
Fed Pract. 2019 June;36(suppl 4):S8-S10
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.
Article PDF Media

Proton Pump Inhibitor Use and Risk of Dementia in the Veteran Population (FULL)

Article Type
Changed
Mon, 06/01/2020 - 15:23
Display Headline
Proton Pump Inhibitor Use and Risk of Dementia in the Veteran Population
There was a statistically significant association between use of proton pump inhibitors and dementia diagnosis in a 11-year retrospective study of patients at the Sioux Falls Veteran Affairs Health Care System.

Proton pump inhibitors (PPIs) have become the mainstay of therapy in the treatment of acid-related disorders since their introduction in 1989. Due to their high potency, excellent tolerability, and generic availability, PPIs have largely replaced histamine-2 receptor antagonists for gastric problems. Since they were first released on the market, the use of PPIs has continued to rise in both the hospital and primary care settings.1 However, this rapid growth has led to the concern of overutilization. A study conducted at the Department of Veterans Affairs (VA) Ann Arbor Health Care System found that out of 946 patients in the ambulatory care setting taking PPIs, only 35% were appropriately prescribed PPIs.2

Although the short-term adverse effects of PPI use seem minimal, chronic PPI use consequences are a growing concern. Chronic PPI use is associated with increased risks of osteoporosis, pneumonia, and Clostridium difficile infections.3 Another long-term risk that has been associated with chronic PPI use is dementia. Dementia is a cognitive syndrome that is characterized by a progressive decline beyond what is expected in normal aging in 1 or more of the cognitive domains of memory, language, orientation, learning capacity, executive function, or social cognition.4 Because it interferes with activities of daily living, dementia is a major cause of disability in the elderly and is an immense burden for caregivers. Currently, about 47 million people globally live with dementia.5 This number is projected to nearly triple by 2050 to 132 million.5 With no cure, identification of risk factors and creation of protective measures are critical in decreasing the prevalence of dementia.

Although the exact pathophysiology behind the link between PPIs and dementia is unknown, several theories exist. One such theory is that PPI-induced vitamin B12 deficiency leads to cognitive decline.6,7 Another theory suggests that PPIs can directly cause dementia by inhibiting enzymes that normally degrade β amyloid.8 This leads to increased levels of β-amyloid plaques, which is a known characteristic of dementia patients. This theory is derived from animal studies that have shown increased amyloid levels in the brains of mice given PPIs.8

Current studies are conflicting regarding the association between PPIs and dementia. Two German prospective, cohort studies found statistically significant increased risks of dementia in patients taking PPIs with hazard ratios (HR) of 1.38 (95% CI, 1.04-1.83) and 1.44 (95% CI, 1.36-1.52), respectively.9,10 A study conducted in Taiwan also found an increased risk of dementia among PPI users with a HR of 1.22 (95% CI, 1.05-1.42).11 On the contrary, other studies have failed to show an increased risk of dementia with PPI use. In fact, Goldstein and colleagues found a decreased risk of dementia in PPI users with a HR of 0.78 (95% CI, 0.76-0.93).12 This study was an observational study conducted in the US using data from the National Alzheimer’s Coordinating Center database.12 Another recent retrospective study conducted in Finland showed that PPI use was not associated with a significantly increased risk of Alzheimer disease.13

Much is unknown about the cause of dementia, and no curative treatment exists. Investigation into potential risk factors for dementia can lead to the development of preventative measures, which can lead to significant improvement in quality of life for both patients and caregivers. Current studies regarding the association between PPIs and dementia are conflicting, and to our knowledge, no study analyzing the effects of PPIs and dementia has been conducted within the veteran population specifically. The objective of the current study is to investigate the association between PPI use and dementia in the veteran population.

Methods

This study is a retrospective, cohort, single-center, chart review study conducted at the Sioux Falls Veteran Affairs Health Care System (SFVAHCS). Data were extracted from the VA electronic health record (EHR) from January 1, 2005 through December 31, 2015. The study included both currently living and deceased veterans who received ≥ 2 documented outpatient visits at the SFVAHCS during the study time frame. Patients also had to be aged ≥ 60 years at the start of the study period. Patients were excluded if they received only a ≤ 30-day PPI prescription. Patients with dementia related to head trauma, acute intoxication, or other known diseases were excluded.

 

 

To analyze the primary endpoint of association between PPI use and dementia, the study compared the rate of dementia in a cohort of veterans who had received an outpatient prescription for a PPI within the study time frame vs the rate of dementia in a random, equal number of veterans who had never been prescribed PPIs within the study time frame. In this study, veterans were classified as having dementia if they had a diagnosis of dementia based on ICD-9 or ICD-10 codes (Table 1), or if they had been prescribed medications used to treat dementia (donepezil, ergoloid mesylates, galantamine, memantine, and rivastigmine).

Secondary endpoints included analysis of the effects of PPI agent, PPI dose, and PPI duration on the risk of dementia. For the PPI dose analysis, cumulative doses were converted into defined daily doses (DDDs) using the World Health Organization calculation to equalize the different potencies of PPI agents (Table 2).14 In addition, the effect of PPI use on vitamin B12 levels was analyzed as an exploratory endpoint to investigate the hypothesis that PPI may be associated with vitamin B12 deficiency, which in turn may be associated with dementia.6,7

Baseline characteristics were collected to determine the variability between the treatment and control group. Data collected included age, gender, past medical history of diseases that may increase risk of dementia, and anticholinergic drug use. Anticholinergic drugs were included if they were classified as having “definite anticholinergic effects” based on the Aging Brain Care Anticholinergic Burden Scale (Appendix).15

 

Statistical Analysis

The primary endpoint was analyzed using a χ2 for association test. For the secondary endpoints, a χ2 for association test was used for endpoints with nominal data, and the Mood median test was used for endpoints with continuous data. The exploratory endpoint analyzing vitamin B12 levels was analyzed with the Mood median test. A P value of < .05 was defined as being statistically significant. Power analysis was not performed since all veterans who met the criteria were included in the study.

Results

Records of 23,656 veterans were included in the study with 11,828 veterans in both the PPI cohort and the non-PPI cohort (Table 3).

Primary Endpoint

Within the PPI group, 1,119 (9.5%) veterans had dementia compared with only 740 (6.3%) veterans in the non-PPI group. There was a statistically significant association between PPI use and dementia (P < .001). These results yielded an odds ratio of 1.55 for dementia risk in PPI users vs nonusers and a relative risk increase of 51.4% for dementia risk with PPI use compared with no PPI use.

 

 

Secondary Endpoints

Users of rabeprazole had the highest rate of dementia (12.8%), followed by lansoprazole (10.9%), omeprazole (9.7%), esomeprazole (7.7%), and pantoprazole (7.0%). The rate of dementia for non-PPI users was 6.3% (P < .001). The median cumulative doses of PPIs were not significant: 597 DDDs (95% CI, 540-630) in the dementia group vs 570 DDDs (95% CI, 540-624) in the nondementia group (P = .79). The median cumulative duration of PPI use in the dementia group was 4.6 years (95% CI, 4.25-4.92) vs 5.3 years (95% CI, 5.08-5.42) in the nondementia group (P < .001).

Exploratory Endpoint

The median B12 level in the PPI group was 521 pg/mL (95% CI, 509-533) compared with 480 pg/mL (95% CI, 465-496) in the non-PPI group (P < .001). However, both groups fell within the normal range for vitamin B12 (200-900 pg/mL).16

Discussion

The aim of this study was to determine whether an association existed between PPI use and dementia. This study showed a statistically significant association between PPI use and dementia within the veteran population. This study also showed a significant association between specific PPI agents and dementia. When analyzing the individual PPI agents, the rabeprazole group yielded the strongest relationship. However, this study was not powered to evaluate and compare risks of dementia between individual PPI agents. More data are needed to determine statistical and clinical significance of associations between individual PPI agents and risk of dementia.

The veterans with dementia had a higher median cumulative PPI dose than did the veterans without dementia; however, the results were not statistically significant. Therefore, the data cannot correlate higher doses of PPI use to increased risk of dementia.

The cumulative duration of PPI use was statistically significant but opposite of the expected outcome. The dementia group had a lower median lifetime duration of PPI use compared with that of the nondementia group. It is difficult to determine the reason for this outcome, but it seems that for this study population, a longer duration of PPI use was not associated with an increased risk of dementia.

Finally, the exploratory endpoint analyzed vitamin B12 levels, since it has been shown that PPI use can lead to vitamin B12 deficiency and that B12 deficiency can lead to dementia.6-8 This study found that the dementia group had significantly higher vitamin B12 levels than the nondementia group. These data suggest that PPI use may not be associated with vitamin B12 deficiency. However, it is important to note that this study was unable to collect data on the use of vitamin B12 supplementation due to the unreliability of over-the-counter (OTC) and non-VA medication use records. Therefore, it is possible that the PPI group had higher rates of B12 deficiency but were effectively treated with B12 supplementation. More research is needed to determine the exact relationship between PPI use, vitamin B12 deficiency, and dementia risk.

Strengths/Limitations

Strengths of this study that support its findings include the large population size. Additionally, the use of the VA EHR allowed for a complete drug dispensing history to be collected, which improves reliability of the data.

 

 

This study also had some limitations. First, the causal relationship of PPI use and dementia cannot be proven using a retrospective cohort design. This study’s design can show association, but it cannot prove causation. Also, due to the retrospective design, exposure to PPI use could not be randomized; thus, correlation between PPI use and dementia may be explained by confounding variables that are not captured within this study. This is especially true since the baseline characteristics were not equally distributed between the 2 groups. In fact, the PPI group had higher rates of many clinical comorbidities. This imbalance may have skewed the results of the primary endpoint. Lastly, OTC PPI use and non-VA PPI prescriptions were not available. Therefore, some of the patients included in the non-PPI group may have been PPI users if they received PPIs from OTC or non-VA sources, which could skew the results.

Conclusion

This study showed a significant association between PPI use and dementia within the veteran study population. The study also showed a significant association between PPI use and dementia within the secondary endpoint of individual PPI agent. Higher cumulative dose and duration of PPI use did not seem to increase risk of dementia. Finally, PPI use was not associated with significantly low vitamin B12 levels. More studies are needed to determine causation of dementia and its risk factors.

Acknowledgments
This material is the result of work supported with resources and the use of facilities at the Sioux Falls VA Health Care System.

References

1. Savarino V, Dulbecco P, de Bortoli N, Ottonello A, Savarino E. The appropriate use of proton pump inhibitors (PPIs): need for a reappraisal. Eur J Intern Med. 2017;37:19-24.

2. Heidelbaugh J, Goldberg K, Inadomi J. Magnitude and economic effect of overuse of antisecretory therapy in the ambulatory care setting. Am J Manag Care. 2010;16(9):e228-e234.

3. Heidelbaugh JJ, Kim AH, Chang R. Walker PC. Overutilization of proton-pump inhibitors: what the clinician needs to know. Therap Adv Gastroenterol. 2012;5(4):219-232.

4. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, (DSM-5). American Psychiatric Association: Washington, DC; 2013.

5. World Health Organization. Dementia. http://www.who.int/mediacentre/factsheets/fs362/en/. Published December 12, 2017. Accessed March 10, 2019.

6. Vogiatzoglou A, Smith AD, Nurk E, et al. Cognitive function in an elderly population: interaction between vitamin B12 status, depression, and apolipoprotein E ε4: the Hordaland Homocysteine Study. Psychosom Med. 2013;75(1):20-29.

7. Lam JR, Schneider JL, Zhao W, Corley DA. Proton pump inhibitor and histamine 2 receptor antagonist use and vitamin B12 deficiency. JAMA. 2013;310(22):2435-2442.

8. Badiola N, Alcalde V, Pujol A, et al. The proton-pump inhibitor lansoprazole enhances amyloid beta production. PLoS One. 2013;8(3):e58837.

9. Haenisch B, von Holt K, Wiese B, et al. Risk of dementia in elderly patients with the use of proton pump inhibitors. Eur Arch Psychiatry Clin Neurosci. 2015;265(5):419-428.

10. Gomm W, von Holt K, Thomé F, et al. Association between proton pump inhibitors with risk of dementia. A pharmacoepidemiological claims data analysis. JAMA Neurol. 2016;73(4):410-416.

11. Tai SY, Chien CY, Wu DC, et al. Risk of dementia from proton pump inhibitor use in Asian population: a nationwide cohort study in Taiwan. PLoS One. 2017;12(2):e0171006.

12. Goldstein FC, Steenland K, Zhao L, Wharton W, Levey AI, Hajjar I. Proton pump inhibitors and risk of mild cognitive impairment and dementia. J Am Geriatr Soc. 2017;65(9):1969-1674.

13. Taipale H, Tolppanen AM, Tiihonen M. Tanskanen A, Tiihonen J, Hartikainen S. No association between proton pump inhibitor use and risk of Alzheimer’s disease. Am J Gastroenterol. 2017;112(12):1801-1808.

14. World Health Organization Collaborating Centre for Drug Statistics Methodology. Definition and general considerations. https://www.whocc.no/ddd/definition_and_general_considera/. Updated February 7, 2018. Accessed March 13, 2019.

15. Indiana University Center for Aging Research, Aging Brain Program. Anticholinergic cognitive burden scale. http://www.idhca.org/wp-content/uploads/2018/02/DESAI_ACB_scale_-_Legal_size_paper.pdf. Updated 2012. Accessed March 10, 2019.

16. US National Library of Medicine, MedlinePlus. Vitamin B12 level. https://medlineplus.gov/ency/article/003705.htm. Updated March 7, 2019. Accessed March 13, 2019.

Article PDF
Author and Disclosure Information

Jenna Welu is a Pharmacy Resident, Justin Metzger is Chief of Pharmacy, Scott Bebensee is a Home-Based Primary Care Clinical Pharmacy Specialist, April Ahrendt is a Systems Redesigner, and Micheal Vasek is a Program Analyst, all at the Sioux Falls VA Health Care System in South Dakota. April Ahrendt is an Academic Assistant Professor at the University of South Dakota School of Health Sciences in Vermillion.
Correspondence: Jenna Welu (jenna.donnelly@ jacks.sdstate.edu)

Author disclosures
The authors report no actual or potential conflicts of interest with regard to this article.

Disclaimer
The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review the complete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

Issue
Federal Practitioner - 36(4)s
Publications
Topics
Page Number
S27-S31
Sections
Author and Disclosure Information

Jenna Welu is a Pharmacy Resident, Justin Metzger is Chief of Pharmacy, Scott Bebensee is a Home-Based Primary Care Clinical Pharmacy Specialist, April Ahrendt is a Systems Redesigner, and Micheal Vasek is a Program Analyst, all at the Sioux Falls VA Health Care System in South Dakota. April Ahrendt is an Academic Assistant Professor at the University of South Dakota School of Health Sciences in Vermillion.
Correspondence: Jenna Welu (jenna.donnelly@ jacks.sdstate.edu)

Author disclosures
The authors report no actual or potential conflicts of interest with regard to this article.

Disclaimer
The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review the complete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

Author and Disclosure Information

Jenna Welu is a Pharmacy Resident, Justin Metzger is Chief of Pharmacy, Scott Bebensee is a Home-Based Primary Care Clinical Pharmacy Specialist, April Ahrendt is a Systems Redesigner, and Micheal Vasek is a Program Analyst, all at the Sioux Falls VA Health Care System in South Dakota. April Ahrendt is an Academic Assistant Professor at the University of South Dakota School of Health Sciences in Vermillion.
Correspondence: Jenna Welu (jenna.donnelly@ jacks.sdstate.edu)

Author disclosures
The authors report no actual or potential conflicts of interest with regard to this article.

Disclaimer
The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review the complete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

Article PDF
Article PDF
Related Articles
There was a statistically significant association between use of proton pump inhibitors and dementia diagnosis in a 11-year retrospective study of patients at the Sioux Falls Veteran Affairs Health Care System.
There was a statistically significant association between use of proton pump inhibitors and dementia diagnosis in a 11-year retrospective study of patients at the Sioux Falls Veteran Affairs Health Care System.

Proton pump inhibitors (PPIs) have become the mainstay of therapy in the treatment of acid-related disorders since their introduction in 1989. Due to their high potency, excellent tolerability, and generic availability, PPIs have largely replaced histamine-2 receptor antagonists for gastric problems. Since they were first released on the market, the use of PPIs has continued to rise in both the hospital and primary care settings.1 However, this rapid growth has led to the concern of overutilization. A study conducted at the Department of Veterans Affairs (VA) Ann Arbor Health Care System found that out of 946 patients in the ambulatory care setting taking PPIs, only 35% were appropriately prescribed PPIs.2

Although the short-term adverse effects of PPI use seem minimal, chronic PPI use consequences are a growing concern. Chronic PPI use is associated with increased risks of osteoporosis, pneumonia, and Clostridium difficile infections.3 Another long-term risk that has been associated with chronic PPI use is dementia. Dementia is a cognitive syndrome that is characterized by a progressive decline beyond what is expected in normal aging in 1 or more of the cognitive domains of memory, language, orientation, learning capacity, executive function, or social cognition.4 Because it interferes with activities of daily living, dementia is a major cause of disability in the elderly and is an immense burden for caregivers. Currently, about 47 million people globally live with dementia.5 This number is projected to nearly triple by 2050 to 132 million.5 With no cure, identification of risk factors and creation of protective measures are critical in decreasing the prevalence of dementia.

Although the exact pathophysiology behind the link between PPIs and dementia is unknown, several theories exist. One such theory is that PPI-induced vitamin B12 deficiency leads to cognitive decline.6,7 Another theory suggests that PPIs can directly cause dementia by inhibiting enzymes that normally degrade β amyloid.8 This leads to increased levels of β-amyloid plaques, which is a known characteristic of dementia patients. This theory is derived from animal studies that have shown increased amyloid levels in the brains of mice given PPIs.8

Current studies are conflicting regarding the association between PPIs and dementia. Two German prospective, cohort studies found statistically significant increased risks of dementia in patients taking PPIs with hazard ratios (HR) of 1.38 (95% CI, 1.04-1.83) and 1.44 (95% CI, 1.36-1.52), respectively.9,10 A study conducted in Taiwan also found an increased risk of dementia among PPI users with a HR of 1.22 (95% CI, 1.05-1.42).11 On the contrary, other studies have failed to show an increased risk of dementia with PPI use. In fact, Goldstein and colleagues found a decreased risk of dementia in PPI users with a HR of 0.78 (95% CI, 0.76-0.93).12 This study was an observational study conducted in the US using data from the National Alzheimer’s Coordinating Center database.12 Another recent retrospective study conducted in Finland showed that PPI use was not associated with a significantly increased risk of Alzheimer disease.13

Much is unknown about the cause of dementia, and no curative treatment exists. Investigation into potential risk factors for dementia can lead to the development of preventative measures, which can lead to significant improvement in quality of life for both patients and caregivers. Current studies regarding the association between PPIs and dementia are conflicting, and to our knowledge, no study analyzing the effects of PPIs and dementia has been conducted within the veteran population specifically. The objective of the current study is to investigate the association between PPI use and dementia in the veteran population.

Methods

This study is a retrospective, cohort, single-center, chart review study conducted at the Sioux Falls Veteran Affairs Health Care System (SFVAHCS). Data were extracted from the VA electronic health record (EHR) from January 1, 2005 through December 31, 2015. The study included both currently living and deceased veterans who received ≥ 2 documented outpatient visits at the SFVAHCS during the study time frame. Patients also had to be aged ≥ 60 years at the start of the study period. Patients were excluded if they received only a ≤ 30-day PPI prescription. Patients with dementia related to head trauma, acute intoxication, or other known diseases were excluded.

 

 

To analyze the primary endpoint of association between PPI use and dementia, the study compared the rate of dementia in a cohort of veterans who had received an outpatient prescription for a PPI within the study time frame vs the rate of dementia in a random, equal number of veterans who had never been prescribed PPIs within the study time frame. In this study, veterans were classified as having dementia if they had a diagnosis of dementia based on ICD-9 or ICD-10 codes (Table 1), or if they had been prescribed medications used to treat dementia (donepezil, ergoloid mesylates, galantamine, memantine, and rivastigmine).

Secondary endpoints included analysis of the effects of PPI agent, PPI dose, and PPI duration on the risk of dementia. For the PPI dose analysis, cumulative doses were converted into defined daily doses (DDDs) using the World Health Organization calculation to equalize the different potencies of PPI agents (Table 2).14 In addition, the effect of PPI use on vitamin B12 levels was analyzed as an exploratory endpoint to investigate the hypothesis that PPI may be associated with vitamin B12 deficiency, which in turn may be associated with dementia.6,7

Baseline characteristics were collected to determine the variability between the treatment and control group. Data collected included age, gender, past medical history of diseases that may increase risk of dementia, and anticholinergic drug use. Anticholinergic drugs were included if they were classified as having “definite anticholinergic effects” based on the Aging Brain Care Anticholinergic Burden Scale (Appendix).15

 

Statistical Analysis

The primary endpoint was analyzed using a χ2 for association test. For the secondary endpoints, a χ2 for association test was used for endpoints with nominal data, and the Mood median test was used for endpoints with continuous data. The exploratory endpoint analyzing vitamin B12 levels was analyzed with the Mood median test. A P value of < .05 was defined as being statistically significant. Power analysis was not performed since all veterans who met the criteria were included in the study.

Results

Records of 23,656 veterans were included in the study with 11,828 veterans in both the PPI cohort and the non-PPI cohort (Table 3).

Primary Endpoint

Within the PPI group, 1,119 (9.5%) veterans had dementia compared with only 740 (6.3%) veterans in the non-PPI group. There was a statistically significant association between PPI use and dementia (P < .001). These results yielded an odds ratio of 1.55 for dementia risk in PPI users vs nonusers and a relative risk increase of 51.4% for dementia risk with PPI use compared with no PPI use.

 

 

Secondary Endpoints

Users of rabeprazole had the highest rate of dementia (12.8%), followed by lansoprazole (10.9%), omeprazole (9.7%), esomeprazole (7.7%), and pantoprazole (7.0%). The rate of dementia for non-PPI users was 6.3% (P < .001). The median cumulative doses of PPIs were not significant: 597 DDDs (95% CI, 540-630) in the dementia group vs 570 DDDs (95% CI, 540-624) in the nondementia group (P = .79). The median cumulative duration of PPI use in the dementia group was 4.6 years (95% CI, 4.25-4.92) vs 5.3 years (95% CI, 5.08-5.42) in the nondementia group (P < .001).

Exploratory Endpoint

The median B12 level in the PPI group was 521 pg/mL (95% CI, 509-533) compared with 480 pg/mL (95% CI, 465-496) in the non-PPI group (P < .001). However, both groups fell within the normal range for vitamin B12 (200-900 pg/mL).16

Discussion

The aim of this study was to determine whether an association existed between PPI use and dementia. This study showed a statistically significant association between PPI use and dementia within the veteran population. This study also showed a significant association between specific PPI agents and dementia. When analyzing the individual PPI agents, the rabeprazole group yielded the strongest relationship. However, this study was not powered to evaluate and compare risks of dementia between individual PPI agents. More data are needed to determine statistical and clinical significance of associations between individual PPI agents and risk of dementia.

The veterans with dementia had a higher median cumulative PPI dose than did the veterans without dementia; however, the results were not statistically significant. Therefore, the data cannot correlate higher doses of PPI use to increased risk of dementia.

The cumulative duration of PPI use was statistically significant but opposite of the expected outcome. The dementia group had a lower median lifetime duration of PPI use compared with that of the nondementia group. It is difficult to determine the reason for this outcome, but it seems that for this study population, a longer duration of PPI use was not associated with an increased risk of dementia.

Finally, the exploratory endpoint analyzed vitamin B12 levels, since it has been shown that PPI use can lead to vitamin B12 deficiency and that B12 deficiency can lead to dementia.6-8 This study found that the dementia group had significantly higher vitamin B12 levels than the nondementia group. These data suggest that PPI use may not be associated with vitamin B12 deficiency. However, it is important to note that this study was unable to collect data on the use of vitamin B12 supplementation due to the unreliability of over-the-counter (OTC) and non-VA medication use records. Therefore, it is possible that the PPI group had higher rates of B12 deficiency but were effectively treated with B12 supplementation. More research is needed to determine the exact relationship between PPI use, vitamin B12 deficiency, and dementia risk.

Strengths/Limitations

Strengths of this study that support its findings include the large population size. Additionally, the use of the VA EHR allowed for a complete drug dispensing history to be collected, which improves reliability of the data.

 

 

This study also had some limitations. First, the causal relationship of PPI use and dementia cannot be proven using a retrospective cohort design. This study’s design can show association, but it cannot prove causation. Also, due to the retrospective design, exposure to PPI use could not be randomized; thus, correlation between PPI use and dementia may be explained by confounding variables that are not captured within this study. This is especially true since the baseline characteristics were not equally distributed between the 2 groups. In fact, the PPI group had higher rates of many clinical comorbidities. This imbalance may have skewed the results of the primary endpoint. Lastly, OTC PPI use and non-VA PPI prescriptions were not available. Therefore, some of the patients included in the non-PPI group may have been PPI users if they received PPIs from OTC or non-VA sources, which could skew the results.

Conclusion

This study showed a significant association between PPI use and dementia within the veteran study population. The study also showed a significant association between PPI use and dementia within the secondary endpoint of individual PPI agent. Higher cumulative dose and duration of PPI use did not seem to increase risk of dementia. Finally, PPI use was not associated with significantly low vitamin B12 levels. More studies are needed to determine causation of dementia and its risk factors.

Acknowledgments
This material is the result of work supported with resources and the use of facilities at the Sioux Falls VA Health Care System.

Proton pump inhibitors (PPIs) have become the mainstay of therapy in the treatment of acid-related disorders since their introduction in 1989. Due to their high potency, excellent tolerability, and generic availability, PPIs have largely replaced histamine-2 receptor antagonists for gastric problems. Since they were first released on the market, the use of PPIs has continued to rise in both the hospital and primary care settings.1 However, this rapid growth has led to the concern of overutilization. A study conducted at the Department of Veterans Affairs (VA) Ann Arbor Health Care System found that out of 946 patients in the ambulatory care setting taking PPIs, only 35% were appropriately prescribed PPIs.2

Although the short-term adverse effects of PPI use seem minimal, chronic PPI use consequences are a growing concern. Chronic PPI use is associated with increased risks of osteoporosis, pneumonia, and Clostridium difficile infections.3 Another long-term risk that has been associated with chronic PPI use is dementia. Dementia is a cognitive syndrome that is characterized by a progressive decline beyond what is expected in normal aging in 1 or more of the cognitive domains of memory, language, orientation, learning capacity, executive function, or social cognition.4 Because it interferes with activities of daily living, dementia is a major cause of disability in the elderly and is an immense burden for caregivers. Currently, about 47 million people globally live with dementia.5 This number is projected to nearly triple by 2050 to 132 million.5 With no cure, identification of risk factors and creation of protective measures are critical in decreasing the prevalence of dementia.

