The American Psychiatric Association (APA) has updated its Privacy Policy and Terms of Use, including with new information specifically addressed to individuals in the European Economic Area. As described in the Privacy Policy and Terms of Use, this website utilizes cookies, including for the purpose of offering an optimal online experience and services tailored to your preferences.

Please read the entire Privacy Policy and Terms of Use. By closing this message, browsing this website, continuing the navigation, or otherwise continuing to use the APA's websites, you confirm that you understand and accept the terms of the Privacy Policy and Terms of Use, including the utilization of cookies.

×
Full Access

Guideline Statements and Implementation

Assessment and Determination of Treatment Plan

Statement 1: Assessment of Possible Schizophrenia

APA recommends (1C) that the initial assessment of a patient with a possible psychotic disorder include the reason the individual is presenting for evaluation; the patient’s goals and preferences for treatment; a review of psychiatric symptoms and trauma history; an assessment of tobacco use and other substance use; a psychiatric treatment history; an assessment of physical health; an assessment of psychosocial and cultural factors; a mental status examination, including cognitive assessment; and an assessment of risk of suicide and aggressive behaviors, as outlined in APA’s Practice Guidelines for the Psychiatric Evaluation of Adults (3rd edition).

Implementation

The importance of the psychiatric evaluation cannot be underestimated because it serves as the initial basis for a therapeutic relationship with the patient and provides information that is crucial to differential diagnosis, shared decision-making about treatment, and educating patients and family members about such factors as illness course and prognosis. APA’s Practice Guidelines for the Psychiatric Evaluation of Adults, 3rd edition (American Psychiatric Association 2016a) describe recommended and suggested elements of assessment for any individual who presents with psychiatric symptoms (Table 1). These elements are by no means comprehensive, and additional areas of inquiry will become apparent as the evaluation unfolds, depending on the responses to initial questions, the presenting concerns, the observations of the clinician during the assessment, the complexity and urgency of clinical decision-making, and other aspects of the clinical context. In many circumstances, aspects of the evaluation will extend across multiple visits (American Psychiatric Association 2016a).

Recommended aspects of the initial psychiatric evaluation

History of present illness

• Reason that the patient is presenting for evaluation, including current symptoms, behaviors, and precipitating factors

• Current psychiatric diagnoses and psychiatric review of systems

Psychiatric history

• Hospitalization and emergency department visits for psychiatric issues, including substance use disorders

• Psychiatric treatments (type, duration, and, where applicable, doses)

• Response and adherence to psychiatric treatments, including psychosocial treatments, pharmacotherapy, and other interventions such as electroconvulsive therapy or transcranial magnetic stimulation

• Prior psychiatric diagnoses and symptoms, including

Hallucinations (including command hallucinations), delusions, and negative symptoms

Aggressive ideas or behaviors (e.g., homicide, domestic or workplace violence, other physically or sexually aggressive threats or acts)

Suicidal ideas, suicide plans, and suicide attempts, including details of each attempt (e.g., context, method, damage, potential lethality, intent) and attempts that were aborted or interrupted

Intentional self-injury in which there was no suicide intent

Impulsivity

Substance use history

• Use of tobacco, alcohol, and other substances (e.g., vaping, marijuana, cocaine, heroin, hallucinogens) and any misuse of prescribed or over-the-counter medications or supplements

• Current or recent substance use disorder or change in use of alcohol or other substances

Medical history

• Whether or not the patient has an ongoing relationship with a primary care health professional

• Allergies or drug sensitivities

• All medications the patient is currently taking or has recently taken and the side effects of these medications (i.e., both prescribed and nonprescribed medications, herbal and nutritional supplements, and vitamins)

• Past or current medical illnesses and related hospitalizations

• Relevant past or current treatments, including surgeries, other procedures, or complementary and alternative medical treatments

• Sexual and reproductive history

• Cardiopulmonary status

• Past or current neurological or neurocognitive disorders or symptoms

• Past physical trauma, including head injuries

• Past or current endocrinological disease

• Past or current infectious disease, including sexually transmitted diseases, HIV, tuberculosis, hepatitis C, and locally endemic infectious diseases such as Lyme disease

• Past or current sleep abnormalities, including sleep apnea

• Past or current symptoms or conditions associated with significant pain and discomfort

• Additional review of systems, as indicated

Family history

• Including history of suicidal behaviors or aggressive behaviors in biological relatives

Personal and social history

• Preferred language and need for an interpreter

• Personal/cultural beliefs, sociocultural environment, and cultural explanations of psychiatric illness

• Presence of psychosocial stressors (e.g., financial, housing, legal, school/occupational, or interpersonal/relationship problems; lack of social support; painful, disfiguring, or terminal medical illness)

• Exposure to physical, sexual, or emotional trauma

• Exposure to violence or aggressive behavior, including combat exposure or childhood abuse

• Legal or disciplinary consequences of past aggressive behaviors

Examination, including mental status examination

• General appearance and nutritional status

• Height, weight, and body mass index (BMI)

• Vital signs

• Skin, including any stigmata of trauma, self-injury, or drug use

• Coordination and gait

• Involuntary movements or abnormalities of motor tone

• Sight and hearing

• Speech, including fluency and articulation

• Mood, degree of hopelessness, and level of anxiety

• Thought content, process, and perceptions, including current hallucinations, delusions, negative symptoms, and insight

• Cognition

• Current suicidal ideas, suicide plans, and suicide intent, including active or passive thoughts of suicide or death

If current suicidal ideas are present, assess patient’s intended course of action if current symptoms worsen; access to suicide methods, including firearms; possible motivations for suicide (e.g., attention or reaction from others, revenge, shame, humiliation, delusional guilt, command hallucinations); reasons for living (e.g., sense of responsibility to children or others, religious beliefs); and quality and strength of the therapeutic alliance

• Current aggressive ideas, including thoughts of physical or sexual aggression or homicide

If current aggressive ideas are present, assess specific individuals or groups toward whom homicidal or aggressive ideas or behaviors have been directed in the past or at present, access to firearms, and impulsivity, including anger management issues

Source. Adapted from APA’s Practice Guidelines for the Psychiatric Evaluation of Adults, 3rd Edition. Arlington VA, American Psychiatric Association, 2016. Copyright © 2016 American Psychiatric Association. Used with permission.

Recommended aspects of the initial psychiatric evaluation

Enlarge table

The specific approach to the interview will depend on many factors, including the patient’s ability to communicate, degree of cooperation, level of insight, illness severity, and ability to recall historical details (American Psychiatric Association 2016a). Such factors as the patient’s health literacy (Clausen et al. 2016) and cultural background (Lewis-Fernández et al. 2016) can also influence the patient’s understanding or interpretation of questions. Typically, a psychiatric evaluation involves a direct interview between the patient and the clinician (American Psychiatric Association 2016a). The use of open-ended empathic questions about the patient’s current life circumstances and reasons for evaluation can provide an initial picture of the individual and serve as a way of establishing rapport. Such questions can be followed up with additional structured inquiry about history, symptoms, or observations made during the assessment.

Throughout the assessment process, it is important to gain an understanding of the patient’s goals, their view of the illness, and preferences for treatment. This information will serve as a starting point for person-centered care and shared decision-making with the patient, family, and other persons of support (Dixon et al. 2016; Hamann and Heres 2019). It will also provide a framework for recovery, which has been defined as “a process of change through which individuals improve their health and wellness, live self-directed lives, and strive to reach their full potential” (Substance Abuse and Mental Health Services Administration 2012a, p. 3). Consequently, discussions of goals should be focused beyond symptom relief and may include goals related to schooling, employment, living situation, relationships, leisure activities, and other aspects of functioning and quality of life. Questions about the patient’s views may help determine whether the patient is aware of having an illness and whether the patient has other explanations for symptoms that are helpful to them (Saks 2009). Patients may have specific views about such topics as medications, other treatment approaches, mechanical restraints, or involuntary treatment based on prior treatment experiences. They may also be able to delineate strategies that have been helpful for them in coping with or managing their symptoms in the past (Cohen et al. 2017). Some patients will have completed a psychiatric advance directive (Murray and Wortzel 2019), which is important to review with the patient if it exists.

In addition to direct interview, patients may be asked to complete electronic or paper-based forms that ask about psychiatric symptoms or key aspects of the history (American Psychiatric Association 2016a). When available, prior medical records, electronic prescription databases, and input from other treating clinicians can add further details to the history or corroborate information obtained in the interview (American Psychiatric Association 2016a).

Family members, friends, and other individuals involved in the patient’s support network can be an important part of the patient’s care team and valuable sources of collateral information about the reason for evaluation, the patient’s past history, and current symptoms and behavior (American Psychiatric Association 2016a). Outreach to family, friends, and others in the support network will typically occur with the patient’s permission. In situations in which the patient is given the opportunity and does not object, necessary information can be shared with family members or other persons involved in the patient’s care or payment for care (Office for Civil Rights 2017b). For example, if a relative or person of support is present with the patient at an appointment, the clinician may discuss information about medications or give education about warning signs of a developing emergency.

In some instances, however, patients may ask that family or others not be contacted. When this is the case, patients can usually identify someone whom they trust to provide additional information, and they are often willing to reconsider contact as treatment proceeds. It is also useful to discuss the reasons that the patient has concerns about contacts with family members or other important people in the patient’s life. For example, a patient may wish to avoid burdening a loved one, may have felt unsupported by a particular family member in the past, or may be experiencing delusional beliefs that involve a family member or friend. The patient may also want to limit the information that clinicians receive about past or recent treatment, symptoms, or behaviors. Even when a patient does not want a specific person to be contacted, the clinician may listen to information provided by that individual, as long as confidential information is not provided to the informant (American Psychiatric Association 2016a). Also, to prevent or lessen a serious and imminent threat to the health or safety of the patient or others, The Principles of Medical Ethics (American Psychiatric Association 2013f) and the Health Insurance Portability and Accountability Act of 1996 (HIPAA; Office for Civil Rights 2017b) permit clinicians to disclose necessary information about a patient to family members, caregivers, law enforcement, or other persons involved with the patient as well as to jails, prisons, and law enforcement officials having lawful custody of the patient. HIPAA also permits health care providers to disclose necessary information to the patient’s family, friends, or other persons involved in the patient’s care or payment for care when such disclosure is judged to be in the best interests of the patient and the patient either is not present or is unable to agree or object to a disclosure because of incapacity or emergency circumstances. Examples of such circumstances are not limited to unconsciousness and may also include such circumstances as temporary psychosis or intoxication with alcohol or other substances (Office for Civil Rights 2017b).

Although it is beyond the scope of this guideline to discuss the differential diagnosis and evaluation of psychotic disorders, many features and aspects of clinical course will enter into such a determination in addition to psychotic symptoms per se. Clinicians should also be mindful that biases can influence assessment and diagnosis, with disparities in diagnosis based on race being particularly common (Olbert et al. 2018; Schwartz and Blankenship 2014). The clinician should be alert to features of the history, including family, developmental, and academic history, that may suggest specific conditions or a need for additional physical or laboratory evaluation. Examples of conditions that can mimic schizophrenia in their initial presentation include neurosyphilis, Huntington’s disease, Wilson’s disease, and anti-N-methyl-d-aspartate (NMDA) receptor encephalitis (Lieberman and First 2018). Individuals with 22q11.2 deletion syndrome have a substantially increased risk of developing schizophrenia (Bassett et al. 2017; McDonald-McGinn et al. 2015; Van et al. 2017). In addition, the presence of a 22q11.2 deletion is associated with an increased likelihood of neurocognitive and physical health impairments (McDonald-McGinn et al. 2015; Moberg et al. 2018; Swillen and McDonald-McGinn 2015), which has implications for treatment (Fung et al. 2015; Mosheva et al. 2019). Psychotic symptoms can also occur in the context of other neurological and systemic illnesses, with or without delirium, and such acute states can at times be mistaken for an acute exacerbation of schizophrenia. Furthermore, because a significant fraction of individuals with psychosis will have a shift in diagnosis over time, the diagnosis may need to be reevaluated as new information about the patient’s illness course and symptoms becomes available (Bromet et al. 2011). Specialty consultation can be helpful in establishing and clarifying diagnosis (Coulter et al. 2019), particularly if the illness symptoms or course appear to be atypical or if the patient is not responding to treatment.

A thorough history is also important for identifying the presence of co-occurring psychiatric conditions or physical disorders that need to be addressed in treatment planning (American Psychiatric Association 2016a; Firth et al. 2019). For example, individuals with serious mental illness have higher rates of smoking, higher rates of heavy smoking, and lower rates of smoking cessation than do community samples (Cook et al. 2014; de Leon and Diaz 2005; Myles et al. 2012; Wium-Andersen et al. 2015). Furthermore, the use of cannabis may be more frequent in individuals with schizophrenia (Koskinen et al. 2010) and associated with greater symptom severity or earlier onset of psychosis (Carney et al. 2017; Large et al. 2011). Other substance use disorders, if present, can also produce or exacerbate symptoms of psychosis (American Psychiatric Association 2016a; Large et al. 2014). Thus, as part of the initial evaluation, it is important to determine whether the patient uses tobacco, cannabis, or other substances such as alcohol, caffeine, cocaine, opioids, sedative-hypnotic agents, stimulants, 3,4-methylenedioxymethamphetamine (MDMA), solvents, androgenic steroids, hallucinogens, or synthetic substances (e.g., “bath salts,” K2, Spice). The route by which substances are used (e.g., ingestion, smoking, vaping, intranasal, intravenous) is similarly important to document.

Mortality is increased in individuals with schizophrenia (Brown et al. 2000; Fazel et al. 2014; Olfson et al. 2015), and the average life span is shortened by a decade or more, with much of this decrease related to increased rates of co-occurring physical conditions (Laursen et al. 2013; Saha et al. 2007; Walker et al. 2015). Adverse health effects of smoking also contribute to an increased risk of mortality among individuals with schizophrenia (Lariscy et al. 2018; Reynolds et al. 2018; Tam et al. 2016). Many other conditions are more frequent in individuals with serious mental illness in general (Janssen et al. 2015; McGinty et al. 2016) and schizophrenia in particular (Henderson et al. 2015), including, but not limited to, poor oral health (Kisely et al. 2015), hepatitis C infection (Chasser et al. 2017; Hauser and Kern 2015; Hughes et al. 2016), HIV infection (Hobkirk et al. 2015; Hughes et al. 2016), cancer (Olfson et al. 2015), sleep apnea (Myles et al. 2016; Stubbs et al. 2016b), obesity (Janssen et al. 2015), diabetes mellitus (Vancampfort et al. 2016a), metabolic syndrome (Vancampfort et al. 2015), and cardiovascular disease (Correll et al. 2017c). These disorders, if present, can contribute to mortality or reduced quality of life, and some may be induced or exacerbated by psychiatric medications. Laboratory tests and physical examination as part of the initial evaluation can help to identify common co-occurring conditions and can serve as a baseline for subsequent monitoring during treatment (Table 2).

Suggested physical and laboratory assessments for patients with schizophrenia

Initial or baseline assessmentsa

Follow-up assessmentsb

Assessments to monitor physical status and detect concomitant physical conditions

Vital signs

Pulse, blood pressure

Pulse, blood pressure, temperature as clinically indicated

Body weight and height

Body weight, height, BMIc

BMIc every visit for 6 months and at least quarterly thereafter

Hematology

CBC, including ANC

CBC, including ANC if clinically indicated (e.g., patients treated with clozapine)

Blood chemistries

Electrolytes, renal function tests, liver function tests, TSH

As clinically indicated

Pregnancy

Pregnancy test for women of childbearing potential

Toxicology

Drug toxicology screen, if clinically indicated

Drug toxicology screen, if clinically indicated

Electrophysiological studies

EEG, if indicated on the basis of neurological examination or history

Imaging

Brain imaging (CT or MRI, with MRI being preferred), if indicated on the basis of neurological examination or historyd

Genetic testing

Chromosomal testing, if indicated on the basis of physical examination or history, including developmental historye

Assessments related to other specific side effects of treatment

Diabetesf

Screening for diabetes risk factors,g fasting blood glucoseh

Fasting blood glucose or hemoglobin A1C at 4 months after initiating a new treatment and at least annually thereafterh

Hyperlipidemia

Lipid paneli

Lipid paneli at 4 months after initiating a new antipsychotic medication and at least annually thereafter

Metabolic syndrome

Determine whether metabolic syndrome criteria are metj

Determine whether metabolic syndrome criteria are metj at 4 months after initiating a new antipsychotic medication and at least annually thereafter

QTc prolongation

ECG before treatment with chlorpromazine, droperidol, iloperidone, pimozide, thioridazine, or ziprasidonek or in the presence of cardiac risk factorsl

ECG with significant change in dose of chlorpromazine, droperidol, iloperidone, pimozide, thioridazine, or ziprasidonek or with the addition of other medications that can affect QTc interval in patients with cardiac risk factorsl or elevated baseline QTc intervals

Hyperprolactinemia

Screening for symptoms of hyperprolactinemiam

Prolactin level, if indicated on the basis of clinical history

Screening for symptoms of hyperprolactinemia at each visit until stable, then yearly if treated with an antipsychotic known to increase prolactinm

Prolactin level, if indicated on the basis of clinical history

Antipsychotic-induced movement disorders

Clinical assessment of akathisia, dystonia, parkinsonism, and other abnormal involuntary movements, including tardive dyskinesian

Clinical assessment of akathisia, dystonia, parkinsonism, and other abnormal involuntary movements, including tardive dyskinesia, at each visitn

Assessment with a structured instrument (e.g., AIMS, DISCUS) if such movements are present

Assessment with a structured instrument (e.g., AIMS, DISCUS) at a minimum of every 6 months in patients at high risk of tardive dyskinesiao and at least every 12 months in other patientsp as well as if a new onset or exacerbation of preexisting movements is detected at any visit

aAPA’s Practice Guidelines for the Psychiatric Evaluation of Adults, 3rd edition (American Psychiatric Association 2016a) recommends that the initial psychiatric evaluation of a patient include assessment of whether or not the patient has an ongoing relationship with a primary care health professional. Preventive care and other tests, such as screening for hepatitis C or HIV, are expected to occur as a part of routine primary care. Nevertheless, determining whether a patient is receiving primary care and inquiring about the patient’s relationship with their primary care practitioner can be a starting point for improved access to quality health care and preventive services.

bAlthough this practice guideline recommends that patients treated with antipsychotic medications be monitored for physical conditions and side effects on a regular basis, there are no absolute criteria for frequency of monitoring. Occurrence of conditions and side effects may be influenced by the patient’s history, preexisting conditions, and use of other medications in addition to antipsychotic agents. Thus, decisions about monitoring patients for physical conditions, specific side effects, or abnormalities in laboratory test results will necessarily depend on the clinical circumstances. In general, assessments related to physical conditions and specific medication-related side effects will be done at the time of initiating or changing antipsychotic medications or when adding other medications that contribute to these side effects.

cBMI may be calculated by using the formula weight in kg/(height in m)2 or the formula 703 × weight in lb/(height in inches)2 or by using a BMI calculator available from the National Heart, Lung, and Blood Institute (www.nhlbi.nih.gov/health/educational/lose_wt/BMI/bmicalc.htm). A person with a BMI of 25–29.9 is considered overweight, and one with a BMI of 30 or higher is considered obese. In addition to BMI, waist circumference can be used as an indicator of risk (> 35 inches for women and > 40 inches for men). Except for patients with a BMI of < 18.5, an increase in BMI of 1 unit would suggest a need for intervention by monitoring weight more closely, engaging the patient in a weight management program, using an adjunctive treatment to reduce weight, or changing the antipsychotic medication.

dFactors that suggest a possible need for imaging include focal neurological signs, new onset of seizures, later age at symptom onset, symptoms suggestive of intracranial pathology (e.g., chronic or severe headaches, nausea, vomiting), and symptoms suggestive of autoimmune encephalitis (e.g., rapid progression of working memory deficits over less than 3 months; decreased or altered level of consciousness, lethargy, or personality change; Graus et al. 2016). In the absence of such indications, decisions about imaging should consider that the yield of routine brain imaging is low, with < 1% of studies showing potentially serious incidental findings or abnormalities that would influence treatment (Cunqueiro et al. 2019; Falkenberg et al. 2017; Forbes et al. 2019; Gibson et al. 2018). On the other hand, routine imaging is a low-risk procedure, and a negative finding can be reassuring to patients and to families. If imaging is ordered, it is rarely necessary to delay other treatment or hospitalization while awaiting imaging results.

eFactors that may suggest a possible need for chromosomal testing (e.g., to identify abnormalities such as 22q11.2 deletion syndrome) include mild dysmorphic features, hypernasal speech, developmental delays, intellectual impairments, learning difficulties, and congenital heart defects (Bassett and Chow 1999; Miller et al. 2010).

fThe U.S. Food and Drug Administration has requested that all manufacturers of second-generation antipsychotic medications (SGAs) include a warning in their product labeling regarding hyperglycemia and diabetes mellitus. Although precise risk estimates for hyperglycemia-related adverse events are not available for each agent, epidemiological studies suggested an increased risk of treatment-emergent adverse events with SGAs, including extreme hyperglycemia. In some patients, this hyperglycemia was associated with ketoacidosis, hyperosmolar coma, or death.

gFactors that indicate an increased risk for undiagnosed diabetes include a BMI > 25, a first-degree relative with diabetes, habitual physical inactivity, being a member of a high-risk ethnic population (African American, Hispanic, American Indian, Asian American, Pacific Islander), history of cardiovascular disease, hypertension (≥ 140/90 mmHg or on therapy for hypertension), high-density lipoprotein cholesterol (HDL-C) < 35 mg/dL (0.90 mmol/L) and/or triglyceride level > 250 mg/dL (2.82 mmol/L), polycystic ovary syndrome (in women), having had gestational diabetes, and other clinical conditions associated with insulin resistance (e.g., severe obesity, acanthosis nigricans) (American Diabetes Association 2018). Symptoms of possible diabetes include frequent urination, excessive thirst, extreme hunger, unusual weight loss, increased fatigue, irritability, and blurry vision.

hWhen screening for the presence of diabetes, criteria for diagnosis include a fasting blood glucose higher than 125 mg/dL, where fasting is defined as no caloric intake for at least 8 hours (American Diabetes Association 2018). Alternatively, a hemoglobin A1C of 6.5% or greater can be used. Other acceptable approaches for diagnosis of diabetes include an oral glucose tolerance test or a random blood glucose of at least 200 mg/dL in conjunction with a hyperglycemic crisis or classic symptoms of hyperglycemia. With all of these approaches, results should be confirmed by repeat testing unless unequivocal hyperglycemia is present. In patients with hemoglobinopathies or conditions associated with increased red blood cell turnover (e.g., second- or third-trimester pregnancy, hemodialysis, recent blood loss, transfusion, erythropoietin therapy), fasting blood glucose should be used rather than hemoglobin A1C. An abnormal value of fasting blood glucose or hemoglobin A1C suggests a need for medical consultation. More frequent monitoring may be indicated in the presence of weight change, symptoms of diabetes, or a random measure of blood glucose > 200 mg/dL.

iAdditional information on screening and management of patients with lipid disorders can be found in the AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APhA/ASPC/NLA Guideline on the Management of Blood Cholesterol (Grundy et al. 2018).

jMetabolic syndrome is currently defined by the presence of at least three of the following five risk factors: elevated waist circumference (defined for the United States and Canada as > 102 cm [40.2 inches] for men and > 88 cm [34.6 inches] for women); elevated triglycerides of ≥ 150 mg/dL (or drug treatment for elevated triglycerides, such as fibrates, nicotinic acid, or high-dose omega-3 fatty acids); reduced HDL-C of < 40 mg/dL in men or < 50 mg/dL in women (or drug treatment for reduced HDL-C, such as fibrates or nicotinic acid); elevated blood pressure (BP) with systolic BP ≥ 130 mmHg and/or diastolic BP ≥ 85 mmHg (or antihypertensive treatment in a patient with a history of hypertension); and elevated fasting glucose ≥ 100 mg/dL (or drug treatment for elevated glucose) (Alberti et al. 2009).

kUsing an adverse drug event causality analysis intended to evaluate the risk of sudden death when taking a specific medication (Woosley et al. 2017), the listed drugs have been categorized as prolonging the QT interval and being clearly associated with a known risk of torsades de pointes, even when taken as recommended (Woosley et al. 2009).

lIn this context, risk factors include non-modifiable (e.g., congenital long QT syndrome, age, sex, family history of sudden cardiac death, personal or family history of structural or functional heart disease, personal history of drug-induced QT prolongation, metabolizer status) and modifiable risk factors (e.g., starvation; bradycardia; risk or presence of hypokalemia, hypomagnesemia, or hypocalcemia; excess dose or rapid intravenous infusion of QTc interval–prolonging drugs; simultaneous use of multiple drugs that prolong QTc intervals; or factors affecting drug metabolism such as drug-drug interactions, acute or chronic kidney disease, or hepatic impairment) (Funk et al. 2018).

mScreening should assess changes in libido, menstrual changes, or galactorrhea in women, and changes in libido or in erectile or ejaculatory function in men.

nAssessment can occur through clinical examination or through the use of a structured evaluative tool such as the Abnormal Involuntary Movement Scale (AIMS; Guy 1976; Munetz and Benjamin 1988) or the Dyskinesia Identification System: Condensed User Scale (DISCUS; Kalachnik and Sprague 1993). For a copy of the AIMS, see www.aacap.org/App_Themes/AACAP/docs/member_resources/toolbox_for_clinical_practice_and_outcomes/monitoring/AIMS.pdf, and for a copy of the DISCUS, see https://portal.ct.gov/-/media/DDS/Health/hcsma20002b.pdf.

oPatients at increased risk for developing abnormal involuntary movements include individuals older than 55 years; women; individuals with a mood disorder, substance use disorder, intellectual disability, or central nervous system injury; individuals with high cumulative exposure to antipsychotic medications, particularly high-potency dopamine D2 receptor antagonists; and patients who experience acute dystonic reactions, clinically significant parkinsonism, or akathisia (Carbon et al. 2017, 2018; Miller et al. 2005; Solmi et al. 2018a). Abnormal involuntary movements can also emerge or worsen with antipsychotic cessation.

pFrequency of monitoring for involuntary movements in individuals receiving treatment with an antipsychotic medication is also subject to local regulations in some jurisdictions.

Abbreviations. AIMS = Abnormal Involuntary Movement Scale; ANC = absolute neutrophil count; BMI = body mass index; CBC = complete blood count; CT = computed tomography; DISCUS = Dyskinesia Identification System: Condensed User Scale; ECG = electrocardiography; EEG = electroencephalogram; MRI = magnetic resonance imaging; QTc = corrected QT interval; TSH = thyroid-stimulating hormone.

Suggested physical and laboratory assessments for patients with schizophrenia

Enlarge table

As part of the initial evaluation, it is also useful to inquire about the course and duration of symptoms prior to treatment (i.e., duration of untreated psychosis) (Penttilä et al. 2014; Register-Brown and Hong 2014; Santesteban-Echarri et al. 2017) and whether the patient has received any mental health treatment. If the patient has received treatment previously, it is important to ask about a broad range of treatments and other approaches to addressing the patient’s symptoms and functioning and to specifically ask about the full range of treatment settings (e.g., outpatient, partial hospitalization, inpatient) and approaches that the patient has found helpful or problematic (American Psychiatric Association 2016a). Although most patients will comment on prior medications, psychotherapy, or psychiatric hospitalizations if asked about treatment history, specific questions may be needed to gather details of such treatments. Prompting may be needed to learn information about the patient’s experiences with other interventions such as psychosocial rehabilitation, supported employment, assertive community treatment (ACT), court-ordered treatment, treatment while incarcerated, substance use treatments, neuromodulatory therapies (e.g., electroconvulsive therapy [ECT], transcranial magnetic stimulation [TMS]), 12-step programs, self-help groups, spiritual healers, and complementary or alternative treatment approaches. Pharmacy databases and patients’ lists of active medications are not likely to include long-acting injectable (LAI) medications (e.g., antipsychotics, naltrexone, buprenorphine) or implants (e.g., buprenorphine, contraceptive agents), over-the-counter medications, herbal products, or nutritional supplements. For each specific type of intervention that the patient has received, it is helpful to learn more about the duration, mode of delivery (e.g., formulation, route, and dose for medications; format, type, and frequency of treatment for psychotherapy), response (including tolerability, changes in quality of life, level of functioning, and symptom response/remission), and degree of adherence.

The psychosocial history reviews the stages of the patient’s life and may include attention to perinatal events, delays in developmental milestones, academic history and performance (including learning difficulties, special education interventions, or disciplinary actions), relationship and sexual history, interpersonal functioning (including in social and family roles, such as parenting), occupational history (including military history), legal history, and identification of major life events (e.g., parental loss, divorce, traumatic experiences, migration history) and psychosocial stressors (e.g., financial, housing, legal, school/occupational, or interpersonal/relationship problems; lack of social support; painful, disfiguring, or terminal medical illness) (American Psychiatric Association 2016a; Barnhill 2014; MacKinnon et al. 2016; Smith et al. 2019). Information about the patient’s family constellation and persons who provide support will serve as a foundation for working collaboratively with the patient and their support network. The cultural history also emphasizes relationships, both familial and nonfamilial, and the role of important cultural and religious influences on the patient’s life (Aggarwal and Lewis-Fernández 2015; American Psychiatric Association 2013a; Lewis-Fernández et al. 2016).

The mental status examination is an integral part of the initial assessment. A full delineation of the mental status examination is beyond the scope of this document, and detailed information on conducting the examination is available elsewhere (American Psychiatric Association 2016a; Barnhill 2014; MacKinnon et al. 2016; Smith et al. 2019; Strub and Black 2000). However, for individuals with possible schizophrenia, a detailed inquiry into hallucinations and delusions will often identify psychotic experiences in addition to the presenting concerns. Negative symptoms and cognitive impairment are common and influence outcomes (Bowie et al. 2006; Green 2016; Jordan et al. 2014; Rabinowitz et al. 2012; Santesteban-Echarri et al. 2017) but may go undetected without specific attention during the evaluation. Negative symptoms also can be difficult to differentiate from lack of interest or reduced motivation due to depression, medication side effects, substance use, or neurological conditions.

Insight is also impaired in a significant proportion of individuals with schizophrenia (Mohamed et al. 2009) and can manifest as a decreased awareness of having a disorder, symptoms, consequences of illness, or a need for treatment (Mintz et al. 2003). Consequently, inquiring about the patient’s degree of insight and judgment will provide information relevant to risk assessment, treatment outcomes, and adherence (Mintz et al. 2003; Mohamed et al. 2009).

Risk assessment is another essential part of the initial psychiatric evaluation (American Psychiatric Association 2004). It requires synthesizing information gathered in the history and mental status examination and identifying modifiable risk factors for suicidal or aggressive behaviors that can serve as targets of intervention in constructing a plan of treatment. Suicidal ideas are common in individuals who have had a psychotic experience (Bromet et al. 2017). Death due to suicide has been estimated to occur in about 4%–10% of individuals with schizophrenia (Drake et al. 1985; Heilä et al. 2005; Hor and Taylor 2010; Inskip et al. 1998; Laursen et al. 2014; Nordentoft et al. 2011; Palmer et al. 2005; Popovic et al. 2014; Tanskanen et al. 2018; Yates et al. 2019), yielding a greater than tenfold increase in standardized mortality ratios (Saha et al. 2007). Among individuals with schizophrenia, suicide attempts and suicide may be more common early in the course of the illness (Popovic et al. 2014) and can occur even before initial treatment for psychosis (Challis et al. 2013).

In individuals with schizophrenia, many of the risk factors that contribute to the risks of suicidal or aggressive behaviors are the same as factors increasing risk in other disorders. For example, in individuals with schizophrenia, an increased risk of suicidal or aggressive behaviors has been associated with male sex, expressed suicidal ideation, a history of attempted suicide or other suicide-related behaviors, and the presence of alcohol use disorder or other substance use disorder (Cassidy et al. 2018; Challis et al. 2013; Fazel et al. 2009a, 2014; Fleischhacker et al. 2014; Hawton et al. 2005; Hor and Taylor 2010; Østergaard et al. 2017; Pompili et al. 2007; Popovic et al. 2014; Roché et al. 2018; Sariaslan et al. 2016; Singh et al. 2012; Swanson et al. 2006; Witt et al. 2013, 2014). Firearm access is an additional contributor to suicide risk (Alban et al. 2018; Anestis and Houtsma 2018; Siegel and Rothman 2016). Additional factors that have been identified as increasing risk for suicide among individuals with schizophrenia include depressive symptoms, hopelessness, agitation or motor restlessness, fear of mental disintegration, recent loss, recency of diagnosis or hospitalization, repeated hospitalizations, high intelligence, young age, and poor adherence to treatment (Cassidy et al. 2018; Fleischhacker et al. 2014; Hawton et al. 2005; Lopez-Morinigo et al. 2014; Pompili et al. 2007; Popovic et al. 2014; Randall et al. 2014). It is not clear whether preserved insight is associated with an increase in suicide risk among individuals with schizophrenia (Hor and Taylor 2010) or whether this is an apparent increase that is mediated by other factors such as hopelessness (López-Moríñigo et al. 2012).

Although reduced risk of suicide was associated with hallucinations in one meta-analysis (Hawton et al. 2005), the presence of auditory command hallucinations may confer increased risk (Harkavy-Friedman et al. 2003; Wong et al. 2013). Command hallucinations can also be relevant when assessing individuals for a risk of aggressive behaviors (McNiel et al. 2000; Swanson et al. 2006), although the relationship between experiencing commands and acting on them is complex (Braham et al. 2004). Persecutory delusions may also contribute to risk of aggression, particularly in the absence of treatment or in association with significant anger (Coid et al. 2013; Keers et al. 2014; Swanson et al. 2006).

Among individuals with psychotic illnesses, prior suicidal threats, angry affect, impulsivity, hostility, recent violent victimization, childhood sexual abuse, medication nonadherence, and a history of involuntary treatment were also associated with an increased risk of aggressive behavior (Buchanan et al. 2019; Large and Nielssen 2011; Reagu et al. 2013; Swanson et al. 2006; Witt et al. 2013, 2014). Other factors associated with a risk of aggression are similar to findings in individuals without psychosis and include male sex, young age, access to firearms, the presence of substance use, traumatic brain injury, a history of attempted suicide or other suicide-related behaviors, and prior aggressive behavior, including that associated with legal consequences (Buchanan et al. 2019; Cassidy et al. 2018; Fazel et al. 2009a, 2009b, 2014; Fleischhacker et al. 2014; Large and Nielssen 2011; Monuteaux et al. 2015; Østergaard et al. 2017; Popovic et al. 2014; Roché et al. 2018; Sariaslan et al. 2016; Short et al. 2013; Singh et al. 2012; Swanson et al. 2006; Witt et al. 2013, 2015).

Balancing of Potential Benefits and Harms in Rating the Strength of the Guideline Statement

Benefits

In an individual with a possible psychotic disorder, a detailed assessment is important in establishing a diagnosis, recognizing co-occurring conditions (including substance use disorders, other psychiatric disorders, and other physical health disorders), identifying psychosocial issues, and developing a plan of treatment that can reduce associated symptoms, morbidity, and mortality.

Harms*

*Harms may include serious adverse events; less serious adverse events that affect tolerability; minor adverse events; negative effects of the intervention on quality of life; barriers and inconveniences associated with treatment; and other negative aspects of the treatment that may influence decision-making by the patient, the clinician, or both.

Some individuals may become anxious, suspicious, or annoyed if asked multiple questions during the evaluation. This could interfere with the therapeutic relationship between the patient and the clinician. Another potential consequence is that time used to focus on a detailed assessment (as outlined in the Practice Guidelines for the Psychiatric Evaluation of Adults, 3rd edition; American Psychiatric Association 2016a) could reduce time available to address other issues of importance to the patient or of relevance to diagnosis and treatment planning.

Patient Preferences

Although there is no specific evidence on patient preferences related to assessment in individuals with a possible psychotic disorder, clinical experience suggests that the majority of patients are cooperative with and accepting of these types of questions as part of an initial assessment.

Balancing of Benefits and Harms

The potential benefits of this guideline statement were viewed as far outweighing the potential harms. This recommendation is also consistent with the APA Practice Guidelines for the Psychiatric Evaluation of Adults, 3rd edition (American Psychiatric Association 2016a). The level of research evidence is rated as low because there is minimal research on the benefits and harms of assessing these aspects of history and examination as part of an initial assessment. Nevertheless, expert opinion suggests that conducting such assessments as part of the initial psychiatric evaluation improves the diagnosis and treatment planning in individuals with a psychiatric disorder. For additional details, see the Practice Guidelines for the Psychiatric Evaluation of Adults. For additional discussion of the research evidence, see Appendix C, Statement 1.

Differences of Opinion Among Writing Group Members

There were no differences of opinion. The writing group voted unanimously in favor of this recommendation.

Review of Available Guidelines From Other Organizations

Relevant guidelines from the following organizations were reviewed: British Association for Psychopharmacology (BAP), Canadian Psychiatric Association and Schizophrenia Society of Canada, National Institute for Health and Care Excellence (NICE), Royal Australian and New Zealand College of Psychiatry (RANZCP), Scottish Intercollegiate Guidelines Network (SIGN), World Federation of Societies of Biological Psychiatry (WFSBP), and Schizophrenia Patient Outcomes Research Team (PORT). Information from them is generally consistent with this guideline statement. Other guidelines on the treatment of schizophrenia incorporate recommendations related to the need for a comprehensive initial assessment (Addington et al. 2017a; National Institute for Health and Care Excellence 2014), including identification of prior and current psychiatric symptoms and diagnoses (Addington et al. 2017a; Hasan et al. 2015; National Institute for Health and Care Excellence 2014), assessment of tobacco use (Addington et al. 2017a; National Institute for Health and Care Excellence 2014), assessment of substance use (Addington et al. 2017a; Barnes et al. 2011; Crockford and Addington 2017; Galletly et al. 2016; National Institute for Health and Care Excellence 2014), physical health history and examination (Addington et al. 2017a; National Institute for Health and Care Excellence 2014), assessment of psychosocial factors (Addington et al. 2017a; Galletly et al. 2016; National Institute for Health and Care Excellence 2014), and mental status examination (Addington et al. 2017a), including assessment of the risk of harm to self or others (Addington et al. 2017a; Hasan et al. 2015; National Institute for Health and Care Excellence 2014). Several other guidelines also provide information on the circumstances in which an electrocardiogram is suggested (Barnes et al. 2011; National Institute for Health and Care Excellence 2014; Pringsheim et al. 2017).

