No evidence of pregnancy, but she is suicidal and depressed after ‘my baby died’

Article Type
Changed
Tue, 12/11/2018 - 15:29
Display Headline
No evidence of pregnancy, but she is suicidal and depressed after ‘my baby died’

CASE Depressed after she says her baby died
Ms. R, age 50, is an African-American woman who presents to a psychiatric hospital under an involuntary commitment executed by local law enforcement. Her sister called the authorities because Ms. R reportedly told her that she is “very depressed” and wants to “end [her] life” by taking an overdose of medications after the death of her newborn 1 week earlier.

Ms. R states that she delivered a child at “full term” in the emergency department of an outside community hospital, and that her current psychiatric symptoms began after the child died from “SIDS” [sudden infant death syndrome] shortly after birth.

Ms. R describes depressive symptoms including depressed mood, anhedonia, decreased energy, feelings of guilt, decreased concentration, poor sleep, and suicidal ideation. She denies substance use or a medical condition that could have induced these symptoms, and denies symptoms of mania, anxiety, or psychosis at admission or during the previous year.

Ms. R reports a history of manic episodes that includes periods of elevated mood or irritability, impulsivity, increased energy, excessive spending despite negative consequences, lack of need for sleep, rapid thoughts, and rapid speech that impaired her social and occupational functioning. Her most recent manic episode was approximately 3 years before this admission. She reports a previous suicide attempt and a history of physical abuse from a former intimate partner.

Neither the findings of a physical examination nor the results of a screening test for serum β-human chorionic gonadotropin (βHCG) are consistent with pregnancy. Ms. R’s medical record reveals that she was hospitalized for a “cardiac workup” a week earlier and requested investigation of possible pregnancy, which was negative. Records also reveal that she had a hysterectomy 10 years earlier.

Although Ms. R’s sister and boyfriend support her claim of pregnancy, the patient’s young adult son refutes it and states that she “does stuff like this for attention.” Her son also reports receiving a forged sonogram picture that his mother found online 1 month earlier. Ms. R presents an obituary from a local newspaper for the child but, on further investigation, the photograph of the infant was discovered to be of another child, also obtained online. Ms. R’s family denies knowledge of potential external reward Ms. R could gain by claiming to be pregnant.


Which of the following diagnoses can be considered after Ms. R’s initial presentation?

   a) somatic symptom disorder
   b) major depressive disorder
   c) bipolar I disorder
   d) delusional disorder


The authors’ observations

Ms. R reported the recent death of a newborn that was incompatible with her medical history. Her family members revealed that Ms. R made an active effort to deceive them about the reported pregnancy. She also exhibited symptoms of a major depressive episode in the context of previous manic episodes and expressed suicidal ideation.

The first step in the diagnostic pathway was to rule out possible medical explanations, including pregnancy, which could account for the patient’s symptoms. Although the serum βHCG level usually returns to non-pregnant levels 2 to 4 weeks after delivery, it can take even longer in some women.1 The absence of βHCG along with the recorded history of hysterectomy indicated that Ms. R was not pregnant at the time of testing or within the preceding few weeks. Once medical anomalies and substance use were ruled out, further classification of the psychiatric condition was undertaken.

One aspect of establishing a diagnosis for Ms. R is determining the presence of psychosis (eg, delusional thinking) (Table 1). Ms. R deliberately fabricated evidence of her pregnancy and manipulated family members, which indicated a low likelihood of delusions and supported a diagnostic alternative to psychosis.

Ms. R has a well-described history of manic episodes with current symptoms of a major depressive episode. The treatment team makes a diagnosis of bipolar I disorder, most recent episode depressed. The depressive symptoms Ms. R described were consistent with bipolar depression but did not explain her report of a pregnancy that is inconsistent with reality.

As is the case with Ms. R, diagnostic clarity often requires observation and evaluation over time. Building a strong therapeutic relationship with Ms. R in the context of an appropriate treatment plan allows the treatment team to explore the origin, motivations, and evolution of her thought content while managing her illness.


Confronting a patient about her false claims is likely to result in which of the following?

   a) spontaneous resolution of symptoms
   b) improved therapeutic alliance
   c) degradation of the patient’s coping mechanism
   d) violent outbursts by the patient

 

 


EVALUATION Confrontation
At admission, Ms. R remains resolute that she was pregnant and is suffering immense psychological distress secondary to the death of her child. Early in the treatment course, she is confronted with evidence indicating that her pregnancy was impossible. Shortly after this interaction, nursing staff alerts the treating physician that Ms. R experienced a “seizure-like spell” characterized by gross non-stereotyped jerking of the upper extremities, intact orientation, retention of bowel and bladder function, and coherent speech consistent with a diagnosis of pseudoseizure.2

Ms. R is transferred to a tertiary care facility for neurologic evaluation and observation. Ms. R repeatedly presents a photograph that she claims to be of her deceased child and implores the allied treatment team to advocate for discharge. Evaluation of Ms. R’s neurologic symptoms revealed no medical explanation for the “seizure-like spell” and she is transferred to the inpatient psychiatric hospital.

Upon return to the inpatient psychiatric unit, Ms. R receives intensive psychological exploration of her symptoms, thought content, and the foundation of her pregnancy claim. Within days, she acknowledges that the pregnancy was “not real” and that she was conscious of this fact in the months prior to hospitalization. She cites turmoil in her romantic relationship as the primary stimulus for her actions.


The authors’ observations

Ms. R’s reported pregnancy was not a delusion, but rather a deceitful exposition constructed with appropriate reality testing and a conscious awareness of the manipulation. This eliminated delusions as the explanation of her pregnancy claim. Although Ms. R initially rejected evidence refuting her belief of pregnancy, she recognized and accepted reality with appropriate intervention.


Factitious disorder vs malingering

Factitious disorder and malingering can present with intentional induction or report of symptoms or signs of a physical abnormality:

Factitious disorder imposed on the self is a willful misrepresentation or fabrication of signs or symptoms of an illness by a person in the absence of obvious personal gain that cannot be explained by a separate physical or mental illness (Table 2).3,4

Malingering is the intentional production or exaggeration of physical or psychological signs or symptoms with obvious secondary gain.

Malingering can be excluded in Ms. R’s case: She did not gain external reward by falsely reporting pregnancy. Although DSM-IV-TR (Table 2) assumes that the motivation for the patient with factitious disorder is to assume the sick role, DSM-5 merely states that the she (he) should present themselves as ill, impaired, or injured.3,4

Ms. R’s treatment team diagnosed factitious disorder imposed on self after careful exclusion of other causes for her symptoms. Bipolar I disorder, most recent episode depressed, also was diagnosed after considering Ms. R’s previous history of manic episodes and depressive symptoms at presentation.

Factitious disorder and other psychiatric conditions often are comorbid. Bipolar disorder, as in Ms. R’s case, as well as major depressive disorder commonly are comorbid with factitious disorder. It is also important to note that factitious disorder often occurs in the context of a personality disorder.5


Which of the following medications are FDA-approved for treating factitious disorder?

   a) olanzapine-fluoxetine combination
   b) lurasidone
   c) valproic acid
   d) all of the above
   e) no medications are approved for treating factitious disorder


TREATMENT Support, drug therapy
Treatment of Ms. R’s factitious disorder consists of psychological interventions via psychotherapy and strengthening of social support. She participates in daily individual therapy sessions as well as several group therapy activities. Ms. R engages with her social worker to facilitate a successful transition to an appropriate support network and access community resources to aid her wellness.

The treatment team feels that her diagnosis of bipolar I disorder, most recent episode depressed, warrants pharmacologic intervention. Ms. R agrees to begin a mood stabilizer, valproic acid, instead of medications FDA-approved to treat bipolar depression, such as lurasidone or quetiapine, because she reports good efficacy and tolerability when she took it during a major depressive episode approximately 4 years earlier.

Valproic acid is started at 250 mg/d and increased to 1,000 mg/d. Ms. R tolerates the medication without observed or reported adverse effects.


The authors’ observations

Managing factitious disorder can be challenging; patients can evoke strong feelings of countertransference during treatment.3,6,7 Providers might feel that the patient does not need to be treated, or that the patient is “not really sick.” This may induce anger and animosity toward the patient (therapeutic nihilism).8 These negative emotions are likely to disrupt the patient–provider relationship and exacerbate the patient’s symptoms.

It is generally accepted that the patient should be made aware of the treatment plan, in an indirect and tactful way, so that the patient does not feel “outed.” Unmasking the patient—the process of instilling insight—is a delicate step and can be a stressful time for the patient.9 A confrontational approach often places the patient’s sick role in doubt and does not address the pathological aspect of the disorder.

 

 

It is rare for a patient to admit to fabricating symptoms; confronted, the patient is likely to double their efforts to maintain the rouse of a fictional disease.10,11 It is important for the treatment team to be aware that patients frequently leave the treatment facility against medical advice, seek a different provider, or even pursue legal action for defamation against the treating physician.

Treating comorbid medical and psychiatric conditions is important for successful management of a patient with factitious disorder. Initiating valproic acid to address Ms. R’s bipolar depression contributed to her overall psychiatric stability. Initial treatment with a medication that is FDA-approved for treating bipolar depression, such as lurasidone, quetiapine, or olanzapine-fluoxetine combination, should be considered as an alternative. We chose valproic acid for Ms. R because of its previous efficacy, good tolerability, and the patient’s high level of comfort with the medication.


Which of the following are risk factors for factitious disorder?
  
a) lengthy medical treatments or hospitalizations as a child
   b) female sex
   c) experience as a health care worker
   d) all of the above


OUTCOME
Stabilization
Successful treatment during Ms. R’s inpatient psychiatric admission results in improved insight, remission of suicidal ideation, and stabilization of mood lability. She is discharged to the care of her family with a plan to follow up with a psychotherapist and psychiatrist. Continued administration of valproic acid continues to be effective after discharge.

Ms. R engages in frequent follow-up with outpatient psychiatric services. She remains engaged in psychotherapy and psychiatric care 1 year after discharge. Ms. R has made no report of pregnancy or required hospitalization during this time. She expresses trust in the mental health care system and acknowledges the role treatment played in her improvement.


BOTTOM LINE
Factitious disorder is a diagnostic and treatment challenge for psychiatrists. Identifying and treating comorbid psychiatric conditions is paramount for symptom resolution. Treatment consisting of acute intervention, psychological care, and frequent follow-up is effective and contributes to a good prognosis.


Related Resources

  • Bursch B. Munchausen by proxy and factitious disorder imposed on another. Psychiatric Times. http://www.psychiatrictimes.com/special-reports/munchausen-proxy-and-factitious-disorder-imposed-another.
  • Feldman M. Playing sick? Untangling the web of Munchausen syndrome, Munchausen by proxy, malingering, and factitious disorder. New York, NY: Brunner-Routledge; 2004.


Drug Brand Names

Lurasidone • Latuda
Quetiapine • Seroquel


Disclosures

The authors report no financial relationships with any company whose products are mentioned in this article or with manufacturers of competing products.
References


1. Reyes FI, Winter JS, Faiman C. Postpartum disappearance of chorionic gonadotropin from the maternal and neonatal circulations. Am J Obstet Gynecol. 1985;153(5):486-489.
2. Avbersek A, Sisodiya S. Does the primary literature provide support for clinical signs used to distinguish psychogenic nonepileptic seizures from epileptic seizures? J Neurol Neurosurg Psychiatry. 2010;81(7):719-725.
3. Diagnostic and statistical manual of mental disorders, 5th ed. Washington, DC: American Psychiatric Association; 2013.
4. Diagnostic and statistical manual of mental disorders, 4th ed, text rev. Washington, DC: American Psychiatric Association; 2000.
5. Kapfhammer HP, Rothenhausler HM, Dietrich E, et al. Artifactual disorders—between deception and self-mutilation. Experiences in consultation psychiatry at a university clinic [in German]. Nervenarzt. 1998;69(5):401-409.
6. Feldman MD, Feldman JM. Tangled in the web: countertransference in the therapy of factitious disorders. Int J Psychiatry Med. 1995;25(4):389-399.
7. Wedel KR. A therapeutic confrontation approach to treating patients with factitious illness. Soc Work. 1971;16(2):69-73.
8. Feldman MD, Hamilton JC, Deemer HN. Factitious disorder. In: Phillips KA, ed. Somatoform and factitious disorder. Washington, DC: American Psychiatric Press; 2001:129-159.
9. Scher LM, Knudsen P, Leamon M. Somatic symptom and related disorders. In: Hales RE, Yudofsky SC, Weiss Roberts L, eds. The American Publishing Psychiatric Publishing textbook of psychiatry. Arlington, VA: American Psychiatric Publishing; 2014:531-556.
10. Lipsitt DR. Introduction. In: Feldman MD, Eisendrath SJ, eds. The spectrum of factitious disorders. Washington, DC: American Psychiatric Press; 1996:xix-xxviii.
11. van der Feltz-Cornelis CM. Confronting patients about a factitious disorder [in Dutch]. Ned Tidjschr Geneeskd. 2000;144(12):545-548.

Article PDF
Author and Disclosure Information

Andrew Pierce, MD
Resident Psychiatrist
University of Florida
College of Medicine
Department of Psychiatry
Gainesville, Florida

Ana Turner, MD
Adjunct Clinical Assistant Professor
University of Florida
College of Medicine
Department of Psychiatry
Gainesville, Florida

Nadia Gilbo, MD
Resident Psychiatrist
Albert Einstein College of Medicine at Yeshiva University
Bronx, New York


Almari Ginory, DO
Assistant Professor
University of Florida
College of Medicine
Department of Psychiatry
Gainesville, Florida


Tessy Korah, MD
Assistant Professor
University of Florida
College of Medicine
Department of Psychiatry
Gainesville, Florida


Rajiv Tandon, MD
Professor
University of Florida
College of Medicine
Department of Psychiatry
Gainesville, Florida
Member, Editorial Board of Current Psychiatry

Issue
Current Psychiatry - 15(7)
Publications
Topics
Page Number
67-68,70-73
Legacy Keywords
pregnancy, pregnant, suicidal, depressed, depressive disorder, depressive disorders, depression, factitious disorder, mallingering,
Sections
Author and Disclosure Information

Andrew Pierce, MD
Resident Psychiatrist
University of Florida
College of Medicine
Department of Psychiatry
Gainesville, Florida

Ana Turner, MD
Adjunct Clinical Assistant Professor
University of Florida
College of Medicine
Department of Psychiatry
Gainesville, Florida

Nadia Gilbo, MD
Resident Psychiatrist
Albert Einstein College of Medicine at Yeshiva University
Bronx, New York


Almari Ginory, DO
Assistant Professor
University of Florida
College of Medicine
Department of Psychiatry
Gainesville, Florida


Tessy Korah, MD
Assistant Professor
University of Florida
College of Medicine
Department of Psychiatry
Gainesville, Florida


Rajiv Tandon, MD
Professor
University of Florida
College of Medicine
Department of Psychiatry
Gainesville, Florida
Member, Editorial Board of Current Psychiatry

Author and Disclosure Information

Andrew Pierce, MD
Resident Psychiatrist
University of Florida
College of Medicine
Department of Psychiatry
Gainesville, Florida

Ana Turner, MD
Adjunct Clinical Assistant Professor
University of Florida
College of Medicine
Department of Psychiatry
Gainesville, Florida

Nadia Gilbo, MD
Resident Psychiatrist
Albert Einstein College of Medicine at Yeshiva University
Bronx, New York


Almari Ginory, DO
Assistant Professor
University of Florida
College of Medicine
Department of Psychiatry
Gainesville, Florida


Tessy Korah, MD
Assistant Professor
University of Florida
College of Medicine
Department of Psychiatry
Gainesville, Florida


Rajiv Tandon, MD
Professor
University of Florida
College of Medicine
Department of Psychiatry
Gainesville, Florida
Member, Editorial Board of Current Psychiatry

Article PDF
Article PDF

CASE Depressed after she says her baby died
Ms. R, age 50, is an African-American woman who presents to a psychiatric hospital under an involuntary commitment executed by local law enforcement. Her sister called the authorities because Ms. R reportedly told her that she is “very depressed” and wants to “end [her] life” by taking an overdose of medications after the death of her newborn 1 week earlier.

Ms. R states that she delivered a child at “full term” in the emergency department of an outside community hospital, and that her current psychiatric symptoms began after the child died from “SIDS” [sudden infant death syndrome] shortly after birth.

Ms. R describes depressive symptoms including depressed mood, anhedonia, decreased energy, feelings of guilt, decreased concentration, poor sleep, and suicidal ideation. She denies substance use or a medical condition that could have induced these symptoms, and denies symptoms of mania, anxiety, or psychosis at admission or during the previous year.

Ms. R reports a history of manic episodes that includes periods of elevated mood or irritability, impulsivity, increased energy, excessive spending despite negative consequences, lack of need for sleep, rapid thoughts, and rapid speech that impaired her social and occupational functioning. Her most recent manic episode was approximately 3 years before this admission. She reports a previous suicide attempt and a history of physical abuse from a former intimate partner.

Neither the findings of a physical examination nor the results of a screening test for serum β-human chorionic gonadotropin (βHCG) are consistent with pregnancy. Ms. R’s medical record reveals that she was hospitalized for a “cardiac workup” a week earlier and requested investigation of possible pregnancy, which was negative. Records also reveal that she had a hysterectomy 10 years earlier.

Although Ms. R’s sister and boyfriend support her claim of pregnancy, the patient’s young adult son refutes it and states that she “does stuff like this for attention.” Her son also reports receiving a forged sonogram picture that his mother found online 1 month earlier. Ms. R presents an obituary from a local newspaper for the child but, on further investigation, the photograph of the infant was discovered to be of another child, also obtained online. Ms. R’s family denies knowledge of potential external reward Ms. R could gain by claiming to be pregnant.


Which of the following diagnoses can be considered after Ms. R’s initial presentation?

   a) somatic symptom disorder
   b) major depressive disorder
   c) bipolar I disorder
   d) delusional disorder


The authors’ observations

Ms. R reported the recent death of a newborn that was incompatible with her medical history. Her family members revealed that Ms. R made an active effort to deceive them about the reported pregnancy. She also exhibited symptoms of a major depressive episode in the context of previous manic episodes and expressed suicidal ideation.

The first step in the diagnostic pathway was to rule out possible medical explanations, including pregnancy, which could account for the patient’s symptoms. Although the serum βHCG level usually returns to non-pregnant levels 2 to 4 weeks after delivery, it can take even longer in some women.1 The absence of βHCG along with the recorded history of hysterectomy indicated that Ms. R was not pregnant at the time of testing or within the preceding few weeks. Once medical anomalies and substance use were ruled out, further classification of the psychiatric condition was undertaken.

One aspect of establishing a diagnosis for Ms. R is determining the presence of psychosis (eg, delusional thinking) (Table 1). Ms. R deliberately fabricated evidence of her pregnancy and manipulated family members, which indicated a low likelihood of delusions and supported a diagnostic alternative to psychosis.

Ms. R has a well-described history of manic episodes with current symptoms of a major depressive episode. The treatment team makes a diagnosis of bipolar I disorder, most recent episode depressed. The depressive symptoms Ms. R described were consistent with bipolar depression but did not explain her report of a pregnancy that is inconsistent with reality.

As is the case with Ms. R, diagnostic clarity often requires observation and evaluation over time. Building a strong therapeutic relationship with Ms. R in the context of an appropriate treatment plan allows the treatment team to explore the origin, motivations, and evolution of her thought content while managing her illness.


Confronting a patient about her false claims is likely to result in which of the following?

   a) spontaneous resolution of symptoms
   b) improved therapeutic alliance
   c) degradation of the patient’s coping mechanism
   d) violent outbursts by the patient

 

 


EVALUATION Confrontation
At admission, Ms. R remains resolute that she was pregnant and is suffering immense psychological distress secondary to the death of her child. Early in the treatment course, she is confronted with evidence indicating that her pregnancy was impossible. Shortly after this interaction, nursing staff alerts the treating physician that Ms. R experienced a “seizure-like spell” characterized by gross non-stereotyped jerking of the upper extremities, intact orientation, retention of bowel and bladder function, and coherent speech consistent with a diagnosis of pseudoseizure.2

Ms. R is transferred to a tertiary care facility for neurologic evaluation and observation. Ms. R repeatedly presents a photograph that she claims to be of her deceased child and implores the allied treatment team to advocate for discharge. Evaluation of Ms. R’s neurologic symptoms revealed no medical explanation for the “seizure-like spell” and she is transferred to the inpatient psychiatric hospital.

Upon return to the inpatient psychiatric unit, Ms. R receives intensive psychological exploration of her symptoms, thought content, and the foundation of her pregnancy claim. Within days, she acknowledges that the pregnancy was “not real” and that she was conscious of this fact in the months prior to hospitalization. She cites turmoil in her romantic relationship as the primary stimulus for her actions.


The authors’ observations

Ms. R’s reported pregnancy was not a delusion, but rather a deceitful exposition constructed with appropriate reality testing and a conscious awareness of the manipulation. This eliminated delusions as the explanation of her pregnancy claim. Although Ms. R initially rejected evidence refuting her belief of pregnancy, she recognized and accepted reality with appropriate intervention.


Factitious disorder vs malingering

Factitious disorder and malingering can present with intentional induction or report of symptoms or signs of a physical abnormality:

Factitious disorder imposed on the self is a willful misrepresentation or fabrication of signs or symptoms of an illness by a person in the absence of obvious personal gain that cannot be explained by a separate physical or mental illness (Table 2).3,4

Malingering is the intentional production or exaggeration of physical or psychological signs or symptoms with obvious secondary gain.

Malingering can be excluded in Ms. R’s case: She did not gain external reward by falsely reporting pregnancy. Although DSM-IV-TR (Table 2) assumes that the motivation for the patient with factitious disorder is to assume the sick role, DSM-5 merely states that the she (he) should present themselves as ill, impaired, or injured.3,4

Ms. R’s treatment team diagnosed factitious disorder imposed on self after careful exclusion of other causes for her symptoms. Bipolar I disorder, most recent episode depressed, also was diagnosed after considering Ms. R’s previous history of manic episodes and depressive symptoms at presentation.

Factitious disorder and other psychiatric conditions often are comorbid. Bipolar disorder, as in Ms. R’s case, as well as major depressive disorder commonly are comorbid with factitious disorder. It is also important to note that factitious disorder often occurs in the context of a personality disorder.5


Which of the following medications are FDA-approved for treating factitious disorder?

   a) olanzapine-fluoxetine combination
   b) lurasidone
   c) valproic acid
   d) all of the above
   e) no medications are approved for treating factitious disorder


TREATMENT Support, drug therapy
Treatment of Ms. R’s factitious disorder consists of psychological interventions via psychotherapy and strengthening of social support. She participates in daily individual therapy sessions as well as several group therapy activities. Ms. R engages with her social worker to facilitate a successful transition to an appropriate support network and access community resources to aid her wellness.

The treatment team feels that her diagnosis of bipolar I disorder, most recent episode depressed, warrants pharmacologic intervention. Ms. R agrees to begin a mood stabilizer, valproic acid, instead of medications FDA-approved to treat bipolar depression, such as lurasidone or quetiapine, because she reports good efficacy and tolerability when she took it during a major depressive episode approximately 4 years earlier.

Valproic acid is started at 250 mg/d and increased to 1,000 mg/d. Ms. R tolerates the medication without observed or reported adverse effects.


The authors’ observations

Managing factitious disorder can be challenging; patients can evoke strong feelings of countertransference during treatment.3,6,7 Providers might feel that the patient does not need to be treated, or that the patient is “not really sick.” This may induce anger and animosity toward the patient (therapeutic nihilism).8 These negative emotions are likely to disrupt the patient–provider relationship and exacerbate the patient’s symptoms.

It is generally accepted that the patient should be made aware of the treatment plan, in an indirect and tactful way, so that the patient does not feel “outed.” Unmasking the patient—the process of instilling insight—is a delicate step and can be a stressful time for the patient.9 A confrontational approach often places the patient’s sick role in doubt and does not address the pathological aspect of the disorder.

 

 

It is rare for a patient to admit to fabricating symptoms; confronted, the patient is likely to double their efforts to maintain the rouse of a fictional disease.10,11 It is important for the treatment team to be aware that patients frequently leave the treatment facility against medical advice, seek a different provider, or even pursue legal action for defamation against the treating physician.

Treating comorbid medical and psychiatric conditions is important for successful management of a patient with factitious disorder. Initiating valproic acid to address Ms. R’s bipolar depression contributed to her overall psychiatric stability. Initial treatment with a medication that is FDA-approved for treating bipolar depression, such as lurasidone, quetiapine, or olanzapine-fluoxetine combination, should be considered as an alternative. We chose valproic acid for Ms. R because of its previous efficacy, good tolerability, and the patient’s high level of comfort with the medication.


Which of the following are risk factors for factitious disorder?
  
a) lengthy medical treatments or hospitalizations as a child
   b) female sex
   c) experience as a health care worker
   d) all of the above


OUTCOME
Stabilization
Successful treatment during Ms. R’s inpatient psychiatric admission results in improved insight, remission of suicidal ideation, and stabilization of mood lability. She is discharged to the care of her family with a plan to follow up with a psychotherapist and psychiatrist. Continued administration of valproic acid continues to be effective after discharge.

Ms. R engages in frequent follow-up with outpatient psychiatric services. She remains engaged in psychotherapy and psychiatric care 1 year after discharge. Ms. R has made no report of pregnancy or required hospitalization during this time. She expresses trust in the mental health care system and acknowledges the role treatment played in her improvement.


BOTTOM LINE
Factitious disorder is a diagnostic and treatment challenge for psychiatrists. Identifying and treating comorbid psychiatric conditions is paramount for symptom resolution. Treatment consisting of acute intervention, psychological care, and frequent follow-up is effective and contributes to a good prognosis.


Related Resources

  • Bursch B. Munchausen by proxy and factitious disorder imposed on another. Psychiatric Times. http://www.psychiatrictimes.com/special-reports/munchausen-proxy-and-factitious-disorder-imposed-another.
  • Feldman M. Playing sick? Untangling the web of Munchausen syndrome, Munchausen by proxy, malingering, and factitious disorder. New York, NY: Brunner-Routledge; 2004.


Drug Brand Names

Lurasidone • Latuda
Quetiapine • Seroquel


Disclosures

The authors report no financial relationships with any company whose products are mentioned in this article or with manufacturers of competing products.