Although the exact pathophysiology behind the link between PPIs and dementia is unknown, several theories exist. One such theory is that PPI-induced vitamin B12 deficiency leads to cognitive decline.6,7 Another theory suggests that PPIs can directly cause dementia by inhibiting enzymes that normally degrade β amyloid.8 This leads to increased levels of β-amyloid plaques, which is a known characteristic of dementia patients. This theory is derived from animal studies that have shown increased amyloid levels in the brains of mice given PPIs.8

Current studies are conflicting regarding the association between PPIs and dementia. Two German prospective, cohort studies found statistically significant increased risks of dementia in patients taking PPIs with hazard ratios (HR) of 1.38 (95% CI, 1.04-1.83) and 1.44 (95% CI, 1.36-1.52), respectively.9,10 A study conducted in Taiwan also found an increased risk of dementia among PPI users with a HR of 1.22 (95% CI, 1.05-1.42).11 On the contrary, other studies have failed to show an increased risk of dementia with PPI use. In fact, Goldstein and colleagues found a decreased risk of dementia in PPI users with a HR of 0.78 (95% CI, 0.76-0.93).12 This study was an observational study conducted in the US using data from the National Alzheimer’s Coordinating Center database.12 Another recent retrospective study conducted in Finland showed that PPI use was not associated with a significantly increased risk of Alzheimer disease.13

Much is unknown about the cause of dementia, and no curative treatment exists. Investigation into potential risk factors for dementia can lead to the development of preventative measures, which can lead to significant improvement in quality of life for both patients and caregivers. Current studies regarding the association between PPIs and dementia are conflicting, and to our knowledge, no study analyzing the effects of PPIs and dementia has been conducted within the veteran population specifically. The objective of the current study is to investigate the association between PPI use and dementia in the veteran population.

Methods

This study is a retrospective, cohort, single-center, chart review study conducted at the Sioux Falls Veteran Affairs Health Care System (SFVAHCS). Data were extracted from the VA electronic health record (EHR) from January 1, 2005 through December 31, 2015. The study included both currently living and deceased veterans who received ≥ 2 documented outpatient visits at the SFVAHCS during the study time frame. Patients also had to be aged ≥ 60 years at the start of the study period. Patients were excluded if they received only a ≤ 30-day PPI prescription. Patients with dementia related to head trauma, acute intoxication, or other known diseases were excluded.

 

 

To analyze the primary endpoint of association between PPI use and dementia, the study compared the rate of dementia in a cohort of veterans who had received an outpatient prescription for a PPI within the study time frame vs the rate of dementia in a random, equal number of veterans who had never been prescribed PPIs within the study time frame. In this study, veterans were classified as having dementia if they had a diagnosis of dementia based on ICD-9 or ICD-10 codes (Table 1), or if they had been prescribed medications used to treat dementia (donepezil, ergoloid mesylates, galantamine, memantine, and rivastigmine).

Secondary endpoints included analysis of the effects of PPI agent, PPI dose, and PPI duration on the risk of dementia. For the PPI dose analysis, cumulative doses were converted into defined daily doses (DDDs) using the World Health Organization calculation to equalize the different potencies of PPI agents (Table 2).14 In addition, the effect of PPI use on vitamin B12 levels was analyzed as an exploratory endpoint to investigate the hypothesis that PPI may be associated with vitamin B12 deficiency, which in turn may be associated with dementia.6,7

Baseline characteristics were collected to determine the variability between the treatment and control group. Data collected included age, gender, past medical history of diseases that may increase risk of dementia, and anticholinergic drug use. Anticholinergic drugs were included if they were classified as having “definite anticholinergic effects” based on the Aging Brain Care Anticholinergic Burden Scale (Appendix).15

 

Statistical Analysis

The primary endpoint was analyzed using a χ2 for association test. For the secondary endpoints, a χ2 for association test was used for endpoints with nominal data, and the Mood median test was used for endpoints with continuous data. The exploratory endpoint analyzing vitamin B12 levels was analyzed with the Mood median test. A P value of < .05 was defined as being statistically significant. Power analysis was not performed since all veterans who met the criteria were included in the study.

Results

Records of 23,656 veterans were included in the study with 11,828 veterans in both the PPI cohort and the non-PPI cohort (Table 3).

Primary Endpoint

Within the PPI group, 1,119 (9.5%) veterans had dementia compared with only 740 (6.3%) veterans in the non-PPI group. There was a statistically significant association between PPI use and dementia (P < .001). These results yielded an odds ratio of 1.55 for dementia risk in PPI users vs nonusers and a relative risk increase of 51.4% for dementia risk with PPI use compared with no PPI use.

 

 

Secondary Endpoints

Users of rabeprazole had the highest rate of dementia (12.8%), followed by lansoprazole (10.9%), omeprazole (9.7%), esomeprazole (7.7%), and pantoprazole (7.0%). The rate of dementia for non-PPI users was 6.3% (P < .001). The median cumulative doses of PPIs were not significant: 597 DDDs (95% CI, 540-630) in the dementia group vs 570 DDDs (95% CI, 540-624) in the nondementia group (P = .79). The median cumulative duration of PPI use in the dementia group was 4.6 years (95% CI, 4.25-4.92) vs 5.3 years (95% CI, 5.08-5.42) in the nondementia group (P < .001).

Exploratory Endpoint

The median B12 level in the PPI group was 521 pg/mL (95% CI, 509-533) compared with 480 pg/mL (95% CI, 465-496) in the non-PPI group (P < .001). However, both groups fell within the normal range for vitamin B12 (200-900 pg/mL).16

Discussion

The aim of this study was to determine whether an association existed between PPI use and dementia. This study showed a statistically significant association between PPI use and dementia within the veteran population. This study also showed a significant association between specific PPI agents and dementia. When analyzing the individual PPI agents, the rabeprazole group yielded the strongest relationship. However, this study was not powered to evaluate and compare risks of dementia between individual PPI agents. More data are needed to determine statistical and clinical significance of associations between individual PPI agents and risk of dementia.

The veterans with dementia had a higher median cumulative PPI dose than did the veterans without dementia; however, the results were not statistically significant. Therefore, the data cannot correlate higher doses of PPI use to increased risk of dementia.

The cumulative duration of PPI use was statistically significant but opposite of the expected outcome. The dementia group had a lower median lifetime duration of PPI use compared with that of the nondementia group. It is difficult to determine the reason for this outcome, but it seems that for this study population, a longer duration of PPI use was not associated with an increased risk of dementia.

Finally, the exploratory endpoint analyzed vitamin B12 levels, since it has been shown that PPI use can lead to vitamin B12 deficiency and that B12 deficiency can lead to dementia.6-8 This study found that the dementia group had significantly higher vitamin B12 levels than the nondementia group. These data suggest that PPI use may not be associated with vitamin B12 deficiency. However, it is important to note that this study was unable to collect data on the use of vitamin B12 supplementation due to the unreliability of over-the-counter (OTC) and non-VA medication use records. Therefore, it is possible that the PPI group had higher rates of B12 deficiency but were effectively treated with B12 supplementation. More research is needed to determine the exact relationship between PPI use, vitamin B12 deficiency, and dementia risk.

Strengths/Limitations

Strengths of this study that support its findings include the large population size. Additionally, the use of the VA EHR allowed for a complete drug dispensing history to be collected, which improves reliability of the data.

 

 

This study also had some limitations. First, the causal relationship of PPI use and dementia cannot be proven using a retrospective cohort design. This study’s design can show association, but it cannot prove causation. Also, due to the retrospective design, exposure to PPI use could not be randomized; thus, correlation between PPI use and dementia may be explained by confounding variables that are not captured within this study. This is especially true since the baseline characteristics were not equally distributed between the 2 groups. In fact, the PPI group had higher rates of many clinical comorbidities. This imbalance may have skewed the results of the primary endpoint. Lastly, OTC PPI use and non-VA PPI prescriptions were not available. Therefore, some of the patients included in the non-PPI group may have been PPI users if they received PPIs from OTC or non-VA sources, which could skew the results.

Conclusion

This study showed a significant association between PPI use and dementia within the veteran study population. The study also showed a significant association between PPI use and dementia within the secondary endpoint of individual PPI agent. Higher cumulative dose and duration of PPI use did not seem to increase risk of dementia. Finally, PPI use was not associated with significantly low vitamin B12 levels. More studies are needed to determine causation of dementia and its risk factors.

Acknowledgments
This material is the result of work supported with resources and the use of facilities at the Sioux Falls VA Health Care System.

References

1. Savarino V, Dulbecco P, de Bortoli N, Ottonello A, Savarino E. The appropriate use of proton pump inhibitors (PPIs): need for a reappraisal. Eur J Intern Med. 2017;37:19-24.

2. Heidelbaugh J, Goldberg K, Inadomi J. Magnitude and economic effect of overuse of antisecretory therapy in the ambulatory care setting. Am J Manag Care. 2010;16(9):e228-e234.

3. Heidelbaugh JJ, Kim AH, Chang R. Walker PC. Overutilization of proton-pump inhibitors: what the clinician needs to know. Therap Adv Gastroenterol. 2012;5(4):219-232.

4. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, (DSM-5). American Psychiatric Association: Washington, DC; 2013.

5. World Health Organization. Dementia. http://www.who.int/mediacentre/factsheets/fs362/en/. Published December 12, 2017. Accessed March 10, 2019.

6. Vogiatzoglou A, Smith AD, Nurk E, et al. Cognitive function in an elderly population: interaction between vitamin B12 status, depression, and apolipoprotein E ε4: the Hordaland Homocysteine Study. Psychosom Med. 2013;75(1):20-29.

7. Lam JR, Schneider JL, Zhao W, Corley DA. Proton pump inhibitor and histamine 2 receptor antagonist use and vitamin B12 deficiency. JAMA. 2013;310(22):2435-2442.

8. Badiola N, Alcalde V, Pujol A, et al. The proton-pump inhibitor lansoprazole enhances amyloid beta production. PLoS One. 2013;8(3):e58837.

9. Haenisch B, von Holt K, Wiese B, et al. Risk of dementia in elderly patients with the use of proton pump inhibitors. Eur Arch Psychiatry Clin Neurosci. 2015;265(5):419-428.

10. Gomm W, von Holt K, Thomé F, et al. Association between proton pump inhibitors with risk of dementia. A pharmacoepidemiological claims data analysis. JAMA Neurol. 2016;73(4):410-416.

11. Tai SY, Chien CY, Wu DC, et al. Risk of dementia from proton pump inhibitor use in Asian population: a nationwide cohort study in Taiwan. PLoS One. 2017;12(2):e0171006.

12. Goldstein FC, Steenland K, Zhao L, Wharton W, Levey AI, Hajjar I. Proton pump inhibitors and risk of mild cognitive impairment and dementia. J Am Geriatr Soc. 2017;65(9):1969-1674.

13. Taipale H, Tolppanen AM, Tiihonen M. Tanskanen A, Tiihonen J, Hartikainen S. No association between proton pump inhibitor use and risk of Alzheimer’s disease. Am J Gastroenterol. 2017;112(12):1801-1808.

14. World Health Organization Collaborating Centre for Drug Statistics Methodology. Definition and general considerations. https://www.whocc.no/ddd/definition_and_general_considera/. Updated February 7, 2018. Accessed March 13, 2019.

15. Indiana University Center for Aging Research, Aging Brain Program. Anticholinergic cognitive burden scale. http://www.idhca.org/wp-content/uploads/2018/02/DESAI_ACB_scale_-_Legal_size_paper.pdf. Updated 2012. Accessed March 10, 2019.

16. US National Library of Medicine, MedlinePlus. Vitamin B12 level. https://medlineplus.gov/ency/article/003705.htm. Updated March 7, 2019. Accessed March 13, 2019.

References

1. Savarino V, Dulbecco P, de Bortoli N, Ottonello A, Savarino E. The appropriate use of proton pump inhibitors (PPIs): need for a reappraisal. Eur J Intern Med. 2017;37:19-24.

2. Heidelbaugh J, Goldberg K, Inadomi J. Magnitude and economic effect of overuse of antisecretory therapy in the ambulatory care setting. Am J Manag Care. 2010;16(9):e228-e234.

3. Heidelbaugh JJ, Kim AH, Chang R. Walker PC. Overutilization of proton-pump inhibitors: what the clinician needs to know. Therap Adv Gastroenterol. 2012;5(4):219-232.

4. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, (DSM-5). American Psychiatric Association: Washington, DC; 2013.

5. World Health Organization. Dementia. http://www.who.int/mediacentre/factsheets/fs362/en/. Published December 12, 2017. Accessed March 10, 2019.

6. Vogiatzoglou A, Smith AD, Nurk E, et al. Cognitive function in an elderly population: interaction between vitamin B12 status, depression, and apolipoprotein E ε4: the Hordaland Homocysteine Study. Psychosom Med. 2013;75(1):20-29.

7. Lam JR, Schneider JL, Zhao W, Corley DA. Proton pump inhibitor and histamine 2 receptor antagonist use and vitamin B12 deficiency. JAMA. 2013;310(22):2435-2442.

8. Badiola N, Alcalde V, Pujol A, et al. The proton-pump inhibitor lansoprazole enhances amyloid beta production. PLoS One. 2013;8(3):e58837.

9. Haenisch B, von Holt K, Wiese B, et al. Risk of dementia in elderly patients with the use of proton pump inhibitors. Eur Arch Psychiatry Clin Neurosci. 2015;265(5):419-428.

10. Gomm W, von Holt K, Thomé F, et al. Association between proton pump inhibitors with risk of dementia. A pharmacoepidemiological claims data analysis. JAMA Neurol. 2016;73(4):410-416.

11. Tai SY, Chien CY, Wu DC, et al. Risk of dementia from proton pump inhibitor use in Asian population: a nationwide cohort study in Taiwan. PLoS One. 2017;12(2):e0171006.

12. Goldstein FC, Steenland K, Zhao L, Wharton W, Levey AI, Hajjar I. Proton pump inhibitors and risk of mild cognitive impairment and dementia. J Am Geriatr Soc. 2017;65(9):1969-1674.

13. Taipale H, Tolppanen AM, Tiihonen M. Tanskanen A, Tiihonen J, Hartikainen S. No association between proton pump inhibitor use and risk of Alzheimer’s disease. Am J Gastroenterol. 2017;112(12):1801-1808.

14. World Health Organization Collaborating Centre for Drug Statistics Methodology. Definition and general considerations. https://www.whocc.no/ddd/definition_and_general_considera/. Updated February 7, 2018. Accessed March 13, 2019.

15. Indiana University Center for Aging Research, Aging Brain Program. Anticholinergic cognitive burden scale. http://www.idhca.org/wp-content/uploads/2018/02/DESAI_ACB_scale_-_Legal_size_paper.pdf. Updated 2012. Accessed March 10, 2019.

16. US National Library of Medicine, MedlinePlus. Vitamin B12 level. https://medlineplus.gov/ency/article/003705.htm. Updated March 7, 2019. Accessed March 13, 2019.

Issue
Federal Practitioner - 36(4)s
Issue
Federal Practitioner - 36(4)s
Page Number
S27-S31
Page Number
S27-S31
Publications
Publications
Topics
Article Type
Display Headline
Proton Pump Inhibitor Use and Risk of Dementia in the Veteran Population
Display Headline
Proton Pump Inhibitor Use and Risk of Dementia in the Veteran Population
Sections
Citation Override
Fed Pract. 2019 June;36(suppl 4):S27-S31
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.
Article PDF Media

Understanding Psychosis in a Veteran With a History of Combat and Multiple Sclerosis (FULL)

Article Type
Changed
Mon, 06/01/2020 - 15:23
Display Headline
Understanding Psychosis in a Veteran With a History of Combat and Multiple Sclerosis

A patient with significant combat history and previous diagnoses of multiple sclerosis and unspecified schizophrenia spectrum and other psychotic disorder was admitted with acute psychosis inconsistent with expected clinical presentations.

Multiple sclerosis (MS) is an immune-mediated neurodegenerative disease that affects > 700,000 people in the US.1 The hallmarks of MS pathology are axonal or neuronal loss, demyelination, and astrocytic gliosis. Of these, axonal or neuronal loss is the main underlying mechanism of permanent clinical disability.

MS also has been associated with an increased prevalence of psychiatric illnesses, with mood disorders affecting up to 40% to 60% of the population, and psychosis being reported in 2% to 4% of patients.2 The link between MS and mood disorders, including bipolar disorder and depression, was documented as early as 1926,with mood disorders hypothesized to be manifestations of central nervous system (CNS) inflammation.3 More recently, inflammation-driven microglia have been hypothesized to impair hippocampal connectivity and activate glucocorticoid-insensitive inflammatory cells that then overstimulate the hypothalamic-pituitary-adrenal axis.4,5

Although the prevalence of psychosis in patients with MS is significantly rarer, averaging between 2% and 4%.6 A Canadian study by Patten and colleagues reviewed data from 2.45 million residents of Alberta and found that those who identified as having MS had a 2% to 3% prevalence of psychosis compared with 0.5% to 1% in the general population.7 The connection between psychosis and MS, similar to that between mood disorders and MS, has been described as a common regional demyelination process. Supporting this, MS manifesting as psychosis has been found to present with distinct magnetic resonance imaging (MRI) findings, such as diffuse periventricular lesions.8 Still, no conclusive criteria have been developed to distinguish MS presenting as psychosis from a primary psychiatric illness, such as schizophrenia.

In patients with combat history, it is possible that both neurodegenerative and psychotic symptoms can be explained by autoantibody formation in response to toxin exposure. When soldiers were deployed to Iraq and Afghanistan, they may have been exposed to multiple toxicities, including depleted uranium, dust and fumes, and numerous infectious diseases.9 Gulf War illness (GWI) or chronic multisymptom illness (CMI) encompass a cluster of symptoms, such as chronic pain, chronic fatigue, irritable bowel syndrome, dermatitis, and seizures, as well as mental health issues such as depression and anxiety experienced following exposure to these combat environments.10,11

In light of this diagnostic uncertainty, the authors detail a case of a patient with significant combat history previously diagnosed with MS and unspecified schizophrenia spectrum and other psychotic disorder (USS & OPD) presenting with acute psychosis.

Case Presentation

A 35-year-old male veteran, with a history of MS, USS & OPD, posttraumatic stress disorder, and traumatic brain injuries (TBIs) was admitted to the psychiatric unit after being found by the police lying in the middle of a busy intersection, internally preoccupied. On admission, he reported a week of auditory hallucinations from birds with whom he had been communicating telepathically, and a recurrent visual hallucination of a tall man in white and purple robes. He had discontinued his antipsychotic medication, aripiprazole 10 mg, a few weeks prior for unknown reasons. He was brought to the hospital by ambulance, where he presented with disorganized thinking, tangential thought process, and active auditory and visual hallucinations. The differential diagnoses included USS & OPD, schizophrenia, schizoaffective disorder and ruled out substance-induced psychotic disorder, and psychosis as a manifestation of MS.

 

 

The patient had 2 psychotic episodes prior to this presentation. He was hospitalized for his first psychotic break in 2015 at age 32, when he had tailed another car “to come back to reality” and ended up in a motor vehicle accident. During that admission, he reported weeks of thought broadcasting, conspiratorial delusions, and racing thoughts. Two years later, he was admitted to a psychiatric intensive care unit for his second episode of severe psychosis. After several trials of different antipsychotic medications, his most recent pharmacologic regimen was aripiprazole 10 mg once daily.

His medical history was complicated by 2 TBIs, in November 2014 and January 2015, with normal computed tomography (CT) scans. He was diagnosed with MS in December 2017, when he presented with intractable emesis, left facial numbness, right upper extremity ataxia, nystagmus, and imbalance. An MRI scan revealed multifocal bilateral hypodensities in his periventricular, subcortical, and brain stem white matter. Multiple areas of hyperintensity were visualized, including in the right periatrial region and left brachium pontis. More than 5 oligoclonal bands on lumbar puncture confirmed the diagnosis.

He was treated with IV methylprednisolone followed by a 2-week prednisone taper. Within 1 week, he returned to the psychiatric unit with worsening symptoms and received a second dose of IV steroids and plasma exchange treatment. In the following months, he completed a course of rituximab infusions and physical therapy for his dysarthria, gait abnormality, and vision impairment.

His social history was notable for multiple first-degree relatives with schizophrenia. He reported a history of sexual and verbal abuse and attempted suicide once at age 13 years by hanging himself with a bathrobe. He left home at age 18 years to serve in the Marine Corps (2001-2006). His service included deployment to Afghanistan, where he received a purple heart. Upon his return, he received BA and MS degrees. He married and had 2 daughters but became estranged from his wife. By his most recent admission, he was unemployed and living with his half-sister.

On the first day of this most recent psychiatric hospitalization, he was restarted on aripiprazole 10 mg daily, and a medicine consult was sought to evaluate the progression of his MS. No new onset neurologic symptoms were noted, but he had possible residual lower extremity hyperreflexia and tandem gait incoordination. The episodes of psychotic and neurologic symptoms appeared independent, given that his psychiatric history preceded the onset of his MS.

The patient reported no visual hallucinations starting day 2, and he no longer endorsed auditory hallucinations by day 3. However, he continued to appear internally preoccupied and was noticed to be pacing around the unit. On day 4 he presented with newly pressured speech and flights of ideas, while his affect remained euthymic and his sleep stayed consistent. In combination with his ongoing pacing, his newfound symptoms were hypothesized to be possibly akathisia, an adverse effect (AE) of aripiprazole. As such, on day 5 his dose was lowered to 5 mg daily. He continued to report no hallucinations and demonstrated progressively increased emotional range. A MRI scan was done on day 6 in case a new lesion could be identified, suggesting a primary MS flare-up; however, the scan identified no enhancing lesions, indicating no ongoing demyelination. After a neurology consult corroborated this conclusion, he was discharged in stable condition on day 7.

As is the case with the majority of patients with MS-induced psychosis, he continued to have relapsing psychiatric disease even after MS treatment had been started. Unfortunately, because this patient had stopped taking his atypical antipsychotic medication several weeks prior to his hospitalization, we cannot clarify whether his psychosis stems from a primary psychiatric vs MS process.

 

 

Discussion

Presently, treatment preferences for MS-related psychosis are divided between atypical antipsychotics and glucocorticoids. Some suggest that the treatment remains similar between MS-related psychosis and primary psychotic disorders in that atypical antipsychotics are the standard of care.12 A variety of atypical antipsychotics have been used successfully in case reports, including zipradisone, risperidone, olanzapine, quetiapine, and aripiprazole.13,14 First-generation antipsychotics and other psychotropic drugs that can precipitate extra-pyramidal AEs are not recommended given their potential additive effect to motor deficits associated with MS.12 Alternatively, several case reports have found that MS-related psychotic symptoms respond to glucocorticoids more effectively, while cautioning that glucocorticoids can precipitate psychosis and depression.15,16 One review article found that 90% of patients who received corticosteroids saw an improvement in their psychotic symptoms.2

Finally, it is possible that our patient’s neuropsychiatric symptoms can be explained by autoantibody formation in response to toxin exposure during his time in Afghanistan. In a pilot study of veterans with GWI, Abou-Donia and colleagues found 2-to-9 fold increase in autoantibody reactivity levels of the following neuronal and glial-specific proteins relative to healthy controls: neurofilament triplet proteins, tubulin, microtubule-associated tau proteins, microtubule-associated protein-2, myelin basic protein, myelin-associated glycoprotein, glial fibrillary acidic protein, and calcium-calmodulin kinase II.17,18 Many of these autoantibodies are longstanding explicit markers for neurodegenerative disorders, given that they target proteins and antigens that support axonal transport and myelination. Still Gulf War veteran status has yet to be explicitly linked to an increased risk of MS,19 making this hypothesis less likely for our patient. Future research should address the clinical and therapeutic implications of different autoantibody levels in combat veterans with psychosis.

Conclusion

For patients with MS, mood disorder and psychotic symptoms should warrant a MRI given the possibility of a psychiatric manifestation of MS relapse. Ultimately, our patient’s presentation was inconsistent with the expected clinical presentations of both a primary psychotic disorder and psychosis as a manifestation of MS. His late age at his first psychotic break is atypical for primary psychotic disease, and the lack of MRI imaging done at his initial psychotic episodes cannot exclude a primary MS diagnosis. Still, his lack of MRI findings at his most recent hospitalization, negative symptomatology, and strong history of schizophrenia make a primary psychotic disorder likely.

Following his future clinical course will be necessary to determine the etiology of his psychotic episodes. Future episodes of psychosis with neurologic symptoms would suggest a primary MS diagnosis and potential benefit of immunosuppressant treatment, whereas repeated psychotic breaks with minimal temporal lobe involvement or demyelination as seen on MRI would be suspicious for separate MS and psychotic disease processes. Further research on treatment regimens for patients experiencing psychosis as a manifestation of MS is still necessary.

References

1. Wallin MT, Culpepper WJ, Campbell JD, et al. The prevalence of MS in the United States: A population-based estimate using health claims data. Neurology. 2019;92(10):e1029-e1040.

2. Camara-Lemarroy CR, Ibarra-Yruegas BE, Rodriguez-Gutierrez R, Berrios-Morales I, Ionete C, Riskind P. The varieties of psychosis in multiple sclerosis: a systematic review of cases. Mult Scler Relat Disord. 2017;12:9-14.

3. Cottrel SS, Wilson SA. The affective symptomatology of disseminated sclerosis: a study of 100 cases. J Neurol Psychopathology. 1926;7(25):1-30.

4. Johansson V, Lundholm C, Hillert J, et al. Multiple sclerosis and psychiatric disorders: comorbidity and sibling risk in a nationwide Swedish cohort. Mult Scler. 2014;20(14):1881-1891.

5. Rossi S, Studer V, Motta C, et al. Neuroinflammation drives anxiety and depression in relapsing-remitting multiple sclerosis. Neurology. 2017;89(13):1338-1347.

6. Gilberthorpe TG, O’Connell KE, Carolan A, et al. The spectrum of psychosis in multiple sclerosis: a clinical case series. Neuropsychiatric disease and treatment. 2017;13:303.

7. Patten SB, Svenson LW, Metz LM. Psychotic disorders in MS: population-based evidence of an association. Neurology 2005;65(7):1123-1125.

8. Kosmidis MH, Giannakou M, Messinis L, Papathanasopoulos P. Psychotic features associated with multiple sclerosis. Int Rev Psychiatry. 2010; 22(1):55-66.

9. US Department of Veterans Affairs. Public health: military exposures. https://www.publichealth.va.gov/exposures/. Updated April 16, 2019. Accessed May 13, 2019.

10. DeBeer BB, Davidson D, Meyer EC, Kimbrel NA, Gulliver SB, Morissette SB. The association between toxic exposures and chronic multisymptom illness in veterans of the wars of Iraq and Afghanistan. J Occup Environ Med. 2017;59(1):54-60.

11. Kang HK, Li B, Mahan CM, Eisen SA, Engel CC. Health of US veterans of 1991 Gulf War: a follow-up survey in 10 years. J Occup Environ Med. 2009;51(4):401-410.

12. Murphy R, O’Donoghue S, Counihan T, et al. Neuropsychiatric syndromes of multiple sclerosis. J Neurol Neurosurg Psychiatry. 2017;88(8):697-708.

13. Davids E, Hartwig U, Gastpar, M. Antipsychotic treatment of psychosis associated with multiple sclerosis. Prog Neuro Psychopharmacol Biol Psychiatry. 2004;28(4):743-744.

14. Lo Fermo S, Barone R, Patti F, et al. Outcome of psychiatric symptoms presenting at onset of multiple sclerosis: a retrospective study. Mult Scler. 2010;16(6):742-748.

15. Enderami A, Fouladi R, Hosseini HS. First-episode psychosis as the initial presentation of multiple sclerosis: a case report. Int Medical Case Rep J. 2018;11:73-76.

16. Fragoso YD, Frota ER, Lopes JS, et al. Severe depression, suicide attempts, and ideation during the use of interferon beta by patients with multiple sclerosis. Clin Neuropharmacol. 2010;33(6):312-316.

17. Abou-Donia MB, Conboy LA, Kokkotou E, et al. Screening for novel central nervous system biomarkers in veterans with Gulf War Illness. Neurotoxicol Teratol. 2017;61:36-46.

18. Abou-Donia MB, Lieberman A, Curtis L. Neural autoantibodies in patients with neurological symptoms and histories of chemical/mold exposures. Toxicol Ind Health. 2018;34(1):44-53.