Quality Measurement Considerations

For patients with psychotic disorders, including schizophrenia, several components of the initial psychiatric evaluation have potential relevance for quality measure development, although such quality measures do not exist at present. A first step toward development of scientifically sound quality measures is identification of discrete indicators that signal the delivery of high-quality care. This step may be challenging to accomplish given the breadth of content within the initial psychiatric assessment and the difficulty in ascertaining evaluation details from chart or administrative data. However, it may still be possible to use available evidence and expert-recommended consensus to develop and specify electronic and clinical data registry quality measures. Additionally, as discussed in the APA Practice Guidelines for the Psychiatric Evaluation of Adults, 3rd edition (American Psychiatric Association 2016a), quality improvement efforts at the local level could assess whether specific aspects of the evaluation such as a risk assessment were completed while still allowing flexibility in the documentation of findings.

Statement 2: Use of Quantitative Measures

APA recommends (1C) that the initial psychiatric evaluation of a patient with a possible psychotic disorder include a quantitative measure to identify and determine the severity of symptoms and impairments of functioning that may be a focus of treatment.

Implementation

APA’s Practice Guidelines for the Psychiatric Evaluation of Adults, 3rd edition (American Psychiatric Association 2016a) provides a general description of the use of quantitative measures as part of the initial psychiatric evaluation. In the assessment of a patient with a possible psychotic disorder, quantitative measures can also be used to help detect and determine the severity of psychosis and associated symptoms. The intent of using a quantitative measure is not to establish a diagnosis but rather to complement other aspects of the screening and assessment process. Depending on the measure, it can aid in treatment planning by providing a structured replicable way to document the patient’s baseline symptoms. It can also help to determine which symptoms should be the target of intervention on the basis of such factors as frequency of occurrence, magnitude, potential for associated harm to the patient or others, and associated distress to the patient.

As treatment proceeds, use of quantitative measures allows more precise tracking of whether nonpharmacological and pharmacological treatments are having their intended effect or whether a shift in the treatment plan is needed. This record of a patient’s response to treatment is of particular value when the treatment is nonstandard (e.g., combination of antipsychotics) or expensive. It can also provide helpful information about the actual effects of prior treatments. In addition, patients’ ratings can be compared with family members’ impressions of treatment effects to clarify the longitudinal course of the patient’s illness.

Much of the treatment-related research in psychiatry has used clinician-rated scales to determine patient outcomes; however, patient-rated scales are typically less time-consuming to administer than clinician-rated scales. In addition, they provide important insights into the patient’s experience that support person-centered care. The use of anchored, self-rated scales with criteria to assess the severity and frequency of symptoms can also help patients become more informed self-observers. However, correlations between patient- and clinician-rated scales are often modest (Harvey 2011; Spitz et al. 2017), suggesting that both types of quantitative measures provide useful information. If a mismatch is noted in the self-assessment of patients as compared with assessments of other observers, this may provide information relevant to outcomes. The accuracy of self-assessments of ability, skills, performance, or decisions (also termed introspective accuracy) is a better predictor of everyday functional deficits than objective measures of neurocognitive or social cognitive performance (Silberstein and Harvey 2019).

The exact frequency at which measures are warranted will depend on clinical circumstances. Use of quantitative measures over time will help assure that key elements of information are collected to guide treatment (Lewis et al. 2019). Consequently, it is preferable to use a consistent approach to quantitative measurement for a given patient because each rating scale defines and measures psychosis and other symptoms differently.

Although recommending a particular scale, patient- or clinician-rated, is outside the scope of this practice guideline, a number of objective, quantitative rating scales to monitor clinical status in schizophrenia are available (American Psychiatric Association 2013a; Rush et al. 2008). The Clinician-Rated Dimensions of Psychosis Symptom Severity (American Psychiatric Association 2013b), which is included in DSM-5 for further research and clinical evaluation, contains eight domains that are rated on a scale from 0 (not present) to 4 (present and severe) on the basis of symptoms in the prior 7 days. A 6-item version of the Positive and Negative Syndrome Scale (PANSS-6; Bech et al. 2018; Østergaard et al. 2018b) consists of the PANSS items for delusions, conceptual disorganization, hallucinations, blunted affect, social withdrawal, and lack of spontaneity and flow of conversation (Kay et al. 1987). Data on the PANSS-6 suggest that it correlates highly with scores on the 30-item version of the Positive and Negative Syndrome Scale (PANSS-30; Østergaard et al. 2018a, 2018b). Furthermore, the PANSS-6 is sensitive to changes with treatment and able to identify symptom remission with a high degree of accuracy (Østergaard et al. 2018a, 2018b) if individuals who are performing ratings are appropriately trained (Opler et al. 2017). Clinician-rated scales may also be selected to assess specific clinical presentations, such as using the Bush-Francis Catatonia Rating Scale for individuals with catatonic features (Bush et al. 1996a). Other clinician-rated scales are commonly used for monitoring psychopathology in research but are likely to be too lengthy for routine clinical use. These include the PANSS-30 (Kay et al. 1987), the Scale for the Assessment of Negative Symptoms (SANS; Andreasen 1984a), the Scale for the Assessment of Positive Symptoms (SAPS; Andreasen 1984b), and the Brief Psychiatric Rating Scale (BPRS; https://mha.ohio.gov/Portals/0/assets/HealthProfessionals/About MH and Addiction Treatment/First Episode Psychosis/Brief-Psychiatric-Rating-Scale.pdf) (Leucht et al. 2005; Overall and Gorham 1962; Ventura et al. 1993).

In terms of patient-rated scales, the DSM-5 Self-Rated Level 1 Cross-Cutting Symptom Measure—Adult (American Psychiatric Association 2013c) includes a total of 23 items in 13 domains, with only 2 items related to psychosis. Nevertheless, it may be useful for identifying and tracking symptoms other than psychosis, including those related to co-occurring disorders. DSM-5 also includes 36-item self- and proxy-administered versions of the World Health Organization Disability Schedule 2.0 (WHODAS 2.0) for assessing functioning difficulties due to health and mental health conditions (American Psychiatric Association 2013d; Üstün et al. 2010). Other options for assessing functioning include the Social and Occupational Functioning Assessment Scale (SOFAS; American Psychiatric Association 2000) and the Personal and Social Performance scale (Morosini et al. 2000). Several versions of Patient-Reported Outcomes Measurement Information System (PROMIS) scales, which address social roles and functioning, are also available (www.healthmeasures.net/explore-measurement-systems/promis). The use of ratings from other informants is particularly helpful in assessing the patient’s level of functioning because individuals with schizophrenia often have a different view of their functioning than do family members or others involved in their lives (Harvey 2011).

For a nonspecific measure of quality of life, patients can be asked to rate their overall (physical and mental) quality of life in the past month on a scale from 0 (“about as bad as dying”) to 10 (“life is perfect”) (Unützer et al. 2002). In individuals with chronic mental illness, the Satisfaction With Life Scale (Diener et al. 1985) has been developed and used to assess life satisfaction and quality of life. Quality of life can also be measured using a scale developed by the World Health Organization, the WHOQOL-BREF (Skevington et al. 2004; WHOQOL Group 1998) (http://depts.washington.edu/seaqol/WHOQOL-BREF). The Centers for Disease Control and Prevention Healthy Days Measure (HRQOL-14) and Healthy Days Core Module (HRQOL-4) (www.cdc.gov/hrqol/hrqol14_measure.htm) have also been used in general population samples to assess physical and emotional symptoms as related to an individual’s perceived sense of well-being (Moriarty et al. 2003).

Rating scales should always be implemented in a way that supports developing and maintaining the therapeutic relationship with the patient. Reviewing scale results with the patient can help foster a collaborative dialogue about progress toward symptom improvement, functioning gains, and recovery goals. Such review may help clinicians, patients, families, and other support persons recognize that improvement is taking place or, conversely, identify issues that need further attention.

If more than one quantitative measure is being used, it is important to minimize duplication of questions and avoid overwhelming the patient with an excessive number of scales to complete. In addition, when choosing among available quantitative measures, objectives of scale use (e.g., screening, documenting baseline symptoms, ongoing monitoring) should be considered. Optimal scale properties (e.g., sensitivity, specificity) will differ depending on the desired purpose, but assessments of scale validity and reliability are typically conducted cross-sectionally in research contexts.

Because many scales ask the patient to rate symptoms over several weeks, they may not be sensitive to change. This can be problematic in acute care settings, where treatment adjustments and symptom improvement can occur fairly quickly. Some symptom-based quantitative measures focus either on symptom frequency over the observation period or on symptom severity. Although these features often increase or decrease in parallel, that is not invariably the case. Other quantitative measures ask the patient to consider both symptom frequency and severity, which can also make the findings difficult to interpret.

Other factors that can affect the statistical reliability and validity of rating scale measures include comorbid illnesses and patient age, language, race, ethnicity, cultural background, literacy, and health literacy. These factors and others can lead patients to misinterpret questions or bias the ratings that they record, either unintentionally (e.g., to please the clinician with their progress) or intentionally (e.g., to obtain controlled substances, to support claims of disability). Thus, the answers to questions and the summative scores on quantitative measures need to be interpreted in the context of the clinical presentation.

The type and extent of quantitative measures used will also be mediated by the clinical setting, the time available for evaluation, and the urgency of the situation. In some clinical contexts, such as a planned outpatient assessment, patients may be asked to complete electronic- or paper-based quantitative measures, either prior to the visit or on arrival at the office (Allen et al. 2009; Harding et al. 2011). Between or prior to visits, electronic approaches (e.g., mobile phone applications, clinical registries, patient portal sites in electronic health records) may also facilitate obtaining quantitative measurements (Lewis et al. 2019; Palmier-Claus et al. 2012; K. Wang et al. 2018). In other clinical contexts, such as acute inpatient settings, electronic modes of data capture may be more cumbersome, and patients may need more assistance in completion of scales. As an alternative, printed versions of scales may be completed by the patient or a proxy or administered by the clinician. In other clinical circumstances, however, printed or electronic versions of quantitative scales may not be readily available or information may not be available to complete all scale items. In emergency settings, use of a quantitative rating scale may need to be postponed until the acute crisis has subsided or until the patient’s clinical status permits a detailed examination.

Although available information suggests that ambulatory patients are generally cooperative, some individuals may be unwilling to complete quantitative measures (Narrow et al. 2013). Severe symptoms, co-occurring psychiatric conditions, low health literacy, reading difficulties, or cognitive impairment may limit some patients’ ability to complete self-report instruments (Harding et al. 2011; Valenstein et al. 2009; Zimmerman et al. 2011). In these circumstances, it may be necessary to place greater reliance on collateral sources of information such as family members, other treating health professionals, or staff members of community residence programs, if applicable. If collateral sources of information are not immediately available, treatment may also need to proceed, with adjustments in the plan, if indicated, as additional knowledge is gained. If time constraints are present, the clinician may wish to focus on rating of relevant target symptoms (e.g., on a Likert scale). In emergent circumstances, safety of the patient and others must take precedence; the initial assessment may need to be brief, with a more detailed assessment and incorporation of quantitative measures once the acute clinical situation has been stabilized.

Balancing of Potential Benefits and Harms in Rating the Strength of the Guideline Statement

Benefits

Clinical decision-making, including but not limited to diagnosis and treatment planning, requires a careful and systematic assessment of the type, frequency, and magnitude of psychiatric symptoms as well as an assessment of the impact of those symptoms on the patient’s day-to-day functioning and quality of life. Intuitively, and by analogy with other medical specialties in which treatment is guided by standardized measurement (e.g., of physiological signs or laboratory tests), the use of a systematic and quantifiable approach to assessment would seemingly produce better patient outcomes and greater standardization of care across patients. As electronic health records become more commonly used, electronic capture of quantitative measures can facilitate use of computerized decision-support systems in guiding evidence-based treatment, catalyzing additional improvements in outcomes and quality of care.

Use of a quantitative measure as part of the initial evaluation can establish baseline information on the patient’s symptoms and level of functioning and can help determine specific targets of treatment in the context of shared decision-making. When administered through paper-based or electronic self-report and as compared with a clinical interview, a quantitative measure may help the clinician to conduct a more consistent and comprehensive review of the multiplicity of symptoms that the patient may be experiencing. Using systematic measures may also increase the efficiency of asking routine questions and allow more time for clinicians to focus on symptoms of greatest severity or issues of most concern to the patient. Such measures may also facilitate collection of information from the patient’s family or other collateral informants on factors such as symptoms or functioning. When used on a longitudinal basis, quantitative measures can help determine whether nonpharmacological and pharmacological treatments are having their intended effect or whether a shift in the treatment plan is needed to address symptoms, treatment-related side effects, level of distress, functioning impairments, or potential for harm to the patient or others.

Without the use of a consistent quantitative measure, recall biases may confound the ability of patients and clinicians to compare past and current levels or patterns of symptoms and functioning. When patients have had substantial improvements in symptoms and functioning, it can be easy to focus on the improvements and overlook residual symptoms or side effects of treatment that are contributing to ongoing impairment or reduced quality of life. Thus, ongoing use of quantitative assessments may foster identification of residual symptoms or impairments and early detection of illness recurrence. Systematic use of quantitative measures can also facilitate communication among treating clinicians and can serve as a basis for enhanced management of populations of patients as well as individual patients. Although mobile apps may be capable of assisting with quantitative measurement, there is no current evidence on which to base recommendations about use of mobile apps in the treatment of schizophrenia.

Harms

The harms of using a quantitative measure include the time required for administration and review. The amount of time available for an initial psychiatric evaluation is typically constrained by clinician availability, cost, and other factors. Under such circumstances, time that is used to obtain quantitative measures could introduce harms by reducing time available to address other issues of importance to the patient or of relevance to clinical decision-making. Overreliance on quantitative measures may also cause other aspects of the patient’s symptoms and clinical presentation to be overlooked.

Some patients may view quantitative measures as impersonal or may feel annoyed by having to complete detailed scales, particularly if done frequently. If a patient feels negatively about quantitative measures, this could alter the therapeutic alliance. In addition, some patients may have difficulty completing self-report scales or may interpret questions incorrectly. Patients may also provide inaccurate information about their symptoms, and relying on inaccurate information can have a negative impact on clinical decision-making, including recommendations for treatment.

Systematic use of measures may require changes in workflow or staffing to distribute scales, increase time needed to review results with the patient, or lead to unreimbursed costs (e.g., to integrate measures into electronic health record systems, to pay to use copyrighted versions of scales).

Patient Preferences

Clinical experience suggests that the majority of patients are cooperative with and accepting of quantitative measures as part of an initial or subsequent assessment. Most patients will be able to appreciate the ways in which the use of quantitative measures will be of benefit to them. For example, in the testing of the DSM-5 Cross-Cutting Symptom Measure as part of the DSM-5 field trials, quantitative measures were found to be acceptable to patients (Clarke et al. 2014; Mościcki et al. 2013), and only a small fraction of individuals felt that measurement of symptoms would not be helpful to their treating clinician (Mościcki et al. 2013).

The fact that the clinician is using a systematic approach to address the patients’ symptoms and functioning sends a positive message that could improve the therapeutic relationship. Especially in developed countries, patients are used to and expect digital, computerized information exchange, including for health-related monitoring and communication. For these patients, the use of quantitative measures within the context of an electronic health record, mobile app, or other computerized technology may be more convenient.

Balancing of Benefits and Harms

The potential benefits of this guideline statement were viewed as far outweighing the potential harms. This recommendation is also consistent with Guideline VII, “Quantitative Assessment,” in the APA’s Practice Guidelines for the Psychiatric Evaluation of Adults, 3rd edition (American Psychiatric Association 2016a). Although quantitative measures have been used for reporting purposes as well as research, the level of research evidence for this recommendation is rated as low because it remains unclear whether routine use of these scales in clinical practice improves overall outcomes. Nonetheless, expert opinion suggests that use of quantitative measures will enhance clinical decision-making and improve treatment outcomes. For additional discussion of the research evidence, see Appendix C, Statement 2.

There is minimal research on the harms of using quantitative measures as part of the psychiatric evaluation as compared with assessment as usual. However, expert opinion suggests that harms of assessment are minimal compared with the benefits of such assessments in improving identification and assessment of psychiatric symptoms. For additional details, see the APA’s Practice Guidelines for the Psychiatric Evaluation of Adults, 3rd edition (American Psychiatric Association 2016a).

Differences of Opinion Among Writing Group Members

There were no differences of opinion. The writing group voted unanimously in favor of this recommendation.

Review of Available Guidelines From Other Organizations

Multiple guidelines from other organizations were reviewed (Addington et al. 2017a, 2017b; Barnes et al. 2011; Buchanan et al. 2010; Crockford and Addington 2017; Galletly et al. 2016; Hasan et al. 2012, 2013, 2015; National Institute for Health and Care Excellence 2014; Norman et al. 2017; Pringsheim et al. 2017; Scottish Intercollegiate Guidelines Network 2013). None of these guidelines specifically recommend using quantitative measures as part of the initial assessment in individuals with schizophrenia, but several guidelines (Barnes et al. 2011; Galletly et al. 2016) do recommend use of rating scales under some circumstances at baseline or as part of ongoing monitoring.

Quality Measurement Considerations

There is insufficient consensus on the most appropriate quantitative measures (i.e., rating scales) to use in assessing individuals with psychotic disorders, including schizophrenia. Nevertheless, data do support the use of clinician-reported, symptom-based ratings to guide treatment. Patient-rated scales may be clinically useful in identifying patient concerns or subjective experiences (e.g., medication side effects). On the basis of these potential benefits, a process-focused internal or health system–based quality improvement measure could determine rates of quantitative measure use, and quality improvement initiatives could be implemented to increase the frequency with which such measures are used in individuals with schizophrenia.

Statement 3: Evidence-Based Treatment Planning

APA recommends (1C) that patients with schizophrenia have a documented, comprehensive, and person-centered treatment plan that includes evidence-based nonpharmacological and pharmacological treatments.

Implementation

When treating individuals with schizophrenia, a person-centered treatment plan should be developed, documented in the medical record, and updated with the patient at appropriate intervals. A person-centered treatment plan can be recorded as part of an evaluation note or progress note and does not need to adhere to a defined development process (e.g., face-to-face multidisciplinary team meeting) or format (e.g., time-specified goals and objectives). Depending on the urgency of the initial clinical presentation, the availability of laboratory results, and other sources of information, the initial treatment plan may need to be augmented over several visits as more details of history and treatment response are obtained.

As treatment proceeds, the treatment plan will require iterative reevaluation and adjustment prompted by such factors as inadequate treatment response, difficulties with tolerability or adherence, impairments in insight, changes in presenting issues or symptoms, or revisions in diagnosis. In adapting treatment to the needs of the individual patient, tailoring of the treatment plan may also be needed on the basis of sociocultural or demographic factors, with an aim of enhancing quality of life or aspects of functioning (e.g., social, academic, occupational). Factors that influence medication metabolism (e.g., age, sex, body weight, renal or hepatic function, smoking status, use of multiple concurrent medications) may also require adjustments to the treatment plan in terms of either typical medication doses or frequency of monitoring. For most individuals with schizophrenia, it is challenging to piece together a coherent picture of the patient’s longitudinal course from medical records. Thus, it is important to note the rationale for any changes in the treatment plan as well as the specific changes that are being made because an accurate history of past and current treatments and responses to them is a key part of future treatment planning.

Aims of Treatment Planning

The overarching aims of treatment planning are 1) to promote and maintain recovery, 2) to maximize quality of life and adaptive functioning, and 3) to reduce or eliminate symptoms. To achieve these aims, it is crucial to identify the patient’s aspirations, goals for treatment, and treatment-related preferences. Depending on prevailing state laws, psychiatric advance directives are one approach to encouraging patients to contemplate and state their preferences about treatment choices (Easter et al. 2017; Kemp et al. 2015; Shields et al. 2014; Wilder et al. 2010). For patients who have completed a psychiatric advance directive, wellness recovery action plan (Copeland 2000), or individualized crisis prevention or safety plan (Stanley and Brown 2012, 2019; Stanley et al. 2018), these documents will be important to review with the patient when crafting a person-centered approach to care. Discussions with the patient, other treating health professionals, family members, and others involved in the patient’s life can each be vital in developing a full picture of the patient and formulating a person-centered treatment plan, using shared decision-making whenever possible. The patient and others may express opinions about specific treatment approaches or identify practical barriers to the patient’s ability to participate in treatment, such as lack of insight, cognitive impairments, disorganization, or inadequate social resources.

Elements of the Treatment Plan

Depending on the clinical circumstances and input from the patient and others, a comprehensive and person-centered treatment plan will typically delineate treatments aimed at improving functioning, reducing positive and negative symptoms, and addressing co-occurring psychiatric symptoms or disorders. In each of these respects, it is essential to consider both nonpharmacological and pharmacological treatment approaches and recognize that a combination of nonpharmacological and pharmacological treatments will likely be needed to optimize outcomes. Other elements of the treatment plan may include the following:

  • determining the most appropriate treatment setting

  • delineating plans for addressing risks of harm to self or others (if present)

  • addressing barriers to adherence

  • engaging family members and others involved in the patient’s life

  • providing information to patients, family members, and others involved in the patient’s life about treatment options, early symptoms of relapse, need for ongoing monitoring, coping strategies, case management services, and community resources, including peer-support programs

  • incorporating goals of treatment related to

    • social support networks

    • interpersonal, family, or intimate relationships

    • parenting status

    • living situation

    • past trauma or victimization

    • school or employment

    • financial considerations, including disability income support, when indicated

    • insurance status

    • legal system involvement

  • identifying additional needs for

    • history or mental status examination

    • physical examination (either by the evaluating clinician or by another health professional)

    • laboratory testing, imaging, electrocardiography (ECG), or other clinical studies (if indicated on the basis of the history, examination, and planned treatments)

  • collaborating with other treating clinicians (including provision of integrated care) to avoid fragmentation of treatment efforts and assure that co-occurring substance use disorders and physical health conditions are managed

Engagement of Family Members and Other Persons of Support

Discussions with the patient, family members, and others will typically occur as part of the initial assessment (see Statement 1), and additional input will be needed as treatment proceeds and the treatment plan is updated. Family members and others involved in the patient’s life may also express specific concerns about the individual’s symptoms or behaviors, which, if present, should be documented and addressed. Most individuals welcome involvement of family members and other persons of support (Cohen et al. 2013; Drapalski et al. 2018; Mueser et al. 2015), and family members can be an important part of the care team. Family members can also be provided with educational materials or directed to organizations that offer education to family members and other persons of support (Mental Health America 2019; National Alliance on Mental Illness 2019a).

Strategies to Promote Adherence

Strategies to promote adherence are always important to consider when developing a patient-centered treatment plan (Ferrando et al. 2014). Maintaining adherence to treatment is often challenging (Acosta et al. 2012; Shafrin et al. 2016; Valenstein et al. 2006), and poor adherence is associated with poor outcomes, including increased risks of relapse, rehospitalization, suicidal and aggressive behaviors, and mortality (Bowtell et al. 2018; Cassidy et al. 2018; Goff et al. 2017; Hawton et al. 2005; Hui et al. 2018; Kishi et al. 2019; Leucht et al. 2012; Thompson et al. 2018; Tiihonen et al. 2018; Vermeulen et al. 2017; Witt et al. 2013). Treatment planning to address adherence will depend on the specific contributing factors and whether reduced adherence is related to medication use, missed appointments, or other aspects of treatment. Issues that may influence adherence include, but are not limited to, lack of awareness of illness, forgetting to take doses, difficulties managing complex regimens (e.g., due to cognitive impairment, frequency of doses, or number of medications), side effects that are of particular importance to the patient (e.g., weight gain, akathisia, sexual dysfunction, effects on cognition), financial barriers (e.g., cost, insurance coverage), perceived risks and benefits of treatment, insufficient understanding of medication benefits for symptoms that are important to the patient, ambivalence or suspiciousness of medications or treatment in general, lack of a perceived need for treatment (e.g., due to feeling good or not viewing self as ill; due to personal, religious, or cultural beliefs), co-occurring conditions (e.g., depression; alcohol, cannabis, or other substance use disorder), high levels of hostility, persecutory delusions, prior difficulties with adherence, prior experiences with treatment (e.g., effectiveness, side effects), limited geographic availability or accessibility of services, financial or insurance constraints on medications or visits, difficulties in the therapeutic relationship, lack of support from significant others for treatment, cultural or family beliefs about illness or treatment, and perceptions of stigma about having an illness and taking medication (Acosta et al. 2012; Ascher-Svanum et al. 2006; Czobor et al. 2015; Foglia et al. 2017; García et al. 2016; Haddad et al. 2014; Hartung et al. 2017; Hatch et al. 2017; Higashi et al. 2013; Kane et al. 2013; MacEwan et al. 2016a; Mueser et al. 2015; Pyne et al. 2014; Shafrin et al. 2016; Velligan et al. 2017; Volavka et al. 2016; Wade et al. 2017). Adherence with appointments can also be influenced by financial barriers, difficulties scheduling visits around work or school schedules, or issues with transportation or with childcare. Addressing these barriers as part of the treatment plan will require active collaboration and problem-solving between the clinician and patient, often with input from the patient’s family and others involved in the patient’s’ life (Mueser et al. 2015).

When assessing adherence, it is important to take a patient-centered approach in inquiring in a nonjudgmental way whether the individual has experienced difficulties with taking medication (Haddad et al. 2014). Obtaining information from patient diaries, patient-completed rating scales, pharmacy records, family members, or other collateral sources of information can be useful supplements to subjective patient reporting (Acosta et al. 2012; Haddad et al. 2014; Hatch et al. 2017; Kane et al. 2013). Tablet counts, monitoring using electronic pill bottle caps, and drug formulations with implanted sensors have also been used to assess adherence with antipsychotic medications (Acosta et al. 2012; Haddad et al. 2014). It can also be useful to obtain medication blood levels. Levels of clozapine have been best studied, but blood levels of other antipsychotic medications are also available. Although the utility of routine therapeutic monitoring is unclear for antipsychotic medications other than clozapine, blood levels may help in establishing whether a patient is taking the medication (Hiemke et al. 2018; Horvitz-Lennon et al. 2017; Lopez and Kane 2013; Lopez et al. 2017; McCutcheon et al. 2018; Predmore et al. 2018). Urine levels of antipsychotic medications can also be used to assess for adherence (Velligan et al. 2006).

In terms of enhancing adherence, a wide range of approaches have been tried. However, evidence on the most effective techniques remains limited (Hartung et al. 2017), and different approaches will likely be needed for different patients. A checklist that includes barriers, facilitators, and motivators for adherence has been developed and may be helpful in promoting discussion and identifying adherence-related factors in individual patients (Pyne et al. 2014). In addition to conducting ongoing monitoring of adherence as treatment proceeds, it can be helpful to focus on optimizing treatment efficacy, addressing side effects and concerns about treatment, adjusting dosing to minimize side effects while maintaining efficacy, providing information about the illness and its treatments, engaging in shared decision-making, fostering a strong therapeutic alliance, and engaging family members and other community and social supports, as appropriate (Acosta et al. 2012; Haddad et al. 2014; Hamann and Heres 2019; Kane et al. 2013; Rezansoff et al. 2017).

For some patients, the formulation of the antipsychotic medication may influence adherence (see Statement 4, Table 3). For example, rapidly dissolving tablets, oral concentrates, or LAI formulations may be preferable for patients who have difficulty swallowing pills or who are ambivalent about medications and inconsistent in swallowing them. For individuals who have difficulty remembering to take medication, LAI formulations of medications can be used, oral medication regimens can be simplified to reduce the number of pills or daily doses, watches or cell phone alarms can be used as reminders to take medications, pillboxes may be filled with the week’s medication, and family or significant others may be enlisted to assist with medication if cognitive impairments are present. Another approach that can be used to improve adherence is behavioral tailoring, which involves cuing oneself to take medications by incorporating adherence into one’s daily routine (Kreyenbuhl et al. 2016; Mueser et al. 2002). If financial issues with medications are affecting adherence, reassessment of the treatment regimen may be needed, or patients’ assistance programs may be pursued (e.g., through pharmaceutical company programs or a discount program such as GoodRx, www.goodrx.com). When a patient does not appear for appointments or is nonadherent in other ways, assertive outreach such as telephone calls or secure messages may be helpful in reengaging the patient in treatment.

Addressing Risks for Suicidal and Aggressive Behavior

Identifying risk factors and estimating risks for suicidal and aggressive behaviors are essential parts of psychiatric evaluation (American Psychiatric Association 2016a and as described in detail in Statement 1, subsection “Implementation”). Despite identification of these risk factors, it is not possible to predict whether an individual patient will engage in aggressive behaviors or attempt or die by suicide. However, when an increased risk for such behaviors is present, it is important that the treatment plan reevaluates the setting of care and implements approaches to target and reduce modifiable risk factors. Although demographic and historical risk factors are static, potentially modifiable risk factors may include poor adherence, core symptoms of schizophrenia (e.g., hallucinations, delusions), co-occurring symptoms (e.g., depression, hopelessness, hostility, impulsivity), and co-occurring diagnoses (e.g., depression, alcohol use disorder, other substance use disorders). Additional elements of the treatment plan can address periods of increased risk (e.g., shortly after diagnosis, during incarceration, subsequent to hospital discharge).

Addressing Tobacco Use and Other Substance Use Disorders

Individuals with schizophrenia have high rates of nicotine dependence (Centers for Disease Control and Prevention 2020; Cook et al. 2014; de Leon and Diaz 2005; Dickerson et al. 2018; Hartz et al. 2014; Myles et al. 2012; Smith et al. 2014; Wium-Andersen et al. 2015), cannabis use (Brunette et al. 2018; Hartz et al. 2014; Hunt et al. 2018; Koskinen et al. 2010; Nesvåg et al. 2015; Toftdahl et al. 2016), and use of alcohol and other substances (Brunette et al. 2018; Hartz et al. 2014; Hunt et al. 2018; Nesvåg et al. 2015; Toftdahl et al. 2016). Smoking is a major contributor to increased mortality in individuals with serious mental illness (Reynolds et al. 2018; Tam et al. 2016), and the adverse health consequences of smoking are well documented (Lariscy et al. 2018; U.S. Department of Health and Human Services 2014; Van Schayck et al. 2017). Thus, smoking cessation is recommended for any individual who smokes. Some studies have assessed smoking cessation approaches targeted to individuals with mental illnesses, but specific evidence in patients with schizophrenia is still limited (Sharma et al. 2017). Thus, smoking cessation approaches will typically follow guidelines for the general population (National Cancer Institute 2019; SAMHSA-HRSA Center of Excellence for Integrated Health Solutions 2018; Siu et al. 2015; Van Schayck et al. 2017; Verbiest et al. 2017). Although quit rates may be lower in individuals with schizophrenia than in the general population (Lum et al. 2018; Streck et al. 2020), health education and motivational interviewing approaches can be helpful in those who are ambivalent about stopping cigarette use (Levounis et al. 2017) or who have had prior unsuccessful attempts at smoking cessation.

Rates of cannabis use and other substance use are also increased among individuals with schizophrenia (Hartz et al. 2014; Hunt et al. 2018; Swartz et al. 2006). Cannabis use has been associated with an increased incidence (Nielsen et al. 2017; Vaucher et al. 2018) and earlier onset (Donoghue et al. 2014; Helle et al. 2016; Kelley et al. 2016; Large et al. 2011) of schizophrenia, and it may also contribute to a higher burden of symptoms (Carney et al. 2017; Oluwoye et al. 2018). Other substance use disorders are associated with a poorer prognosis in individuals with schizophrenia (Brunette et al. 2018; Conus et al. 2017; Weibell et al. 2017) and, as noted previously, can contribute to risk of suicide or aggressive behavior. Thus, it is important for the treatment plan to address substance use disorders when they are present. Screening, Brief Intervention, and Referral to Treatment (SBIRT) can be integrated into a range of clinical settings (Agerwala and McCance-Katz 2012; SAMHSA-HRSA Center of Excellence for Integrated Health Solutions 2019). Often, a comprehensive integrated treatment model is suggested in which the same clinicians or team of clinicians provide treatment for schizophrenia as well as treatment of substance use disorders. However, if an integrated treatment is unavailable, the treatment plan should address both disorders with communication and collaboration among treating clinicians. For patients who do not recognize the need for treatment of a substance use disorder, a stagewise motivational approach can be pursued (Catley et al. 2016; Levounis et al. 2017).

Addressing Other Concomitant Psychiatric Symptoms and Diagnoses

Depressive symptoms are common in individuals with schizophrenia and should be addressed as part of treatment planning. The approach to treating depression will be grounded in a careful differential diagnosis that considers the possible contributions of demoralization, negative symptoms of schizophrenia, side effects of antipsychotic medications, substance intoxication or withdrawal, physical health condition, or a co-occurring major depressive episode. Depressive symptoms that occur during an acute episode of psychosis often improve as psychotic symptoms respond to treatment.

Evidence on the use of antidepressants to treat depression in individuals with schizophrenia comes from multiple trials, many of which have small sample sizes or factors that increase the risk of bias in the findings (Dondé et al. 2018; Gregory et al. 2017; Helfer et al. 2016). Nevertheless, a meta-analysis suggests that the addition of antidepressant medications results in small beneficial effects on symptoms of depression, quality of life, and response rates as well as on positive symptoms, negative symptoms, and overall symptoms (Helfer et al. 2016). These effects were more prominent in patients with more severe depressive symptoms. Furthermore, antidepressant treatment did not appear to be associated with exacerbation of psychosis or significant differences in adverse effects (Helfer et al. 2016). Nonpharmacological treatments for depression in schizophrenia have been less well studied but could also be incorporated into treatment planning (Dondé et al. 2018; Opoka and Lincoln 2017).

Treatments for posttraumatic stress disorder (Brand et al. 2018; Sin et al. 2017b) and anxiety (Howells et al. 2017) in individuals with schizophrenia have been less well studied. Nevertheless, many individuals with schizophrenia will have experienced violent victimization (de Vries et al. 2019; Morgan et al. 2016; Roy et al. 2014) or childhood adversity (Bonoldi et al. 2013; Schalinski et al. 2019; Trotta et al. 2015; Varese et al. 2012), and the impact of these experiences needs to be considered as part of a patient-centered treatment plan (Center for Substance Abuse Treatment 2014). When anxiety symptoms are present in individuals with schizophrenia, the possible contributions of psychotic symptoms, medication side effects, substance intoxication or withdrawal, or co-occurring anxiety disorders may suggest an approach to treatment. Given the relative safety of adjunctive antidepressant medications in individuals with schizophrenia and depression, these medications may be considered if otherwise indicated to treat posttraumatic stress disorder or an anxiety disorder. On the other hand, studies on the use of benzodiazepines in schizophrenia are limited (Dold et al. 2012), and long-term use of benzodiazepines may be associated with increased risk of poorer outcomes, including side effects (Dold et al. 2013; Fond et al. 2018; Fontanella et al. 2016; Tiihonen et al. 2016) or misuse (Maust et al. 2019). Nonpharmacological treatments for posttraumatic stress disorder in individuals with schizophrenia have been less well studied but may have modest benefits and do not appear to have significant adverse effects as compared with usual care (Brand et al. 2018; Sin et al. 2017b).

In terms of the use of stimulants to treat preexisting attention-deficit/hyperactivity disorder in individuals with schizophrenia, available evidence is also very limited but suggests a potential for worsening of psychotic symptoms as well as potential for development of a stimulant use disorder (Sara et al. 2014; Solmi et al. 2019). Thus, if stimulant medications are used, monitoring for these possible adverse effects is warranted as part of the treatment plan.

Addressing Other Concomitant Health Conditions

As described in Statement 1, other health conditions are more frequent in individuals with serious mental illness in general (Firth et al. 2019; Janssen et al. 2015; McGinty et al. 2016) and schizophrenia in particular (Henderson et al. 2015). Such disorders include but are not limited to poor oral health (Kisely et al. 2015), hepatitis C infection (Chasser et al. 2017; Hauser and Kern 2015; Hughes et al. 2016), HIV infection (Hobkirk et al. 2015; Hughes et al. 2016), cancer (Olfson et al. 2015), sleep apnea (Myles et al. 2016; Stubbs et al. 2016b), obesity (Janssen et al. 2015), diabetes mellitus (Vancampfort et al. 2016a), metabolic syndrome (Vancampfort et al. 2015), and cardiovascular disease (Correll et al. 2017c). These disorders, if present, can contribute to mortality or reduced quality of life, and some may be induced or exacerbated by psychiatric medications. Impairments in renal and hepatic function, if present, can influence treatment recommendations.

Table 2 (see Statement 1) provides a discussion of suggested physical and laboratory assessments for patients with schizophrenia as part of initial evaluation and follow-up assessments. Such assessments are important for prevention, early recognition, and treatment of abnormalities such as glucose dysregulation, hyperlipidemia, and metabolic syndrome. It is important that patients have access to primary care clinicians who can work with the psychiatrist to diagnose and treat concurrent physical health conditions (American Psychiatric Association 2016a), but the psychiatrist may also provide ongoing monitoring and treatment of common medical conditions in conjunction with primary care clinicians (Druss et al. 2018).

Pregnancy and Postpartum Period

Women with childbearing potential and at risk for pregnancy should be assisted in obtaining effective contraception if pregnancy is not desired. For women who are planning to become pregnant or who are pregnant or in the postpartum period, it is essential to collaborate with the patient, her obstetrician-gynecologist or other obstetric practitioner, and, if involved, her partner or other persons of support. For women who are breastfeeding, collaboration with the infant’s pediatrician is similarly important. The overall goal is to develop a plan of care aimed at optimizing outcomes for both the patient and her infant. In addition, during pregnancy and postpartum, frequent reassessment will be needed to determine whether any modifications to the treatment plan are indicated.