CASE Depressed after she says her baby died
Ms. R, age 50, is an African-American woman who presents to a psychiatric hospital under an involuntary commitment executed by local law enforcement. Her sister called the authorities because Ms. R reportedly told her that she is “very depressed” and wants to “end [her] life” by taking an overdose of medications after the death of her newborn 1 week earlier.

Ms. R states that she delivered a child at “full term” in the emergency department of an outside community hospital, and that her current psychiatric symptoms began after the child died from “SIDS” [sudden infant death syndrome] shortly after birth.

Ms. R describes depressive symptoms including depressed mood, anhedonia, decreased energy, feelings of guilt, decreased concentration, poor sleep, and suicidal ideation. She denies substance use or a medical condition that could have induced these symptoms, and denies symptoms of mania, anxiety, or psychosis at admission or during the previous year.

Ms. R reports a history of manic episodes that includes periods of elevated mood or irritability, impulsivity, increased energy, excessive spending despite negative consequences, lack of need for sleep, rapid thoughts, and rapid speech that impaired her social and occupational functioning. Her most recent manic episode was approximately 3 years before this admission. She reports a previous suicide attempt and a history of physical abuse from a former intimate partner.

Neither the findings of a physical examination nor the results of a screening test for serum β-human chorionic gonadotropin (βHCG) are consistent with pregnancy. Ms. R’s medical record reveals that she was hospitalized for a “cardiac workup” a week earlier and requested investigation of possible pregnancy, which was negative. Records also reveal that she had a hysterectomy 10 years earlier.

Although Ms. R’s sister and boyfriend support her claim of pregnancy, the patient’s young adult son refutes it and states that she “does stuff like this for attention.” Her son also reports receiving a forged sonogram picture that his mother found online 1 month earlier. Ms. R presents an obituary from a local newspaper for the child but, on further investigation, the photograph of the infant was discovered to be of another child, also obtained online. Ms. R’s family denies knowledge of potential external reward Ms. R could gain by claiming to be pregnant.


Which of the following diagnoses can be considered after Ms. R’s initial presentation?

   a) somatic symptom disorder
   b) major depressive disorder
   c) bipolar I disorder
   d) delusional disorder


The authors’ observations

Ms. R reported the recent death of a newborn that was incompatible with her medical history. Her family members revealed that Ms. R made an active effort to deceive them about the reported pregnancy. She also exhibited symptoms of a major depressive episode in the context of previous manic episodes and expressed suicidal ideation.

The first step in the diagnostic pathway was to rule out possible medical explanations, including pregnancy, which could account for the patient’s symptoms. Although the serum βHCG level usually returns to non-pregnant levels 2 to 4 weeks after delivery, it can take even longer in some women.1 The absence of βHCG along with the recorded history of hysterectomy indicated that Ms. R was not pregnant at the time of testing or within the preceding few weeks. Once medical anomalies and substance use were ruled out, further classification of the psychiatric condition was undertaken.

One aspect of establishing a diagnosis for Ms. R is determining the presence of psychosis (eg, delusional thinking) (Table 1). Ms. R deliberately fabricated evidence of her pregnancy and manipulated family members, which indicated a low likelihood of delusions and supported a diagnostic alternative to psychosis.

Ms. R has a well-described history of manic episodes with current symptoms of a major depressive episode. The treatment team makes a diagnosis of bipolar I disorder, most recent episode depressed. The depressive symptoms Ms. R described were consistent with bipolar depression but did not explain her report of a pregnancy that is inconsistent with reality.

As is the case with Ms. R, diagnostic clarity often requires observation and evaluation over time. Building a strong therapeutic relationship with Ms. R in the context of an appropriate treatment plan allows the treatment team to explore the origin, motivations, and evolution of her thought content while managing her illness.


Confronting a patient about her false claims is likely to result in which of the following?

   a) spontaneous resolution of symptoms
   b) improved therapeutic alliance
   c) degradation of the patient’s coping mechanism
   d) violent outbursts by the patient

 

 


EVALUATION Confrontation
At admission, Ms. R remains resolute that she was pregnant and is suffering immense psychological distress secondary to the death of her child. Early in the treatment course, she is confronted with evidence indicating that her pregnancy was impossible. Shortly after this interaction, nursing staff alerts the treating physician that Ms. R experienced a “seizure-like spell” characterized by gross non-stereotyped jerking of the upper extremities, intact orientation, retention of bowel and bladder function, and coherent speech consistent with a diagnosis of pseudoseizure.2

Ms. R is transferred to a tertiary care facility for neurologic evaluation and observation. Ms. R repeatedly presents a photograph that she claims to be of her deceased child and implores the allied treatment team to advocate for discharge. Evaluation of Ms. R’s neurologic symptoms revealed no medical explanation for the “seizure-like spell” and she is transferred to the inpatient psychiatric hospital.

Upon return to the inpatient psychiatric unit, Ms. R receives intensive psychological exploration of her symptoms, thought content, and the foundation of her pregnancy claim. Within days, she acknowledges that the pregnancy was “not real” and that she was conscious of this fact in the months prior to hospitalization. She cites turmoil in her romantic relationship as the primary stimulus for her actions.


The authors’ observations

Ms. R’s reported pregnancy was not a delusion, but rather a deceitful exposition constructed with appropriate reality testing and a conscious awareness of the manipulation. This eliminated delusions as the explanation of her pregnancy claim. Although Ms. R initially rejected evidence refuting her belief of pregnancy, she recognized and accepted reality with appropriate intervention.


Factitious disorder vs malingering

Factitious disorder and malingering can present with intentional induction or report of symptoms or signs of a physical abnormality:

Factitious disorder imposed on the self is a willful misrepresentation or fabrication of signs or symptoms of an illness by a person in the absence of obvious personal gain that cannot be explained by a separate physical or mental illness (Table 2).3,4

Malingering is the intentional production or exaggeration of physical or psychological signs or symptoms with obvious secondary gain.

Malingering can be excluded in Ms. R’s case: She did not gain external reward by falsely reporting pregnancy. Although DSM-IV-TR (Table 2) assumes that the motivation for the patient with factitious disorder is to assume the sick role, DSM-5 merely states that the she (he) should present themselves as ill, impaired, or injured.3,4

Ms. R’s treatment team diagnosed factitious disorder imposed on self after careful exclusion of other causes for her symptoms. Bipolar I disorder, most recent episode depressed, also was diagnosed after considering Ms. R’s previous history of manic episodes and depressive symptoms at presentation.

Factitious disorder and other psychiatric conditions often are comorbid. Bipolar disorder, as in Ms. R’s case, as well as major depressive disorder commonly are comorbid with factitious disorder. It is also important to note that factitious disorder often occurs in the context of a personality disorder.5


Which of the following medications are FDA-approved for treating factitious disorder?

   a) olanzapine-fluoxetine combination
   b) lurasidone
   c) valproic acid
   d) all of the above
   e) no medications are approved for treating factitious disorder


TREATMENT Support, drug therapy
Treatment of Ms. R’s factitious disorder consists of psychological interventions via psychotherapy and strengthening of social support. She participates in daily individual therapy sessions as well as several group therapy activities. Ms. R engages with her social worker to facilitate a successful transition to an appropriate support network and access community resources to aid her wellness.

The treatment team feels that her diagnosis of bipolar I disorder, most recent episode depressed, warrants pharmacologic intervention. Ms. R agrees to begin a mood stabilizer, valproic acid, instead of medications FDA-approved to treat bipolar depression, such as lurasidone or quetiapine, because she reports good efficacy and tolerability when she took it during a major depressive episode approximately 4 years earlier.

Valproic acid is started at 250 mg/d and increased to 1,000 mg/d. Ms. R tolerates the medication without observed or reported adverse effects.


The authors’ observations

Managing factitious disorder can be challenging; patients can evoke strong feelings of countertransference during treatment.3,6,7 Providers might feel that the patient does not need to be treated, or that the patient is “not really sick.” This may induce anger and animosity toward the patient (therapeutic nihilism).8 These negative emotions are likely to disrupt the patient–provider relationship and exacerbate the patient’s symptoms.

It is generally accepted that the patient should be made aware of the treatment plan, in an indirect and tactful way, so that the patient does not feel “outed.” Unmasking the patient—the process of instilling insight—is a delicate step and can be a stressful time for the patient.9 A confrontational approach often places the patient’s sick role in doubt and does not address the pathological aspect of the disorder.

 

 

It is rare for a patient to admit to fabricating symptoms; confronted, the patient is likely to double their efforts to maintain the rouse of a fictional disease.10,11 It is important for the treatment team to be aware that patients frequently leave the treatment facility against medical advice, seek a different provider, or even pursue legal action for defamation against the treating physician.

Treating comorbid medical and psychiatric conditions is important for successful management of a patient with factitious disorder. Initiating valproic acid to address Ms. R’s bipolar depression contributed to her overall psychiatric stability. Initial treatment with a medication that is FDA-approved for treating bipolar depression, such as lurasidone, quetiapine, or olanzapine-fluoxetine combination, should be considered as an alternative. We chose valproic acid for Ms. R because of its previous efficacy, good tolerability, and the patient’s high level of comfort with the medication.


Which of the following are risk factors for factitious disorder?
  
a) lengthy medical treatments or hospitalizations as a child
   b) female sex
   c) experience as a health care worker
   d) all of the above


OUTCOME
Stabilization
Successful treatment during Ms. R’s inpatient psychiatric admission results in improved insight, remission of suicidal ideation, and stabilization of mood lability. She is discharged to the care of her family with a plan to follow up with a psychotherapist and psychiatrist. Continued administration of valproic acid continues to be effective after discharge.

Ms. R engages in frequent follow-up with outpatient psychiatric services. She remains engaged in psychotherapy and psychiatric care 1 year after discharge. Ms. R has made no report of pregnancy or required hospitalization during this time. She expresses trust in the mental health care system and acknowledges the role treatment played in her improvement.


BOTTOM LINE
Factitious disorder is a diagnostic and treatment challenge for psychiatrists. Identifying and treating comorbid psychiatric conditions is paramount for symptom resolution. Treatment consisting of acute intervention, psychological care, and frequent follow-up is effective and contributes to a good prognosis.


Related Resources

  • Bursch B. Munchausen by proxy and factitious disorder imposed on another. Psychiatric Times. http://www.psychiatrictimes.com/special-reports/munchausen-proxy-and-factitious-disorder-imposed-another.
  • Feldman M. Playing sick? Untangling the web of Munchausen syndrome, Munchausen by proxy, malingering, and factitious disorder. New York, NY: Brunner-Routledge; 2004.


Drug Brand Names

Lurasidone • Latuda
Quetiapine • Seroquel


Disclosures

The authors report no financial relationships with any company whose products are mentioned in this article or with manufacturers of competing products.
References


1. Reyes FI, Winter JS, Faiman C. Postpartum disappearance of chorionic gonadotropin from the maternal and neonatal circulations. Am J Obstet Gynecol. 1985;153(5):486-489.
2. Avbersek A, Sisodiya S. Does the primary literature provide support for clinical signs used to distinguish psychogenic nonepileptic seizures from epileptic seizures? J Neurol Neurosurg Psychiatry. 2010;81(7):719-725.
3. Diagnostic and statistical manual of mental disorders, 5th ed. Washington, DC: American Psychiatric Association; 2013.
4. Diagnostic and statistical manual of mental disorders, 4th ed, text rev. Washington, DC: American Psychiatric Association; 2000.
5. Kapfhammer HP, Rothenhausler HM, Dietrich E, et al. Artifactual disorders—between deception and self-mutilation. Experiences in consultation psychiatry at a university clinic [in German]. Nervenarzt. 1998;69(5):401-409.
6. Feldman MD, Feldman JM. Tangled in the web: countertransference in the therapy of factitious disorders. Int J Psychiatry Med. 1995;25(4):389-399.
7. Wedel KR. A therapeutic confrontation approach to treating patients with factitious illness. Soc Work. 1971;16(2):69-73.
8. Feldman MD, Hamilton JC, Deemer HN. Factitious disorder. In: Phillips KA, ed. Somatoform and factitious disorder. Washington, DC: American Psychiatric Press; 2001:129-159.
9. Scher LM, Knudsen P, Leamon M. Somatic symptom and related disorders. In: Hales RE, Yudofsky SC, Weiss Roberts L, eds. The American Publishing Psychiatric Publishing textbook of psychiatry. Arlington, VA: American Psychiatric Publishing; 2014:531-556.
10. Lipsitt DR. Introduction. In: Feldman MD, Eisendrath SJ, eds. The spectrum of factitious disorders. Washington, DC: American Psychiatric Press; 1996:xix-xxviii.
11. van der Feltz-Cornelis CM. Confronting patients about a factitious disorder [in Dutch]. Ned Tidjschr Geneeskd. 2000;144(12):545-548.

References


1. Reyes FI, Winter JS, Faiman C. Postpartum disappearance of chorionic gonadotropin from the maternal and neonatal circulations. Am J Obstet Gynecol. 1985;153(5):486-489.
2. Avbersek A, Sisodiya S. Does the primary literature provide support for clinical signs used to distinguish psychogenic nonepileptic seizures from epileptic seizures? J Neurol Neurosurg Psychiatry. 2010;81(7):719-725.
3. Diagnostic and statistical manual of mental disorders, 5th ed. Washington, DC: American Psychiatric Association; 2013.
4. Diagnostic and statistical manual of mental disorders, 4th ed, text rev. Washington, DC: American Psychiatric Association; 2000.
5. Kapfhammer HP, Rothenhausler HM, Dietrich E, et al. Artifactual disorders—between deception and self-mutilation. Experiences in consultation psychiatry at a university clinic [in German]. Nervenarzt. 1998;69(5):401-409.
6. Feldman MD, Feldman JM. Tangled in the web: countertransference in the therapy of factitious disorders. Int J Psychiatry Med. 1995;25(4):389-399.
7. Wedel KR. A therapeutic confrontation approach to treating patients with factitious illness. Soc Work. 1971;16(2):69-73.
8. Feldman MD, Hamilton JC, Deemer HN. Factitious disorder. In: Phillips KA, ed. Somatoform and factitious disorder. Washington, DC: American Psychiatric Press; 2001:129-159.
9. Scher LM, Knudsen P, Leamon M. Somatic symptom and related disorders. In: Hales RE, Yudofsky SC, Weiss Roberts L, eds. The American Publishing Psychiatric Publishing textbook of psychiatry. Arlington, VA: American Psychiatric Publishing; 2014:531-556.
10. Lipsitt DR. Introduction. In: Feldman MD, Eisendrath SJ, eds. The spectrum of factitious disorders. Washington, DC: American Psychiatric Press; 1996:xix-xxviii.
11. van der Feltz-Cornelis CM. Confronting patients about a factitious disorder [in Dutch]. Ned Tidjschr Geneeskd. 2000;144(12):545-548.

Issue
Current Psychiatry - 15(7)
Issue
Current Psychiatry - 15(7)
Page Number
67-68,70-73
Page Number
67-68,70-73
Publications
Publications
Topics
Article Type
Display Headline
No evidence of pregnancy, but she is suicidal and depressed after ‘my baby died’
Display Headline
No evidence of pregnancy, but she is suicidal and depressed after ‘my baby died’
Legacy Keywords
pregnancy, pregnant, suicidal, depressed, depressive disorder, depressive disorders, depression, factitious disorder, mallingering,
Legacy Keywords
pregnancy, pregnant, suicidal, depressed, depressive disorder, depressive disorders, depression, factitious disorder, mallingering,
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Pregnant and nursing patients benefit from ‘ambitious’ changes to drug labeling for safety

Article Type
Changed
Tue, 12/11/2018 - 15:01
Display Headline
Pregnant and nursing patients benefit from ‘ambitious’ changes to drug labeling for safety

In December 2014, the FDA issued draft guidance for sweeping changes to labeling of pharmaceutical treatments in regard to pregnancy and lactation information. These changes are now in effect for use in practice.1 The undertaking has been years in the making, and is truly ambitious.

The outdated system of letter categories (A, B, C, D, X) falls short of clinical needs in several ways:

  • the quality and volume of data can be lacking
  • comparative risk is not described
  • using letters can led to oversimplification or, in some cases, exaggeration of risk and safety (Box).

Other drawbacks include infrequent updating of information and omission of information about baseline rates of reproductive-related adverse events, to provide a more meaningful context for risk assessment.

A note before we continue discussion of labeling: Recognize that pregnancy itself is inherently risky; poor outcomes are, regrettably, not uncommon. The rate of birth defects in the United States is approximately 3%, and obstetric complications, such as prematurity, are common.2,3


New system described
The new labeling content has been described in the FDA’s Pregnancy and Lactation Labeling Rule (also called the “final rule”), issued in December 2014. For each medication, there will be subsections in the labeling:

  • Pregnancy
  • Lactation
  • Females and Males of Reproductive Potential.

In addition, FDA instructions now state that labeling:

  • must be updated when new information becomes available
  • needs to include evaluation of human data that becomes available mainly after the drug is approved
  • needs to include information about the background rates of adverse events related to reproduction.

Labeling in pregnancy. As an example, the “Pregnancy” section of every label contains 3 subsections, all of great clinical importance. First is information about pregnancy exposure registries, with a listing of scientifically acceptable registries (if a registry is available for that drug) and contact information; this section focuses on the high value of data that are systematically and prospectively collected. The second summarizes risk associated with the drug during pregnancy, based on available human, animal, and pharmacologic data. Third is a discussion of clinical considerations.

Need for appropriate controls. Psychiatric disorders increase the risk of pregnancy complications, and often are associated with variables that might increase the risk of a poor pregnancy outcome. For example, a patient who has a psychiatric disorder might be less likely to seek prenatal care, take a prenatal vitamin, and sleep and eat well; she also might use alcohol, tobacco, or other substances of abuse.

The medical literature on the reproductive safety of psychotropic medications is fraught with confounding variables other than the medications themselves. These include variables that, taken alone, might confer a poorer outcome on the fetus or newborn of a pregnant or lactating woman who has a psychiatric illness (to the extent that she uses psychotropics during a pregnancy), compared with what would be seen in (1) a healthy woman who is not taking such medication or (2) the general population.

On the new labels, detailed statements on human data include information from clinical trials, pregnancy exposure registries, and epidemiologic studies. Labels are also to include:

  • incidence of adverse events
  • effect of dosage
  • effect of duration of exposure
  • effect of gestational timing of exposure.

The labels emphasize quantifying risk relative to the risk of the same outcome in infants born to women who have not been exposed to the particular drug, but who have the disease or condition for which the drug is indicated (ie, appropriate controls).

Clinical considerations are to include information on the following related to the specific medication (when that information is known):

  • more information for prescribers, to further risk-benefit counseling
  • disease-associated maternal-fetal risks
  • dosage adjustments during pregnancy and postpartum
  • maternal adverse reactions
  • fetal and neonatal adverse reactions
  • labor and delivery.

Clearly, this overdue shift in providing information regarding reproductive safety has the potential to inform clinicians and patients in a meaningful way about the risks and benefits of specific treatments during pregnancy and lactation. Translating that information into practice is daunting, however.


Important aspects of implementation

Pregnancy exposure registries will play a crucial role.
For most medications, no systematic registry has been established; to do so, rigorous methodology is required to acquire prospective data and account for confounding variables.4 Appropriate control groups also are required to yield data that are useful and interpretable. Primary outcomes require verification, such as review of medical records. Last, registries must be well-conducted and therefore adequately funded, yet labeling changes have not been accompanied by funding requirements set forth by regulators to pharmaceutical manufacturers.

Labeling must be updated continually. Furthermore, it is unclear who will review data for precision and comprehensiveness.

 

 

Data need to be understandable to health care providers across disciplines and to patients with varying levels of education for the label to have a meaningful impact on clinical care.

As noted, there is no mandate for funding the meticulous pharmacovigilance required to provide definitive data for labeling. It is unclear if the potential benefits of the new labeling can be reaped without adequate financing of the pharmacovigilance mechanisms required to inform patients adequately.


Role of pregnancy registries

Over the past 2 decades, pregnancy registries have emerged as a rapid, systematic means of collecting important reproductive safety data on the risk for major malformations after prenatal exposure to a medication or a class of medications.5,6 Such registries enhance the rigor of available cohort studies and other analyses of reproductive safety data that have been derived from large administrative databases.

NPRAA and NPRAD. Recently, the National Pregnancy Registry for Atypical Antipsychotics (NPRAA) and the National Pregnancy Registry for Antidepressants (NPRAD) were established in an effort to obtain reproductive safety data about fetal exposure to second-generation antipsychotics (SGAs) and to newer antidepressants.7 Based at Massachusetts General Hospital in Boston, NPRAA and NPRAD systematically and prospectively evaluate the risk of malformations among infants who have been exposed in utero to an SGA or an antidepressant.

The structure of both registries are the same, modeled after the North American Antiepileptic Drug Registry.5,8 Data are collected prospectively from pregnant women, age 18 to 45, by means of 3 telephone interviews conducted proximate to enrollment, at 7 months’ gestation, and at 2 or 3 months’ postpartum.

Participants include (1) pregnant women who have a history of fetal exposure to an SGA or an antidepressant, or both, and (2) a comparison group of non-exposed pregnant women who have a history of a psychiatric illness. Authorization for release of medical records is obtained for obstetric care, labor and delivery, and neonatal care (≤6 months of age).

Information on the presence of major malformations is abstracted from the medical record, along with other data on neonatal and maternal health outcomes. Identified cases of a congenital malformation are sent to a dysmorphologist, who has been blinded to drug exposure, for final adjudication. Release of findings is dictated by a governing Scientific Advisory Board.

Results so far. Results are available from the NPRAA.9 As of December 2014, 487 women were enrolled: 353 who used an SGA and 134 comparison women. Medical records were obtained for 82.2% of participants. A total of 303 women completed the study and were eligible for inclusion in the analysis. Findings include:

  • Of 214 live births with first-trimester exposure to an SGA, 3 major malformations were confirmed. In the control group (n = 89), 1 major malformation was confirmed
  • The absolute risk of a major malformation was 1.4% for an exposed infant and 1.1% for an unexposed infant
  • The odds ratio for a major malformation, comparing exposed infants with unexposed infants, was 1.25 (95% CI, 0.13–12.19).

It is reasonable, therefore, to conclude that, as a class, SGAs are not major teratogens. Although the confidence intervals around the odds ratio estimate remain wide, with the probability for change over the course of the study, it is unlikely that risk will rise to the level of known major teratogens, such as valproate and thalidomide.10,11


Help with decision-making

Given recent FDA guidance about the importance of pregnancy registries (www.fda.gov/pregnancyregistries), such carefully collected data might help clinicians and patients make informed choices about treatment. Future efforts of NPRAA and NPRAD will focus on sustaining growth in enrollment of participants so that the reproductive safety of SGAs and newer antidepressants can be delineated more clearly.

Last, you can refer potential participants to NPRAA and NPRAD by calling 1-866-961-2388. More information is available at www.womensmentalhealth.org.


Related Resources

  • Sahin L, Nallani SC, Tassinari MS. Medication use in pregnancy and the pregnancy and lactation labeling rule [published online April 15, 2016]. Clin Pharmacol Ther. doi: 10.1002/cpt.380.
  • Burt VK. Evidence-based pregnancy registries: good for babies and their mothers. Am J Psychiatry. 2016;173(3):208-210.
  • Wood W. What to tell your bipolar disorder patient who wants to breast-feed. Current Psychiatry. 2015;14(4):30-33.


Drug Brand Names

Thalidomide • Thalomid
Valproate • Depakote


Disclosures

Dr. Freeman receives grant or research support from JayMac Pharmaceuticals and Takeda Pharmaceuticals, and is a consultant to JDS Therapeutics and SAGE Therapeutics. She is a member of the Current Psychiatry Editorial Board.

Dr. Viguera receives grant or research support from Alkermes, Bristol-Myers Squibb/Otsuka America Pharmaceutical, and Sunovion Pharmaceuticals, Inc.

Dr. Cohen receives grant or research support from Alkermes, AstraZeneca, Bristol-Myers Squibb/Otsuka America Pharmaceutical, Cephalon, Ortho-McNeil-Janssen, Sunovion Pharmaceuticals, Inc., and Takeda/Lundbeck. He is a consultant to JDS Therapeutics.

A statement of commercial sponsorship of the National Pregnancy Registry for Atypical Antipsychotics appears at: https://womensmentalhealth.org/clinical-and-research-programs/pregnancyregistry/atypicalantipsychotic.

References


1. U.S. Department of Health and Human Services; Food and Drug Administration; Center for Drug Evaluation and Research (CDER); Center for Biologic Evaluation and Research (CBER). Pregnancy, lactation, and reproductive potential: labeling for human prescription drug and biological products—content and format: guidance for industry. http://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/UCM425398.pdf. Published December 2014. Accessed June 7, 2016.
2. Centers for Disease Control and Prevention. Birth defects. http://www.cdc.gov/ncbddd/birthdefects/facts.html. Updated September 21, 2005. Accessed June 7, 2016.
3. Centers for Disease Control and Prevention. Preterm birth. http://www.cdc.gov/reproductivehealth/maternalinfanthealth/pretermbirth.htm. Updated December 4, 2015. Accessed June 7, 2016.
4. U.S. Department of Health and Human Services; Food and Drug Administration; Center for Drug Evaluation and Research (CDER); Center for Biologic Evaluation and Research (CBER). Guidance for industry: establishing pregnancy exposure registries. http://www.fda.gov/downloads/ScienceResearch/SpecialTopics/WomensHealthResearch/UCM133332.pdf. Published August 2002. Accessed June 7, 2016.
5. Holmes LB, Wyszynski DF. North American antiepileptic drug pregnancy registry. Epilepsia. 2004;45(11):1465.
6. Tomson T, Battino D, Craig J, et al; ILAE Commission on Therapeutic Strategies. Pregnancy registries: differences, similarities, and possible harmonization. Epilepsia. 2010;51(5):909-915.
7. Cohen LS, Viguera AC, McInerney KA, et al. Establishment of the National Pregnancy Registry for Atypical Antipsychotics. J Clin Psychiatry. 2015;76(7):986-989.
8. Holmes LB, Wyszynski DF, Lieberman E. The AED (antiepileptic drug) pregnancy registry: a 6-year experience. Arch Neurol. 2004;61(5):673-678.
9. Cohen LS, Viguera AC, McInerney KA, et al. Reproductive safety of second-generation antipsychotics: current data from the Massachusetts General Hospital National Pregnancy Registry for Atypical Antipsychotics. Am J Psychiatry. 2016;173(3):263-270.
10. McBride WG. Thalidomide and congenital abnormalities. Lancet. 1961;2(7216):1358.
11. Wyszynski DF, Nambisan M, Surve T, et al; Antiepileptic Drug Pregnancy Registry. Increased rate of major malformations in offspring exposed to valproate during pregnancy. Neurology. 2005;64(6):961-965.