19. Wallin MT, Kurtzke JF, Culpepper WJ, et al. Multiple sclerosis in Gulf War era veterans. 2. Military deployment and risk of multiple sclerosis in the first Gulf War. Neuroepidemiology. 2014;42(4):226-234.

Article PDF
Author and Disclosure Information

Angela Lee is a Medical Student, and Kalpana Nathan is a Clinical Associate Professor (Affiliated) in the Department of Psychiatry and Behavioral Sciences, both at Stanford University School of Medicine in California. Kalpana Nathan also is an Attending Psychiatrist in the Veterans Affairs Palo Alto Health Care System in California.
Correspondence: Angela Lee (angelal4@stanford.edu)

Author disclosures
The authors report no actual or potential conflicts of interest with regard to this article.

Disclaimer
The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review the complete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

Issue
Federal Practitioner - 36(4)s
Publications
Topics
Page Number
S32-S35
Sections
Author and Disclosure Information

Angela Lee is a Medical Student, and Kalpana Nathan is a Clinical Associate Professor (Affiliated) in the Department of Psychiatry and Behavioral Sciences, both at Stanford University School of Medicine in California. Kalpana Nathan also is an Attending Psychiatrist in the Veterans Affairs Palo Alto Health Care System in California.
Correspondence: Angela Lee (angelal4@stanford.edu)

Author disclosures
The authors report no actual or potential conflicts of interest with regard to this article.

Disclaimer
The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review the complete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

Author and Disclosure Information

Angela Lee is a Medical Student, and Kalpana Nathan is a Clinical Associate Professor (Affiliated) in the Department of Psychiatry and Behavioral Sciences, both at Stanford University School of Medicine in California. Kalpana Nathan also is an Attending Psychiatrist in the Veterans Affairs Palo Alto Health Care System in California.
Correspondence: Angela Lee (angelal4@stanford.edu)

Author disclosures
The authors report no actual or potential conflicts of interest with regard to this article.

Disclaimer
The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review the complete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

Article PDF
Article PDF
Related Articles

A patient with significant combat history and previous diagnoses of multiple sclerosis and unspecified schizophrenia spectrum and other psychotic disorder was admitted with acute psychosis inconsistent with expected clinical presentations.

A patient with significant combat history and previous diagnoses of multiple sclerosis and unspecified schizophrenia spectrum and other psychotic disorder was admitted with acute psychosis inconsistent with expected clinical presentations.

Multiple sclerosis (MS) is an immune-mediated neurodegenerative disease that affects > 700,000 people in the US.1 The hallmarks of MS pathology are axonal or neuronal loss, demyelination, and astrocytic gliosis. Of these, axonal or neuronal loss is the main underlying mechanism of permanent clinical disability.

MS also has been associated with an increased prevalence of psychiatric illnesses, with mood disorders affecting up to 40% to 60% of the population, and psychosis being reported in 2% to 4% of patients.2 The link between MS and mood disorders, including bipolar disorder and depression, was documented as early as 1926,with mood disorders hypothesized to be manifestations of central nervous system (CNS) inflammation.3 More recently, inflammation-driven microglia have been hypothesized to impair hippocampal connectivity and activate glucocorticoid-insensitive inflammatory cells that then overstimulate the hypothalamic-pituitary-adrenal axis.4,5

Although the prevalence of psychosis in patients with MS is significantly rarer, averaging between 2% and 4%.6 A Canadian study by Patten and colleagues reviewed data from 2.45 million residents of Alberta and found that those who identified as having MS had a 2% to 3% prevalence of psychosis compared with 0.5% to 1% in the general population.7 The connection between psychosis and MS, similar to that between mood disorders and MS, has been described as a common regional demyelination process. Supporting this, MS manifesting as psychosis has been found to present with distinct magnetic resonance imaging (MRI) findings, such as diffuse periventricular lesions.8 Still, no conclusive criteria have been developed to distinguish MS presenting as psychosis from a primary psychiatric illness, such as schizophrenia.

In patients with combat history, it is possible that both neurodegenerative and psychotic symptoms can be explained by autoantibody formation in response to toxin exposure. When soldiers were deployed to Iraq and Afghanistan, they may have been exposed to multiple toxicities, including depleted uranium, dust and fumes, and numerous infectious diseases.9 Gulf War illness (GWI) or chronic multisymptom illness (CMI) encompass a cluster of symptoms, such as chronic pain, chronic fatigue, irritable bowel syndrome, dermatitis, and seizures, as well as mental health issues such as depression and anxiety experienced following exposure to these combat environments.10,11

In light of this diagnostic uncertainty, the authors detail a case of a patient with significant combat history previously diagnosed with MS and unspecified schizophrenia spectrum and other psychotic disorder (USS & OPD) presenting with acute psychosis.

Case Presentation

A 35-year-old male veteran, with a history of MS, USS & OPD, posttraumatic stress disorder, and traumatic brain injuries (TBIs) was admitted to the psychiatric unit after being found by the police lying in the middle of a busy intersection, internally preoccupied. On admission, he reported a week of auditory hallucinations from birds with whom he had been communicating telepathically, and a recurrent visual hallucination of a tall man in white and purple robes. He had discontinued his antipsychotic medication, aripiprazole 10 mg, a few weeks prior for unknown reasons. He was brought to the hospital by ambulance, where he presented with disorganized thinking, tangential thought process, and active auditory and visual hallucinations. The differential diagnoses included USS & OPD, schizophrenia, schizoaffective disorder and ruled out substance-induced psychotic disorder, and psychosis as a manifestation of MS.

 

 

The patient had 2 psychotic episodes prior to this presentation. He was hospitalized for his first psychotic break in 2015 at age 32, when he had tailed another car “to come back to reality” and ended up in a motor vehicle accident. During that admission, he reported weeks of thought broadcasting, conspiratorial delusions, and racing thoughts. Two years later, he was admitted to a psychiatric intensive care unit for his second episode of severe psychosis. After several trials of different antipsychotic medications, his most recent pharmacologic regimen was aripiprazole 10 mg once daily.

His medical history was complicated by 2 TBIs, in November 2014 and January 2015, with normal computed tomography (CT) scans. He was diagnosed with MS in December 2017, when he presented with intractable emesis, left facial numbness, right upper extremity ataxia, nystagmus, and imbalance. An MRI scan revealed multifocal bilateral hypodensities in his periventricular, subcortical, and brain stem white matter. Multiple areas of hyperintensity were visualized, including in the right periatrial region and left brachium pontis. More than 5 oligoclonal bands on lumbar puncture confirmed the diagnosis.

He was treated with IV methylprednisolone followed by a 2-week prednisone taper. Within 1 week, he returned to the psychiatric unit with worsening symptoms and received a second dose of IV steroids and plasma exchange treatment. In the following months, he completed a course of rituximab infusions and physical therapy for his dysarthria, gait abnormality, and vision impairment.

His social history was notable for multiple first-degree relatives with schizophrenia. He reported a history of sexual and verbal abuse and attempted suicide once at age 13 years by hanging himself with a bathrobe. He left home at age 18 years to serve in the Marine Corps (2001-2006). His service included deployment to Afghanistan, where he received a purple heart. Upon his return, he received BA and MS degrees. He married and had 2 daughters but became estranged from his wife. By his most recent admission, he was unemployed and living with his half-sister.

On the first day of this most recent psychiatric hospitalization, he was restarted on aripiprazole 10 mg daily, and a medicine consult was sought to evaluate the progression of his MS. No new onset neurologic symptoms were noted, but he had possible residual lower extremity hyperreflexia and tandem gait incoordination. The episodes of psychotic and neurologic symptoms appeared independent, given that his psychiatric history preceded the onset of his MS.

The patient reported no visual hallucinations starting day 2, and he no longer endorsed auditory hallucinations by day 3. However, he continued to appear internally preoccupied and was noticed to be pacing around the unit. On day 4 he presented with newly pressured speech and flights of ideas, while his affect remained euthymic and his sleep stayed consistent. In combination with his ongoing pacing, his newfound symptoms were hypothesized to be possibly akathisia, an adverse effect (AE) of aripiprazole. As such, on day 5 his dose was lowered to 5 mg daily. He continued to report no hallucinations and demonstrated progressively increased emotional range. A MRI scan was done on day 6 in case a new lesion could be identified, suggesting a primary MS flare-up; however, the scan identified no enhancing lesions, indicating no ongoing demyelination. After a neurology consult corroborated this conclusion, he was discharged in stable condition on day 7.

As is the case with the majority of patients with MS-induced psychosis, he continued to have relapsing psychiatric disease even after MS treatment had been started. Unfortunately, because this patient had stopped taking his atypical antipsychotic medication several weeks prior to his hospitalization, we cannot clarify whether his psychosis stems from a primary psychiatric vs MS process.

 

 

Discussion

Presently, treatment preferences for MS-related psychosis are divided between atypical antipsychotics and glucocorticoids. Some suggest that the treatment remains similar between MS-related psychosis and primary psychotic disorders in that atypical antipsychotics are the standard of care.12 A variety of atypical antipsychotics have been used successfully in case reports, including zipradisone, risperidone, olanzapine, quetiapine, and aripiprazole.13,14 First-generation antipsychotics and other psychotropic drugs that can precipitate extra-pyramidal AEs are not recommended given their potential additive effect to motor deficits associated with MS.12 Alternatively, several case reports have found that MS-related psychotic symptoms respond to glucocorticoids more effectively, while cautioning that glucocorticoids can precipitate psychosis and depression.15,16 One review article found that 90% of patients who received corticosteroids saw an improvement in their psychotic symptoms.2

Finally, it is possible that our patient’s neuropsychiatric symptoms can be explained by autoantibody formation in response to toxin exposure during his time in Afghanistan. In a pilot study of veterans with GWI, Abou-Donia and colleagues found 2-to-9 fold increase in autoantibody reactivity levels of the following neuronal and glial-specific proteins relative to healthy controls: neurofilament triplet proteins, tubulin, microtubule-associated tau proteins, microtubule-associated protein-2, myelin basic protein, myelin-associated glycoprotein, glial fibrillary acidic protein, and calcium-calmodulin kinase II.17,18 Many of these autoantibodies are longstanding explicit markers for neurodegenerative disorders, given that they target proteins and antigens that support axonal transport and myelination. Still Gulf War veteran status has yet to be explicitly linked to an increased risk of MS,19 making this hypothesis less likely for our patient. Future research should address the clinical and therapeutic implications of different autoantibody levels in combat veterans with psychosis.

Conclusion

For patients with MS, mood disorder and psychotic symptoms should warrant a MRI given the possibility of a psychiatric manifestation of MS relapse. Ultimately, our patient’s presentation was inconsistent with the expected clinical presentations of both a primary psychotic disorder and psychosis as a manifestation of MS. His late age at his first psychotic break is atypical for primary psychotic disease, and the lack of MRI imaging done at his initial psychotic episodes cannot exclude a primary MS diagnosis. Still, his lack of MRI findings at his most recent hospitalization, negative symptomatology, and strong history of schizophrenia make a primary psychotic disorder likely.

Following his future clinical course will be necessary to determine the etiology of his psychotic episodes. Future episodes of psychosis with neurologic symptoms would suggest a primary MS diagnosis and potential benefit of immunosuppressant treatment, whereas repeated psychotic breaks with minimal temporal lobe involvement or demyelination as seen on MRI would be suspicious for separate MS and psychotic disease processes. Further research on treatment regimens for patients experiencing psychosis as a manifestation of MS is still necessary.

Multiple sclerosis (MS) is an immune-mediated neurodegenerative disease that affects > 700,000 people in the US.1 The hallmarks of MS pathology are axonal or neuronal loss, demyelination, and astrocytic gliosis. Of these, axonal or neuronal loss is the main underlying mechanism of permanent clinical disability.

MS also has been associated with an increased prevalence of psychiatric illnesses, with mood disorders affecting up to 40% to 60% of the population, and psychosis being reported in 2% to 4% of patients.2 The link between MS and mood disorders, including bipolar disorder and depression, was documented as early as 1926,with mood disorders hypothesized to be manifestations of central nervous system (CNS) inflammation.3 More recently, inflammation-driven microglia have been hypothesized to impair hippocampal connectivity and activate glucocorticoid-insensitive inflammatory cells that then overstimulate the hypothalamic-pituitary-adrenal axis.4,5

Although the prevalence of psychosis in patients with MS is significantly rarer, averaging between 2% and 4%.6 A Canadian study by Patten and colleagues reviewed data from 2.45 million residents of Alberta and found that those who identified as having MS had a 2% to 3% prevalence of psychosis compared with 0.5% to 1% in the general population.7 The connection between psychosis and MS, similar to that between mood disorders and MS, has been described as a common regional demyelination process. Supporting this, MS manifesting as psychosis has been found to present with distinct magnetic resonance imaging (MRI) findings, such as diffuse periventricular lesions.8 Still, no conclusive criteria have been developed to distinguish MS presenting as psychosis from a primary psychiatric illness, such as schizophrenia.

In patients with combat history, it is possible that both neurodegenerative and psychotic symptoms can be explained by autoantibody formation in response to toxin exposure. When soldiers were deployed to Iraq and Afghanistan, they may have been exposed to multiple toxicities, including depleted uranium, dust and fumes, and numerous infectious diseases.9 Gulf War illness (GWI) or chronic multisymptom illness (CMI) encompass a cluster of symptoms, such as chronic pain, chronic fatigue, irritable bowel syndrome, dermatitis, and seizures, as well as mental health issues such as depression and anxiety experienced following exposure to these combat environments.10,11

In light of this diagnostic uncertainty, the authors detail a case of a patient with significant combat history previously diagnosed with MS and unspecified schizophrenia spectrum and other psychotic disorder (USS & OPD) presenting with acute psychosis.

Case Presentation

A 35-year-old male veteran, with a history of MS, USS & OPD, posttraumatic stress disorder, and traumatic brain injuries (TBIs) was admitted to the psychiatric unit after being found by the police lying in the middle of a busy intersection, internally preoccupied. On admission, he reported a week of auditory hallucinations from birds with whom he had been communicating telepathically, and a recurrent visual hallucination of a tall man in white and purple robes. He had discontinued his antipsychotic medication, aripiprazole 10 mg, a few weeks prior for unknown reasons. He was brought to the hospital by ambulance, where he presented with disorganized thinking, tangential thought process, and active auditory and visual hallucinations. The differential diagnoses included USS & OPD, schizophrenia, schizoaffective disorder and ruled out substance-induced psychotic disorder, and psychosis as a manifestation of MS.

 

 

The patient had 2 psychotic episodes prior to this presentation. He was hospitalized for his first psychotic break in 2015 at age 32, when he had tailed another car “to come back to reality” and ended up in a motor vehicle accident. During that admission, he reported weeks of thought broadcasting, conspiratorial delusions, and racing thoughts. Two years later, he was admitted to a psychiatric intensive care unit for his second episode of severe psychosis. After several trials of different antipsychotic medications, his most recent pharmacologic regimen was aripiprazole 10 mg once daily.

His medical history was complicated by 2 TBIs, in November 2014 and January 2015, with normal computed tomography (CT) scans. He was diagnosed with MS in December 2017, when he presented with intractable emesis, left facial numbness, right upper extremity ataxia, nystagmus, and imbalance. An MRI scan revealed multifocal bilateral hypodensities in his periventricular, subcortical, and brain stem white matter. Multiple areas of hyperintensity were visualized, including in the right periatrial region and left brachium pontis. More than 5 oligoclonal bands on lumbar puncture confirmed the diagnosis.

He was treated with IV methylprednisolone followed by a 2-week prednisone taper. Within 1 week, he returned to the psychiatric unit with worsening symptoms and received a second dose of IV steroids and plasma exchange treatment. In the following months, he completed a course of rituximab infusions and physical therapy for his dysarthria, gait abnormality, and vision impairment.

His social history was notable for multiple first-degree relatives with schizophrenia. He reported a history of sexual and verbal abuse and attempted suicide once at age 13 years by hanging himself with a bathrobe. He left home at age 18 years to serve in the Marine Corps (2001-2006). His service included deployment to Afghanistan, where he received a purple heart. Upon his return, he received BA and MS degrees. He married and had 2 daughters but became estranged from his wife. By his most recent admission, he was unemployed and living with his half-sister.

On the first day of this most recent psychiatric hospitalization, he was restarted on aripiprazole 10 mg daily, and a medicine consult was sought to evaluate the progression of his MS. No new onset neurologic symptoms were noted, but he had possible residual lower extremity hyperreflexia and tandem gait incoordination. The episodes of psychotic and neurologic symptoms appeared independent, given that his psychiatric history preceded the onset of his MS.

The patient reported no visual hallucinations starting day 2, and he no longer endorsed auditory hallucinations by day 3. However, he continued to appear internally preoccupied and was noticed to be pacing around the unit. On day 4 he presented with newly pressured speech and flights of ideas, while his affect remained euthymic and his sleep stayed consistent. In combination with his ongoing pacing, his newfound symptoms were hypothesized to be possibly akathisia, an adverse effect (AE) of aripiprazole. As such, on day 5 his dose was lowered to 5 mg daily. He continued to report no hallucinations and demonstrated progressively increased emotional range. A MRI scan was done on day 6 in case a new lesion could be identified, suggesting a primary MS flare-up; however, the scan identified no enhancing lesions, indicating no ongoing demyelination. After a neurology consult corroborated this conclusion, he was discharged in stable condition on day 7.

As is the case with the majority of patients with MS-induced psychosis, he continued to have relapsing psychiatric disease even after MS treatment had been started. Unfortunately, because this patient had stopped taking his atypical antipsychotic medication several weeks prior to his hospitalization, we cannot clarify whether his psychosis stems from a primary psychiatric vs MS process.

 

 

Discussion

Presently, treatment preferences for MS-related psychosis are divided between atypical antipsychotics and glucocorticoids. Some suggest that the treatment remains similar between MS-related psychosis and primary psychotic disorders in that atypical antipsychotics are the standard of care.12 A variety of atypical antipsychotics have been used successfully in case reports, including zipradisone, risperidone, olanzapine, quetiapine, and aripiprazole.13,14 First-generation antipsychotics and other psychotropic drugs that can precipitate extra-pyramidal AEs are not recommended given their potential additive effect to motor deficits associated with MS.12 Alternatively, several case reports have found that MS-related psychotic symptoms respond to glucocorticoids more effectively, while cautioning that glucocorticoids can precipitate psychosis and depression.15,16 One review article found that 90% of patients who received corticosteroids saw an improvement in their psychotic symptoms.2

Finally, it is possible that our patient’s neuropsychiatric symptoms can be explained by autoantibody formation in response to toxin exposure during his time in Afghanistan. In a pilot study of veterans with GWI, Abou-Donia and colleagues found 2-to-9 fold increase in autoantibody reactivity levels of the following neuronal and glial-specific proteins relative to healthy controls: neurofilament triplet proteins, tubulin, microtubule-associated tau proteins, microtubule-associated protein-2, myelin basic protein, myelin-associated glycoprotein, glial fibrillary acidic protein, and calcium-calmodulin kinase II.17,18 Many of these autoantibodies are longstanding explicit markers for neurodegenerative disorders, given that they target proteins and antigens that support axonal transport and myelination. Still Gulf War veteran status has yet to be explicitly linked to an increased risk of MS,19 making this hypothesis less likely for our patient. Future research should address the clinical and therapeutic implications of different autoantibody levels in combat veterans with psychosis.

Conclusion

For patients with MS, mood disorder and psychotic symptoms should warrant a MRI given the possibility of a psychiatric manifestation of MS relapse. Ultimately, our patient’s presentation was inconsistent with the expected clinical presentations of both a primary psychotic disorder and psychosis as a manifestation of MS. His late age at his first psychotic break is atypical for primary psychotic disease, and the lack of MRI imaging done at his initial psychotic episodes cannot exclude a primary MS diagnosis. Still, his lack of MRI findings at his most recent hospitalization, negative symptomatology, and strong history of schizophrenia make a primary psychotic disorder likely.

Following his future clinical course will be necessary to determine the etiology of his psychotic episodes. Future episodes of psychosis with neurologic symptoms would suggest a primary MS diagnosis and potential benefit of immunosuppressant treatment, whereas repeated psychotic breaks with minimal temporal lobe involvement or demyelination as seen on MRI would be suspicious for separate MS and psychotic disease processes. Further research on treatment regimens for patients experiencing psychosis as a manifestation of MS is still necessary.

References

1. Wallin MT, Culpepper WJ, Campbell JD, et al. The prevalence of MS in the United States: A population-based estimate using health claims data. Neurology. 2019;92(10):e1029-e1040.

2. Camara-Lemarroy CR, Ibarra-Yruegas BE, Rodriguez-Gutierrez R, Berrios-Morales I, Ionete C, Riskind P. The varieties of psychosis in multiple sclerosis: a systematic review of cases. Mult Scler Relat Disord. 2017;12:9-14.

3. Cottrel SS, Wilson SA. The affective symptomatology of disseminated sclerosis: a study of 100 cases. J Neurol Psychopathology. 1926;7(25):1-30.

4. Johansson V, Lundholm C, Hillert J, et al. Multiple sclerosis and psychiatric disorders: comorbidity and sibling risk in a nationwide Swedish cohort. Mult Scler. 2014;20(14):1881-1891.

5. Rossi S, Studer V, Motta C, et al. Neuroinflammation drives anxiety and depression in relapsing-remitting multiple sclerosis. Neurology. 2017;89(13):1338-1347.

6. Gilberthorpe TG, O’Connell KE, Carolan A, et al. The spectrum of psychosis in multiple sclerosis: a clinical case series. Neuropsychiatric disease and treatment. 2017;13:303.

7. Patten SB, Svenson LW, Metz LM. Psychotic disorders in MS: population-based evidence of an association. Neurology 2005;65(7):1123-1125.

8. Kosmidis MH, Giannakou M, Messinis L, Papathanasopoulos P. Psychotic features associated with multiple sclerosis. Int Rev Psychiatry. 2010; 22(1):55-66.

9. US Department of Veterans Affairs. Public health: military exposures. https://www.publichealth.va.gov/exposures/. Updated April 16, 2019. Accessed May 13, 2019.

10. DeBeer BB, Davidson D, Meyer EC, Kimbrel NA, Gulliver SB, Morissette SB. The association between toxic exposures and chronic multisymptom illness in veterans of the wars of Iraq and Afghanistan. J Occup Environ Med. 2017;59(1):54-60.

11. Kang HK, Li B, Mahan CM, Eisen SA, Engel CC. Health of US veterans of 1991 Gulf War: a follow-up survey in 10 years. J Occup Environ Med. 2009;51(4):401-410.

12. Murphy R, O’Donoghue S, Counihan T, et al. Neuropsychiatric syndromes of multiple sclerosis. J Neurol Neurosurg Psychiatry. 2017;88(8):697-708.

13. Davids E, Hartwig U, Gastpar, M. Antipsychotic treatment of psychosis associated with multiple sclerosis. Prog Neuro Psychopharmacol Biol Psychiatry. 2004;28(4):743-744.

14. Lo Fermo S, Barone R, Patti F, et al. Outcome of psychiatric symptoms presenting at onset of multiple sclerosis: a retrospective study. Mult Scler. 2010;16(6):742-748.

15. Enderami A, Fouladi R, Hosseini HS. First-episode psychosis as the initial presentation of multiple sclerosis: a case report. Int Medical Case Rep J. 2018;11:73-76.

16. Fragoso YD, Frota ER, Lopes JS, et al. Severe depression, suicide attempts, and ideation during the use of interferon beta by patients with multiple sclerosis. Clin Neuropharmacol. 2010;33(6):312-316.

17. Abou-Donia MB, Conboy LA, Kokkotou E, et al. Screening for novel central nervous system biomarkers in veterans with Gulf War Illness. Neurotoxicol Teratol. 2017;61:36-46.

18. Abou-Donia MB, Lieberman A, Curtis L. Neural autoantibodies in patients with neurological symptoms and histories of chemical/mold exposures. Toxicol Ind Health. 2018;34(1):44-53.

19. Wallin MT, Kurtzke JF, Culpepper WJ, et al. Multiple sclerosis in Gulf War era veterans. 2. Military deployment and risk of multiple sclerosis in the first Gulf War. Neuroepidemiology. 2014;42(4):226-234.

References

1. Wallin MT, Culpepper WJ, Campbell JD, et al. The prevalence of MS in the United States: A population-based estimate using health claims data. Neurology. 2019;92(10):e1029-e1040.

2. Camara-Lemarroy CR, Ibarra-Yruegas BE, Rodriguez-Gutierrez R, Berrios-Morales I, Ionete C, Riskind P. The varieties of psychosis in multiple sclerosis: a systematic review of cases. Mult Scler Relat Disord. 2017;12:9-14.

3. Cottrel SS, Wilson SA. The affective symptomatology of disseminated sclerosis: a study of 100 cases. J Neurol Psychopathology. 1926;7(25):1-30.

4. Johansson V, Lundholm C, Hillert J, et al. Multiple sclerosis and psychiatric disorders: comorbidity and sibling risk in a nationwide Swedish cohort. Mult Scler. 2014;20(14):1881-1891.

5. Rossi S, Studer V, Motta C, et al. Neuroinflammation drives anxiety and depression in relapsing-remitting multiple sclerosis. Neurology. 2017;89(13):1338-1347.

6. Gilberthorpe TG, O’Connell KE, Carolan A, et al. The spectrum of psychosis in multiple sclerosis: a clinical case series. Neuropsychiatric disease and treatment. 2017;13:303.

7. Patten SB, Svenson LW, Metz LM. Psychotic disorders in MS: population-based evidence of an association. Neurology 2005;65(7):1123-1125.

8. Kosmidis MH, Giannakou M, Messinis L, Papathanasopoulos P. Psychotic features associated with multiple sclerosis. Int Rev Psychiatry. 2010; 22(1):55-66.

9. US Department of Veterans Affairs. Public health: military exposures. https://www.publichealth.va.gov/exposures/. Updated April 16, 2019. Accessed May 13, 2019.

10. DeBeer BB, Davidson D, Meyer EC, Kimbrel NA, Gulliver SB, Morissette SB. The association between toxic exposures and chronic multisymptom illness in veterans of the wars of Iraq and Afghanistan. J Occup Environ Med. 2017;59(1):54-60.

11. Kang HK, Li B, Mahan CM, Eisen SA, Engel CC. Health of US veterans of 1991 Gulf War: a follow-up survey in 10 years. J Occup Environ Med. 2009;51(4):401-410.

12. Murphy R, O’Donoghue S, Counihan T, et al. Neuropsychiatric syndromes of multiple sclerosis. J Neurol Neurosurg Psychiatry. 2017;88(8):697-708.

13. Davids E, Hartwig U, Gastpar, M. Antipsychotic treatment of psychosis associated with multiple sclerosis. Prog Neuro Psychopharmacol Biol Psychiatry. 2004;28(4):743-744.

14. Lo Fermo S, Barone R, Patti F, et al. Outcome of psychiatric symptoms presenting at onset of multiple sclerosis: a retrospective study. Mult Scler. 2010;16(6):742-748.

15. Enderami A, Fouladi R, Hosseini HS. First-episode psychosis as the initial presentation of multiple sclerosis: a case report. Int Medical Case Rep J. 2018;11:73-76.

16. Fragoso YD, Frota ER, Lopes JS, et al. Severe depression, suicide attempts, and ideation during the use of interferon beta by patients with multiple sclerosis. Clin Neuropharmacol. 2010;33(6):312-316.