As with any decisions related to the use of psychiatric medications prior to conception, during pregnancy, or while breastfeeding, it is essential to consider the potential benefits of treatment as well as the potential harms of untreated illness and the potential for negative fetal or neonatal effects. Untreated or inadequately treated maternal psychiatric illness can result in poor adherence to prenatal care, inadequate nutrition, increased alcohol or tobacco use, and disruptions to the family environment and mother-infant bonding (ACOG Committee on Practice Bulletins—Obstetrics 2008; American Academy of Pediatrics and the American College of Obstetricians and Gynecologists 2017; Tosato et al. 2017). For women with childbearing potential, decisions about medications and advice about contraceptive practices should consider the potential effects if pregnancy were to occur. Some medications are best avoided in women with childbearing potential; for example, valproic acid should be avoided because of its teratogenic effects (Briggs et al. 2017) and association with maternal metabolic syndrome.

All psychotropic medications studied to date cross the placenta, are present in amniotic fluid, and enter human breast milk (American Academy of Pediatrics and the American College of Obstetricians and Gynecologists 2017). In addition, the period from the third through the eighth week of gestation is associated with greatest risk for teratogenesis (American Academy of Pediatrics and the American College of Obstetricians and Gynecologists 2017). If a woman becomes pregnant while taking an antipsychotic medication, consideration should be given to consulting an obstetrician-gynecologist or maternal/fetal medicine subspecialist in addition to discussion with the prescribing clinician before indicated psychotropic medications are stopped to determine whether the risks of stopping the medication outweigh any possible fetal risks (American Academy of Pediatrics and the American College of Obstetricians and Gynecologists 2017; U.S. Food and Drug Administration 2011a). For many women, the eighth week of gestation will already have passed before obstetric care begins, and stopping medication will not avoid or reduce teratogenic risk. Thus, for a woman with schizophrenia, the benefits of continued treatment with antipsychotic medications in minimizing relapse will generally outweigh the potential for fetal risk (Briggs et al. 2017).

Knowledge of the effects of antipsychotic medications is limited to observational and registry-based studies. Although limited information is known about newer second-generation antipsychotic medications (SGAs), first-generation antipsychotic medications (FGAs) have been in wide use for more than 40 years, and older SGAs have been available for several decades. The available data suggest that these medications have minimal risk in terms of teratogenic or toxic effects on the fetus (ACOG Committee on Practice Bulletins—Obstetrics 2008; Briggs et al. 2017; Chisolm and Payne 2016). There does appear to be a risk of withdrawal symptoms or neurological effects of antipsychotic medications in the newborn if an antipsychotic medication is used in the third trimester (Briggs et al. 2017; U.S. Food and Drug Administration 2011a). Symptoms may include agitation, abnormally increased or decreased muscle tone, tremor, sleepiness, severe difficulty breathing, and difficulty in feeding (Briggs et al. 2017; U.S. Food and Drug Administration 2011a). Nevertheless, tapering of antipsychotic medication late in pregnancy is not advisable because of the associated risk of relapse. In some newborns, the symptoms subside within hours or days and do not require specific treatment; other newborns may require longer hospital stays (U.S. Food and Drug Administration 2011a). The possibility of these effects signals the importance of close monitoring of the newborn in conjunction with the infant’s pediatrician. As noted above, however, the benefits of treatment for the mother and the longer-term benefits of treatment for the infant (e.g., enhanced mother-infant bonding, better adherence to prenatal care and nutrition, lesser rates of prenatal alcohol or tobacco use) will generally favor continuing and not tapering antipsychotic treatment.

A number of other considerations are relevant when treating women with an antipsychotic medication during pregnancy. In general, symptoms should be managed with the lowest effective dose, although it is preferable to maintain efficacy using a single medication at a higher dose rather than using multiple medications at lower doses (American Academy of Pediatrics and the American College of Obstetricians and Gynecologists 2017). If a patient’s symptoms are well controlled on a specific medication, it is usually not advisable to switch to a different antipsychotic medication, even if more safety information is available for a different drug (Chisolm and Payne 2016). Changing medications exposes the fetus to two different medications and also increases possibilities for symptom relapse in the patient.

As with all women who are pregnant, regular prenatal care is essential to ensuring optimal maternal-fetal outcomes (American Academy of Pediatrics and the American College of Obstetricians and Gynecologists 2017; American College of Obstetricians and Gynecologists 2018). Close monitoring for symptom recurrence and for side effects is important during pregnancy and in the postpartum period because the physiological alterations of pregnancy may affect the absorption, distribution, metabolism, and elimination of medications and may necessitate adjustments in medication doses (ACOG Committee on Practice Bulletins—Obstetrics 2008; Chisolm and Payne 2016). Women who are taking antipsychotic medications are also at increased risk of obesity and hyperglycemia, and folate supplementation to reduce risks of neural tube defects and assessment for diabetes during pregnancy are important elements of prenatal care (Briggs et al. 2017).

In terms of breastfeeding, limited information is available, but infants may be exposed to clinically significant levels of medication in breast milk, and the long-term effects of such exposure are not known (Sachs et al. 2013). Accordingly, mothers who wish to breastfeed their infants should review the potential benefits of breastfeeding as well as potential risks in the context of shared decision-making (American College of Obstetricians and Gynecologists’ Committee on Obstetric Practice and the Breastfeeding Expert Work Group 2016; Sachs et al. 2013), with associated monitoring of growth and development by the infant’s pediatrician (Sachs et al. 2013).

Additional information related to the use of antipsychotic medications during pregnancy and while breastfeeding can be found at the websites of the U.S. Food and Drug Administration (FDA; www.fda.gov), REPROTOX (www.reprotox.org), the Teratogen Information System (TERIS; www.depts.washington.edu/terisweb/teris), and the U.S. National Library of Medicine’s LactMed (www.ncbi.nlm.nih.gov/books/NBK501922). For women who have been treated with an SGA during pregnancy, enrolling in the National Pregnancy Registry for Atypical Antipsychotics is suggested (MGH Center for Women’s Mental Health 2019).

Determining a Treatment Setting

In determining a treatment setting, considerations for individuals with schizophrenia are similar to those for individuals with other diagnoses. Thus, in general, patients should be cared for in the least restrictive setting that is likely to be safe and to allow for effective treatment. If inpatient care is deemed essential, efforts should be made to hospitalize patients voluntarily. However, if hospitalization is deemed essential but is not accepted voluntarily by the patient, state or jurisdictional requirements for involuntary hospitalization should be followed. Indications for hospitalization usually include the patient posing a serious threat of harm to self or others or being unable to care for himself or herself and needing constant supervision or support as a result. Other possible indications for hospitalization include psychiatric or other medical problems that make outpatient treatment unsafe or ineffective or new onset of psychosis that warrants initial inpatient stabilization to promote reduction of acute symptoms and permit engagement in treatment.

For individuals with schizophrenia and other significant health issues, determination of a treatment setting will require weighing the pluses and minuses of possible settings to identify the optimal location for care. For example, individuals who require significant medical or surgical interventions or monitoring that are not typically available on a psychiatric inpatient service will likely be better served on a general hospital unit or in an intensive care setting with input from consultation-liaison psychiatrists. Considerable efforts may be needed to help staff who are unfamiliar with psychotic disorders engage with the patient (Freudenreich et al. 2019). In other circumstances, management of the patient on an inpatient psychiatric service in collaboration with consultants of other medical specialties will be optimal.

Less restrictive settings may be indicated when a patient does not meet criteria for inpatient treatment but requires more monitoring or assistance than is available in routine outpatient care. Such settings and programs may include ACT (Substance Abuse and Mental Health Services Administration 2008), assisted outpatient treatment, intensive outpatient treatment, partial hospitalization, and day hospitalization.

Involuntary Treatment Considerations

Under some circumstances, individuals may not wish to participate in treatment or take medications, even if they have severe symptoms. In states where psychiatric advance directives are available, patients may be able to state their preferences about treatment choices while they have capacity in the event of future decompensation and an inability to participate in decision-making. Even in the absence of a psychiatric advance directive, patients can often be helped to accept pharmacological treatment over time and with psychotherapeutic interactions that are aimed toward identifying subjectively distressing symptoms that have previously responded to treatment. Family members and other persons of support can also be helpful in encouraging the patient to engage in treatment.

Prevailing state laws will determine other steps to take if an individual lacks capacity but requires treatment. Some states have processes by which pharmacological treatment may be administered involuntarily, whereas in other states a judicial hearing may be needed to obtain permission to treat a patient who lacks capacity.

For a small subgroup of patients with repeated relapses, rehospitalizations, or even reincarcerations associated with nonadherence or impairments in insight, involuntary outpatient commitment may warrant inclusion in the treatment plan to improve adherence, prevent psychiatric deterioration, enhance outcomes, and promote recovery (American Psychiatric Association 2015; Gaynes et al. 2015; Harris et al. 2019; Segal et al. 2017a, 2017b; Swartz et al. 2017). Involuntary outpatient commitment (which also may be referred to as assisted outpatient treatment, mandated community treatment, outpatient court-ordered treatment, or a community treatment order) is increasingly available but varies among countries (Burns et al. 2013; Harris et al. 2019) and jurisdictions within the United States (Meldrum et al. 2016) in its criteria and implementation. Effective implementation requires adequate resources and individualized treatment planning (American Psychiatric Association 2015) if psychiatric (Gaynes et al. 2015; Harris et al. 2019; Segal et al. 2017a; Swartz et al. 2017) and physical health (Segal et al. 2017b) benefits are to be realized. As with any form of involuntary treatment, decisions about involuntary outpatient commitment require balancing ethical considerations related to patient autonomy and self-determination with considerations about the individual’s best interest (American Psychiatric Association 2015).

Addressing Needs of Patients With Schizophrenia in Correctional Settings

Rates of serious mental illness, including schizophrenia, are higher in correctional settings (e.g., prisons, jails, police lockups, detention facilities) than in the general population (Al-Rousan et al. 2017; Bebbington et al. 2017; Bradley-Engen et al. 2010; Hall et al. 2019; Steadman et al. 2009). Careful assessment and treatment planning are essential when individuals with schizophrenia are in correctional settings. Although some aspects of treatment may need to be adjusted to conform with unique aspects of correctional settings (Tamburello et al. 2018), many individuals experience gaps in care during incarceration (Fries et al. 2013; Reingle Gonzalez and Connell 2014; Wilper et al. 2009), and access should be preserved to essential elements of treatment, including antipsychotic medications (American Psychiatric Association 2009b) and treatment for concomitant substance use disorders (American Psychiatric Association 2007).

While in the correctional system, individuals with schizophrenia may be withdrawn or disorganized or behave in a disruptive manner. These behaviors may result in disciplinary infractions, which may lead the individual with schizophrenia to be placed in a locked-down setting. Such units are often called administrative segregation, disciplinary segregation, or restricted housing units (Krelstein 2002; Semenza and Grosholz 2019) and have been conceptualized as having three main characteristics: social isolation, sensory deprivation, and confinement (Zubek et al. 1969). Each of these elements can vary significantly, but inmates typically spend an average of 23 hours per day in a cell, have limited human interaction and minimal or no access to programs, and are maintained in an environment that is designed to exert maximum control over the person, which has raised broader ethical considerations about the long-term use of such settings (Ahalt and Williams 2016; Ahalt et al. 2017; American Psychiatric Association 2017, 2018; American Public Health Association 2013; Cloud et al. 2015; National Commission on Correctional Health Care 2016).

Inmates’ responses to the segregation experience differ, and relevant scientific literature is sparse (Kapoor and Trestman 2016; O’Keefe et al. 2013). In addition, mental health clinicians working in such facilities frequently report that inmates without preexisting serious mental disorders develop irritability, anxiety, and other dysphoric symptoms when housed in these units for long periods of time (Metzner 2002). Difficulties in providing appropriate and adequate access to mental health care and treatment are especially problematic in any segregation environment and are related to logistical issues that frequently include inadequate office space and limited access to inmates because of security issues (Metzner 2003; Metzner and Fellner 2010). In addition, because of their inherently punitive structure, such units typically provide very little support or access to relevant treatment modalities or a therapeutic milieu. Furthermore, rates of self-injury and suicide appear to be higher in such settings than elsewhere in the correctional system (Baillargeon et al. 2009b; Glowa-Kollisch et al. 2016; Kaba et al. 2014; Way et al. 2005). Consequently, persons with schizophrenia should generally not be placed in a 23-hour/day lockdown for behaviors that are directly related to schizophrenia because such an intervention is likely to exacerbate rather than reduce psychotic symptoms, as well as increase rather than reduce disruptive behaviors (American College of Correctional Physicians 2013; American Psychiatric Association 2016b, 2017; American Public Health Association 2013; National Commission on Correctional Health Care 2016).

Individuals with schizophrenia, like other individuals with serious mental illness, are at increased risk for symptom relapse and gaps in treatment on release from a correctional setting. Services are often needed to reduce the likelihood of recidivism and maintain continuity of care for treatment of schizophrenia and concomitant disorders (e.g., substance use disorders, other medical conditions). Thus, discharge planning is a crucial aspect of care for inmates with schizophrenia, particularly for those who have been incarcerated for significant periods of time. Often, inmates with schizophrenia have been alienated from systems of care and psychosocial supports prior to arrest, and this estrangement is compounded by incarceration. As a result, inmates will likely need assistance around the time of discharge, which can encompass various domains, including housing, treatment needs, financial support, and obtaining supplemental security income/social security disability and related Medicaid benefits (American Psychiatric Association 2009c; Angell et al. 2014; Baillargeon et al. 2009a, 2010; Draine et al. 2010; Gertner et al. 2019; Morrissey et al. 2016; Wenzlow et al. 2011).

Balancing of Potential Benefits and Harms in Rating the Strength of the Guideline Statement

Benefits

Development and documentation of a comprehensive, person-centered treatment plan assures that the clinician has considered the available nonpharmacological and pharmacological options for treatment and has identified those treatments that are best suited to the needs of the individual patient, with a goal of improving overall outcome. It may also assist in forming a therapeutic relationship, eliciting patient preferences, permitting education about possible treatments, setting expectations for treatment, and establishing a framework for shared decision-making. Documentation of a treatment plan promotes accurate communication among all those caring for the patient and can serve as a reminder of prior discussions about treatment.

Harms

The only identifiable harm from this recommendation relates to the time spent in discussion and documentation that may reduce the opportunity to focus on other aspects of the evaluation.

Patient Preferences

Clinical experience suggests that patients are cooperative with and accepting of efforts to establish treatment plans.

Balancing of Benefits and Harms

The potential benefits of this guideline statement were viewed as far outweighing the potential harms. The level of research evidence is rated as low because no information is available on the harms of such an approach. There is also minimal research on whether developing and documenting a specific treatment plan improves outcomes as compared with assessment and documentation as usual. However, indirect evidence, including expert opinion, supports the benefits of comprehensive treatment planning. For additional discussion of the research evidence, see Appendix C, Statement 3.

Differences of Opinion Among Writing Group Members

There were no differences of opinion. The writing group voted unanimously in favor of this recommendation.

Review of Available Guidelines From Other Organizations

Information from other guidelines (Addington et al. 2017a, 2017b; Barnes et al. 2011; Buchanan et al. 2010; Crockford and Addington 2017; Galletly et al. 2016; Hasan et al. 2012, 2013, 2015; National Institute for Health and Care Excellence 2014; Norman et al. 2017; Pringsheim et al. 2017; Scottish Intercollegiate Guidelines Network 2013) is generally consistent with this guideline statement in either explicitly or implicitly recommending development of a person-centered treatment plan that includes evidence-based nonpharmacological and pharmacological treatments.

Quality Measurement Considerations

It is not known whether psychiatrists and other mental health professionals typically document a comprehensive and person-centered treatment plan that includes evidence-based nonpharmacological and pharmacological treatments, and there is likely to be variability. Although a well-defined and scientifically sound quality measure could be developed to assess for the implementation of an evidence-based treatment plan that meets consensus-based features of person-centered care, clinical judgment would still be needed to determine whether a documented treatment plan is comprehensive and adapted to individual needs and preferences. Manual review of charts to evaluate for the presence of such a person-centered treatment plan would be burdensome and time-consuming to implement.

A quality measure could assess the presence or absence of text in the medical record that would reflect treatment planning. When considering the development of such quality measures, there should be a thorough examination of the potential for unintended negative consequences, such as increased documentation burden or overuse of standardized language that meets the quality measure criteria but would inaccurately reflect what occurred in practice.

Pharmacotherapy

Statement 4: Antipsychotic Medications

APA recommends (1A) that patients with schizophrenia be treated with an antipsychotic medication and monitored for effectiveness and side effects.*

*This guideline statement should be implemented in the context of a person-centered treatment plan that includes evidence-based nonpharmacological and pharmacological treatments for schizophrenia.

Implementation

Selection of an Antipsychotic Medication

General Principles

In the treatment of schizophrenia, antipsychotic medication is one important component. The choice of an antipsychotic agent depends on many factors that are specific to an individual patient. Thus, before initiating treatment with antipsychotic medication, it is recommended that as part of selecting a medication, the treating clinician gather information on the patient’s treatment-related preferences and prior treatment responses and then discuss with the patient the potential benefits and risks of medication as compared with other management options. Many patients will wish family members or other persons of support to be involved in this discussion. The depth of this discussion will, of course, be determined by the patient’s condition. Even with agitated patients and patients with thought disorder, however, the therapeutic alliance will be enhanced if the patient and physician can identify target symptoms (e.g., anxiety, poor sleep, and, for patients with insight, hallucinations and delusions) that are subjectively distressing and that antipsychotics can ameliorate. Mentioning the possibility of acute side effects (e.g., dizziness, sedation, restlessness) helps patients identify and report side-effect occurrence and also may help maintain a therapeutic alliance. Patients with schizophrenia often have attentional and other cognitive impairments that may be more severe during an acute illness exacerbation, so it is helpful to return to the topic of identification of target symptoms and discussion of acute and longer-term side effects on multiple occasions as treatment proceeds.

An evidence-based ranking of FGAs and SGAs or an algorithmic approach to antipsychotic selection is not possible because of the significant heterogeneity in clinical trial designs, the limited numbers of head-to-head comparisons of antipsychotic medications, and the limited clinical trial data for a number of the antipsychotic medications. By the same token, it is not possible to note a preference for either SGAs or FGAs. Although there may be clinically meaningful distinctions in response to and tolerability of different antipsychotic medications in an individual patient, there is no definitive evidence that one antipsychotic will have consistently superior efficacy compared with another, with the possible exception of clozapine. Although data in first-episode schizophrenia are more limited, there appears to be no difference in response among the SGAs that have been studied (McDonagh et al. 2017; Zhu et al. 2017). Furthermore, there is no reliable strategy to predict response or risk of side effects with one agent compared with another. Consequently, the choice of a particular antipsychotic agent will typically occur in the context of discussion with the patient about the likely benefits and possible side effects of medication options and will incorporate patient preferences; the patient’s past responses to treatment (including symptom response and tolerability); the medication’s side-effect profile (see Table 6); the presence of physical health conditions that may be affected by medication side effects; and other medication-related factors such as available formulations, potential for drug-drug interactions, receptor binding profiles, and pharmacokinetic considerations (Tables 3–9).

Antipsychotic medications: available oral and short-acting intramuscular formulations and dosing considerationsa,b

Trade namec

Available U.S. formulations (mg, unless otherwise noted)

Initial dose (mg/day)

Typical dose range (mg/day)

Maximum daily dose (mg/day)

Commentsd,e,f,g

First-generation antipsychotics

Chlorpromazine

Thorazine

Tablet: 10, 25, 50, 100, 200

25–100

200–800

Oral: 1,000–2,000

IM dosing is typically 25–50 mg per upper outer quadrant of gluteal with 200 mg/day maximum. Do not inject subcutaneously. Use much lower IM doses than oral doses because oral first-pass metabolism is significant.

Short-acting injection (HCl): 25/mL (1 mL, 2 mL)

Fluphenazine

Prolixin

Tablet: 1, 2.5, 5, 10

2.5–10

6–20

Oral: 40

Short-acting IM dose is 33%–50% of oral dose. Dilute oral concentrate immediately before use to ensure palatability and stability.

Oral concentrate: 5/mL (120 mL)

IM: 10

Elixir: 2.5/5 mL (60 mL)

Short-acting injection (HCl): 2.5/mL (10 mL)

Haloperidol

Haldol

Tablet: 0.5, 1, 2, 5, 10, 20

1–15

5–20

Oral:100

2–5 mg IM can be given every 4–8 hours.

Oral concentrate: 2/mL (5 mL, 15 mL, 120 mL)

IM: 20

Short-acting injection (lactate): 5/mL (1 mL, 10 mL)

Loxapine

Loxitane

Capsule: 5, 10, 25, 50

20

60–100h

Oral: 250

Oral inhalation formulation (Adasuve) to treat agitation requires REMS program because of potential for bronchospasm.

Aerosol powder breath-activated inhalation: 10

Aerosol: 10

Molindone

Moban

Tablet: 5, 10, 25

50–75

30–100h

225

Perphenazine

Trilafon

Tablet: 2, 4, 8, 16

8–16

8–32

64

CYP2D6 poor metabolizers will have higher blood concentrations.

Pimozide

Orap

Tablet: 1, 2

0.5–2

2–4

10

Pimozide does not have an FDA indication for schizophrenia but is sometimes used off-label or to treat delusional disorders such as delusional parasitosis. Avoid concomitant use of CYP1A2 or CYP3A4 inducers or inhibitors. Perform CYP2D6 genotyping if doses greater than 4 mg/day are used. In poor CYP2D6 metabolizers, do not give more than 4 mg/day and do not increase dose earlier than 14 days.

Thioridazine

Mellaril

Tablet: 10, 25, 50, 100

150–300

300–800g

800

Use is associated with dose-related QTc prolongation. Baseline ECG and serum potassium level are recommended. Avoid use if QTc interval is > 450 msec or with concomitant use of drugs that prolong the QTc interval or inhibit CYP2D6. Reserve use for patients who do not show an acceptable response to adequate courses of treatment with other antipsychotic drugs.

Thiothixene

Navane

Capsule: 1, 2, 5, 10

6–10

15–30

60

Smoking may reduce levels via CYP1A2 induction.

Trifluoperazine

Stelazine

Tablet: 1, 2, 5, 10

4–10

15–20

50

Smoking may reduce levels via CYP1A2 induction.

Second-generation antipsychotics

Aripiprazole

Abilify

Tablet: 2, 5, 10, 15, 20, 30

10–15

10–15

30

Adjust dose if a poor CYP2D6 metabolizer or with concomitant use of a CYP3A4 inhibitor, CYP3A4 inducer, or CYP2D6 inhibitor. Tablet and oral solution may be interchanged on a mg-per-mg basis, up to 25 mg. Doses using 30 mg tablets should be exchanged for 25 mg oral solution. Orally disintegrating tablets (Abilify Discmelt) are bioequivalent to the immediate-release tablets (Abilify). Mycite tablet cannot be split or crushed.

Tablet, disintegrating: 10, 15

Tablet with ingestible event marker (Mycite): 2, 5, 10, 15, 20, 30

Solution: 1/mL (150 mL)

Asenapine

Saphris

Tablet, sublingual: 2.5, 5, 10

10

20

20

Consider dose adjustment in smokers and with concomitant use of CYP1A2 inhibitors. Do not split, crush, or swallow. Place under tongue and allow to dissolve completely. Do not eat or drink for 10 minutes after administration to ensure absorption.

Asenapine

Secuado

Transdermal system: 3.8 mg/ 24 hours, 5.7 mg/24 hours, 7.6 mg/24 hours

3.8

3.8–7.6

7.6

Consider dose adjustment in smokers and with concomitant use of CYP1A2 inhibitors. A dose of 3.8 mg/24 hours corresponds to 5 mg twice daily of sublingual asenapine; 7.6 mg/24 hours corresponds to 10 mg twice daily of sublingual asenapine. Apply to clean, dry, and intact skin on the upper arm, upper back, abdomen, or hip; rotate sites when applying a new transdermal system. Do not cut. Do not apply external heat sources to the transdermal system.

Brexpiprazole

Rexulti

Tablet: 0.25, 0.5, 1, 2, 3, 4

1

2–4

4

Adjust dose if a poor CYP2D6 metabolizer or with concomitant use of moderate/strong CYP2D6 inhibitors, strong CYP3A4 inhibitors, or strong CYP3A4 inducers.

Cariprazine

Vraylar

Capsule: 1.5, 3, 4.5, 6

1.5

1.5–6

6i

Adjust dose with concomitant use of a strong CYP3A4 inhibitor or inducer.

Clozapine

Clozaril, FazaClo, Versacloz

Tablet: 25, 50, 100, 200

12.5 –25

300–450e

900

Prescribers must complete clozapine REMS education (www.clozapinerems.com) and follow requirements for a baseline CBC and ANC and for ANC monitoring before and during treatment. When initiating clozapine, increase in 25–50 mg/day increments for 2 weeks, then further increments not exceeding 100 mg up to twice weekly. For treatment interruptions of 2 or more days, restart at 12.5 mg once or twice daily. Retitration can occur more rapidly than with initial treatment. With treatment interruptions of more than 30 days, recommendations for initial titration and monitoring frequency should be followed. Adjust dose with concomitant use of strong CYP1A2 inhibitors and with strong CYP3A4 inducers. Smoking reduces clozapine levels via CYP1A2 induction. Clozapine levels can be informative in making dose adjustments.j

Tablet, disintegrating: 12.5, 25, 100, 150, 200

Oral suspension: 50/mL (100 mL)

Iloperidone

Fanapt

Tablet: 1, 2, 4, 6, 8, 10, 12

2

12–24

24

Titrate slowly (no more than 4 mg/day increase in dose). Follow initial titration approach if more than 3-day gap in treatment. Adjust dose with concomitant use of strong CYP2D6 or CYP3A4 inhibitors and reduce dose by 50% in CYP2D6 poor metabolizers.

Lurasidone

Latuda

Tablet: 20, 40, 60, 80, 120

40

40–120

160

Administer with food ( 350 calories). Adjust dose for concomitant use of moderate to strong CYP3A4 inhibitors or inducers.

Olanzapine

Zyprexa

Tablet: 2.5, 5, 7.5, 10, 15, 20

5–10

10–20

20k

Short-acting IM formulation is used primarily for agitation, with usual dose of 2.5–10 mg IM and maximum dosage of 30 mg/day. Administer IM slowly, deep into muscle. Do not use subcutaneously. Concomitant use of IM olanzapine with parenteral benzodiazepines is not recommended because of potential for excessive sedation and cardiorespiratory depression. Smokers may require a 30% greater daily dose than nonsmokers because of CYP1A2 induction. Women may need lower daily doses. Approximately 40% of an oral dose is removed by first-pass metabolism as compared with IM dose. IM elimination half-life is ~1.5 times greater in the elderly. Oral dissolving tablet dissolves rapidly in saliva and may be swallowed with or without liquid. Olanzapine may be administered with or without food/meals.

Tablet, disintegrating: 5, 10, 15, 20

Short-acting IM powder for solution: 10/2 mL

Paliperidone

Invega

Tablet, extended release: 1.5, 3, 6, 9

6

3–12

12

If exceeding 6 mg daily, increases of 3 mg/day are recommended at intervals of more than 5 days, up to a maximum of 12 mg/day. Uses OROS; do not split or crush tablet. Use of extended-release tablet is not recommended with preexisting severe gastrointestinal narrowing disorders. Tablet shell is expelled in the stool.

Quetiapine

Seroquel

Tablet, immediate release: 25, 50, 100, 200, 300, 400

Immediate release: 50

400–800

800

Dosage is once daily for extended release and divided dosing for immediate release. Do not split or crush extended-release tablets. Immediate-release tablets are marginally affected by food, whereas extended-release tablets are significantly affected by a high-fat meal. Give extended-release tablets without food or with < 300 calories. Retitrate for gap in treatment of more than 1 week. Adjust dose for concomitant use of strong CYP3A4 inhibitors or inducers.

Tablet, extended release: 50, 150, 200, 300, 400

Extended release: 300

Risperidone

Risperdal

Tablet: 0.25, 0.5, 1, 2, 3, 4

2

2–8

8l

Use lower initial doses and slower titration rates with CrCl < 30 mL/min or severe hepatic impairment (Child-Pugh class C). Fraction of free risperidone is increased with hepatic impairment, and the initial starting dose is 0.5 mg twice daily, which may be increased in increments of 0.5 mg or less, administered twice daily. With renal or hepatic impairment, increase in intervals of 1 week or more for doses > 1.5 mg twice daily. Adjust dose with concomitant use of inducers or inhibitors of CYP2D6. Check labeling for compatible liquids with oral solution. Do not split or crush oral disintegrating tablets. Inform patients with phenylketonuria that oral disintegrating tablets contain phenylalanine.

Tablet, disintegrating: 0.25, 0.5, 1, 2, 3, 4

Oral solution: 1/mL (30 mL)

Ziprasidone

Geodon

Capsule: 20, 40, 60, 80

40

80–160

320

Give capsules with > 500 calories of food. No data suggest improved efficacy at higher doses. See labeling for reconstitution and storage of IM formulation. Short-acting IM formulation is used primarily for agitation, with usual dosage of 20 mg/day and maximum dosage of 40 mg/day.

Solution reconstituted, IM: 20

aThis table and subsequent medication-related tables include information compiled from multiple sources. Detailed information on such issues as dose regimen, dose adjustments, medication administration procedures, handling precautions, and storage can be found in product labeling. It is recommended that readers consult product labeling information for authoritative information on these medications.

bLong-acting injectable formulations of antipsychotic medications are described separately in Tables 7, 8, and 9. Droperidol is a first-generation antipsychotic medication but is not included because it is available only in a parenteral formulation for short-term use, primarily for treatment of agitation or postoperative nausea and vomiting. Pimavanserin is a second-generation antipsychotic but is not included because it is FDA indicated for the treatment of hallucinations and delusions associated with Parkinson’s disease psychosis (Nuplazid [10 and 34 mg pimavanserin] 2018; Nuplazid [17 mg pimavanserin] 2018). Mesoridazine and triflupromazine were previously marketed in the United States but are no longer available. Other antipsychotic medications and other formulations of the listed medications may be available in Canada.

cThe most common U.S. trade names are included for reference only. At the time of publication, some of these products may be manufactured only as generic products.

dElderly patients with dementia-related psychosis treated with antipsychotics are at an increased risk of death compared with placebo, and an FDA black box warning applies to all antipsychotic medications. Antipsychotic agents with an indication for augmentation treatment in major depressive disorder (e.g., aripiprazole, brexpiprazole) have an additional black box warning related to increased risk of suicidal thinking/behaviors.

eMay be taken without regard to food or other medications unless specifically noted.

fTablets can be crushed or split unless specifically noted.

gAs described by Pugh et al. (1973), Child-Pugh class A corresponds to a Child-Pugh score of 5–6, class B corresponds to a Child-Pugh score of 7–9, and class C corresponds to a Child-Pugh score of 10–15.

hUsually given in divided doses.

iUp to 9 mg/day has been studied in clinical trials.

jClozapine levels should be drawn after at least 3 days on a stable dose and about 12 hours after the last dose. Levels associated with efficacy show individual variation, but efficacy typically begins at a level above 250 ng/mL, with the most efficacy seen at levels higher than 350 ng/mL.

kOlanzapine has been used at higher dosages, typically up to 30 mg/day, although some case series describe use of up to 60 mg/day.

lDosages of risperidone up to 16 mg/day have been studied in clinical trials; however, doses > 6 mg for twice-daily dosing do not appear to confer additional benefit and have a higher incidence of extrapyramidal symptoms than do lower doses.

Abbreviations. ANC  =  absolute neutrophil count; CBC = complete blood count; CrCl = creatinine clearance; CYP = cytochrome P450; ECG = electrocardiography; HCl = hydrochloride; OROS = osmotic-controlled release oral delivery system; QTc = corrected QT interval; REMS = risk evaluation and mitigation strategy.

Antipsychotic medications: available oral and short-acting intramuscular formulations and dosing considerationsa,b

Enlarge table

Antipsychotic medications: pharmacokinetics/pharmacodynamics of oral and short-acting intramuscular formulations

Trade name

Oral bioavailability

Time to peak level

Protein binding

Metabolic enzymes/transporters

Metabolites

Elimination half-life in adults

Excretion

Hepatic impairmenta

Renal impairment

First-generation antipsychotics

Chlorpromazine

Thorazine

32%

2.8 hours

90%–99%

CYP2D6 (major), CYP1A2 (minor), CYP3A4 (minor) substrate

NOR2CPZ, NOR2CPZ SULF, and 3-OH CPZ

Biphasic: initial 2 hours, terminal 30 hours

Primarily renal (< 1% as unchanged drug)

Use with caution

Use with caution; not dialyzable

Fluphenazine

Prolixin

2.7%

Oral: 2 hours

99%

CYP2D6 (major) substrate

7-hydroxyfluphenazine, fluphenazine sulfoxide

4.4–16.4 hours

Renal and fecal; exact proportion unclear

Contraindicated by manufacturer

Use with caution

IM: 1.5–2 hours

Haloperidol

Haldol

60%–70%

Oral: 2–6 hours

89%–93%

CYP2D6 (major), CYP3A4 (major), CYP1A2 (minor) substrate; 50%–60% glucuronidation

Hydroxymetabolite-reduced haloperidol

14–37 hours

15% fecal, 30% renal (1% as unchanged drug)

No dose adjustments noted

No dose adjustments noted

IM: 20 minutes

Loxapine

Loxitane

99%

1.5–3 hours

97%

CYP1A2 (minor), CYP2D6 (minor), CYP3A4 (minor) substrate; P-glycoprotein inhibitor

N-desmethyl loxapine (amoxapine), 8-hydroxyloxapine

Biphasic: initial 5 hours, terminal 19 hours

Renal and fecal

No dose adjustments noted

No dose adjustments noted

Molindone

Moban

Unclear

1.5 hours

76%

CYP2D6 substrate

Multiple

1.5 hours

Renal and fecal

Use with caution

No dose adjustments noted

Perphenazine

Trilafon

20%–40%

Perphenazine: 1–3 hours

91%–99%

CYP2D6 (major), CYP1A2 (minor), CYP2C19 (minor), CYP2C9 (minor), CYP3A4 (minor) substrate

7-hydroxyperphenazine (responsible for 70% of the activity)

Perphenazine: 9–12 hours

5% fecal, 70% renal

Contraindicated in liver damage

Use with caution

7-hydroxyperphenazine: 2–4 hours

7-hydroxyperphenazine: 10–19 hours

Pimozide

Orap

 50%

6–8 hours

99%

CYP1A2 (major), CYP2D6 (major), CYP3A4 (major) substrate

Unknown activity: 4-bis-(4- fluorophenyl) butyric acid, 1-(4-piperidyl)-2-benzimidazolinone

55 hours

Primarily renal

Use with caution

Use with caution

Thioridazine

Mellaril

25%–33%

1–4 hours

96%–99%

CYP2D6 (major) substrate and moderate inhibitor, CYP2C19 (minor) substrate

Mesoridazine (twice as potent as thioridazine), sulforidazine

21–24 hours

Minimal renal

Use with caution

No dose adjustments noted

Thiothixene

Navane

~ 50%; erratic absorption

1–2 hours

90%

CYP1A2 (major) substrate

None noted

34 hours

Feces (unchanged drug and metabolites)

No dose adjustments noted

No dose adjustments noted

Trifluoperazine

Stelazine

Erratic absorption

1.5–6 hours

90%–99%

CYP1A2 (major) substrate

N-desmethyltrifluoperazine, 7-hydroxytrifluoperazine, and other metabolites

3–12 hours

Renal

Contraindicated in hepatic disease

No dose adjustments noted

Second-generation antipsychotics

Aripiprazole

Abilify

87%

3–5 hours

> 99%

CYP2D6 (major), CYP3A4 (major) substrate

Dehydro- aripiprazole

75 hours,

55% fecal, 25% renal

No dose adjustments noted

No dose adjustments noted

94 hours dehydroaripiprazole

146 hours in poor CYP2D6 metabolizers

Asenapine

Saphris

35%

0.5–1.5 hours

95%

CYP1A2 (major), CYP2D6 (minor), CYP3A4 (minor) substrate; glucuronidation by UGT1A4; CYP2D6 weak inhibitor

Inactive: N(+)- glucuronide, N-desmethyl-asenapine, and N-desmethyl-asenapine N-carbamoyl glucuronide

24 hours

40% fecal, 50% renal

Contraindicated in severe hepatic impairment (Child-Pugh class C)

No dose adjustments noted

Asenapine

Secuado

Not applicable

Not available

95%

CYP1A2 (major), CYP2D6 (minor), CYP3A4 (minor) substrate; glucuronidation by UGT1A4; CYP2D6 weak inhibitor

Inactive: N(+)- glucuronide, N-desmethyl-asenapine, and N-desmethyl-asenapine N-carbamoyl glucuronide

24 hours

40% fecal, 50% renal

Contraindicated in severe hepatic impairment (Child-Pugh class C)

No dose adjustments noted

Brexpiprazole

Rexulti

95%

4 hours

> 99%

CYP3A4 (major), CYP2D6 (major) substrate

Inactive: DM-3411

91 hours

46% fecal, 25% renal

Moderate to severe impairment (Child-Pugh class B or C): use maximum dosage of 2 mg/day in MDD and 3 mg/day in schizophrenia

CrCl < 60 mL/minute: use maximum dosage of 2 mg/day in MDD and 3 mg/day in schizophrenia

Cariprazine

Vraylar

High

3–6 hours

91%–97%

CYP3A4 (major), CYP2D6 (minor) substrate

Desmethyl cariprazine (DCAR), didesmethyl cariprazine (DDCAR)

Cariprazine: 2–4 days

21% renal

Severe impairment (Child-Pugh class C): not recommended

CrCl < 30 mL/minute: not recommended

DCAR: 1–2 days

evere impairment (Child-Pugh class C): not recommended

DDCAR: 1–3 weeks

Clozapine

Clozaril, FazaClo, Versacloz

27%–60%

2.2–2.5 hours (range: 1–6 hours)

97%

CYP1A2 (major), CYP2A6 (minor), CYP2C19 (minor), CYP2C9 (minor), CYP2D6 (minor), CYP3A4 (minor) substrate

N-desmethylclozapine (active), hydroxylated and N-oxide derivatives (inactive)

4–66 hours (steady state 12 hours)

30% fecal, 50% renal

Significant impairment: dose reduction may be necessary

Significant impairment: dose reduction may be necessary

Iloperidone

Fanapt

96%

2–4 hours

92%–97%

CYP2D6 (major), CYP3A4 (minor) substrate, CYP3A4 weak inhibitor

P88

Extensive metabolizers: iloperidone 18 hours, P88 26 hours, P95 23 hours

~ 20% fecal, ~ 50% renal

Moderate impairment: use with caution

No dose adjustments noted

P95

Poor metabolizers: iloperidone 33 hours, P88 37 hours, P95 31 hours

Severe impairment: not recommended

No dose adjustments noted

Lurasidone

Latuda

9–19%

1–3 hours

99%

CYP3A4 (major) substrate, CYP3A4 weak inhibitor

ID-14283, ID-14326 (active); ID-20219, ID-20220 (inactive)

Lurasidone 18–40 hours

ID-14283: 7.5–10 hours

~ 80% fecal, ~ 9% renal

Moderate to severe hepatic impairment (Child-Pugh class B and class C): use 20 mg/day initially, with maximum dose of 80 mg/day and 40 mg/day, respectively

For CrCl < 50 mL/minute: initial 20 mg/day, maximum dose 80 mg/day

Olanzapine

Zyprexa

> 57%

Oral: 6 hours

93%

CYP1A2 (major), CYP2D6 (minor) substrate; metabolized via direct glucuronidation

10-N- glucuronide, 4-N-desmethyl olanzapine (inactive)

30 hours

30% fecal, 57% renal

Use with caution

Not removed by dialysis

IM: 15–45 minutes

Paliperidone

Invega

28%

24 hours

74%

P-glycoprotein/ABCB1, CYP2D6 (minor), CYP3A4 (minor) substrate

Activity unclear: M1, M9, M10, M11, M12, M16

23 hours; 24–51 hours with renal impairment (CrCl < 80 mL/minute)

11% fecal, 80% renal

Mild to moderate: no adjustment necessary

Not recommended for CrCl < 10 mL/minute; for CrCl 10–49 mL/minute and CrCl 50–79 mL/minute, use maximum dosage of 3 mg/day and 6 mg/day, respectively

Severe: not studied

Quetiapine

Seroquel

100%

Immediate release: 1.5 hours

83%

CYP3A4 (major), CYP2D6 (minor) substrate

Active: norquetiapine, 7-hydroxyquetiapine

Quetiapine: 6–7 hours

20% fecal, 73% renal

Immediate release: initial 25 mg/day dose, increase by 25–50 mg/day to effective dose

No dose adjustments noted

Extended release: 6 hours

Inactive: quetiapine sulfoxide (major), parent acid metabolite

Norquetiapine: 12 hours

Extended release: initial 50 mg/day, increase by 50 mg/day to effective dose

Risperidone

Risperdal

Absolute: 70%

1 hour

90%

CYP2D6 (major), CYP3A4 (minor), P-glycoprotein/ABCB1 substrate, N-dealkylation (minor), CYP2D6 weak inhibitor

Active: 9- hydroxy-risperidone

Risperidone: 3–20 hours

14% fecal, 70% renal

Mild or moderate impairment (Child-Pugh class A or B): reduce dose

Mild or moderate impairment (CrCl   30 mL/minute): reduce dose

Tablet relative to oral solution: 94%

9-hydroxy-risperidone: 21–30 hours

Severe impairment (Child-Pugh class C): initial 0.5 mg twice a day, increase by no more than 0.5 mg twice a day; may increase to total dosage > 1.5 mg twice a day at 1 week or greater

Severe impairment (CrCl < 30 mL/minute): initial 0.5 mg twice a day, increase by no more than 0.5 mg twice a day; may increase to dosage > 1.5 mg twice a day at 1 week or greater

Ziprasidone

Geodon

Oral with food: 60%

Oral: 6–8 hours

> 99%

CYP1A2 (minor), CYP3A4 (minor) substrate, glutathione, aldehyde oxidase

Active: benzisothiazole sulfoxide (major), benzisothiazole sulfone (major), ziprasidone sulfoxide, S-methyl- dihydroziprasidone

Oral: 7 hours

66% fecal, 20% renal

Use with caution

No oral dose adjustments noted

IM:100%

IM: 60 minutes

IM: 2–5 hours

IM formulation contains a renally cleared excipient, cyclodextrin; use with caution

aAs described by Pugh et al. (1973), Child-Pugh class A corresponds to a Child-Pugh score of 5–6, class B corresponds to a Child-Pugh score of 7–9, and class C corresponds to a Child-Pugh score of 10–15.