Article PDF
Author and Disclosure Information

Marlene P. Freeman, MD
Associate Professor of Psychiatry
Harvard Medical School
Associate Director
Perinatal and Reproductive Psychiatry Program
Massachusetts General Hospital
Boston, Massachusetts

Adele C. Viguera, MD, MPH
Assistant Professor
Cleveland Clinic Lerner College of Medicine
Director, Women’s Mental Health
Neurological Institute
Cleveland Clinic
Cleveland, Ohio

Lee S. Cohen, MD
Edmund N. and Carroll M. Carpenter Professor of Psychiatry
Harvard Medical School
Director
Center for Women’s Mental Health
Massachusetts General Hospital
Boston, Massachusetts

Issue
Current Psychiatry - 15(7)
Publications
Page Number
37-40
Legacy Keywords
pregnancy, lactation, pregnant, antipsychotic, antipsychotics, National Pregnancy Registry for Atypical Antipsychotics (NPRAA), the National Pregnancy Registry for Antidepressants (NPRAD), FDA
Sections
Author and Disclosure Information

Marlene P. Freeman, MD
Associate Professor of Psychiatry
Harvard Medical School
Associate Director
Perinatal and Reproductive Psychiatry Program
Massachusetts General Hospital
Boston, Massachusetts

Adele C. Viguera, MD, MPH
Assistant Professor
Cleveland Clinic Lerner College of Medicine
Director, Women’s Mental Health
Neurological Institute
Cleveland Clinic
Cleveland, Ohio

Lee S. Cohen, MD
Edmund N. and Carroll M. Carpenter Professor of Psychiatry
Harvard Medical School
Director
Center for Women’s Mental Health
Massachusetts General Hospital
Boston, Massachusetts

Author and Disclosure Information

Marlene P. Freeman, MD
Associate Professor of Psychiatry
Harvard Medical School
Associate Director
Perinatal and Reproductive Psychiatry Program
Massachusetts General Hospital
Boston, Massachusetts

Adele C. Viguera, MD, MPH
Assistant Professor
Cleveland Clinic Lerner College of Medicine
Director, Women’s Mental Health
Neurological Institute
Cleveland Clinic
Cleveland, Ohio

Lee S. Cohen, MD
Edmund N. and Carroll M. Carpenter Professor of Psychiatry
Harvard Medical School
Director
Center for Women’s Mental Health
Massachusetts General Hospital
Boston, Massachusetts

Article PDF
Article PDF

In December 2014, the FDA issued draft guidance for sweeping changes to labeling of pharmaceutical treatments in regard to pregnancy and lactation information. These changes are now in effect for use in practice.1 The undertaking has been years in the making, and is truly ambitious.

The outdated system of letter categories (A, B, C, D, X) falls short of clinical needs in several ways:

  • the quality and volume of data can be lacking
  • comparative risk is not described
  • using letters can led to oversimplification or, in some cases, exaggeration of risk and safety (Box).

Other drawbacks include infrequent updating of information and omission of information about baseline rates of reproductive-related adverse events, to provide a more meaningful context for risk assessment.

A note before we continue discussion of labeling: Recognize that pregnancy itself is inherently risky; poor outcomes are, regrettably, not uncommon. The rate of birth defects in the United States is approximately 3%, and obstetric complications, such as prematurity, are common.2,3


New system described
The new labeling content has been described in the FDA’s Pregnancy and Lactation Labeling Rule (also called the “final rule”), issued in December 2014. For each medication, there will be subsections in the labeling:

  • Pregnancy
  • Lactation
  • Females and Males of Reproductive Potential.

In addition, FDA instructions now state that labeling:

  • must be updated when new information becomes available
  • needs to include evaluation of human data that becomes available mainly after the drug is approved
  • needs to include information about the background rates of adverse events related to reproduction.

Labeling in pregnancy. As an example, the “Pregnancy” section of every label contains 3 subsections, all of great clinical importance. First is information about pregnancy exposure registries, with a listing of scientifically acceptable registries (if a registry is available for that drug) and contact information; this section focuses on the high value of data that are systematically and prospectively collected. The second summarizes risk associated with the drug during pregnancy, based on available human, animal, and pharmacologic data. Third is a discussion of clinical considerations.

Need for appropriate controls. Psychiatric disorders increase the risk of pregnancy complications, and often are associated with variables that might increase the risk of a poor pregnancy outcome. For example, a patient who has a psychiatric disorder might be less likely to seek prenatal care, take a prenatal vitamin, and sleep and eat well; she also might use alcohol, tobacco, or other substances of abuse.

The medical literature on the reproductive safety of psychotropic medications is fraught with confounding variables other than the medications themselves. These include variables that, taken alone, might confer a poorer outcome on the fetus or newborn of a pregnant or lactating woman who has a psychiatric illness (to the extent that she uses psychotropics during a pregnancy), compared with what would be seen in (1) a healthy woman who is not taking such medication or (2) the general population.

On the new labels, detailed statements on human data include information from clinical trials, pregnancy exposure registries, and epidemiologic studies. Labels are also to include:

  • incidence of adverse events
  • effect of dosage
  • effect of duration of exposure
  • effect of gestational timing of exposure.

The labels emphasize quantifying risk relative to the risk of the same outcome in infants born to women who have not been exposed to the particular drug, but who have the disease or condition for which the drug is indicated (ie, appropriate controls).

Clinical considerations are to include information on the following related to the specific medication (when that information is known):

  • more information for prescribers, to further risk-benefit counseling
  • disease-associated maternal-fetal risks
  • dosage adjustments during pregnancy and postpartum
  • maternal adverse reactions
  • fetal and neonatal adverse reactions
  • labor and delivery.

Clearly, this overdue shift in providing information regarding reproductive safety has the potential to inform clinicians and patients in a meaningful way about the risks and benefits of specific treatments during pregnancy and lactation. Translating that information into practice is daunting, however.


Important aspects of implementation

Pregnancy exposure registries will play a crucial role.
For most medications, no systematic registry has been established; to do so, rigorous methodology is required to acquire prospective data and account for confounding variables.4 Appropriate control groups also are required to yield data that are useful and interpretable. Primary outcomes require verification, such as review of medical records. Last, registries must be well-conducted and therefore adequately funded, yet labeling changes have not been accompanied by funding requirements set forth by regulators to pharmaceutical manufacturers.

Labeling must be updated continually. Furthermore, it is unclear who will review data for precision and comprehensiveness.

 

 

Data need to be understandable to health care providers across disciplines and to patients with varying levels of education for the label to have a meaningful impact on clinical care.

As noted, there is no mandate for funding the meticulous pharmacovigilance required to provide definitive data for labeling. It is unclear if the potential benefits of the new labeling can be reaped without adequate financing of the pharmacovigilance mechanisms required to inform patients adequately.


Role of pregnancy registries

Over the past 2 decades, pregnancy registries have emerged as a rapid, systematic means of collecting important reproductive safety data on the risk for major malformations after prenatal exposure to a medication or a class of medications.5,6 Such registries enhance the rigor of available cohort studies and other analyses of reproductive safety data that have been derived from large administrative databases.

NPRAA and NPRAD. Recently, the National Pregnancy Registry for Atypical Antipsychotics (NPRAA) and the National Pregnancy Registry for Antidepressants (NPRAD) were established in an effort to obtain reproductive safety data about fetal exposure to second-generation antipsychotics (SGAs) and to newer antidepressants.7 Based at Massachusetts General Hospital in Boston, NPRAA and NPRAD systematically and prospectively evaluate the risk of malformations among infants who have been exposed in utero to an SGA or an antidepressant.

The structure of both registries are the same, modeled after the North American Antiepileptic Drug Registry.5,8 Data are collected prospectively from pregnant women, age 18 to 45, by means of 3 telephone interviews conducted proximate to enrollment, at 7 months’ gestation, and at 2 or 3 months’ postpartum.

Participants include (1) pregnant women who have a history of fetal exposure to an SGA or an antidepressant, or both, and (2) a comparison group of non-exposed pregnant women who have a history of a psychiatric illness. Authorization for release of medical records is obtained for obstetric care, labor and delivery, and neonatal care (≤6 months of age).

Information on the presence of major malformations is abstracted from the medical record, along with other data on neonatal and maternal health outcomes. Identified cases of a congenital malformation are sent to a dysmorphologist, who has been blinded to drug exposure, for final adjudication. Release of findings is dictated by a governing Scientific Advisory Board.

Results so far. Results are available from the NPRAA.9 As of December 2014, 487 women were enrolled: 353 who used an SGA and 134 comparison women. Medical records were obtained for 82.2% of participants. A total of 303 women completed the study and were eligible for inclusion in the analysis. Findings include:

  • Of 214 live births with first-trimester exposure to an SGA, 3 major malformations were confirmed. In the control group (n = 89), 1 major malformation was confirmed
  • The absolute risk of a major malformation was 1.4% for an exposed infant and 1.1% for an unexposed infant
  • The odds ratio for a major malformation, comparing exposed infants with unexposed infants, was 1.25 (95% CI, 0.13–12.19).

It is reasonable, therefore, to conclude that, as a class, SGAs are not major teratogens. Although the confidence intervals around the odds ratio estimate remain wide, with the probability for change over the course of the study, it is unlikely that risk will rise to the level of known major teratogens, such as valproate and thalidomide.10,11


Help with decision-making

Given recent FDA guidance about the importance of pregnancy registries (www.fda.gov/pregnancyregistries), such carefully collected data might help clinicians and patients make informed choices about treatment. Future efforts of NPRAA and NPRAD will focus on sustaining growth in enrollment of participants so that the reproductive safety of SGAs and newer antidepressants can be delineated more clearly.

Last, you can refer potential participants to NPRAA and NPRAD by calling 1-866-961-2388. More information is available at www.womensmentalhealth.org.


Related Resources

  • Sahin L, Nallani SC, Tassinari MS. Medication use in pregnancy and the pregnancy and lactation labeling rule [published online April 15, 2016]. Clin Pharmacol Ther. doi: 10.1002/cpt.380.
  • Burt VK. Evidence-based pregnancy registries: good for babies and their mothers. Am J Psychiatry. 2016;173(3):208-210.
  • Wood W. What to tell your bipolar disorder patient who wants to breast-feed. Current Psychiatry. 2015;14(4):30-33.


Drug Brand Names

Thalidomide • Thalomid
Valproate • Depakote


Disclosures

Dr. Freeman receives grant or research support from JayMac Pharmaceuticals and Takeda Pharmaceuticals, and is a consultant to JDS Therapeutics and SAGE Therapeutics. She is a member of the Current Psychiatry Editorial Board.

Dr. Viguera receives grant or research support from Alkermes, Bristol-Myers Squibb/Otsuka America Pharmaceutical, and Sunovion Pharmaceuticals, Inc.

Dr. Cohen receives grant or research support from Alkermes, AstraZeneca, Bristol-Myers Squibb/Otsuka America Pharmaceutical, Cephalon, Ortho-McNeil-Janssen, Sunovion Pharmaceuticals, Inc., and Takeda/Lundbeck. He is a consultant to JDS Therapeutics.

A statement of commercial sponsorship of the National Pregnancy Registry for Atypical Antipsychotics appears at: https://womensmentalhealth.org/clinical-and-research-programs/pregnancyregistry/atypicalantipsychotic.

In December 2014, the FDA issued draft guidance for sweeping changes to labeling of pharmaceutical treatments in regard to pregnancy and lactation information. These changes are now in effect for use in practice.1 The undertaking has been years in the making, and is truly ambitious.

The outdated system of letter categories (A, B, C, D, X) falls short of clinical needs in several ways:

  • the quality and volume of data can be lacking
  • comparative risk is not described
  • using letters can led to oversimplification or, in some cases, exaggeration of risk and safety (Box).

Other drawbacks include infrequent updating of information and omission of information about baseline rates of reproductive-related adverse events, to provide a more meaningful context for risk assessment.

A note before we continue discussion of labeling: Recognize that pregnancy itself is inherently risky; poor outcomes are, regrettably, not uncommon. The rate of birth defects in the United States is approximately 3%, and obstetric complications, such as prematurity, are common.2,3


New system described
The new labeling content has been described in the FDA’s Pregnancy and Lactation Labeling Rule (also called the “final rule”), issued in December 2014. For each medication, there will be subsections in the labeling:

  • Pregnancy
  • Lactation
  • Females and Males of Reproductive Potential.

In addition, FDA instructions now state that labeling:

  • must be updated when new information becomes available
  • needs to include evaluation of human data that becomes available mainly after the drug is approved
  • needs to include information about the background rates of adverse events related to reproduction.

Labeling in pregnancy. As an example, the “Pregnancy” section of every label contains 3 subsections, all of great clinical importance. First is information about pregnancy exposure registries, with a listing of scientifically acceptable registries (if a registry is available for that drug) and contact information; this section focuses on the high value of data that are systematically and prospectively collected. The second summarizes risk associated with the drug during pregnancy, based on available human, animal, and pharmacologic data. Third is a discussion of clinical considerations.

Need for appropriate controls. Psychiatric disorders increase the risk of pregnancy complications, and often are associated with variables that might increase the risk of a poor pregnancy outcome. For example, a patient who has a psychiatric disorder might be less likely to seek prenatal care, take a prenatal vitamin, and sleep and eat well; she also might use alcohol, tobacco, or other substances of abuse.

The medical literature on the reproductive safety of psychotropic medications is fraught with confounding variables other than the medications themselves. These include variables that, taken alone, might confer a poorer outcome on the fetus or newborn of a pregnant or lactating woman who has a psychiatric illness (to the extent that she uses psychotropics during a pregnancy), compared with what would be seen in (1) a healthy woman who is not taking such medication or (2) the general population.

On the new labels, detailed statements on human data include information from clinical trials, pregnancy exposure registries, and epidemiologic studies. Labels are also to include:

  • incidence of adverse events
  • effect of dosage
  • effect of duration of exposure
  • effect of gestational timing of exposure.

The labels emphasize quantifying risk relative to the risk of the same outcome in infants born to women who have not been exposed to the particular drug, but who have the disease or condition for which the drug is indicated (ie, appropriate controls).

Clinical considerations are to include information on the following related to the specific medication (when that information is known):

  • more information for prescribers, to further risk-benefit counseling
  • disease-associated maternal-fetal risks
  • dosage adjustments during pregnancy and postpartum
  • maternal adverse reactions
  • fetal and neonatal adverse reactions
  • labor and delivery.

Clearly, this overdue shift in providing information regarding reproductive safety has the potential to inform clinicians and patients in a meaningful way about the risks and benefits of specific treatments during pregnancy and lactation. Translating that information into practice is daunting, however.


Important aspects of implementation

Pregnancy exposure registries will play a crucial role.
For most medications, no systematic registry has been established; to do so, rigorous methodology is required to acquire prospective data and account for confounding variables.4 Appropriate control groups also are required to yield data that are useful and interpretable. Primary outcomes require verification, such as review of medical records. Last, registries must be well-conducted and therefore adequately funded, yet labeling changes have not been accompanied by funding requirements set forth by regulators to pharmaceutical manufacturers.

Labeling must be updated continually. Furthermore, it is unclear who will review data for precision and comprehensiveness.

 

 

Data need to be understandable to health care providers across disciplines and to patients with varying levels of education for the label to have a meaningful impact on clinical care.

As noted, there is no mandate for funding the meticulous pharmacovigilance required to provide definitive data for labeling. It is unclear if the potential benefits of the new labeling can be reaped without adequate financing of the pharmacovigilance mechanisms required to inform patients adequately.


Role of pregnancy registries

Over the past 2 decades, pregnancy registries have emerged as a rapid, systematic means of collecting important reproductive safety data on the risk for major malformations after prenatal exposure to a medication or a class of medications.5,6 Such registries enhance the rigor of available cohort studies and other analyses of reproductive safety data that have been derived from large administrative databases.

NPRAA and NPRAD. Recently, the National Pregnancy Registry for Atypical Antipsychotics (NPRAA) and the National Pregnancy Registry for Antidepressants (NPRAD) were established in an effort to obtain reproductive safety data about fetal exposure to second-generation antipsychotics (SGAs) and to newer antidepressants.7 Based at Massachusetts General Hospital in Boston, NPRAA and NPRAD systematically and prospectively evaluate the risk of malformations among infants who have been exposed in utero to an SGA or an antidepressant.

The structure of both registries are the same, modeled after the North American Antiepileptic Drug Registry.5,8 Data are collected prospectively from pregnant women, age 18 to 45, by means of 3 telephone interviews conducted proximate to enrollment, at 7 months’ gestation, and at 2 or 3 months’ postpartum.

Participants include (1) pregnant women who have a history of fetal exposure to an SGA or an antidepressant, or both, and (2) a comparison group of non-exposed pregnant women who have a history of a psychiatric illness. Authorization for release of medical records is obtained for obstetric care, labor and delivery, and neonatal care (≤6 months of age).

Information on the presence of major malformations is abstracted from the medical record, along with other data on neonatal and maternal health outcomes. Identified cases of a congenital malformation are sent to a dysmorphologist, who has been blinded to drug exposure, for final adjudication. Release of findings is dictated by a governing Scientific Advisory Board.

Results so far. Results are available from the NPRAA.9 As of December 2014, 487 women were enrolled: 353 who used an SGA and 134 comparison women. Medical records were obtained for 82.2% of participants. A total of 303 women completed the study and were eligible for inclusion in the analysis. Findings include:

  • Of 214 live births with first-trimester exposure to an SGA, 3 major malformations were confirmed. In the control group (n = 89), 1 major malformation was confirmed
  • The absolute risk of a major malformation was 1.4% for an exposed infant and 1.1% for an unexposed infant
  • The odds ratio for a major malformation, comparing exposed infants with unexposed infants, was 1.25 (95% CI, 0.13–12.19).

It is reasonable, therefore, to conclude that, as a class, SGAs are not major teratogens. Although the confidence intervals around the odds ratio estimate remain wide, with the probability for change over the course of the study, it is unlikely that risk will rise to the level of known major teratogens, such as valproate and thalidomide.10,11


Help with decision-making

Given recent FDA guidance about the importance of pregnancy registries (www.fda.gov/pregnancyregistries), such carefully collected data might help clinicians and patients make informed choices about treatment. Future efforts of NPRAA and NPRAD will focus on sustaining growth in enrollment of participants so that the reproductive safety of SGAs and newer antidepressants can be delineated more clearly.

Last, you can refer potential participants to NPRAA and NPRAD by calling 1-866-961-2388. More information is available at www.womensmentalhealth.org.


Related Resources

  • Sahin L, Nallani SC, Tassinari MS. Medication use in pregnancy and the pregnancy and lactation labeling rule [published online April 15, 2016]. Clin Pharmacol Ther. doi: 10.1002/cpt.380.
  • Burt VK. Evidence-based pregnancy registries: good for babies and their mothers. Am J Psychiatry. 2016;173(3):208-210.
  • Wood W. What to tell your bipolar disorder patient who wants to breast-feed. Current Psychiatry. 2015;14(4):30-33.


Drug Brand Names

Thalidomide • Thalomid
Valproate • Depakote


Disclosures

Dr. Freeman receives grant or research support from JayMac Pharmaceuticals and Takeda Pharmaceuticals, and is a consultant to JDS Therapeutics and SAGE Therapeutics. She is a member of the Current Psychiatry Editorial Board.

Dr. Viguera receives grant or research support from Alkermes, Bristol-Myers Squibb/Otsuka America Pharmaceutical, and Sunovion Pharmaceuticals, Inc.

Dr. Cohen receives grant or research support from Alkermes, AstraZeneca, Bristol-Myers Squibb/Otsuka America Pharmaceutical, Cephalon, Ortho-McNeil-Janssen, Sunovion Pharmaceuticals, Inc., and Takeda/Lundbeck. He is a consultant to JDS Therapeutics.

A statement of commercial sponsorship of the National Pregnancy Registry for Atypical Antipsychotics appears at: https://womensmentalhealth.org/clinical-and-research-programs/pregnancyregistry/atypicalantipsychotic.

References


1. U.S. Department of Health and Human Services; Food and Drug Administration; Center for Drug Evaluation and Research (CDER); Center for Biologic Evaluation and Research (CBER). Pregnancy, lactation, and reproductive potential: labeling for human prescription drug and biological products—content and format: guidance for industry. http://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/UCM425398.pdf. Published December 2014. Accessed June 7, 2016.
2. Centers for Disease Control and Prevention. Birth defects. http://www.cdc.gov/ncbddd/birthdefects/facts.html. Updated September 21, 2005. Accessed June 7, 2016.
3. Centers for Disease Control and Prevention. Preterm birth. http://www.cdc.gov/reproductivehealth/maternalinfanthealth/pretermbirth.htm. Updated December 4, 2015. Accessed June 7, 2016.
4. U.S. Department of Health and Human Services; Food and Drug Administration; Center for Drug Evaluation and Research (CDER); Center for Biologic Evaluation and Research (CBER). Guidance for industry: establishing pregnancy exposure registries. http://www.fda.gov/downloads/ScienceResearch/SpecialTopics/WomensHealthResearch/UCM133332.pdf. Published August 2002. Accessed June 7, 2016.
5. Holmes LB, Wyszynski DF. North American antiepileptic drug pregnancy registry. Epilepsia. 2004;45(11):1465.
6. Tomson T, Battino D, Craig J, et al; ILAE Commission on Therapeutic Strategies. Pregnancy registries: differences, similarities, and possible harmonization. Epilepsia. 2010;51(5):909-915.
7. Cohen LS, Viguera AC, McInerney KA, et al. Establishment of the National Pregnancy Registry for Atypical Antipsychotics. J Clin Psychiatry. 2015;76(7):986-989.
8. Holmes LB, Wyszynski DF, Lieberman E. The AED (antiepileptic drug) pregnancy registry: a 6-year experience. Arch Neurol. 2004;61(5):673-678.
9. Cohen LS, Viguera AC, McInerney KA, et al. Reproductive safety of second-generation antipsychotics: current data from the Massachusetts General Hospital National Pregnancy Registry for Atypical Antipsychotics. Am J Psychiatry. 2016;173(3):263-270.
10. McBride WG. Thalidomide and congenital abnormalities. Lancet. 1961;2(7216):1358.
11. Wyszynski DF, Nambisan M, Surve T, et al; Antiepileptic Drug Pregnancy Registry. Increased rate of major malformations in offspring exposed to valproate during pregnancy. Neurology. 2005;64(6):961-965.

References


1. U.S. Department of Health and Human Services; Food and Drug Administration; Center for Drug Evaluation and Research (CDER); Center for Biologic Evaluation and Research (CBER). Pregnancy, lactation, and reproductive potential: labeling for human prescription drug and biological products—content and format: guidance for industry. http://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/UCM425398.pdf. Published December 2014. Accessed June 7, 2016.
2. Centers for Disease Control and Prevention. Birth defects. http://www.cdc.gov/ncbddd/birthdefects/facts.html. Updated September 21, 2005. Accessed June 7, 2016.
3. Centers for Disease Control and Prevention. Preterm birth. http://www.cdc.gov/reproductivehealth/maternalinfanthealth/pretermbirth.htm. Updated December 4, 2015. Accessed June 7, 2016.
4. U.S. Department of Health and Human Services; Food and Drug Administration; Center for Drug Evaluation and Research (CDER); Center for Biologic Evaluation and Research (CBER). Guidance for industry: establishing pregnancy exposure registries. http://www.fda.gov/downloads/ScienceResearch/SpecialTopics/WomensHealthResearch/UCM133332.pdf. Published August 2002. Accessed June 7, 2016.
5. Holmes LB, Wyszynski DF. North American antiepileptic drug pregnancy registry. Epilepsia. 2004;45(11):1465.
6. Tomson T, Battino D, Craig J, et al; ILAE Commission on Therapeutic Strategies. Pregnancy registries: differences, similarities, and possible harmonization. Epilepsia. 2010;51(5):909-915.
7. Cohen LS, Viguera AC, McInerney KA, et al. Establishment of the National Pregnancy Registry for Atypical Antipsychotics. J Clin Psychiatry. 2015;76(7):986-989.
8. Holmes LB, Wyszynski DF, Lieberman E. The AED (antiepileptic drug) pregnancy registry: a 6-year experience. Arch Neurol. 2004;61(5):673-678.
9. Cohen LS, Viguera AC, McInerney KA, et al. Reproductive safety of second-generation antipsychotics: current data from the Massachusetts General Hospital National Pregnancy Registry for Atypical Antipsychotics. Am J Psychiatry. 2016;173(3):263-270.
10. McBride WG. Thalidomide and congenital abnormalities. Lancet. 1961;2(7216):1358.
11. Wyszynski DF, Nambisan M, Surve T, et al; Antiepileptic Drug Pregnancy Registry. Increased rate of major malformations in offspring exposed to valproate during pregnancy. Neurology. 2005;64(6):961-965.

Issue
Current Psychiatry - 15(7)
Issue
Current Psychiatry - 15(7)
Page Number
37-40
Page Number
37-40
Publications
Publications
Article Type
Display Headline
Pregnant and nursing patients benefit from ‘ambitious’ changes to drug labeling for safety
Display Headline
Pregnant and nursing patients benefit from ‘ambitious’ changes to drug labeling for safety
Legacy Keywords
pregnancy, lactation, pregnant, antipsychotic, antipsychotics, National Pregnancy Registry for Atypical Antipsychotics (NPRAA), the National Pregnancy Registry for Antidepressants (NPRAD), FDA
Legacy Keywords
pregnancy, lactation, pregnant, antipsychotic, antipsychotics, National Pregnancy Registry for Atypical Antipsychotics (NPRAA), the National Pregnancy Registry for Antidepressants (NPRAD), FDA
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Adding obinutuzumab to bendamustine boosts progression-free survival in rituximab-refractory indolent non-Hodgkin lymphoma

Article Type
Changed
Fri, 01/04/2019 - 09:52
Display Headline
Adding obinutuzumab to bendamustine boosts progression-free survival in rituximab-refractory indolent non-Hodgkin lymphoma

Obinutuzumab and bendamustine followed by obinutuzumab maintenance therapy was superior to bendamustine monotherapy based on progression-free survival in rituximab-refractory patients with indolent non-Hodgkin lymphoma, based on a study published online in the Lancet Oncology.