17. Abou-Donia MB, Conboy LA, Kokkotou E, et al. Screening for novel central nervous system biomarkers in veterans with Gulf War Illness. Neurotoxicol Teratol. 2017;61:36-46.

18. Abou-Donia MB, Lieberman A, Curtis L. Neural autoantibodies in patients with neurological symptoms and histories of chemical/mold exposures. Toxicol Ind Health. 2018;34(1):44-53.

19. Wallin MT, Kurtzke JF, Culpepper WJ, et al. Multiple sclerosis in Gulf War era veterans. 2. Military deployment and risk of multiple sclerosis in the first Gulf War. Neuroepidemiology. 2014;42(4):226-234.

Issue
Federal Practitioner - 36(4)s
Issue
Federal Practitioner - 36(4)s
Page Number
S32-S35
Page Number
S32-S35
Publications
Publications
Topics
Article Type
Display Headline
Understanding Psychosis in a Veteran With a History of Combat and Multiple Sclerosis
Display Headline
Understanding Psychosis in a Veteran With a History of Combat and Multiple Sclerosis
Sections
Citation Override
Fed Pract. 2019 June;36(suppl 4):S32-S35
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.
Article PDF Media

Early and Accurate Identification of Parkinson Disease Among US Veterans (FULL)

Article Type
Changed
Mon, 06/01/2020 - 15:23
Display Headline
Early and Accurate Identification of Parkinson Disease Among US Veterans
Early and accurate identification and management of veterans at risk for Parkinson disease is an important priority area for the US Department of Veterans Affairs because of the substantial impact on quality of life and disability-adjusted life years.

Parkinson disease (PD) affects about 680,000 in the US, including > 110,000 veterans (Caroline Tanner, MD, PhD, unpublished data).1 In the next 10 years, this number is expected to double, in part because of the aging of the US population.1 Although the classic diagnostic criteria emphasize motor symptoms that include tremor, gait disturbance, and paucity of movement, there is increasing recognition that disease pathology begins decades before the development of motor impairment.2

Pathologic studies confirm that by the onset of motor symptoms, at least 30% of nigrostriatal neurons are lost or dysfunctional.3-5 Similarly, the Braak staging hypothesis posits initial deposition of Lewy bodies in the olfactory bulb and the dorsal motor nucleus of the vagus nerve, followed by prion-like spread through the brain stem into the midbrain/substantia nigra, and finally into the cortex (Figure 1).6

The decades-long prodromal or preclinical phase represents a unique opportunity for early identification of those at highest risk for developing the motor symptoms of Parkinson disease.7 Accurate identification, ideally before the onset of manifest motor disability, would not only improve prognostic counseling of veterans and families, but also could allow for early enrollment into trials of potentially disease-modifying therapeutic agents. Thus, early and accurate identification of PD is an important goal of the care of veterans with potential PD.

 

Prodromal Symptoms

Prodromal PD, as defined by the International Parkinson Disease and Movement Disorders Society (MDS), focuses on nonmotor symptoms that herald the onset of manifest motor PD.8 The most commonly assessed nonmotor features include olfaction, constipation, sleep disturbance, and mood disorders.

Olfaction is impaired in > 90% of patients with motor PD at the time of diagnosis; by contrast, the prevalence of hyposmia in the general population ranges from 20% to 50%, with higher rates in older adults and in smokers.9-11 Thus, olfaction appears to be a relatively sensitive, though nonspecific, prodromal feature. Importantly, subjective report of hyposmia is poorly reliable, so a number of different tests have been developed for objective assessment of olfactory dysfunction.12 The 12-item Brief Smell Identification Test (B-SIT), derived from the longer University of Pennsylvania Smell Identification Test, is a “scratch-and-sniff” forced multiple choice test that can be self-administered by cooperative patients.13,14 The B-SIT has been validated in multiple ethnic and cultural groups and shows high discrimination between PD subjects and controls.13,15 Of note, olfactory impairment appears to be associated with risk of cognitive decline in PD, further emphasizing the need for accurate assessment to guide prognosis.16

Like hyposmia, constipation can be noted long before the diagnosis of manifest motor PD.17 After adjustment for lifestyle factors, constipated individuals have up to 4.5-fold increased odds of developing PD, and those with constipation suffer worsened disease outcomes and health-related quality of life.17-20 Some groups have demonstrated alterations in gut microbiota of those with prodromal PD, which suggests local inflammatory processes and intestinal permeability may contribute to protein misfolding and disease development.21,22 This also raises the intriguing possibility that dietary alterations may be neuroprotective or neurorestorative, although this has yet to be tested in humans.23,24

Like constipation, mood changes can precede the appearance of manifest motor PD.25,26 Case control studies suggest a higher risk of developing PD among individuals who were previously diagnosed with depression or anxiety, particularly in the 1 to 2 years prior to PD diagnosis.27-29 Both apathy and anxiety are associated with striatal dopamine dysfunction, particularly in the right caudate nucleus, which suggests that mood changes are directly related to disease pathology.30,31

 

 

Of the prodromal features, rapid eye movement sleep behavior disorder (RBD) is associated with the highest risk of conversion to motor PD.8 Up to 80% of older men with socalled idiopathic RBD develop a parkinsonian syndrome within 20 years; risk is divided about equally between idiopathic PD and dementia with Lewy bodies (DLB).32 Collateral history from a bed-partner is usually sufficient to make the diagnosis, although, this is often confounded by the prevalence of nightmares in those with posttraumatic stress disorder in the veteran population.32 Thus, in suspected cases, obtaining a polysomnogram can aid in distinguishing between idiopathic PC and DLB.33 Given the specificity of RBD as a marker of synuclein deposition and the high risk of progression to a degenerative syndrome, accurate diagnosis and counseling is imperative.

Each of the prodromal nonmotor features of PD are at best moderately sensitive or specific in isolation, but in concert, they can be used to develop a Parkinson risk score. For instance, the MDS prodromal criteria combine individual likelihood ratios into Bayesian analysis to determine a combined probability of PD, which can be further stratified to probable or possible prodromal PD (probability > 80%, > 50%, respectively).8 These criteria have been applied to several independent cohorts and demonstrate high sensitivity and specificity, especially over time.34,35 Applicability in a veteran population has yet to be determined.

Use of Imaging in Diagnosis

Although clinical diagnostic criteria and prodromal features can improve diagnostic accuracy, it can be extremely challenging to distinguish idiopathic PD from nondegenerative parkinsonism or atypical syndromes (see below). Compared with the gold standard of pathologic assessment, the clinical diagnostic accuracy for PD ranges from 73% for nonexperts to 80% for fellowship-trained movement disorders specialists.36 Thus, objective biomarkers are sought to improve diagnostic accuracy both for clinical care as well as for research purposes, such as enrollment into clinical trials.

Multiple potential imaging biomarkers for preclinical PD can aid in early diagnosis and help differentiate PD from related but distinct disorders. While beyond the scope of this review, these techniques have recently been reviewed.7 Of these, the most widely available and accurate is dopamine transporter (DAT) imaging, which uses a radioiodinated ligand that binds to DAT on striatal dopaminergic terminals; binding is detected through single photon emission computed tomography (SPECT) scanning. Thus, a SPECT DaTscan (GE Healthcare Bio-Sciences, Little Chalfont, England) directly assesses the integrity of the presynaptic nigrostriatal system and is well correlated with severity of motor and nonmotor parkinsonism.37,38

In individuals with suspected prodromal PD, abnormal DaTscans are associated with faster progression to manifest motor PD.39 However, it should be noted that a number of medications, several of which are commonly utilized in the veteran population, can affect the outcome of a DaTscan.40 Some of these medications only mildly affect the outcome, so the physician interpreting the scan should be made aware of their use, while others need to be held for days to weeks so as not to invalidate the DaTscan. DaTscan also do not differentiate between PD and atypical degenerative parkinsonisms such as multiple system atrophy (MSA), DLB, progressive supranuclear palsy (PSP), or corticobasal syndrome (CBS). Nevertheless, these scans can be used to distinguish degenerative parkinsonisms from other conditions that can be difficult to distinguish clinically from PD, including essential tremor, normal pressure hydrocephalus, vascular parkinsonism, or druginduced parkinsonism (DIP).

DIP usually is caused by blockade of postsynaptic dopamine receptors by antipsychotic medications, which are prescribed to as many as 1 in 4 older veterans; antiemetic agents such as metoclopramide are also potential offenders if used chronically.41 The risk of DIP appears to be associated with the D2 binding affinity of the drug. Thus, of the newer atypical antipsychotics, clozapine and quetiapine appear to have the lowest risk, while ziprasidone and aripiprazole have the highest binding affinity and therefore the highest risk.42 In many patients, parkinsonism persists even after discontinuation of the offending agent, suggesting that in at least a subset of patients, DIP may be an “unmasking” of latent PD rather than a true adverse effect of the medication. The prodromal features discussed above can be used to distinguish isolated DIP from unmasked latent PD.43 In a study we conducted in veterans at the Michael J. Crescenz VA Medical Center in Philadelphia, Pennsylvania, hyposmia in particular was shown to be highly predictive of an underlying dopaminergic deficit with an odds ratio of 63.44

Other important considerations in the differential diagnosis of PD are the atypical degenerative parkinsonian syndromes, formerly called Parkinson plus syndromes. These may be further divided into the synucleinopathies (MSA, DLB) or the tauopathies (PSP, CBS), depending on the predominant amyloidogenic protein. Early in the disease, the atypical syndromes and idiopathic PD may be clinically indistinguishable, although the atypical syndromes tend to progress more rapidly and often have a less robust response to levodopa.

 

 

Radiologic and fluid biomarkers for the atypical syndromes are under active investigation; at present the most accessible study is magnetic resonance imaging (MRI), which may show characteristic features such as degeneration of the pontocerebellar fibers in MSA or midbrain atrophy in PSP.45,46 By contrast, standard MRI sequences in idiopathic PD are usually normal, although high-resolution (7 tesla) imaging can reveal loss of neuromelanin in the substantia nigra.47 MRI also can be useful in the workup of suspected normal pressure hydrocephalus or vascular parkinsonism, which would show disproportionate ventriculomegaly with transependymal flow, or white matter lesions in the basal ganglia, respectively.

Data-Based Identification of Preclinical PD

The integration of clinical motor or prodromal features with biomarker data has led to the development of several large-scale clinical and administrative databases to identify PD. The Parkinson Progression Markers Initiative initially enrolled only de novo clinically identified people with PD, but it expanded to include a prodromal cohort who are being assessed for rates of conversion to PD.48 Similarly, metabolic imaging can be combined with prodromal symptoms, such as hyposmia or RBD, to predict risk for phenoconversion into manifest motor PD.49

The PREDICT-PD study synthesizes mood symptoms, RBD, smell testing, genotyping, and keyboard-tapping tasks to divide individuals into high-, middle-, and low-risk groups; interim analysis at 3 years of follow-up (N = 842) demonstrated a hazard ratio of 4.39 (95% CI, 1.03-18.68) for the diagnosis of PD in the highrisk group compared with the low-risk group.50 Lastly, administrative claims data for prodromal features, such as constipation, RBD, and mood symptoms, is highly predictive of eventual PD diagnosis.51 VA databases accessed through the Corporate Data Warehouse are complementary sources of information to nonveteranspecific Medicare databases; to our knowledge there has not yet been a comprehensive search of VA databases to identify veterans with preclinical PD.

Risk Factors Associated With Military Service

A number of potential environmental risk factors may increase the risk of developing Parkinson disease for veterans. Perhaps the most commonly recognized is pesticide exposure, particularly given the presumptive service connections established by the VA for Parkinson disease and exposure to Agent Orange or contaminated water at Camp Lejeune.52,53 Both dioxin, the toxic ingredient in Agent Orange, and the solvents trichloroethylene and perchloroethylene, found in the water supply at Camp Lejeune, interfere with mitochondrial function leading to oxidative stress and apoptosis of nigrostriatal neurons.54,55 Other potential exposures, which are not necessarily limited to the veteran population, include rotenone, a phytochemical used to kill fish in reservoirs, and paraquat, an herbicide that may directly promote synuclein aggregation.56,57 Veterans who have reported exposure to these or other environmental chemicals in civilian life should be carefully assessed for the presence of motor PD or prodromal features.

Traumatic brain injury (TBI) also may be a risk factor for PD, which may be particularly relevant for veterans who had served in Iraq or Afghanistan. Retrospective claims data suggest a strong association between PD and recent TBI in the 5 to 10 years prior to motor PD diagnosis.58,59 A recent assessment of combat veterans with TBI found that even mild TBI was associated with a 56% increased risk of PD, while moderate-to-severe TBI was associated with an 83% higher risk of PD.60 The pathologic mechanism for this link is unclear, but post-TBI inflammatory processes may lead to the formation of reactive oxygen species and/or glutamatergic excitotoxicity, thus leading to secondary injury in the nigrostriatal pathway.61 As with prodromal symptoms, the risk of PD related to environmental risk factors may be synergistic; repetitive TBI may be more damaging than a single injury, and a combination of TBI and pesticide exposure markedly increases PD risk beyond the risk of TBI or the risk of pesticides alone.62 Recently, parkinsonism, including Parkinson disease, was recognized as a service connected condition for veterans with a servicerelated moderate or severe TBI.63

Conclusion

Because of the substantial impact on quality of life and disability-adjusted life years, early and accurate identification and management of veterans at risk for PD is an important priority area for the VA. The 10-year cost of PD-related benefits through the VA was estimated at $3.5 billion in fiscal year 2010, and that number is likely to rise in coming years, due to the aging population as well as synergistic effects of independent risk factors described above.64 In response, the VA has created a network of specialty care sites, known as Parkinson Disease Research, Education, and Clinical Centers (PADRECCs) located in Philadelphia, Pennsylvania; Richmond, Virginia; Houston, Texas; West Los Angeles and San Francisco, California; and Seattle, Washington/ Portland, Oregon (www.parkinsons.va.gov).

The PADRECCs are supplemented by a National VA PD Consortium network of VA physicians trained in PD management (Figure 2). Studies, including one investigating care of veterans with PD, have demonstrated that involvement of specialty care services early in the course of PD leads to improved patient outcomes.65,66 In addition to patient-facing resources such as support groups and specialized physical/occupational/speech therapy, PADRECCs and the consortium sites are national leaders in PD education and clinical trials and provide high-quality, multidisciplinary care for veterans with PD.67 Thus, veterans with significant risk factors or prodromal symptoms of PD should be referred into the PADRECC/Consortium network in order to maximize their quality of care and quality of life.

References

1.  Marras C, Beck JC, Bower JH, et al; Parkinson’s Foundation P4 Group. Prevalence of Parkinson’s disease across North America. NPJ Parkinsons Dis. 2018;4:21.

2.  Postuma RB, Berg D, Stern M, et al. MDS clinical diagnostic criteria for Parkinson’s disease. Mov Disord. 2015;30(12):1591-1601.

3.  Fearnley JM, Lees AJ. Ageing and Parkinson’s disease: substantia nigra regional selectivity. Brain. 1991;114(pt 5):2283-2301.

4.  Greffard S, Verny M, Bonnet A-M, et al. Motor score of the Unified Parkinson Disease Rating Scale as a good predictor of Lewy body-associated neuronal loss in the substantia nigra. Arch Neurol. 2006;63(4):584-588.

5.  Hilker R, Schweitzer K, Coburger S, et al. Nonlinear progression of Parkinson disease as determined by serial positron emission tomographic imaging of striatal fluorodopa F 18 activity. Arch Neurol. 2005;62(3):378-382.

6.  Braak H, Del Tredici K, Rüb U, de Vos RAI, Jansen Steur ENH, Braak E. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol Aging. 2003;24(2):197-211.

7.  Mantri S, Morley JF, Siderowf AD. The importance of preclinical diagnostics in Parkinson disease. Parkinsonism Relat Disord. 2018;pii:S1353-8020(18)30396-1. [Epub ahead of print]

8.  Berg D, Postuma RB, Adler CH, et al. MDS research criteria for prodromal Parkinson’s disease. Mov Disord. 2015;30(12):1600-1611.

9.  Haehner A, Boesveldt S, Berendse HW, et al. Prevalence of smell loss in Parkinson’s disease – a multicenter study. Parkinsonism Relat Disord. 2009;15(7):490-494.

10.  Mullol J, Alobid I, Mariño-Sánchez F, et al. Furthering the understanding of olfaction, prevalence of loss of smell and risk factors: a population-based survey (OLFACAT study). BMJ Open. 2012;2(6).pii:e001256.

11.  Doty RL, Shaman P, Applebaum SL, Giberson R, Siksorski L, Rosenberg L. Smell identification ability: changes with age. Science. 1984;226(4681):1441-1443.

12.  Doty RL. Olfactory dysfunction in Parkinson disease. Nat Rev Neurol. 2012;8(6):329-339.

13.  Double KL, Rowe DB, Hayes M, et al. Identifying the pattern of olfactory deficits in Parkinson disease using the brief smell identification test. Arch Neurol. 2003;60(4):545-549.

14.  Doty RL, Shaman P, Dann M. Development of the University of Pennsylvania Smell Identification Test: a standardized microencapsulated test of olfactory function. Physiol Behav. 1984;32(3):489-502.

15.  Morley JF, Cohen A, Silveira-Moriyama L, et al. Optimizing olfactory testing for the diagnosis of Parkinson’s disease: item analysis of the University of Pennsylvania smell identification test. NPJ Parkinsons Dis. 2018;4:2.

16.  Fullard ME, Tran B, Xie SX, et al. Olfactory impairment predicts cognitive decline in early Parkinson’s disease. Parkinsonism Relat Disord. 2016;25:45-51.

17.  Savica R, Carlin JM, Grossardt BR, et al. Medical records documentation of constipation preceding Parkinson disease: a case-control study. Neurology. 2009;73(21):1752-1758.

18.  Abbott RD, Petrovitch H, White LR, et al. Frequency of bowel movements and the future risk of Parkinson’s disease. Neurology. 2001;57(3):456-462.

19.  Stocchi F, Torti M. Constipation in Parkinson’s disease. Int Rev Neurobiol. 2017;134:811-826.

20.  Yu QJ, Yu SY, Zuo LJ, et al. Parkinson disease with constipation: clinical features and relevant factors. Sci Rep. 2018;8(1):567.

21.  Hill-Burns EM, Debelius JW, Morton JT, et al. Parkinson’s disease and Parkinson’s disease medications have distinct signatures of the gut microbiome. Mov Disord. 2017;32(5):739-749.

22.  Mulak A, Bonaz B. Brain-gut-microbiota axis in Parkinson’s disease. World J Gastroenterol. 2015;21(37): 10609-10620.

23.  Shah SP, Duda JE. Dietary modifications in Parkinson’s disease: a neuroprotective intervention? Med Hypotheses. 2015;85(6):1002-1005.

24.  Perez-Pardo P, de Jong EM, Broersen LM, et al. Promising effects of neurorestorative diets on motor, cognitive, and gastrointestinal dysfunction after symptom development in a mouse model of Parkinson’s disease. Front Aging Neurosci. 2017;9:57.

25.  Fang F, Xu Q, Park Y, et al. Depression and the subsequent risk of Parkinson’s disease in the NIH-AARP Diet and Health Study. Mov Disord. 2010;25(9):1157-1162.

26.  Leentjens AFG, Van den Akker M, Metsemakers JFM, Lousberg R, Verhey FRJ. Higher incidence of depression preceding the onset of Parkinson’s disease: a register study. Mov Disord. 2003;18(4):414-418.

27.  Alonso A, Rodriguez LAG, Logroscino G, Hernán MA. Use of antidepressants and the risk of Parkinson’s disease: a prospective study. J Neurol Neurosurg Psychiatry. 2009;80(6):671-674.

28.  Weisskopf MG, Chen H, Schwarzschild MA, Kawachi I, Ascherio A. Prospective study of phobic anxiety and risk of Parkinson’s disease. Mov Disord. 2003;18(6):646-651.

29.  Darweesh SK, Verlinden VJ, Stricker BH, Hofman A, Koudstaal PJ, Ikram MA. Trajectories of prediagnostic functioning in Parkinson’s disease. Brain. 2017;140(2):429-441.

30.  Santangelo G, Vitale C, Picillo M, et al. Apathy and striatal dopamine transporter levels in de-novo, untreated Parkinson’s disease patients. Parkinsonism Relat Disord. 2015;21(5):489-493.

31.  Erro R, Pappatà S, Amboni M, et al. Anxiety is associated with striatal dopamine transporter availability in newly diagnosed untreated Parkinson’s disease patients. Parkinsonism Relat Disord. 2012;18(9):1034-1038.

32.  Schenck CH, Boeve BF, Mahowald MW. Delayed emergence of a parkinsonian disorder or dementia in 81% of older men initially diagnosed with idiopathic rapid eye movement sleep behavior disorder: a 16-year update on a previously reported series. Sleep Med. 2013;14(8):
744-748.

33.  Melendez J, Hesselbacher S, Sharafkhaneh A, Hirshkowitz M. Assessment of REM sleep behavior disorder in veterans with posttraumatic stress disorder. Chest. 2011;140(4):967A.

34.  Pilotto A, Heinzel S, Suenkel U, et al. Application of the movement disorder society prodromal Parkinson’s disease research criteria in 2 independent prospective cohorts. Mov Disord. 2017;32(7):1025-1034.

35.  Fereshtehnejad S-M, Montplaisir JY, Pelletier A, Gagnon J-F, Berg D, Postuma RB. Validation of the MDS research criteria for prodromal Parkinson’s disease: Longitudinal assessment in a REM sleep behavior disorder (RBD) cohort. Mov Disord. 2017;32(6):865-873.

36. Rizzo G, Copetti M, Arcuti S, Martino D, Fontana A, Logroscino G. Accuracy of clinical diagnosis of Parkinson disease: a systematic review and meta-analysis. Neurology. 2016;86(6):566-576.

37.  Moccia M, Pappatà S, Picillo M, et al. Dopamine transporter availability in motor subtypes of de novo drug-naïve Parkinson’s disease. J Neurol. 2014;261(11):2112-2118.

38.  Siepel FJ, Brønnick KS, Booij J, et al. Cognitive executive impairment and dopaminergic deficits in de novo Parkinson’s disease. Mov Disord. 2014;29(14):1802-1808.

39. Iranzo A, Valldeoriola F, Lomeña F, et al. Serial dopamine transporter imaging of nigrostriatal function in patients with idiopathic rapid-eye-movement sleep behaviour disorder: a prospective study. Lancet Neurol. 2011;10(9):797-805.

40.  Booij J, Kemp P. Dopamine transporter imaging with [(123)I]FP-CIT SPECT: potential effects of drugs. Eur J Nucl Med Mol Imaging. 2008;35(2):424-438.

41.  Gellad WF, Aspinall SL, Handler SM, et al. Use of antipsychotics among older residents in VA nursing homes. Med Care. 2012;50(11):954-960.

42.  Mauri MC, Paletta S, Maffini M, et al. Clinical pharmacology of atypical antipsychotics: an update. EXCLI J. 2014;13:1163-1191.

43.  Morley JF, Duda JE. Use of hyposmia and other non-motor symptoms to distinguish between drug-induced parkinsonism and Parkinson’s disease. J Parkinsons Dis. 2014;4(2):169-173.

44.  Morley JF, Cheng G, Dubroff JG, Wood S, Wilkinson JR, Duda JE. Olfactory impairment predicts underlying dopaminergic deficit in presumed drug-induced parkinsonism. Mov Disord Clin Pract. 2017;4(4):603-606.

45.  Whitwell JL, Höglinger GU, Antonini A, et al; Movement Disorder Society-endorsed PSP Study Group. Radiological biomarkers for diagnosis in PSP: where are we and where do we need to be? Mov Disord. 2017;32(7):955-971.

46.  Laurens B, Constantinescu R, Freeman R, et al. Fluid biomarkers in multiple system atrophy: A review of the MSA Biomarker Initiative. Neurobiol Dis. 2015;80:29-41.

47.  Barber TR, Klein JC, Mackay CE, Hu MTM. Neuroimaging in pre-motor Parkinson’s disease. NeuroImage Clin. 2017;15:215-227.

48.  Parkinson Progression Marker Initiative. The Parkinson Progression Marker Initiative (PPMI). Prog Neurobiol. 2011;95(4):629-635.

49.  Meles SK, Vadasz D, Renken RJ, et al. FDG PET, dopamine transporter SPECT, and olfaction: combining biomarkers in REM sleep behavior disorder. Mov Disord. 2017;32(10):1482-1486.

50.  Noyce AJ, R’Bibo L, Peress L, et al. PREDICT‐PD: an online approach to prospectively identify risk indicators of Parkinson’s disease. Mov Disord. 2017 Feb; 32(2): 219–226.

51.  Searles Nielsen S, Warden MN, Camacho-Soto A, Willis AW, Wright BA, Racette BA. A predictive model to identify Parkinson disease from administrative claims data. Neurology. 2017;89(14):1448-1456.

52.  Institute of Medicine. Veterans and Agent Orange: Update 2012. National Academies Press: Washington, DC; 2013.

53.  Department of Veterans Affairs. Diseases associated with exposure to Contaminants in the Water Supply at Camp Lejeune. Final rule. Fed Regist. 2017;82(9):4173-4185.

54.  Goldman SM, Quinlan PJ, Ross GW, et al. Solvent exposures and Parkinson disease risk in twins. Ann Neurol. 2012;71(6):776-784.

55.  Liu M, Shin EJ, Dang DK, et al. Trichloroethylene and Parkinson’s disease: risk assessment. Mol Neurobiol. 2018;55(7):6201-6214.

56.  Betarbet R, Sherer TB, MacKenzie G, Garcia-Osuna M, Panov AV, Greenamyre JT. Chronic systemic pesticide exposure reproduces features of Parkinson’s disease. Nat Neurosci. 2000;3(12):1301-1306.

57.  Manning-Bog AB, McCormack AL, Li J, Uversky VN, Fink AL, Monte DAD. The herbicide paraquat causes up-regulation and aggregation of alpha-synuclein in mice: paraquat and alpha-synuclein. J Biol Chem. 2002;277(3):1641-1644.

58.  Camacho-Soto A, Warden MN, Searles Nielsen S, et al. Traumatic brain injury in the prodromal period of Parkinson’s disease: a large epidemiological study using medicare data. Ann Neurol. 2017;82(5):744-754.

59.  Gardner RC, Burke JF, Nettiksimmons J, Goldman S, Tanner CM, Yaffe K. Traumatic brain injury in later life increases risk for Parkinson disease. Ann Neurol. 2015;77(6):987-995.

60.  Gardner RC, Byers AL, Barnes DE, Li Y, Boscardin J, Yaffe K. Mild TBI and risk of Parkinson disease: a Chronic Effects of Neurotrauma Consortium Study. Neurology. 2018;90(20):e1771-e1779.

61.  Cruz-Haces M, Tang J, Acosta G, Fernandez J, Shi R. Pathological correlations between traumatic brain injury and chronic neurodegenerative diseases. Transl Neurodegener. 2017;6:20.

62.  Lee PC, Bordelon Y, Bronstein J, Ritz B. Traumatic brain injury, paraquat exposure, and their relationship to Parkinson disease. Neurology. 2012;79(20):2061-2066.

63.  Disabilities that are proximately due to, or aggravated by, service-connected disease or injury. 38 CFR §3.310.

64.  Diseases Associated With Exposure to Certain Herbicide Agents (Hairy Cell Leukemia and Other Chronic B-Cell Leukemias, Parkinson’s Disease and Ischemic Heart Disease). Federal Regist. 2010;75(173):53202-53216. To be codified at 38 CFR §3.

65.  Cheng EM, Swarztrauber K, Siderowf AD, et al. Association of specialist involvement and quality of care for Parkinson’s disease. Mov Disord. 2007;22(4):515-522.