Abbreviations. CrCl = creatinine clearance; CYP = cytochrome P450; MDD = major depressive disorder; UGT = uridine 5’-diphospho-glucuronosyltransferase.

Antipsychotic medications: pharmacokinetics/pharmacodynamics of oral and short-acting intramuscular formulations

Enlarge table

Antipsychotic receptor binding properties

Trade name

D1

D2

D3

D4

D5

5-HT1A

5-HT2A

5-HT2C

5-HT7

H1

Musc M1

α1

α2

Comments

First-generation antipsychotics

Chlorpromazine

Thorazine

+

+++

+++

++

+

0

+++

++

++

+++

++

+++

+

Fluphenazine

Prolixin

++

++++

++++

++

++

+

++

+

+++

++

0

+++

0

Haloperidol

Haldol

+

+++

+++

+++

+

0

++

0

+

0

0

++

0

Loxapine

Loxitane

++

++

++

+++

++

0

+++

++

++

+++

+

++

0

Molindone

Moban

0

++

++

0

0

0

0

0

0

0

0

+

Perphenazine

Trilafon

++

++++

++++

++

0

+++

+

++

+++

0

++

+

Pimozide

Orap

0

++++

+++

++

+

++

0

++++

+

+

+

+

Moderate activity at dopamine transporter

Thioridazine

Mellaril

++

++

+++

++

+

+

++

++

++

++

+++

+++

+

Thiothixene

Navane

+

++++

++++

+

+

+

++

0

++

+++

0

++

0

Trifluoperazine

Stelazine

+

+++

++++

++

+

++

+

+

++

+

++

0

Second-generation antipsychotics

Aripiprazole

Abilify

+

////

+++

+

0

///

+++

++

++

++

0

++

+

Asenapine

Saphris, Secuado

+++

+++

++++

+++

+++

++++

++++

++++

+++

0

+++

+++

Brexpiprazole

Rexulti

+

///

+++

++++

////

++++

++

+++

++

0

+++

++++

Cariprazine

Vraylar

///

++++

///

++

+

+

++

0

+

Clozapine

Clozaril, FazaClo, Versacloz

+

+

+

++

+

/

+++

++

++

+++

///

+++

+

Iloperidone

Fanapt

+

++

++

++

+

//

++++

++

++

+

0

+++

+++

Lurasidone

Latuda

+

+++

++

++

/

++++

+

++++

0

0

++

++

Olanzapine

Zyprexa

++

++

++

++

++

0

+++

++

+

+++

+++

++

+

Paliperidone

Invega

+

+++

+++

++

++

+

++++

++

+++

+++

0

+++

++

Quetiapine

Seroquel

0

+

+

0

0

/

+

0

+

+++

+

++

0

Risperidone

Risperdal

+

+++

+++

+++

+

+

++++

++

+++

++

0

+++

+++

Ziprasidone

Geodon

+

+++

+++

++

+

///

++++

++++

+++

++

0

+++

+

Weak activity at norepinephrine and serotonin transporter

Note. ++++ = very strong binding (Ki < 1 nM); +++ = strong binding (1 nM  Ki < 10 nM); ++ = moderate binding (10 nM  Ki < 100 nM); + = weak binding (100 nM  Ki < 1,000 nM); 0 = very weak or negligible binding (Ki  1,000 nM). For partial agonists, / is used instead of + to denote relative binding values.

Antipsychotic receptor binding properties

Enlarge table

Antipsychotic medications: relative side effects of oral formulations

Trade name

Akathisia

Parkinsonism

Dystonia

Tardive dyskinesia

Hyperprolactinemiaa

Anticholinergic

Sedation

First-generation antipsychotics

Chlorpromazine

Thorazine

++

++

++

+++

+

+++

+++

Fluphenazine

Prolixin

+++

+++

+++

+++

+++

+

+

Haloperidol

Haldol

+++

+++

+++

+++

+++

+

+

Loxapine

Loxitane

++

++

++

++

++

++

++

Molindone

Moban

++

++

++

++

++

+

++

Perphenazine

Trilafon

++

++

++

++

++

++

++

Pimozide

Orap

+++

+++

++

+++

+++

+

+

Thioridazine

Mellaril

+

+

+

+

++

+++

+++

Thiothixene

Navane

+++

+++

+++

+++

+++

+

+

Trifluoperazine

Stelazine

++

++

++

++

++

++

+

Second-generation antipsychotics

Aripiprazole

Abilify

++

+

+

+

+

+

+

Asenapine

Saphris

++

+

++

++

++

+

++

Brexpiprazole

Rexulti

++

+

+

+

+

+

++

Cariprazine

Vraylar

++

+

+

+

+

++

++

Clozapine

Clozaril, FazaClo, Versacloz

+

+

+

+

+

+++

+++

Iloperidone

Fanapt

+

+

+

+

++

+

++

Lurasidone

Latuda

++

++

++

++

+

+

++

Olanzapine

Zyprexa

++

++

+

+

++

++

+++

Paliperidone

Invega

++

++

++

++

+++

+

+

Quetiapine

Seroquel

+

+

+

+

+

++

+++

Risperidone

Risperdal

++

++

++

++

+++

+

++

Ziprasidone

Geodon

++

+

+

+

++

+

++

First-generation antipsychotics

Chlorpromazine

Thorazine

++

+++

+++

++

+

++

Fluphenazine

Prolixin

+

+

++

++

+

+

Haloperidol

Haldol

+

+

++

++

+

+

Loxapine

Loxitane

+

++

++

+

+

+

Molindone

Moban

+

+

++

+

+

+

Perphenazine

Trilafon

+

++

++

++

+

+

Pimozide

Orap

+++

+

+++

+

+

+

Thioridazine

Mellaril

++

+++

+++

++

+

+

Pigmentary retinopathy; high rates of sexual dysfunction; avoid use if QTc interval is > 450 msec or with concomitant use of drugs that prolong the QTc interval or inhibit CYP2D6

Thiothixene

Navane

+++

+

++

+

+

+

Trifluoperazine

Stelazine

+

+

++

++

+

+

Second-generation antipsychotics

Aripiprazole

Abilify

+

+

+

+

+

+

FDA safety alert for impulse control disorders (e.g., gambling, binge eating); may reduce hyperprolactinemia with other antipsychotics

Asenapine

Saphris

+

++

++

++

++

++

Oral hypoesthesia

Brexpiprazole

Rexulti

+

+

++

+

++

+

Cariprazine

Vraylar

+

+

++

++

+

+

Clozapine

Clozaril, FazaClo, Versacloz

+++

+++

++

+++

+++

+++

Increased salivation common; high rate of sexual dysfunction; severe constipation and paralytic ileus possible; fever can occur with initiation; myocarditis is infrequent; cardiomyopathy and severe neutropenia are rare

Iloperidone

Fanapt

+

+++

+++

++

+

++

Lurasidone

Latuda

+

+

+

+

++

++

Dose-related creatinine increase in some patients

Olanzapine

Zyprexa

++

++

++

+++

+++

+++

Paliperidone

Invega

+

++

++

++

++

+

Quetiapine

Seroquel

++

++

++

++

+++

++

Risperidone

Risperdal

+

++

++

++

+

++

Intraoperative floppy iris syndrome reported

Ziprasidone

Geodon

+

++

+++

+

+

+

aIn general, rates of sexual dysfunction parallel rates of hyperprolactinemia except where noted in comments.

Note. + = seldom; ++ = sometimes; +++ = often. CYP = cytochrome P450.

Antipsychotic medications: relative side effects of oral formulations

Enlarge table

Long-acting injectable antipsychotic medications: availability and injection-related considerationsa

Trade name

Available strengthsb (mg, unless otherwise noted)

How supplied

Injection site and techniquec

Reactions at injection sited

Comments

First-generation antipsychotics

Fluphenazine

Prolixin Decanoate

25/mL (5 mL)

Vial, sesame oil vehicle with 1.2% benzyl alcohol

Deep IM gluteal or deltoid injection; use of Z-track technique recommendede

Skin reactions reported

Monitor for hypotension. In sesame oil, be alert for allergy. For detailed instructions on needle size and product handling, refer to labeling.

Haloperidol

Haldol Decanoate

50/mL (1 mL, 5 mL), 100/mL (1 mL, 5 mL)

Vial, sesame oil vehicle with 1.2% benzyl alcohol

Deep IM gluteal or deltoid injection; use of Z-track technique recommendede

Inflammation and nodules reported, especially with dose > 100 mg/mL

Do not administer more than 3 mL per injection site. In sesame oil, be alert for allergy. For detailed instructions on needle size, refer to labeling.

Second-generation antipsychotics

Aripiprazole monohydrate

Abilify Maintena

300, 400

Kit with either prefilled syringe or single-use vial

Slow IM injection into gluteal or deltoid muscle

Occasional redness, swelling, induration (mild to moderate)

Rotate injection sites. Do not massage muscle after injection. For detailed instructions on needle size and product reconstitution, refer to labeling.

Aripiprazole lauroxil

Aristada Initio

675/2.4 mL

Kit with prefilled syringe

IM deltoid or gluteal muscle

Common: pain

Infrequent: induration, swelling, redness

Only to be used as a single dose to initiate Aristada treatment or to reinitiate treatment following a missed dose of Aristada. Not for repeat dosing. Not interchangeable with Aristada. Avoid concomitant injection of Aristada Initio and Aristada into the same deltoid or gluteal muscle. Refer to labeling for detailed instructions on injection site, needle length, and instructions to ensure a uniform suspension.

Aripiprazole lauroxil

Aristada

441/1.6 mL, 662/2.4 mL, 882 /3.2 mL, 1,064/3.9 mL

Kit with prefilled syringe

IM deltoid or gluteal muscle for 441 mg

IM gluteal muscle for 662 mg, 882 mg, or 1,064 mg

Common: pain

Infrequent: induration, swelling, redness

Not interchangeable with Aristada Initio. Avoid concomitant injection of Aristada Initio and Aristada into the same deltoid or gluteal muscle. Refer to labeling for detailed instructions on injection site, needle length, and instructions to ensure a uniform suspension.

Olanzapine

Zyprexa Relprevv

210, 300, 405

Kit with vial containing diluent and vial with powder for reconstituting suspension

Deep IM gluteal injection only; do not administer subcutaneously

Infrequent induration or mass at injection site

Because of risk of postinjection delirium/sedation syndrome, must be given in a registered health care facility with ready access to emergency response services, and patient must be observed for at least 3 hours postinjection and accompanied on discharge. Requires use of FDA REMS program (www.zyprexarelprevv program.com/public/Default.aspx). Do not massage muscle after injection. The combined effects of age, smoking, and biological sex may lead to significant pharmacokinetic differences. For detailed instructions on product handling and reconstitution, refer to labeling.

Paliperidone palmitate

Invega Sustenna

39/0.25 mL, 78/0.5 mL, 117/0.75 mL, 156/1 mL, 234/1.5 mL

Kit with prefilled syringe

IM only; slow, deep IM deltoid injection for first 2 doses, then deep deltoid or gluteal injection (upper outer quadrant) thereafter

Occasional redness, swelling, induration

The two initial deltoid IM injections help attain therapeutic concentrations rapidly. Alternate deltoid injections (right and left deltoid muscle). For detailed instructions on needle size and product reconstitution, refer to labeling.

Paliperidone palmitate

Invega Trinza

273/0.875 mL, 410/1.315 mL, 546/1.75 mL, 819/2.625 mL

Kit with prefilled syringe

IM only; slow, deep IM deltoid or gluteal injection

Infrequent redness or swelling

Shake prefilled syringe for 15 seconds within 5 minutes prior to administration. For detailed instructions on needle size, product handling, and reconstitution, refer to labeling.

Risperidone

Risperdal Consta

12.5, 25, 37.5, 50

Kit with prefilled syringe and vial for reconstitution

Deep IM injection into the deltoid or gluteal (upper outer quadrant)

Occasional redness, swelling, induration

Alternate injection sites. Refrigerate and store at 2°C–8°C and protect from light. Vial should come to room temperature for at least 30 minutes before reconstituting. May be stored at 25°C for up to 7 days prior to administration. For detailed instructions on product handling and reconstitution, refer to labeling.

Risperidone

Perseris

90/0.6 mL, 120/0.8 mL

Kit with prefilled syringes containing powder and diluent

Abdominal subcutaneous injection only

Lump at injection site may persist for several weeks

Alternate injection sites. Inject only in area without skin conditions, irritation, reddening, bruising, infection, or scarring; do not rub or massage injection sites. Store at 2°C–8°C and protect from light. Allow package to come to room temperature for at least 15 minutes before injection. For detailed instructions on product handling and reconstitution, refer to labeling.

aThis table and the subsequent table on long-acting injectable antipsychotic medications include information compiled from multiple sources. It is recommended that readers consult product labeling information for authoritative information on these medications. Detailed information on issues such as dose regimen, dose adjustments, medication administration procedures, appropriate needle size based on injection site and patient weight, product reconstitution, handling precautions, and storage can also be found in product labeling.

bAvailable strengths are based on U.S. products; strengths and products available in other countries may differ.

cEach injection must be administered only by a health care professional. Long-acting injectable antipsychotic medications should never be administered intravenously.

dPain at injection site noted for all products.

eSource. Government of South Australia Health: Injection Sites. Available at: www.sahealth.sa.gov.au/wps/wcm/connect/public+content/sa+health+internet/clinical+resources/ clinical+programs+and+practice+guidelines/medicines+and+drugs/injection+techniques#Z%20track. Accessed July 21, 2020. Northumberland, Tyne and Wear NHS Foundation Trust: Standard for the Assessment and Management of Physical Health. Appendix 2: Injection Sites. Available at: www.ntw.nhs.uk/content/uploads/2015/06/AMPH-PGN-10-IMI-App2-Injection-Sites-V01-iss-Sep7.pdf. Accessed January 20, 2019.

Abbreviations. REMS = risk evaluation and mitigation strategy.

Long-acting injectable antipsychotic medications: availability and injection-related considerationsa

Enlarge table

Long-acting injectable antipsychotic medications: dosing

Trade name

Dose conversions

Initial dose (mg)

Typical dose (mg)

Maximum dose (mg)

Dosing frequency

Need for initial oral supplementation

Comments

First-generation antipsychotics

Fluphenazine

Prolixin Decanoate

For each 10 mg/day oral, give 12.5 mg decanoate every 3 weeks

6.25–25 every 2 weeks

6.25–25 every 2–4 weeks

100

2–4 weeks

Decrease oral dose by half after first injection, then discontinue with second injection

Increase in 12.5 mg increments if doses > 50 mg are needed.

Haloperidol

Haldol Decanoate

For each 5 mg/day oral, give 50–75 mg decanoate every 4 weeks

Determined by oral dose and/or risk of relapse up to a maximum of 100 mg

50–200 (10–15 times previous oral dose)

450/month

4 weeks

Taper and discontinue after two to three injections

If initial dose is > 100 mg, split into two injections separated by 3–7 days.

Second-generation antipsychotics

Aripiprazole monohydrate

Abilify Maintena

Not applicable

400

400

400/month

Monthly

Continue oral for 14 days after initial injection

Follow labeling if scheduled injections are missed. Dose adjust for poor CYP2D6 metabolizers, for those taking CYP2D6 and/or CYP3A4 inhibitors, or because of adverse effects. Avoid use with CYP3A4 inducers.

Aripiprazole lauroxil

Aristada Initio

Not applicable

675

675

675

Single dose to initiate Aristada treatment or reinitiate treatment after a missed Aristada dose. Not for repeated dosing.

Must be administered in conjunction with one 30 mg dose of oral aripiprazole

For patients who have never taken aripiprazole, establish tolerability with oral aripiprazole before use. Aristada Initio and Aristada are not interchangeable. See labeling for dose adjustments.

Aripiprazole lauroxil

Aristada

10 mg/day orally, give 441 mg IM/month

Monthly: 441, 662, 882

Monthly: 441, 662, 882

882/month

Monthly: 441, 662, 882

There are two ways to initiate treatment:

Aristada Initio and Aristada are not interchangeable. The first Aristada injection may be given on the same day as Aristada Initio or up to 10 days thereafter.

15 mg/day orally, give 662 mg/month IM, 882 mg IM every 6 weeks, or 1,064 mg IM every 2 months

Every 6 weeks: 882

Every 6 weeks: 882

Every 6 weeks: 882

1. Give one IM injection of Aristada Initio 675 mg and one dose of oral aripiprazole 30 mg

20 mg/day or greater orally, give 882 mg/month IM

Every 2 months: 1,064

Every 2 months: 1,064

Every 2 months: 1,064

or

2. Give 21 days of oral aripiprazole in conjunction with the first Aristada injection

Olanzapine

Zyprexa Relprevv

10 mg/day orally, 210 mg every 2 weeks for four doses or 405 mg every 4 weeks

Determined by oral dose

150 mg, 210 mg, or 300 mg every 2 weeks or 300 mg or 405 mg every 4 weeks

300 mg every 2 weeks or 405 mg every 4 weeks

2–4 weeks

Not required

Give 150 mg every 4 weeks in patients who may have sensitivity to side effects or slower metabolism. Smokers may require a greater daily dose than nonsmokers, and women may need lower daily doses than expected.

15 mg/day orally, 300 mg every 2 weeks for four doses

20 mg/day orally, 300 mg every 2 weeks

Paliperidone palmitate

Invega Sustenna

3 mg oral paliperidone, give 39–78 mg IM

234 mg IM on day 1 and 156 mg IM 1 week later, both administered in the deltoid muscle

78–234 mg monthly beginning at week 5

234 mg/month

Monthly

Not required

Contains range of particle sizes for rapid and delayed absorption. For changes to oral or other LAI to Sustenna, see labeling. Doses are expressed as amount of paliperidone palmitate rather than as paliperidone. Avoid using with a strong inducer of CYP3A4 and/or P-glycoprotein.

6 mg oral, give 117 mg IM

9 mg oral, give 156 mg IM

12 mg oral, give 234 mg IM

Paliperidone palmitate

Invega Trinza

Conversion from monthly Invega Sustenna to every 3-month injections of Invega Trinza:

78 mg, give 273 mg

117 mg, give 410 mg

156 mg, give 546 mg

234 mg, give 819 mg

Dependent on last dose of monthly paliperidone

273–819

819/3 months

Every 3 months

Not applicable

Change to Trinza after at least four Invega Sustenna doses (with two doses at same strength). For changes from IM Trinza to oral or to IM Sustenna, see labeling. Doses are expressed as amount of paliperidone palmitate rather than as paliperidone. Avoid using with a strong inducer of CYP3A4 and/or P-glycoprotein.

Risperidone

Risperdal Consta

Oral risperidone to Risperdal Consta IM:

 3 mg/day, give 25 mg/2 weeks

> 3 to  5 mg/day, give 37.5 mg/ 2 weeks

> 5 mg/day, give 50 mg/2 weeks

25 every 2 weeks

25–50 every 2 weeks

50 every 2 weeks

2 weeks

Continue oral for 3 weeks (21 days)

Upward dose adjustment should not be made more frequently than every 4 weeks.

Risperidone

Perseris

Oral risperidone to subcutaneous risperidone extended release:

3 mg/day, give 90 mg/month

4 mg/day, give 120 mg/month

Determined by oral dose

90–120 monthly

120/month

Monthly

Neither a loading dose nor oral overlap is needed

May not be appropriate for patients taking less than 3 mg or more than 4 mg of oral risperidone daily. Adjust dose with concomitant CYP2D6 inhibitors or CYP3A4 inducers.

Abbreviations. CYP = cytochrome P450; LAI = long-acting injectable.

Long-acting injectable antipsychotic medications: dosing

Enlarge table

Long-acting injectable antipsychotic medications: pharmacological characteristics

Trade name

Time to peak plasma level

Time to steady state

Elimination half-life

Commentsa

First-generation antipsychotics

Fluphenazine

Prolixin Decanoate

8–10 hours

2 months

6–9 days for single injection and 14–26 days for multiple doses

Major CYP2D6 substrate

Haloperidol

Haldol Decanoate

6 days

3–4 months

21 days

Major CYP2D6 and CYP3A4 substrate

Second-generation antipsychotics

Aripiprazole monohydrate

Abilify Maintena

4 days (deltoid); 5–7 days (gluteal)

By fourth dose

300 mg: 29.9 days

Give no sooner than 26 days between injections.

400 mg: 46.5 days (400 mg) with gluteal injection

Major CYP2D6 and CYP3A4 substrate

Aripiprazole lauroxil

Aristada Initio

16–35 days (median 27 days)

Not applicable

15–18 days

Not interchangeable with Aristada because of differing pharmacokinetic profiles

CYP2D6 and CYP3A4 substrate

Aripiprazole lauroxil

Aristada

Not available

4 months

53.9–57.2 days

Not interchangeable with Aristada Initio because of differing pharmacokinetic profiles

CYP2D6 and CYP3A4 substrate

Olanzapine

Zyprexa Relprevv

7 days

~ 3 months

30 days

Major CYP1A2 substrate

Paliperidone palmitate

Invega Sustenna

13 days

2–3 months

25–49 days; increased in renal disease

CrCl 50–79 mL/minute: initiate at 156 mg on day 1, followed by 117 mg 1 week later, both administered in the deltoid muscle. Maintenance dose of 78 mg. Use not recommended in patients with CrCl < 50 mL/minute.

Substrate of P-glycoprotein/ABCB1

Paliperidone palmitate

Invega Trinza

30–33 days

Not applicable

84–95 days with deltoid injection; 118–139 days with gluteal injection; increased in renal disease

Do not use in patients with CrCl < 50 mL/minute.

Substrate of P-glycoprotein/ABCB1

Risperidone

Risperdal Consta

29–31 days

2 months

3–6 days; increased in renal or hepatic disease

For renal/hepatic impairment: initiate with oral dosing (0.5 mg twice a day for 1 week, then 1 mg twice a day or 2 mg daily for 1 week); if tolerated, begin 25 mg IM every 2 weeks and continue oral dosing for 21 days. An initial IM dose of 12.5 mg may also be considered.

Major substrate of CYP2D6 and minor substrate of CYP3A4 (minor) substrate; weak CYP2D6 inhibitor

Risperidone

Perseris

Two peaks: 4–6 hours and 10–14 days

2 months

9–11 days

For renal/hepatic impairment: use with caution with renal impairment; has not been studied. If oral risperidone is tolerated and effective at doses up to 3 mg/day, 90 mg/month can be considered.

Major CYP2D6 substrate and minor CYP3A4 substrate; weak CYP2D6 inhibitor

aIf a dose of a long-acting injectable antipsychotic medication is missed, refer to product labeling for information on adjustments to medication dose or administration frequency.

Abbreviations. CrCl = creatinine clearance; CYP = cytochrome P450.

Long-acting injectable antipsychotic medications: pharmacological characteristics

Enlarge table

Factors Influencing Choice of an Antipsychotic Medication

Available Drug Formulations

Medication choice may be influenced by available formulations of specific medications, such as oral concentrates or rapidly dissolving tablets for patients who have difficulty swallowing pills or who are ambivalent about medications and inconsistent in swallowing them. Use of ingestible sensors with associated monitoring technology may assist in evaluating ingestion, although the FDA notes that improvements in adherence have not yet been shown (U.S. Food and Drug Administration 2017). LAI formulations may be preferred by some patients (Heres et al. 2007; Patel et al. 2009; Walburn et al. 2001) and may be particularly useful for patients with a history of poor or uncertain adherence (see Statement 10). Short-acting parenteral formulations of antipsychotic agents are available for short-term use in individuals who are unable to take oral medications or for emergency administration in acutely agitated patients.

Drug-Drug Interactions and Metabolism

Careful attention must be paid to the potential for interactions of antipsychotic agents with other prescribed medications. For example, when multiple medications are prescribed, side effects (e.g., sedation, anticholinergic effects) can be additive. In addition, drug interactions can influence the amount of free drug in the blood that is available to act at receptors. Because most antipsychotic medications are highly bound to plasma proteins, the addition of other protein-bound medications will displace drug molecules from proteins, resulting in a greater proportion of unbound drug in the blood. Another common cause of drug-drug interactions relates to interactions at metabolic enzymes such as cytochrome P450 (CYP) enzymes, UDP-glucuronosyltransferases, and flavin-containing monooxygenases (Ouzzine et al. 2014; Phillips and Shephard 2017; Rowland et al. 2013; Zanger and Schwab 2013). In particular, hepatic metabolism of antipsychotic medications via CYP enzymes has been widely studied (see Lexicomp 2019; Micromedex 2019). Medications may compete with each other for the same CYP enzyme, or they may induce or inhibit the activity of CYP enzymes, altering levels of drugs that are metabolized through that route. For antipsychotic medications that have active metabolites, shifts in CYP enzyme activity can influence the relative amounts of the active metabolite. Consequently, when a patient is taking multiple medications, it is useful to check for possible drug-drug interactions using electronic drug interaction software (e.g., Web-based software, drug interaction checking embedded in electronic health record software).

In addition to drug-drug interactions, a number of other factors can influence CYP enzymes and thereby affect antipsychotic medication levels in blood. For example, smoking tobacco or marijuana induces CYP1A2, resulting in a corresponding reduction of levels of drugs that are metabolized through that enzyme, including clozapine and olanzapine (Anderson and Chan 2016; Kroon 2007; Scherf-Clavel et al. 2019). Conversely, with cessation of smoking (either intentionally or with admission to a smoke-free facility), there will be corresponding increases in the levels of drugs metabolized via CYP1A2. These shifts in blood levels can be quite significant and contribute to shifts in medication effectiveness or toxicity. Several of the main phytocannabinoids in marijuana (e.g., Δ9-tetrahydrocannabinol, cannabidiol) are metabolized via CYP3A4, and cannabidiol may also inhibit CYP2C19 (Anderson and Chan 2016).

Furthermore, levels of antipsychotic medications, the relative proportions of active metabolites, and other pharmacokinetic properties such as medication or active metabolite half-life can be influenced by genetic differences in metabolic enzyme activity. Polymorphisms of CYP2D6 have been subjected to the most study (Eum et al. 2016; Zhou 2009), show substantial variation in their occurrence in the population by ethnicity (Bertilsson 2007; Gaedigk et al. 2017), and are likely to have the greatest potential for impact on antipsychotic medication metabolism. Polymorphisms of the ATP-binding cassette subfamily B member 1 (ABCB1) gene, which affects P-glycoprotein membrane transport, may influence brain concentrations of drugs, including antipsychotic agents (Moons et al. 2011). Although the applicability of gene polymorphism testing to the clinical choice of an antipsychotic medication is still being explored (Bousman and Dunlop 2018; Koopmans et al. 2018; Lagishetty et al. 2016), the FDA has incorporated testing for CYP2D6 polymorphisms into its labeling recommendations for dosing of pimozide on the basis of the increased risk of electrocardiographic changes in poor metabolizers at doses higher than 4 mg/day (0.05 mg/kg/day in children) (Rogers et al. 2012; U.S. Food and Drug Administration 2011b). In addition, product labeling for a number of other antipsychotic medications refers to a need for dose adjustments based on metabolizer status (U.S. Food and Drug Administration 2019). Additional information on the clinical pharmacogenomics of antipsychotic medications is available through the Clinical Pharmacogenetics Implementation Consortium (https://cpicpgx.org; Relling and Klein 2011), the Pharmacogene Variation Consortium (www.pharmvar.org; Gaedigk et al. 2018), and the Pharmacogenomics Knowledgebase (www.pharmgkb.org; Whirl-Carrillo et al. 2012).

Pharmacokinetic Properties

The absorption of some antipsychotic medications is affected by the presence of food in the stomach (see Statement 4, Tables 3 and 4). Some individuals may have difficulty in adhering to appropriate meal size or content, which could influence choice of these medications.

The half-life of an antipsychotic medication is another pharmacokinetic property that may be useful to consider in choosing among antipsychotic agents. Antipsychotic agents with a short half-life (see Statement 4, Tables 3 and 4) are more likely to require divided dosing in contrast to antipsychotic medications with a half-life that is closer to 24 hours. An oral antipsychotic medication with a longer half-life or an LAI may be preferable for patients who are prone to forget doses or who are intermittently nonadherent to treatment. Nevertheless, if an antipsychotic medication (or active metabolite) half-life is significantly longer than 24 hours, it is important to be aware that steady state may not be reached for some time. This can complicate interpreting the patient’s response to adjustments in doses in terms of therapeutic benefits and side effects. Additional caution may be needed when an antipsychotic medication with a long half-life is chosen for older individuals, for an individual who is taking other medications that may affect drug metabolism, or for individuals with renal or hepatic impairment.

Older individuals often exhibit additional physiological changes relative to younger persons, including a reduced cardiac output (and concomitant reduction in renal and hepatic blood flow), reduced glomerular filtration rate, possible reduction in hepatic metabolism, and increased fat content. These changes may alter the absorption, distribution, metabolism, and excretion of medications and may also result in prolonged drug effects and greater sensitivity to medications, in terms of both therapeutic response and side effects (Kaiser 2015).

Side-Effect Profile

The side-effect profile of an antipsychotic agent is a significant factor in the choice of a specific medication (see Statement 4, Table 6). Often, a patient will express concerns about a particular side effect of medication (e.g., weight gain). A specific side effect (e.g., akathisia, weight gain, sedation, orthostatic hypotension, sexual dysfunction) may also have limited a patient’s treatment adherence or ability to function in the past. If a patient has a concomitant physical condition (e.g., diabetes, cardiac conduction abnormalities, a seizure disorder), choice of medication will need to consider the likelihood of exacerbating an existing health condition. Older individuals may be more sensitive to some medication side effects, such as tardive dyskinesia, orthostatic hypotension, or anticholinergic effects of medications. Thus, a medication that has a lower likelihood of these side effects might be preferred. In contrast, there may be circumstances in which a medication side effect may be helpful. For example, in a patient who is not sleeping well, a more sedating antipsychotic might be chosen and administered at bedtime. Regardless of the initial side effect–related considerations in the choice of an antipsychotic medication, it is important to continue to monitor for side effects as treatment proceeds and to have additional discussions with the patient about side effects as they relate to treatment preferences.

Initiation of Treatment With an Antipsychotic Medication

The initial goal of acute treatment with an antipsychotic medication is to reduce acute symptoms, with the aim of returning the individual to his or her baseline level of functioning. Later, maintenance treatment will aim to prevent recurrence of symptoms and maximize functioning and quality of life.

The initial dose of medication will depend on such factors as the medication formulation, the characteristics of the patient, and whether a prior trial of antipsychotic medication has occurred. With the exception of clozapine, the dose of most antipsychotic medications can be increased relatively quickly to a typical therapeutic dose once an initial dose has been tolerated. For patients who have previously been treated with an oral or LAI antipsychotic medication, more rapid resumption of an effective medication dose is often appropriate. Although as-needed or emergency administration of antipsychotic medication may, at times, be useful in individuals with acute agitation, it can also reduce tolerability and contribute to a perception that premature dose increases are needed.

Younger individuals who are experiencing a first episode of psychosis may be more likely to gain weight or develop adverse metabolic effects of antipsychotic medications (Correll et al. 2014; Jensen et al. 2019). This can influence selection of an initial medication. In such individuals, a lower initial medication dose may help in minimizing acute side effects of antipsychotic medication and may improve a patient’s willingness to continue with treatment (Czobor et al. 2015; Gaebel et al. 2010). Because a first episode of psychosis may respond more rapidly and may require a lower medication dose than later episodes do, use of a lower initial medication dose is also reasonable (Takeuchi et al. 2019). In older individuals, particularly those with concomitant physical health issues who are receiving multiple medications, recommended starting doses of medication are one-quarter to one-half of the usual adult starting dose on the basis of pharmacokinetic considerations (Howard et al. 2000).

Determining the optimal dose of antipsychotic medication during acute treatment is complicated by the fact that there is usually a delay between initiation of treatment and full therapeutic response. Patients may take between 2 and 4 weeks to show an initial response and longer periods of time to show full or optimal response. Once a therapeutic dose of the antipsychotic medication is reached, overly rapid or premature escalation of medication doses can affect tolerability. Premature dose increases can also create the false impression of enhanced efficacy due to a higher dose when the observed response is actually related to elapsed time at a steady state level of medication. Available evidence suggests that patients who have not exhibited at least a 20% reduction in symptoms (or minimal improvement) by about 2 weeks on a therapeutic dose are unlikely to be much improved at 4–6 weeks as reflected by at least a 50% reduction in symptoms (Samara et al. 2015). Consequently, monitoring of the patient’s clinical status for 2–4 weeks is warranted on a therapeutic dose unless the patient is having uncomfortable side effects.