After a median follow-up of 22 months in the obinutuzumab plus bendamustine group and 20 months in the bendamustine monotherapy group, progression-free survival was significantly longer with obinutuzumab plus bendamustine (median not reached; 95% confidence interval, 22.5 months – not estimable) than with bendamustine monotherapy (14.9 months, range, 12.8-16.6; hazard ratio, 0.55; 95% CI 0.40-0.74; P = .0001). About two-thirds of the nearly 400 patients in both study arms had grade 3-5 adverse events.

The anti-CD20 monoclonal antibody obinutuzumab is an option when patients with indolent non-Hodgkin lymphoma relapse or don’t achieve adequate disease control with rituximab-based treatment, wrote Laurie H. Sehn, MD, of the British Columbia Cancer Agency and the University of British Columbia, Vancouver, and her colleagues.

In an open-label, randomized, phase III study called GADOLIN, patients with CD20-positive indolent non-Hodgkin lymphoma were stratified by indolent non-Hodgkin lymphoma subtype, rituximab-refractory type, number of previous therapies, and geographic region.

For the study, 194 patients were assigned to obinutuzumab plus bendamustine and 202 to bendamustine monotherapy. Trial participants received six 28-day cycles with either obinutuzumab plus bendamustine (obinutuzumab 1,000 mg on days 1, 8, and 15, cycle 1; and on day 1, cycles 2-6) plus bendamustine (90 mg/m2 per day on days 1 and 2, cycles 1-6) or bendamustine monotherapy (120 mg/m2 per day on days 1 and 2 of all cycles). Patients in the obinutuzumab plus bendamustine group whose disease did not progress received obinutuzumab maintenance therapy of 1,000 mg once every 2 months for up to 2 years.

Grade 3-5 adverse events occurred in 68% of 194 patients in the obinutuzumab plus bendamustine group and in 62% of 198 patients in the bendamustine monotherapy group. Grade 3 or worse neutropenia affected 33% of the obinutuzumab plus bendamustine group and 26% of the bendamustine monotherapy group. Other grade 3 or worse events included thrombocytopenia in 11% and 16%, anemia in 8% and 10%, and infusion-related reactions in 11% and 6%. Serious adverse events occurred in 38% in the obinutuzumab plus bendamustine group and in 33% in the bendamustine monotherapy group. Adverse events resulted in death in 6% of patients in each group.

The study was funded by Hoffmann-La Roche. Genentech, the maker of obinutuzumab (Gazyva) in the United States, is a wholly owned member of the Roche Group. Dr. Sehn receives honoraria and is a consultant or advisor to Genentech as well as several other drug companies.

[email protected]

On Twitter @maryjodales

References

Author and Disclosure Information

Publications
Topics
Sections
Author and Disclosure Information

Author and Disclosure Information

Obinutuzumab and bendamustine followed by obinutuzumab maintenance therapy was superior to bendamustine monotherapy based on progression-free survival in rituximab-refractory patients with indolent non-Hodgkin lymphoma, based on a study published online in the Lancet Oncology.

After a median follow-up of 22 months in the obinutuzumab plus bendamustine group and 20 months in the bendamustine monotherapy group, progression-free survival was significantly longer with obinutuzumab plus bendamustine (median not reached; 95% confidence interval, 22.5 months – not estimable) than with bendamustine monotherapy (14.9 months, range, 12.8-16.6; hazard ratio, 0.55; 95% CI 0.40-0.74; P = .0001). About two-thirds of the nearly 400 patients in both study arms had grade 3-5 adverse events.

The anti-CD20 monoclonal antibody obinutuzumab is an option when patients with indolent non-Hodgkin lymphoma relapse or don’t achieve adequate disease control with rituximab-based treatment, wrote Laurie H. Sehn, MD, of the British Columbia Cancer Agency and the University of British Columbia, Vancouver, and her colleagues.

In an open-label, randomized, phase III study called GADOLIN, patients with CD20-positive indolent non-Hodgkin lymphoma were stratified by indolent non-Hodgkin lymphoma subtype, rituximab-refractory type, number of previous therapies, and geographic region.

For the study, 194 patients were assigned to obinutuzumab plus bendamustine and 202 to bendamustine monotherapy. Trial participants received six 28-day cycles with either obinutuzumab plus bendamustine (obinutuzumab 1,000 mg on days 1, 8, and 15, cycle 1; and on day 1, cycles 2-6) plus bendamustine (90 mg/m2 per day on days 1 and 2, cycles 1-6) or bendamustine monotherapy (120 mg/m2 per day on days 1 and 2 of all cycles). Patients in the obinutuzumab plus bendamustine group whose disease did not progress received obinutuzumab maintenance therapy of 1,000 mg once every 2 months for up to 2 years.

Grade 3-5 adverse events occurred in 68% of 194 patients in the obinutuzumab plus bendamustine group and in 62% of 198 patients in the bendamustine monotherapy group. Grade 3 or worse neutropenia affected 33% of the obinutuzumab plus bendamustine group and 26% of the bendamustine monotherapy group. Other grade 3 or worse events included thrombocytopenia in 11% and 16%, anemia in 8% and 10%, and infusion-related reactions in 11% and 6%. Serious adverse events occurred in 38% in the obinutuzumab plus bendamustine group and in 33% in the bendamustine monotherapy group. Adverse events resulted in death in 6% of patients in each group.

The study was funded by Hoffmann-La Roche. Genentech, the maker of obinutuzumab (Gazyva) in the United States, is a wholly owned member of the Roche Group. Dr. Sehn receives honoraria and is a consultant or advisor to Genentech as well as several other drug companies.

[email protected]

On Twitter @maryjodales

Obinutuzumab and bendamustine followed by obinutuzumab maintenance therapy was superior to bendamustine monotherapy based on progression-free survival in rituximab-refractory patients with indolent non-Hodgkin lymphoma, based on a study published online in the Lancet Oncology.

After a median follow-up of 22 months in the obinutuzumab plus bendamustine group and 20 months in the bendamustine monotherapy group, progression-free survival was significantly longer with obinutuzumab plus bendamustine (median not reached; 95% confidence interval, 22.5 months – not estimable) than with bendamustine monotherapy (14.9 months, range, 12.8-16.6; hazard ratio, 0.55; 95% CI 0.40-0.74; P = .0001). About two-thirds of the nearly 400 patients in both study arms had grade 3-5 adverse events.

The anti-CD20 monoclonal antibody obinutuzumab is an option when patients with indolent non-Hodgkin lymphoma relapse or don’t achieve adequate disease control with rituximab-based treatment, wrote Laurie H. Sehn, MD, of the British Columbia Cancer Agency and the University of British Columbia, Vancouver, and her colleagues.

In an open-label, randomized, phase III study called GADOLIN, patients with CD20-positive indolent non-Hodgkin lymphoma were stratified by indolent non-Hodgkin lymphoma subtype, rituximab-refractory type, number of previous therapies, and geographic region.

For the study, 194 patients were assigned to obinutuzumab plus bendamustine and 202 to bendamustine monotherapy. Trial participants received six 28-day cycles with either obinutuzumab plus bendamustine (obinutuzumab 1,000 mg on days 1, 8, and 15, cycle 1; and on day 1, cycles 2-6) plus bendamustine (90 mg/m2 per day on days 1 and 2, cycles 1-6) or bendamustine monotherapy (120 mg/m2 per day on days 1 and 2 of all cycles). Patients in the obinutuzumab plus bendamustine group whose disease did not progress received obinutuzumab maintenance therapy of 1,000 mg once every 2 months for up to 2 years.

Grade 3-5 adverse events occurred in 68% of 194 patients in the obinutuzumab plus bendamustine group and in 62% of 198 patients in the bendamustine monotherapy group. Grade 3 or worse neutropenia affected 33% of the obinutuzumab plus bendamustine group and 26% of the bendamustine monotherapy group. Other grade 3 or worse events included thrombocytopenia in 11% and 16%, anemia in 8% and 10%, and infusion-related reactions in 11% and 6%. Serious adverse events occurred in 38% in the obinutuzumab plus bendamustine group and in 33% in the bendamustine monotherapy group. Adverse events resulted in death in 6% of patients in each group.

The study was funded by Hoffmann-La Roche. Genentech, the maker of obinutuzumab (Gazyva) in the United States, is a wholly owned member of the Roche Group. Dr. Sehn receives honoraria and is a consultant or advisor to Genentech as well as several other drug companies.

[email protected]

On Twitter @maryjodales

References

References

Publications
Publications
Topics
Article Type
Display Headline
Adding obinutuzumab to bendamustine boosts progression-free survival in rituximab-refractory indolent non-Hodgkin lymphoma
Display Headline
Adding obinutuzumab to bendamustine boosts progression-free survival in rituximab-refractory indolent non-Hodgkin lymphoma
Sections
Article Source

FROM THE LANCET ONCOLOGY

PURLs Copyright

Inside the Article

Vitals

Key clinical point: Obinutuzumab is an option when patients with indolent non-Hodgkin lymphoma relapse or don’t achieve adequate disease control with rituximab-based treatment.

Major finding: Progression-free survival was significantly longer with obinutuzumab plus bendamustine (median not reached; 95% CI, 22.5 months – not estimable) than with bendamustine monotherapy (14.9 months, range,12.8-16.6 months; hazard ratio, 0.55; 95% CI, 0.40-0.74; P = ·0001).

Data source: An open-label, randomized, phase III study of 396 patients.

Disclosures: The study was funded by Hoffmann-La Roche. Genentech, the maker of obinutuzumab (Gazyva) in the United States, is a wholly owned member of the Roche Group. Dr. Sehn receives honoraria and is a consultant or adviser to Genentech as well as several other drug companies.

LETTER: 6 Tips When Practicing Telemedicine

Article Type
Changed
Fri, 09/14/2018 - 12:03
Display Headline
LETTER: 6 Tips When Practicing Telemedicine

In early 2014, I decided to use the six state licenses I had obtained as a locum tenens physician to start practicing telemedicine. Since then, I have worked with several telemedicine platforms. I have found that telemedicine companies differ dramatically in their overall ease of use for the provider. Here are my top tips for deciding which telemedicine company to work with.

  1. Technology support: Telemedicine is dependent on technology. If it is difficult to get help from tech support, do not credential with the company. Tech support is your lifeline to your patients. Make sure you can get help right away if you are having problems finishing or starting a consult. Companies that send automatic emails saying they will get back to you within 24 hours are the most difficult to work with.
  2. Nursing support: All of the telemedicine companies that I have worked with have amazing nurses, but some are overwhelmed with work. Telemedicine nurses are able to connect to your patients via direct callback numbers in a way that you cannot connect. They are able to call in prescriptions to pharmacies if the platform is down or if the patient put in the wrong pharmacy information. Make sure that the company has a nurse that is able to call you back right away. A few telemedicine companies are understaffed with nurses, and it can take hours for a callback. If the key to telemedicine is volume, this is frustrating to deal with.
  3. Chief complaints: Many telemedicine companies are moving away from making the “chief complaint” visible to providers before choosing to take the consult. For me, this is a big red flag. It can be as simple as, “I have a cold.” I like this because if I see a patient who says, “I have abdominal pain,” I know to triage them first.
  4. Volume: Telemedicine is great for staying connected to outpatient medicine. If you are looking to work on a telemedicine platform for your main source of income, then volume is key. A lot of telemedicine companies will tell you how many calls they get per day; the key question is how many calls they get for the states that you are licensed in and how many providers they have licensed in those states. If you want higher volume, then ask if they will pay for your license in states with higher needs (some will). If you are willing to pay to be licensed in additional states, make sure the volume is high enough to make that extra out-of-pocket cost worth it.
  5. Malpractice coverage: Many companies provide malpractice coverage as part of their credentialing package. If they do not, make sure your malpractice coverage covers you for telemedicine.
  6. Documentation: Documentation during your telemedicine consult is arguably even more important than in an outpatient visit. Everything is on the phone or by video, so make sure, in the subjective area, that you are quoting what the patient is telling you. You are not able to do a physical exam, so your recommendations will be based on what the patient is saying.

Have fun! Telemedicine has been really enjoyable for me. I like being able to have the time to educate my patients about things like antibiotics. I enjoy the technological aspects and understanding all of the different platforms. Telemedicine gives you a unique opportunity to practice your skills from the comfort of your own home. TH


Geeta Arora, MD, locum tenens hospitalist

Issue
The Hospitalist - 2016(06)
Publications
Sections

In early 2014, I decided to use the six state licenses I had obtained as a locum tenens physician to start practicing telemedicine. Since then, I have worked with several telemedicine platforms. I have found that telemedicine companies differ dramatically in their overall ease of use for the provider. Here are my top tips for deciding which telemedicine company to work with.

  1. Technology support: Telemedicine is dependent on technology. If it is difficult to get help from tech support, do not credential with the company. Tech support is your lifeline to your patients. Make sure you can get help right away if you are having problems finishing or starting a consult. Companies that send automatic emails saying they will get back to you within 24 hours are the most difficult to work with.
  2. Nursing support: All of the telemedicine companies that I have worked with have amazing nurses, but some are overwhelmed with work. Telemedicine nurses are able to connect to your patients via direct callback numbers in a way that you cannot connect. They are able to call in prescriptions to pharmacies if the platform is down or if the patient put in the wrong pharmacy information. Make sure that the company has a nurse that is able to call you back right away. A few telemedicine companies are understaffed with nurses, and it can take hours for a callback. If the key to telemedicine is volume, this is frustrating to deal with.
  3. Chief complaints: Many telemedicine companies are moving away from making the “chief complaint” visible to providers before choosing to take the consult. For me, this is a big red flag. It can be as simple as, “I have a cold.” I like this because if I see a patient who says, “I have abdominal pain,” I know to triage them first.
  4. Volume: Telemedicine is great for staying connected to outpatient medicine. If you are looking to work on a telemedicine platform for your main source of income, then volume is key. A lot of telemedicine companies will tell you how many calls they get per day; the key question is how many calls they get for the states that you are licensed in and how many providers they have licensed in those states. If you want higher volume, then ask if they will pay for your license in states with higher needs (some will). If you are willing to pay to be licensed in additional states, make sure the volume is high enough to make that extra out-of-pocket cost worth it.
  5. Malpractice coverage: Many companies provide malpractice coverage as part of their credentialing package. If they do not, make sure your malpractice coverage covers you for telemedicine.
  6. Documentation: Documentation during your telemedicine consult is arguably even more important than in an outpatient visit. Everything is on the phone or by video, so make sure, in the subjective area, that you are quoting what the patient is telling you. You are not able to do a physical exam, so your recommendations will be based on what the patient is saying.

Have fun! Telemedicine has been really enjoyable for me. I like being able to have the time to educate my patients about things like antibiotics. I enjoy the technological aspects and understanding all of the different platforms. Telemedicine gives you a unique opportunity to practice your skills from the comfort of your own home. TH


Geeta Arora, MD, locum tenens hospitalist

In early 2014, I decided to use the six state licenses I had obtained as a locum tenens physician to start practicing telemedicine. Since then, I have worked with several telemedicine platforms. I have found that telemedicine companies differ dramatically in their overall ease of use for the provider. Here are my top tips for deciding which telemedicine company to work with.

  1. Technology support: Telemedicine is dependent on technology. If it is difficult to get help from tech support, do not credential with the company. Tech support is your lifeline to your patients. Make sure you can get help right away if you are having problems finishing or starting a consult. Companies that send automatic emails saying they will get back to you within 24 hours are the most difficult to work with.
  2. Nursing support: All of the telemedicine companies that I have worked with have amazing nurses, but some are overwhelmed with work. Telemedicine nurses are able to connect to your patients via direct callback numbers in a way that you cannot connect. They are able to call in prescriptions to pharmacies if the platform is down or if the patient put in the wrong pharmacy information. Make sure that the company has a nurse that is able to call you back right away. A few telemedicine companies are understaffed with nurses, and it can take hours for a callback. If the key to telemedicine is volume, this is frustrating to deal with.
  3. Chief complaints: Many telemedicine companies are moving away from making the “chief complaint” visible to providers before choosing to take the consult. For me, this is a big red flag. It can be as simple as, “I have a cold.” I like this because if I see a patient who says, “I have abdominal pain,” I know to triage them first.
  4. Volume: Telemedicine is great for staying connected to outpatient medicine. If you are looking to work on a telemedicine platform for your main source of income, then volume is key. A lot of telemedicine companies will tell you how many calls they get per day; the key question is how many calls they get for the states that you are licensed in and how many providers they have licensed in those states. If you want higher volume, then ask if they will pay for your license in states with higher needs (some will). If you are willing to pay to be licensed in additional states, make sure the volume is high enough to make that extra out-of-pocket cost worth it.
  5. Malpractice coverage: Many companies provide malpractice coverage as part of their credentialing package. If they do not, make sure your malpractice coverage covers you for telemedicine.
  6. Documentation: Documentation during your telemedicine consult is arguably even more important than in an outpatient visit. Everything is on the phone or by video, so make sure, in the subjective area, that you are quoting what the patient is telling you. You are not able to do a physical exam, so your recommendations will be based on what the patient is saying.

Have fun! Telemedicine has been really enjoyable for me. I like being able to have the time to educate my patients about things like antibiotics. I enjoy the technological aspects and understanding all of the different platforms. Telemedicine gives you a unique opportunity to practice your skills from the comfort of your own home. TH


Geeta Arora, MD, locum tenens hospitalist

Issue
The Hospitalist - 2016(06)
Issue
The Hospitalist - 2016(06)
Publications
Publications
Article Type
Display Headline
LETTER: 6 Tips When Practicing Telemedicine
Display Headline
LETTER: 6 Tips When Practicing Telemedicine
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)

Team maps chromatin landscape in CLL

Article Type
Changed
Tue, 06/28/2016 - 05:00
Display Headline
Team maps chromatin landscape in CLL

Micrograph showing CLL

Researchers say they have performed the first large-scale analysis of the chromatin landscape in chronic lymphocytic leukemia (CLL).

And, in doing so, they have identified shared gene regulatory networks as well as heterogeneity between patients and CLL subtypes.

The group says this work should enable deeper investigation into chromatin regulation in CLL and the identification of therapeutically relevant mechanisms of disease.

The work has been published in Nature Communications.

The researchers performed chromatin accessibility mapping—via the assay for transposase-accessible chromatin using sequencing (ATAC-seq)—on 88 CLL samples from 55 patients.

For 10 of the samples, the team also established histone profiles using ChIPmentation for 3 histone marks (H3K4me1, H3K27ac, and H3K27me3) and transcriptome profiles using RNA sequencing.

The researchers then developed a bioinformatic method for linking the chromatin profiles to clinical annotations and molecular diagnostics data, and they analyzed gene regulatory networks that underlie the major disease subtypes of CLL.

The work revealed a “shared core” of regulatory regions in CLL patients as well as variations between the samples.

Furthermore, the chromatin profiles and gene regulatory networks accurately predicted IGHV mutation status and pinpointed differences between IGVH-mutated and IGVH-unmutated CLL.

“Our study has been able to dissect the variability that exists in the epigenome of CLL patients and helped to identify disease-specific changes, which will hopefully be informative for distinguishing disease subtypes or identifying suitable treatments,” said study author Jonathan Strefford, PhD, of the University of Southampton in the UK.

“Epigenetics can offer a useful doorway into ways of improving disease diagnosis and more personalized treatment choices for patients.”

Publications
Topics

Micrograph showing CLL

Researchers say they have performed the first large-scale analysis of the chromatin landscape in chronic lymphocytic leukemia (CLL).

And, in doing so, they have identified shared gene regulatory networks as well as heterogeneity between patients and CLL subtypes.

The group says this work should enable deeper investigation into chromatin regulation in CLL and the identification of therapeutically relevant mechanisms of disease.

The work has been published in Nature Communications.

The researchers performed chromatin accessibility mapping—via the assay for transposase-accessible chromatin using sequencing (ATAC-seq)—on 88 CLL samples from 55 patients.

For 10 of the samples, the team also established histone profiles using ChIPmentation for 3 histone marks (H3K4me1, H3K27ac, and H3K27me3) and transcriptome profiles using RNA sequencing.

The researchers then developed a bioinformatic method for linking the chromatin profiles to clinical annotations and molecular diagnostics data, and they analyzed gene regulatory networks that underlie the major disease subtypes of CLL.

The work revealed a “shared core” of regulatory regions in CLL patients as well as variations between the samples.

Furthermore, the chromatin profiles and gene regulatory networks accurately predicted IGHV mutation status and pinpointed differences between IGVH-mutated and IGVH-unmutated CLL.

“Our study has been able to dissect the variability that exists in the epigenome of CLL patients and helped to identify disease-specific changes, which will hopefully be informative for distinguishing disease subtypes or identifying suitable treatments,” said study author Jonathan Strefford, PhD, of the University of Southampton in the UK.

“Epigenetics can offer a useful doorway into ways of improving disease diagnosis and more personalized treatment choices for patients.”

Micrograph showing CLL

Researchers say they have performed the first large-scale analysis of the chromatin landscape in chronic lymphocytic leukemia (CLL).

And, in doing so, they have identified shared gene regulatory networks as well as heterogeneity between patients and CLL subtypes.

The group says this work should enable deeper investigation into chromatin regulation in CLL and the identification of therapeutically relevant mechanisms of disease.

The work has been published in Nature Communications.

The researchers performed chromatin accessibility mapping—via the assay for transposase-accessible chromatin using sequencing (ATAC-seq)—on 88 CLL samples from 55 patients.

For 10 of the samples, the team also established histone profiles using ChIPmentation for 3 histone marks (H3K4me1, H3K27ac, and H3K27me3) and transcriptome profiles using RNA sequencing.

The researchers then developed a bioinformatic method for linking the chromatin profiles to clinical annotations and molecular diagnostics data, and they analyzed gene regulatory networks that underlie the major disease subtypes of CLL.

The work revealed a “shared core” of regulatory regions in CLL patients as well as variations between the samples.

Furthermore, the chromatin profiles and gene regulatory networks accurately predicted IGHV mutation status and pinpointed differences between IGVH-mutated and IGVH-unmutated CLL.

“Our study has been able to dissect the variability that exists in the epigenome of CLL patients and helped to identify disease-specific changes, which will hopefully be informative for distinguishing disease subtypes or identifying suitable treatments,” said study author Jonathan Strefford, PhD, of the University of Southampton in the UK.

“Epigenetics can offer a useful doorway into ways of improving disease diagnosis and more personalized treatment choices for patients.”

Publications
Publications
Topics
Article Type
Display Headline
Team maps chromatin landscape in CLL
Display Headline
Team maps chromatin landscape in CLL
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica

EBV-CTL product classified as ATMP

Article Type
Changed
Tue, 06/28/2016 - 05:00
Display Headline
EBV-CTL product classified as ATMP

An EBV-infected cell (green/red)

among uninfected cells (blue)

Image courtesy of Benjamin

Chaigne-Delalande

A cytotoxic T-lymphocyte product that targets Epstein-Barr virus (EBV-CTLs) has been classified as an advanced therapy medicinal product (ATMP) by the European Medicines Agency (EMA).

The EBV-CTLs are being developed by Atara Biotherapeutics, Inc., to treat patients with EBV post-transplant lymphoproliferative disorder (EBV-PTLD).

ATMP classification was established to regulate cell and gene therapy and tissue-engineered medicinal products, support the development of these products, and provide a benchmark for the level of quality compliance for pharmaceutical practices.

ATMP classification can provide developers with scientific regulatory guidance, help clarify the applicable regulatory framework and development path, and provide access to all relevant services and incentives offered by the EMA. It can also be advantageous when submitting clinical trial dossiers to national regulatory authorities within the European Union.

About EBV-CTLs

Atara Bio’s EBV-CTL product utilizes a technology in which T cells are collected from the blood of third-party donors and then exposed to EBV antigens. The activated T cells are then expanded, characterized, and stored for future use in a partially HLA-matched patient.

In the context of EBV-PTLD, the EBV-CTLs find the cancer cells expressing EBV and kill them.

Atara Bio’s EBV-CTL product is currently being studied in phase 2 trials of patients with EBV-associated cancers, including PTLD and nasopharyngeal carcinoma.

Results of a phase 1/2 study of EBV-CTLs were presented at the APHON 37th Annual Conference and Exhibit and the 2015 ASCO Annual Meeting.

Atara Bio’s EBV-CTL product has orphan designation in the European Union and the US, as well as breakthrough designation in the US.

Publications
Topics

An EBV-infected cell (green/red)

among uninfected cells (blue)

Image courtesy of Benjamin

Chaigne-Delalande

A cytotoxic T-lymphocyte product that targets Epstein-Barr virus (EBV-CTLs) has been classified as an advanced therapy medicinal product (ATMP) by the European Medicines Agency (EMA).

The EBV-CTLs are being developed by Atara Biotherapeutics, Inc., to treat patients with EBV post-transplant lymphoproliferative disorder (EBV-PTLD).

ATMP classification was established to regulate cell and gene therapy and tissue-engineered medicinal products, support the development of these products, and provide a benchmark for the level of quality compliance for pharmaceutical practices.

ATMP classification can provide developers with scientific regulatory guidance, help clarify the applicable regulatory framework and development path, and provide access to all relevant services and incentives offered by the EMA. It can also be advantageous when submitting clinical trial dossiers to national regulatory authorities within the European Union.

About EBV-CTLs

Atara Bio’s EBV-CTL product utilizes a technology in which T cells are collected from the blood of third-party donors and then exposed to EBV antigens. The activated T cells are then expanded, characterized, and stored for future use in a partially HLA-matched patient.

In the context of EBV-PTLD, the EBV-CTLs find the cancer cells expressing EBV and kill them.

Atara Bio’s EBV-CTL product is currently being studied in phase 2 trials of patients with EBV-associated cancers, including PTLD and nasopharyngeal carcinoma.

Results of a phase 1/2 study of EBV-CTLs were presented at the APHON 37th Annual Conference and Exhibit and the 2015 ASCO Annual Meeting.

Atara Bio’s EBV-CTL product has orphan designation in the European Union and the US, as well as breakthrough designation in the US.

An EBV-infected cell (green/red)

among uninfected cells (blue)

Image courtesy of Benjamin

Chaigne-Delalande

A cytotoxic T-lymphocyte product that targets Epstein-Barr virus (EBV-CTLs) has been classified as an advanced therapy medicinal product (ATMP) by the European Medicines Agency (EMA).

The EBV-CTLs are being developed by Atara Biotherapeutics, Inc., to treat patients with EBV post-transplant lymphoproliferative disorder (EBV-PTLD).

ATMP classification was established to regulate cell and gene therapy and tissue-engineered medicinal products, support the development of these products, and provide a benchmark for the level of quality compliance for pharmaceutical practices.