66.  Qamar MA, Harington G, Trump S, Johnson J, Roberts F, Frost E. Multidisciplinary care in Parkinson’s disease. Int Rev Neurobiol. 2017;132:511-523.

67.  Pogoda TK, Cramer IE, Meterko M, et al. Patient and organizational factors related to education and support use by veterans with Parkinson’s disease. Mov Disord. 2009;24(13):1916-1924.

Article PDF
Author and Disclosure Information

Sneha Mantri is Assistant Professor of Neurology at Duke University in Durham, North Carolina. John Duda is National Parkinson’s Disease Research, Education, and Clinical Center (PADRECC) Director and Chair of the National VA Parkinson’s Disease Consortium; and James Morley is Associate Director of Research, PADRECC; both at the Corporal Michael J. Crescenz VA Medical Center in Philadelphia, Pennsylvania. John Duda is Associate Professor of Neurology and James Morley is Assistant Professor of Neurology, both at the Perelman School of Medicine, University of Pennsylvania in Philadelphia.
Correspondence: Sneha Mantri (sneha.mantri@duke.edu)

Author disclosures
The authors report no actual or potential conflicts of interest with regard to this article.

Disclaimer
The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of
its agencies.

Issue
Federal Practitioner - 36(4)s
Publications
Topics
Page Number
S18-S23
Sections
Author and Disclosure Information

Sneha Mantri is Assistant Professor of Neurology at Duke University in Durham, North Carolina. John Duda is National Parkinson’s Disease Research, Education, and Clinical Center (PADRECC) Director and Chair of the National VA Parkinson’s Disease Consortium; and James Morley is Associate Director of Research, PADRECC; both at the Corporal Michael J. Crescenz VA Medical Center in Philadelphia, Pennsylvania. John Duda is Associate Professor of Neurology and James Morley is Assistant Professor of Neurology, both at the Perelman School of Medicine, University of Pennsylvania in Philadelphia.
Correspondence: Sneha Mantri (sneha.mantri@duke.edu)

Author disclosures
The authors report no actual or potential conflicts of interest with regard to this article.

Disclaimer
The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of
its agencies.

Author and Disclosure Information

Sneha Mantri is Assistant Professor of Neurology at Duke University in Durham, North Carolina. John Duda is National Parkinson’s Disease Research, Education, and Clinical Center (PADRECC) Director and Chair of the National VA Parkinson’s Disease Consortium; and James Morley is Associate Director of Research, PADRECC; both at the Corporal Michael J. Crescenz VA Medical Center in Philadelphia, Pennsylvania. John Duda is Associate Professor of Neurology and James Morley is Assistant Professor of Neurology, both at the Perelman School of Medicine, University of Pennsylvania in Philadelphia.
Correspondence: Sneha Mantri (sneha.mantri@duke.edu)

Author disclosures
The authors report no actual or potential conflicts of interest with regard to this article.

Disclaimer
The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of
its agencies.

Article PDF
Article PDF
Related Articles
Early and accurate identification and management of veterans at risk for Parkinson disease is an important priority area for the US Department of Veterans Affairs because of the substantial impact on quality of life and disability-adjusted life years.
Early and accurate identification and management of veterans at risk for Parkinson disease is an important priority area for the US Department of Veterans Affairs because of the substantial impact on quality of life and disability-adjusted life years.

Parkinson disease (PD) affects about 680,000 in the US, including > 110,000 veterans (Caroline Tanner, MD, PhD, unpublished data).1 In the next 10 years, this number is expected to double, in part because of the aging of the US population.1 Although the classic diagnostic criteria emphasize motor symptoms that include tremor, gait disturbance, and paucity of movement, there is increasing recognition that disease pathology begins decades before the development of motor impairment.2

Pathologic studies confirm that by the onset of motor symptoms, at least 30% of nigrostriatal neurons are lost or dysfunctional.3-5 Similarly, the Braak staging hypothesis posits initial deposition of Lewy bodies in the olfactory bulb and the dorsal motor nucleus of the vagus nerve, followed by prion-like spread through the brain stem into the midbrain/substantia nigra, and finally into the cortex (Figure 1).6

The decades-long prodromal or preclinical phase represents a unique opportunity for early identification of those at highest risk for developing the motor symptoms of Parkinson disease.7 Accurate identification, ideally before the onset of manifest motor disability, would not only improve prognostic counseling of veterans and families, but also could allow for early enrollment into trials of potentially disease-modifying therapeutic agents. Thus, early and accurate identification of PD is an important goal of the care of veterans with potential PD.

 

Prodromal Symptoms

Prodromal PD, as defined by the International Parkinson Disease and Movement Disorders Society (MDS), focuses on nonmotor symptoms that herald the onset of manifest motor PD.8 The most commonly assessed nonmotor features include olfaction, constipation, sleep disturbance, and mood disorders.

Olfaction is impaired in > 90% of patients with motor PD at the time of diagnosis; by contrast, the prevalence of hyposmia in the general population ranges from 20% to 50%, with higher rates in older adults and in smokers.9-11 Thus, olfaction appears to be a relatively sensitive, though nonspecific, prodromal feature. Importantly, subjective report of hyposmia is poorly reliable, so a number of different tests have been developed for objective assessment of olfactory dysfunction.12 The 12-item Brief Smell Identification Test (B-SIT), derived from the longer University of Pennsylvania Smell Identification Test, is a “scratch-and-sniff” forced multiple choice test that can be self-administered by cooperative patients.13,14 The B-SIT has been validated in multiple ethnic and cultural groups and shows high discrimination between PD subjects and controls.13,15 Of note, olfactory impairment appears to be associated with risk of cognitive decline in PD, further emphasizing the need for accurate assessment to guide prognosis.16

Like hyposmia, constipation can be noted long before the diagnosis of manifest motor PD.17 After adjustment for lifestyle factors, constipated individuals have up to 4.5-fold increased odds of developing PD, and those with constipation suffer worsened disease outcomes and health-related quality of life.17-20 Some groups have demonstrated alterations in gut microbiota of those with prodromal PD, which suggests local inflammatory processes and intestinal permeability may contribute to protein misfolding and disease development.21,22 This also raises the intriguing possibility that dietary alterations may be neuroprotective or neurorestorative, although this has yet to be tested in humans.23,24

Like constipation, mood changes can precede the appearance of manifest motor PD.25,26 Case control studies suggest a higher risk of developing PD among individuals who were previously diagnosed with depression or anxiety, particularly in the 1 to 2 years prior to PD diagnosis.27-29 Both apathy and anxiety are associated with striatal dopamine dysfunction, particularly in the right caudate nucleus, which suggests that mood changes are directly related to disease pathology.30,31

 

 

Of the prodromal features, rapid eye movement sleep behavior disorder (RBD) is associated with the highest risk of conversion to motor PD.8 Up to 80% of older men with socalled idiopathic RBD develop a parkinsonian syndrome within 20 years; risk is divided about equally between idiopathic PD and dementia with Lewy bodies (DLB).32 Collateral history from a bed-partner is usually sufficient to make the diagnosis, although, this is often confounded by the prevalence of nightmares in those with posttraumatic stress disorder in the veteran population.32 Thus, in suspected cases, obtaining a polysomnogram can aid in distinguishing between idiopathic PC and DLB.33 Given the specificity of RBD as a marker of synuclein deposition and the high risk of progression to a degenerative syndrome, accurate diagnosis and counseling is imperative.

Each of the prodromal nonmotor features of PD are at best moderately sensitive or specific in isolation, but in concert, they can be used to develop a Parkinson risk score. For instance, the MDS prodromal criteria combine individual likelihood ratios into Bayesian analysis to determine a combined probability of PD, which can be further stratified to probable or possible prodromal PD (probability > 80%, > 50%, respectively).8 These criteria have been applied to several independent cohorts and demonstrate high sensitivity and specificity, especially over time.34,35 Applicability in a veteran population has yet to be determined.

Use of Imaging in Diagnosis

Although clinical diagnostic criteria and prodromal features can improve diagnostic accuracy, it can be extremely challenging to distinguish idiopathic PD from nondegenerative parkinsonism or atypical syndromes (see below). Compared with the gold standard of pathologic assessment, the clinical diagnostic accuracy for PD ranges from 73% for nonexperts to 80% for fellowship-trained movement disorders specialists.36 Thus, objective biomarkers are sought to improve diagnostic accuracy both for clinical care as well as for research purposes, such as enrollment into clinical trials.

Multiple potential imaging biomarkers for preclinical PD can aid in early diagnosis and help differentiate PD from related but distinct disorders. While beyond the scope of this review, these techniques have recently been reviewed.7 Of these, the most widely available and accurate is dopamine transporter (DAT) imaging, which uses a radioiodinated ligand that binds to DAT on striatal dopaminergic terminals; binding is detected through single photon emission computed tomography (SPECT) scanning. Thus, a SPECT DaTscan (GE Healthcare Bio-Sciences, Little Chalfont, England) directly assesses the integrity of the presynaptic nigrostriatal system and is well correlated with severity of motor and nonmotor parkinsonism.37,38

In individuals with suspected prodromal PD, abnormal DaTscans are associated with faster progression to manifest motor PD.39 However, it should be noted that a number of medications, several of which are commonly utilized in the veteran population, can affect the outcome of a DaTscan.40 Some of these medications only mildly affect the outcome, so the physician interpreting the scan should be made aware of their use, while others need to be held for days to weeks so as not to invalidate the DaTscan. DaTscan also do not differentiate between PD and atypical degenerative parkinsonisms such as multiple system atrophy (MSA), DLB, progressive supranuclear palsy (PSP), or corticobasal syndrome (CBS). Nevertheless, these scans can be used to distinguish degenerative parkinsonisms from other conditions that can be difficult to distinguish clinically from PD, including essential tremor, normal pressure hydrocephalus, vascular parkinsonism, or druginduced parkinsonism (DIP).

DIP usually is caused by blockade of postsynaptic dopamine receptors by antipsychotic medications, which are prescribed to as many as 1 in 4 older veterans; antiemetic agents such as metoclopramide are also potential offenders if used chronically.41 The risk of DIP appears to be associated with the D2 binding affinity of the drug. Thus, of the newer atypical antipsychotics, clozapine and quetiapine appear to have the lowest risk, while ziprasidone and aripiprazole have the highest binding affinity and therefore the highest risk.42 In many patients, parkinsonism persists even after discontinuation of the offending agent, suggesting that in at least a subset of patients, DIP may be an “unmasking” of latent PD rather than a true adverse effect of the medication. The prodromal features discussed above can be used to distinguish isolated DIP from unmasked latent PD.43 In a study we conducted in veterans at the Michael J. Crescenz VA Medical Center in Philadelphia, Pennsylvania, hyposmia in particular was shown to be highly predictive of an underlying dopaminergic deficit with an odds ratio of 63.44

Other important considerations in the differential diagnosis of PD are the atypical degenerative parkinsonian syndromes, formerly called Parkinson plus syndromes. These may be further divided into the synucleinopathies (MSA, DLB) or the tauopathies (PSP, CBS), depending on the predominant amyloidogenic protein. Early in the disease, the atypical syndromes and idiopathic PD may be clinically indistinguishable, although the atypical syndromes tend to progress more rapidly and often have a less robust response to levodopa.

 

 

Radiologic and fluid biomarkers for the atypical syndromes are under active investigation; at present the most accessible study is magnetic resonance imaging (MRI), which may show characteristic features such as degeneration of the pontocerebellar fibers in MSA or midbrain atrophy in PSP.45,46 By contrast, standard MRI sequences in idiopathic PD are usually normal, although high-resolution (7 tesla) imaging can reveal loss of neuromelanin in the substantia nigra.47 MRI also can be useful in the workup of suspected normal pressure hydrocephalus or vascular parkinsonism, which would show disproportionate ventriculomegaly with transependymal flow, or white matter lesions in the basal ganglia, respectively.

Data-Based Identification of Preclinical PD

The integration of clinical motor or prodromal features with biomarker data has led to the development of several large-scale clinical and administrative databases to identify PD. The Parkinson Progression Markers Initiative initially enrolled only de novo clinically identified people with PD, but it expanded to include a prodromal cohort who are being assessed for rates of conversion to PD.48 Similarly, metabolic imaging can be combined with prodromal symptoms, such as hyposmia or RBD, to predict risk for phenoconversion into manifest motor PD.49

The PREDICT-PD study synthesizes mood symptoms, RBD, smell testing, genotyping, and keyboard-tapping tasks to divide individuals into high-, middle-, and low-risk groups; interim analysis at 3 years of follow-up (N = 842) demonstrated a hazard ratio of 4.39 (95% CI, 1.03-18.68) for the diagnosis of PD in the highrisk group compared with the low-risk group.50 Lastly, administrative claims data for prodromal features, such as constipation, RBD, and mood symptoms, is highly predictive of eventual PD diagnosis.51 VA databases accessed through the Corporate Data Warehouse are complementary sources of information to nonveteranspecific Medicare databases; to our knowledge there has not yet been a comprehensive search of VA databases to identify veterans with preclinical PD.

Risk Factors Associated With Military Service

A number of potential environmental risk factors may increase the risk of developing Parkinson disease for veterans. Perhaps the most commonly recognized is pesticide exposure, particularly given the presumptive service connections established by the VA for Parkinson disease and exposure to Agent Orange or contaminated water at Camp Lejeune.52,53 Both dioxin, the toxic ingredient in Agent Orange, and the solvents trichloroethylene and perchloroethylene, found in the water supply at Camp Lejeune, interfere with mitochondrial function leading to oxidative stress and apoptosis of nigrostriatal neurons.54,55 Other potential exposures, which are not necessarily limited to the veteran population, include rotenone, a phytochemical used to kill fish in reservoirs, and paraquat, an herbicide that may directly promote synuclein aggregation.56,57 Veterans who have reported exposure to these or other environmental chemicals in civilian life should be carefully assessed for the presence of motor PD or prodromal features.

Traumatic brain injury (TBI) also may be a risk factor for PD, which may be particularly relevant for veterans who had served in Iraq or Afghanistan. Retrospective claims data suggest a strong association between PD and recent TBI in the 5 to 10 years prior to motor PD diagnosis.58,59 A recent assessment of combat veterans with TBI found that even mild TBI was associated with a 56% increased risk of PD, while moderate-to-severe TBI was associated with an 83% higher risk of PD.60 The pathologic mechanism for this link is unclear, but post-TBI inflammatory processes may lead to the formation of reactive oxygen species and/or glutamatergic excitotoxicity, thus leading to secondary injury in the nigrostriatal pathway.61 As with prodromal symptoms, the risk of PD related to environmental risk factors may be synergistic; repetitive TBI may be more damaging than a single injury, and a combination of TBI and pesticide exposure markedly increases PD risk beyond the risk of TBI or the risk of pesticides alone.62 Recently, parkinsonism, including Parkinson disease, was recognized as a service connected condition for veterans with a servicerelated moderate or severe TBI.63

Conclusion

Because of the substantial impact on quality of life and disability-adjusted life years, early and accurate identification and management of veterans at risk for PD is an important priority area for the VA. The 10-year cost of PD-related benefits through the VA was estimated at $3.5 billion in fiscal year 2010, and that number is likely to rise in coming years, due to the aging population as well as synergistic effects of independent risk factors described above.64 In response, the VA has created a network of specialty care sites, known as Parkinson Disease Research, Education, and Clinical Centers (PADRECCs) located in Philadelphia, Pennsylvania; Richmond, Virginia; Houston, Texas; West Los Angeles and San Francisco, California; and Seattle, Washington/ Portland, Oregon (www.parkinsons.va.gov).

The PADRECCs are supplemented by a National VA PD Consortium network of VA physicians trained in PD management (Figure 2). Studies, including one investigating care of veterans with PD, have demonstrated that involvement of specialty care services early in the course of PD leads to improved patient outcomes.65,66 In addition to patient-facing resources such as support groups and specialized physical/occupational/speech therapy, PADRECCs and the consortium sites are national leaders in PD education and clinical trials and provide high-quality, multidisciplinary care for veterans with PD.67 Thus, veterans with significant risk factors or prodromal symptoms of PD should be referred into the PADRECC/Consortium network in order to maximize their quality of care and quality of life.

Parkinson disease (PD) affects about 680,000 in the US, including > 110,000 veterans (Caroline Tanner, MD, PhD, unpublished data).1 In the next 10 years, this number is expected to double, in part because of the aging of the US population.1 Although the classic diagnostic criteria emphasize motor symptoms that include tremor, gait disturbance, and paucity of movement, there is increasing recognition that disease pathology begins decades before the development of motor impairment.2

Pathologic studies confirm that by the onset of motor symptoms, at least 30% of nigrostriatal neurons are lost or dysfunctional.3-5 Similarly, the Braak staging hypothesis posits initial deposition of Lewy bodies in the olfactory bulb and the dorsal motor nucleus of the vagus nerve, followed by prion-like spread through the brain stem into the midbrain/substantia nigra, and finally into the cortex (Figure 1).6

The decades-long prodromal or preclinical phase represents a unique opportunity for early identification of those at highest risk for developing the motor symptoms of Parkinson disease.7 Accurate identification, ideally before the onset of manifest motor disability, would not only improve prognostic counseling of veterans and families, but also could allow for early enrollment into trials of potentially disease-modifying therapeutic agents. Thus, early and accurate identification of PD is an important goal of the care of veterans with potential PD.

 

Prodromal Symptoms

Prodromal PD, as defined by the International Parkinson Disease and Movement Disorders Society (MDS), focuses on nonmotor symptoms that herald the onset of manifest motor PD.8 The most commonly assessed nonmotor features include olfaction, constipation, sleep disturbance, and mood disorders.

Olfaction is impaired in > 90% of patients with motor PD at the time of diagnosis; by contrast, the prevalence of hyposmia in the general population ranges from 20% to 50%, with higher rates in older adults and in smokers.9-11 Thus, olfaction appears to be a relatively sensitive, though nonspecific, prodromal feature. Importantly, subjective report of hyposmia is poorly reliable, so a number of different tests have been developed for objective assessment of olfactory dysfunction.12 The 12-item Brief Smell Identification Test (B-SIT), derived from the longer University of Pennsylvania Smell Identification Test, is a “scratch-and-sniff” forced multiple choice test that can be self-administered by cooperative patients.13,14 The B-SIT has been validated in multiple ethnic and cultural groups and shows high discrimination between PD subjects and controls.13,15 Of note, olfactory impairment appears to be associated with risk of cognitive decline in PD, further emphasizing the need for accurate assessment to guide prognosis.16

Like hyposmia, constipation can be noted long before the diagnosis of manifest motor PD.17 After adjustment for lifestyle factors, constipated individuals have up to 4.5-fold increased odds of developing PD, and those with constipation suffer worsened disease outcomes and health-related quality of life.17-20 Some groups have demonstrated alterations in gut microbiota of those with prodromal PD, which suggests local inflammatory processes and intestinal permeability may contribute to protein misfolding and disease development.21,22 This also raises the intriguing possibility that dietary alterations may be neuroprotective or neurorestorative, although this has yet to be tested in humans.23,24

Like constipation, mood changes can precede the appearance of manifest motor PD.25,26 Case control studies suggest a higher risk of developing PD among individuals who were previously diagnosed with depression or anxiety, particularly in the 1 to 2 years prior to PD diagnosis.27-29 Both apathy and anxiety are associated with striatal dopamine dysfunction, particularly in the right caudate nucleus, which suggests that mood changes are directly related to disease pathology.30,31

 

 

Of the prodromal features, rapid eye movement sleep behavior disorder (RBD) is associated with the highest risk of conversion to motor PD.8 Up to 80% of older men with socalled idiopathic RBD develop a parkinsonian syndrome within 20 years; risk is divided about equally between idiopathic PD and dementia with Lewy bodies (DLB).32 Collateral history from a bed-partner is usually sufficient to make the diagnosis, although, this is often confounded by the prevalence of nightmares in those with posttraumatic stress disorder in the veteran population.32 Thus, in suspected cases, obtaining a polysomnogram can aid in distinguishing between idiopathic PC and DLB.33 Given the specificity of RBD as a marker of synuclein deposition and the high risk of progression to a degenerative syndrome, accurate diagnosis and counseling is imperative.

Each of the prodromal nonmotor features of PD are at best moderately sensitive or specific in isolation, but in concert, they can be used to develop a Parkinson risk score. For instance, the MDS prodromal criteria combine individual likelihood ratios into Bayesian analysis to determine a combined probability of PD, which can be further stratified to probable or possible prodromal PD (probability > 80%, > 50%, respectively).8 These criteria have been applied to several independent cohorts and demonstrate high sensitivity and specificity, especially over time.34,35 Applicability in a veteran population has yet to be determined.

Use of Imaging in Diagnosis

Although clinical diagnostic criteria and prodromal features can improve diagnostic accuracy, it can be extremely challenging to distinguish idiopathic PD from nondegenerative parkinsonism or atypical syndromes (see below). Compared with the gold standard of pathologic assessment, the clinical diagnostic accuracy for PD ranges from 73% for nonexperts to 80% for fellowship-trained movement disorders specialists.36 Thus, objective biomarkers are sought to improve diagnostic accuracy both for clinical care as well as for research purposes, such as enrollment into clinical trials.

Multiple potential imaging biomarkers for preclinical PD can aid in early diagnosis and help differentiate PD from related but distinct disorders. While beyond the scope of this review, these techniques have recently been reviewed.7 Of these, the most widely available and accurate is dopamine transporter (DAT) imaging, which uses a radioiodinated ligand that binds to DAT on striatal dopaminergic terminals; binding is detected through single photon emission computed tomography (SPECT) scanning. Thus, a SPECT DaTscan (GE Healthcare Bio-Sciences, Little Chalfont, England) directly assesses the integrity of the presynaptic nigrostriatal system and is well correlated with severity of motor and nonmotor parkinsonism.37,38

In individuals with suspected prodromal PD, abnormal DaTscans are associated with faster progression to manifest motor PD.39 However, it should be noted that a number of medications, several of which are commonly utilized in the veteran population, can affect the outcome of a DaTscan.40 Some of these medications only mildly affect the outcome, so the physician interpreting the scan should be made aware of their use, while others need to be held for days to weeks so as not to invalidate the DaTscan. DaTscan also do not differentiate between PD and atypical degenerative parkinsonisms such as multiple system atrophy (MSA), DLB, progressive supranuclear palsy (PSP), or corticobasal syndrome (CBS). Nevertheless, these scans can be used to distinguish degenerative parkinsonisms from other conditions that can be difficult to distinguish clinically from PD, including essential tremor, normal pressure hydrocephalus, vascular parkinsonism, or druginduced parkinsonism (DIP).

DIP usually is caused by blockade of postsynaptic dopamine receptors by antipsychotic medications, which are prescribed to as many as 1 in 4 older veterans; antiemetic agents such as metoclopramide are also potential offenders if used chronically.41 The risk of DIP appears to be associated with the D2 binding affinity of the drug. Thus, of the newer atypical antipsychotics, clozapine and quetiapine appear to have the lowest risk, while ziprasidone and aripiprazole have the highest binding affinity and therefore the highest risk.42 In many patients, parkinsonism persists even after discontinuation of the offending agent, suggesting that in at least a subset of patients, DIP may be an “unmasking” of latent PD rather than a true adverse effect of the medication. The prodromal features discussed above can be used to distinguish isolated DIP from unmasked latent PD.43 In a study we conducted in veterans at the Michael J. Crescenz VA Medical Center in Philadelphia, Pennsylvania, hyposmia in particular was shown to be highly predictive of an underlying dopaminergic deficit with an odds ratio of 63.44

Other important considerations in the differential diagnosis of PD are the atypical degenerative parkinsonian syndromes, formerly called Parkinson plus syndromes. These may be further divided into the synucleinopathies (MSA, DLB) or the tauopathies (PSP, CBS), depending on the predominant amyloidogenic protein. Early in the disease, the atypical syndromes and idiopathic PD may be clinically indistinguishable, although the atypical syndromes tend to progress more rapidly and often have a less robust response to levodopa.

 

 

Radiologic and fluid biomarkers for the atypical syndromes are under active investigation; at present the most accessible study is magnetic resonance imaging (MRI), which may show characteristic features such as degeneration of the pontocerebellar fibers in MSA or midbrain atrophy in PSP.45,46 By contrast, standard MRI sequences in idiopathic PD are usually normal, although high-resolution (7 tesla) imaging can reveal loss of neuromelanin in the substantia nigra.47 MRI also can be useful in the workup of suspected normal pressure hydrocephalus or vascular parkinsonism, which would show disproportionate ventriculomegaly with transependymal flow, or white matter lesions in the basal ganglia, respectively.

Data-Based Identification of Preclinical PD

The integration of clinical motor or prodromal features with biomarker data has led to the development of several large-scale clinical and administrative databases to identify PD. The Parkinson Progression Markers Initiative initially enrolled only de novo clinically identified people with PD, but it expanded to include a prodromal cohort who are being assessed for rates of conversion to PD.48 Similarly, metabolic imaging can be combined with prodromal symptoms, such as hyposmia or RBD, to predict risk for phenoconversion into manifest motor PD.49

The PREDICT-PD study synthesizes mood symptoms, RBD, smell testing, genotyping, and keyboard-tapping tasks to divide individuals into high-, middle-, and low-risk groups; interim analysis at 3 years of follow-up (N = 842) demonstrated a hazard ratio of 4.39 (95% CI, 1.03-18.68) for the diagnosis of PD in the highrisk group compared with the low-risk group.50 Lastly, administrative claims data for prodromal features, such as constipation, RBD, and mood symptoms, is highly predictive of eventual PD diagnosis.51 VA databases accessed through the Corporate Data Warehouse are complementary sources of information to nonveteranspecific Medicare databases; to our knowledge there has not yet been a comprehensive search of VA databases to identify veterans with preclinical PD.

Risk Factors Associated With Military Service

A number of potential environmental risk factors may increase the risk of developing Parkinson disease for veterans. Perhaps the most commonly recognized is pesticide exposure, particularly given the presumptive service connections established by the VA for Parkinson disease and exposure to Agent Orange or contaminated water at Camp Lejeune.52,53 Both dioxin, the toxic ingredient in Agent Orange, and the solvents trichloroethylene and perchloroethylene, found in the water supply at Camp Lejeune, interfere with mitochondrial function leading to oxidative stress and apoptosis of nigrostriatal neurons.54,55 Other potential exposures, which are not necessarily limited to the veteran population, include rotenone, a phytochemical used to kill fish in reservoirs, and paraquat, an herbicide that may directly promote synuclein aggregation.56,57 Veterans who have reported exposure to these or other environmental chemicals in civilian life should be carefully assessed for the presence of motor PD or prodromal features.

Traumatic brain injury (TBI) also may be a risk factor for PD, which may be particularly relevant for veterans who had served in Iraq or Afghanistan. Retrospective claims data suggest a strong association between PD and recent TBI in the 5 to 10 years prior to motor PD diagnosis.58,59 A recent assessment of combat veterans with TBI found that even mild TBI was associated with a 56% increased risk of PD, while moderate-to-severe TBI was associated with an 83% higher risk of PD.60 The pathologic mechanism for this link is unclear, but post-TBI inflammatory processes may lead to the formation of reactive oxygen species and/or glutamatergic excitotoxicity, thus leading to secondary injury in the nigrostriatal pathway.61 As with prodromal symptoms, the risk of PD related to environmental risk factors may be synergistic; repetitive TBI may be more damaging than a single injury, and a combination of TBI and pesticide exposure markedly increases PD risk beyond the risk of TBI or the risk of pesticides alone.62 Recently, parkinsonism, including Parkinson disease, was recognized as a service connected condition for veterans with a servicerelated moderate or severe TBI.63

Conclusion

Because of the substantial impact on quality of life and disability-adjusted life years, early and accurate identification and management of veterans at risk for PD is an important priority area for the VA. The 10-year cost of PD-related benefits through the VA was estimated at $3.5 billion in fiscal year 2010, and that number is likely to rise in coming years, due to the aging population as well as synergistic effects of independent risk factors described above.64 In response, the VA has created a network of specialty care sites, known as Parkinson Disease Research, Education, and Clinical Centers (PADRECCs) located in Philadelphia, Pennsylvania; Richmond, Virginia; Houston, Texas; West Los Angeles and San Francisco, California; and Seattle, Washington/ Portland, Oregon (www.parkinsons.va.gov).