Initiation of treatment with clozapine is a notable exception to this general approach because it requires a slow dose titration to minimize the risks of seizure, orthostatic hypotension, and excessive sedation (Clozaril 2019). Large, rapid increases in clozapine dosage have led to cardiovascular collapse and death, particularly in patients taking respiratory depressant medications such as benzodiazepines. From a starting dose of 12.5 mg once or twice daily, the daily clozapine dosage can be increased by, at most, 25–50 mg/day to a target dose of 300–450 mg/day (Clozaril 2019). Subsequent dose increases, if needed, should be of 100 mg or less, once or twice weekly. A slower rate of titration may be needed for patients with an initial episode of schizophrenia and in those who are older, severely debilitated, or sensitive to side effects. Those with a preexisting central nervous system condition, including individuals with 22q11.2 deletion syndrome, also warrant a slower rate of titration and may have an increased risk of seizures at usual doses. Use of divided doses can be helpful in reducing side effects during initial dose titration, although many patients are ultimately treated with a single dose at bedtime to minimize daytime sedation and facilitate adherence (Takeuchi et al. 2016). Although efficacy is often seen at a dosage of 300–450 mg/day, some individuals may need higher dosages of clozapine, to a maximum daily dose of 900 mg, for full response. Blood levels of clozapine can be helpful to obtain if making adjustments to clozapine doses (see Statement 7).

With clozapine, safety monitoring during treatment is important in order to minimize the risk of adverse events. The U.S. Clozapine Risk Evaluation and Mitigation Strategy (REMS) Program (www.clozapinerems.com)1 includes required training that must be completed by prescribers (Clozapine REMS Program 2019a), resource materials (Clozapine REMS Program 2019b), and a shared patient registry for all clozapine manufacturers’ products that permits tracking of absolute neutrophil counts (ANCs) and documentation of decisions about continued treatment. The Clozapine REMS site provides instructions about threshold values for ANCs in hematologically normal individuals and in those with benign ethnic neutropenia, which is most common in individuals of African descent and is associated with normal ANCs that are lower than standard reference ranges (Clozapine REMS Program 2014). The site also describes the required frequencies for ANC monitoring, which vary with ANC values. Because the highest risk of severe neutropenia (ANC < 500/μL) occurs within the initial 6 months of clozapine treatment (Alvir et al. 1993; Clozapine REMS Program 2019c; Myles et al. 2018), the frequency of ANC monitoring is also reduced with longer treatment duration. In patients who have stopped or interrupted treatment with clozapine for 30 days or more, the initial dose titration for clozapine and the monitoring frequency for treatment initiation should be followed.

1For Canadian prescribers, use the appropriate Canadian clozapine registry, not the U.S. Clozapine REMS Program.

With clozapine as well as with other antipsychotic medications, some common early side effects such as sedation, postural hypotension, or nausea may improve or resolve after the first several days or weeks of treatment, and patients can be encouraged to tolerate or temporarily manage these short-term effects. Other side effects, notably parkinsonism and akathisia, are likely to persist with long-term treatment, and additional approaches to management may be needed (see Statements 12 and 13).

If treatment with an LAI antipsychotic medication is planned, a trial of the oral formulation of the same medication is usually given to assure tolerability. The conversion from an oral dose of medication to a corresponding dose of an LAI antipsychotic depends on the specific medication (Meyer 2017), and product labeling for each medication describes approximate conversion ratios and whether a period of concomitant oral and LAI medication is needed.

Strategies to Address Initial Nonresponse or Partial Response to Antipsychotic Treatment

If a patient is showing response within several weeks of treatment initiation, continuing with the same medication and monitoring for continued improvement is appropriate. However, if there is no significant improvement after several weeks of treatment (e.g., < 20% improvement in symptoms) or if improvement plateaus before substantial improvement is achieved (e.g., > 50% improvement in symptoms, minimal impairment in functioning), it is important to consider whether factors are present that are influencing treatment response. Such factors may include concomitant substance use, rapid medication metabolism, poor medication absorption, interactions with other medications, and other effects on drug metabolism (e.g., smoking) that could affect blood levels of medication.

Difficulties with adherence are a common contributor to reduced response. (For a detailed discussion of adherence, see Statement 3.) When adherence is poor or uncertain, use of an LAI formulation of an antipsychotic may improve adherence as well as response. Determination of the blood concentration of the drug may also be helpful if the patient is being treated with a medication (e.g., clozapine) for which blood level has some correlation with clinical response. For other antipsychotic medications, a blood level can help to determine if poor adherence or subtherapeutic levels may be contributing to poor response (Bishara et al. 2013; de Oliveira et al. 1996; Hiemke et al. 2004; Lopez and Kane 2013; McCutcheon et al. 2018; Melkote et al. 2018; Sparshatt et al. 2010; Uchida et al. 2011b; Van Putten et al. 1991). Depending on the patient’s symptoms, the possibility of another concomitant disorder should be considered. For example, in a patient with negative symptoms, an untreated major depressive disorder may also be present.

If no factors that would affect treatment response have been identified, raising the dose for a finite period, such as 2–4 weeks, can be tried. Although the incremental efficacy of higher doses has not been established (Samara et al. 2018), some patients may show benefit if they are able to tolerate a higher dose of antipsychotic medication without significant side effects. If dose adjustment does not result in an adequate response, a different antipsychotic medication should be considered. Tables 5 and 6 in Statement 4 can be consulted to identify antipsychotic medications with other receptor binding profiles or different side effects. Because each patient responds differently to antipsychotic medications in terms of therapeutic effects and side effects, adequate trials of multiple antipsychotic medications may be needed before antipsychotic treatment is optimized, and it can be helpful to advise patients of this possibility.

If a patient has had minimal or no response to two trials of antipsychotic medication of 2–4 weeks’ duration at an adequate dose (Howes et al. 2017; Samara et al. 2015), a trial of clozapine is recommended (see Statement 7). A trial of clozapine is also recommended for a patient with a persistent risk of suicide that has not responded to other treatments and is suggested for a patient with a persistent risk of aggressive behavior that has not responded to other treatments (see Statements 8 and 9). A trial of clozapine may also be appropriate in individuals who show a response to treatment (i.e., have more than a 20% reduction in symptoms) yet still have significant symptoms or impairments in functioning (Howes et al. 2017). In fact, clozapine is often underused (Carruthers et al. 2016; Latimer et al. 2013; Olfson et al. 2016; Stroup et al. 2014; Tang et al. 2017), and many patients would benefit from earlier consideration of clozapine initiation.

For individuals with treatment-resistant schizophrenia who are unable to tolerate clozapine or are not interested in pursuing a trial of clozapine, the limited available evidence suggests no benefit from high doses of antipsychotic medication, and treatment-related side effects are likely to be increased (Dold et al. 2015). However, a trial of a different antipsychotic medication may be helpful, particularly if there is no response or only a partial response to the most recently used medication.

Augmentation treatment can also be considered, although a trial of clozapine should not be delayed by multiple attempts at augmentation therapy. Particularly for patients with negative symptoms or depression, augmentation of antipsychotic therapy with an antidepressant medication may also be helpful (Helfer et al. 2016; Stroup et al. 2019). Use of a benzodiazepine, such as lorazepam, is also suggested in patients who exhibit catatonia (Bush et al. 1996b; Fink 2013; Pelzer et al. 2018; Unal et al. 2017). Other augmentation approaches (e.g., antipsychotics, anticonvulsants, benzodiazepines, lithium) have also been studied, although evidence is mixed and primarily from small, short-term open-label studies (Correll et al. 2017b; Galling et al. 2017; Ortiz-Orendain et al. 2017). For combination therapy with two antipsychotic medications, data from a large nationwide cohort study suggest that emergency visits and rehospitalization rates may be reduced in individuals receiving polypharmacy as compared with monotherapy (Tiihonen et al. 2019). In addition, there is no evidence that combining drugs is any more harmful than using a single medication, beyond the common side effects from each drug. Nevertheless, if multiple drugs are used, monitoring for benefits and side effects is important, and it is preferable to limit changes in dose to one drug at a time. In addition, if a patient experiences an exacerbation of symptoms while on a stable dose of medication, a reconsideration of the treatment plan is warranted rather than simply adding medications to the existing regimen.

Monitoring During Treatment With an Antipsychotic Medication

During treatment with an antipsychotic medication, it is important to monitor medication adherence, therapeutic benefits of treatment, and treatment-related side effects. The patient’s clinical status can also be affected by changes in physical health, adjustments to other psychotropic and nonpsychotropic medications, and other factors, such as cessation or resumption of smoking.

Adherence to antipsychotic treatment is a common problem that affects treatment outcomes. There are many barriers to treatment adherence as well as facilitators and motivators of adherence, each of which will differ for an individual patient (Hatch et al. 2017; Kane et al. 2013; Pyne et al. 2014). Thus, it is important to take a patient-centered approach in inquiring in a nonjudgmental way whether the individual has experienced difficulties with taking medication since the last visit. (For a detailed discussion of factors related to adherence, see Statement 3.)

Monitoring of treatment response is also essential for identifying whether there are reductions in the severity of functional impairments or target symptoms, including positive symptoms, negative symptoms, and other symptoms that are a focus of treatment. Use of a quantitative measure (see Statement 2) can assist in determining whether the antipsychotic medication is producing therapeutic benefits, including reductions in symptom severity and improvements in functioning. If a lack of response or a partial response is noted, additional assessment will be needed to identify and address possible contributors as described in the subsection “Strategies to Address Initial Nonresponse or Partial Response to Antipsychotic Treatment.” If an antipsychotic medication dose is being decreased, monitoring can help detect a return of symptoms prior to a more serious relapse.

Monitoring for the presence of side effects is also important throughout the course of antipsychotic treatment. Some side effects (e.g., sedation, nausea) are prominent with treatment initiation but dissipate, at least to some extent, with continued treatment. Other side effects (e.g., hypotension, akathisia) may be present initially and increase in severity with titration of the medication dose. Still other side effects emerge only after longer periods of treatment (e.g., tardive dyskinesia) or become more noticeable to patients as their acute symptoms are better controlled (e.g., sexual dysfunction). Table 2 in Statement 1 gives suggestions for baseline assessments and monitoring frequencies for some side effects, clinical measurements, and laboratory studies. Specific attention may need to be given to clinical workflow to assure that indicated monitoring is conducted because rates of follow-up testing and screening for metabolic side effects of treatment appear to be low (Morrato et al. 2009). Patients should also be asked about other common side effects of antipsychotic medications, which may vary with the specific medication that is prescribed (see Statement 4, Table 6).

Use of rating scales can help assure that patients are asked about side effects in a systematic fashion. Although the clinician-rated Udvalg for Kliniske Undersogelser (UKU) Side Effect Rating Scale (Lingjaerde et al. 1987) is often used to assess side effects of antipsychotic medications in clinical trials (van Strien et al. 2015), it can be time-consuming to administer. However, a self-rated version of the UKU Side Effect Rating Scale (Lindström et al. 2001) is also available (http://scnp.org/fileadmin/SCNP/SCNP/UKU/UKU-Pat-%20English%20.pdf). Another self-rating scale, the Glasgow Antipsychotic Side-effect Scale, has two versions: one for use in patients treated with clozapine (www.sussexpartnership.nhs.uk/sites/default/files/documents/gass_for_clozapine .pdf) (Hynes et al. 2015) and one for patients treated with other antipsychotic medications (www.southernhealth.nhs.uk/_resources/assets/inline/full/0/38222.pdf) (Waddell and Taylor 2008). Other rating scales are aimed at identifying and assessing the severity of a specific type of side effect. For example, the clinician-administered Abnormal Involuntary Movement Scale (AIMS; www.aacap.org/App_Themes/AACAP/docs/member_resources/toolbox_for_clinical_practice_ and_outcomes/monitoring/AIMS.pdf) (Guy 1976) or the Dyskinesia Identification System: Condensed User Scale (DISCUS; https://portal.ct.gov/-/media/DDS/Health/hcsma20002b.pdf) (Kalachnik and Sprague 1993) can complement clinical assessment in identifying and monitoring tardive dyskinesia and other abnormal movements. Another example, the self-rated Changes in Sexual Functioning Questionnaire (www.dbsalliance.org/wp-content/uploads/2019/02/Restoring_Intimacy_CSFQ_ Handout.pdf) (Clayton et al. 1997a, 1997b; Depression and Bipolar Support Alliance 2019; Keller et al. 2006), can help to identify sexual side effects of antipsychotic treatment, which is an issue that patients may find difficult to discuss yet can lead them to discontinue treatment.

Treatment-Emergent Side Effects of Antipsychotic Medications

As with most medications, antipsychotic medications have been associated with a number of side effects that can develop as treatment proceeds. Table 6 in Statement 4 shows the relative tendencies for antipsychotic medications to be associated with specific side effects. In addition, each of these side effects is described in further detail below.

Early in the course of treatment, common side effects include sedation; orthostatic changes in blood pressure; and anticholinergic side effects such as dry mouth, constipation, and difficulty with urination.

Of the side effects related to dopamine D2 receptor antagonist effects of antipsychotics, acute dystonia also appears early in treatment. It is particularly common with high-potency antipsychotic medications (e.g., haloperidol, fluphenazine) and can be life-threatening if associated with laryngospasm. Neuroleptic malignant syndrome (NMS) can also be life-threatening because of associated hyperthermia and autonomic instability. It typically occurs within the first month of antipsychotic treatment, with resumption of treatment, or with an increase in the dose of antipsychotic medication. Akathisia and medication-induced parkinsonism can also occur in the initial weeks of treatment or after increases in medication doses. Hyperprolactinemia, related to D2 receptor antagonism in the hypothalamic-pituitary axis, can lead to breast enlargement, galactorrhea, sexual dysfunction, and, in women, menstrual disturbances. These elevations in prolactin also occur in the initial weeks to months of treatment. On the other hand, tardive syndromes, including tardive dyskinesia, develop later, often months or even years after treatment initiation.

Side effects related to metabolic syndrome are common. Generally, they are observed in the initial months of treatment, but they can also occur later in treatment. These side effects include weight gain; hyperlipidemia; and glucose dysregulation, including development of diabetes mellitus.

Clozapine treatment is associated with a number of side effects that are less commonly seen with other antipsychotic medications. Severe neutropenia is most often seen early in treatment and is potentially life-threatening; however, with current regulatory requirements for monitoring ANC levels during treatment, it is rare. When seizures occur with clozapine, it is typically with very high doses or blood levels of clozapine, rapid increases in clozapine dose, or shifts in medication levels (related to drug-drug interactions or effects of smoking on drug metabolism). Myocarditis is infrequent and generally occurs early in treatment. Cardiomyopathy is rare and generally occurs later in the treatment course. Gastrointestinal effects of clozapine can also be significant, and in some patients it is associated with fecal impaction or paralytic ileus. Sialorrhea and tachycardia are each commonly observed during treatment with clozapine but can generally be managed conservatively.

Allergic and Dermatological Side Effects

Cutaneous allergic reactions occur infrequently with antipsychotic medications, but hypersensitivity can manifest as maculopapular erythematous rashes, typically of the trunk, face, neck, and extremities. Medication discontinuation or administration of an antihistamine is usually effective in reversing these symptoms.

In terms of other dermatological side effects, on rare occasions, thioridazine treatment is noted to be associated with hyperpigmentation of the skin. Dermatological reactions, including hyperpigmentation and cutaneous reactions, have also been reported with risperidone, clozapine, olanzapine, quetiapine, and haloperidol (Bliss and Warnock 2013). Photosensitivity reactions, resulting in severe sunburn, are also most commonly observed with low-potency phenothiazine medications. A blue-gray discoloration of the skin in body areas exposed to sunlight has been reported in patients receiving long-term chlorpromazine treatment. Consequently, patients who are taking these medications should be instructed to avoid excessive sunlight and use sunscreen.

Cardiovascular Effects

Hyperlipidemia

There is some evidence that certain antipsychotic medications, particularly clozapine and olanzapine, may increase the risk for hyperlipidemias (Buhagiar and Jabbar 2019; Bushe and Paton 2005; Meyer and Koro 2004; Mitchell et al. 2013a). However, there is also a suggestion that some patients may have a dyslipidemia prior to starting on antipsychotic treatment (Misiak et al. 2017; Pillinger et al. 2017b; Yan et al. 2013). Some patients develop an elevation of triglyceride levels in association with antipsychotic treatment that rarely is sufficiently high as to be associated with development of pancreatitis (Alastal et al. 2016). It is unclear whether triglyceridemia with antipsychotic treatment is a direct result of the medication or an indirect result of increased triglycerides in the blood with concomitant diabetes (Yan et al. 2013). In any patient with hyperlipidemia, it is also important to assess for other contributors to metabolic syndrome (Mitchell et al. 2012, 2013b) and ensure that the patient is receiving treatment with a lipid-lowering agent, as clinically indicated.

Myocarditis and Cardiomyopathy

Myocarditis and cardiomyopathy have been reported in some patients treated with clozapine and have resulted in death in some individuals. The etiology of these cardiac effects is unclear, although an immune-mediated mechanism has been suggested (Røge et al. 2012). For myocarditis, the reported incidence has varied from 0.015% to 8.5% (Bellissima et al. 2018). For reasons that are unclear, the highest rates have been reported in Australia (Ronaldson et al. 2015); rates elsewhere appear to be much lower. For example, an early study using the U.S. Clozaril National Registry found 17 confirmed cases of myocarditis in a total of 189,405 individuals who had received clozapine (La Grenade et al. 2001). A recent national registry study of outpatients in Denmark found 1 of 3,262 (0.03%) clozapine-treated patients developed myocarditis in the initial 2 months of treatment (Rohde et al. 2018). These authors estimated that a maximum of 0.28% of patients treated with clozapine would experience fatality due to clozapine-associated myocarditis, which is comparable to rates of cardiac adverse effects with other antipsychotic medications. For cardiomyopathy, the reported incidence is even less clear but appears to be considerably lower than rates of clozapine-associated myocarditis (Higgins et al. 2019; Khan et al. 2017; Rohde et al. 2018; Ronaldson et al. 2015).

Although cardiomyopathy has been reported throughout the course of clozapine treatment, the onset of myocarditis typically occurs during the first month of treatment and is heralded by shortness of breath, tachycardia, and fever (Bellissima et al. 2018; Ronaldson et al. 2015). Other features can include fatigue, chest pain, palpitations, and peripheral edema. Diagnosis can be challenging because of the nonspecific nature of these symptoms. For example, primary tachycardia is common with clozapine treatment without signifying underlying cardiac disease. Fever can also occur with clozapine initiation yet often resolves quickly and without evidence of myocarditis (Bruno et al. 2015; Lowe et al. 2007; Pui-yin Chung et al. 2008).

Recommendations for monitoring have varied, but there is no evidence or consensus that preemptive screening is necessary or helpful. However, if myocarditis or cardiomyopathy is suspected, a recent systematic review suggests seeking cardiology consultation as well as monitoring C-reactive protein and troponin (I and T subtypes) and obtaining an electrocardiogram as indicated (Knoph et al. 2018). Cardiac magnetic resonance imaging may also be indicated in some individuals.

In patients who do develop myocarditis or cardiomyopathy in conjunction with clozapine treatment, clozapine is typically discontinued. Subsequent decisions about resuming clozapine are individualized and should be based on the benefits and risks of treatment as compared with other therapeutic alternatives.

Orthostatic Hypotension

Orthostatic hypotension, a drop in blood pressure when changing from lying down or sitting to standing, is dose-related and due to the α-receptor-blocking effects of antipsychotic medications. When severe, orthostatic hypotension can cause syncope, dizziness, or falls. Older or severely debilitated patients, patients in the dose-titration phase of clozapine therapy, and patients with peripheral vascular disease or a compromised cardiovascular status may be at particular risk. Patients who experience orthostatic hypotension must be cautioned to sit on the edge of the bed for a minute before standing up, to move slowly when going from lying down or sitting to standing, and to seek assistance when needed. Management strategies for orthostatic hypotension include using supportive measures (e.g., use of support stockings, increased dietary salt and fluid intake); reducing the speed of antipsychotic dose titration; decreasing or dividing doses of antipsychotic medication; switching to an antipsychotic medication without antiadrenergic effects; and, as a last resort, administration of the salt/fluid-retaining corticosteroid fludrocortisone to increase intravascular volume (Mar and Raj 2018; Shen et al. 2017). For patients who are receiving concomitant antihypertensive treatment, adjustments to the dose of these medications may be needed.

QTc Prolongation

The QT interval on the electrocardiogram reflects the length of time required for ventricular repolarization and varies with heart rate (Funk et al. 2018). Several approaches exist for calculating a QT interval corrected for heart rate (QTc). Although the Bazett formula remains the one most widely used for drug monitoring and research, alternative correction formulas, such as the Fridericia and Framingham formulas, have been shown to most accurately correct for rate and improve prediction of mortality. In order to accurately predict risk, clinicians should be familiar with an alternative correction formula (Aytemir et al. 1999; Rautaharju et al. 2009; Vandenberk et al. 2016). Significant prolongation of the QTc interval is associated with increased risk for a ventricular tachyarrhythmia, torsades de pointes (TdP), which can lead to life-threatening consequences (e.g., ventricular fibrillation, sudden death). When the QTc interval is prolonged, a decision about the antipsychotic medication choice or changes requires a comprehensive risk-benefit assessment. A QTc interval > 500 msec is sometimes viewed as a threshold for concern; however, “there is no absolute QTc interval at which a psychotropic should not be used” (Funk et al. 2018, p. 2).

Studies that have examined the risks of QTc prolongation with antipsychotic treatment have varied in study quality, sample sizes, and the physical health of study subjects. Sources are available that categorize medications on the basis of their level of risk for QTc prolongation and TdP (Woosley et al. 2009), but the quality of data that informs such categorizations is also variable (Funk et al. 2018). Nevertheless, among the FGAs, chlorpromazine, droperidol, thioridazine, and pimozide appear to be associated with the greatest risk of QTc prolongation. The FDA recommends that thioridazine be used only when patients have not had a clinically acceptable response to other available antipsychotics (U.S. National Library of Medicine 2018b). Pimozide labeling also includes specific instructions related to medication dosing and QTc interval prolongation (U.S. National Library of Medicine 2018a). Orally administered haloperidol has been associated with only a mild increase in QTc interval length in healthy individuals; however, the risk of QTc interval prolongation and TdP appears to be greater with intravenous administration in medically ill individuals (Funk et al. 2018). Most SGAs have also been associated with some QTc interval prolongation, with ziprasidone and iloperidone appearing to have the greatest likelihood of QTc prolongation. The FDA has required that a warning about QTc prolongation be included with product labeling for ziprasidone (Geodon 2018), quetiapine (Quetiapine 2011; Seroquel XR 2019), iloperidone (Fanapt 2017), and paliperidone (Invega 2018, 2019; Invega Sustenna 2018a, 2018b; Invega Trinza 2018a, 2018b).

Factors to consider when making a determination about selecting or changing antipsychotic medications include whether the patient is taking other medications that are known to prolong QTc intervals; whether the patient has factors that would influence drug metabolism, leading to higher blood levels of a drug (e.g., poor metabolizer status, pharmacokinetic drug-drug interactions, hepatic or renal disease, drug toxicity); whether the patient is known to have a significant cardiac risk factor (e.g., congenital long QT syndrome, structural or functional cardiac disease, bradycardia, family history of sudden cardiac death); and other factors associated with an increased risk of TdP (e.g., female sex; advanced age; personal history of drug-induced QTc prolongation; severe acute illness; starvation; risk or presence of hypokalemia, hypomagnesemia, or hypocalcemia) (Funk et al. 2018). For individuals with these risk factors, antipsychotic medications with a regulatory warning or those with a known risk of QTc prolongation are not recommended for use if safer medication alternatives are available. Input from cardiology consultants should be considered when significant cardiac disease or other risk factors for QTc prolongation are present, although routine cardiology consultation is not indicated for patients without cardiac risk factors (Funk et al. 2018).

Tachycardia

Tachycardia can be primary (e.g., with clozapine), a reflex response to orthostatic hypotension, or a result of anticholinergic effects. It appears to be particularly common in individuals who are treated with clozapine (Lally et al. 2016a), but it may also be seen in individuals treated with other antipsychotic medications, particularly low-potency phenothiazines. Although healthy patients may be able to tolerate some increase in resting pulse rate, this may not be the case for patients with preexisting heart disease. In patients with significant tachycardia (heart rates above 110–120 bpm), ECG is warranted, as is an assessment for other potential causes of tachycardia (e.g., fever, anemia, smoking, hyperthyroidism, respiratory disease, cardiovascular disorders, caffeine and other stimulants, side effects of other medications). Early in treatment with clozapine, the possibility of myocarditis should be considered. Management strategies for tachycardia with antipsychotic medications include reducing the dose of medication, discontinuing medications with anticholinergic or stimulant properties, and using the strategies described above to reduce any contributing orthostatic hypotension. Case reports have discussed the use of medications such as β-blocking agents for persistent and significant tachycardia with clozapine. Nevertheless, treatment is not indicated unless the patient is symptomatic or the patient’s heart rate is substantially greater than 120 bpm because data from more rigorous studies are not available and these medications can contribute to other side effects, such as orthostatic hypotension (Lally et al. 2016a). If tachycardia is accompanied by pain, shortness of breath, fever, or signs of a myocardial infarction or heart rhythm problem, emergency assessment is essential.

Endocrine Side Effects

Glucose Dysregulation and Diabetes Mellitus

Evidence from meta-analyses of RCTs, population-based studies, and case-control studies suggests that some antipsychotic medications, clozapine and olanzapine in particular, are associated with an increased risk of hyperglycemia and diabetes (Hirsch et al. 2017; Ward and Druss 2015; Whicher et al. 2018; Zhang et al. 2017). Complicating the evaluation of antipsychotic-related risk of diabetes is that some patients with first-episode psychosis seem to have abnormal glucose regulation that precedes antipsychotic treatment (Greenhalgh et al. 2017; Perry et al. 2016; Pillinger et al. 2017a). In addition, obesity and treatment-related weight gain may contribute to diabetes risk. Nevertheless, there are some patients without other known risk factors who develop insulin resistance early in the course of antipsychotic treatment. In some individuals, diabetic ketoacidosis and nonketotic hyperosmolar coma have been reported in the absence of a known diagnosis of diabetes (Guenette et al. 2013; Kato et al. 2015; Liao and Phan 2014; Polcwiartek et al. 2016; Vuk et al. 2017). Given the rare occurrence of extreme hyperglycemia, ketoacidosis, hyperosmolar coma, or death and the suggestion from epidemiological studies of an increased risk of treatment-emergent adverse events with SGAs, the FDA has requested that all manufacturers of SGA medications include a warning in their product labeling regarding hyperglycemia and diabetes mellitus (Rostack 2003). When individuals with schizophrenia do develop diabetes, management principles should follow current guidelines for any patient with diabetes (Holt and Mitchell 2015; Scott et al. 2012). Given the existence of frequent health disparities for individuals with serious mental illness (Mangurian et al. 2016; Scott et al. 2012), clinicians can also help in ensuring that patients are obtaining appropriate diabetes care and encourage patients to engage in lifestyle interventions to improve diabetes self-management (Cimo et al. 2012). In any patient with diabetes, it is also important to assess for other contributors to metabolic syndrome (Mitchell et al. 2012, 2013b).

Hyperprolactinemia

Prolactin elevation is frequent in patients treated with antipsychotics (Ajmal et al. 2014; Cookson et al. 2012; Kinon et al. 2003; Lally et al. 2017a; Leucht et al. 2013; Rubio-Abadal et al. 2016), which increase prolactin secretion by blocking the inhibitory actions of dopamine on lactotrophic cells in the anterior pituitary. Consequently, hyperprolactinemia is observed more frequently with the use of antipsychotics that are more potent at blocking dopamine receptors (Tsuboi et al. 2013).

In both men and women, prolactin-related disruption of the hypothalamic-pituitary-gonadal axis can lead to decreased sexual interest and impaired sexual function (Kirino 2017; Rubio-Abadal et al. 2016). Other effects of hyperprolactinemia may include breast tenderness, breast enlargement, and lactation (Ajmal et al. 2014; Cookson et al. 2012). Because prolactin also regulates gonadal function, hyperprolactinemia can lead to decreased production of gonadal hormones, including estrogen and testosterone, resulting in disruption or elimination of menstrual cycles in women. In addition, in lactating mothers, suppression of prolactin may be detrimental, and the potential for this effect should be considered.

The long-term clinical consequences of chronic elevation of prolactin are poorly understood. Chronic hypogonadal states may increase the risk of osteopenia/osteoporosis and fractures may be increased in individuals with schizophrenia, but a direct link to antipsychotic-induced hyperprolactinemia has not been established (Bolton et al. 2017; Stubbs et al. 2014, 2015; Tseng et al. 2015; Weaver et al. 2019). In addition, some concern has been expressed about potential effects of hyperprolactinemia on the risk of breast or endometrial cancer; however, the available evidence suggests that such risks, if they exist, are likely to be small (De Hert et al. 2016; Froes Brandao et al. 2016; Klil-Drori et al. 2017; Pottegård et al. 2018; Wang et al. 2002).

If a patient is experiencing clinical symptoms of prolactin elevation, the dose of antipsychotic medication may be reduced, or the medication regimen may be switched to an antipsychotic with less effect on prolactin such as one with partial agonist activity at dopamine receptors (Ajmal et al. 2014; Grigg et al. 2017; Yoon et al. 2016). Administration of a dopamine agonist such as bromocriptine may also be considered.

Sexual Function Disturbances

A majority of patients with schizophrenia report some difficulties with sexual function. Although multiple factors are likely to contribute and rates vary widely depending on the study, it is clear that antipsychotic treatment contributes to sexual dysfunction (de Boer et al. 2015; La Torre et al. 2013; Marques et al. 2012; Serretti and Chiesa 2011; van Dijk et al. 2018). Effects of antipsychotic agents on sexual function may be mediated directly via drug actions on adrenergic and serotonergic receptors or indirectly through effects on prolactin and gonadal hormones (Kirino 2017; Knegtering et al. 2008; Rubio-Abadal et al. 2016). Loss of libido and anorgasmia can occur in men and in women; erectile dysfunction and ejaculatory disturbances also occur in men (La Torre et al. 2013; Marques et al. 2012; Serretti and Chiesa 2011; van Dijk et al. 2018). Retrograde ejaculation has also been reported with specific antipsychotic medications (e.g., thioridazine, risperidone) (Chouinard et al. 1993; de Boer et al. 2015; Kotin et al. 1976). In addition, it is important to note that priapism can also occur in association with antipsychotic treatment, particularly in individuals with other underlying risk factors such as sickle cell disease (Burnett and Bivalacqua 2011; Sood et al. 2008).

Despite the high rates of occurrence of sexual dysfunction with antipsychotic medication, many patients will not spontaneously report such difficulties. Thus, it is important to ask patients specifically about these side effects. Structured rating scales also exist to assess sexual side effects during antipsychotic treatment, and these can be used to supplement information obtained via interview (Clayton et al. 1997a, 1997b; de Boer et al. 2014; Depression and Bipolar Support Alliance 2019; Keller et al. 2006). Education about sexual side effects of medication can also be provided to the patient to communicate that these symptoms may occur but can be addressed (de Boer et al. 2015).

When sexual side effects of antipsychotic therapy are of significant concern to the patient, a reduction in medication dose or change in medication may be considered in addition to an assessment of other potential contributing factors (e.g., hyperprolactinemia, other medications, psychological factors) (de Boer et al. 2015; La Torre et al. 2013). Priapism, if it occurs, requires urgent urological consultation.

Gastrointestinal Side Effects

The most common gastrointestinal side effects of antipsychotic medications are related to anticholinergic side effects and include dry mouth and constipation, as noted in the subsection “Treatment-Emergent Side Effects of Antipsychotic Medications.” Patients and families should be educated about monitoring for constipation, and, if present, constipation should be reported promptly to clinicians. With clozapine in particular, gastrointestinal hypomotility can be severe and can result in fecal impaction or paralytic ileus (Every-Palmer and Ellis 2017; Leung et al. 2017; Palmer et al. 2008). Thus, if constipation is severe or does not resolve, the patient should obtain urgent medical care.

To prevent development of constipation in patients at increased risk (e.g., older patients, patients treated with clozapine), it is useful to minimize the doses and number of contributory medications such as other anticholinergic medications and opioids. A stool softener (e.g., docusate [Colace]) can be started. Activity and exercise should be encouraged to stimulate motility.

If constipation does develop, initial treatment can include stool softeners (e.g., docusate [Colace]) or osmotic laxatives (e.g., lactulose [Enulose], polyethylene glycol [Miralax], bisacodyl [Dulcolax]). Second-line treatments include stimulant laxatives (e.g., senna [Senokot], senna tea, cascara, sodium picosulfate). If constipation persists, an enema (e.g., Fleet) should be considered. A combination of treatments may be needed to treat constipation and then to prevent its recurrence.

Hepatic effects have also been reported with antipsychotic medications, including elevation of liver enzyme levels and cholestatic jaundice (U.S. Department of Health and Human Services and U.S. National Library of Medicine 2017b). Cholestatic jaundice is rare and has been reported primarily with chlorpromazine (U.S. Department of Health and Human Services and U.S. National Library of Medicine 2017a). It usually occurs within the first month after the initiation of treatment and generally requires discontinuation of treatment. However, given the relative infrequency of antipsychotic-induced jaundice, other etiologies for jaundice should be evaluated before the cause is judged to be antipsychotic medication.

Hematological Effects

Hematological effects are of greatest concern with clozapine; however, they have also been reported with other antipsychotic agents and may include inhibition of leukopoiesis, purpura, hemolytic anemia, and pancytopenia (Balon and Berchou 1986; Myles et al. 2019; Pisciotta 1969). For example, with chlorpromazine, transient benign leukopenia (white blood cell count < 3,500/mm3) is common, and severe neutropenia has been reported in 0.08% of patients, typically within the first few months of treatment (Pisciotta 1969).

There is no clear etiology of severe neutropenia or agranulocytosis with antipsychotic medications. With clozapine, a complex polygenic trait appears likely, perhaps involving the human leukocyte antigen locus or a group of hepatic transporter genes (de With et al. 2017; Legge et al. 2017). Initial estimates suggested that severe neutropenia would develop in 1%–2% of patients treated with clozapine, with fatal agranulocytosis in approximately 15% of those individuals (Alvir et al. 1993; Honigfeld et al. 1998). However, data from the initial 5 years of monitoring through clozapine registries showed a rate of severe neutropenia of 0.38%, with death occurring in only 3.1% of those cases (Honigfeld et al. 1998). A recent meta-analysis suggested an incidence of severe neutropenia in 0.9% of clozapine-treated patients, with a case fatality rate for individuals with severe neutropenia of 2.1% (Myles et al. 2018). For clozapine-treated patients as a group, the incidence of death due to severe neutropenia was 0.013% (Myles et al. 2018), suggesting that clozapine is quite safe with appropriate monitoring. Nevertheless, patients who are receiving clozapine should be advised to report any sign of infection (e.g., sore throat, fever, weakness, lethargy) immediately so that a decision can be made about obtaining additional evaluation.

If severe neutropenia does develop, it is usually reversible if clozapine is discontinued immediately and secondary complications (e.g., sepsis) are given intensive treatment. Granulocyte colony–stimulating factor has been used to accelerate granulopoietic function and shorten recovery time (Lally et al. 2017c; Myles et al. 2017).

Although there have been reports of successful resumption of clozapine after severe neutropenia, the risk of recurrence remains high (Lally et al. 2017b; Manu et al. 2018). For patients with a good clinical response to clozapine after multiple unsuccessful trials of other antipsychotic medications, the benefits and risks of rechallenge require thorough consideration and discussion with the patient and involved family members. Under such circumstances, case reports have suggested using granulocyte colony–stimulating factor to reduce the risk of recurrence, although evidence is limited (Lally et al. 2017b).

Neurological Side Effects

Acute Dystonia

Medication-induced acute dystonia is defined in DSM-5 as

[a]bnormal and prolonged contraction of the muscles of the eyes (oculogyric crisis), head, neck (torticollis or retrocollis), limbs, or trunk developing within a few days of starting or raising the dosage of a medication (such as a neuroleptic) or after reducing the dosage of a medication used to treat extrapyramidal symptoms. (American Psychiatric Association 2013a, p. 711)

A dystonic spasm of the axial muscles along the spinal cord can result in opisthotonos, in which the head, neck, and spinal column are hyperextended in an arched position. Rarely, acute dystonia can also present as life-threatening laryngospasm, which results in an inability to breathe (Ganesh et al. 2015; Koek and Pi 1989). Acute dystonia is sudden in onset and painful and can cause patients great distress. Because of the dramatic appearance of acute dystonia, health professionals who are unfamiliar with the condition may incorrectly attribute these reactions to catatonic signs or unusual behavior on the part of patients, and oculogyric crises can sometimes be misinterpreted as indicative of seizure activity.

In individuals treated with FGAs, it is estimated that up to 10% of patients may experience an acute dystonic episode, and with SGAs, rates of acute dystonia may be less than 2% (Martino et al. 2018; Miller et al. 2008; Satterthwaite et al. 2008). Additional factors that increase the risk of acute dystonia with antipsychotic medication include young age, male sex, ethnicity, recent cocaine use, high medication dose, and intramuscular route of medication administration (Gray and Pi 1998; Spina et al. 1993; van Harten et al. 1999). For further discussion of acute dystonia, including its treatment, see Statement 11.

Akathisia

Medication-induced acute akathisia is defined in DSM-5 as

[s]ubjective complaints of restlessness, often accompanied by observed excessive movements (e.g., fidgety movements of the legs, rocking from foot to foot, pacing, inability to sit or stand still), developing within a few weeks of starting or raising the dosage of a medication (such as a neuroleptic) or after reducing the dosage of a medication used to treat extrapyramidal symptoms. (American Psychiatric Association 2013a, p. 711)

Akathisia is sometimes difficult to distinguish from psychomotor agitation associated with psychosis, leading to a cycle of increasing doses of antipsychotic medication that lead to further increases in akathisia. Even in mild forms in which the patient is able to control most movements, akathisia is often extremely distressing to patients and is a frequent cause of nonadherence with antipsychotic treatment. If allowed to persist, akathisia can contribute to feelings of dysphoria and, in some instances, suicidal behaviors. The reported rates of akathisia vary from 10%–15% to as many as one-third of patients treated with antipsychotic medication, even when SGAs are used (Juncal-Ruiz et al. 2017; Martino et al. 2018; Mentzel et al. 2017; Miller et al. 2008). For further discussion of akathisia, including its treatment, see Statement 13.