ATMP classification can provide developers with scientific regulatory guidance, help clarify the applicable regulatory framework and development path, and provide access to all relevant services and incentives offered by the EMA. It can also be advantageous when submitting clinical trial dossiers to national regulatory authorities within the European Union.

About EBV-CTLs

Atara Bio’s EBV-CTL product utilizes a technology in which T cells are collected from the blood of third-party donors and then exposed to EBV antigens. The activated T cells are then expanded, characterized, and stored for future use in a partially HLA-matched patient.

In the context of EBV-PTLD, the EBV-CTLs find the cancer cells expressing EBV and kill them.

Atara Bio’s EBV-CTL product is currently being studied in phase 2 trials of patients with EBV-associated cancers, including PTLD and nasopharyngeal carcinoma.

Results of a phase 1/2 study of EBV-CTLs were presented at the APHON 37th Annual Conference and Exhibit and the 2015 ASCO Annual Meeting.

Atara Bio’s EBV-CTL product has orphan designation in the European Union and the US, as well as breakthrough designation in the US.

Publications
Publications
Topics
Article Type
Display Headline
EBV-CTL product classified as ATMP
Display Headline
EBV-CTL product classified as ATMP
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica

P vivax evolving differently in different regions

Article Type
Changed
Tue, 06/28/2016 - 05:00
Display Headline
P vivax evolving differently in different regions

Blood smear showing

Plasmodium vivax

Image by Mae Melvin

Genomic research suggests the malaria parasite Plasmodium vivax is evolving rapidly to adapt to conditions in different geographic locations.

Researchers studied more than 200 parasite samples from across the Asia-Pacific region and found that P vivax has evolved differently in different areas.

The team identified substantial differences in the frequency of copy number variations (CNVs) in samples from western Thailand, western Cambodia, and Papua Indonesia.

They believe this is a result of the different antimalarial drugs used in these regions.

The researchers described this work in Nature Genetics.

“For so long, it’s not been possible to study P vivax genomes in detail, on a large-scale, but now we can, and we’re seeing the effect that drug use has on how parasites are evolving,” said study author Dominic Kwiatkowski, of the Wellcome Trust Sanger Institute in the UK.

He and his colleagues studied the genomes of 228 parasite samples, identifying the strains carried by each patient and revealing their infection history. Most samples came from Southeast Asia (Thailand, Cambodia, Vietnam, Laos, Myanmar, and Malaysia) and Oceania (Papua Indonesia and Papua New Guinea), but the team also studied samples from China, India, Sri Lanka, Brazil, and Madagascar.

The researchers performed detailed population genetic analyses using 148 samples from western Thailand, western Cambodia, and Papua Indonesia. This revealed CNVs in 9 regions of the core genome, and the frequency of the 4 most common CNVs varied greatly according to geographical location.

The first common CNV was a 9-kb deletion on chromosome 8 that includes the first 3 exons of a gene encoding a cytoadherence-linked asexual protein. The CNV was present in 73% of Papua Indonesia samples, 6% of western Cambodia samples, and 3% of western Thailand samples.

The second common CNV was a 7-kb duplication on chromosome 6 that encompasses pvdbp, the gene that encodes the Duffy-binding protein, which mediates P vivax’s invasion of erythrocytes. It was present in 5% of Papua Indonesia samples, 35% of western Cambodia samples, and 25% of western Thailand samples.

The third common CNV was a 37-kb duplication on chromosome 10 that includes pvmdr1, which has been associated with resistance to mefloquine and is homologous to the pfmdr1 amplification responsible for mefloquine resistance in P falciparum. This CNV was only present in samples from western Thailand.

The fourth common CNV was a 3-kb duplication on chromosome 14 that includes the gene PVX_101445. It was found only in Papua Indonesia samples.

“Our study shows that the strongest evidence of evolution is in Papua, Indonesia, where resistance of P vivax to chloroquine is now rampant,” said Ric Price, MD, of the University of Oxford in the UK.

“These data provide crucial information from which we can start to identify the mechanisms of drug resistance in P vivax.”

“We can see in the genome that drug resistance is a huge driver for evolution,” added Richard Pearson, PhD, of the Wellcome Trust Sanger Institute.

“Intriguingly, in some places, this process appears to be happening in response to drugs used primarily to treat a different malaria parasite, P falciparum. Although the exact cause isn’t known, this is a worrying sign that drug resistance is becoming deeply entrenched in the parasite population.”

The researchers said there are a few possible reasons why P vivax may be evolving to evade drugs used against P falciparum.

Many people carry mixed infections of both species of parasite, so, in treating one species, the other automatically gets exposed to the drug. Another culprit may be unsupervised drug use—where many people take the most readily available, rather than the most suitable, antimalarial drug.

 

 

Another finding from this study was that, when the researchers identified patients who were carrying multiple strains of parasite, the genomic data made it possible to determine how closely the different strains were related to one another.

“This means that we can now start to pull apart the genetic complexity of individual Plasmodium vivax infections and work out whether the parasites came from one or more mosquito bites,” Kwiatkowski said. “It provides a way of addressing fundamental questions about how P vivax is transmitted and how it persists within a community and, in particular, about the biology of relapsing infections.”

Publications
Topics

Blood smear showing

Plasmodium vivax

Image by Mae Melvin

Genomic research suggests the malaria parasite Plasmodium vivax is evolving rapidly to adapt to conditions in different geographic locations.

Researchers studied more than 200 parasite samples from across the Asia-Pacific region and found that P vivax has evolved differently in different areas.

The team identified substantial differences in the frequency of copy number variations (CNVs) in samples from western Thailand, western Cambodia, and Papua Indonesia.

They believe this is a result of the different antimalarial drugs used in these regions.

The researchers described this work in Nature Genetics.

“For so long, it’s not been possible to study P vivax genomes in detail, on a large-scale, but now we can, and we’re seeing the effect that drug use has on how parasites are evolving,” said study author Dominic Kwiatkowski, of the Wellcome Trust Sanger Institute in the UK.

He and his colleagues studied the genomes of 228 parasite samples, identifying the strains carried by each patient and revealing their infection history. Most samples came from Southeast Asia (Thailand, Cambodia, Vietnam, Laos, Myanmar, and Malaysia) and Oceania (Papua Indonesia and Papua New Guinea), but the team also studied samples from China, India, Sri Lanka, Brazil, and Madagascar.

The researchers performed detailed population genetic analyses using 148 samples from western Thailand, western Cambodia, and Papua Indonesia. This revealed CNVs in 9 regions of the core genome, and the frequency of the 4 most common CNVs varied greatly according to geographical location.

The first common CNV was a 9-kb deletion on chromosome 8 that includes the first 3 exons of a gene encoding a cytoadherence-linked asexual protein. The CNV was present in 73% of Papua Indonesia samples, 6% of western Cambodia samples, and 3% of western Thailand samples.

The second common CNV was a 7-kb duplication on chromosome 6 that encompasses pvdbp, the gene that encodes the Duffy-binding protein, which mediates P vivax’s invasion of erythrocytes. It was present in 5% of Papua Indonesia samples, 35% of western Cambodia samples, and 25% of western Thailand samples.

The third common CNV was a 37-kb duplication on chromosome 10 that includes pvmdr1, which has been associated with resistance to mefloquine and is homologous to the pfmdr1 amplification responsible for mefloquine resistance in P falciparum. This CNV was only present in samples from western Thailand.

The fourth common CNV was a 3-kb duplication on chromosome 14 that includes the gene PVX_101445. It was found only in Papua Indonesia samples.

“Our study shows that the strongest evidence of evolution is in Papua, Indonesia, where resistance of P vivax to chloroquine is now rampant,” said Ric Price, MD, of the University of Oxford in the UK.

“These data provide crucial information from which we can start to identify the mechanisms of drug resistance in P vivax.”

“We can see in the genome that drug resistance is a huge driver for evolution,” added Richard Pearson, PhD, of the Wellcome Trust Sanger Institute.

“Intriguingly, in some places, this process appears to be happening in response to drugs used primarily to treat a different malaria parasite, P falciparum. Although the exact cause isn’t known, this is a worrying sign that drug resistance is becoming deeply entrenched in the parasite population.”

The researchers said there are a few possible reasons why P vivax may be evolving to evade drugs used against P falciparum.

Many people carry mixed infections of both species of parasite, so, in treating one species, the other automatically gets exposed to the drug. Another culprit may be unsupervised drug use—where many people take the most readily available, rather than the most suitable, antimalarial drug.

 

 

Another finding from this study was that, when the researchers identified patients who were carrying multiple strains of parasite, the genomic data made it possible to determine how closely the different strains were related to one another.

“This means that we can now start to pull apart the genetic complexity of individual Plasmodium vivax infections and work out whether the parasites came from one or more mosquito bites,” Kwiatkowski said. “It provides a way of addressing fundamental questions about how P vivax is transmitted and how it persists within a community and, in particular, about the biology of relapsing infections.”

Blood smear showing

Plasmodium vivax

Image by Mae Melvin

Genomic research suggests the malaria parasite Plasmodium vivax is evolving rapidly to adapt to conditions in different geographic locations.

Researchers studied more than 200 parasite samples from across the Asia-Pacific region and found that P vivax has evolved differently in different areas.

The team identified substantial differences in the frequency of copy number variations (CNVs) in samples from western Thailand, western Cambodia, and Papua Indonesia.

They believe this is a result of the different antimalarial drugs used in these regions.

The researchers described this work in Nature Genetics.

“For so long, it’s not been possible to study P vivax genomes in detail, on a large-scale, but now we can, and we’re seeing the effect that drug use has on how parasites are evolving,” said study author Dominic Kwiatkowski, of the Wellcome Trust Sanger Institute in the UK.

He and his colleagues studied the genomes of 228 parasite samples, identifying the strains carried by each patient and revealing their infection history. Most samples came from Southeast Asia (Thailand, Cambodia, Vietnam, Laos, Myanmar, and Malaysia) and Oceania (Papua Indonesia and Papua New Guinea), but the team also studied samples from China, India, Sri Lanka, Brazil, and Madagascar.

The researchers performed detailed population genetic analyses using 148 samples from western Thailand, western Cambodia, and Papua Indonesia. This revealed CNVs in 9 regions of the core genome, and the frequency of the 4 most common CNVs varied greatly according to geographical location.

The first common CNV was a 9-kb deletion on chromosome 8 that includes the first 3 exons of a gene encoding a cytoadherence-linked asexual protein. The CNV was present in 73% of Papua Indonesia samples, 6% of western Cambodia samples, and 3% of western Thailand samples.

The second common CNV was a 7-kb duplication on chromosome 6 that encompasses pvdbp, the gene that encodes the Duffy-binding protein, which mediates P vivax’s invasion of erythrocytes. It was present in 5% of Papua Indonesia samples, 35% of western Cambodia samples, and 25% of western Thailand samples.

The third common CNV was a 37-kb duplication on chromosome 10 that includes pvmdr1, which has been associated with resistance to mefloquine and is homologous to the pfmdr1 amplification responsible for mefloquine resistance in P falciparum. This CNV was only present in samples from western Thailand.

The fourth common CNV was a 3-kb duplication on chromosome 14 that includes the gene PVX_101445. It was found only in Papua Indonesia samples.

“Our study shows that the strongest evidence of evolution is in Papua, Indonesia, where resistance of P vivax to chloroquine is now rampant,” said Ric Price, MD, of the University of Oxford in the UK.

“These data provide crucial information from which we can start to identify the mechanisms of drug resistance in P vivax.”

“We can see in the genome that drug resistance is a huge driver for evolution,” added Richard Pearson, PhD, of the Wellcome Trust Sanger Institute.

“Intriguingly, in some places, this process appears to be happening in response to drugs used primarily to treat a different malaria parasite, P falciparum. Although the exact cause isn’t known, this is a worrying sign that drug resistance is becoming deeply entrenched in the parasite population.”

The researchers said there are a few possible reasons why P vivax may be evolving to evade drugs used against P falciparum.

Many people carry mixed infections of both species of parasite, so, in treating one species, the other automatically gets exposed to the drug. Another culprit may be unsupervised drug use—where many people take the most readily available, rather than the most suitable, antimalarial drug.

 

 

Another finding from this study was that, when the researchers identified patients who were carrying multiple strains of parasite, the genomic data made it possible to determine how closely the different strains were related to one another.

“This means that we can now start to pull apart the genetic complexity of individual Plasmodium vivax infections and work out whether the parasites came from one or more mosquito bites,” Kwiatkowski said. “It provides a way of addressing fundamental questions about how P vivax is transmitted and how it persists within a community and, in particular, about the biology of relapsing infections.”

Publications
Publications
Topics
Article Type
Display Headline
P vivax evolving differently in different regions
Display Headline
P vivax evolving differently in different regions
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica

Immunotherapy drugs linked to rheumatic diseases

Article Type
Changed
Tue, 06/28/2016 - 05:00
Display Headline
Immunotherapy drugs linked to rheumatic diseases

Vials of drug

Photo by Bill Branson

Several case reports have suggested that cancer patients taking the immunotherapy drugs nivolumab and ipilimumab may have a higher-than-normal risk of developing rheumatic diseases.

Between 2012 and 2016, 13 patients at the Johns Hopkins Kimmel Cancer Center who were taking one or both drugs developed inflammatory arthritis or sicca syndrome, a set of autoimmune conditions causing dry eyes and mouth.

The cases were described in Annals of Rheumatic Diseases.

Nivolumab and ipilimumab are both designed to turn off the molecular “checkpoints” some cancers—including lymphoma—use to evade the immune system. When the drugs work, they allow the immune system to detect and attack tumor cells. However, they also turn up the activity of the immune system as a whole and can therefore trigger immune-related side effects.

Clinical trials of ipilimumab and nivolumab have indicated that the drugs confer an increased risk of inflammatory bowel diseases, lung inflammation, autoimmune thyroid disease, and pituitary gland inflammation.

However, those trials were designed primarily to determine efficacy against cancer and not to fully examine all features of rheumatologic side effects, said Laura C. Cappelli, MD, of the Johns Hopkins University School of Medicine in Baltimore, Maryland.

With this in mind, she and her colleagues decided to take a closer look at 13 adults (older than 18) who were treated at the Johns Hopkins Kimmel Cancer Center and reported rheumatologic symptoms after their treatment with nivolumab and/or ipilimumab.

Eight patients were taking both ipilimumab and nivolumab, and 5 were taking 1 of the 2 drugs. They were receiving the drugs to treat melanoma (n=6), non-small-cell lung cancer (n=5), small-cell lung cancer (n=1), and renal cell carcinoma (n=1).

Nine of the patients developed inflammatory arthritis—4 with synovitis confirmed via imaging and 4 with inflammatory synovial fluid—and the remaining 4 patients were diagnosed with sicca syndrome. Other immune-related adverse events included pneumonitis, colitis, interstitial nephritis, and thyroiditis.

The researchers said this is the largest published case series showing a link between checkpoint inhibitors and rheumatic diseases.

The patients described in this case report make up about 1.3% of all patients treated with the drugs—singly or in combination—at The Johns Hopkins Hospital from 2012 to 2016. However, the researchers believe that rate is likely an underestimation of how common rheumatic diseases are in patients taking immune checkpoint inhibitors.

“We keep having referrals coming in from our oncologists as more patients are treated with these drugs,” said Clifton Bingham, MD, of the Johns Hopkins University School of Medicine.

“In particular, as more patients are treated with combinations of multiple immunotherapies, we expect the rate to go up.”

Dr Cappelli said she wants the case report to raise awareness among patients and clinicians that rheumatologic side effects may occur with checkpoint inhibitors.

“It is important when weighing the risk-benefit ratio of prescribing these drugs,” she said. “And it’s important for people to be on the lookout for symptoms so they can see a rheumatologist early in an effort to prevent or limit joint damage.”

Drs Cappelli and Bingham and their colleagues are planning further collaboration with Johns Hopkins oncologists to better track the incidence of rheumatic disease in patients taking immunotherapy drugs and determine whether any particular characteristics put cancer patients at higher risk of such complications.

Publications
Topics

Vials of drug

Photo by Bill Branson

Several case reports have suggested that cancer patients taking the immunotherapy drugs nivolumab and ipilimumab may have a higher-than-normal risk of developing rheumatic diseases.

Between 2012 and 2016, 13 patients at the Johns Hopkins Kimmel Cancer Center who were taking one or both drugs developed inflammatory arthritis or sicca syndrome, a set of autoimmune conditions causing dry eyes and mouth.

The cases were described in Annals of Rheumatic Diseases.

Nivolumab and ipilimumab are both designed to turn off the molecular “checkpoints” some cancers—including lymphoma—use to evade the immune system. When the drugs work, they allow the immune system to detect and attack tumor cells. However, they also turn up the activity of the immune system as a whole and can therefore trigger immune-related side effects.

Clinical trials of ipilimumab and nivolumab have indicated that the drugs confer an increased risk of inflammatory bowel diseases, lung inflammation, autoimmune thyroid disease, and pituitary gland inflammation.

However, those trials were designed primarily to determine efficacy against cancer and not to fully examine all features of rheumatologic side effects, said Laura C. Cappelli, MD, of the Johns Hopkins University School of Medicine in Baltimore, Maryland.

With this in mind, she and her colleagues decided to take a closer look at 13 adults (older than 18) who were treated at the Johns Hopkins Kimmel Cancer Center and reported rheumatologic symptoms after their treatment with nivolumab and/or ipilimumab.

Eight patients were taking both ipilimumab and nivolumab, and 5 were taking 1 of the 2 drugs. They were receiving the drugs to treat melanoma (n=6), non-small-cell lung cancer (n=5), small-cell lung cancer (n=1), and renal cell carcinoma (n=1).

Nine of the patients developed inflammatory arthritis—4 with synovitis confirmed via imaging and 4 with inflammatory synovial fluid—and the remaining 4 patients were diagnosed with sicca syndrome. Other immune-related adverse events included pneumonitis, colitis, interstitial nephritis, and thyroiditis.

The researchers said this is the largest published case series showing a link between checkpoint inhibitors and rheumatic diseases.

The patients described in this case report make up about 1.3% of all patients treated with the drugs—singly or in combination—at The Johns Hopkins Hospital from 2012 to 2016. However, the researchers believe that rate is likely an underestimation of how common rheumatic diseases are in patients taking immune checkpoint inhibitors.

“We keep having referrals coming in from our oncologists as more patients are treated with these drugs,” said Clifton Bingham, MD, of the Johns Hopkins University School of Medicine.

“In particular, as more patients are treated with combinations of multiple immunotherapies, we expect the rate to go up.”

Dr Cappelli said she wants the case report to raise awareness among patients and clinicians that rheumatologic side effects may occur with checkpoint inhibitors.

“It is important when weighing the risk-benefit ratio of prescribing these drugs,” she said. “And it’s important for people to be on the lookout for symptoms so they can see a rheumatologist early in an effort to prevent or limit joint damage.”

Drs Cappelli and Bingham and their colleagues are planning further collaboration with Johns Hopkins oncologists to better track the incidence of rheumatic disease in patients taking immunotherapy drugs and determine whether any particular characteristics put cancer patients at higher risk of such complications.

Vials of drug

Photo by Bill Branson

Several case reports have suggested that cancer patients taking the immunotherapy drugs nivolumab and ipilimumab may have a higher-than-normal risk of developing rheumatic diseases.

Between 2012 and 2016, 13 patients at the Johns Hopkins Kimmel Cancer Center who were taking one or both drugs developed inflammatory arthritis or sicca syndrome, a set of autoimmune conditions causing dry eyes and mouth.

The cases were described in Annals of Rheumatic Diseases.

Nivolumab and ipilimumab are both designed to turn off the molecular “checkpoints” some cancers—including lymphoma—use to evade the immune system. When the drugs work, they allow the immune system to detect and attack tumor cells. However, they also turn up the activity of the immune system as a whole and can therefore trigger immune-related side effects.

Clinical trials of ipilimumab and nivolumab have indicated that the drugs confer an increased risk of inflammatory bowel diseases, lung inflammation, autoimmune thyroid disease, and pituitary gland inflammation.

However, those trials were designed primarily to determine efficacy against cancer and not to fully examine all features of rheumatologic side effects, said Laura C. Cappelli, MD, of the Johns Hopkins University School of Medicine in Baltimore, Maryland.

With this in mind, she and her colleagues decided to take a closer look at 13 adults (older than 18) who were treated at the Johns Hopkins Kimmel Cancer Center and reported rheumatologic symptoms after their treatment with nivolumab and/or ipilimumab.

Eight patients were taking both ipilimumab and nivolumab, and 5 were taking 1 of the 2 drugs. They were receiving the drugs to treat melanoma (n=6), non-small-cell lung cancer (n=5), small-cell lung cancer (n=1), and renal cell carcinoma (n=1).

Nine of the patients developed inflammatory arthritis—4 with synovitis confirmed via imaging and 4 with inflammatory synovial fluid—and the remaining 4 patients were diagnosed with sicca syndrome. Other immune-related adverse events included pneumonitis, colitis, interstitial nephritis, and thyroiditis.

The researchers said this is the largest published case series showing a link between checkpoint inhibitors and rheumatic diseases.

The patients described in this case report make up about 1.3% of all patients treated with the drugs—singly or in combination—at The Johns Hopkins Hospital from 2012 to 2016. However, the researchers believe that rate is likely an underestimation of how common rheumatic diseases are in patients taking immune checkpoint inhibitors.

“We keep having referrals coming in from our oncologists as more patients are treated with these drugs,” said Clifton Bingham, MD, of the Johns Hopkins University School of Medicine.

“In particular, as more patients are treated with combinations of multiple immunotherapies, we expect the rate to go up.”

Dr Cappelli said she wants the case report to raise awareness among patients and clinicians that rheumatologic side effects may occur with checkpoint inhibitors.

“It is important when weighing the risk-benefit ratio of prescribing these drugs,” she said. “And it’s important for people to be on the lookout for symptoms so they can see a rheumatologist early in an effort to prevent or limit joint damage.”

Drs Cappelli and Bingham and their colleagues are planning further collaboration with Johns Hopkins oncologists to better track the incidence of rheumatic disease in patients taking immunotherapy drugs and determine whether any particular characteristics put cancer patients at higher risk of such complications.

Publications
Publications
Topics
Article Type
Display Headline
Immunotherapy drugs linked to rheumatic diseases
Display Headline
Immunotherapy drugs linked to rheumatic diseases
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica

ICU Transfer Delay and Outcome

Article Type
Changed
Mon, 01/02/2017 - 19:34
Display Headline
Association between intensive care unit transfer delay and hospital mortality: A multicenter investigation

Patients on hospital wards may become critically ill due to worsening of the underlying condition that was the cause of their admission or acquisition of a new hospital‐acquired illness. Once physiologic deterioration occurs, some patients are evaluated and quickly transferred to the intensive care unit (ICU), whereas others are left on the wards until further deterioration occurs. Because many critical illness syndromes benefit from early intervention, such as sepsis and respiratory failure, early transfer to the ICU for treatment may improve patient outcomes, and conversely, delays in ICU transfer may lead to increased mortality and length of stay (LOS) in critically ill ward patients.[1, 2] However, the timeliness of that transfer is dependent on numerous changing variables, such as ICU bed availability, clinician identification of the deterioration, and clinical judgment regarding the appropriate transfer thresholds.[2, 3, 4, 5, 6, 7] As a result, there is a large degree of heterogeneity in the severity of illness of patients at the time of ICU transfer and in patient outcomes.[6, 8]

Previous studies investigating the association between delayed ICU transfer and patient outcomes have typically utilized the time of consultation by the ICU team to denote the onset of critical illness.[5, 6, 9, 10] However, the decision to transfer a patient to the ICU is often subjective, and previous studies have found an alarmingly high rate of errors in diagnosis and management of critically ill ward patients, including the failure to call for help.[2, 11] Therefore, a more objective tool for quantifying critical illness is necessary for determining the onset of critical illness and quantifying the association of transfer delay with patient outcomes.

Early warning scores, which are designed to detect critical illness on the wards, represent objective measures of critical illness that can be easily calculated in ward patients.[12] The aim of this study was to utilize the electronic Cardiac Arrest Risk Triage (eCART) score, a previously published, statistically derived early warning score that utilizes demographic, vital sign, and laboratory data, as an objective measure of critical illness to estimate the effect of delayed ICU transfer on patient outcomes in a large, multicenter database.[13] We chose 6 hours as the cutoff for delay in this study a priori because it is a threshold noted to be an important time period in critical illness syndromes, such as sepsis.[14, 15]

METHODS

All patients admitted to the medical‐surgical wards at 5 hospitals between November 2008 and January 2013 were eligible for inclusion in this observational cohort study. Further details of the hospital populations have been previously described.[13] A waiver of consent was granted by NorthShore University HealthSystem (IRB #EH11‐258) and the University of Chicago Institutional Review Board (IRB #16995A) based on general impracticability and minimal harm. Collection of patient information was designed to comply with the Health Insurance Portability and Accountability Act of 1996 (HIPAA) regulations.

Defining the Onset of Critical Illness

The eCART score, a statistically derived early warning score that is calculated based on patient demographic, vital sign, and laboratory data, was used as an objective measure of critical illness.[13] Score calculation was performed utilizing demographic information from administrative databases and time‐ and location‐stamped vital signs and laboratory results from data warehouses at the respective institutions. In this study, a score was calculated for each time‐stamped point in the entire dataset. Of note, eCART was not used in this population for patient care as this was a retrospective observational study. An eCART score at the 95% specificity cutoff for ICU transfer from the entire dataset defined a ward patient as critically ill, a definition created a priori and before any data analysis was performed.

Defining ICU Transfer Delay and Study Outcomes

The period of time from when a patient first reached this predefined eCART score to ICU transfer was calculated for each patient, up to a maximum of 24 hours. Transfer to the ICU greater than 6 hours after reaching the critical eCART score was defined a priori as a delayed transfer to allow comparisons between patients with nondelayed and delayed transfer. A patient who suffered a ward cardiac arrest with attempted resuscitation was counted as an ICU transfer at the time of arrest. If a patient experienced more than 1 ICU transfer during the admission, then only the first ward to ICU transfer was used. The primary outcome of the study was in‐hospital mortality, and secondary outcomes were ICU mortality and hospital LOS.