The PADRECCs are supplemented by a National VA PD Consortium network of VA physicians trained in PD management (Figure 2). Studies, including one investigating care of veterans with PD, have demonstrated that involvement of specialty care services early in the course of PD leads to improved patient outcomes.65,66 In addition to patient-facing resources such as support groups and specialized physical/occupational/speech therapy, PADRECCs and the consortium sites are national leaders in PD education and clinical trials and provide high-quality, multidisciplinary care for veterans with PD.67 Thus, veterans with significant risk factors or prodromal symptoms of PD should be referred into the PADRECC/Consortium network in order to maximize their quality of care and quality of life.

References

1.  Marras C, Beck JC, Bower JH, et al; Parkinson’s Foundation P4 Group. Prevalence of Parkinson’s disease across North America. NPJ Parkinsons Dis. 2018;4:21.

2.  Postuma RB, Berg D, Stern M, et al. MDS clinical diagnostic criteria for Parkinson’s disease. Mov Disord. 2015;30(12):1591-1601.

3.  Fearnley JM, Lees AJ. Ageing and Parkinson’s disease: substantia nigra regional selectivity. Brain. 1991;114(pt 5):2283-2301.

4.  Greffard S, Verny M, Bonnet A-M, et al. Motor score of the Unified Parkinson Disease Rating Scale as a good predictor of Lewy body-associated neuronal loss in the substantia nigra. Arch Neurol. 2006;63(4):584-588.

5.  Hilker R, Schweitzer K, Coburger S, et al. Nonlinear progression of Parkinson disease as determined by serial positron emission tomographic imaging of striatal fluorodopa F 18 activity. Arch Neurol. 2005;62(3):378-382.

6.  Braak H, Del Tredici K, Rüb U, de Vos RAI, Jansen Steur ENH, Braak E. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol Aging. 2003;24(2):197-211.

7.  Mantri S, Morley JF, Siderowf AD. The importance of preclinical diagnostics in Parkinson disease. Parkinsonism Relat Disord. 2018;pii:S1353-8020(18)30396-1. [Epub ahead of print]

8.  Berg D, Postuma RB, Adler CH, et al. MDS research criteria for prodromal Parkinson’s disease. Mov Disord. 2015;30(12):1600-1611.

9.  Haehner A, Boesveldt S, Berendse HW, et al. Prevalence of smell loss in Parkinson’s disease – a multicenter study. Parkinsonism Relat Disord. 2009;15(7):490-494.

10.  Mullol J, Alobid I, Mariño-Sánchez F, et al. Furthering the understanding of olfaction, prevalence of loss of smell and risk factors: a population-based survey (OLFACAT study). BMJ Open. 2012;2(6).pii:e001256.

11.  Doty RL, Shaman P, Applebaum SL, Giberson R, Siksorski L, Rosenberg L. Smell identification ability: changes with age. Science. 1984;226(4681):1441-1443.

12.  Doty RL. Olfactory dysfunction in Parkinson disease. Nat Rev Neurol. 2012;8(6):329-339.

13.  Double KL, Rowe DB, Hayes M, et al. Identifying the pattern of olfactory deficits in Parkinson disease using the brief smell identification test. Arch Neurol. 2003;60(4):545-549.

14.  Doty RL, Shaman P, Dann M. Development of the University of Pennsylvania Smell Identification Test: a standardized microencapsulated test of olfactory function. Physiol Behav. 1984;32(3):489-502.

15.  Morley JF, Cohen A, Silveira-Moriyama L, et al. Optimizing olfactory testing for the diagnosis of Parkinson’s disease: item analysis of the University of Pennsylvania smell identification test. NPJ Parkinsons Dis. 2018;4:2.

16.  Fullard ME, Tran B, Xie SX, et al. Olfactory impairment predicts cognitive decline in early Parkinson’s disease. Parkinsonism Relat Disord. 2016;25:45-51.

17.  Savica R, Carlin JM, Grossardt BR, et al. Medical records documentation of constipation preceding Parkinson disease: a case-control study. Neurology. 2009;73(21):1752-1758.

18.  Abbott RD, Petrovitch H, White LR, et al. Frequency of bowel movements and the future risk of Parkinson’s disease. Neurology. 2001;57(3):456-462.

19.  Stocchi F, Torti M. Constipation in Parkinson’s disease. Int Rev Neurobiol. 2017;134:811-826.

20.  Yu QJ, Yu SY, Zuo LJ, et al. Parkinson disease with constipation: clinical features and relevant factors. Sci Rep. 2018;8(1):567.

21.  Hill-Burns EM, Debelius JW, Morton JT, et al. Parkinson’s disease and Parkinson’s disease medications have distinct signatures of the gut microbiome. Mov Disord. 2017;32(5):739-749.

22.  Mulak A, Bonaz B. Brain-gut-microbiota axis in Parkinson’s disease. World J Gastroenterol. 2015;21(37): 10609-10620.

23.  Shah SP, Duda JE. Dietary modifications in Parkinson’s disease: a neuroprotective intervention? Med Hypotheses. 2015;85(6):1002-1005.

24.  Perez-Pardo P, de Jong EM, Broersen LM, et al. Promising effects of neurorestorative diets on motor, cognitive, and gastrointestinal dysfunction after symptom development in a mouse model of Parkinson’s disease. Front Aging Neurosci. 2017;9:57.

25.  Fang F, Xu Q, Park Y, et al. Depression and the subsequent risk of Parkinson’s disease in the NIH-AARP Diet and Health Study. Mov Disord. 2010;25(9):1157-1162.

26.  Leentjens AFG, Van den Akker M, Metsemakers JFM, Lousberg R, Verhey FRJ. Higher incidence of depression preceding the onset of Parkinson’s disease: a register study. Mov Disord. 2003;18(4):414-418.

27.  Alonso A, Rodriguez LAG, Logroscino G, Hernán MA. Use of antidepressants and the risk of Parkinson’s disease: a prospective study. J Neurol Neurosurg Psychiatry. 2009;80(6):671-674.

28.  Weisskopf MG, Chen H, Schwarzschild MA, Kawachi I, Ascherio A. Prospective study of phobic anxiety and risk of Parkinson’s disease. Mov Disord. 2003;18(6):646-651.

29.  Darweesh SK, Verlinden VJ, Stricker BH, Hofman A, Koudstaal PJ, Ikram MA. Trajectories of prediagnostic functioning in Parkinson’s disease. Brain. 2017;140(2):429-441.

30.  Santangelo G, Vitale C, Picillo M, et al. Apathy and striatal dopamine transporter levels in de-novo, untreated Parkinson’s disease patients. Parkinsonism Relat Disord. 2015;21(5):489-493.

31.  Erro R, Pappatà S, Amboni M, et al. Anxiety is associated with striatal dopamine transporter availability in newly diagnosed untreated Parkinson’s disease patients. Parkinsonism Relat Disord. 2012;18(9):1034-1038.

32.  Schenck CH, Boeve BF, Mahowald MW. Delayed emergence of a parkinsonian disorder or dementia in 81% of older men initially diagnosed with idiopathic rapid eye movement sleep behavior disorder: a 16-year update on a previously reported series. Sleep Med. 2013;14(8):
744-748.

33.  Melendez J, Hesselbacher S, Sharafkhaneh A, Hirshkowitz M. Assessment of REM sleep behavior disorder in veterans with posttraumatic stress disorder. Chest. 2011;140(4):967A.

34.  Pilotto A, Heinzel S, Suenkel U, et al. Application of the movement disorder society prodromal Parkinson’s disease research criteria in 2 independent prospective cohorts. Mov Disord. 2017;32(7):1025-1034.

35.  Fereshtehnejad S-M, Montplaisir JY, Pelletier A, Gagnon J-F, Berg D, Postuma RB. Validation of the MDS research criteria for prodromal Parkinson’s disease: Longitudinal assessment in a REM sleep behavior disorder (RBD) cohort. Mov Disord. 2017;32(6):865-873.

36. Rizzo G, Copetti M, Arcuti S, Martino D, Fontana A, Logroscino G. Accuracy of clinical diagnosis of Parkinson disease: a systematic review and meta-analysis. Neurology. 2016;86(6):566-576.

37.  Moccia M, Pappatà S, Picillo M, et al. Dopamine transporter availability in motor subtypes of de novo drug-naïve Parkinson’s disease. J Neurol. 2014;261(11):2112-2118.

38.  Siepel FJ, Brønnick KS, Booij J, et al. Cognitive executive impairment and dopaminergic deficits in de novo Parkinson’s disease. Mov Disord. 2014;29(14):1802-1808.

39. Iranzo A, Valldeoriola F, Lomeña F, et al. Serial dopamine transporter imaging of nigrostriatal function in patients with idiopathic rapid-eye-movement sleep behaviour disorder: a prospective study. Lancet Neurol. 2011;10(9):797-805.

40.  Booij J, Kemp P. Dopamine transporter imaging with [(123)I]FP-CIT SPECT: potential effects of drugs. Eur J Nucl Med Mol Imaging. 2008;35(2):424-438.

41.  Gellad WF, Aspinall SL, Handler SM, et al. Use of antipsychotics among older residents in VA nursing homes. Med Care. 2012;50(11):954-960.

42.  Mauri MC, Paletta S, Maffini M, et al. Clinical pharmacology of atypical antipsychotics: an update. EXCLI J. 2014;13:1163-1191.

43.  Morley JF, Duda JE. Use of hyposmia and other non-motor symptoms to distinguish between drug-induced parkinsonism and Parkinson’s disease. J Parkinsons Dis. 2014;4(2):169-173.

44.  Morley JF, Cheng G, Dubroff JG, Wood S, Wilkinson JR, Duda JE. Olfactory impairment predicts underlying dopaminergic deficit in presumed drug-induced parkinsonism. Mov Disord Clin Pract. 2017;4(4):603-606.

45.  Whitwell JL, Höglinger GU, Antonini A, et al; Movement Disorder Society-endorsed PSP Study Group. Radiological biomarkers for diagnosis in PSP: where are we and where do we need to be? Mov Disord. 2017;32(7):955-971.

46.  Laurens B, Constantinescu R, Freeman R, et al. Fluid biomarkers in multiple system atrophy: A review of the MSA Biomarker Initiative. Neurobiol Dis. 2015;80:29-41.

47.  Barber TR, Klein JC, Mackay CE, Hu MTM. Neuroimaging in pre-motor Parkinson’s disease. NeuroImage Clin. 2017;15:215-227.

48.  Parkinson Progression Marker Initiative. The Parkinson Progression Marker Initiative (PPMI). Prog Neurobiol. 2011;95(4):629-635.

49.  Meles SK, Vadasz D, Renken RJ, et al. FDG PET, dopamine transporter SPECT, and olfaction: combining biomarkers in REM sleep behavior disorder. Mov Disord. 2017;32(10):1482-1486.

50.  Noyce AJ, R’Bibo L, Peress L, et al. PREDICT‐PD: an online approach to prospectively identify risk indicators of Parkinson’s disease. Mov Disord. 2017 Feb; 32(2): 219–226.

51.  Searles Nielsen S, Warden MN, Camacho-Soto A, Willis AW, Wright BA, Racette BA. A predictive model to identify Parkinson disease from administrative claims data. Neurology. 2017;89(14):1448-1456.

52.  Institute of Medicine. Veterans and Agent Orange: Update 2012. National Academies Press: Washington, DC; 2013.

53.  Department of Veterans Affairs. Diseases associated with exposure to Contaminants in the Water Supply at Camp Lejeune. Final rule. Fed Regist. 2017;82(9):4173-4185.

54.  Goldman SM, Quinlan PJ, Ross GW, et al. Solvent exposures and Parkinson disease risk in twins. Ann Neurol. 2012;71(6):776-784.

55.  Liu M, Shin EJ, Dang DK, et al. Trichloroethylene and Parkinson’s disease: risk assessment. Mol Neurobiol. 2018;55(7):6201-6214.

56.  Betarbet R, Sherer TB, MacKenzie G, Garcia-Osuna M, Panov AV, Greenamyre JT. Chronic systemic pesticide exposure reproduces features of Parkinson’s disease. Nat Neurosci. 2000;3(12):1301-1306.

57.  Manning-Bog AB, McCormack AL, Li J, Uversky VN, Fink AL, Monte DAD. The herbicide paraquat causes up-regulation and aggregation of alpha-synuclein in mice: paraquat and alpha-synuclein. J Biol Chem. 2002;277(3):1641-1644.

58.  Camacho-Soto A, Warden MN, Searles Nielsen S, et al. Traumatic brain injury in the prodromal period of Parkinson’s disease: a large epidemiological study using medicare data. Ann Neurol. 2017;82(5):744-754.

59.  Gardner RC, Burke JF, Nettiksimmons J, Goldman S, Tanner CM, Yaffe K. Traumatic brain injury in later life increases risk for Parkinson disease. Ann Neurol. 2015;77(6):987-995.

60.  Gardner RC, Byers AL, Barnes DE, Li Y, Boscardin J, Yaffe K. Mild TBI and risk of Parkinson disease: a Chronic Effects of Neurotrauma Consortium Study. Neurology. 2018;90(20):e1771-e1779.

61.  Cruz-Haces M, Tang J, Acosta G, Fernandez J, Shi R. Pathological correlations between traumatic brain injury and chronic neurodegenerative diseases. Transl Neurodegener. 2017;6:20.

62.  Lee PC, Bordelon Y, Bronstein J, Ritz B. Traumatic brain injury, paraquat exposure, and their relationship to Parkinson disease. Neurology. 2012;79(20):2061-2066.

63.  Disabilities that are proximately due to, or aggravated by, service-connected disease or injury. 38 CFR §3.310.

64.  Diseases Associated With Exposure to Certain Herbicide Agents (Hairy Cell Leukemia and Other Chronic B-Cell Leukemias, Parkinson’s Disease and Ischemic Heart Disease). Federal Regist. 2010;75(173):53202-53216. To be codified at 38 CFR §3.

65.  Cheng EM, Swarztrauber K, Siderowf AD, et al. Association of specialist involvement and quality of care for Parkinson’s disease. Mov Disord. 2007;22(4):515-522.

66.  Qamar MA, Harington G, Trump S, Johnson J, Roberts F, Frost E. Multidisciplinary care in Parkinson’s disease. Int Rev Neurobiol. 2017;132:511-523.

67.  Pogoda TK, Cramer IE, Meterko M, et al. Patient and organizational factors related to education and support use by veterans with Parkinson’s disease. Mov Disord. 2009;24(13):1916-1924.

References

1.  Marras C, Beck JC, Bower JH, et al; Parkinson’s Foundation P4 Group. Prevalence of Parkinson’s disease across North America. NPJ Parkinsons Dis. 2018;4:21.

2.  Postuma RB, Berg D, Stern M, et al. MDS clinical diagnostic criteria for Parkinson’s disease. Mov Disord. 2015;30(12):1591-1601.

3.  Fearnley JM, Lees AJ. Ageing and Parkinson’s disease: substantia nigra regional selectivity. Brain. 1991;114(pt 5):2283-2301.

4.  Greffard S, Verny M, Bonnet A-M, et al. Motor score of the Unified Parkinson Disease Rating Scale as a good predictor of Lewy body-associated neuronal loss in the substantia nigra. Arch Neurol. 2006;63(4):584-588.

5.  Hilker R, Schweitzer K, Coburger S, et al. Nonlinear progression of Parkinson disease as determined by serial positron emission tomographic imaging of striatal fluorodopa F 18 activity. Arch Neurol. 2005;62(3):378-382.

6.  Braak H, Del Tredici K, Rüb U, de Vos RAI, Jansen Steur ENH, Braak E. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol Aging. 2003;24(2):197-211.

7.  Mantri S, Morley JF, Siderowf AD. The importance of preclinical diagnostics in Parkinson disease. Parkinsonism Relat Disord. 2018;pii:S1353-8020(18)30396-1. [Epub ahead of print]

8.  Berg D, Postuma RB, Adler CH, et al. MDS research criteria for prodromal Parkinson’s disease. Mov Disord. 2015;30(12):1600-1611.

9.  Haehner A, Boesveldt S, Berendse HW, et al. Prevalence of smell loss in Parkinson’s disease – a multicenter study. Parkinsonism Relat Disord. 2009;15(7):490-494.

10.  Mullol J, Alobid I, Mariño-Sánchez F, et al. Furthering the understanding of olfaction, prevalence of loss of smell and risk factors: a population-based survey (OLFACAT study). BMJ Open. 2012;2(6).pii:e001256.

11.  Doty RL, Shaman P, Applebaum SL, Giberson R, Siksorski L, Rosenberg L. Smell identification ability: changes with age. Science. 1984;226(4681):1441-1443.

12.  Doty RL. Olfactory dysfunction in Parkinson disease. Nat Rev Neurol. 2012;8(6):329-339.

13.  Double KL, Rowe DB, Hayes M, et al. Identifying the pattern of olfactory deficits in Parkinson disease using the brief smell identification test. Arch Neurol. 2003;60(4):545-549.

14.  Doty RL, Shaman P, Dann M. Development of the University of Pennsylvania Smell Identification Test: a standardized microencapsulated test of olfactory function. Physiol Behav. 1984;32(3):489-502.

15.  Morley JF, Cohen A, Silveira-Moriyama L, et al. Optimizing olfactory testing for the diagnosis of Parkinson’s disease: item analysis of the University of Pennsylvania smell identification test. NPJ Parkinsons Dis. 2018;4:2.

16.  Fullard ME, Tran B, Xie SX, et al. Olfactory impairment predicts cognitive decline in early Parkinson’s disease. Parkinsonism Relat Disord. 2016;25:45-51.

17.  Savica R, Carlin JM, Grossardt BR, et al. Medical records documentation of constipation preceding Parkinson disease: a case-control study. Neurology. 2009;73(21):1752-1758.

18.  Abbott RD, Petrovitch H, White LR, et al. Frequency of bowel movements and the future risk of Parkinson’s disease. Neurology. 2001;57(3):456-462.

19.  Stocchi F, Torti M. Constipation in Parkinson’s disease. Int Rev Neurobiol. 2017;134:811-826.

20.  Yu QJ, Yu SY, Zuo LJ, et al. Parkinson disease with constipation: clinical features and relevant factors. Sci Rep. 2018;8(1):567.

21.  Hill-Burns EM, Debelius JW, Morton JT, et al. Parkinson’s disease and Parkinson’s disease medications have distinct signatures of the gut microbiome. Mov Disord. 2017;32(5):739-749.

22.  Mulak A, Bonaz B. Brain-gut-microbiota axis in Parkinson’s disease. World J Gastroenterol. 2015;21(37): 10609-10620.

23.  Shah SP, Duda JE. Dietary modifications in Parkinson’s disease: a neuroprotective intervention? Med Hypotheses. 2015;85(6):1002-1005.

24.  Perez-Pardo P, de Jong EM, Broersen LM, et al. Promising effects of neurorestorative diets on motor, cognitive, and gastrointestinal dysfunction after symptom development in a mouse model of Parkinson’s disease. Front Aging Neurosci. 2017;9:57.

25.  Fang F, Xu Q, Park Y, et al. Depression and the subsequent risk of Parkinson’s disease in the NIH-AARP Diet and Health Study. Mov Disord. 2010;25(9):1157-1162.

26.  Leentjens AFG, Van den Akker M, Metsemakers JFM, Lousberg R, Verhey FRJ. Higher incidence of depression preceding the onset of Parkinson’s disease: a register study. Mov Disord. 2003;18(4):414-418.

27.  Alonso A, Rodriguez LAG, Logroscino G, Hernán MA. Use of antidepressants and the risk of Parkinson’s disease: a prospective study. J Neurol Neurosurg Psychiatry. 2009;80(6):671-674.

28.  Weisskopf MG, Chen H, Schwarzschild MA, Kawachi I, Ascherio A. Prospective study of phobic anxiety and risk of Parkinson’s disease. Mov Disord. 2003;18(6):646-651.

29.  Darweesh SK, Verlinden VJ, Stricker BH, Hofman A, Koudstaal PJ, Ikram MA. Trajectories of prediagnostic functioning in Parkinson’s disease. Brain. 2017;140(2):429-441.

30.  Santangelo G, Vitale C, Picillo M, et al. Apathy and striatal dopamine transporter levels in de-novo, untreated Parkinson’s disease patients. Parkinsonism Relat Disord. 2015;21(5):489-493.

31.  Erro R, Pappatà S, Amboni M, et al. Anxiety is associated with striatal dopamine transporter availability in newly diagnosed untreated Parkinson’s disease patients. Parkinsonism Relat Disord. 2012;18(9):1034-1038.

32.  Schenck CH, Boeve BF, Mahowald MW. Delayed emergence of a parkinsonian disorder or dementia in 81% of older men initially diagnosed with idiopathic rapid eye movement sleep behavior disorder: a 16-year update on a previously reported series. Sleep Med. 2013;14(8):
744-748.

33.  Melendez J, Hesselbacher S, Sharafkhaneh A, Hirshkowitz M. Assessment of REM sleep behavior disorder in veterans with posttraumatic stress disorder. Chest. 2011;140(4):967A.

34.  Pilotto A, Heinzel S, Suenkel U, et al. Application of the movement disorder society prodromal Parkinson’s disease research criteria in 2 independent prospective cohorts. Mov Disord. 2017;32(7):1025-1034.

35.  Fereshtehnejad S-M, Montplaisir JY, Pelletier A, Gagnon J-F, Berg D, Postuma RB. Validation of the MDS research criteria for prodromal Parkinson’s disease: Longitudinal assessment in a REM sleep behavior disorder (RBD) cohort. Mov Disord. 2017;32(6):865-873.

36. Rizzo G, Copetti M, Arcuti S, Martino D, Fontana A, Logroscino G. Accuracy of clinical diagnosis of Parkinson disease: a systematic review and meta-analysis. Neurology. 2016;86(6):566-576.

37.  Moccia M, Pappatà S, Picillo M, et al. Dopamine transporter availability in motor subtypes of de novo drug-naïve Parkinson’s disease. J Neurol. 2014;261(11):2112-2118.

38.  Siepel FJ, Brønnick KS, Booij J, et al. Cognitive executive impairment and dopaminergic deficits in de novo Parkinson’s disease. Mov Disord. 2014;29(14):1802-1808.

39. Iranzo A, Valldeoriola F, Lomeña F, et al. Serial dopamine transporter imaging of nigrostriatal function in patients with idiopathic rapid-eye-movement sleep behaviour disorder: a prospective study. Lancet Neurol. 2011;10(9):797-805.

40.  Booij J, Kemp P. Dopamine transporter imaging with [(123)I]FP-CIT SPECT: potential effects of drugs. Eur J Nucl Med Mol Imaging. 2008;35(2):424-438.

41.  Gellad WF, Aspinall SL, Handler SM, et al. Use of antipsychotics among older residents in VA nursing homes. Med Care. 2012;50(11):954-960.

42.  Mauri MC, Paletta S, Maffini M, et al. Clinical pharmacology of atypical antipsychotics: an update. EXCLI J. 2014;13:1163-1191.

43.  Morley JF, Duda JE. Use of hyposmia and other non-motor symptoms to distinguish between drug-induced parkinsonism and Parkinson’s disease. J Parkinsons Dis. 2014;4(2):169-173.

44.  Morley JF, Cheng G, Dubroff JG, Wood S, Wilkinson JR, Duda JE. Olfactory impairment predicts underlying dopaminergic deficit in presumed drug-induced parkinsonism. Mov Disord Clin Pract. 2017;4(4):603-606.

45.  Whitwell JL, Höglinger GU, Antonini A, et al; Movement Disorder Society-endorsed PSP Study Group. Radiological biomarkers for diagnosis in PSP: where are we and where do we need to be? Mov Disord. 2017;32(7):955-971.

46.  Laurens B, Constantinescu R, Freeman R, et al. Fluid biomarkers in multiple system atrophy: A review of the MSA Biomarker Initiative. Neurobiol Dis. 2015;80:29-41.

47.  Barber TR, Klein JC, Mackay CE, Hu MTM. Neuroimaging in pre-motor Parkinson’s disease. NeuroImage Clin. 2017;15:215-227.

48.  Parkinson Progression Marker Initiative. The Parkinson Progression Marker Initiative (PPMI). Prog Neurobiol. 2011;95(4):629-635.

49.  Meles SK, Vadasz D, Renken RJ, et al. FDG PET, dopamine transporter SPECT, and olfaction: combining biomarkers in REM sleep behavior disorder. Mov Disord. 2017;32(10):1482-1486.

50.  Noyce AJ, R’Bibo L, Peress L, et al. PREDICT‐PD: an online approach to prospectively identify risk indicators of Parkinson’s disease. Mov Disord. 2017 Feb; 32(2): 219–226.

51.  Searles Nielsen S, Warden MN, Camacho-Soto A, Willis AW, Wright BA, Racette BA. A predictive model to identify Parkinson disease from administrative claims data. Neurology. 2017;89(14):1448-1456.

52.  Institute of Medicine. Veterans and Agent Orange: Update 2012. National Academies Press: Washington, DC; 2013.

53.  Department of Veterans Affairs. Diseases associated with exposure to Contaminants in the Water Supply at Camp Lejeune. Final rule. Fed Regist. 2017;82(9):4173-4185.

54.  Goldman SM, Quinlan PJ, Ross GW, et al. Solvent exposures and Parkinson disease risk in twins. Ann Neurol. 2012;71(6):776-784.

55.  Liu M, Shin EJ, Dang DK, et al. Trichloroethylene and Parkinson’s disease: risk assessment. Mol Neurobiol. 2018;55(7):6201-6214.

56.  Betarbet R, Sherer TB, MacKenzie G, Garcia-Osuna M, Panov AV, Greenamyre JT. Chronic systemic pesticide exposure reproduces features of Parkinson’s disease. Nat Neurosci. 2000;3(12):1301-1306.

57.  Manning-Bog AB, McCormack AL, Li J, Uversky VN, Fink AL, Monte DAD. The herbicide paraquat causes up-regulation and aggregation of alpha-synuclein in mice: paraquat and alpha-synuclein. J Biol Chem. 2002;277(3):1641-1644.

58.  Camacho-Soto A, Warden MN, Searles Nielsen S, et al. Traumatic brain injury in the prodromal period of Parkinson’s disease: a large epidemiological study using medicare data. Ann Neurol. 2017;82(5):744-754.

59.  Gardner RC, Burke JF, Nettiksimmons J, Goldman S, Tanner CM, Yaffe K. Traumatic brain injury in later life increases risk for Parkinson disease. Ann Neurol. 2015;77(6):987-995.

60.  Gardner RC, Byers AL, Barnes DE, Li Y, Boscardin J, Yaffe K. Mild TBI and risk of Parkinson disease: a Chronic Effects of Neurotrauma Consortium Study. Neurology. 2018;90(20):e1771-e1779.

61.  Cruz-Haces M, Tang J, Acosta G, Fernandez J, Shi R. Pathological correlations between traumatic brain injury and chronic neurodegenerative diseases. Transl Neurodegener. 2017;6:20.