Parkinsonism

Medication-induced parkinsonism, which is termed neuroleptic-induced parkinsonism and other medication-induced parkinsonism in DSM-5, is defined as

[p]arkinsonian tremor, muscular rigidity, akinesia (i.e., loss of movement or difficulty initiating movement), or bradykinesia (i.e., slowing movement) developing within a few weeks of starting or raising the dosage of a medication (e.g., a neuroleptic) or after reducing the dosage of a medication used to treat extrapyramidal symptoms. (American Psychiatric Association 2013a, p. 709)

These symptoms of medication-induced parkinsonism are dose-dependent and generally resolve with discontinuation of antipsychotic medication. It is important to appreciate that medication-induced parkinsonism can affect emotional and cognitive function, at times in the absence of detectable motor symptoms. As a result, it can be difficult to distinguish the negative symptoms of schizophrenia or concomitant depression from medication-induced parkinsonism. In addition, emotional and cognitive features of medication-induced parkinsonism can be subjectively unpleasant and can contribute to poor medication adherence (Acosta et al. 2012; Ascher-Svanum et al. 2006). For further discussion of medication-induced parkinsonism, including its treatment, see Statement 12.

Neuroleptic Malignant Syndrome

NMS is characterized by a classic triad of rigidity, hyperthermia (> 100.4°F/38.0°C on at least two occasions, measured orally), and sympathetic nervous system lability, including hypertension and tachycardia, in the context of exposure to a dopamine antagonist (or withdrawal of a dopamine agonist), typically within 72 hours of symptom development (American Psychiatric Association 2013a; Gurrera et al. 2011, 2017). In addition, NMS is associated with an elevated level of serum creatine kinase (typically, at least four times the upper limit of normal), tachypnea, change in mental status (e.g., delirium, stupor), and lack of another identified etiology for the symptoms. Notably, however, the onset and clinical features of NMS can vary and may make recognition more difficult. If misdiagnosed and mistreated, NMS can be fatal (Berman 2011; Rosebush and Stewart 1989; Strawn et al. 2007).

Other diagnostic considerations in patients presenting with possible NMS include malignant catatonia, malignant hyperthermia (in association with anesthetic administration), heat stroke (for which patients treated with antipsychotics have a heightened susceptibility), serotonin syndrome (in patients also taking serotonergic drugs such as selective serotonin reuptake inhibitors), benign elevations in the level of serum creatine kinase, fever in association with clozapine treatment, alcohol or sedative withdrawal, anticholinergic syndrome, hyperthermia associated with use of stimulants and hallucinogens, central nervous system infections, limbic encephalitis, and inflammatory or autoimmune conditions (American Psychiatric Association 2013a; Berman 2011; Rosebush and Stewart 1989; Strawn et al. 2007).

NMS has been reported with almost all medications that block dopamine receptors, but a greater risk of occurrence appears to be associated with high-potency FGAs (Schneider et al. 2018; Stübner et al. 2004). Risk also may be increased by use of short-acting intramuscular formulations of antipsychotic medications, use of higher total drug dosages, or rapid increases in the dosage of the antipsychotic medication (Keck et al. 1989; Sachdev et al. 1997; Viejo et al. 2003). Additional risk factors for NMS include acute agitation, dehydration, exhaustion, iron deficiency, physical illness, preexisting neurological disability, and a prior episode of NMS (American Psychiatric Association 2013a; Keck et al. 1989; Sachdev et al. 1997; Strawn et al. 2007).

Because NMS is rare, with an estimated incidence of 0.01%–0.02% among individuals treated with antipsychotics (Schneider et al. 2018; Stübner et al. 2004), most evidence regarding NMS treatment comes from single case reports or case series. Antipsychotic medications should always be discontinued, and supportive treatment to maintain hydration and to treat the fever and cardiovascular, renal, or other symptoms should be provided (American Psychiatric Association 2013a; Berman 2011; Strawn et al. 2007). NMS is usually self-limited, with resolution within a week of medication discontinuation in the majority of patients; however, prolonged symptoms of NMS do occur and may be associated with use of LAI antipsychotic medications (Caroff and Mann 1988; Caroff et al. 2000).

In addition to antipsychotic discontinuation and supportive care, a number of approaches have been used to treat NMS, although evidence is limited to case reports and case series (Pileggi and Cook 2016; Strawn et al. 2007). Benzodiazepines, such as lorazepam, have been used because of their benefits in treating catatonia and the parallels between malignant catatonia and NMS. As a postsynaptic D2 receptor agonist, bromocriptine has been used to counteract the dopamine antagonist effects of the antipsychotic medication. Dantrolene, a direct-acting skeletal muscle relaxant, has also been used, particularly in severe cases of NMS, because of its benefits in treating malignant hyperthermia. When NMS has not responded to these interventions or when catatonic symptoms persist after the resolution of NMS, case reports suggest that ECT can be beneficial (Caroff et al. 2000; Pileggi and Cook 2016; Strawn et al. 2007; Wittenauer Welsh et al. 2016). Assistance with emergency management of NMS is recommended and can be obtained through NMSContact (www.mhaus.org/nmsis/nmscontact).

Once NMS has resolved, caution is needed when resuming an antipsychotic medication because recurrence has been reported (Rosebush and Stewart 1989; Strawn et al. 2007; Susman and Addonizio 1988). Generally, when treatment is resumed, doses are increased gradually, and a medication other than the precipitating agent is used, typically one with a lower potency at blocking dopamine D2 receptors.

Seizures

Among the antipsychotic medications, clozapine is associated with the greatest likelihood of a seizure, and patients with a history of an idiopathic or medication-induced seizure may have a higher risk (Alldredge 1999; Devinsky and Pacia 1994; Wong and Delva 2007). Although generalized tonic-clonic seizures are most frequent, other types of seizures may occur. Seizures may also be preceded by myoclonus or drop attacks.

The seizure risk with clozapine is increased by rapid increases in dose as well as at high blood levels or doses of the drug. The overall seizure rate is 2.8%; with low-dose treatment (< 300 mg/day) the risk is 1%, with medium doses (300–599 mg/day) the risk is 2.7%, and with high doses (> 599 mg/day) the risk is 4.4% (Devinsky et al. 1991). Therefore, a slow initial titration of clozapine dose is essential, and patients should be cautioned not to drive or engage in other potentially hazardous activities while clozapine is being titrated. In individuals at high risk of seizure, prophylactic treatment with an anticonvulsant medication can be considered. FGAs can also lower the seizure threshold in a dose-related manner and result in the development of generalized tonic-clonic seizures (Alldredge 1999). Nevertheless, at usual dose ranges, seizure rates are below 1% for all FGAs.

In patients who do experience a seizure while taking clozapine or another antipsychotic medication, neurological consultation will be important for delineating the risks of a further seizure, determining whether anticonvulsant therapy (e.g., valproate) is indicated, and collaborating with the clinician in determining whether changes to the patient’s antipsychotic regimen are indicated (Alldredge 1999; Wong and Delva 2007).

Tardive Syndromes, Including Tardive Dyskinesia

Tardive syndromes are persistent abnormal involuntary movement disorders caused by sustained exposure to antipsychotic medication, the most common of which are tardive dyskinesia, tardive dystonia, and tardive akathisia (Frei et al. 2018). They begin later in treatment than acute dystonia, akathisia, or medication-induced parkinsonism, and they persist and may even increase, despite reduction in dose or discontinuation of the antipsychotic medication. Typically, tardive dyskinesia presents as “[i]nvoluntary athetoid or choreiform movements (lasting at least a few weeks) generally of the tongue, lower face and jaw, and extremities (but sometimes involving the pharyngeal, diaphragmatic, or trunk muscles)” (American Psychiatric Association 2013a, p. 712), whereas tardive dystonia and tardive akathisia resemble their acute counterparts in phenomenology.

Tardive dyskinesia has been reported after exposure to any of the available antipsychotic medications (Carbon et al. 2017, 2018). It occurs at a rate of approximately 4%–8% per year in adult patients treated with FGAs (Carbon et al. 2018; Woods et al. 2010), a risk that appears to be at least three times that observed with SGAs (Carbon et al. 2018; O’Brien 2016; Woods et al. 2010). Various factors are associated with greater vulnerability to tardive dyskinesia, including age greater than 55 years; female sex; white or African race/ethnicity; presence of a mood disorder, intellectual disability, or central nervous system injury; and past or current akathisia, clinically significant parkinsonism, or acute dystonic reactions (Patterson-Lomba et al. 2019; Solmi et al. 2018a).

Although the majority of patients who develop tardive dyskinesia have mild symptoms, a small proportion will develop symptoms of moderate or severe degree. Tardive dyskinesia can have significant effects on quality of life and can be associated with social withdrawal (McEvoy et al. 2019). Although the impact appears to be influenced by the severity of tardive dyskinesia, individuals with mild symptoms can also experience negative effects on quality of life.

Evaluation of the risk of tardive dyskinesia is complicated by the fact that dyskinetic movements may be observed with a reduction in antipsychotic medication dose, which is termed a withdrawal-emergent dyskinesia (American Psychiatric Association 2013a). Fluctuations in symptoms are also common and may be influenced by such factors as psychosocial stressors. Furthermore, spontaneous dyskinesias, which are clinically indistinguishable from tardive dyskinesia, have been described in elderly patients before the advent of antipsychotic medications and in up to 20% of never-medicated patients with chronic schizophrenia (Blanchet et al. 2004; Crow et al. 1982; Fenton et al. 1997; Saltz et al. 1991). In longer-term studies, findings are often confounded by the sequential or concomitant use of more than one antipsychotic medication and the lack of systematic prospective assessments for the presence of a movement disorder (Tarsy and Baldessarini 2006). Nevertheless, evaluation for the presence of tardive syndromes is important in order to identify them, minimize worsening, and institute clinically indicated treatment. For further discussion of tardive syndromes, including their treatment, see Statement 14.

Ophthalmological Effects

The most common ophthalmological effects of antipsychotic medications are related to the anticholinergic effects of these agents and include blurred vision and exacerbation of open-angle glaucoma. Pigmentary retinopathies and corneal opacities can occur with chronic administration of the low-potency medications thioridazine and chlorpromazine, particularly at high doses (e.g., more than 800 mg/day of thioridazine) (Matsuo et al. 2016). With SGAs, including quetiapine, evidence does not suggest any increase in the likelihood of cataract development (Laties et al. 2015; Pakzad-Vaezi et al. 2013). If patients do undergo cataract surgery, however, there have been case reports of intraoperative floppy-iris syndrome in individuals treated with antipsychotic medications, a complication that has been associated with use of medications that block α1-adrenergic receptors (Chatziralli and Sergentanis 2011). Although adverse ophthalmological effects of antipsychotic medications are infrequent, encouraging regular eye care is important to maintaining good vision for individuals with schizophrenia (Viertiö et al. 2007), particularly because of high rates of diabetes and other health conditions that can affect sight.

Other Side Effects

Anticholinergic Effects

The anticholinergic effects of some antipsychotic medications (along with the anticholinergic effects of antiparkinsonian medications, if concurrently administered) can produce a variety of peripheral side effects, including dry mouth, blurred vision, constipation, tachycardia, urinary retention, and effects on thermoregulation (e.g., hyperthermia in hot weather) (Nasrallah and Tandon 2017; Ozbilen and Adams 2009). Central anticholinergic effects can include impaired learning and memory and slowed cognition (Ang et al. 2017; Vinogradov et al. 2009).

Because most anticholinergic side effects are mild and tolerable, they are often overlooked. Nevertheless, they can have multiple implications for patients, including impaired quality of life and significant health complications (Salahudeen et al. 2015). For example, dry mouth is associated with an increased risk for multiple dental complications (Singh and Papas 2014), and drinking high-calorie fluids in response to dry mouth can contribute to weight gain. The muscarinic receptor antagonist properties of antipsychotic drugs can be particularly problematic in older individuals and can contribute to problems such as urinary retention, confusion, fecal impaction, and anticholinergic toxicity (with delirium, somnolence, and hallucinations) (Nasrallah and Tandon 2017). Anticholinergic properties of antipsychotic or antiparkinsonian medications can also precipitate acute angle-closure glaucoma (Lachkar and Bouassida 2007), although patients with treated glaucoma seem to be able to tolerate these medications with careful monitoring (Bower et al. 2018).

The propensity of an antipsychotic medication to cause anticholinergic effects should be considered when choosing an antipsychotic agent initially, particularly in older individuals or those with physical conditions that may confer a greater risk of anticholinergic complications. In selecting a medication, it is also important to keep in mind the total anticholinergic burden from antipsychotic medications, antiparkinsonian medications, urological medications (e.g., oxybutynin), nonselective antihistamines (e.g., hydroxyzine, diphenhydramine), and other medications with anticholinergic side effects. For this reason, antiparkinsonian medications with anticholinergic properties are not typically administered on a prophylactic basis. When anticholinergic side effects do occur, they are often dose-related and thus may improve with lowering of the dose or administering the medications that have anticholinergic properties in divided doses. For additional discussion of anticholinergic properties of antiparkisonian medications, see Statement 12.

Fever

Fever (> 38°C) should prompt assessment for possible etiologies, including NMS or infection. In hot weather, the possibility of heat stroke should be considered in patients who do not have access to air-conditioned environments due to the increased risk of heat-related events in individuals with psychiatric illness (Bouchama et al. 2007) and the effects of some antipsychotics and anticholinergic agents on thermoregulation (Martin-Latry et al. 2007). In patients who are treated with clozapine, a brief self-limiting fever may occur during the first few weeks of treatment and responds to supportive measures (Bruno et al. 2015; Lowe et al. 2007; Pui-yin Chung et al. 2008). However, it is also essential to assess for the presence of potentially life-threatening complications, including NMS, severe neutropenia, and myocarditis.

Sedation

Sedation is a very common side effect of antipsychotic medications (Citrome 2017a; Leucht et al. 2013). This effect may be related to antagonist effects of those drugs on histamine, adrenergic, and dopamine receptors (Michl et al. 2014). Sedation is most pronounced in the initial phases of treatment, and many patients develop some tolerance to the sedating effects with continued administration. For agitated patients, the sedating effects of these medications in the initial phase of treatment can have therapeutic benefits. Bedtime sedation can also be desirable for patients who are having difficulty sleeping. However, persistent sedation, including daytime drowsiness, increased sleep time, and reduced cognitive acuity, can interfere with social, recreational, and vocational function.

Lowering of the daily dose, consolidation of divided doses into one evening dose, or changing to a less sedating antipsychotic medication may be effective in reducing the severity of sedation. Coffee or other caffeine can be helpful in the morning but can also interact with medications (e.g., contribute to tachycardia; raise blood levels of medications, including clozapine). Adding a stimulant medication is not typically helpful and can lead to additional side effects. If sedation or the risk of sedation is significant (e.g., during initial clozapine titration), patients should be cautioned not to drive or engage in potentially hazardous activities.

Sialorrhea

Sialorrhea (or hypersalivation) is a frequent side effect of clozapine (Maher et al. 2016) but can also be observed with other antipsychotic medications (Essali et al. 2013). The etiology of sialorrhea is unclear but may relate to decreased saliva clearance, although actions on muscarinic or α-adrenergic receptors have also been postulated (Ekström et al. 2010). Sialorrhea can contribute to reductions in quality of life and can also be associated with complications such as aspiration pneumonia (Dzahini et al. 2018; Kaplan et al. 2018; Stoecker et al. 2017).

During the day, patients can be encouraged to chew sugarless gum, which stimulates the swallowing reflex. Because sialorrhea may be more bothersome at night, patients may be advised to place a towel on their pillow and change to a clean towel in the middle of the night to minimize discomfort. Pharmacological approaches to address sialorrhea come from small studies and case reports and include use of low-dose or topical anticholinergic medications, such as glycopyrrolate or sublingual ophthalmic atropine 1% drops (Bird et al. 2011; Liang et al. 2010; Man et al. 2017). Diphenhydramine has also been studied (Chen et al. 2019); however, because clozapine and other antipsychotics can have significant anticholinergic properties themselves and anticholinergics have small effects on sialorrhea, the use of agents with added anticholinergic effects should be approached cautiously. Terazosin and, in severe refractory cases, botulinum toxin have also been used (Bird et al. 2011; Liang et al. 2010; Man et al. 2017).

Weight Gain

Weight gain occurs with most antipsychotic agents and appears to relate to actions of these medications as histamine H1 receptor antagonists, although actions on serotonin and muscarinic receptors may also play a role (He et al. 2013; Kroeze et al. 2003; Michl et al. 2014; Olten and Bloch 2018). Reviews and meta-analyses have compared average weight gains with antipsychotic treatment and the proportion of patients who gain 7% or more of body weight (Bak et al. 2014; Leucht et al. 2013; Zhang et al. 2013). Nevertheless, there is substantial variability in the amount of weight gain that will occur in an individual patient who is treated with a specific antipsychotic medication. Typically, weight gain is progressive over the first 6 months of treatment, although some patients continue to gain weight indefinitely (Alvarez-Jimenez et al. 2008). In addition, younger individuals who are experiencing a first episode of psychosis may be more likely than older individuals to gain weight with antipsychotic medication (Correll et al. 2014; Jensen et al. 2019). In identifying individuals with schizophrenia who experience weight gain with antipsychotic treatment, self-reported awareness may be less effective than objective measurement (Gao et al. 2016).

Obesity, in general, can contribute to an increase in risk for mortality and morbidity, including increased rates of cardiovascular disease, hypertension, cancers, diabetes, osteoarthritis, and sleep apnea (Aune et al. 2016; Bellou et al. 2018; Jehan et al. 2018; Lauby-Secretan et al. 2016; Stringhini et al. 2017). Consequently, weight gain with antipsychotic medications is also likely to contribute to an increase in physical health conditions and mortality. Prevention of weight gain should, thus, be a high priority because weight loss is difficult for most patients. Efforts should be made to intervene proactively with weight gain of 5–10 pounds because people who are obese rarely lose more than 10% of body weight with weight loss regimens.

A number of studies have evaluated the effectiveness of specific interventions to prevent or treat antipsychotic-induced weight gain (Caemmerer et al. 2012; Das et al. 2012; de Silva et al. 2016; Gierisch et al. 2014; Mahmood et al. 2013; Manu et al. 2015; Mizuno et al. 2014; Mukundan et al. 2010; Vancampfort et al. 2019; Zheng et al. 2015). Nutritional interventions have shown small but consistent benefits (Bonfioli et al. 2012), although patients who are willing to enroll in and are able to adhere to such studies may not be representative. Nevertheless, nutritional approaches may be suggested for their benefits for overall health as well as for weight. Such approaches include specialized behavioral health interventions, in-person community interventions (e.g., Weight Watchers), services that include meal delivery (e.g., Jenny Craig), and Internet-based interventions (e.g., Omada Health). In addition, some programs have begun to integrate dietitians into the treatment team, given the nutritional challenges that exist for many individuals with serious mental illness (Teasdale et al. 2017). Other nonpharmacological approaches that have been studied include exercise and cognitive-behavioral therapy approaches (Bonfioli et al. 2012; Caemmerer et al. 2012; Das et al. 2012).

In collaboration with the patient’s primary care clinician, medication strategies for weight loss can be considered. Of the pharmacological treatments that have been assessed, metformin has been studied most often. It has been shown to be safe in individuals without hyperglycemia, shows modest benefits on weight (with average weight loss of 3–4 kg), and can reverse metabolic abnormalities in patients with obesity or other metabolic problems (Das et al. 2012; de Silva et al. 2016; McGinty et al. 2016; Mizuno et al. 2014; Siskind et al. 2016a; Vancampfort et al. 2019; Zheng et al. 2015; Zhuo et al. 2018). However, most studies have been small, and follow-up periods have not been longer than 6 months. Modest benefit has also been seen in several studies of glucagon-like peptide-1 receptor agonists (Siskind et al. 2019) and small studies of topiramate (Mahmood et al. 2013; Mizuno et al. 2014; Vancampfort et al. 2019; Zhuo et al. 2018). Other medications have been examined in small trials or case series, with less consistent findings (Mizuno et al. 2014). This limited evidence and the modest benefit of these pharmacological treatments need to be considered in light of potential adverse effects.

Another consideration for a patient who has experienced significant weight gain with antipsychotic treatment is to change or augment treatment with a medication with lower weight-gain liability (Vancampfort et al. 2019). When possible, other medications that can cause weight gain (e.g., valproate) should be tapered and discontinued. Such decisions need to consider the extent of the patient’s response to the current medication regimen, the risks to the patient if relapse occurs with a medication change, and the likelihood that a medication change will be beneficial in terms of weight loss or other side effects (Manu et al. 2015; Mukundan et al. 2010; Newcomer et al. 2013; Vancampfort et al. 2019). In any patient with weight gain, it is also important to assess for other contributors to metabolic syndrome (Mitchell et al. 2012, 2013b).

The benefits of exercise appear to be small in terms of weight loss in individuals with schizophrenia (Firth et al. 2015; Pearsall et al. 2014; Vancampfort et al. 2017, 2019). Nevertheless, many individuals with schizophrenia do not engage in physical activity (Stubbs et al. 2016a; Vancampfort et al. 2016b), and exercise can be suggested for its benefits to overall health, improved cardiorespiratory fitness, and other aspects of functioning (Dauwan et al. 2016; Firth et al. 2015, 2017; Vancampfort et al. 2017, 2019). Health promotion coaching interventions focused on individuals with mental illness, such as the In SHAPE program, can also be pursued and may be associated with weight loss and reduced cardiovascular risk (Bartels et al. 2015; Naslund et al. 2016; Van Citters et al. 2010; Verhaeghe et al. 2013).

Balancing of Potential Benefits and Harms in Rating the Strength of the Guideline Statement

Benefits

Use of an antipsychotic medication in the treatment of schizophrenia can improve positive and negative symptoms of psychosis (high strength of research evidence) and can also lead to reductions in depression and improvements in quality of life and functioning (moderate strength of research evidence). A meta-analysis of double-blind, randomized, placebo-controlled trials showed a medium effect size for overall efficacy (Leucht et al. 2017), with the greatest effect on positive symptoms. The rates of achieving any response or a good response were also significantly greater in patients who received an antipsychotic medication. In addition, the proportion of individuals who dropped out of treatment for any reason and for lack of efficacy was significantly less in those who were treated with an antipsychotic medication. Research evidence from head-to-head comparison studies and network meta-analysis (McDonagh et al. 2017) showed no consistent evidence that favored a specific antipsychotic medication, with the possible exception of clozapine.

Harms

The harms of using an antipsychotic medication in the treatment of schizophrenia include sedation, side effects mediated through dopamine receptor blockade (e.g., acute dystonia, akathisia, parkinsonism, tardive syndromes, NMS, hyperprolactinemia), disturbances in sexual function, anticholinergic effects, weight gain, glucose abnormalities, hyperlipidemia, orthostatic hypotension, tachycardia, and QTc prolongation. Clozapine has additional harms associated with its use, including sialorrhea, seizures, neutropenia (which can be severe and life-threatening), myocarditis, and cardiomyopathy. Among the antipsychotic medications, there is variability in the rates at which each of these effects occurs, and no specific medication appears to be devoid of possible side effects.

Patient Preferences

Clinical experience suggests that many patients are cooperative with and accepting of antipsychotic medications as part of a treatment plan. A survey of patient preferences reported that patients viewed an ability to think more clearly and an ability to stop hallucinations or paranoia as important efficacy-related reasons to take an antipsychotic medication (Achtyes et al. 2018). However, patients also reported concerns about side effects, particularly weight gain, sedation, and restlessness, as reasons that they might not wish to take antipsychotic medications. Some patients might also choose not to take an antipsychotic medication when they are feeling well or if they do not view themselves as having a condition that requires treatment. Some patients may also prefer one medication over another medication on the basis of prior treatment experiences or other factors.

Balancing of Benefits and Harms

The potential benefits of this guideline statement were viewed as far outweighing the potential harms. Although harms of antipsychotic medications can be significant, the impact of schizophrenia on patients’ lives is also substantial, and consistent benefits of antipsychotic treatment were found. Harms of treatment can be mitigated by selecting medications on the basis of individual characteristics and preferences of patients as well as by choosing a medication on the basis of its side-effect profile, pharmacological characteristics, and other factors. For clozapine, the additional benefits of treatment were viewed as outweighing the additional rare but serious harms and the need for ANC monitoring to reduce the likelihood of severe neutropenia. For additional discussion of the research evidence, see Appendix C, Statement 4.

Differences of Opinion Among Writing Group Members

There were no differences of opinion. The writing group voted unanimously in favor of this recommendation.

Review of Available Guidelines From Other Organizations

Information from other guidelines is consistent with this guideline statement. Other guidelines on the treatment of schizophrenia (BAP, Canadian Schizophrenia Guidelines [CSG], NICE, PORT, RANZCP, SIGN, WFSBP) all recommend use of an antipsychotic medication in the treatment of schizophrenia, with the selection of a specific medication on an individualized basis with consideration of medication characteristics, patient characteristics, and patient preferences (Addington et al. 2017a, 2017b; Barnes et al. 2011; Buchanan et al. 2010; Crockford and Addington 2017; Galletly et al. 2016; Hasan et al. 2012; National Institute for Health and Care Excellence 2014; Pringsheim et al. 2017; Remington et al. 2017; Scottish Intercollegiate Guidelines Network 2013). Each guideline also recommends monitoring during the course of treatment to assess therapeutic response and treatment-related side effects.

Quality Measurement Considerations

In clinical practice, almost all individuals with schizophrenia are offered an antipsychotic medication. Thus, a quality measure is unlikely to enhance outcomes if it examines only whether an individual with schizophrenia is offered or receives an initial prescription for antipsychotic treatment. An existing National Quality Forum–endorsed measure, “Adherence to Antipsychotic Medications for Individuals with Schizophrenia” (NQF #1879, www.qualityforum.org/QPS/1879), is aimed at assessing whether an antipsychotic medication is continued once it is begun. For individuals who are at least 18 years old and who have a diagnosis of schizophrenia or schizoaffective disorder, this measure assesses the percentage who have been dispensed an antipsychotic medication (as reflected by at least two such prescriptions being filled) and who had a proportion of covered days of at least 0.8 during a 12 consecutive month measurement period. By requiring ongoing prescribing of antipsychotic medication, this measure is more likely to be associated with improvements in outcomes for patients. Nevertheless, this measure does have several limitations. It uses pharmacy claims data or electronic prescription orders to examine whether a medication has been prescribed, but such measures do not guarantee treatment adherence. For instance, a prescriber could submit an antipsychotic medication prescription and the patient could fill the prescription at the pharmacy, but the patient might not actually take the medication. This measure also does not determine the adequacy of the medication or the medication dose and could be met through continuous prescriptions of a subtherapeutic dose or clinically ineffective antipsychotic. Another limitation is the difficulty in determining the proportion of covered days in a 12 consecutive month period, particularly when patients have transitions in care between settings or treating clinicians.

Quality measures, quality improvement initiatives, or electronic decision supports may be appropriate for monitoring side effects of antipsychotic treatment. Evidence suggests that rates of guideline concordant monitoring are low for metabolic risk factors, including lipids, diabetes, and weight (Mitchell et al. 2012; Morrato et al. 2009). Several measures endorsed by NQF address such monitoring. NQF #1932, “Diabetes Screening for People With Schizophrenia or Bipolar Disorder Who Are Using Antipsychotic Medications” (www.qualityforum.org/QPS/1932) measures the percentage of patients ages 18–64 years with schizophrenia or bipolar disorder “who were dispensed an antipsychotic medication and had a diabetes screening test during the measurement year.” Because this measure is focused on screening, it excludes monitoring of individuals who had diabetes in the measurement year or in the preceding year.

NQF #1927, “Cardiovascular Health Screening for People With Schizophrenia or Bipolar Disorder Who Are Prescribed Antipsychotic Medications” (www.qualityforum.org/QPS/1927) measures the percentage of individuals ages 25–64 years with schizophrenia or bipolar disorder “who were prescribed any antipsychotic medication and who received a cardiovascular health screening during the measurement year,” where cardiovascular health screening consists of “one or more LDL-C screenings performed during the measurement year.” Individuals are excluded from this screening measure for having evidence of preexisting cardiovascular disease as defined by precise criteria in the measure text. Presently, the specific elements of these criteria can often be challenging to determine from unstructured electronic health records.

Two additional NQF-approved measures, NQF #1933 (www.qualityforum.org/QPS/1933) and NQF #1934 (www.qualityforum.org/QPS/1934) address cardiovascular and diabetes monitoring, respectively, for individuals with preexisting cardiovascular disease or diabetes. Each of these measures is limited to individuals who are ages 18–64 years. The cardiovascular monitoring measure requires that individuals receive “an LDL-C test performed during the measurement year,” whereas the diabetes monitoring measure requires “[o]ne or more HbA1c tests and one or more LDL-C tests performed during the measurement year.” These measures have been tested for feasibility, usability, reliability, and validity at the health plan, integrated delivery system, and population levels; however, before holding individual clinicians or facilities accountable for the delivered quality of care, the measures would need additional testing at these levels.

Statement 5: Continuing Medications

APA recommends (1A) that patients with schizophrenia whose symptoms have improved with an antipsychotic medication continue to be treated with an antipsychotic medication.*

*This guideline statement should be implemented in the context of a person-centered treatment plan that includes evidence-based nonpharmacological and pharmacological treatments for schizophrenia.

Implementation

For individuals with a diagnosis of schizophrenia whose symptoms have improved with an antipsychotic medication, there are a number of benefits to maintenance treatment, including reduced risks of relapse (Bowtell et al. 2018; Goff et al. 2017; Hui et al. 2018; Kishi et al. 2019; Leucht et al. 2012; Thompson et al. 2018), rehospitalization (Tiihonen et al. 2018), and death (Tiihonen et al. 2018; Vermeulen et al. 2017). When administered on a long-term basis, however, antipsychotic medications are also associated with a greater incidence of weight gain, sedation, and movement disorders (Leucht et al. 2012). In addition, some studies have raised questions about whether long-term antipsychotic treatment might be associated with other adverse effects on functioning or health, including loss of brain volume (Davidson 2018; Goff et al. 2017). These data are heterogeneous, and when compared with withholding treatment, there was minimal evidence to suggest negative effects of maintenance treatments on outcomes (Goff et al. 2017; Huhtaniska et al. 2017). In addition, it may be possible to mitigate these risks by preventive interventions (e.g., early intervention for weight gain, screening for lipid and glucose abnormalities) and careful monitoring for side effects of medication. Nevertheless, as treatment proceeds, the pluses and minuses of continuing treatment with an antipsychotic medication should be reviewed with the patient in the context of shared decision-making. It will typically be beneficial to include family members or other persons of support in such discussions (Hamann and Heres 2019).

Despite the benefits of continued antipsychotic treatment for the majority of patients, maintaining adherence to an antipsychotic medication can be difficult (Acosta et al. 2012; Shafrin et al. 2016; Valenstein et al. 2006). Barriers to, facilitators of, and motivators of treatment adherence will differ for each patient. Engaging family members or other persons of support can be helpful in fostering adherence (Mueser et al. 2015). Other approaches to assessing and enhancing adherence are described in detail in Statement 3.

For some patients, the formulation of the antipsychotic medication may influence adherence (see Statement 4, Table 3). For example, rapidly dissolving tablets or oral concentrates may be preferable for patients who have difficulty swallowing pills or who are ambivalent about medications and inconsistent in swallowing them. LAI formulations may be preferred by some patients (Heres et al. 2007; Patel et al. 2009; Walburn et al. 2001) and may be particularly useful for patients with a history of poor or uncertain adherence (see Statement 10).

It is also important to assess the ongoing benefits and side effects of treatment that may indicate a need for adjustments to medication doses or changes in medications. The use of quantitative measures can be helpful in systematically assessing each of these realms (see Statement 2). The optimal dose of medication is one that provides the best medication benefits yet is tolerable in terms of medication side effects. For some patients, adjustments in dose will be required during the course of treatment to maintain this balance (Essock et al. 2006). Such factors as addition or discontinuation of interacting medications, changes in smoking status, changes in patient body mass, changes in renal or hepatic status, or changes in drug absorption (e.g., with bariatric surgery) may influence medication pharmacokinetics and require increases or decreases in medication dose. In order to minimize the risk of extended side effects, when medications with a long half-life are used and particularly when LAI antipsychotic medications are used, considerations about changes in dose need to consider the extended actions of these medications.

Evidence on the rationale and approach to planned reductions of medication doses is limited. Unless a medication requires emergent discontinuation, gradual reductions in doses are preferable, with close monitoring for recurrent symptoms.

Shared decision-making discussions with the patient should consider the patient’s recovery goals, the potential benefits of medication changes or dose reductions in terms of changes or diminution of side effects, and the potential harms of medication changes or dose reductions (Davidson 2018). The longitudinal course of the patient’s episode and the certainty of the diagnosis should also be considered. There may be some individuals with a brief episode of psychosis or uncertain psychotic diagnosis (e.g., possible substance-induced psychosis or mood-related psychosis) who may not require continuing antipsychotic treatment. On the other hand, individuals with chronic symptoms, repeated relapses, and clear diagnostic features of schizophrenia will likely have poorer outcomes if medications are stopped. In addition to symptom recurrence and relapse, medication cessation may be associated with hospitalization, legal difficulties, reduced likelihood of response with reinstatement of treatment, or poorer psychosocial outcomes (Correll et al. 2018; Hui et al. 2018; Takeuchi et al. 2019; Wilper et al. 2009). It will typically be beneficial to include family members or other persons of support in discussions of medication changes or dose reductions.

Balancing of Potential Benefits and Harms in Rating the Strength of the Guideline Statement

Benefits

Use of an antipsychotic medication that has already been associated with symptom response can maintain improvements in symptoms as well as promote enhanced functioning and quality of life (high strength of research evidence). Long-term treatment with an antipsychotic medication has also been associated with a reduction in mortality as compared with no antipsychotic treatment in individuals with schizophrenia. In contrast, discontinuation of antipsychotic treatment can be associated with increases in symptoms and risk of hospitalization and poorer long-term outcomes, including greater mortality in the long term (low strength of research evidence).

Harms

The harms of continuing use of an antipsychotic medication can vary depending on whether the patient is experiencing any significant side effects from the medication that would have long-term untoward effects. For patients whose medications are well tolerated, long-term risks include tardive syndromes from antipsychotic medications. For other patients, long-term risks will vary according to the specific side effect, with metabolic effects of antipsychotic medication serving as a possible contributor to long-term health risks. Some studies have raised concerns about changes in brain region volumes with antipsychotic treatment, but these findings are heterogeneous and inconsistent.

Patient Preferences

Clinical experience suggests that many patients are cooperative with and accepting of antipsychotic medications as part of a treatment plan. This is particularly true when the medication has been associated with a response in symptoms. Indeed, a survey of patient preferences reported that patients viewed an ability to think more clearly and an ability to stop hallucinations or paranoia as important efficacy-related reasons to take an antipsychotic medication (Achtyes et al. 2018). Patients are also likely to value the long-term benefits that have been shown with continued antipsychotic treatment, including reductions in relapses, hospitalizations, and mortality. However, patients also report concerns about side effects, particularly weight gain, sedation, and restlessness, that can make them reluctant to take antipsychotic medications on a long-term basis. In addition, some patients may choose not to take an antipsychotic medication when they are feeling well or if they do not view themselves as having a condition that requires treatment.

Balancing of Benefits and Harms

The potential benefits of this guideline statement were viewed as far outweighing the potential harms. Although harms of antipsychotic medications can be significant and the long-term effects of antipsychotic medications are not well studied, the impact of schizophrenia on patients’ lives is also substantial, and consistent benefits of continued antipsychotic treatment were found. Overall, rates of mortality appear to be reduced by ongoing treatment with an antipsychotic medication as compared with no treatment. In addition, harms of treatment can be mitigated by using the lowest effective dose; by selecting medications on the basis of individual characteristics and preferences of patients; and by choosing a medication on the basis of its side-effect profile, pharmacological characteristics, and other factors. For additional discussion of the research evidence, see Appendix C, Statement 5.

Differences of Opinion Among Writing Group Members

There were no differences of opinion. The writing group voted unanimously in favor of this recommendation.

Review of Available Guidelines From Other Organizations

Information from other guidelines is consistent with this guideline statement. Other guidelines on the treatment of schizophrenia (BAP, NICE, PORT, SIGN, WFSBP) recommend continued use of an antipsychotic medication in the treatment of schizophrenia once symptom response has been achieved (Barnes et al. 2011; Buchanan et al. 2010; Hasan et al. 2013; National Institute for Health and Care Excellence 2014; Scottish Intercollegiate Guidelines Network 2013). Other guidelines (RANZCP, BAP, SIGN, NICE) also suggest the use of LAI antipsychotic medications on the basis of patient preference or when adherence has been poor or uncertain (Barnes et al. 2011; Galletly et al. 2016; National Institute for Health and Care Excellence 2014; Scottish Intercollegiate Guidelines Network 2013). Use of a gradual reduction in dose, including a gradual cross-taper when changing medications, is noted by several guidelines (BAP, NICE, SIGN) along with an emphasis on close monitoring for signs of relapse (Barnes et al. 2011; National Institute for Health and Care Excellence 2014; Scottish Intercollegiate Guidelines Network 2013).

Quality Measurement Considerations

See Statement 4 for a discussion of quality measures related to initiation and ongoing use of an antipsychotic medication.

Statement 6: Continuing the Same Medications

APA suggests (2B) that patients with schizophrenia whose symptoms have improved with an antipsychotic medication continue to be treated with the same antipsychotic medication.*

*This guideline statement should be implemented in the context of a person-centered treatment plan that includes evidence-based nonpharmacological and pharmacological treatments for schizophrenia.