Statistical Analysis

Patient characteristics were compared between patients who experienced delayed and nondelayed ICU transfers using t tests, Wilcoxon rank sums, and [2] tests, as appropriate. The association between length of transfer delay and in‐hospital mortality was calculated using logistic regression, with adjustment for age, sex, and surgical status. In a post hoc sensitivity analysis, additional adjustments were made using each patient's first eCART score on the ward, the individual vital signs and laboratory variables from eCART, and whether the ICU transfer was due to a cardiac arrest on the wards. In addition, an interaction term between time to transfer and the initial eCART on the ward was added to determine if the association between delay and mortality varied by baseline severity. The change in eCART score over time was plotted from 12 hours before the time of first reaching the critical value until ICU transfer for those in the delayed and nondelayed groups using restricted cubic splines to compare the trajectories of severity of illness between these 2 groups. In addition, a linear regression model was fit to investigate the association between the eCART slope in the 8 hours prior to the critical eCART value until ICU transfer and the timing of ICU transfer delay. Statistical analyses were performed using Stata version 12.1 (StataCorp, College Station, TX), and all tests of significance used a 2‐sided P<0.05.

RESULTS

A total of 269,999 admissions had documented vital signs on the hospital wards during the study period, including 11,995 patients who were either transferred from the wards to the ICU (n=11,636) or who suffered a cardiac arrest on the wards (n=359) during their initial ward stay. Of these patients, 3789 reached an eCART score at the 95% specificity cutoff (critical eCART score of 60) within 24 hours of transfer. The median time from first critical eCART value to ICU transfer was 5.4 hours (interquartile range (IQR), 214 hours; mean, 8 hours). Compared to patients without delayed ICU transfer, those with delayed transfer were slightly older (median age, 73 [IQR, 6083] years vs 71 [IQR, 5882] years; P=0.002), whereas all other characteristics were similar (Table 1). Table 2 shows comparisons of vital sign and laboratory results for delayed and nondelayed transfers at the time of ICU transfer. As shown, patients with delayed transfer had lower median respiratory rate, blood pressure, heart rate, and hemoglobin, but higher median white blood cell count and creatinine.

Comparisons of Patient Characteristics Among All ICU Transfer Patients and Nondelayed (Within Six Hours) and Delayed Transfers Who Reached the Critical CART Score
Characteristic Transferred Within 6 Hours, n=2,055 Transfer Delayed, n=1,734 P Value
  • NOTE: Data shown are mean (standard deviation) unless otherwise noted; n refers to the number of patients in each group. Abbreviations: eCART, electronic Cardiac Arrest Risk Triage; ICU, intensive care unit; IQR, interquartile range; LOS, length of stay. *For patients who survived to hospital discharge

Age, median (IQR), y 71 (5882) 73 (6083) 0.002
Female sex, n (%) 1,018 (49.5) 847 (48.8) 0.67
Race, n (%) 0.72
Black 467 (22.7) 374 (21.6)
White 1,141 (55.5) 971 (56.0)
Other/unknown 447 (21.8) 389 (22.4)
Surgical patient, n (%) 572 (27.8) 438 (25.2) 0.07
Hospital LOS prior to first critical eCART, median (IQR), d 1.5 (0.33.7) 1.6 (0.43.9) 0.04
Total hospital LOS, median (IQR), d* 11 (719) 13 (821) <0.001
Died during admission, n (%) 503 (24.5) 576 (33.2) <0.001
Comparison of Physiologic Variables at The time of ICU Transfer Between Nondelayed and Delayed ICU Transfers
Transferred Within 6 Hours, n=2,055 Transfer Delayed, n=1,734 P Value
  • NOTE: Abbreviations: Alk phos, alkaline phosphatase; BUN, blood urea nitrogen; Cr, creatinine; eCART, electronic Cardiac Arrest Risk Triage; GFR, glomerular filtration rate; ICU, intensive care unit; IQR, interquartile range; K+, potassium; SGOT, serum glutamic‐oxaloacetic transaminase; SGPT, serum glutamic pyruvic transaminase; WBC, white blood cells.

  • All data are median (IQR) unless otherwise noted.

Respiratory rate, breaths/min 23 (1830) 22 (1828) <0.001
Systolic blood pressure, mm Hg 111 (92134) 109 (92128) 0.002
Diastolic blood pressure, mm Hg 61 (5075) 59 (4971) <0.001
Heart rate, beats/min 106 (88124) 101 (85117) <0.001
Oxygen saturation, median (IQR), % 97 (9499) 97 (9599) 0.15
Temperature, F 98.0 (97.299.1) 98.0 (97.199.0) 0.001
Alert mental status, number of observations (%) 1,749 (85%) 1,431 (83%) <0.001
eCART score at time of ICU transfer 61 (26122) 48 (21121) 0.914
WBC 10.3 (7.514.5) 11.7 (8.117.0) <0.001
Hemoglobin 10.7 (9.312.0) 10.3 (9.111.6) <0.001
Platelet 215 (137275) 195 (120269) 0.017
Sodium 137 (134140) 137 (134141) 0.70
K+ 4.1 (3.84.6) 4.2 (3.84.7) 0.006
Anion Gap 10 (813) 10 (814) <0.001
CO2 24 (2026) 23 (1826) <0.001
BUN 24 (1640) 32 (1853) <0.001
Cr 1.2 (0.92.0) 1.5 (1.02.7) <0.001
GFR 70 (7070) 70 (5170) <0.001
Glucose 123 (106161) 129 (105164) 0.48
Calcium 8.5 (7.98.8) 8.2 (7.78.7) <0.001
SGOT 26 (2635) 26 (2644) 0.001
SGPT 21 (2127) 21 (2033) 0.002
Total bilirubin 0.7 (0.71.0) 0.7 (0.71.3) <0.001
Alk phos 80 (8096) 80 (79111) 0.175
Albumin 3.0 (2.73.0) 3.0 (2.43.0) <0.001

Delayed transfer occurred in 46% of patients (n=1734) and was associated with increased in‐hospital mortality (33.2% vs 24.5%, P<0.001). This relationship was linear, with each 1‐hour increase in transfer delay associated with a 3% increase in the odds of in‐hospital death (P<0.001) (Figure 1). The association between length of transfer delay and hospital mortality remained unchanged after controlling for age, sex, surgical status, initial eCART score on the wards, vital signs, laboratory values, and whether the ICU transfer was due to a cardiac arrest (3% increase per hour, P<0.001). This association did not vary based on the initial eCART score on the wards (P=0.71 for interaction). Additionally, despite having similar median hospital lengths of stay prior to first critical eCART score (1.6 vs 1.5 days, P=0.04), patients experiencing delayed ICU transfer who survived to discharge had a longer median hospital LOS by 2 days compared to those with nondelayed transfer who survived to discharge (median LOS, 13 (821) days vs 11 (719) days, P=0.01). The change in eCART score over time in the 12 hours before first reaching the critical eCART score until ICU transfer is shown in Figure 2 for patients with delayed and nondelayed transfer. As shown, patients transferred within 6 hours had a more rapid rise in eCART score prior to ICU transfer compared to those with a delayed transfer. This difference in trajectories between delayed and nondelayed patients was similar in patients with low (<13), intermediate (1359), and high (60) initial eCART scores on the wards. A regression model investigating the association between eCART slope prior to ICU transfer and time to ICU transfer demonstrated that a steeper slope was significantly associated with a decreased time to ICU transfer (P<0.01).

Figure 1
Association between length of intensive care unit (ICU) transfer delay and hospital mortality. Abbreviations: CI, confidence interval; eCART, electronic Cardiac Arrest Risk Triage.
Figure 2
Change in electronic Cardiac Arrest Risk Triage (eCART) score over time for the 12 hours prior to reaching the critical eCART value until intensive care unit (ICU) transfer for patients with delayed versus nondelayed ICU transfer. Time 0 denotes first critical eCART value.

DISCUSSION

We found that a delay in transfer to the ICU after reaching a predefined objective threshold of critical illness was associated with a significant increase in hospital mortality and hospital LOS. We also discovered a significant association between critical illness trajectory and delays in transfer, suggesting that caregivers may not recognize more subtle trends in critical illness. This work highlights the importance of timely transfer to the ICU for critically ill ward patients, which can be affected by several factors such as ICU bed availability and caregiver recognition and triage decisions. Our findings have significant implications for patient safety on the wards and provide further evidence for implementing early warning scores into practice to aid with clinical decision making.

Our findings of increased mortality with delayed ICU transfer are consistent with previous studies.[1, 5, 9] For example, Young et al. compared ICU mortality between delayed and nondelayed transfers in 91 consecutive patients with noncardiac diagnoses at a community hospital.[1] They also used predefined criteria for critical illness, and found that delayed transfers had a higher ICU mortality than nondelayed patients (41% vs 11%). However, their criteria for critical illness only had a specificity of 13% for predicting ICU transfer, compared to 95% in our study, suggesting that our threshold is more consistent with critical illness. Another study, by Cardoso and colleagues, investigated the impact of delayed ICU admission due to bed shortages on ICU mortality in 401 patients at a university hospital.[9] Of those patients deemed appropriate for transfer to the ICU but who had to wait for a bed to become available, the median wait time for a bed was 18 hours. They found that each hour of waiting was associated with a 1.5% increase in ICU death. A similar study by Robert and colleagues investigated the impact of delayed or refused ICU admission due to a lack of bed availability.[5] Patients deemed too sick (or too well) to benefit from ICU transfer were excluded. Twenty‐eightday and 60‐day mortality were higher in the admitted group compared to those not admitted, although this finding was not statistically significant. In addition, patients later admitted to the ICU once a bed became available (median wait time, 6 hours; n=89) had higher 28‐day mortality than those admitted immediately (adjusted odds ratio, 1.78; P=0.05). Several other studies have investigated the impact of ICU refusal for reasons that included bed shortages, and found increased mortality in those not admitted to the ICU.[16, 17] However, many of these studies included patients deemed too sick or too well to be transferred to the ICU in the group of nonadmitted patients. Our study adds to this literature by utilizing a highly specific objective measure of critical illness and by including all patients on the wards who reached this threshold, rather than only those for whom a consult was requested.

There are several potential explanations for our finding of increased mortality with delayed ICU transfer. First, those with delayed transfer might be different in some way from those transferred immediately. For example, we found that those with delayed transfer were older. The finding that increasing age is associated with a delay in ICU transfer is interesting, and may reflect physiologic differences in older patients compared to younger ones. For example, older patients have a lower maximum heart rate and thus may not develop the same level of vital sign abnormalities that younger patients do, causing them to be inappropriately left on the wards for too long.[18] In addition, patients with delayed transfer had more deranged renal function and lower blood pressure. It is unknown whether these organ dysfunctions would have been prevented by earlier transfer and to what degree they were related to chronic conditions. However, delayed transfer was still associated with increased mortality even after controlling for age, vital sign and laboratory values, and eCART on ward admission. It may also be possible that patients with delayed transfer received early and appropriate treatment on the wards but failed to improve and thus required ICU transfer. We did not have access to orders in this large database, so this theory will need to be investigated in future work. Finally, the most likely explanation for our findings is that earlier identification and treatment improves outcomes of critically ill patients on the wards, which is consistent with the findings of previous studies.[1, 5, 9, 10] Our study demonstrates that early identification of critical illness is crucial, and that delayed treatment can rapidly lead to increased mortality and LOS.

Our comparison of eCART score trajectory showed that patients transferred within 6 hours of onset of critical illness had a more rapid rise in eCART score over the preceding time period, whereas patients who experienced transfer delay showed a slower increase in eCART score. One explanation for this finding is that patients who decompensate more rapidly are in turn more readily recognizable to providers, whereas patients who experience a more insidious clinical deterioration are recognized later in the process, which then leads to a delay in escalation of care. This hypothesis underlines the importance of utilizing an objective marker of illness that is calculated longitudinally and in real time, as opposed to relying upon provider recognition alone. In fact, we have recently demonstrated that eCART is more accurate and identifies patients earlier than standard rapid response team activation.[19]

There are several important implications of our findings. First, it highlights the potential impact that early warning scores, particular those that are evidence based, can have on the outcomes of hospitalized patients. Second, it suggests that it is important to include age in early warning scores. Previous studies have been mixed as to whether the inclusion of age improves detection of outcomes on the wards, although the method of inclusion of age has been variable in terms of its weighting.[20, 21, 22] Our study found that older patients were more likely to be left on the wards longer prior to ICU transfer after becoming critically ill. By incorporating age into early warning scores, both accuracy and early recognition of critical illness may be improved. Finally, our finding that the trends of the eCART score differed among patients who were immediately transferred to the ICU, and who had a delay in their transfer, suggests that adding vital sign trends to early warning scores may further improve their accuracy and ability to serve as clinical decision support tools.

Our study is unique in that we used an objective measure of critical illness and then examined outcomes after patients reached this threshold on the wards. This overcomes the subjectivity of using evaluation by the ICU team or rapid response team as the starting point, as previous studies have shown a failure to call for help when patients become critically ill on the wards.[2, 11, 23] By using the eCART score, which contains commonly collected electronic health record data and can be calculated electronically in real time, we were able to calculate the score for patients on the wards and in the ICU. This allowed us to examine trends in the eCART score over time to find clues as to why some patients are transferred late to the ICU and why these late transfers have worse outcomes than those transferred earlier. Another strength is the large multicenter database used for the analysis, which included an urban tertiary care hospital, suburban teaching hospitals, and a community nonteaching hospital.

Our study has several limitations. First, we utilized just 1 of many potential measures of critical illness and a cutoff that only included one‐third of patients ultimately transferred to the ICU. However, by using the eCART score, we were able to track a patient's physiologic status over time and remove the variability that comes with using subjective definitions of critical illness. Furthermore, we utilized a high‐specificity cutoff for eCART to ensure that transferred patients had significantly deranged physiology and to avoid including planned transfers to the ICU. It is likely that some patients who were critically ill with less deranged physiology that would have benefitted from earlier transfer were excluded from the study. Second, we were unable to determine the cause of physiologic deterioration for patients in our study due to the large number of included patients. In addition, we did not have code status, comorbidities, or reason for ICU admission available in the dataset. It is likely that the impact of delayed transfer varies by the indication for ICU admission and chronic disease burden. It is also possible that controlling for these unmeasured factors could negate the beneficial association seen for earlier ICU admission. However, our finding of such a strong relationship between time to transfer and mortality after controlling for several important variables suggests that early recognition of critical illness is beneficial to many patients on the wards. Third, due to its observational nature, our study cannot estimate the true impact of timely ICU transfer on critically ill ward patient outcomes. Future clinical trials will be needed to determine the impact of electronic early warning scores on patient outcomes.

In conclusion, delayed ICU transfer is associated with significantly increased hospital LOS and mortality. This association highlights the need for ongoing work toward both the implementation of an evidence‐based risk stratification tool as well as development of effective critical care outreach resources for patients decompensating on the wards. Real‐time use of a validated early warning score, such as eCART, could potentially lead to more timely ICU transfer for critically ill patients and reduced rates of preventable in‐hospital death.

Acknowledgements

The authors thank Timothy Holper, Justin Lakeman, and Contessa Hsu for assistance with data extraction and technical support; Poome Chamnankit, MS, CNP, Kelly Bhatia, MSN, ACNP, and Audrey Seitman, MSN, ACNP for performing manual chart review of cardiac arrest patients; and Nicole Twu for administrative support.

Disclosures: This research was funded in part by an institutional Clinical and Translational Science Award grant (UL1 RR024999, PI: Dr. Julian Solway). Dr. Churpek is supported by a career development award from the National Heart, Lung, and Blood Institute (K08 HL121080). Drs. Churpek and Edelson have a patent pending (ARCD. P0535US.P2) for risk stratification algorithms for hospitalized patients. In addition, Dr. Edelson has received research support from Philips Healthcare (Andover, MA), the American Heart Association (Dallas, TX), and Laerdal Medical (Stavanger, Norway). She has ownership interest in Quant HC (Chicago, IL), which is developing products for risk stratification of hospitalized patients. Drs. Churpek and Wendlandt had full access to all the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis. Preliminary versions of these data were presented at the 2015 meeting of the Society of Hospital Medicine (March 31, 2015, National Harbor, MD).

Files
References
  1. Young MP, Gooder VJ, McBride K, James B, Fisher ES. Inpatient transfers to the intensive care unit: delays are associated with increased mortality and morbidity. J Gen Intern Med. 2003;18(2):7783.
  2. McQuillan P, Pilkington S, Allan A, et al. Confidential inquiry into quality of care before admission to intensive care. BMJ. 1998;316(7148):18531858.
  3. Town JA, Churpek MM, Yuen TC, Huber MT, Kress JP, Edelson DP. Relationship between ICU bed availability, ICU readmission, and cardiac arrest in the general wards. Crit Care Med. 2014;42(9):20372041.
  4. Simchen E, Sprung CL, Galai N, et al. Survival of critically ill patients hospitalized in and out of intensive care units under paucity of intensive care unit beds. Crit Care Med. 2004;32(8):16541661.
  5. Robert R, Reignier J, Tournoux‐Facon C, et al. Refusal of intensive care unit admission due to a full unit: impact on mortality. Am J Respir Crit Care Med. 2012;185(10):10811087.
  6. Sprung CL, Geber D, Eidelman LA, et al. Evaluation of triage decisions for intensive care admission. Crit Care Med. 1999;27(6):10731079.
  7. Garrouste‐Orgeas M, Montuclard L, Timsit JF, et al. Predictors of intensive care unit refusal in French intensive care units: a multiple‐center study. Crit Care Med. 2005;33(4):750755.
  8. Zimmerman JE, Kramer AA, McNair DS, Malila FM. Acute Physiology and Chronic Health Evaluation (APACHE) IV: hospital mortality assessment for today's critically ill patients. Crit Care Med. 2006;34(5):12971310.
  9. Cardoso LT, Grion CM, Matsuo T, et al. Impact of delayed admission to intensive care units on mortality of critically ill patients: a cohort study. Crit Care. 2011;15(1):R28.
  10. Iapichino G, Corbella D, Minelli C, et al. Reasons for refusal of admission to intensive care and impact on mortality. Intensive Care Med. 2010;36(10):17721779.
  11. Hodgetts TJ, Kenward G, Vlackonikolis I, et al. Incidence, location and reasons for avoidable in‐hospital cardiac arrest in a district general hospital. Resuscitation. 2002;54(2):115123.
  12. Churpek MM, Yuen TC, Edelson DP. Risk stratification of hospitalized patients on the wards. Chest. 2013;143(6):17581765.
  13. Churpek MM, Yuen TC, Winslow C, et al. Multicenter development and validation of a risk stratification tool for ward patients. Am J Respir Crit Care Med. 2014;190(6):649655.
  14. Rivers E, Nguyen B, Havstad S, et al. Early goal‐directed therapy in the treatment of severe sepsis and septic shock. N Engl J Med. 2001;345(19):13681377.
  15. Dellinger RP, Levy MM, Rhodes A, et al. Surviving sepsis campaign: international guidelines for management of severe sepsis and septic shock: 2012. Crit Care Med. 2013;41(2):580637.
  16. Vanhecke TE, Gandhi M, McCullough PA, et al. Outcomes of patients considered for, but not admitted to, the intensive care unit. Crit Care Med. 2008;36(3):812817.
  17. Metcalfe MA, Sloggett A, McPherson K. Mortality among appropriately referred patients refused admission to intensive‐care units. Lancet. 1997;350(9070):711.
  18. Churpek MM, Yuen TC, Winslow C, Hall J, Edelson DP. Differences in vital signs between elderly and nonelderly patients prior to ward cardiac arrest. Crit Care Med. 2015;43(4):816822.
  19. Kang MA, Churpek MM, Zadravecz FJ, Adhikari R, Twu NM, Edelson DP. Real‐time risk prediction on the wards: a feasibility study [published April 13, 2016]. Crit Care Med. doi: 10.1097/CCM.0000000000001716.
  20. Smith GB, Prytherch DR, Schmidt PE, et al. Should age be included as a component of track and trigger systems used to identify sick adult patients? Resuscitation. 2008;78(2):109115.
  21. Duckitt RW, Buxton‐Thomas R, Walker J, et al. Worthing physiological scoring system: derivation and validation of a physiological early‐warning system for medical admissions. An observational, population‐based single‐centre study. Br J Anaesth. 2007;98(6):769774.
  22. Subbe CP, Kruger M, Rutherford P, Gemmel L. Validation of a modified Early Warning Score in medical admissions. QJM. 2001;94(10):521526.
  23. Hillman K, Chen J, Cretikos M, et al. Introduction of the medical emergency team (MET) system: a cluster‐randomised controlled trial. Lancet. 2005;365(9477):20912097.
Article PDF
Issue
Journal of Hospital Medicine - 11(11)
Page Number
757-762
Sections
Files
Files
Article PDF
Article PDF

Patients on hospital wards may become critically ill due to worsening of the underlying condition that was the cause of their admission or acquisition of a new hospital‐acquired illness. Once physiologic deterioration occurs, some patients are evaluated and quickly transferred to the intensive care unit (ICU), whereas others are left on the wards until further deterioration occurs. Because many critical illness syndromes benefit from early intervention, such as sepsis and respiratory failure, early transfer to the ICU for treatment may improve patient outcomes, and conversely, delays in ICU transfer may lead to increased mortality and length of stay (LOS) in critically ill ward patients.[1, 2] However, the timeliness of that transfer is dependent on numerous changing variables, such as ICU bed availability, clinician identification of the deterioration, and clinical judgment regarding the appropriate transfer thresholds.[2, 3, 4, 5, 6, 7] As a result, there is a large degree of heterogeneity in the severity of illness of patients at the time of ICU transfer and in patient outcomes.[6, 8]

Previous studies investigating the association between delayed ICU transfer and patient outcomes have typically utilized the time of consultation by the ICU team to denote the onset of critical illness.[5, 6, 9, 10] However, the decision to transfer a patient to the ICU is often subjective, and previous studies have found an alarmingly high rate of errors in diagnosis and management of critically ill ward patients, including the failure to call for help.[2, 11] Therefore, a more objective tool for quantifying critical illness is necessary for determining the onset of critical illness and quantifying the association of transfer delay with patient outcomes.

Early warning scores, which are designed to detect critical illness on the wards, represent objective measures of critical illness that can be easily calculated in ward patients.[12] The aim of this study was to utilize the electronic Cardiac Arrest Risk Triage (eCART) score, a previously published, statistically derived early warning score that utilizes demographic, vital sign, and laboratory data, as an objective measure of critical illness to estimate the effect of delayed ICU transfer on patient outcomes in a large, multicenter database.[13] We chose 6 hours as the cutoff for delay in this study a priori because it is a threshold noted to be an important time period in critical illness syndromes, such as sepsis.[14, 15]

METHODS

All patients admitted to the medical‐surgical wards at 5 hospitals between November 2008 and January 2013 were eligible for inclusion in this observational cohort study. Further details of the hospital populations have been previously described.[13] A waiver of consent was granted by NorthShore University HealthSystem (IRB #EH11‐258) and the University of Chicago Institutional Review Board (IRB #16995A) based on general impracticability and minimal harm. Collection of patient information was designed to comply with the Health Insurance Portability and Accountability Act of 1996 (HIPAA) regulations.

Defining the Onset of Critical Illness

The eCART score, a statistically derived early warning score that is calculated based on patient demographic, vital sign, and laboratory data, was used as an objective measure of critical illness.[13] Score calculation was performed utilizing demographic information from administrative databases and time‐ and location‐stamped vital signs and laboratory results from data warehouses at the respective institutions. In this study, a score was calculated for each time‐stamped point in the entire dataset. Of note, eCART was not used in this population for patient care as this was a retrospective observational study. An eCART score at the 95% specificity cutoff for ICU transfer from the entire dataset defined a ward patient as critically ill, a definition created a priori and before any data analysis was performed.

Defining ICU Transfer Delay and Study Outcomes

The period of time from when a patient first reached this predefined eCART score to ICU transfer was calculated for each patient, up to a maximum of 24 hours. Transfer to the ICU greater than 6 hours after reaching the critical eCART score was defined a priori as a delayed transfer to allow comparisons between patients with nondelayed and delayed transfer. A patient who suffered a ward cardiac arrest with attempted resuscitation was counted as an ICU transfer at the time of arrest. If a patient experienced more than 1 ICU transfer during the admission, then only the first ward to ICU transfer was used. The primary outcome of the study was in‐hospital mortality, and secondary outcomes were ICU mortality and hospital LOS.

Statistical Analysis

Patient characteristics were compared between patients who experienced delayed and nondelayed ICU transfers using t tests, Wilcoxon rank sums, and [2] tests, as appropriate. The association between length of transfer delay and in‐hospital mortality was calculated using logistic regression, with adjustment for age, sex, and surgical status. In a post hoc sensitivity analysis, additional adjustments were made using each patient's first eCART score on the ward, the individual vital signs and laboratory variables from eCART, and whether the ICU transfer was due to a cardiac arrest on the wards. In addition, an interaction term between time to transfer and the initial eCART on the ward was added to determine if the association between delay and mortality varied by baseline severity. The change in eCART score over time was plotted from 12 hours before the time of first reaching the critical value until ICU transfer for those in the delayed and nondelayed groups using restricted cubic splines to compare the trajectories of severity of illness between these 2 groups. In addition, a linear regression model was fit to investigate the association between the eCART slope in the 8 hours prior to the critical eCART value until ICU transfer and the timing of ICU transfer delay. Statistical analyses were performed using Stata version 12.1 (StataCorp, College Station, TX), and all tests of significance used a 2‐sided P<0.05.

RESULTS

A total of 269,999 admissions had documented vital signs on the hospital wards during the study period, including 11,995 patients who were either transferred from the wards to the ICU (n=11,636) or who suffered a cardiac arrest on the wards (n=359) during their initial ward stay. Of these patients, 3789 reached an eCART score at the 95% specificity cutoff (critical eCART score of 60) within 24 hours of transfer. The median time from first critical eCART value to ICU transfer was 5.4 hours (interquartile range (IQR), 214 hours; mean, 8 hours). Compared to patients without delayed ICU transfer, those with delayed transfer were slightly older (median age, 73 [IQR, 6083] years vs 71 [IQR, 5882] years; P=0.002), whereas all other characteristics were similar (Table 1). Table 2 shows comparisons of vital sign and laboratory results for delayed and nondelayed transfers at the time of ICU transfer. As shown, patients with delayed transfer had lower median respiratory rate, blood pressure, heart rate, and hemoglobin, but higher median white blood cell count and creatinine.