62.  Lee PC, Bordelon Y, Bronstein J, Ritz B. Traumatic brain injury, paraquat exposure, and their relationship to Parkinson disease. Neurology. 2012;79(20):2061-2066.

63.  Disabilities that are proximately due to, or aggravated by, service-connected disease or injury. 38 CFR §3.310.

64.  Diseases Associated With Exposure to Certain Herbicide Agents (Hairy Cell Leukemia and Other Chronic B-Cell Leukemias, Parkinson’s Disease and Ischemic Heart Disease). Federal Regist. 2010;75(173):53202-53216. To be codified at 38 CFR §3.

65.  Cheng EM, Swarztrauber K, Siderowf AD, et al. Association of specialist involvement and quality of care for Parkinson’s disease. Mov Disord. 2007;22(4):515-522.

66.  Qamar MA, Harington G, Trump S, Johnson J, Roberts F, Frost E. Multidisciplinary care in Parkinson’s disease. Int Rev Neurobiol. 2017;132:511-523.

67.  Pogoda TK, Cramer IE, Meterko M, et al. Patient and organizational factors related to education and support use by veterans with Parkinson’s disease. Mov Disord. 2009;24(13):1916-1924.

Issue
Federal Practitioner - 36(4)s
Issue
Federal Practitioner - 36(4)s
Page Number
S18-S23
Page Number
S18-S23
Publications
Publications
Topics
Article Type
Display Headline
Early and Accurate Identification of Parkinson Disease Among US Veterans
Display Headline
Early and Accurate Identification of Parkinson Disease Among US Veterans
Sections
Citation Override
Fed Pract. 2019 June;36(suppl 4):S18-S23
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Tue, 06/04/2019 - 15:45
Un-Gate On Date
Tue, 06/04/2019 - 15:45
Use ProPublica
CFC Schedule Remove Status
Tue, 06/04/2019 - 15:45
Hide sidebar & use full width
render the right sidebar.
Article PDF Media

Hemolytic Uremic Syndrome With Severe Neurologic Complications in an Adult (FULL)

Article Type
Changed
Mon, 06/01/2020 - 15:23
Display Headline
Hemolytic Uremic Syndrome With Severe Neurologic Complications in an Adult

The case of a female presenting with Shiga toxin-producing Escherichia coli and hemolytic uremic syndrome highlights a severe neurologic complication that canbe associated with these conditions.

Hemolytic uremic syndrome (HUS) is a rare illness that can be acquired through the consumption of food products contaminated with strains of Shiga toxin-producing Escherichia coli (E coli; STEC).1 Between 6% and 15% of individuals infected with STEC develop HUS, with children affected more frequently than adults.2,3 This strain of E coli releases Shiga toxin into the systemic circulation, which causes a thrombotic microangiopathy resulting in the characteristic HUS triad of symptoms: acute renal insufficiency, thrombocytopenia, and hemolytic anemia.4-6

Although neurologic features are common in HUS, they have not been extensively studied, particularly in adults. We report a case of STEC 0157:H7 subtype HUS in an adult with severe neurologic complications. This case highlights the neurological sequelae in an adult with typical STEC-HUS. The use of treatment modalities, such as plasmapheresis and eculizumab, and their use in adult typical STEC-HUS also is explored.

Case

A 53-year-old white woman with no pertinent past medical history presented to the Bay Pines Veterans Affairs Healthcare System Emergency Department with a 2-day history of abdominal pain, vomiting, nausea, diarrhea, and bright bloody stools. She returned from a cruise to the Bahamas 3 days prior, where she ate local foods, including salads. She reported no fever, shortness of breath, chest pain, headache, and cognitive difficulties. She presented with a normal mental status and neurologic exam. Apart from leukocytosis and elevated glucose level, her laboratory results at initial presentation were normal, (Table). A stool sample showed occult blood with white blood cell counts (WBCs) but was negative for Clostridium difficile. She was started on ciprofloxacin 400 mg and metronidazole 500 mg on the day of admission.

Hematuria was found on hospital day 2. On hospital day 4, the patient exhibited word finding difficulties. Blood studies revealed anemia, thrombocytopenia, leukocytosis, and increasing blood urea nitrogen (BUN) and creatinine. A computed tomography scan of the head was normal. Laboratory analysis showed schistocytes in the peripheral blood smear.

The patient’s cognitive functioning deteriorated on hospital day 5. She was not oriented to time or place. Her laboratory results showed complement level C3 at 70 mg/dL (ref: 83-193 mg/dL) complement C4 at 12 mg/dL (ref: 15-57mg/dL). The patient exhibited oliguria and hyponatremia, as well as both metabolic and respiratory acidosis; dialysis was then initiated. Results from the stool sample that was collected on hospital day 1 were received and tested positive for Shiga toxin.

At this point, the patient’s presentation of hemolytic anemia and thrombocytopenia in the setting of acute bloody diarrheal illness with known Shiga toxin, schistocytes on blood smear, and lack of pertinent medical history for other causes of this presentation made STEC-HUS the leading differential diagnosis. Plasmapheresis was ordered and performed on hospital day 6 and 7. Shiga toxin was no longer detected in the stool and antibiotics were stopped on hospital day 7.

The patient’s progressive deterioration in mental status continued on hospital day 8. She was not oriented to time or place, unable to perform simple calculations, and could not spell the word “hand” backwards. Physicians observed repetitive jerking motions of the upper extremities that were worse on the left side. An electroencephalogram (EEG) revealed right hemispheric sharp waves that were thought to be epileptiform (Figure 1). The patient began taking levetiracetam 1500 mg IV with 750 mg bid maintenance for seizure control. Plasmapheresis was discontinued due to her continued neurologic deterioration on this therapy. Consequently, eculizumab 900 mg IV was given along with the Neisseria meningitidis (N meningitidis) vaccine and a 19-day course of azithromycin 250 mg po as prophylaxis for encapsulated bacteria.

The patient continued to seize on hospital days 10 through 13. Oculocephalic maneuvers showed a tendency to keep her eyes deviated to the right. Her pupils continued to react to light. A repeat EEG showed diffuse slowing (5-6 Hz) with no epileptic activity seen (Figure 2). A second dose of eculizumab 900 mg IV was administered on hospital day 15. The patient experienced cardiac arrest on hospital day 16 and was successfully resuscitated. On hospital day 25 (10 days after receiving her second dose of eculizumab), the patient was able to speak and follow simple commands but exhibited difficulty concentrating and poor impulse control.

The patient was alert and oriented to person, place, time, and situation on hospital day 28 (6 days after the third and final dose of eculizumab). A neurologic exam was significant only for a slight intention tremor. She was continued on levetiracetam with a plan to be maintained on the medication for the next 6 months for seizure control. She was discharged on hospital day 30.

Twenty-eight days postdischarge (57 days postadmission), the patient showed marked recovery. She had returned to her previous employment as a business administrator on a part-time basis and exhibited no deficiencies in executive functioning or handling activities of daily living. Although she had been very active prior to this illness, she now experienced decreased physical and mental endurance; however, this gradually improved with physical therapy. She planned on returning to work on a full-time basis when she had regained her stamina. She also noticed difficulties in retaining short term memory since her discharge but believed that these symptoms were remitting. On examination her mental status and neurologic exam was significant for inability to continue serial 7s, left sided 4/5 muscle strength in quadriceps and thumb to 5th metacarpal adduction, bilateral 1+ reflexes in muscle groups tested (triceps, biceps, brachioradialis, patellar, and Achilles), loss of dull pinprick sensation bilaterally at web of hands, deficit in tandem gait while looking away, and slight intention tremor on finger to nose testing bilaterally (with left hand tremor more pronounced than right). Her complete blood count was normal. Her recovery continues to be monitored in an outpatient setting.

 

 

Discussion

HUS is characterized by 3 core clinical features: microangiopathic hemolytic anemia, thrombocytopenia, and acute kidney injury.4 Schistocytes are seen on peripheral blood smear and occur due to the passage of red blood cells over the microvascular thrombi induced by the disease. HUS can be classified as typical, atypical, or occurring with a coexisting disease. Typical HUS is associated with STEC 0157:H7 subtype, a bacterium known to be acquired through contaminated food and via human-to-human transmission.6-8 In the case of typical STEC 0157:H7, the bacterium releases a verotoxin that damages the vascular endothelium, thereby leading to activation of the coagulation cascade and eventually the formation of thrombi.4 It has been hypothesized that the Shiga toxin also activates the alternative complement pathway directly, which could contribute to thrombosis.9 This would explain the findings of low complement levels in our patient. Atypical HUS is primarily attributable to mutations in the alternative complement pathway. Causes for the third type of HUS can include Streptococcus pneumoniae, HIV, drug toxicity, and alterations in the metabolism of cobalamin C.

Epidemiologically, 15.3% of children aged < 5 years develop typical HUS after exposure to STEC compared with 1.2% of adults aged 18 to 59 years. The median age of patients who developed HUS from STEC exposure was 4 years compared with 16 years for those who did not develop HUS.2

Neurologic manifestations increase mortality for HUS patients.10 These have been described in the pediatric population as alteration in consciousness (85%), seizures (71%), pyramidal syndrome (52%), and extrapyramidal syndrome with hypertonia (42%).11 Brain imaging in children has demonstrated hemorrhagic lesions involving the pons, basal ganglia, and occipital cortex.11 Blood flow to areas such as the cerebellum, brainstem, and orbitofrontal area can be compromised.10 Adult patients with HUS can present without lesions on cranial magnetic resonance imaging (MRI), but instead with transient symmetric vasogenic edema of the central brain stem.12 Unfortunately in this case, MRI was not performed because it was thought to provide limited aid in diagnosis and to avoid unnecessary testing for the acutely ill patient.

The underlying pathophysiology of neurologic manifestations in patients may be due to a metabolic disturbance, toxin-mediated damage of the vascular endothelium, or toxin-induced cytokine release resulting in death of neural cells and subsequent neuroinflammation. However, the most likely mechanism is parenchymal ischemic changes related to microangiopathy.11,13 Pediatric patients often experience seizures and altered mental status, and their EEGs display delta waves.13 This patient’s diffuse slowing on her second EEG and altered mental status suggests that the neuropathologic mechanisms for typical HUS in adults may be similar to those in children.

HUS Treatment

The treatment and management of adults with typical STEC-HUS is evolving. The patient was first suspected to have an infectious colitis and empiric antibiotics were initiated. Some studies suggest that antibiotic administration may worsen the course of HUS in children as it may lead to release and subsequent absorption of Shiga toxin in the intestine.9,14 However, there is little evidence to suggest harm or efficacy of administration in adults. It is unclear what role antibiotic administration played in the recovery time of HUS given the co-administration of other treatments such as eculizumab and plasmapheresis, but it does appear to have helped with the initial E coli infection.

 

 

Plasmapheresis was subsequently administered, due to its documented benefit in the treatment of HUS.15 However, it should be noted that even though plasmapheresis is currently used in patients with CNS involvement, it remains unproven with conflicting information on its efficacy.3,16 The mechanism of action is unclear, but it has been hypothesized that plasmapheresis prevents microangiopathy caused by microthrombi.3,16 For this reason, eculizumab is becoming the mainstay for treatment of STEC-HUS with neurologic complications given the lack of well researched alternative treatments. In this case study, the use of plasmapheresis did not result in clinical improvement, and was abandoned after 2 days of treatment.

Eculizumab is a humanized, recombinant monoclonal IgG antibody that is a terminal complement inhibitor of the alternative complement system at the final step to cleave C5.17 The Shiga toxin may directly activate the complement system via the alternative pathway, which can result in uncontrolled platelet and white blood cell activation and depletion, endothelial cell damage, and hemolysis. The galvanized complement system leads to a series of cascading events that contribute to organ damage and death.9 Eculizumab is FDA approved for use in atypical HUS.18 It also can be used off-label to treat typical-HUS in adults with neurologic complications.

Eculizumab interferes with the immune response against encapsulated bacteria because it inhibits the alternative complement pathway. Thus, vaccination against N meningitides is recommended 2 weeks prior to the administration of eculizumab. However, in situations where the risks of delaying eculizumab for 2 weeks are greater than the risk of developing an N meningitides infection, eculizumab may be given without delay.18 Given the rapid deterioration of our patient’s condition, the vaccine and eculizumab were given together with prophylactic azithromycin. Although penicillin is the standard for prophylaxis in this situation, the patient’s penicillin allergy led to the use of azithromycin 250 mg po once a day. Literature also suggests azithromycin reduces the carriage duration of E coli-induced colitis.19 As such, it is possible that some improvement in the patient’s condition could be attributed to the elimination of the pathogen and toxin.

 

Conclusion

Three doses of eculizumab were administered at weekly intervals, with the first dose on hospital day 8 and the final dose on hospital day 22. Prior to the first dose, the patient displayed significant decline in mental status with EEG findings of right hemisphere epileptogenic discharges. After her third dose, she was found to have a drastically improved mental status exam and a normal EEG. One week later, she was discharged home. At the time of her 1-month follow-up, she was independent in all activities of daily living and had returned to part-time work. Apart from subtle cognitive changes, the remainder of her neurologic exam was normal.

There is evidence that supports the efficacy of eculizumab in children with HUS with neurologic symptoms on dialysis.20 However, its use in adults is not well established.21 This patient required dialysis and had neurologic symptoms similar to pediatric patients described in the literature, and responded similarly to the eculizumab. The rationale for the use of eculizumab in STEC-HUS also is evidenced by in vitro demonstrations of complement activation in STEC-HUS.22-25 This case report adds to the literature supporting the use of eculizumab in adult patients with typical HUS with neurological complications. Further research is necessary to develop guidelines in the treatment of adult STEC-HUS with regards to neurologic complications.

Acknowledgments

The authors would like to thank Pete DiStaso, REEGT for his work on obtaining the electroencephalograms and Anthony Rinaldi, PsyD; Julie Cessnapalas, PsyD; and Syed Faizan Sagheer for proof-reading the article.

References

1. Tarr PI, Gordon CA, Chandler WL. Shiga-toxin-producing Escherichia coli and haemolytic uraemic syndrome. Lancet. 2005;365(9464):1073-1086.

2. Gould LH, Demma L, Jones TF, et al. Hemolytic uremic syndrome and death in persons with Escherichia coli O157:H7 infection, foodborne diseases active surveillance network sites, 2000-2006. Clin Infect Dis. 2009;49(10):1480-1485.

3. Boyce TG, Swerdlow DL, Griffin PM. Escherichia coli O157:H7 and the hemolytic-uremic syndrome. N Engl J Med. 1995;333(6):364-368.

4. Rondeau E, Peraldi MN. Escherichia coli and the hemolytic-uremic syndrome. N Engl J Med. 1996;335(9):660-662.

5. Te Loo DM, van Hinsbergh VW, van den Heuvel LP, Monnens LA. Detection of verocytotoxin bound to circulating polymorphonuclear leukocytes of patients with hemolytic uremic syndrome. J Am Soc Nephrol. 2001;12(4):800-806.

6. Tran SL, Jenkins C, Livrelli V, Schüller S. Shiga toxin 2 translocation across intestinal epithelium is linked to virulence of Shiga toxin-producing Escherichia coli in humans. Microbiology. 2018;164(4):509-516.

7. Jokiranta TS. HUS and atypical HUS. Blood. 2017;129(21):2847-2856.

8. Ferens WA, Hovde CJ. Escherichia coli O157:H7: animal reservoir and sources of human infection. Foodborne Pathog Dis. 2011;8(4):465-487.

9. Percheron L, Gramada R, Tellier S, et al. Eculizumab treatment in severe pediatric STEC-HUS: a multicenter retrospective study. Pediatr Nephrol. 2018;33(8):1385-1394.

10. Hosaka T, Nakamagoe K, Tamaoka A. Hemolytic uremic syndrome-associated encephalopathy successfully treated with corticosteroids. Intern Med. 2017;56(21):2937-2941.

11. Nathanson S, Kwon T, Elmaleh M, et al. Acute neurological involvement in diarrhea-associated hemolytic uremic syndrome. Clin J Am Soc Nephrol. 2010;5(7):1218-1228.

12. Wengenroth M, Hoeltje J, Repenthin J, et al. Central nervous system involvement in adults with epidemic hemolytic uremic syndrome. AJNR Am J Neuroradiol. 2013;34(5):1016-1021, S1.

13. Eriksson KJ, Boyd SG, Tasker RC. Acute neurology and neurophysiology of haemolytic-uraemic syndrome. Arch Dis Child. 2001;84(5):434-435.

14. Wong CS, Jelacic S, Habeeb RL, Watkins SL, Tarr PI. The risk of the hemolytic-uremic syndrome after antibiotic treatment of Escherichia coli O157:H7 infections. N Engl J Med. 2000;342(26):1930-1936.

15. Nguyen TC, Kiss JE, Goldman JR, Carcillo JA. The role of plasmapheresis in critical illness. Crit Care Clin. 2012;28(3):453-468, vii.

16. Loos S, Ahlenstiel T, Kranz B, et al. An outbreak of Shiga toxin-producing Escherichia coli O104:H4 hemolytic uremic syndrome in Germany: presentation and short-term outcome in children. Clin Infect Dis. 2012;55(6):753-759.

17. Hossain MA, Cheema A, Kalathil S, et al. Atypical hemolytic uremic syndrome: Laboratory characteristics, complement-amplifying conditions, renal biopsy, and genetic mutations. Saudi J Kidney Dis Transpl. 2018;29(2):276-283.

18. Soliris (eculizumab) [package insert]. Cheshire, CT: Alexion Pharmaceuticals, Inc; 2011.

19. Keenswijk W, Raes A, Vande Walle J. Is eculizumab efficacious in Shigatoxin-associated hemolytic uremic syndrome? A narrative review of current evidence. Eur J Pediatr. 2018;177(3):311-318.

20. Lapeyraque AL, Malina M, Fremeaux-Bacchi V, et al. Eculizumab in severe Shiga-toxin-associated HUS. N Engl J Med. 2011;364(26):2561-2563.

21. Pape L, Hartmann H, Bange FC, Suerbaum S, Bueltmann E, Ahlenstiel-Grunow T. Eculizumab in typical hemolytic uremic syndrome (HUS) with neurological involvement. Medicine (Baltimore). 2015;94(24):e1000.

22. Kim Y, Miller K, Michael AF. Breakdown products of C3 and factor B in hemolytic-uremic syndrome. J Lab Clin Med. 1977;89(4):845-850.

23. Monnens L, Molenaar J, Lambert PH, Proesmans W, van Munster P. The complement system in hemolytic-uremic syndrome in childhood. Clin Nephrol. 1980;13(4):168-171.

24. Thurman JM, Marians R, Emlen W, et al. Alternative pathway of complement in children with diarrhea-associated hemolytic uremic syndrome. Clin J Am Soc Nephrol. 2009;4(12):1920-1924.

25. Ståhl AL, Sartz L, Karpman D. Complement activation on platelet-leukocyte complexes and microparticles in enterohemorrhagic Escherichia coli-induced hemolytic uremic syndrome. Blood. 2011;117(20):5503-5513.

Article PDF
Author and Disclosure Information

Ayan Nasir, Nimai Patel, Samantha Prabakaran, S. Hamad Sagheer, and Steven Troy are Medical Students, Esther Baldinger is an Assistant Professor, and Alfred Frontera is an Associate Professor, all at the University of Central Florida College of Medicine in Orlando. Esther Baldinger and Alfred Frontera are Neurologists at the Bay Pines VA Healthcare System in Florida.
Correspondence: S. Hamad Sagheer (s.sagheer@knights.ucf.edu) * Co-lead authors.

Author disclosures
The authors report no actual or potential conflicts of interest with regard to this article.

Disclaimer
The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review the complete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

Issue
Federal Practitioner - 36(4)s
Publications
Topics
Page Number
S36-S41
Sections
Author and Disclosure Information

Ayan Nasir, Nimai Patel, Samantha Prabakaran, S. Hamad Sagheer, and Steven Troy are Medical Students, Esther Baldinger is an Assistant Professor, and Alfred Frontera is an Associate Professor, all at the University of Central Florida College of Medicine in Orlando. Esther Baldinger and Alfred Frontera are Neurologists at the Bay Pines VA Healthcare System in Florida.
Correspondence: S. Hamad Sagheer (s.sagheer@knights.ucf.edu) * Co-lead authors.

Author disclosures
The authors report no actual or potential conflicts of interest with regard to this article.

Disclaimer
The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review the complete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

Author and Disclosure Information

Ayan Nasir, Nimai Patel, Samantha Prabakaran, S. Hamad Sagheer, and Steven Troy are Medical Students, Esther Baldinger is an Assistant Professor, and Alfred Frontera is an Associate Professor, all at the University of Central Florida College of Medicine in Orlando. Esther Baldinger and Alfred Frontera are Neurologists at the Bay Pines VA Healthcare System in Florida.
Correspondence: S. Hamad Sagheer (s.sagheer@knights.ucf.edu) * Co-lead authors.

Author disclosures
The authors report no actual or potential conflicts of interest with regard to this article.

Disclaimer
The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review the complete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

Article PDF
Article PDF
Related Articles

The case of a female presenting with Shiga toxin-producing Escherichia coli and hemolytic uremic syndrome highlights a severe neurologic complication that canbe associated with these conditions.

The case of a female presenting with Shiga toxin-producing Escherichia coli and hemolytic uremic syndrome highlights a severe neurologic complication that canbe associated with these conditions.

Hemolytic uremic syndrome (HUS) is a rare illness that can be acquired through the consumption of food products contaminated with strains of Shiga toxin-producing Escherichia coli (E coli; STEC).1 Between 6% and 15% of individuals infected with STEC develop HUS, with children affected more frequently than adults.2,3 This strain of E coli releases Shiga toxin into the systemic circulation, which causes a thrombotic microangiopathy resulting in the characteristic HUS triad of symptoms: acute renal insufficiency, thrombocytopenia, and hemolytic anemia.4-6

Although neurologic features are common in HUS, they have not been extensively studied, particularly in adults. We report a case of STEC 0157:H7 subtype HUS in an adult with severe neurologic complications. This case highlights the neurological sequelae in an adult with typical STEC-HUS. The use of treatment modalities, such as plasmapheresis and eculizumab, and their use in adult typical STEC-HUS also is explored.

Case

A 53-year-old white woman with no pertinent past medical history presented to the Bay Pines Veterans Affairs Healthcare System Emergency Department with a 2-day history of abdominal pain, vomiting, nausea, diarrhea, and bright bloody stools. She returned from a cruise to the Bahamas 3 days prior, where she ate local foods, including salads. She reported no fever, shortness of breath, chest pain, headache, and cognitive difficulties. She presented with a normal mental status and neurologic exam. Apart from leukocytosis and elevated glucose level, her laboratory results at initial presentation were normal, (Table). A stool sample showed occult blood with white blood cell counts (WBCs) but was negative for Clostridium difficile. She was started on ciprofloxacin 400 mg and metronidazole 500 mg on the day of admission.

Hematuria was found on hospital day 2. On hospital day 4, the patient exhibited word finding difficulties. Blood studies revealed anemia, thrombocytopenia, leukocytosis, and increasing blood urea nitrogen (BUN) and creatinine. A computed tomography scan of the head was normal. Laboratory analysis showed schistocytes in the peripheral blood smear.

The patient’s cognitive functioning deteriorated on hospital day 5. She was not oriented to time or place. Her laboratory results showed complement level C3 at 70 mg/dL (ref: 83-193 mg/dL) complement C4 at 12 mg/dL (ref: 15-57mg/dL). The patient exhibited oliguria and hyponatremia, as well as both metabolic and respiratory acidosis; dialysis was then initiated. Results from the stool sample that was collected on hospital day 1 were received and tested positive for Shiga toxin.

At this point, the patient’s presentation of hemolytic anemia and thrombocytopenia in the setting of acute bloody diarrheal illness with known Shiga toxin, schistocytes on blood smear, and lack of pertinent medical history for other causes of this presentation made STEC-HUS the leading differential diagnosis. Plasmapheresis was ordered and performed on hospital day 6 and 7. Shiga toxin was no longer detected in the stool and antibiotics were stopped on hospital day 7.

The patient’s progressive deterioration in mental status continued on hospital day 8. She was not oriented to time or place, unable to perform simple calculations, and could not spell the word “hand” backwards. Physicians observed repetitive jerking motions of the upper extremities that were worse on the left side. An electroencephalogram (EEG) revealed right hemispheric sharp waves that were thought to be epileptiform (Figure 1). The patient began taking levetiracetam 1500 mg IV with 750 mg bid maintenance for seizure control. Plasmapheresis was discontinued due to her continued neurologic deterioration on this therapy. Consequently, eculizumab 900 mg IV was given along with the Neisseria meningitidis (N meningitidis) vaccine and a 19-day course of azithromycin 250 mg po as prophylaxis for encapsulated bacteria.

The patient continued to seize on hospital days 10 through 13. Oculocephalic maneuvers showed a tendency to keep her eyes deviated to the right. Her pupils continued to react to light. A repeat EEG showed diffuse slowing (5-6 Hz) with no epileptic activity seen (Figure 2). A second dose of eculizumab 900 mg IV was administered on hospital day 15. The patient experienced cardiac arrest on hospital day 16 and was successfully resuscitated. On hospital day 25 (10 days after receiving her second dose of eculizumab), the patient was able to speak and follow simple commands but exhibited difficulty concentrating and poor impulse control.

The patient was alert and oriented to person, place, time, and situation on hospital day 28 (6 days after the third and final dose of eculizumab). A neurologic exam was significant only for a slight intention tremor. She was continued on levetiracetam with a plan to be maintained on the medication for the next 6 months for seizure control. She was discharged on hospital day 30.

Twenty-eight days postdischarge (57 days postadmission), the patient showed marked recovery. She had returned to her previous employment as a business administrator on a part-time basis and exhibited no deficiencies in executive functioning or handling activities of daily living. Although she had been very active prior to this illness, she now experienced decreased physical and mental endurance; however, this gradually improved with physical therapy. She planned on returning to work on a full-time basis when she had regained her stamina. She also noticed difficulties in retaining short term memory since her discharge but believed that these symptoms were remitting. On examination her mental status and neurologic exam was significant for inability to continue serial 7s, left sided 4/5 muscle strength in quadriceps and thumb to 5th metacarpal adduction, bilateral 1+ reflexes in muscle groups tested (triceps, biceps, brachioradialis, patellar, and Achilles), loss of dull pinprick sensation bilaterally at web of hands, deficit in tandem gait while looking away, and slight intention tremor on finger to nose testing bilaterally (with left hand tremor more pronounced than right). Her complete blood count was normal. Her recovery continues to be monitored in an outpatient setting.

 

 

Discussion

HUS is characterized by 3 core clinical features: microangiopathic hemolytic anemia, thrombocytopenia, and acute kidney injury.4 Schistocytes are seen on peripheral blood smear and occur due to the passage of red blood cells over the microvascular thrombi induced by the disease. HUS can be classified as typical, atypical, or occurring with a coexisting disease. Typical HUS is associated with STEC 0157:H7 subtype, a bacterium known to be acquired through contaminated food and via human-to-human transmission.6-8 In the case of typical STEC 0157:H7, the bacterium releases a verotoxin that damages the vascular endothelium, thereby leading to activation of the coagulation cascade and eventually the formation of thrombi.4 It has been hypothesized that the Shiga toxin also activates the alternative complement pathway directly, which could contribute to thrombosis.9 This would explain the findings of low complement levels in our patient. Atypical HUS is primarily attributable to mutations in the alternative complement pathway. Causes for the third type of HUS can include Streptococcus pneumoniae, HIV, drug toxicity, and alterations in the metabolism of cobalamin C.