Implementation

As noted in Statement 5, it is important for treatment with an antipsychotic medication to be maintained once symptoms have improved. Specifically, for individuals with a diagnosis of schizophrenia, there are a number of benefits to continued treatment with an antipsychotic medication, including reduced risks of relapse (Bowtell et al. 2018; Goff et al. 2017; Hui et al. 2018; Kishi et al. 2019; Leucht et al. 2012; Thompson et al. 2018), rehospitalization (Tiihonen et al. 2018), and death (Tiihonen et al. 2018; Vermeulen et al. 2017). Implicitly, continued treatment with an effective and tolerable medication would be preferable to potential destabilization or treatment discontinuation. This inference is also consistent with clinical observations that individualizing choice of an antipsychotic medication is important. In clinical trials, a change to a different medication has been associated with earlier discontinuation of treatment as compared with continuation of the same antipsychotic medication (Essock et al. 2006; Stroup et al. 2011).

For these reasons, it will be optimal to continue on the same medication for most patients. Nevertheless, under some circumstances, it may be necessary to consider a change from one antipsychotic medication to another one. For example, a patient may have experienced some degree of response to initial treatment but may still have significant symptoms or difficulties in functioning that would warrant a trial of a different medication. Another reason to change medications would be to initiate treatment with an LAI antipsychotic if the current oral medication is unavailable in an LAI formulation (see Statement 10). A medication change may also be considered because of patient preferences, medication availability, or side effects. Given the long-term health risks of metabolic syndrome and obesity, weight gain and development of diabetes or metabolic syndrome are common reasons that a change to a different medication may be discussed.

In a randomized study that examined the effects of switching from olanzapine, quetiapine, or risperidone to aripiprazole to reduce metabolic risk, a change to aripiprazole was associated with improvements in non-HDL cholesterol, serum triglycerides, and weight as well as a small reduction in 10-year risk of coronary heart disease but no difference in the odds of having metabolic syndrome (Stroup et al. 2011, 2013). Individuals who switched to aripiprazole, as compared with those who remained on their initial medication, had a higher rate of discontinuing treatment but showed no significant increases in symptoms or hospitalizations. In addition, the Clinical Antipsychotic Trials of Intervention Effectiveness (CATIE) showed that individuals who experienced issues with medication efficacy or tolerability in an early phase of the trial could still go on to do well with a different medication in a subsequent phase of the trial (McEvoy et al. 2006; Rosenheck et al. 2009; Stroup et al. 2007, 2009).

These findings suggest that a change in medication can be of benefit to patients under some circumstances but also suggest that the possible benefits and risks of a medication change should be reviewed with the patient in the context of shared decision-making. Such discussions with the patient should consider the patient’s recovery goals, the potential benefits of medication changes or dose reductions in terms of changes in or diminution of side effects, and the potential harms of medication changes or dose reductions (Davidson 2018). It will typically be beneficial to include family members or other persons of support in such discussions.

Only a limited amount of research has explored the optimal approach for changing antipsychotic medications when warranted. The typical approach is a gradual cross-taper in which the second antipsychotic medication is begun and gradually increased in dose as the initial antipsychotic medication is gradually tapered. However, the few studies that are available do not suggest differences between gradual discontinuation as compared with immediate discontinuation of the first medication (Takeuchi et al. 2017a). In addition, no differences have been seen between starting the second antipsychotic and discontinuing the first antipsychotic at the same time as compared with starting the second antipsychotic and waiting before discontinuing the first antipsychotic agent (Takeuchi et al. 2017b). Regardless of the approach that is taken, careful monitoring is essential to avoid the risks of reduced adherence and clinical destabilization if a change in medications is undertaken. Depending on the pharmacological properties of the medications, including pharmacokinetic and receptor binding profiles (see Statement 4, Tables 4 and 5), side effects of medications may also emerge (e.g., insomnia with a shift to a less sedating medication, withdrawal dyskinesia with a shift to a medication with less prominent dopamine D2 receptor blockade) (Cerovecki et al. 2013).

Balancing of Potential Benefits and Harms in Rating the Strength of the Guideline Statement

Benefits

Use of an antipsychotic medication that has already been associated with symptom response can maintain improvements in symptoms as well as promote enhanced functioning and quality of life. In contrast, changes in antipsychotic treatment can be associated with early treatment discontinuation, increases in symptoms, clinical destabilization, and worsening of treatment tolerability.

Harms

The harms of continuing use of the same antipsychotic medication can vary depending on whether the patient is experiencing any significant side effects from the medication that would have long-term untoward effects. Continuing the same medication could lead to greater long-term risks such as metabolic effects or tardive syndromes from antipsychotic medications, but this would depend on the side-effect profile of the medication. In some instances, changing to a different medication could worsen long-term side-effect risk rather than reduce such risks.

Patient Preferences

Clinical experience suggests that most patients prefer to continue to take an antipsychotic medication that has led to a response in symptoms. Once they have found a medication that is effective and well tolerated, many individuals experience anxiety if they are unable to continue with that medication because of realistic concerns about a possible return of symptoms, reductions in functioning, risk of hospitalization, and other potential consequences of medication changes. However, other patients may not wish to remain on a given antipsychotic medication because of concerns about side effects or other factors that make continued treatment difficult.

Balancing of Benefits and Harms

The potential benefits of this guideline statement were viewed as likely to outweigh the potential harms. Although most patients prefer to continue taking the same medication once their symptoms have responded, there are reasons that a change in medication may be indicated, and factors such as medication side-effect profiles, medication availability, and patient preferences for specific medications also may play a role in decisions to continue with the same medication. For additional discussion of the research evidence, see Appendix C, Statement 6.

Differences of Opinion Among Writing Group Members

There were no differences of opinion. The writing group voted unanimously in favor of this suggestion.

Review of Available Guidelines From Other Organizations

Information from other guidelines is consistent with this guideline statement. Other guidelines on the treatment of schizophrenia (SIGN, WFSBP) note that treatment should usually continue with the same antipsychotic medication that led to the best response and had the best individual side-effect profile, given the risk of destabilization with switching an antipsychotic regimen (Hasan et al. 2013; Scottish Intercollegiate Guidelines Network 2013).

Quality Measurement Considerations

As a suggestion, this guideline statement is not appropriate for use as a quality measure or for electronic decision support. However, health plans may wish to implement internal process measures to assess and reduce rates at which changes to stable medication regimens are made on the basis of nonclinical factors such as pre-authorization requirements or formulary changes.

Statement 7: Clozapine in Treatment-Resistant Schizophrenia

APA recommends (1B) that patients with treatment-resistant schizophrenia be treated with clozapine.*

*This guideline statement should be implemented in the context of a person-centered treatment plan that includes evidence-based nonpharmacological and pharmacological treatments for schizophrenia.

Implementation

Identification of Treatment-Resistant Schizophrenia

Clozapine is recommended for individuals with treatment-resistant schizophrenia, but there is considerable variation in definitions of treatment-resistant schizophrenia in clinical trials and in practice (Howes et al. 2017). For the purpose of future research trials, the Treatment Response and Resistance in Psychosis (TRRIP) Working Group conducted a detailed systematic review of clinical trials in treatment-resistant schizophrenia and used a consensus-based approach to establish minimum and optimum criteria for identifying treatment-resistant schizophrenia (Howes et al. 2017). In addition to a diagnosis of schizophrenia, the identification of treatment-resistant schizophrenia rests on the persistence of significant symptoms despite adequate pharmacological treatment (Howes et al. 2017). More specifically, the TRRIP Working Group recommended that symptoms be of at least 12 weeks’ duration in total, be of at least moderate severity, and be associated with at least moderate functional impairment as determined by validated rating scales (e.g., PANSS, BPRS, or SANS and SAPS for symptoms and a score < 60 on the SOFAS as a measure of functioning). If a prospective medication trial of at least 6 weeks at adequate dose has not led to symptom reduction of more than 20%, this provides additional evidence of treatment resistance. It is helpful to note whether the persistent symptoms include positive, negative, or cognitive symptoms because responses to these symptom domains may differ.

In terms of treatment adequacy, the TRRIP Working Group recommended that at least two antipsychotic trials should be conducted with different antipsychotic medications with at least 6 weeks at a therapeutic dosage of medication for each and with adherence of at least 80% of prescribed dosages (Howes et al. 2017). A therapeutic dosage of medication was defined as the midpoint of the target range for acute treatment of schizophrenia according to the manufacturer’s product labeling or the equivalent of at least 600 mg of chlorpromazine per day (Howes et al. 2017). (For tables of dose equivalents, see College of Psychiatric and Neurologic Pharmacists 2019; Leucht et al. 2014, 2015; Rothe et al. 2018.) The consensus criteria include at least one antipsychotic blood level to assess adherence, obtaining information on adherence from at least two sources (e.g., pill counts, dispensing chart reviews, patient/carer reports), and obtaining information on past treatment response from patient/carer reports and other sources.

For clinical purposes, a common definition is that a patient’s symptoms have shown no response or partial and suboptimal response to two antipsychotic medication trials of at least 6 weeks each at an adequate dosage of medication, and some definitions specify using medications from different classes (e.g., SGA vs. FGA). However, if there is no significant improvement after several weeks of treatment (e.g., < 20% improvement in symptoms), the likelihood of substantial improvement (e.g., > 50% improvement in symptoms) is small (Howes et al. 2017; Samara et al. 2015), and a longer trial of the medication may not be warranted. It should also be noted that a medication trial cannot be viewed as adequate if truncated in terms of duration or dosage because of poor tolerability or if limited by poor adherence. Accordingly, some experts suggest a trial of an LAI antipsychotic medication before deciding that a patient’s symptoms are treatment-resistant.

Initiation of Treatment With Clozapine

After a patient is identified as having treatment-resistant schizophrenia, the clinician should engage the patient in discussion about clozapine treatment. A trial of clozapine may also be appropriate in individuals who show a response to treatment (i.e., have at least a 20% reduction in symptoms) yet still have significant symptoms or impairments in functioning (Howes et al. 2017). In fact, clozapine is often underused (Carruthers et al. 2016; Latimer et al. 2013; Olfson et al. 2016; Stroup et al. 2014; Tang et al. 2017), and many patients would benefit from earlier consideration of clozapine initiation.

Discussion of clozapine should emphasize principles of shared decision-making; including family members or other persons of support in such discussions is often beneficial. In terms of a patient’s recovery goals, most individuals value an ability to think more clearly and stop hallucinations or delusions when deciding about medication changes (Achtyes et al. 2018). In addition, most patients who receive clozapine view it positively. For example, one large survey of individuals with schizophrenia or schizoaffective disorder who were taking an antipsychotic medication found that the vast majority of those taking clozapine adhered to treatment and found it helpful, whereas only approximately 5% found it not helpful (Siskind et al. 2017a). In contrast, most other antipsychotic medications were viewed less positively (Siskind et al. 2017a). Nevertheless, it is important to identify patient concerns about clozapine and address them insofar as is possible. For example, patients may express concerns about the burdens of required blood work and may encounter logistical barriers such as access to transportation (Farooq et al. 2019; Gee et al. 2017; Verdoux et al. 2018). However, they may be willing to consider clozapine if logistical barriers can be overcome or if given the information that blood monitoring requirements become less frequent over time. Concerns about other side effects, such as weight gain or somnolence, may also contribute to a reluctance to switch to clozapine (Achtyes et al. 2018). It can be helpful to have an open discussion of these side effects and a well-defined plan for monitoring as treatment proceeds. Peer-run support groups that directly address living with side effects can help patients develop strategies for coping with side effects.

Clinicians may also have concerns about clozapine that can serve as a barrier to treatment. For example, many clinicians have limited experience in using clozapine and sometimes express concerns about paperwork burdens, patient adherence with monitoring, and side effects (Daod et al. 2019; Farooq et al. 2019; Gee et al. 2017; Kelly and Love 2019; Kelly et al. 2018; Leung et al. 2019; Verdoux et al. 2018; Warnez and Alessi-Severini 2014). Many clinicians overestimate the likelihood of severe neutropenia and are reluctant to begin clozapine on an outpatient basis (Farooq et al. 2019). Education about the use of clozapine and its side effects can be useful in addressing clinician-related prescribing barriers.

When initiating treatment with clozapine, a slow dose titration is essential to minimize the risks of seizure, orthostatic hypotension, and excessive sedation (Clozaril 2019). Large, rapid increases in clozapine dosage have led to cardiovascular collapse and death, particularly in patients taking respiratory depressant medications such as benzodiazepines. From a starting dosage of 12.5 mg once or twice daily, the dosage of clozapine can be increased by, at most, 25–50 mg/day to a target dosage of 300–450 mg/day (Clozaril 2019). Subsequent dose increases, if needed, should be of 100 mg or less, once or twice weekly. Although efficacy is often seen at a dosage of 300–450 mg/day, some individuals may need higher dosages of clozapine, to a maximum daily dose of 900 mg, for full response. A slower rate of titration may be needed for patients with an initial episode of schizophrenia and in those who are older, severely debilitated, or sensitive to side effects. Those with a preexisting central nervous system condition, including individuals with 22q11.2 deletion syndrome, also warrant a slower rate of titration and may have an increased risk of seizures at usual doses. Use of divided doses can be helpful in reducing side effects during initial dose titration, although many patients are ultimately treated with a single dose at bedtime to minimize daytime sedation and facilitate adherence (Takeuchi et al. 2016).

Monitoring for therapeutic benefits and side effects of clozapine should occur throughout the dose titration phase (see Statement 4). Because titration of clozapine proceeds slowly, the therapeutic benefits may not be noticed immediately, and side effects may be more prominent than benefits. Thus, it can be helpful to provide patients with education and reassurance about the expected timetable of therapeutic effects of clozapine.

If clozapine is being resumed after a gap in treatment of 48 hours or more, it should be restarted at 12.5 mg once or twice daily. If that dosage is well tolerated, the dose may be increased to a therapeutic range more quickly than recommended for initial treatment. If a decision is made to stop clozapine, it is best to taper the dose unless the medication is being stopped for medically urgent reasons (e.g., severe neutropenia, myocarditis, NMS).

Use of Clozapine Levels During Treatment With Clozapine

While the dose of clozapine is being titrated, it is useful to obtain blood levels of clozapine and its major active metabolite, norclozapine (N-desmethylclozapine) (Couchman et al. 2010). Blood levels can also be helpful if there are questions about medication adherence, less efficacy or more side effects than expected, potential medication interactions, or other factors that may be influencing clozapine levels. Although there is substantial variation between individuals, clozapine levels on a specific dosage will generally be greater in nonsmokers than in smokers, in heavy caffeine users than in nonusers, in women than in men, and in older individuals than in younger individuals (Carrillo et al. 1998; Ismail et al. 2012). In addition, changing between different generic forms of clozapine can lead to a 5%–10% difference in blood levels. Levels of clozapine should be drawn at steady state (3 days or more after a dose change) and at a trough in medication levels (about 12 hours after the last dose). Typically, patients will receive a bedtime dose of clozapine and then have a level drawn the following morning before receiving an additional dose. There is not an absolute level of clozapine that is associated with either efficacy or toxicity (Remington et al. 2013; Spina et al. 2000; Stark and Scott 2012; Suzuki et al. 2011; VanderZwaag et al. 1996). In most patients, efficacy will be highest at levels greater than 350 ng/mL of clozapine, but some patients will show response or prevention of relapse at levels as low as 200 ng/mL. The risk of developing seizures increases with the blood level of clozapine.

As with the results of any laboratory test, interpretation of clozapine levels should consider the clinical context. For example, if a clozapine level is much higher than expected, assess for dose-related side effects and clinical evidence of toxicity. If the patient’s clinical status does not suggest signs of clozapine toxicity, then determine the timing of the level (e.g., peak vs. trough) and identify any potential for drug interactions, changes in smoking status, or incorrect specimen labeling. If levels are much lower than expected, such factors as poor adherence, rapid metabolism, drug interactions, or changes in smoking status may also be relevant.

Typically, the level of norclozapine will be reported along with the blood level of clozapine. Norclozapine is the major active metabolite of clozapine and appears to differ from clozapine in efficacy and side effects. Nevertheless, the value of norclozapine levels in guiding clinical decisions is unclear. Because the half-life of norclozapine is greater than the half-life of clozapine, a clozapine:norclozapine ratio less than 0.5 can suggest poor adherence over the previous day or rapid metabolism of clozapine (e.g., via CYP1A2 induction). A clozapine:norclozapine ratio greater than 3.0 could suggest that metabolic pathways are saturated or inhibited by a concomitant medication. Shifts in the ratio of clozapine to norclozapine can also result from other drug-drug interactions or if nontrough levels are obtained (Couchman et al. 2010; Ellison and Dufresne 2015).

Monitoring for Side Effects During Treatment With Clozapine

With clozapine, safety monitoring during treatment is important to minimize the risk of adverse events. The Clozapine REMS Program (www.clozapinerems.com)1 is required for prescribing of clozapine in the United States. The REMS program includes required training that must be completed by prescribers (Clozapine REMS Program 2019a), resource materials (Clozapine REMS Program 2019b), and a shared patient registry for all clozapine manufacturers’ products that permits tracking of ANCs and documentation of decisions about continued treatment. The Clozapine REMS site provides instructions about threshold values for ANCs in hematologically normal individuals and in those with benign ethnic neutropenia, which is most common in individuals of African descent and is associated with ANCs that are lower than standard reference ranges (Clozapine REMS Program 2014). It also describes the required frequencies for ANC monitoring, which vary with ANC values. In patients who have stopped or interrupted treatment with clozapine for 30 days or more, the initial dose titration for clozapine and the monitoring frequency for treatment initiation should be followed.

1For Canadian prescribers, use the appropriate Canadian clozapine registry, not the U.S. Clozapine REMS Program.

Because the highest risk of severe neutropenia (ANC < 500/μL) occurs within the initial 6 months of clozapine treatment (Alvir et al. 1993; Clozapine REMS Program 2019c; Myles et al. 2018), ANC monitoring is more frequent early in treatment and is required less often with longer treatment duration. The need for ANC monitoring can be a common practical issue for patients because of the time and transportation needed to obtain blood tests at a laboratory. The availability of point-of-care testing for white blood cell counts may mitigate these barriers for patients and facilitate treatment with clozapine.

In addition to neutropenia, clozapine treatment can be associated with several other important side effects. Potentially serious cardiac complications of clozapine treatment include myocarditis and cardiomyopathy. Myocarditis is infrequent and generally occurs during the first month of treatment, whereas cardiomyopathy is rare and generally occurs later in the treatment course (Bellissima et al. 2018; Ronaldson et al. 2015). Myocarditis usually is heralded by shortness of breath, tachycardia, and fever, but diagnosis can be challenging because of the nonspecific nature of other symptoms, which can include fatigue, chest pain, palpitations, peripheral edema, and hypereosinophilia. In patients who do develop myocarditis or cardiomyopathy in conjunction with clozapine treatment, clozapine is typically discontinued. Subsequent decisions about resuming clozapine are individualized and are based on the benefits and risks of treatment as compared with other therapeutic alternatives.

In patients who are treated with clozapine, a brief self-limiting fever (> 38°C) may also occur during the first few weeks of clozapine treatment; this fever responds to supportive measures (Bruno et al. 2015; Lowe et al. 2007; Pui-yin Chung et al. 2008). However, in a febrile patient, it is essential to assess for the presence of potentially life-threatening complications, including NMS, severe neutropenia, infection, and myocarditis.

Other potentially serious side effects of clozapine treatment include seizures, orthostatic hypotension, and gastrointestinal effects. When seizures occur with clozapine, they typically occur with very high doses of clozapine, rapid increases in clozapine dose, or shifts in medication levels (related to drug-drug interactions or effects of smoking on drug metabolism) (Devinsky et al. 1991). Therefore, a slow initial titration of clozapine dose is essential, and patients should be cautioned not to drive or engage in other potentially hazardous activities while clozapine is being titrated. If a seizure does occur with clozapine, dose adjustment may be needed, or adjunctive anticonvulsant medication (e.g., valproate) may be considered in conjunction with neurological consultation (Alldredge 1999; Wong and Delva 2007).

Orthostatic hypotension can also occur with clozapine and is most common with treatment initiation. Older patients and patients with peripheral vascular disease or a compromised cardiovascular status may be at particular risk. When severe, orthostatic hypotension can cause syncope, dizziness, or falls. Patients who experience orthostatic hypotension must be cautioned to sit on the edge of the bed for a minute before standing up, move slowly when going from lying down or sitting to standing, and seek assistance when needed. Management strategies for orthostatic hypotension include supportive measures (e.g., use of support stockings, increased dietary salt and fluid intake), reducing the speed of clozapine dose titration, and decreasing or dividing doses of clozapine. As a last resort, administration of the salt/fluid-retaining corticosteroid fludrocortisone can be considered to increase intravascular volume, but it is important to be mindful of the potential for immunosuppressive effects and development of diabetes with this medication (Mar and Raj 2018; Shen et al. 2017). For patients who are receiving concomitant antihypertensive treatment, adjustments to the dose of these medications may be needed.

Gastrointestinal effects of clozapine can also be significant and in some patients are associated with fecal impaction or paralytic ileus (Every-Palmer and Ellis 2017; Leung et al. 2017). Thus, the patient should obtain urgent medical care if experiencing constipation that is severe or does not resolve. To prevent development of constipation, it is useful to minimize the doses and number of contributory medications such as other anticholinergic medications and opioids. Activity and exercise should be encouraged to stimulate motility. A stool softener (e.g., docusate [Colace]) can be started for patients at increased risk (e.g., older patients). If constipation does develop, initial treatment can include stool softeners (e.g., docusate [Colace]) or osmotic laxatives (e.g., lactulose [Enulose], polyethylene glycol [Miralax], bisacody, [Dulcolax]). Second-line treatments include stimulant laxatives (e.g., senna [Senokot], senna tea, cascara, sodium picosulfate). If constipation persists, an enema (e.g., Fleet) should be considered. A combination of treatments may be needed to treat constipation and then to prevent its recurrence.

Side effects related to metabolic syndrome are common and generally are observed in the initial months of treatment but can also occur later in treatment. These side effects include weight gain (Alvarez-Jimenez et al. 2008; Leucht et al. 2013; Zhang et al. 2013); hyperlipidemia (Buhagiar and Jabbar 2019; Bushe and Paton 2005; Meyer and Koro 2004; Mitchell et al. 2013a); and glucose dysregulation, including development of diabetes mellitus (Hirsch et al. 2017; Ward and Druss 2015; Whicher et al. 2018; Zhang et al. 2017). Monitoring of body mass index, hemoglobin A1c, and lipid levels is important during clozapine treatment, as outlined in Statement 1, Table 2. If diabetes or hyperlipidemia is identified, it should be treated, typically by the patient’s primary care clinician. When weight gain occurs, it is usually progressive over the first 6 months of treatment, although some patients continue to gain weight indefinitely (Alvarez-Jimenez et al. 2008). Prevention of weight gain should, thus, be a high priority because weight loss is difficult for most patients. Efforts should be made to intervene proactively with weight gain of 5–10 pounds, and other medications that can cause weight gain (e.g., valproate) should be tapered and discontinued when possible. Dietary interventions, such as specialized behavioral health interventions, in-person community interventions (e.g., Weight Watchers), services that include meal delivery (e.g., Jenny Craig), or Internet-based interventions (e.g., Omada Health), should be suggested (Bonfioli et al. 2012).

In collaboration with the patient’s primary care clinician, metformin or nonstimulant medications for weight loss can be considered. Metformin has been shown to be safe in individuals without hyperglycemia and can reduce body weight and reverse metabolic abnormalities in patients with obesity or other metabolic problems (Das et al. 2012; de Silva et al. 2016; McGinty et al. 2016; Mizuno et al. 2014; Siskind et al. 2016a; Vancampfort et al. 2019; Zheng et al. 2015; Zhuo et al. 2018). Fewer studies have been done with glucagon-like peptide-1 agonist medications, but the available data suggest that body weight and metabolic risk factors are reduced by these medications as compared with placebo (Siskind et al. 2019). The benefits of exercise appear to be small in terms of weight loss in individuals with schizophrenia (Firth et al. 2015; Pearsall et al. 2014; Vancampfort et al. 2017, 2019). Nevertheless, many individuals with schizophrenia do not engage in physical activity (Stubbs et al. 2016a; Vancampfort et al. 2016b), and exercise can be suggested for its benefits for overall health, improved cardiorespiratory fitness, and other aspects of functioning (Dauwan et al. 2016; Firth et al. 2015, 2017; Vancampfort et al. 2017, 2019).

Sedation, sialorrhea, and tachycardia are each commonly observed during treatment with clozapine but can generally be managed conservatively. Sedation is most pronounced in the initial phases of treatment with clozapine, and many patients develop some tolerance to the sedating effects with continued administration. However, persistent sedation, including daytime drowsiness and increased sleep time, can interfere with social, recreational, and vocational function. Lowering of the daily dose, consolidating divided doses into one evening dose, or changing to a less sedating antipsychotic medication may be effective in reducing the severity of sedation. Coffee or other forms of caffeine can be helpful in the morning but can also interact with medications (e.g., contribute to tachycardia; raise blood levels of medications, including clozapine). Adding a stimulant medication is not typically helpful and can lead to additional side effects. If sedation or the risk of sedation is significant (e.g., during initial clozapine titration), patients should be cautioned not to drive or engage in potentially hazardous activities.

Sialorrhea (or hypersalivation) is also a frequent side effect of clozapine that can contribute to reductions in quality of life and complications such as aspiration pneumonia (Dzahini et al. 2018; Kaplan et al. 2018; Stoecker et al. 2017). Because sialorrhea may be more bothersome at night, patients may be advised to place a towel on their pillow and change to a clean towel in the middle of the night to minimize discomfort. During the day, patients can be encouraged to chew sugarless gum, which stimulates the swallowing reflex. Pharmacological approaches to address sialorrhea come from small studies and case reports and include use of low-dose or topical anticholinergic medications, such as glycopyrrolate or sublingual ophthalmic atropine 1% drops (Bird et al. 2011; Liang et al. 2010; Man et al. 2017). Diphenhydramine has also been studied (Chen et al. 2019); however, because clozapine and other antipsychotics can have significant anticholinergic properties themselves and anticholinergics have small effects on sialorrhea, the use of agents with added anticholinergic effects should be approached cautiously. Terazosin and, in severe refractory cases, botulinum toxin have also been used (Bird et al. 2011; Liang et al. 2010; Man et al. 2017).

Healthy patients can usually tolerate some increase in resting pulse rate, although this may not be the case for patients with preexisting heart disease. In patients with significant tachycardia (heart rates above 110–120 bpm), an ECG is warranted, as is assessment for other potential causes of tachycardia (e.g., fever, anemia, smoking, hyperthyroidism, respiratory disease, cardiovascular disorders, caffeine, other stimulants, and side effects of other medications). Early in treatment with clozapine, the possibility of myocarditis should be considered. If tachycardia is accompanied by pain, shortness of breath, fever, or signs of a myocardial infarction or heart rhythm problem, emergency assessment is essential. Management strategies for tachycardia with any antipsychotic medication include reducing the dose of medication; discontinuing medications with anticholinergic or stimulant properties; and addressing orthostatic hypotension, if present. Case reports have discussed the use of medications such as β-blocking agents for persistent and significant tachycardia with clozapine. Nevertheless, treatment is not indicated unless the patient is symptomatic or the patient’s heart rate is substantially greater than 120 bpm because data from more rigorous studies are not available and these medications can contribute to other side effects such as orthostatic hypotension (Lally et al. 2016a).

Side effects related to dopamine D2 receptor antagonism (e.g., acute dystonia, akathisia, medication-induced parkinsonism, NMS, tardive syndromes, hyperprolactinemia) can occur but are less frequent with clozapine than with many other antipsychotic medications. For additional information on the recognition and management of these side effects, see Statement 4, subsection “Treatment-Emergent Side Effects of Antipsychotic Medications.”

Other Approaches to Treatment-Resistant Schizophrenia

For all patients with treatment-resistant schizophrenia, it is important to conduct a review of the treatment plan at periodic intervals (see Statement 3). In addition to a review of prior medication trials, it is essential to review the psychosocial treatments that a patient has received and whether addition of one or more psychosocial interventions would be of benefit. For example, some patients may not have received cognitive-behavioral therapy for psychosis (CBTp) as recommended in Statement 16 or initial benefits of CBTp may have faded if CBTp was stopped. Under such circumstances, treatment with CBTp may be warranted (Burns et al. 2014; Morrison et al. 2018). Engaging families or persons of support in CBTp can also be helpful (Turkington and Spencer 2019). A similar review of potential additions to the treatment plan can occur with other psychosocial treatments.

Optimizing Treatment With Clozapine

Although studies suggest that at least one-third of individuals with treatment-resistant schizophrenia will respond to clozapine (Kahn et al. 2018; McEvoy et al. 2006), some patients will not have a complete response. Before concluding that a patient has not responded to clozapine, it is important to assure that an adequate target dose has been reached (typically 300–450 mg/day) and that steady state levels of clozapine and norclozapine appear sufficient to produce therapeutic benefit. Although no absolute level of clozapine is associated with efficacy (Remington et al. 2013; Spina et al. 2000; Suzuki et al. 2011; VanderZwaag et al. 1996), if no response is evident and clozapine is well tolerated, the clozapine dose should be increased to achieve a clozapine level of greater than 350 ng/mL. In general, this dose of medication should be continued for at least 8 weeks to determine response, although further increases in dose can also be made, as tolerated. If there continues to be no evidence of benefit, as for any patient treated with clozapine, the value of the medication should be assessed periodically in terms of the patient’s response, the medication side effects, and the availability of any newer treatment options. Longitudinal use of a quantitative measure (see Statement 2) can be helpful in assessing functioning and overall response and identifying specific symptoms that have or have not responded to treatment.

Continuing Clozapine and Augmenting With Another Medication

For individuals who do not respond to clozapine alone, the evidence base for other treatments is limited, although a number of options have been tried. Augmentation of clozapine with another medication has shown no significant benefit in double-blind trials, although some benefit was noted in open-label trials and meta-analyses of trials that were generally of low quality (Barber et al. 2017; Correll et al. 2017b; Galling et al. 2017; Sinclair and Adams 2014; Siskind et al. 2018; Veerman et al. 2014; Wagner et al. 2019a). Studies have included augmentation with other antipsychotic medications (FGAs and SGAs), anticonvulsants, and other medications. If a trial of augmentation therapy is undertaken, it is important to consider the potential additive effects of the medications on side effects and the potential for drug-drug interactions. Periodic review of the patient’s medication regimen is also important in order to identify and reduce or discontinue medications that are not effective, that are no longer necessary, or that are contributing to an inordinate burden of side effects. As noted in the previous subsection, longitudinal use of a quantitative measure can assist in making such determinations.

Augmenting Clozapine or Another Antipsychotic Medication With Electroconvulsive Therapy

There is also evidence for benefits of ECT in combination with clozapine as compared with clozapine alone in most (Ahmed et al. 2017; Grover et al. 2015; Lally et al. 2016b; Petrides et al. 2015; Pompili et al. 2013; G. Wang et al. 2018), but not all (Melzer-Ribeiro et al. 2017), studies. Rates of headache and reports of memory impairment were more frequent with clozapine plus ECT than with clozapine alone; however, symptomatic improvement and rates of remission at the end of treatment were significantly greater in the group that received adjunctive ECT. For this reason, ECT could be considered for clozapine-resistant schizophrenia, particularly in patients who also have catatonia or significant suicide risk or who require a rapid response because of the severity of their psychiatric or medical condition. For individuals who show a response to ECT, treatment with ECT on a maintenance basis could be considered as an adjunct to clozapine.

Some studies have shown evidence for benefits of ECT in combination with antipsychotic medications other than clozapine (Ahmed et al. 2017; Ali et al. 2019; Pompili et al. 2013; Sinclair et al. 2019; Zheng et al. 2016). Particularly in patients who also have catatonia or significant suicide risk or who require a rapid response due to the severity of their psychiatric or medical condition, ECT could be considered. For individuals who show a response to ECT, treatment with ECT on a maintenance basis could be considered.

Although studies have also been done with TMS for treatment of hallucinations and for treatment of negative symptoms, at present there is insufficient evidence of benefit to suggest use of TMS in individuals with schizophrenia (Dollfus et al. 2016; Dougall et al. 2015; He et al. 2017).

Balancing of Potential Benefits and Harms in Rating the Strength of the Guideline Statement

Benefits

Use of clozapine in individuals with treatment-resistant schizophrenia can be associated with reductions in psychotic symptoms, higher rates of treatment response, and lower rates of treatment discontinuation due to lack of efficacy (low to moderate strength of research evidence) as well as lower rates of self-harm, suicide attempts, or hospitalizations to prevent suicide (moderate strength of research evidence). Overall rates of hospitalization are also reduced during treatment with clozapine as compared with other oral antipsychotic medications (low strength of research evidence). All-cause mortality is also reduced in individuals treated with clozapine as compared with other individuals with treatment-resistant schizophrenia (moderate strength of research evidence).

Harms

Although overall rates of adverse events do not differ with clozapine as compared with risperidone (low strength of research evidence), clozapine does have a higher risk of study withdrawal due to adverse events than some other SGAs (low strength of research evidence). Specific harms of using clozapine include rare but serious effects, including severe neutropenia, myocarditis, cardiomyopathy, and NMS. These harms cannot be eliminated, but risks of severe neutropenia are lessened by required ANC monitoring. Early attention to and recognition of NMS and cardiac complications of clozapine use may also reduce risk. Seizures are also more frequent with clozapine than other antipsychotics but can be minimized by slow titration of the clozapine dose, avoidance of very high clozapine doses, and attention to pharmacokinetic factors that may lead to rapid shifts in clozapine levels. Constipation can also be significant with clozapine and in some patients is associated with fecal impaction or paralytic ileus. Other side effects that are more common with clozapine than other antipsychotic medications include sialorrhea, tachycardia, fever, dizziness, sedation, and weight gain. Rates of hyperglycemia and diabetes may also be increased.

Patient Preferences

Clinical experience suggests that many patients are cooperative with and accepting of clozapine as part of a treatment plan; however, other patients may express concerns about the burdens of required blood work, including logistical barriers such as access to transportation (Farooq et al. 2019; Gee et al. 2017; Verdoux et al. 2018). Concerns about other side effects, such as weight gain or somnolence, may also contribute to a reluctance to switch to clozapine (Achtyes et al. 2018). On the other hand, when deciding about medication changes, most patients value an ability to think more clearly and stop hallucinations or delusions (Achtyes et al. 2018; Kuhnigk et al. 2012; Levitan et al. 2015), and most patients who receive clozapine view it positively. For example, one large survey of individuals with schizophrenia or schizoaffective disorder who were taking an antipsychotic medication found that the vast majority of those taking clozapine adhered to treatment and found it helpful, whereas only approximately 5% found it not helpful (Siskind et al. 2017a). In contrast, most other antipsychotic medications were viewed less positively (Siskind et al. 2017a). In addition, in the CATIE study, clozapine and combination antipsychotic treatment regimens were frequently selected by patients who stopped a previous medication because of inadequate therapeutic effect and by those with relatively severe symptoms (Stroup et al. 2009).

Balancing of Benefits and Harms

The potential benefits of this guideline statement were viewed as far outweighing the potential harms. For individuals with treatment-resistant schizophrenia, the risks of inadequately treated illness are substantial in terms of reduced quality of life (Kennedy et al. 2014) and increased mortality (Cho et al. 2019; Vermeulen et al. 2019; Wimberley et al. 2017) as well as negative effects for informal caregivers (Brain et al. 2018). Even in individuals who have had an inadequate response to other antipsychotic medications, a substantial fraction shows a clinically relevant response to clozapine. With careful monitoring to minimize the risk of harms from clozapine, the benefits of clozapine in patients with treatment-resistant schizophrenia were viewed as significantly outweighing the harms of treatment. For additional discussion of the research evidence, see Appendix C, Statement 7.

Differences of Opinion Among Writing Group Members

There were no differences of opinion. The writing group voted unanimously in favor of this recommendation.

Review of Available Guidelines From Other Organizations

Practice guidelines (BAP, CSG, NICE, RANZCP, SIGN, WFSBP, PORT) are consistent in recommending clozapine for individuals with treatment-resistant schizophrenia (Barnes et al. 2011; Buchanan et al. 2010; Galletly et al. 2016; Hasan et al. 2012; National Institute for Health and Care Excellence 2014; Scottish Intercollegiate Guidelines Network 2013). In terms of other therapies for treatment-resistant schizophrenia, several guidelines (SIGN, WFSBP, BAP) recommend augmentation treatment with an antidepressant for treatment-resistant illness associated with negative symptoms (Barnes et al. 2011; Hasan et al. 2012; Scottish Intercollegiate Guidelines Network 2013). For individuals with catatonia, the WFSBP recommends benzodiazepines and ECT (Hasan et al. 2012). In addition, ECT is mentioned in several guidelines (e.g., SIGN, RANZCP, WFSBP) as being appropriate in individuals with treatment-resistant schizophrenia.

Quality Measurement Considerations

Studies suggest that clozapine is underused and that a significant proportion of individuals with treatment-resistant schizophrenia do not receive treatment with clozapine, although there is significant variation between and within countries (Addington et al. 2012; Bachmann et al. 2017; Carruthers et al. 2016; Keller et al. 2014; Olfson et al. 2016; Stroup et al. 2014; Tang et al. 2017). Thus, internal quality improvement programs may wish to focus on ways to increase use of clozapine in individuals with treatment-resistant schizophrenia and track rates of clozapine use in this patient population. Internal quality improvement programs could also focus on increasing use of quantitative measures (i.e., rating scales) to improve identification of individuals with treatment-resistant schizophrenia and facilitate systematic longitudinal tracking of functioning, symptoms, and side-effect burdens. If quality measures are considered for development at the provider, facility, health plan, integrated delivery system, or population level, testing of feasibility, usability, reliability, and validity would be essential prior to use for purposes of accountability.

Electronic decision support would be challenging to implement and would depend on accurate and consistent entry of structured information. Nevertheless, in combination with rating scale data and prior prescribing histories, electronic decision support could help identify individuals with treatment-resistant illness who would benefit from a trial of clozapine.