Comparisons of Patient Characteristics Among All ICU Transfer Patients and Nondelayed (Within Six Hours) and Delayed Transfers Who Reached the Critical CART Score
Characteristic Transferred Within 6 Hours, n=2,055 Transfer Delayed, n=1,734 P Value
  • NOTE: Data shown are mean (standard deviation) unless otherwise noted; n refers to the number of patients in each group. Abbreviations: eCART, electronic Cardiac Arrest Risk Triage; ICU, intensive care unit; IQR, interquartile range; LOS, length of stay. *For patients who survived to hospital discharge

Age, median (IQR), y 71 (5882) 73 (6083) 0.002
Female sex, n (%) 1,018 (49.5) 847 (48.8) 0.67
Race, n (%) 0.72
Black 467 (22.7) 374 (21.6)
White 1,141 (55.5) 971 (56.0)
Other/unknown 447 (21.8) 389 (22.4)
Surgical patient, n (%) 572 (27.8) 438 (25.2) 0.07
Hospital LOS prior to first critical eCART, median (IQR), d 1.5 (0.33.7) 1.6 (0.43.9) 0.04
Total hospital LOS, median (IQR), d* 11 (719) 13 (821) <0.001
Died during admission, n (%) 503 (24.5) 576 (33.2) <0.001
Comparison of Physiologic Variables at The time of ICU Transfer Between Nondelayed and Delayed ICU Transfers
Transferred Within 6 Hours, n=2,055 Transfer Delayed, n=1,734 P Value
  • NOTE: Abbreviations: Alk phos, alkaline phosphatase; BUN, blood urea nitrogen; Cr, creatinine; eCART, electronic Cardiac Arrest Risk Triage; GFR, glomerular filtration rate; ICU, intensive care unit; IQR, interquartile range; K+, potassium; SGOT, serum glutamic‐oxaloacetic transaminase; SGPT, serum glutamic pyruvic transaminase; WBC, white blood cells.

  • All data are median (IQR) unless otherwise noted.

Respiratory rate, breaths/min 23 (1830) 22 (1828) <0.001
Systolic blood pressure, mm Hg 111 (92134) 109 (92128) 0.002
Diastolic blood pressure, mm Hg 61 (5075) 59 (4971) <0.001
Heart rate, beats/min 106 (88124) 101 (85117) <0.001
Oxygen saturation, median (IQR), % 97 (9499) 97 (9599) 0.15
Temperature, F 98.0 (97.299.1) 98.0 (97.199.0) 0.001
Alert mental status, number of observations (%) 1,749 (85%) 1,431 (83%) <0.001
eCART score at time of ICU transfer 61 (26122) 48 (21121) 0.914
WBC 10.3 (7.514.5) 11.7 (8.117.0) <0.001
Hemoglobin 10.7 (9.312.0) 10.3 (9.111.6) <0.001
Platelet 215 (137275) 195 (120269) 0.017
Sodium 137 (134140) 137 (134141) 0.70
K+ 4.1 (3.84.6) 4.2 (3.84.7) 0.006
Anion Gap 10 (813) 10 (814) <0.001
CO2 24 (2026) 23 (1826) <0.001
BUN 24 (1640) 32 (1853) <0.001
Cr 1.2 (0.92.0) 1.5 (1.02.7) <0.001
GFR 70 (7070) 70 (5170) <0.001
Glucose 123 (106161) 129 (105164) 0.48
Calcium 8.5 (7.98.8) 8.2 (7.78.7) <0.001
SGOT 26 (2635) 26 (2644) 0.001
SGPT 21 (2127) 21 (2033) 0.002
Total bilirubin 0.7 (0.71.0) 0.7 (0.71.3) <0.001
Alk phos 80 (8096) 80 (79111) 0.175
Albumin 3.0 (2.73.0) 3.0 (2.43.0) <0.001

Delayed transfer occurred in 46% of patients (n=1734) and was associated with increased in‐hospital mortality (33.2% vs 24.5%, P<0.001). This relationship was linear, with each 1‐hour increase in transfer delay associated with a 3% increase in the odds of in‐hospital death (P<0.001) (Figure 1). The association between length of transfer delay and hospital mortality remained unchanged after controlling for age, sex, surgical status, initial eCART score on the wards, vital signs, laboratory values, and whether the ICU transfer was due to a cardiac arrest (3% increase per hour, P<0.001). This association did not vary based on the initial eCART score on the wards (P=0.71 for interaction). Additionally, despite having similar median hospital lengths of stay prior to first critical eCART score (1.6 vs 1.5 days, P=0.04), patients experiencing delayed ICU transfer who survived to discharge had a longer median hospital LOS by 2 days compared to those with nondelayed transfer who survived to discharge (median LOS, 13 (821) days vs 11 (719) days, P=0.01). The change in eCART score over time in the 12 hours before first reaching the critical eCART score until ICU transfer is shown in Figure 2 for patients with delayed and nondelayed transfer. As shown, patients transferred within 6 hours had a more rapid rise in eCART score prior to ICU transfer compared to those with a delayed transfer. This difference in trajectories between delayed and nondelayed patients was similar in patients with low (<13), intermediate (1359), and high (60) initial eCART scores on the wards. A regression model investigating the association between eCART slope prior to ICU transfer and time to ICU transfer demonstrated that a steeper slope was significantly associated with a decreased time to ICU transfer (P<0.01).

Figure 1
Association between length of intensive care unit (ICU) transfer delay and hospital mortality. Abbreviations: CI, confidence interval; eCART, electronic Cardiac Arrest Risk Triage.
Figure 2
Change in electronic Cardiac Arrest Risk Triage (eCART) score over time for the 12 hours prior to reaching the critical eCART value until intensive care unit (ICU) transfer for patients with delayed versus nondelayed ICU transfer. Time 0 denotes first critical eCART value.

DISCUSSION

We found that a delay in transfer to the ICU after reaching a predefined objective threshold of critical illness was associated with a significant increase in hospital mortality and hospital LOS. We also discovered a significant association between critical illness trajectory and delays in transfer, suggesting that caregivers may not recognize more subtle trends in critical illness. This work highlights the importance of timely transfer to the ICU for critically ill ward patients, which can be affected by several factors such as ICU bed availability and caregiver recognition and triage decisions. Our findings have significant implications for patient safety on the wards and provide further evidence for implementing early warning scores into practice to aid with clinical decision making.

Our findings of increased mortality with delayed ICU transfer are consistent with previous studies.[1, 5, 9] For example, Young et al. compared ICU mortality between delayed and nondelayed transfers in 91 consecutive patients with noncardiac diagnoses at a community hospital.[1] They also used predefined criteria for critical illness, and found that delayed transfers had a higher ICU mortality than nondelayed patients (41% vs 11%). However, their criteria for critical illness only had a specificity of 13% for predicting ICU transfer, compared to 95% in our study, suggesting that our threshold is more consistent with critical illness. Another study, by Cardoso and colleagues, investigated the impact of delayed ICU admission due to bed shortages on ICU mortality in 401 patients at a university hospital.[9] Of those patients deemed appropriate for transfer to the ICU but who had to wait for a bed to become available, the median wait time for a bed was 18 hours. They found that each hour of waiting was associated with a 1.5% increase in ICU death. A similar study by Robert and colleagues investigated the impact of delayed or refused ICU admission due to a lack of bed availability.[5] Patients deemed too sick (or too well) to benefit from ICU transfer were excluded. Twenty‐eightday and 60‐day mortality were higher in the admitted group compared to those not admitted, although this finding was not statistically significant. In addition, patients later admitted to the ICU once a bed became available (median wait time, 6 hours; n=89) had higher 28‐day mortality than those admitted immediately (adjusted odds ratio, 1.78; P=0.05). Several other studies have investigated the impact of ICU refusal for reasons that included bed shortages, and found increased mortality in those not admitted to the ICU.[16, 17] However, many of these studies included patients deemed too sick or too well to be transferred to the ICU in the group of nonadmitted patients. Our study adds to this literature by utilizing a highly specific objective measure of critical illness and by including all patients on the wards who reached this threshold, rather than only those for whom a consult was requested.

There are several potential explanations for our finding of increased mortality with delayed ICU transfer. First, those with delayed transfer might be different in some way from those transferred immediately. For example, we found that those with delayed transfer were older. The finding that increasing age is associated with a delay in ICU transfer is interesting, and may reflect physiologic differences in older patients compared to younger ones. For example, older patients have a lower maximum heart rate and thus may not develop the same level of vital sign abnormalities that younger patients do, causing them to be inappropriately left on the wards for too long.[18] In addition, patients with delayed transfer had more deranged renal function and lower blood pressure. It is unknown whether these organ dysfunctions would have been prevented by earlier transfer and to what degree they were related to chronic conditions. However, delayed transfer was still associated with increased mortality even after controlling for age, vital sign and laboratory values, and eCART on ward admission. It may also be possible that patients with delayed transfer received early and appropriate treatment on the wards but failed to improve and thus required ICU transfer. We did not have access to orders in this large database, so this theory will need to be investigated in future work. Finally, the most likely explanation for our findings is that earlier identification and treatment improves outcomes of critically ill patients on the wards, which is consistent with the findings of previous studies.[1, 5, 9, 10] Our study demonstrates that early identification of critical illness is crucial, and that delayed treatment can rapidly lead to increased mortality and LOS.

Our comparison of eCART score trajectory showed that patients transferred within 6 hours of onset of critical illness had a more rapid rise in eCART score over the preceding time period, whereas patients who experienced transfer delay showed a slower increase in eCART score. One explanation for this finding is that patients who decompensate more rapidly are in turn more readily recognizable to providers, whereas patients who experience a more insidious clinical deterioration are recognized later in the process, which then leads to a delay in escalation of care. This hypothesis underlines the importance of utilizing an objective marker of illness that is calculated longitudinally and in real time, as opposed to relying upon provider recognition alone. In fact, we have recently demonstrated that eCART is more accurate and identifies patients earlier than standard rapid response team activation.[19]

There are several important implications of our findings. First, it highlights the potential impact that early warning scores, particular those that are evidence based, can have on the outcomes of hospitalized patients. Second, it suggests that it is important to include age in early warning scores. Previous studies have been mixed as to whether the inclusion of age improves detection of outcomes on the wards, although the method of inclusion of age has been variable in terms of its weighting.[20, 21, 22] Our study found that older patients were more likely to be left on the wards longer prior to ICU transfer after becoming critically ill. By incorporating age into early warning scores, both accuracy and early recognition of critical illness may be improved. Finally, our finding that the trends of the eCART score differed among patients who were immediately transferred to the ICU, and who had a delay in their transfer, suggests that adding vital sign trends to early warning scores may further improve their accuracy and ability to serve as clinical decision support tools.

Our study is unique in that we used an objective measure of critical illness and then examined outcomes after patients reached this threshold on the wards. This overcomes the subjectivity of using evaluation by the ICU team or rapid response team as the starting point, as previous studies have shown a failure to call for help when patients become critically ill on the wards.[2, 11, 23] By using the eCART score, which contains commonly collected electronic health record data and can be calculated electronically in real time, we were able to calculate the score for patients on the wards and in the ICU. This allowed us to examine trends in the eCART score over time to find clues as to why some patients are transferred late to the ICU and why these late transfers have worse outcomes than those transferred earlier. Another strength is the large multicenter database used for the analysis, which included an urban tertiary care hospital, suburban teaching hospitals, and a community nonteaching hospital.

Our study has several limitations. First, we utilized just 1 of many potential measures of critical illness and a cutoff that only included one‐third of patients ultimately transferred to the ICU. However, by using the eCART score, we were able to track a patient's physiologic status over time and remove the variability that comes with using subjective definitions of critical illness. Furthermore, we utilized a high‐specificity cutoff for eCART to ensure that transferred patients had significantly deranged physiology and to avoid including planned transfers to the ICU. It is likely that some patients who were critically ill with less deranged physiology that would have benefitted from earlier transfer were excluded from the study. Second, we were unable to determine the cause of physiologic deterioration for patients in our study due to the large number of included patients. In addition, we did not have code status, comorbidities, or reason for ICU admission available in the dataset. It is likely that the impact of delayed transfer varies by the indication for ICU admission and chronic disease burden. It is also possible that controlling for these unmeasured factors could negate the beneficial association seen for earlier ICU admission. However, our finding of such a strong relationship between time to transfer and mortality after controlling for several important variables suggests that early recognition of critical illness is beneficial to many patients on the wards. Third, due to its observational nature, our study cannot estimate the true impact of timely ICU transfer on critically ill ward patient outcomes. Future clinical trials will be needed to determine the impact of electronic early warning scores on patient outcomes.

In conclusion, delayed ICU transfer is associated with significantly increased hospital LOS and mortality. This association highlights the need for ongoing work toward both the implementation of an evidence‐based risk stratification tool as well as development of effective critical care outreach resources for patients decompensating on the wards. Real‐time use of a validated early warning score, such as eCART, could potentially lead to more timely ICU transfer for critically ill patients and reduced rates of preventable in‐hospital death.

Acknowledgements

The authors thank Timothy Holper, Justin Lakeman, and Contessa Hsu for assistance with data extraction and technical support; Poome Chamnankit, MS, CNP, Kelly Bhatia, MSN, ACNP, and Audrey Seitman, MSN, ACNP for performing manual chart review of cardiac arrest patients; and Nicole Twu for administrative support.

Disclosures: This research was funded in part by an institutional Clinical and Translational Science Award grant (UL1 RR024999, PI: Dr. Julian Solway). Dr. Churpek is supported by a career development award from the National Heart, Lung, and Blood Institute (K08 HL121080). Drs. Churpek and Edelson have a patent pending (ARCD. P0535US.P2) for risk stratification algorithms for hospitalized patients. In addition, Dr. Edelson has received research support from Philips Healthcare (Andover, MA), the American Heart Association (Dallas, TX), and Laerdal Medical (Stavanger, Norway). She has ownership interest in Quant HC (Chicago, IL), which is developing products for risk stratification of hospitalized patients. Drs. Churpek and Wendlandt had full access to all the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis. Preliminary versions of these data were presented at the 2015 meeting of the Society of Hospital Medicine (March 31, 2015, National Harbor, MD).

Patients on hospital wards may become critically ill due to worsening of the underlying condition that was the cause of their admission or acquisition of a new hospital‐acquired illness. Once physiologic deterioration occurs, some patients are evaluated and quickly transferred to the intensive care unit (ICU), whereas others are left on the wards until further deterioration occurs. Because many critical illness syndromes benefit from early intervention, such as sepsis and respiratory failure, early transfer to the ICU for treatment may improve patient outcomes, and conversely, delays in ICU transfer may lead to increased mortality and length of stay (LOS) in critically ill ward patients.[1, 2] However, the timeliness of that transfer is dependent on numerous changing variables, such as ICU bed availability, clinician identification of the deterioration, and clinical judgment regarding the appropriate transfer thresholds.[2, 3, 4, 5, 6, 7] As a result, there is a large degree of heterogeneity in the severity of illness of patients at the time of ICU transfer and in patient outcomes.[6, 8]

Previous studies investigating the association between delayed ICU transfer and patient outcomes have typically utilized the time of consultation by the ICU team to denote the onset of critical illness.[5, 6, 9, 10] However, the decision to transfer a patient to the ICU is often subjective, and previous studies have found an alarmingly high rate of errors in diagnosis and management of critically ill ward patients, including the failure to call for help.[2, 11] Therefore, a more objective tool for quantifying critical illness is necessary for determining the onset of critical illness and quantifying the association of transfer delay with patient outcomes.

Early warning scores, which are designed to detect critical illness on the wards, represent objective measures of critical illness that can be easily calculated in ward patients.[12] The aim of this study was to utilize the electronic Cardiac Arrest Risk Triage (eCART) score, a previously published, statistically derived early warning score that utilizes demographic, vital sign, and laboratory data, as an objective measure of critical illness to estimate the effect of delayed ICU transfer on patient outcomes in a large, multicenter database.[13] We chose 6 hours as the cutoff for delay in this study a priori because it is a threshold noted to be an important time period in critical illness syndromes, such as sepsis.[14, 15]

METHODS

All patients admitted to the medical‐surgical wards at 5 hospitals between November 2008 and January 2013 were eligible for inclusion in this observational cohort study. Further details of the hospital populations have been previously described.[13] A waiver of consent was granted by NorthShore University HealthSystem (IRB #EH11‐258) and the University of Chicago Institutional Review Board (IRB #16995A) based on general impracticability and minimal harm. Collection of patient information was designed to comply with the Health Insurance Portability and Accountability Act of 1996 (HIPAA) regulations.

Defining the Onset of Critical Illness

The eCART score, a statistically derived early warning score that is calculated based on patient demographic, vital sign, and laboratory data, was used as an objective measure of critical illness.[13] Score calculation was performed utilizing demographic information from administrative databases and time‐ and location‐stamped vital signs and laboratory results from data warehouses at the respective institutions. In this study, a score was calculated for each time‐stamped point in the entire dataset. Of note, eCART was not used in this population for patient care as this was a retrospective observational study. An eCART score at the 95% specificity cutoff for ICU transfer from the entire dataset defined a ward patient as critically ill, a definition created a priori and before any data analysis was performed.

Defining ICU Transfer Delay and Study Outcomes

The period of time from when a patient first reached this predefined eCART score to ICU transfer was calculated for each patient, up to a maximum of 24 hours. Transfer to the ICU greater than 6 hours after reaching the critical eCART score was defined a priori as a delayed transfer to allow comparisons between patients with nondelayed and delayed transfer. A patient who suffered a ward cardiac arrest with attempted resuscitation was counted as an ICU transfer at the time of arrest. If a patient experienced more than 1 ICU transfer during the admission, then only the first ward to ICU transfer was used. The primary outcome of the study was in‐hospital mortality, and secondary outcomes were ICU mortality and hospital LOS.

Statistical Analysis

Patient characteristics were compared between patients who experienced delayed and nondelayed ICU transfers using t tests, Wilcoxon rank sums, and [2] tests, as appropriate. The association between length of transfer delay and in‐hospital mortality was calculated using logistic regression, with adjustment for age, sex, and surgical status. In a post hoc sensitivity analysis, additional adjustments were made using each patient's first eCART score on the ward, the individual vital signs and laboratory variables from eCART, and whether the ICU transfer was due to a cardiac arrest on the wards. In addition, an interaction term between time to transfer and the initial eCART on the ward was added to determine if the association between delay and mortality varied by baseline severity. The change in eCART score over time was plotted from 12 hours before the time of first reaching the critical value until ICU transfer for those in the delayed and nondelayed groups using restricted cubic splines to compare the trajectories of severity of illness between these 2 groups. In addition, a linear regression model was fit to investigate the association between the eCART slope in the 8 hours prior to the critical eCART value until ICU transfer and the timing of ICU transfer delay. Statistical analyses were performed using Stata version 12.1 (StataCorp, College Station, TX), and all tests of significance used a 2‐sided P<0.05.

RESULTS

A total of 269,999 admissions had documented vital signs on the hospital wards during the study period, including 11,995 patients who were either transferred from the wards to the ICU (n=11,636) or who suffered a cardiac arrest on the wards (n=359) during their initial ward stay. Of these patients, 3789 reached an eCART score at the 95% specificity cutoff (critical eCART score of 60) within 24 hours of transfer. The median time from first critical eCART value to ICU transfer was 5.4 hours (interquartile range (IQR), 214 hours; mean, 8 hours). Compared to patients without delayed ICU transfer, those with delayed transfer were slightly older (median age, 73 [IQR, 6083] years vs 71 [IQR, 5882] years; P=0.002), whereas all other characteristics were similar (Table 1). Table 2 shows comparisons of vital sign and laboratory results for delayed and nondelayed transfers at the time of ICU transfer. As shown, patients with delayed transfer had lower median respiratory rate, blood pressure, heart rate, and hemoglobin, but higher median white blood cell count and creatinine.

Comparisons of Patient Characteristics Among All ICU Transfer Patients and Nondelayed (Within Six Hours) and Delayed Transfers Who Reached the Critical CART Score
Characteristic Transferred Within 6 Hours, n=2,055 Transfer Delayed, n=1,734 P Value
  • NOTE: Data shown are mean (standard deviation) unless otherwise noted; n refers to the number of patients in each group. Abbreviations: eCART, electronic Cardiac Arrest Risk Triage; ICU, intensive care unit; IQR, interquartile range; LOS, length of stay. *For patients who survived to hospital discharge

Age, median (IQR), y 71 (5882) 73 (6083) 0.002
Female sex, n (%) 1,018 (49.5) 847 (48.8) 0.67
Race, n (%) 0.72
Black 467 (22.7) 374 (21.6)
White 1,141 (55.5) 971 (56.0)
Other/unknown 447 (21.8) 389 (22.4)
Surgical patient, n (%) 572 (27.8) 438 (25.2) 0.07
Hospital LOS prior to first critical eCART, median (IQR), d 1.5 (0.33.7) 1.6 (0.43.9) 0.04
Total hospital LOS, median (IQR), d* 11 (719) 13 (821) <0.001
Died during admission, n (%) 503 (24.5) 576 (33.2) <0.001
Comparison of Physiologic Variables at The time of ICU Transfer Between Nondelayed and Delayed ICU Transfers
Transferred Within 6 Hours, n=2,055 Transfer Delayed, n=1,734 P Value
  • NOTE: Abbreviations: Alk phos, alkaline phosphatase; BUN, blood urea nitrogen; Cr, creatinine; eCART, electronic Cardiac Arrest Risk Triage; GFR, glomerular filtration rate; ICU, intensive care unit; IQR, interquartile range; K+, potassium; SGOT, serum glutamic‐oxaloacetic transaminase; SGPT, serum glutamic pyruvic transaminase; WBC, white blood cells.

  • All data are median (IQR) unless otherwise noted.

Respiratory rate, breaths/min 23 (1830) 22 (1828) <0.001
Systolic blood pressure, mm Hg 111 (92134) 109 (92128) 0.002
Diastolic blood pressure, mm Hg 61 (5075) 59 (4971) <0.001
Heart rate, beats/min 106 (88124) 101 (85117) <0.001
Oxygen saturation, median (IQR), % 97 (9499) 97 (9599) 0.15
Temperature, F 98.0 (97.299.1) 98.0 (97.199.0) 0.001
Alert mental status, number of observations (%) 1,749 (85%) 1,431 (83%) <0.001
eCART score at time of ICU transfer 61 (26122) 48 (21121) 0.914
WBC 10.3 (7.514.5) 11.7 (8.117.0) <0.001
Hemoglobin 10.7 (9.312.0) 10.3 (9.111.6) <0.001
Platelet 215 (137275) 195 (120269) 0.017
Sodium 137 (134140) 137 (134141) 0.70
K+ 4.1 (3.84.6) 4.2 (3.84.7) 0.006
Anion Gap 10 (813) 10 (814) <0.001
CO2 24 (2026) 23 (1826) <0.001
BUN 24 (1640) 32 (1853) <0.001
Cr 1.2 (0.92.0) 1.5 (1.02.7) <0.001
GFR 70 (7070) 70 (5170) <0.001
Glucose 123 (106161) 129 (105164) 0.48
Calcium 8.5 (7.98.8) 8.2 (7.78.7) <0.001
SGOT 26 (2635) 26 (2644) 0.001
SGPT 21 (2127) 21 (2033) 0.002
Total bilirubin 0.7 (0.71.0) 0.7 (0.71.3) <0.001
Alk phos 80 (8096) 80 (79111) 0.175
Albumin 3.0 (2.73.0) 3.0 (2.43.0) <0.001

Delayed transfer occurred in 46% of patients (n=1734) and was associated with increased in‐hospital mortality (33.2% vs 24.5%, P<0.001). This relationship was linear, with each 1‐hour increase in transfer delay associated with a 3% increase in the odds of in‐hospital death (P<0.001) (Figure 1). The association between length of transfer delay and hospital mortality remained unchanged after controlling for age, sex, surgical status, initial eCART score on the wards, vital signs, laboratory values, and whether the ICU transfer was due to a cardiac arrest (3% increase per hour, P<0.001). This association did not vary based on the initial eCART score on the wards (P=0.71 for interaction). Additionally, despite having similar median hospital lengths of stay prior to first critical eCART score (1.6 vs 1.5 days, P=0.04), patients experiencing delayed ICU transfer who survived to discharge had a longer median hospital LOS by 2 days compared to those with nondelayed transfer who survived to discharge (median LOS, 13 (821) days vs 11 (719) days, P=0.01). The change in eCART score over time in the 12 hours before first reaching the critical eCART score until ICU transfer is shown in Figure 2 for patients with delayed and nondelayed transfer. As shown, patients transferred within 6 hours had a more rapid rise in eCART score prior to ICU transfer compared to those with a delayed transfer. This difference in trajectories between delayed and nondelayed patients was similar in patients with low (<13), intermediate (1359), and high (60) initial eCART scores on the wards. A regression model investigating the association between eCART slope prior to ICU transfer and time to ICU transfer demonstrated that a steeper slope was significantly associated with a decreased time to ICU transfer (P<0.01).

Figure 1
Association between length of intensive care unit (ICU) transfer delay and hospital mortality. Abbreviations: CI, confidence interval; eCART, electronic Cardiac Arrest Risk Triage.
Figure 2
Change in electronic Cardiac Arrest Risk Triage (eCART) score over time for the 12 hours prior to reaching the critical eCART value until intensive care unit (ICU) transfer for patients with delayed versus nondelayed ICU transfer. Time 0 denotes first critical eCART value.

DISCUSSION

We found that a delay in transfer to the ICU after reaching a predefined objective threshold of critical illness was associated with a significant increase in hospital mortality and hospital LOS. We also discovered a significant association between critical illness trajectory and delays in transfer, suggesting that caregivers may not recognize more subtle trends in critical illness. This work highlights the importance of timely transfer to the ICU for critically ill ward patients, which can be affected by several factors such as ICU bed availability and caregiver recognition and triage decisions. Our findings have significant implications for patient safety on the wards and provide further evidence for implementing early warning scores into practice to aid with clinical decision making.