Epidemiologically, 15.3% of children aged < 5 years develop typical HUS after exposure to STEC compared with 1.2% of adults aged 18 to 59 years. The median age of patients who developed HUS from STEC exposure was 4 years compared with 16 years for those who did not develop HUS.2

Neurologic manifestations increase mortality for HUS patients.10 These have been described in the pediatric population as alteration in consciousness (85%), seizures (71%), pyramidal syndrome (52%), and extrapyramidal syndrome with hypertonia (42%).11 Brain imaging in children has demonstrated hemorrhagic lesions involving the pons, basal ganglia, and occipital cortex.11 Blood flow to areas such as the cerebellum, brainstem, and orbitofrontal area can be compromised.10 Adult patients with HUS can present without lesions on cranial magnetic resonance imaging (MRI), but instead with transient symmetric vasogenic edema of the central brain stem.12 Unfortunately in this case, MRI was not performed because it was thought to provide limited aid in diagnosis and to avoid unnecessary testing for the acutely ill patient.

The underlying pathophysiology of neurologic manifestations in patients may be due to a metabolic disturbance, toxin-mediated damage of the vascular endothelium, or toxin-induced cytokine release resulting in death of neural cells and subsequent neuroinflammation. However, the most likely mechanism is parenchymal ischemic changes related to microangiopathy.11,13 Pediatric patients often experience seizures and altered mental status, and their EEGs display delta waves.13 This patient’s diffuse slowing on her second EEG and altered mental status suggests that the neuropathologic mechanisms for typical HUS in adults may be similar to those in children.

HUS Treatment

The treatment and management of adults with typical STEC-HUS is evolving. The patient was first suspected to have an infectious colitis and empiric antibiotics were initiated. Some studies suggest that antibiotic administration may worsen the course of HUS in children as it may lead to release and subsequent absorption of Shiga toxin in the intestine.9,14 However, there is little evidence to suggest harm or efficacy of administration in adults. It is unclear what role antibiotic administration played in the recovery time of HUS given the co-administration of other treatments such as eculizumab and plasmapheresis, but it does appear to have helped with the initial E coli infection.

 

 

Plasmapheresis was subsequently administered, due to its documented benefit in the treatment of HUS.15 However, it should be noted that even though plasmapheresis is currently used in patients with CNS involvement, it remains unproven with conflicting information on its efficacy.3,16 The mechanism of action is unclear, but it has been hypothesized that plasmapheresis prevents microangiopathy caused by microthrombi.3,16 For this reason, eculizumab is becoming the mainstay for treatment of STEC-HUS with neurologic complications given the lack of well researched alternative treatments. In this case study, the use of plasmapheresis did not result in clinical improvement, and was abandoned after 2 days of treatment.

Eculizumab is a humanized, recombinant monoclonal IgG antibody that is a terminal complement inhibitor of the alternative complement system at the final step to cleave C5.17 The Shiga toxin may directly activate the complement system via the alternative pathway, which can result in uncontrolled platelet and white blood cell activation and depletion, endothelial cell damage, and hemolysis. The galvanized complement system leads to a series of cascading events that contribute to organ damage and death.9 Eculizumab is FDA approved for use in atypical HUS.18 It also can be used off-label to treat typical-HUS in adults with neurologic complications.

Eculizumab interferes with the immune response against encapsulated bacteria because it inhibits the alternative complement pathway. Thus, vaccination against N meningitides is recommended 2 weeks prior to the administration of eculizumab. However, in situations where the risks of delaying eculizumab for 2 weeks are greater than the risk of developing an N meningitides infection, eculizumab may be given without delay.18 Given the rapid deterioration of our patient’s condition, the vaccine and eculizumab were given together with prophylactic azithromycin. Although penicillin is the standard for prophylaxis in this situation, the patient’s penicillin allergy led to the use of azithromycin 250 mg po once a day. Literature also suggests azithromycin reduces the carriage duration of E coli-induced colitis.19 As such, it is possible that some improvement in the patient’s condition could be attributed to the elimination of the pathogen and toxin.

 

Conclusion

Three doses of eculizumab were administered at weekly intervals, with the first dose on hospital day 8 and the final dose on hospital day 22. Prior to the first dose, the patient displayed significant decline in mental status with EEG findings of right hemisphere epileptogenic discharges. After her third dose, she was found to have a drastically improved mental status exam and a normal EEG. One week later, she was discharged home. At the time of her 1-month follow-up, she was independent in all activities of daily living and had returned to part-time work. Apart from subtle cognitive changes, the remainder of her neurologic exam was normal.

There is evidence that supports the efficacy of eculizumab in children with HUS with neurologic symptoms on dialysis.20 However, its use in adults is not well established.21 This patient required dialysis and had neurologic symptoms similar to pediatric patients described in the literature, and responded similarly to the eculizumab. The rationale for the use of eculizumab in STEC-HUS also is evidenced by in vitro demonstrations of complement activation in STEC-HUS.22-25 This case report adds to the literature supporting the use of eculizumab in adult patients with typical HUS with neurological complications. Further research is necessary to develop guidelines in the treatment of adult STEC-HUS with regards to neurologic complications.

Acknowledgments

The authors would like to thank Pete DiStaso, REEGT for his work on obtaining the electroencephalograms and Anthony Rinaldi, PsyD; Julie Cessnapalas, PsyD; and Syed Faizan Sagheer for proof-reading the article.

Hemolytic uremic syndrome (HUS) is a rare illness that can be acquired through the consumption of food products contaminated with strains of Shiga toxin-producing Escherichia coli (E coli; STEC).1 Between 6% and 15% of individuals infected with STEC develop HUS, with children affected more frequently than adults.2,3 This strain of E coli releases Shiga toxin into the systemic circulation, which causes a thrombotic microangiopathy resulting in the characteristic HUS triad of symptoms: acute renal insufficiency, thrombocytopenia, and hemolytic anemia.4-6

Although neurologic features are common in HUS, they have not been extensively studied, particularly in adults. We report a case of STEC 0157:H7 subtype HUS in an adult with severe neurologic complications. This case highlights the neurological sequelae in an adult with typical STEC-HUS. The use of treatment modalities, such as plasmapheresis and eculizumab, and their use in adult typical STEC-HUS also is explored.

Case

A 53-year-old white woman with no pertinent past medical history presented to the Bay Pines Veterans Affairs Healthcare System Emergency Department with a 2-day history of abdominal pain, vomiting, nausea, diarrhea, and bright bloody stools. She returned from a cruise to the Bahamas 3 days prior, where she ate local foods, including salads. She reported no fever, shortness of breath, chest pain, headache, and cognitive difficulties. She presented with a normal mental status and neurologic exam. Apart from leukocytosis and elevated glucose level, her laboratory results at initial presentation were normal, (Table). A stool sample showed occult blood with white blood cell counts (WBCs) but was negative for Clostridium difficile. She was started on ciprofloxacin 400 mg and metronidazole 500 mg on the day of admission.

Hematuria was found on hospital day 2. On hospital day 4, the patient exhibited word finding difficulties. Blood studies revealed anemia, thrombocytopenia, leukocytosis, and increasing blood urea nitrogen (BUN) and creatinine. A computed tomography scan of the head was normal. Laboratory analysis showed schistocytes in the peripheral blood smear.

The patient’s cognitive functioning deteriorated on hospital day 5. She was not oriented to time or place. Her laboratory results showed complement level C3 at 70 mg/dL (ref: 83-193 mg/dL) complement C4 at 12 mg/dL (ref: 15-57mg/dL). The patient exhibited oliguria and hyponatremia, as well as both metabolic and respiratory acidosis; dialysis was then initiated. Results from the stool sample that was collected on hospital day 1 were received and tested positive for Shiga toxin.

At this point, the patient’s presentation of hemolytic anemia and thrombocytopenia in the setting of acute bloody diarrheal illness with known Shiga toxin, schistocytes on blood smear, and lack of pertinent medical history for other causes of this presentation made STEC-HUS the leading differential diagnosis. Plasmapheresis was ordered and performed on hospital day 6 and 7. Shiga toxin was no longer detected in the stool and antibiotics were stopped on hospital day 7.

The patient’s progressive deterioration in mental status continued on hospital day 8. She was not oriented to time or place, unable to perform simple calculations, and could not spell the word “hand” backwards. Physicians observed repetitive jerking motions of the upper extremities that were worse on the left side. An electroencephalogram (EEG) revealed right hemispheric sharp waves that were thought to be epileptiform (Figure 1). The patient began taking levetiracetam 1500 mg IV with 750 mg bid maintenance for seizure control. Plasmapheresis was discontinued due to her continued neurologic deterioration on this therapy. Consequently, eculizumab 900 mg IV was given along with the Neisseria meningitidis (N meningitidis) vaccine and a 19-day course of azithromycin 250 mg po as prophylaxis for encapsulated bacteria.

The patient continued to seize on hospital days 10 through 13. Oculocephalic maneuvers showed a tendency to keep her eyes deviated to the right. Her pupils continued to react to light. A repeat EEG showed diffuse slowing (5-6 Hz) with no epileptic activity seen (Figure 2). A second dose of eculizumab 900 mg IV was administered on hospital day 15. The patient experienced cardiac arrest on hospital day 16 and was successfully resuscitated. On hospital day 25 (10 days after receiving her second dose of eculizumab), the patient was able to speak and follow simple commands but exhibited difficulty concentrating and poor impulse control.

The patient was alert and oriented to person, place, time, and situation on hospital day 28 (6 days after the third and final dose of eculizumab). A neurologic exam was significant only for a slight intention tremor. She was continued on levetiracetam with a plan to be maintained on the medication for the next 6 months for seizure control. She was discharged on hospital day 30.

Twenty-eight days postdischarge (57 days postadmission), the patient showed marked recovery. She had returned to her previous employment as a business administrator on a part-time basis and exhibited no deficiencies in executive functioning or handling activities of daily living. Although she had been very active prior to this illness, she now experienced decreased physical and mental endurance; however, this gradually improved with physical therapy. She planned on returning to work on a full-time basis when she had regained her stamina. She also noticed difficulties in retaining short term memory since her discharge but believed that these symptoms were remitting. On examination her mental status and neurologic exam was significant for inability to continue serial 7s, left sided 4/5 muscle strength in quadriceps and thumb to 5th metacarpal adduction, bilateral 1+ reflexes in muscle groups tested (triceps, biceps, brachioradialis, patellar, and Achilles), loss of dull pinprick sensation bilaterally at web of hands, deficit in tandem gait while looking away, and slight intention tremor on finger to nose testing bilaterally (with left hand tremor more pronounced than right). Her complete blood count was normal. Her recovery continues to be monitored in an outpatient setting.

 

 

Discussion

HUS is characterized by 3 core clinical features: microangiopathic hemolytic anemia, thrombocytopenia, and acute kidney injury.4 Schistocytes are seen on peripheral blood smear and occur due to the passage of red blood cells over the microvascular thrombi induced by the disease. HUS can be classified as typical, atypical, or occurring with a coexisting disease. Typical HUS is associated with STEC 0157:H7 subtype, a bacterium known to be acquired through contaminated food and via human-to-human transmission.6-8 In the case of typical STEC 0157:H7, the bacterium releases a verotoxin that damages the vascular endothelium, thereby leading to activation of the coagulation cascade and eventually the formation of thrombi.4 It has been hypothesized that the Shiga toxin also activates the alternative complement pathway directly, which could contribute to thrombosis.9 This would explain the findings of low complement levels in our patient. Atypical HUS is primarily attributable to mutations in the alternative complement pathway. Causes for the third type of HUS can include Streptococcus pneumoniae, HIV, drug toxicity, and alterations in the metabolism of cobalamin C.

Epidemiologically, 15.3% of children aged < 5 years develop typical HUS after exposure to STEC compared with 1.2% of adults aged 18 to 59 years. The median age of patients who developed HUS from STEC exposure was 4 years compared with 16 years for those who did not develop HUS.2

Neurologic manifestations increase mortality for HUS patients.10 These have been described in the pediatric population as alteration in consciousness (85%), seizures (71%), pyramidal syndrome (52%), and extrapyramidal syndrome with hypertonia (42%).11 Brain imaging in children has demonstrated hemorrhagic lesions involving the pons, basal ganglia, and occipital cortex.11 Blood flow to areas such as the cerebellum, brainstem, and orbitofrontal area can be compromised.10 Adult patients with HUS can present without lesions on cranial magnetic resonance imaging (MRI), but instead with transient symmetric vasogenic edema of the central brain stem.12 Unfortunately in this case, MRI was not performed because it was thought to provide limited aid in diagnosis and to avoid unnecessary testing for the acutely ill patient.

The underlying pathophysiology of neurologic manifestations in patients may be due to a metabolic disturbance, toxin-mediated damage of the vascular endothelium, or toxin-induced cytokine release resulting in death of neural cells and subsequent neuroinflammation. However, the most likely mechanism is parenchymal ischemic changes related to microangiopathy.11,13 Pediatric patients often experience seizures and altered mental status, and their EEGs display delta waves.13 This patient’s diffuse slowing on her second EEG and altered mental status suggests that the neuropathologic mechanisms for typical HUS in adults may be similar to those in children.

HUS Treatment

The treatment and management of adults with typical STEC-HUS is evolving. The patient was first suspected to have an infectious colitis and empiric antibiotics were initiated. Some studies suggest that antibiotic administration may worsen the course of HUS in children as it may lead to release and subsequent absorption of Shiga toxin in the intestine.9,14 However, there is little evidence to suggest harm or efficacy of administration in adults. It is unclear what role antibiotic administration played in the recovery time of HUS given the co-administration of other treatments such as eculizumab and plasmapheresis, but it does appear to have helped with the initial E coli infection.

 

 

Plasmapheresis was subsequently administered, due to its documented benefit in the treatment of HUS.15 However, it should be noted that even though plasmapheresis is currently used in patients with CNS involvement, it remains unproven with conflicting information on its efficacy.3,16 The mechanism of action is unclear, but it has been hypothesized that plasmapheresis prevents microangiopathy caused by microthrombi.3,16 For this reason, eculizumab is becoming the mainstay for treatment of STEC-HUS with neurologic complications given the lack of well researched alternative treatments. In this case study, the use of plasmapheresis did not result in clinical improvement, and was abandoned after 2 days of treatment.

Eculizumab is a humanized, recombinant monoclonal IgG antibody that is a terminal complement inhibitor of the alternative complement system at the final step to cleave C5.17 The Shiga toxin may directly activate the complement system via the alternative pathway, which can result in uncontrolled platelet and white blood cell activation and depletion, endothelial cell damage, and hemolysis. The galvanized complement system leads to a series of cascading events that contribute to organ damage and death.9 Eculizumab is FDA approved for use in atypical HUS.18 It also can be used off-label to treat typical-HUS in adults with neurologic complications.

Eculizumab interferes with the immune response against encapsulated bacteria because it inhibits the alternative complement pathway. Thus, vaccination against N meningitides is recommended 2 weeks prior to the administration of eculizumab. However, in situations where the risks of delaying eculizumab for 2 weeks are greater than the risk of developing an N meningitides infection, eculizumab may be given without delay.18 Given the rapid deterioration of our patient’s condition, the vaccine and eculizumab were given together with prophylactic azithromycin. Although penicillin is the standard for prophylaxis in this situation, the patient’s penicillin allergy led to the use of azithromycin 250 mg po once a day. Literature also suggests azithromycin reduces the carriage duration of E coli-induced colitis.19 As such, it is possible that some improvement in the patient’s condition could be attributed to the elimination of the pathogen and toxin.

 

Conclusion

Three doses of eculizumab were administered at weekly intervals, with the first dose on hospital day 8 and the final dose on hospital day 22. Prior to the first dose, the patient displayed significant decline in mental status with EEG findings of right hemisphere epileptogenic discharges. After her third dose, she was found to have a drastically improved mental status exam and a normal EEG. One week later, she was discharged home. At the time of her 1-month follow-up, she was independent in all activities of daily living and had returned to part-time work. Apart from subtle cognitive changes, the remainder of her neurologic exam was normal.

There is evidence that supports the efficacy of eculizumab in children with HUS with neurologic symptoms on dialysis.20 However, its use in adults is not well established.21 This patient required dialysis and had neurologic symptoms similar to pediatric patients described in the literature, and responded similarly to the eculizumab. The rationale for the use of eculizumab in STEC-HUS also is evidenced by in vitro demonstrations of complement activation in STEC-HUS.22-25 This case report adds to the literature supporting the use of eculizumab in adult patients with typical HUS with neurological complications. Further research is necessary to develop guidelines in the treatment of adult STEC-HUS with regards to neurologic complications.

Acknowledgments

The authors would like to thank Pete DiStaso, REEGT for his work on obtaining the electroencephalograms and Anthony Rinaldi, PsyD; Julie Cessnapalas, PsyD; and Syed Faizan Sagheer for proof-reading the article.

References

1. Tarr PI, Gordon CA, Chandler WL. Shiga-toxin-producing Escherichia coli and haemolytic uraemic syndrome. Lancet. 2005;365(9464):1073-1086.

2. Gould LH, Demma L, Jones TF, et al. Hemolytic uremic syndrome and death in persons with Escherichia coli O157:H7 infection, foodborne diseases active surveillance network sites, 2000-2006. Clin Infect Dis. 2009;49(10):1480-1485.

3. Boyce TG, Swerdlow DL, Griffin PM. Escherichia coli O157:H7 and the hemolytic-uremic syndrome. N Engl J Med. 1995;333(6):364-368.

4. Rondeau E, Peraldi MN. Escherichia coli and the hemolytic-uremic syndrome. N Engl J Med. 1996;335(9):660-662.

5. Te Loo DM, van Hinsbergh VW, van den Heuvel LP, Monnens LA. Detection of verocytotoxin bound to circulating polymorphonuclear leukocytes of patients with hemolytic uremic syndrome. J Am Soc Nephrol. 2001;12(4):800-806.

6. Tran SL, Jenkins C, Livrelli V, Schüller S. Shiga toxin 2 translocation across intestinal epithelium is linked to virulence of Shiga toxin-producing Escherichia coli in humans. Microbiology. 2018;164(4):509-516.

7. Jokiranta TS. HUS and atypical HUS. Blood. 2017;129(21):2847-2856.

8. Ferens WA, Hovde CJ. Escherichia coli O157:H7: animal reservoir and sources of human infection. Foodborne Pathog Dis. 2011;8(4):465-487.

9. Percheron L, Gramada R, Tellier S, et al. Eculizumab treatment in severe pediatric STEC-HUS: a multicenter retrospective study. Pediatr Nephrol. 2018;33(8):1385-1394.

10. Hosaka T, Nakamagoe K, Tamaoka A. Hemolytic uremic syndrome-associated encephalopathy successfully treated with corticosteroids. Intern Med. 2017;56(21):2937-2941.

11. Nathanson S, Kwon T, Elmaleh M, et al. Acute neurological involvement in diarrhea-associated hemolytic uremic syndrome. Clin J Am Soc Nephrol. 2010;5(7):1218-1228.

12. Wengenroth M, Hoeltje J, Repenthin J, et al. Central nervous system involvement in adults with epidemic hemolytic uremic syndrome. AJNR Am J Neuroradiol. 2013;34(5):1016-1021, S1.

13. Eriksson KJ, Boyd SG, Tasker RC. Acute neurology and neurophysiology of haemolytic-uraemic syndrome. Arch Dis Child. 2001;84(5):434-435.

14. Wong CS, Jelacic S, Habeeb RL, Watkins SL, Tarr PI. The risk of the hemolytic-uremic syndrome after antibiotic treatment of Escherichia coli O157:H7 infections. N Engl J Med. 2000;342(26):1930-1936.

15. Nguyen TC, Kiss JE, Goldman JR, Carcillo JA. The role of plasmapheresis in critical illness. Crit Care Clin. 2012;28(3):453-468, vii.

16. Loos S, Ahlenstiel T, Kranz B, et al. An outbreak of Shiga toxin-producing Escherichia coli O104:H4 hemolytic uremic syndrome in Germany: presentation and short-term outcome in children. Clin Infect Dis. 2012;55(6):753-759.

17. Hossain MA, Cheema A, Kalathil S, et al. Atypical hemolytic uremic syndrome: Laboratory characteristics, complement-amplifying conditions, renal biopsy, and genetic mutations. Saudi J Kidney Dis Transpl. 2018;29(2):276-283.

18. Soliris (eculizumab) [package insert]. Cheshire, CT: Alexion Pharmaceuticals, Inc; 2011.

19. Keenswijk W, Raes A, Vande Walle J. Is eculizumab efficacious in Shigatoxin-associated hemolytic uremic syndrome? A narrative review of current evidence. Eur J Pediatr. 2018;177(3):311-318.

20. Lapeyraque AL, Malina M, Fremeaux-Bacchi V, et al. Eculizumab in severe Shiga-toxin-associated HUS. N Engl J Med. 2011;364(26):2561-2563.

21. Pape L, Hartmann H, Bange FC, Suerbaum S, Bueltmann E, Ahlenstiel-Grunow T. Eculizumab in typical hemolytic uremic syndrome (HUS) with neurological involvement. Medicine (Baltimore). 2015;94(24):e1000.

22. Kim Y, Miller K, Michael AF. Breakdown products of C3 and factor B in hemolytic-uremic syndrome. J Lab Clin Med. 1977;89(4):845-850.

23. Monnens L, Molenaar J, Lambert PH, Proesmans W, van Munster P. The complement system in hemolytic-uremic syndrome in childhood. Clin Nephrol. 1980;13(4):168-171.

24. Thurman JM, Marians R, Emlen W, et al. Alternative pathway of complement in children with diarrhea-associated hemolytic uremic syndrome. Clin J Am Soc Nephrol. 2009;4(12):1920-1924.

25. Ståhl AL, Sartz L, Karpman D. Complement activation on platelet-leukocyte complexes and microparticles in enterohemorrhagic Escherichia coli-induced hemolytic uremic syndrome. Blood. 2011;117(20):5503-5513.

References

1. Tarr PI, Gordon CA, Chandler WL. Shiga-toxin-producing Escherichia coli and haemolytic uraemic syndrome. Lancet. 2005;365(9464):1073-1086.

2. Gould LH, Demma L, Jones TF, et al. Hemolytic uremic syndrome and death in persons with Escherichia coli O157:H7 infection, foodborne diseases active surveillance network sites, 2000-2006. Clin Infect Dis. 2009;49(10):1480-1485.

3. Boyce TG, Swerdlow DL, Griffin PM. Escherichia coli O157:H7 and the hemolytic-uremic syndrome. N Engl J Med. 1995;333(6):364-368.

4. Rondeau E, Peraldi MN. Escherichia coli and the hemolytic-uremic syndrome. N Engl J Med. 1996;335(9):660-662.

5. Te Loo DM, van Hinsbergh VW, van den Heuvel LP, Monnens LA. Detection of verocytotoxin bound to circulating polymorphonuclear leukocytes of patients with hemolytic uremic syndrome. J Am Soc Nephrol. 2001;12(4):800-806.

6. Tran SL, Jenkins C, Livrelli V, Schüller S. Shiga toxin 2 translocation across intestinal epithelium is linked to virulence of Shiga toxin-producing Escherichia coli in humans. Microbiology. 2018;164(4):509-516.

7. Jokiranta TS. HUS and atypical HUS. Blood. 2017;129(21):2847-2856.

8. Ferens WA, Hovde CJ. Escherichia coli O157:H7: animal reservoir and sources of human infection. Foodborne Pathog Dis. 2011;8(4):465-487.

9. Percheron L, Gramada R, Tellier S, et al. Eculizumab treatment in severe pediatric STEC-HUS: a multicenter retrospective study. Pediatr Nephrol. 2018;33(8):1385-1394.

10. Hosaka T, Nakamagoe K, Tamaoka A. Hemolytic uremic syndrome-associated encephalopathy successfully treated with corticosteroids. Intern Med. 2017;56(21):2937-2941.

11. Nathanson S, Kwon T, Elmaleh M, et al. Acute neurological involvement in diarrhea-associated hemolytic uremic syndrome. Clin J Am Soc Nephrol. 2010;5(7):1218-1228.

12. Wengenroth M, Hoeltje J, Repenthin J, et al. Central nervous system involvement in adults with epidemic hemolytic uremic syndrome. AJNR Am J Neuroradiol. 2013;34(5):1016-1021, S1.

13. Eriksson KJ, Boyd SG, Tasker RC. Acute neurology and neurophysiology of haemolytic-uraemic syndrome. Arch Dis Child. 2001;84(5):434-435.

14. Wong CS, Jelacic S, Habeeb RL, Watkins SL, Tarr PI. The risk of the hemolytic-uremic syndrome after antibiotic treatment of Escherichia coli O157:H7 infections. N Engl J Med. 2000;342(26):1930-1936.

15. Nguyen TC, Kiss JE, Goldman JR, Carcillo JA. The role of plasmapheresis in critical illness. Crit Care Clin. 2012;28(3):453-468, vii.

16. Loos S, Ahlenstiel T, Kranz B, et al. An outbreak of Shiga toxin-producing Escherichia coli O104:H4 hemolytic uremic syndrome in Germany: presentation and short-term outcome in children. Clin Infect Dis. 2012;55(6):753-759.

17. Hossain MA, Cheema A, Kalathil S, et al. Atypical hemolytic uremic syndrome: Laboratory characteristics, complement-amplifying conditions, renal biopsy, and genetic mutations. Saudi J Kidney Dis Transpl. 2018;29(2):276-283.

18. Soliris (eculizumab) [package insert]. Cheshire, CT: Alexion Pharmaceuticals, Inc; 2011.

19. Keenswijk W, Raes A, Vande Walle J. Is eculizumab efficacious in Shigatoxin-associated hemolytic uremic syndrome? A narrative review of current evidence. Eur J Pediatr. 2018;177(3):311-318.

20. Lapeyraque AL, Malina M, Fremeaux-Bacchi V, et al. Eculizumab in severe Shiga-toxin-associated HUS. N Engl J Med. 2011;364(26):2561-2563.

21. Pape L, Hartmann H, Bange FC, Suerbaum S, Bueltmann E, Ahlenstiel-Grunow T. Eculizumab in typical hemolytic uremic syndrome (HUS) with neurological involvement. Medicine (Baltimore). 2015;94(24):e1000.

22. Kim Y, Miller K, Michael AF. Breakdown products of C3 and factor B in hemolytic-uremic syndrome. J Lab Clin Med. 1977;89(4):845-850.

23. Monnens L, Molenaar J, Lambert PH, Proesmans W, van Munster P. The complement system in hemolytic-uremic syndrome in childhood. Clin Nephrol. 1980;13(4):168-171.

24. Thurman JM, Marians R, Emlen W, et al. Alternative pathway of complement in children with diarrhea-associated hemolytic uremic syndrome. Clin J Am Soc Nephrol. 2009;4(12):1920-1924.

25. Ståhl AL, Sartz L, Karpman D. Complement activation on platelet-leukocyte complexes and microparticles in enterohemorrhagic Escherichia coli-induced hemolytic uremic syndrome. Blood. 2011;117(20):5503-5513.

Issue
Federal Practitioner - 36(4)s
Issue
Federal Practitioner - 36(4)s
Page Number
S36-S41
Page Number
S36-S41
Publications
Publications
Topics
Article Type
Display Headline
Hemolytic Uremic Syndrome With Severe Neurologic Complications in an Adult
Display Headline
Hemolytic Uremic Syndrome With Severe Neurologic Complications in an Adult
Sections
Citation Override
Fed Pract. 2019 June;36(suppl 4):S36-S41
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.
Article PDF Media