Our findings of increased mortality with delayed ICU transfer are consistent with previous studies.[1, 5, 9] For example, Young et al. compared ICU mortality between delayed and nondelayed transfers in 91 consecutive patients with noncardiac diagnoses at a community hospital.[1] They also used predefined criteria for critical illness, and found that delayed transfers had a higher ICU mortality than nondelayed patients (41% vs 11%). However, their criteria for critical illness only had a specificity of 13% for predicting ICU transfer, compared to 95% in our study, suggesting that our threshold is more consistent with critical illness. Another study, by Cardoso and colleagues, investigated the impact of delayed ICU admission due to bed shortages on ICU mortality in 401 patients at a university hospital.[9] Of those patients deemed appropriate for transfer to the ICU but who had to wait for a bed to become available, the median wait time for a bed was 18 hours. They found that each hour of waiting was associated with a 1.5% increase in ICU death. A similar study by Robert and colleagues investigated the impact of delayed or refused ICU admission due to a lack of bed availability.[5] Patients deemed too sick (or too well) to benefit from ICU transfer were excluded. Twenty‐eightday and 60‐day mortality were higher in the admitted group compared to those not admitted, although this finding was not statistically significant. In addition, patients later admitted to the ICU once a bed became available (median wait time, 6 hours; n=89) had higher 28‐day mortality than those admitted immediately (adjusted odds ratio, 1.78; P=0.05). Several other studies have investigated the impact of ICU refusal for reasons that included bed shortages, and found increased mortality in those not admitted to the ICU.[16, 17] However, many of these studies included patients deemed too sick or too well to be transferred to the ICU in the group of nonadmitted patients. Our study adds to this literature by utilizing a highly specific objective measure of critical illness and by including all patients on the wards who reached this threshold, rather than only those for whom a consult was requested.

There are several potential explanations for our finding of increased mortality with delayed ICU transfer. First, those with delayed transfer might be different in some way from those transferred immediately. For example, we found that those with delayed transfer were older. The finding that increasing age is associated with a delay in ICU transfer is interesting, and may reflect physiologic differences in older patients compared to younger ones. For example, older patients have a lower maximum heart rate and thus may not develop the same level of vital sign abnormalities that younger patients do, causing them to be inappropriately left on the wards for too long.[18] In addition, patients with delayed transfer had more deranged renal function and lower blood pressure. It is unknown whether these organ dysfunctions would have been prevented by earlier transfer and to what degree they were related to chronic conditions. However, delayed transfer was still associated with increased mortality even after controlling for age, vital sign and laboratory values, and eCART on ward admission. It may also be possible that patients with delayed transfer received early and appropriate treatment on the wards but failed to improve and thus required ICU transfer. We did not have access to orders in this large database, so this theory will need to be investigated in future work. Finally, the most likely explanation for our findings is that earlier identification and treatment improves outcomes of critically ill patients on the wards, which is consistent with the findings of previous studies.[1, 5, 9, 10] Our study demonstrates that early identification of critical illness is crucial, and that delayed treatment can rapidly lead to increased mortality and LOS.

Our comparison of eCART score trajectory showed that patients transferred within 6 hours of onset of critical illness had a more rapid rise in eCART score over the preceding time period, whereas patients who experienced transfer delay showed a slower increase in eCART score. One explanation for this finding is that patients who decompensate more rapidly are in turn more readily recognizable to providers, whereas patients who experience a more insidious clinical deterioration are recognized later in the process, which then leads to a delay in escalation of care. This hypothesis underlines the importance of utilizing an objective marker of illness that is calculated longitudinally and in real time, as opposed to relying upon provider recognition alone. In fact, we have recently demonstrated that eCART is more accurate and identifies patients earlier than standard rapid response team activation.[19]

There are several important implications of our findings. First, it highlights the potential impact that early warning scores, particular those that are evidence based, can have on the outcomes of hospitalized patients. Second, it suggests that it is important to include age in early warning scores. Previous studies have been mixed as to whether the inclusion of age improves detection of outcomes on the wards, although the method of inclusion of age has been variable in terms of its weighting.[20, 21, 22] Our study found that older patients were more likely to be left on the wards longer prior to ICU transfer after becoming critically ill. By incorporating age into early warning scores, both accuracy and early recognition of critical illness may be improved. Finally, our finding that the trends of the eCART score differed among patients who were immediately transferred to the ICU, and who had a delay in their transfer, suggests that adding vital sign trends to early warning scores may further improve their accuracy and ability to serve as clinical decision support tools.

Our study is unique in that we used an objective measure of critical illness and then examined outcomes after patients reached this threshold on the wards. This overcomes the subjectivity of using evaluation by the ICU team or rapid response team as the starting point, as previous studies have shown a failure to call for help when patients become critically ill on the wards.[2, 11, 23] By using the eCART score, which contains commonly collected electronic health record data and can be calculated electronically in real time, we were able to calculate the score for patients on the wards and in the ICU. This allowed us to examine trends in the eCART score over time to find clues as to why some patients are transferred late to the ICU and why these late transfers have worse outcomes than those transferred earlier. Another strength is the large multicenter database used for the analysis, which included an urban tertiary care hospital, suburban teaching hospitals, and a community nonteaching hospital.

Our study has several limitations. First, we utilized just 1 of many potential measures of critical illness and a cutoff that only included one‐third of patients ultimately transferred to the ICU. However, by using the eCART score, we were able to track a patient's physiologic status over time and remove the variability that comes with using subjective definitions of critical illness. Furthermore, we utilized a high‐specificity cutoff for eCART to ensure that transferred patients had significantly deranged physiology and to avoid including planned transfers to the ICU. It is likely that some patients who were critically ill with less deranged physiology that would have benefitted from earlier transfer were excluded from the study. Second, we were unable to determine the cause of physiologic deterioration for patients in our study due to the large number of included patients. In addition, we did not have code status, comorbidities, or reason for ICU admission available in the dataset. It is likely that the impact of delayed transfer varies by the indication for ICU admission and chronic disease burden. It is also possible that controlling for these unmeasured factors could negate the beneficial association seen for earlier ICU admission. However, our finding of such a strong relationship between time to transfer and mortality after controlling for several important variables suggests that early recognition of critical illness is beneficial to many patients on the wards. Third, due to its observational nature, our study cannot estimate the true impact of timely ICU transfer on critically ill ward patient outcomes. Future clinical trials will be needed to determine the impact of electronic early warning scores on patient outcomes.

In conclusion, delayed ICU transfer is associated with significantly increased hospital LOS and mortality. This association highlights the need for ongoing work toward both the implementation of an evidence‐based risk stratification tool as well as development of effective critical care outreach resources for patients decompensating on the wards. Real‐time use of a validated early warning score, such as eCART, could potentially lead to more timely ICU transfer for critically ill patients and reduced rates of preventable in‐hospital death.

Acknowledgements

The authors thank Timothy Holper, Justin Lakeman, and Contessa Hsu for assistance with data extraction and technical support; Poome Chamnankit, MS, CNP, Kelly Bhatia, MSN, ACNP, and Audrey Seitman, MSN, ACNP for performing manual chart review of cardiac arrest patients; and Nicole Twu for administrative support.

Disclosures: This research was funded in part by an institutional Clinical and Translational Science Award grant (UL1 RR024999, PI: Dr. Julian Solway). Dr. Churpek is supported by a career development award from the National Heart, Lung, and Blood Institute (K08 HL121080). Drs. Churpek and Edelson have a patent pending (ARCD. P0535US.P2) for risk stratification algorithms for hospitalized patients. In addition, Dr. Edelson has received research support from Philips Healthcare (Andover, MA), the American Heart Association (Dallas, TX), and Laerdal Medical (Stavanger, Norway). She has ownership interest in Quant HC (Chicago, IL), which is developing products for risk stratification of hospitalized patients. Drs. Churpek and Wendlandt had full access to all the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis. Preliminary versions of these data were presented at the 2015 meeting of the Society of Hospital Medicine (March 31, 2015, National Harbor, MD).

References
  1. Young MP, Gooder VJ, McBride K, James B, Fisher ES. Inpatient transfers to the intensive care unit: delays are associated with increased mortality and morbidity. J Gen Intern Med. 2003;18(2):7783.
  2. McQuillan P, Pilkington S, Allan A, et al. Confidential inquiry into quality of care before admission to intensive care. BMJ. 1998;316(7148):18531858.
  3. Town JA, Churpek MM, Yuen TC, Huber MT, Kress JP, Edelson DP. Relationship between ICU bed availability, ICU readmission, and cardiac arrest in the general wards. Crit Care Med. 2014;42(9):20372041.
  4. Simchen E, Sprung CL, Galai N, et al. Survival of critically ill patients hospitalized in and out of intensive care units under paucity of intensive care unit beds. Crit Care Med. 2004;32(8):16541661.
  5. Robert R, Reignier J, Tournoux‐Facon C, et al. Refusal of intensive care unit admission due to a full unit: impact on mortality. Am J Respir Crit Care Med. 2012;185(10):10811087.
  6. Sprung CL, Geber D, Eidelman LA, et al. Evaluation of triage decisions for intensive care admission. Crit Care Med. 1999;27(6):10731079.
  7. Garrouste‐Orgeas M, Montuclard L, Timsit JF, et al. Predictors of intensive care unit refusal in French intensive care units: a multiple‐center study. Crit Care Med. 2005;33(4):750755.
  8. Zimmerman JE, Kramer AA, McNair DS, Malila FM. Acute Physiology and Chronic Health Evaluation (APACHE) IV: hospital mortality assessment for today's critically ill patients. Crit Care Med. 2006;34(5):12971310.
  9. Cardoso LT, Grion CM, Matsuo T, et al. Impact of delayed admission to intensive care units on mortality of critically ill patients: a cohort study. Crit Care. 2011;15(1):R28.
  10. Iapichino G, Corbella D, Minelli C, et al. Reasons for refusal of admission to intensive care and impact on mortality. Intensive Care Med. 2010;36(10):17721779.
  11. Hodgetts TJ, Kenward G, Vlackonikolis I, et al. Incidence, location and reasons for avoidable in‐hospital cardiac arrest in a district general hospital. Resuscitation. 2002;54(2):115123.
  12. Churpek MM, Yuen TC, Edelson DP. Risk stratification of hospitalized patients on the wards. Chest. 2013;143(6):17581765.
  13. Churpek MM, Yuen TC, Winslow C, et al. Multicenter development and validation of a risk stratification tool for ward patients. Am J Respir Crit Care Med. 2014;190(6):649655.
  14. Rivers E, Nguyen B, Havstad S, et al. Early goal‐directed therapy in the treatment of severe sepsis and septic shock. N Engl J Med. 2001;345(19):13681377.
  15. Dellinger RP, Levy MM, Rhodes A, et al. Surviving sepsis campaign: international guidelines for management of severe sepsis and septic shock: 2012. Crit Care Med. 2013;41(2):580637.
  16. Vanhecke TE, Gandhi M, McCullough PA, et al. Outcomes of patients considered for, but not admitted to, the intensive care unit. Crit Care Med. 2008;36(3):812817.
  17. Metcalfe MA, Sloggett A, McPherson K. Mortality among appropriately referred patients refused admission to intensive‐care units. Lancet. 1997;350(9070):711.
  18. Churpek MM, Yuen TC, Winslow C, Hall J, Edelson DP. Differences in vital signs between elderly and nonelderly patients prior to ward cardiac arrest. Crit Care Med. 2015;43(4):816822.
  19. Kang MA, Churpek MM, Zadravecz FJ, Adhikari R, Twu NM, Edelson DP. Real‐time risk prediction on the wards: a feasibility study [published April 13, 2016]. Crit Care Med. doi: 10.1097/CCM.0000000000001716.
  20. Smith GB, Prytherch DR, Schmidt PE, et al. Should age be included as a component of track and trigger systems used to identify sick adult patients? Resuscitation. 2008;78(2):109115.
  21. Duckitt RW, Buxton‐Thomas R, Walker J, et al. Worthing physiological scoring system: derivation and validation of a physiological early‐warning system for medical admissions. An observational, population‐based single‐centre study. Br J Anaesth. 2007;98(6):769774.
  22. Subbe CP, Kruger M, Rutherford P, Gemmel L. Validation of a modified Early Warning Score in medical admissions. QJM. 2001;94(10):521526.
  23. Hillman K, Chen J, Cretikos M, et al. Introduction of the medical emergency team (MET) system: a cluster‐randomised controlled trial. Lancet. 2005;365(9477):20912097.
References
  1. Young MP, Gooder VJ, McBride K, James B, Fisher ES. Inpatient transfers to the intensive care unit: delays are associated with increased mortality and morbidity. J Gen Intern Med. 2003;18(2):7783.
  2. McQuillan P, Pilkington S, Allan A, et al. Confidential inquiry into quality of care before admission to intensive care. BMJ. 1998;316(7148):18531858.
  3. Town JA, Churpek MM, Yuen TC, Huber MT, Kress JP, Edelson DP. Relationship between ICU bed availability, ICU readmission, and cardiac arrest in the general wards. Crit Care Med. 2014;42(9):20372041.
  4. Simchen E, Sprung CL, Galai N, et al. Survival of critically ill patients hospitalized in and out of intensive care units under paucity of intensive care unit beds. Crit Care Med. 2004;32(8):16541661.
  5. Robert R, Reignier J, Tournoux‐Facon C, et al. Refusal of intensive care unit admission due to a full unit: impact on mortality. Am J Respir Crit Care Med. 2012;185(10):10811087.
  6. Sprung CL, Geber D, Eidelman LA, et al. Evaluation of triage decisions for intensive care admission. Crit Care Med. 1999;27(6):10731079.
  7. Garrouste‐Orgeas M, Montuclard L, Timsit JF, et al. Predictors of intensive care unit refusal in French intensive care units: a multiple‐center study. Crit Care Med. 2005;33(4):750755.
  8. Zimmerman JE, Kramer AA, McNair DS, Malila FM. Acute Physiology and Chronic Health Evaluation (APACHE) IV: hospital mortality assessment for today's critically ill patients. Crit Care Med. 2006;34(5):12971310.
  9. Cardoso LT, Grion CM, Matsuo T, et al. Impact of delayed admission to intensive care units on mortality of critically ill patients: a cohort study. Crit Care. 2011;15(1):R28.
  10. Iapichino G, Corbella D, Minelli C, et al. Reasons for refusal of admission to intensive care and impact on mortality. Intensive Care Med. 2010;36(10):17721779.
  11. Hodgetts TJ, Kenward G, Vlackonikolis I, et al. Incidence, location and reasons for avoidable in‐hospital cardiac arrest in a district general hospital. Resuscitation. 2002;54(2):115123.
  12. Churpek MM, Yuen TC, Edelson DP. Risk stratification of hospitalized patients on the wards. Chest. 2013;143(6):17581765.
  13. Churpek MM, Yuen TC, Winslow C, et al. Multicenter development and validation of a risk stratification tool for ward patients. Am J Respir Crit Care Med. 2014;190(6):649655.
  14. Rivers E, Nguyen B, Havstad S, et al. Early goal‐directed therapy in the treatment of severe sepsis and septic shock. N Engl J Med. 2001;345(19):13681377.
  15. Dellinger RP, Levy MM, Rhodes A, et al. Surviving sepsis campaign: international guidelines for management of severe sepsis and septic shock: 2012. Crit Care Med. 2013;41(2):580637.
  16. Vanhecke TE, Gandhi M, McCullough PA, et al. Outcomes of patients considered for, but not admitted to, the intensive care unit. Crit Care Med. 2008;36(3):812817.
  17. Metcalfe MA, Sloggett A, McPherson K. Mortality among appropriately referred patients refused admission to intensive‐care units. Lancet. 1997;350(9070):711.
  18. Churpek MM, Yuen TC, Winslow C, Hall J, Edelson DP. Differences in vital signs between elderly and nonelderly patients prior to ward cardiac arrest. Crit Care Med. 2015;43(4):816822.
  19. Kang MA, Churpek MM, Zadravecz FJ, Adhikari R, Twu NM, Edelson DP. Real‐time risk prediction on the wards: a feasibility study [published April 13, 2016]. Crit Care Med. doi: 10.1097/CCM.0000000000001716.
  20. Smith GB, Prytherch DR, Schmidt PE, et al. Should age be included as a component of track and trigger systems used to identify sick adult patients? Resuscitation. 2008;78(2):109115.
  21. Duckitt RW, Buxton‐Thomas R, Walker J, et al. Worthing physiological scoring system: derivation and validation of a physiological early‐warning system for medical admissions. An observational, population‐based single‐centre study. Br J Anaesth. 2007;98(6):769774.
  22. Subbe CP, Kruger M, Rutherford P, Gemmel L. Validation of a modified Early Warning Score in medical admissions. QJM. 2001;94(10):521526.
  23. Hillman K, Chen J, Cretikos M, et al. Introduction of the medical emergency team (MET) system: a cluster‐randomised controlled trial. Lancet. 2005;365(9477):20912097.
Issue
Journal of Hospital Medicine - 11(11)
Issue
Journal of Hospital Medicine - 11(11)
Page Number
757-762
Page Number
757-762
Article Type
Display Headline
Association between intensive care unit transfer delay and hospital mortality: A multicenter investigation
Display Headline
Association between intensive care unit transfer delay and hospital mortality: A multicenter investigation
Sections
Article Source
© 2016 Society of Hospital Medicine
Disallow All Ads
Correspondence Location
Address for correspondence and reprint requests: Matthew M. Churpek, MD, University of Chicago Medical Center, Section of Pulmonary and Critical Care Medicine, 5841 South Maryland Avenue, MC 6076, Chicago, IL 60637; Telephone: 773‐702‐1092; Fax: 773‐702‐6500; E‐mail: [email protected]
Content Gating
Gated (full article locked unless allowed per User)
Gating Strategy
First Peek Free
Article PDF Media
Media Files

Oophorectomy cost-effective at 4% lifetime ovarian cancer risk

Article Type
Changed
Fri, 01/18/2019 - 16:01
Display Headline
Oophorectomy cost-effective at 4% lifetime ovarian cancer risk

Premenopausal risk-reducing salpingo-oophorectomy becomes cost-effective in women who have a 4% or greater lifetime risk of ovarian cancer, according to a modeling study published online in the Journal of Medical Genetics.

The procedure, which is usually undertaken in women aged over 35 years who have completed their families, is available in the United Kingdom to women with a greater than 10% lifetime risk of ovarian cancer. However, the researchers, led by Dr. Ranjit Manchanda of Barts Cancer Institute at Queen Mary University of London, suggested that this threshold has not been tested for cost-effectiveness.

The decision analysis model evaluated lifetime costs as well as the effects of risk-reducing salpingo-oophorectomy in 40-year-old premenopausal women by comparing it with no procedure in women whose lifetime ovarian cancer risk ranged from 2%-10%. The final outcomes were development of breast cancer, ovarian cancer, and excess deaths from coronary heart disease, while cost-effectiveness was judged against the National Institute for Health and Care Excellence threshold of £20,000-£30,000 per quality-adjusted life-years (QALY).

Researchers found that premenopausal risk-reducing salpingo-oophorectomy was cost-effective in women with a 4% or greater lifetime risk of ovarian cancer, largely because of the reduction in their risk of breast cancer. At this level of risk, surgery gained 42.7 days of life-expectancy, with an incremental cost-effectiveness ratio of £19,536($26,186)/QALY.

Premenopausal risk-reducing salpingo-oophorectomy was not cost-effective at the baseline risk rate of 2%, with an incremental cost-effectiveness ratio of £46,480($62,267)/QALY and 19.9 days gain in life expectancy (J Med Genetics 2016 June 27. doi: 10.1136/jmedgenet-2016-103800).

The cost-effectiveness was predicated on the assumption of at least an 80% compliance rate with hormone therapy (HT) in women who underwent the procedure; without HT, the cost-effectiveness threshold increased to a lifetime risk of over 8.2%.

“Our results are of major significance for clinical practice and risk management in view of declining genetic testing costs and the improvements in estimating an individual’s OC risk,” the authors wrote.

“With routine clinical testing for certain moderate penetrance genes around the corner and lack of an effective OC screening programme, these findings are timely as it provides evidence supporting a surgical prevention strategy for ‘lower-risk’ (lifetime risk less than 10%) individuals,” noted Dr. Manchanda and colleagues.

They stressed that symptom levels after salpingo-oophorectomy, particularly for sexual function, were still higher even in women taking HT compared to those who hadn’t undergone salpingo-oophorectomy.

“This limitation needs to be discussed as part of informed consent for the surgical procedure and incorporated into [the risk-reducing salpingo-oophorectomy] decision-making process,” they wrote.

One author declared a financial interest in Abcodia, which has an interest in ovarian cancer screening and biomarkers for screening and risk prediction. No other conflicts of interest were declared.

Click for Credit Link
Publications
Topics
Click for Credit Link
Click for Credit Link

Premenopausal risk-reducing salpingo-oophorectomy becomes cost-effective in women who have a 4% or greater lifetime risk of ovarian cancer, according to a modeling study published online in the Journal of Medical Genetics.

The procedure, which is usually undertaken in women aged over 35 years who have completed their families, is available in the United Kingdom to women with a greater than 10% lifetime risk of ovarian cancer. However, the researchers, led by Dr. Ranjit Manchanda of Barts Cancer Institute at Queen Mary University of London, suggested that this threshold has not been tested for cost-effectiveness.

The decision analysis model evaluated lifetime costs as well as the effects of risk-reducing salpingo-oophorectomy in 40-year-old premenopausal women by comparing it with no procedure in women whose lifetime ovarian cancer risk ranged from 2%-10%. The final outcomes were development of breast cancer, ovarian cancer, and excess deaths from coronary heart disease, while cost-effectiveness was judged against the National Institute for Health and Care Excellence threshold of £20,000-£30,000 per quality-adjusted life-years (QALY).

Researchers found that premenopausal risk-reducing salpingo-oophorectomy was cost-effective in women with a 4% or greater lifetime risk of ovarian cancer, largely because of the reduction in their risk of breast cancer. At this level of risk, surgery gained 42.7 days of life-expectancy, with an incremental cost-effectiveness ratio of £19,536($26,186)/QALY.

Premenopausal risk-reducing salpingo-oophorectomy was not cost-effective at the baseline risk rate of 2%, with an incremental cost-effectiveness ratio of £46,480($62,267)/QALY and 19.9 days gain in life expectancy (J Med Genetics 2016 June 27. doi: 10.1136/jmedgenet-2016-103800).

The cost-effectiveness was predicated on the assumption of at least an 80% compliance rate with hormone therapy (HT) in women who underwent the procedure; without HT, the cost-effectiveness threshold increased to a lifetime risk of over 8.2%.

“Our results are of major significance for clinical practice and risk management in view of declining genetic testing costs and the improvements in estimating an individual’s OC risk,” the authors wrote.

“With routine clinical testing for certain moderate penetrance genes around the corner and lack of an effective OC screening programme, these findings are timely as it provides evidence supporting a surgical prevention strategy for ‘lower-risk’ (lifetime risk less than 10%) individuals,” noted Dr. Manchanda and colleagues.

They stressed that symptom levels after salpingo-oophorectomy, particularly for sexual function, were still higher even in women taking HT compared to those who hadn’t undergone salpingo-oophorectomy.

“This limitation needs to be discussed as part of informed consent for the surgical procedure and incorporated into [the risk-reducing salpingo-oophorectomy] decision-making process,” they wrote.

One author declared a financial interest in Abcodia, which has an interest in ovarian cancer screening and biomarkers for screening and risk prediction. No other conflicts of interest were declared.

Premenopausal risk-reducing salpingo-oophorectomy becomes cost-effective in women who have a 4% or greater lifetime risk of ovarian cancer, according to a modeling study published online in the Journal of Medical Genetics.

The procedure, which is usually undertaken in women aged over 35 years who have completed their families, is available in the United Kingdom to women with a greater than 10% lifetime risk of ovarian cancer. However, the researchers, led by Dr. Ranjit Manchanda of Barts Cancer Institute at Queen Mary University of London, suggested that this threshold has not been tested for cost-effectiveness.

The decision analysis model evaluated lifetime costs as well as the effects of risk-reducing salpingo-oophorectomy in 40-year-old premenopausal women by comparing it with no procedure in women whose lifetime ovarian cancer risk ranged from 2%-10%. The final outcomes were development of breast cancer, ovarian cancer, and excess deaths from coronary heart disease, while cost-effectiveness was judged against the National Institute for Health and Care Excellence threshold of £20,000-£30,000 per quality-adjusted life-years (QALY).

Researchers found that premenopausal risk-reducing salpingo-oophorectomy was cost-effective in women with a 4% or greater lifetime risk of ovarian cancer, largely because of the reduction in their risk of breast cancer. At this level of risk, surgery gained 42.7 days of life-expectancy, with an incremental cost-effectiveness ratio of £19,536($26,186)/QALY.

Premenopausal risk-reducing salpingo-oophorectomy was not cost-effective at the baseline risk rate of 2%, with an incremental cost-effectiveness ratio of £46,480($62,267)/QALY and 19.9 days gain in life expectancy (J Med Genetics 2016 June 27. doi: 10.1136/jmedgenet-2016-103800).

The cost-effectiveness was predicated on the assumption of at least an 80% compliance rate with hormone therapy (HT) in women who underwent the procedure; without HT, the cost-effectiveness threshold increased to a lifetime risk of over 8.2%.

“Our results are of major significance for clinical practice and risk management in view of declining genetic testing costs and the improvements in estimating an individual’s OC risk,” the authors wrote.

“With routine clinical testing for certain moderate penetrance genes around the corner and lack of an effective OC screening programme, these findings are timely as it provides evidence supporting a surgical prevention strategy for ‘lower-risk’ (lifetime risk less than 10%) individuals,” noted Dr. Manchanda and colleagues.

They stressed that symptom levels after salpingo-oophorectomy, particularly for sexual function, were still higher even in women taking HT compared to those who hadn’t undergone salpingo-oophorectomy.

“This limitation needs to be discussed as part of informed consent for the surgical procedure and incorporated into [the risk-reducing salpingo-oophorectomy] decision-making process,” they wrote.

One author declared a financial interest in Abcodia, which has an interest in ovarian cancer screening and biomarkers for screening and risk prediction. No other conflicts of interest were declared.

Publications
Publications
Topics
Article Type
Display Headline
Oophorectomy cost-effective at 4% lifetime ovarian cancer risk
Display Headline
Oophorectomy cost-effective at 4% lifetime ovarian cancer risk
Click for Credit Status
Active
Article Source

FROM THE JOURNAL OF MEDICAL GENETICS

Disallow All Ads
Vitals

Key clinical point: Premenopausal risk-reducing salpingo-oophorectomy becomes cost-effective in women who have a 4% or greater lifetime risk of ovarian cancer.

Major finding: Premenopausal risk-reducing salpingo-oophorectomy in women with a 4% or greater lifetime risk of ovarian cancer gained 42.7 days of life expectancy, with an incremental cost-effectiveness ratio of £19,536($26,186)/QALY.

Data source: Decision analysis model.

Disclosures: One author declared a financial interest in Abcodia, which has an interest in ovarian cancer screening and biomarkers for screening and risk prediction. No other conflicts of interest were declared.