Statement 8: Clozapine in Suicide Risk

APA recommends (1B) that patients with schizophrenia be treated with clozapine if the risk for suicide attempts or suicide remains substantial despite other treatments.*

*This guideline statement should be implemented in the context of a person-centered treatment plan that includes evidence-based nonpharmacological and pharmacological treatments for schizophrenia.

Implementation

Treatment with clozapine can be effective in reducing suicidal behavior if risk remains substantial despite other treatments. In addition, treatment with clozapine can be effective in reducing rates of suicide attempts and suicide in individuals with schizophrenia, regardless of whether formal criteria for treatment resistance have been met. Risk factors for suicidal behavior in individuals with schizophrenia are described in Statement 1, subsection “Implementation.” Although demographic and historical risk factors are static, a number of other risk factors are potentially modifiable and can serve as targets of intervention in constructing a plan of treatment (for additional details, see Statement 3). As in other circumstances in which patients do not appear to be responding fully to treatment, attention to adherence is crucial (for additional details, see Statement 3). For details of initiating and monitoring clozapine treatment, see Statement 7, subsections “Initiation of Treatment With Clozapine,” “Use of Clozapine Levels During Treatment With Clozapine,” and “Monitoring for Side Effects During Treatment With Clozapine.”

Balancing of Potential Benefits and Harms in Rating the Strength of the Guideline Statement

Benefits

In individuals with schizophrenia who are at significant risk for suicide attempts or suicide, use of clozapine can be associated with lower rates of self-harm, suicide attempts, or hospitalization to prevent suicide (moderate strength of research evidence). Additional benefits of clozapine treatment include higher rates of treatment response (low to moderate strength of research evidence) and reductions in psychotic symptoms, all-cause mortality, overall hospitalization rates, and treatment discontinuation due to lack of efficacy (low to moderate strength of research evidence).

Harms

Although overall rates of adverse events do not differ with clozapine as compared with risperidone (low strength of research evidence), clozapine does have a higher risk of study withdrawal due to adverse events than some other SGAs (low strength of research evidence). Specific harms of using clozapine include rare but serious effects, including severe neutropenia, myocarditis, cardiomyopathy, and NMS. These harms cannot be eliminated, but risks of severe neutropenia are lessened by required ANC monitoring. Early attention to and recognition of NMS and cardiac complications of clozapine use may also reduce risk. Seizures are also more frequent with clozapine than other antipsychotics but can be minimized by slow titration of the clozapine dose, avoidance of very high clozapine doses, and attention to pharmacokinetic factors that may lead to rapid shifts in clozapine levels. Constipation can also be significant with clozapine and in some patients is associated with fecal impaction or paralytic ileus. Other side effects that are more common with clozapine than other antipsychotic medications include sialorrhea, tachycardia, fever, dizziness, sedation, and weight gain. Rates of hyperglycemia and diabetes may also be increased.

Patient Preferences

Clinical experience suggests that many patients are cooperative with and accepting of clozapine as part of a treatment plan; however, other patients may express concerns about the burdens of required blood work, including logistical barriers such as access to transportation (Farooq et al. 2019; Gee et al. 2017; Verdoux et al. 2018). Concerns about other side effects, such as weight gain or somnolence, may also contribute to a reluctance to switch to clozapine (Achtyes et al. 2018). On the other hand, when deciding about medication changes, most patients value an ability to think more clearly and stop hallucinations or delusions (Achtyes et al. 2018; Kuhnigk et al. 2012; Levitan et al. 2015), and most patients who receive clozapine view it positively. For example, one large survey of individuals with schizophrenia or schizoaffective disorder who were taking an antipsychotic medication found that the vast majority of those taking clozapine adhered to treatment and found it helpful, whereas only approximately 5% found it not helpful (Siskind et al. 2017a). In contrast, most other antipsychotic medications were viewed less positively (Siskind et al. 2017a).

Balancing of Benefits and Harms

The potential benefits of this recommendation were viewed as far outweighing the potential harms. For individuals at significant risk for suicide attempts or suicide despite other treatments, the benefit of clozapine in reducing suicide-related risk is significant. With careful monitoring to minimize the risk of harms from clozapine, the benefit of clozapine in such patients was viewed as significantly outweighing the harms of treatment. For additional discussion of the research evidence, see Appendix C, Statement 8.

Differences of Opinion Among Writing Group Members

There were no differences of opinion. The writing group voted unanimously in favor of this recommendation.

Review of Available Guidelines From Other Organizations

Other guidelines do not specifically mention the use of clozapine for individuals with schizophrenia who are at substantial risk for suicide attempts or suicide despite other treatment. Guidelines (BAP, CSG, NICE, RANZCP, SIGN, WFSBP, PORT) are consistent, however, in recommending clozapine for individuals with treatment-resistant schizophrenia (Barnes et al. 2011; Buchanan et al. 2010; Galletly et al. 2016; Hasan et al. 2012; National Institute for Health and Care Excellence 2014; Scottish Intercollegiate Guidelines Network 2013).

Quality Measurement Considerations

Studies suggest that clozapine is underused and that a significant proportion of individuals with treatment-resistant schizophrenia do not receive treatment with clozapine, although there is significant variation between and within countries (Addington et al. 2012; Bachmann et al. 2017; Carruthers et al. 2016; Keller et al. 2014; Olfson et al. 2016; Stroup et al. 2014; Tang et al. 2017). Given low utilization of clozapine in general (Addington et al. 2012; Bachmann et al. 2017; Carruthers et al. 2016; Keller et al. 2014; Olfson et al. 2016; Stroup et al. 2014; Tang et al. 2017) and the high rates of suicidal ideas among individuals with treatment-resistant schizophrenia (Kennedy et al. 2014), it is likely that many individuals at significant suicide risk are not receiving treatment with clozapine. Thus, internal quality improvement programs may wish to focus on ways to increase and track use of clozapine in individuals with schizophrenia who have significant suicide risk that persists despite other treatments. Internal quality improvement programs could also focus on increasing the use of quantitative measures to improve identification and monitoring of individuals with risk factors for suicide.

If quality measures are considered for development at the provider, facility, health plan, integrated delivery system, or population level, testing of feasibility, usability, reliability, and validity would be essential prior to use for purposes of accountability. In particular, it is currently not possible to identify from most administrative data people at increased risk for suicide. Clinical assessment or patient self-report data are likely to be required.

Electronic decision support using passive alerts may be able to prompt clinicians to consider clozapine; however, such prompts would be challenging to implement because they would depend on accurate and consistent entry of structured information about diagnosis, suicidal ideation, and suicide attempts. Nevertheless, in combination with rating scale data, electronic decision support could help identify individuals with schizophrenia and significant suicide risk who would benefit from a trial of clozapine.

Statement 9: Clozapine in Aggressive Behavior

APA suggests (2C) that patients with schizophrenia be treated with clozapine if the risk for aggressive behavior remains substantial despite other treatments.*

*This guideline statement should be implemented in the context of a person-centered treatment plan that includes evidence-based nonpharmacological and pharmacological treatments for schizophrenia.

Implementation

Treatment with clozapine can be effective in reducing aggressive behavior if risk remains substantial despite other treatments. As in other circumstances in which patients do not appear to be responding fully to treatment, attention to adherence is crucial (for additional details, see Statement 3). Risk factors for aggressive behavior in individuals with schizophrenia are described in Statement 1, subsection “Implementation.” Although demographic and historical risk factors are static, a number of other risk factors are potentially modifiable and can serve as targets of intervention in constructing a plan of treatment (for additional details, see Statement 3). For details of initiating and monitoring clozapine treatment, see Statement 7, subsections “Initiation of Treatment With Clozapine,” “Use of Clozapine Levels During Treatment With Clozapine,” and “Monitoring for Side Effects During Treatment With Clozapine.”

Balancing of Potential Benefits and Harms in Rating the Strength of the Guideline Statement

Benefits

In individuals with schizophrenia who are at significant risk for aggressive behavior, use of clozapine may reduce the likelihood of aggressive behaviors (low strength of research evidence). Additional benefits of clozapine treatment include higher rates of treatment response (low to moderate strength of research evidence); reductions in psychotic symptoms, all-cause mortality, overall hospitalization rates, and treatment discontinuation due to lack of efficacy (low to moderate strength of research evidence); and lower rates of self-harm, suicide attempts, or hospitalizations to prevent suicide (moderate strength of research evidence).

Harms

Although overall rates of adverse events do not differ with clozapine as compared with risperidone (low strength of research evidence), clozapine does have a higher risk of study withdrawal due to adverse events than some other SGAs (low strength of research evidence). Specific harms of using clozapine include rare but serious effects, including severe neutropenia, myocarditis, cardiomyopathy, and NMS. These harms cannot be eliminated, but risks of severe neutropenia are lessened by required ANC monitoring. Early attention to and recognition of NMS and cardiac complications of clozapine use may also reduce risk. Seizures are also more frequent with clozapine than other antipsychotics but can be minimized by slow titration of the clozapine dose, avoidance of very high clozapine doses, and attention to pharmacokinetic factors that may lead to rapid shifts in clozapine levels. Constipation can also be significant with clozapine and in some patients is associated with fecal impaction or paralytic ileus. Other side effects that are more common with clozapine than other antipsychotic medications include sialorrhea, tachycardia, fever, dizziness, sedation, and weight gain. Rates of hyperglycemia and diabetes may also be increased.

Patient Preferences

Clinical experience suggests that many patients are cooperative with and accepting of clozapine as part of a treatment plan; however, other patients may express concerns about the burdens of required blood work, including logistical barriers such as access to transportation (Farooq et al. 2019; Gee et al. 2017; Verdoux et al. 2018). Concerns about other side effects, such as weight gain or somnolence, may also contribute to a reluctance to switch to clozapine (Achtyes et al. 2018). On the other hand, when deciding about medication changes, most patients value an ability to think more clearly and stop hallucinations or delusions (Achtyes et al. 2018; Kuhnigk et al. 2012; Levitan et al. 2015), and most patients who receive clozapine view it positively. For example, one large survey of individuals with schizophrenia or schizoaffective disorder who were taking an antipsychotic medication found that the vast majority of those taking clozapine adhered to treatment and found it helpful, whereas only approximately 5% found it not helpful (Siskind et al. 2017a). In contrast, most other antipsychotic medications were viewed less positively (Siskind et al. 2017a).

Balancing of Benefits and Harms

The potential benefits of this guideline statement were viewed as likely to outweigh the potential harms. For individuals at significant risk for aggressive behavior despite other treatments, there appears to be some benefit of clozapine in reducing aggression risk. In addition, clozapine may lead to indirect reductions in the risk of aggressive behavior by reducing other contributory risk factors for aggression such as hallucinations and delusions. Thus, with consideration of patient preferences and careful monitoring to minimize the risk of harms from clozapine, the benefit of clozapine in such patients was viewed as likely to outweigh the harms of treatment. For additional discussion of the research evidence, see Appendix C, Statement 9.

Differences of Opinion Among Writing Group Members

There were no differences of opinion. The writing group voted unanimously in favor of this suggestion.

Review of Available Guidelines From Other Organizations

Information from other guidelines is consistent with this guideline statement. The SIGN, RANZCP, BAP, and PORT guidelines all suggest consideration of clozapine for individuals with hostility or aggressive behaviors that do not respond to other interventions (Barnes et al. 2011; Buchanan et al. 2010; Galletly et al. 2016; Hasan et al. 2012; Scottish Intercollegiate Guidelines Network 2013). In addition, guidelines (BAP, CSG, NICE, RANZCP, SIGN, WFSBP, PORT) are consistent in recommending clozapine for individuals with treatment-resistant schizophrenia (Barnes et al. 2011; Buchanan et al. 2010; Galletly et al. 2016; Hasan et al. 2012; National Institute for Health and Care Excellence 2014; Scottish Intercollegiate Guidelines Network 2013).

Quality Measurement Considerations

As a suggestion, this guideline statement is not appropriate for use as a quality measure for purposes of accountability. Electronic decision support using passive alerts may be able to prompt clinicians to consider clozapine; however, such prompts would be challenging to implement because they would depend on accurate and consistent entry of structured information about diagnosis and risk factors for aggression. Nevertheless, in combination with rating scale data, electronic decision support could help identify individuals with schizophrenia and significant aggression risk who may benefit from a trial of clozapine.

Statement 10: Long-Acting Injectable Antipsychotic Medications

APA suggests (2B) that patients receive treatment with a long-acting injectable antipsychotic medication if they prefer such treatment or if they have a history of poor or uncertain adherence.*

*This guideline statement should be implemented in the context of a person-centered treatment plan that includes evidence-based nonpharmacological and pharmacological treatments for schizophrenia.

Implementation

LAI formulations of antipsychotic medications can provide a number of benefits for patients, families, and clinicians, yet they are often underutilized (Brown et al. 2014; Correll et al. 2016; Lawson et al. 2015; Sultana et al. 2019). Racial differences also exist in the proportion of individuals who are treated with LAI antipsychotic medications, with greater use of these formulations in Black patients than in white patients (Brown et al. 2014; Lawson et al. 2015).

With LAI antipsychotic medications, there is greater assurance that a patient will receive medication continuously because there are fewer opportunities to miss a medication dose and clinicians will be immediately aware of a missed visit or injection, yielding greater time for intervention before symptoms recur (Correll et al. 2016; Velligan et al. 2010; West et al. 2008). Presumably due to improved adherence, advantages of LAI antipsychotics include the potential for a decreased risk of mortality; reduced risk of hospitalization; and decreased rates of treatment discontinuation, including discontinuation due to inefficacy (see Appendix C, Statement 10). Other benefits for patients include a subjective sense of better symptom control, greater convenience as a result of needing to take fewer medications daily, and reduced conflict with family members or other persons of support related to medication-related reminders (Caroli et al. 2011; Correll et al. 2016; Iyer et al. 2013; Yeo et al. 2019). Although some patients may not wish to experience the discomfort associated with receiving injections of medications, this is not a major barrier for most patients. In addition, discomfort can often be minimized by using SGA LAIs rather than FGA LAIs, which have sesame oil–based vehicles, or by using an LAI with a small injection volume or lower administration frequency (Correll et al. 2016).

Consistent with principles of patient-centered care, it may be preferable to educate patients about the availability of LAI antipsychotic medications when discussing other aspects of antipsychotic treatment. Indeed, many patients will accept and may prefer LAIs if provided with information about the pluses and minuses of LAIs in the context of shared decision-making (Caroli et al. 2011; Heres et al. 2007; Kane et al. 2019; Waddell and Taylor 2009; Weiden et al. 2015; Yeo et al. 2019). Preference rates for LAIs are even higher among individuals who have personal experience in receiving an LAI formulation of an antipsychotic medication (Heres et al. 2007; Patel et al. 2009; Waddell and Taylor 2009).

Discussions about LAI antipsychotic medications often occur with patients who have had difficulty in adhering to oral medications. However, such discussions can also take place at other junctures. For example, if an individual has not responded to treatment with an oral antipsychotic medication, a trial of an LAI may be warranted (Howes et al. 2017; see Statement 4, subsection “Strategies to Address Initial Nonresponse or Partial Response to Antipsychotic Treatment”) because breaks in the continuity of oral medication therapy can be unrecognized (Lopez et al. 2017). An LAI formulation of an antipsychotic may also be considered when patients are transitioning between settings (e.g., at inpatient discharge, on release from a correctional facility), when future adherence is uncertain and the risk of reduced adherence may be increased. Although LAI antipsychotic medications have typically been used in individuals with multiple episodes of schizophrenia, some studies have used an LAI antipsychotic formulation earlier in the course of illness (Kane et al. 2019; Subotnik et al. 2015), when rates of poor adherence may be greater. Earlier discussion of an LAI may also be considered in individuals who are at increased risk of poor adherence due to a limited awareness of needing treatment or a co-occurring substance use disorder (García et al. 2016; Velligan et al. 2009).

If a decision is made to initiate treatment with an LAI, aspects of medication selection are similar to those for selection of an oral medication in terms of considering prior response, prior tolerability, pharmacological considerations, and side-effect profiles (see Statement 4). Patients may also have specific preferences and values related to the frequency of injection and the type and location of the injection (e.g., IM deltoid or gluteal sites, subcutaneous abdominal site; Heres et al. 2012). Because the oral and LAI formulations of a specific antipsychotic medication are comparable, a trial of the same oral antipsychotic will typically occur first to assure efficacy and tolerability (SMI Adviser 2019). Patients will experience medication side effects for a longer period of time after drug discontinuation with LAIs than with oral medications because of pharmacokinetic differences in the formulations, but this is not usually a problem if the oral formulation has first been well tolerated. Nevertheless, caution is warranted in a patient who has experienced NMS previously (Correll et al. 2016).

The conversion from an oral dose of medication to a corresponding dose of an LAI antipsychotic depends on the specific medication (see Statement 4, Tables 7, 8, and 9; Meyer 2013, 2017); product labeling for each medication describes approximate conversion ratios and whether a period of concomitant oral and LAI medication is needed. If the patient is taking an oral medication that lacks a corresponding LAI formulation, a change in antipsychotic medication may be needed if an LAI formulation is clinically indicated or preferred (see Statement 6).

When administering LAI antipsychotic injections, it is important to follow recommendations for safe injection practices (Centers for Disease Control and Prevention 2019a, 2019b) and infection control precautions (Centers for Disease Control and Prevention 2016, 2019b) as well as instructions from product labeling. Product labeling also contains important information on storage and reconstitution of LAI antipsychotic formulations as well as information on how to handle missed or late doses of medications (see Statement 4, Tables 7–9).

There are several barriers related to the use of LAI formulations of antipsychotic medications. For patients, there may be logistical barriers (e.g., access to transportation, childcare, school or work schedules) that depend on the frequency of appointments needed to receive an LAI antipsychotic medication. Other logistical barriers for patients may relate to such factors as cost or insurance authorizations for LAI antipsychotic agents. For clinicians, lack of knowledge and limited experience in using LAI antipsychotic medications contribute to underuse (Correll et al. 2016). Skill and experience in administering injections may be lacking, and nursing staff may not be available to give injections.

Additional barriers relate to the decision to suggest an LAI antipsychotic medication. Clinicians often do not consider LAI antipsychotic medications as a treatment option (Hamann et al. 2014; Heres et al. 2006; Kirschner et al. 2013; Weiden et al. 2015), even when such use is appropriate. Furthermore, clinicians may overestimate patients’ adherence with oral medications when considering relative benefits of LAIs (Correll et al. 2016; Lopez et al. 2017) or underestimate the acceptability of LAIs to patients (Hamann et al. 2010; Iyer et al. 2013; Patel et al. 2010a, 2010b; Weiden et al. 2015). At an organizational level, there may be a lack of resources, space, or trained personnel to administer injections (Correll et al. 2016; Velligan et al. 2011). Thus, workflows may need to be adjusted, partnerships may need to be developed with primary care clinicians to administer LAI antipsychotic injections, or other concerted efforts may be needed to address logistical and clinical barriers to LAI antipsychotic use.

Balancing of Potential Benefits and Harms in Rating the Strength of the Guideline Statement

Benefits

Use of an LAI antipsychotic medication in the treatment of schizophrenia may be associated with improved outcomes. Although meta-analyses of head-to-head RCTs comparing LAI with oral antipsychotics (McDonagh et al. 2017) and other meta-analyses of RCTs (Kishi et al. 2016a, 2016b; Kishimoto et al. 2014; Ostuzzi et al. 2017) do not show evidence of benefit from LAIs relative to oral antipsychotic medications, observational data from nationwide registry databases (Taipale et al. 2018a, 2018b; Tiihonen et al. 2011, 2017), cohort studies (Kishimoto et al. 2018), and “mirror image” studies (Kishimoto et al. 2013) suggest that use of LAI antipsychotic agents as compared with oral antipsychotic medications is associated with a decreased risk of mortality, reduced risk of hospitalization, and decreased rates of study discontinuation (including discontinuation due to inefficacy).

Harms

The harms of using an LAI antipsychotic medication in the treatment of schizophrenia are generally comparable to the harms of using an oral formulation of the same medication. For some patients, side effects with LAIs may be less problematic because peaks and troughs of medication levels will be less prominent than with oral medications because of the pharmacokinetic differences in the medication formulations. On the other hand, patients may experience medication side effects for a longer period of time with LAIs than with oral medications, again because of pharmacokinetic differences. In addition, patients may experience injection-related side effects, including pain, swelling, redness, or induration at the injection site, with LAI antipsychotic agents.

Patient Preferences

Clinical experience suggests that many patients are cooperative with and accepting of an LAI antipsychotic medication as part of a treatment plan, particularly when the option of an LAI and the pluses and minuses of an LAI antipsychotic are reviewed in the context of shared decision-making (Caroli et al. 2011; Heres et al. 2007; Kane et al. 2019; Waddell and Taylor 2009; Weiden et al. 2015; Yeo et al. 2019). Attitudes about LAIs are typically more positive among patients who have previously received or currently receive an LAI antipsychotic medication than in those who have never been treated with an LAI medication (Patel et al. 2009). Many patients prefer the convenience of receiving an infrequent injection rather than needing to remember to take oral medications. They also value the potential benefits of an LAI antipsychotic medication in terms of better subjective symptom reduction or improved long-term outcomes. On the other hand, some patients may not wish to experience the discomfort associated with receiving injections of medications.

Balancing of Benefits and Harms

The potential benefits of this guideline statement were viewed as likely to outweigh the potential harms. The outcomes associated with use of an LAI formulation of an antipsychotic medication are at least as good as using an oral formulation of the medication and may be better, particularly in terms of treatment discontinuation, rehospitalization, and mortality risk. Many experts infer that the relative benefits of LAI antipsychotic medications as compared with equivalent oral formulations are related to improved adherence (Velligan et al. 2010; West et al. 2008), although specific data to test this supposition are not available. Nevertheless, use of an LAI antipsychotic may have additional advantages in patients who have difficulty with adherence or in whom adherence is uncertain. The side effects of treatment with an LAI antipsychotic medication are also comparable to side effects with the corresponding oral medication. For additional discussion of the research evidence, see Appendix C, Statement 10.

Differences of Opinion Among Writing Group Members

There were no differences of opinion. The writing group voted unanimously in favor of this suggestion.

Review of Available Guidelines From Other Organizations

Information from other guidelines is consistent with this guideline statement (Barnes et al. 2011; Buchanan et al. 2010; Galletly et al. 2016; Hasan et al. 2012; National Institute for Health and Care Excellence 2014; Remington et al. 2017; Scottish Intercollegiate Guidelines Network 2013). Other guidelines on the treatment of schizophrenia suggest use of LAIs based on patient preference (BAP, CSG, NICE, PORT, RANZCP, SIGN) or in the context of poor or uncertain adherence (BAP, NICE, RANZCP, SIGN). The RANZCP and WFSBP guidelines also note that LAIs should be considered if there has been a poor response to oral medication, and the RANZCP guideline notes that LAIs should be offered to patients early in the clinical course of schizophrenia.

Quality Measurement Considerations

As a suggestion, this guideline statement is not appropriate for use as a quality measure. However, only a small proportion of individuals with schizophrenia receive an LAI antipsychotic medication in clinical practice (Brown et al. 2014; Correll et al. 2016; Lawson et al. 2015; Sultana et al. 2019). Because adherence is poor or uncertain in many individuals with schizophrenia, patients may benefit from or prefer to receive an LAI antipsychotic medication if one is offered. Consequently, electronic decision support could suggest that clinicians consider an LAI antipsychotic medication if poor adherence is documented or with repeated hospitalizations or emergency visits. In addition, health care organizations may wish to ensure that they will provide treatment with an LAI antipsychotic medication to patients when appropriate. Health care organizations and health plans may also wish to implement internal process measures to assess and increase rates at which LAI antipsychotics are used.

Statement 11: Anticholinergic Medications for Acute Dystonia

APA recommends (1C) that patients who have acute dystonia associated with antipsychotic therapy be treated with an anticholinergic medication.

Implementation

Medication-induced acute dystonia is defined in DSM-5 as

[a]bnormal and prolonged contraction of the muscles of the eyes (oculogyric crisis), head, neck (torticollis or retrocollis), limbs, or trunk developing within a few days of starting or raising the dosage of a medication (such as a neuroleptic) or after reducing the dosage of a medication used to treat extrapyramidal symptoms. (American Psychiatric Association 2013a, p. 711)

A dystonic spasm of the axial muscles along the spinal cord can result in opisthotonos, in which the head, neck, and spinal column are hyperextended in an arched position. Rarely, acute dystonia can also present as life-threatening laryngospasm, which results in an inability to breathe (Ganesh et al. 2015; Koek and Pi 1989). Acute dystonia is sudden in onset and painful and can cause patients great distress. Because of the dramatic appearance of acute dystonia, health professionals who are unfamiliar with the condition may incorrectly attribute these reactions to catatonic signs or unusual behavior on the part of patients, and oculogyric crises can sometimes be misinterpreted as indicative of seizure activity. In individuals treated with FGAs, it is estimated that up to 10% of patients may experience an acute dystonic episode, and with SGAs, rates of acute dystonia may be less than 2% (Martino et al. 2018; Miller et al. 2008; Satterthwaite et al. 2008). Additional factors that increase the risk of acute dystonia with antipsychotic medication include young age, male sex, ethnicity, recent cocaine use, high medication dose, and intramuscular route of medication administration (Gray and Pi 1998; Spina et al. 1993; van Harten et al. 1999).

There are a limited number of clinical studies of anticholinergic medications in acute dystonia associated with antipsychotic therapy. Nevertheless, a large amount of clinical experience suggests that acute dystonia can be reversed by administration of diphenhydramine, a histamine receptor antagonist with anticholinergic properties. Typically, it is administered intramuscularly to treat acute dystonia, but it can also be administered intravenously in emergent situations, as with acute dystonia associated with laryngospasm. Alternatively, benztropine can also be administered intramuscularly. Once the acute dystonia has resolved, it may be necessary to continue an oral anticholinergic medication to prevent recurrence, at least until other changes in medications can take place such as reducing the dose of medication or changing to an antipsychotic medication that is less likely to be associated with acute dystonia. Typically, a medication such as benztropine or trihexyphenidyl is used for this purpose because of the shorter half-life of oral diphenhydramine and a need for more frequent dosing. For additional details of dosing and use of these medications, see Statement 12, Table 10. Regardless of the anticholinergic medication that is chosen, it is important to use the lowest dose that is able to treat acute dystonia and continue the anticholinergic medication for the shortest time needed to prevent dystonia from recurring. After several weeks to months, anticholinergic medications can sometimes be reduced or withdrawn without recurrence of dystonia or worsening of other antipsychotic-induced neurological symptoms (Desmarais et al. 2012). Medications with anticholinergic effects can result in multiple difficulties for patients, including impaired quality of life, impaired cognition, and significant health complications (Salahudeen et al. 2015). Dry mouth due to anticholinergic effects is associated with an increased risk for multiple dental complications (Singh and Papas 2014), and drinking high-calorie fluids in response to dry mouth can contribute to weight gain. Medications with anticholinergic effects can also precipitate acute angle-closure glaucoma (Lachkar and Bouassida 2007), although patients with treated glaucoma seem to be able to tolerate these medications with careful monitoring (Bower et al. 2018). Other peripheral side effects of anticholinergic medications can include blurred vision, constipation, tachycardia, urinary retention, and effects on thermoregulation (e.g., hyperthermia in hot weather) (Nasrallah and Tandon 2017; Ozbilen and Adams 2009), and central anticholinergic effects can include impaired learning and memory and slowed cognition (Ang et al. 2017; Vinogradov et al. 2009). Older individuals can be particularly sensitive to these anticholinergic effects and can develop problems such as urinary retention, confusion, fecal impaction, and anticholinergic toxicity (with delirium, somnolence, and hallucinations) (Nasrallah and Tandon 2017). In addition, it is important to consider the anticholinergic side effects associated with other medications that a patient is taking, such as antipsychotic medications, some antidepressant medications, urological medications (e.g., oxybutynin), and nonselective antihistamines (e.g., hydroxyzine, diphenhydramine).

Balancing of Potential Benefits and Harms in Rating the Strength of the Guideline Statement

Benefits

In individuals who have acute dystonia associated with antipsychotic therapy, the use of medications with anticholinergic properties (including diphenhydramine, benztropine, and trihexyphenidyl) can be associated with rapid symptom relief. In addition, continuing treatment with an anticholinergic medication can prevent the return of dystonia until other adjustments to the treatment regimen can be made to minimize the risk of recurrence.

Harms

The harms of using a medication with anticholinergic properties to treat acute dystonia include side effects such as dry mouth, blurred vision, precipitation of acute angle-closure glaucoma, constipation (and in some cases fecal impaction), tachycardia, urinary retention, effects on thermoregulation (e.g., hyperthermia in hot weather), impaired learning and memory, slowed cognition, and anticholinergic toxicity (with delirium, somnolence, and hallucinations). These harms are likely to be greater in older individuals and may be augmented in individuals taking other medications with anticholinergic properties.

Patient Preferences

Clinical experience suggests that acute dystonia associated with antipsychotic therapy is very uncomfortable for patients, and most of them are frightened by it. As a result, patients are typically cooperative with and accepting of acute treatment with an anticholinergic agent. They may also be willing to take one of these medications to prevent the return of dystonia. However, some patients may be troubled by side effects such as blurred vision, dry mouth, and constipation and may wish to avoid more significant side effects associated with anticholinergic medications.

Balancing of Benefits and Harms

The potential benefits of this guideline statement were viewed as far outweighing the potential harms. For the majority of patients who are experiencing acute dystonia associated with antipsychotic therapy, the rapid relief of symptoms with anticholinergic treatment outweighs the side effects associated with these medications, at least on a short-term basis. In patients who experience acute laryngeal dystonia, rapid administration of a medication with anticholinergic properties, such as diphenhydramine, can be lifesaving. Nevertheless, the long-term benefits and harms of anticholinergic medications are less clear, and, in this context, harms may outweigh benefits. For additional discussion of the research evidence, see Appendix C, Statement 11.

Differences of Opinion Among Writing Group Members

Eight writing group members voted to recommend this statement. One writing group member disagreed with this statement out of concern that a reduction in antipsychotic medication dose or a change in medication may be preferable to immediate use of an anticholinergic medication in some situations. In addition, one writing group member expressed concern that the use of the phrase “anticholinergic medication” in the statement may be misleading because diphenhydramine is typically viewed as an antihistamine but may be preferable to other anticholinergic medications to treat acute dystonia.

Review of Available Guidelines From Other Organizations

The guidelines of the WFSBP are in agreement with this recommendation, noting that acute dystonia responds dramatically to administration of anticholinergic or antihistaminic medication (Hasan et al. 2013). The guideline of the BAP notes that use for acute dystonia “should be determined on an individual basis, taking account of factors such as the patient’s history of extrapyramidal side effects and the risk of anticholinergic side effects” (Barnes et al. 2011, p. 7).

Quality Measurement Considerations

This guideline statement is not appropriate for use as a quality measure or as part of electronic clinical decision support. Even with short-term treatment, some patients could have the potential to develop significant anticholinergic side effects, which would need to be incorporated into exclusion and exemption criteria. With reductions in the use of high doses of high-potency FGAs, the frequency of acute dystonia is significantly reduced. Any measure would apply only to a small number of individuals, which would complicate testing for feasibility, usability, reliability, and validity.

Statement 12: Treatments for Parkinsonism

APA suggests (2C) the following options for patients who have parkinsonism associated with antipsychotic therapy: lowering the dosage of the antipsychotic medication, switching to another antipsychotic medication, or treating with an anticholinergic medication.

Implementation

Medication-induced parkinsonism, which is termed neuroleptic-induced parkinsonism and other medication-induced parkinsonism in DSM-5, is defined as

[p]arkinsonian tremor, muscular rigidity, akinesia (i.e., loss of movement or difficulty initiating movement), or bradykinesia (i.e., slowing movement) developing within a few weeks of starting or raising the dosage of a medication (e.g., a neuroleptic) or after reducing the dosage of a medication used to treat extrapyramidal symptoms. (American Psychiatric Association 2013a, p. 709)

These symptoms of medication-induced parkinsonism are dose-dependent and generally resolve with discontinuation of antipsychotic medication. It is important to appreciate that medication-induced parkinsonism can affect emotional and cognitive function, at times in the absence of detectable motor symptoms. As a result, it can be difficult to distinguish the negative symptoms of schizophrenia or concomitant depression from medication-induced parkinsonism. In addition, emotional and cognitive features of medication-induced parkinsonism can be subjectively unpleasant and can contribute to poor medication adherence (Acosta et al. 2012; Ascher-Svanum et al. 2006).

A number of approaches can be taken when a patient is experiencing medication-induced parkinsonism. A reduction in the dose of the antipsychotic medication, if feasible, is often helpful in reducing parkinsonism. In some individuals, it may be appropriate to change the antipsychotic medication to one with a lower likelihood of parkinsonism (see Statement 4, Table 6). For individuals who are highly sensitive to medication-induced parkinsonism, clozapine may be considered. However, before reducing the dose of medication or changing to another antipsychotic medication, the benefits of reduced parkinsonism should be weighed against the potential for an increase in psychotic symptoms. Careful monitoring for symptom recurrence is always important when making changes or reducing doses of antipsychotic medications, and use of quantitative measures can be helpful in this regard, as described in Statement 2.

The use of an anticholinergic medication is another option, either on a short-term basis, until a change in dose or a change in medication can occur, or on a longer-term basis, if a change in dose or change in medication is not feasible. In most circumstances, an anticholinergic medication will be started only after parkinsonian symptoms are apparent. However, some individuals may be at increased risk of developing parkinsonism (e.g., those with significant parkinsonism with prior treatment), and prophylactic use of an anticholinergic medication may occasionally be warranted.

Typically, a medication such as benztropine or trihexyphenidyl is used to treat medication-induced parkinsonism because diphenhydramine has a shorter half-life and greater likelihood of sedation. However, oral or intramuscular diphenhydramine can also be used on an acute basis. Additional details on these medications are provided in Table 10. It should also be noted that different symptoms of parkinsonism (e.g., rigidity, tremors, akinesia) may have a differential response to anticholinergic medications, and different treatment approaches may be needed to address each of these symptoms. Parkinsonism should also be distinguished from tardive dyskinesia, which can be worsened by use of anticholinergic medications (Bergman and Soares-Weiser 2018; Cogentin 2013).

Medications for treatment of medication-induced parkinsonisma

Amantadine

Benztropine mesylate

Diphenhydramine

Trihexyphenidyl hydrochloride

Trade nameb

Symmetrel

Cogentin

Benadryl

Artane

Typical use

Parkinsonism

Acute dystonia, parkinsonism

Acute dystonia, parkinsonism

Acute dystonia, parkinsonism

Mechanism of action

Uncompetitive NMDA receptor antagonist (weak)

Muscarinic antagonist

Histamine H1 antagonist

Muscarinic antagonist

Available formulations (mg, unless otherwise noted)

Tablet: 100

Tablet: 0.5, 1, 2

Capsule: 25, 50

Oral elixir: 0.4/mL (473 mL)

Tablet, extended release: 129, 193, 258

Solution, injection: 1/mL (2 mL)

Oral elixir: 12.5/5 mL

Tablet: 2, 5

Capsule: 100

Oral solution: 12.5/ 5 mL, 6.25/1 mL

Capsule, liquid filled: 100

Tablet: 25, 50

Capsule, extended release: 68.5, 137

Solution, injection: 50/1 mL

Oral syrup: 50/5 mL

Other brand-name formulations are available for allergy relief

Typical dose range (mg/day)

Immediate-release tablet or capsule: 100–300

Tablet: 0.5–6.0

Oral: 75–200

Oral: 5–15

Extended-release tablet: 129–322

Solution, injection: 1–2

Solution, injection: 10–50

Bioavailability

86%–94%

29%

40%–70%

100%

Time to peak level (hours)

Immediate release: 2–4

Oral: 7

1–4

1.3

Extended release: 7.5–12

IM: minutes

Protein binding

67%

95%

76%–85%

Not known

Metabolism

Primarily renal

Hepatic

Hepatic

Not known

Metabolic enzymes/transporters

Substrate of organic cation transporter 2

Substrate of CYP2D6 (minor)

Extensively hepatic N-demethylation via CYP2D6; minor demethylation via CYP1A2, CYP2C9, and CYP2C19; inhibits CYP2D6 (weak)

None known

Metabolites

Multiple; unknown activity

Not known

Inactive

Not known

Elimination half-life (hours)

16–17

7

4–8

4

Excretion

Urine 85% unchanged; 0.6% fecal

Urine

Urine (as metabolites and unchanged drug)

Urine and bile

Hepatic impairment

No dose adjustments noted in labeling

No dose adjustments noted in labeling

No dose adjustments noted in labeling

No dose adjustments noted in labeling

Renal impairment

Elimination half-life increases with renal impairment

No dose adjustments noted in labeling

No dose adjustments noted in labeling; however, dosing interval may need to be increased or dosage reduced in older individuals and those with renal impairments

No dose adjustments noted in labeling

Comments

Negligible removal by dialysis; do not crush or divide extended-release products

Onset of action with IV dose is comparable to IM

Total daily dose typically divided into 3–4 doses per day

Maximum daily dose 300 mg for oral and 400 mg for IM/IV, with 100 mg maximum dose for IV/IM

IV dose at a rate of 25 mg/minute; IM dose by deep IM injection because subcutaneous or intradermal injection can cause local necrosis

aThis table includes information compiled from multiple sources. Detailed information on such issues as dose regimen, dose adjustments, medication administration procedures, handling precautions, and storage can be found in product labeling. It is recommended that readers consult product labeling information for authoritative information on these medications.

bThe most common U.S. trade names are included for reference only. At the time of publication, some of these products may be manufactured only as generic products. Other medications or other formulations of the listed medications may be available in Canada.

Abbreviations. CYP = cytochrome P450; NMDA = N-methyl-d-aspartate.

Medications for treatment of medication-induced parkinsonisma

Enlarge table

If an anticholinergic medication is used, it is important to adjust the medication to the lowest dose that is able to treat the parkinsonian symptoms. In addition, it is also important to use the medication for the shortest time necessary. After several weeks to months, anticholinergic medications can sometimes be reduced or withdrawn without recurrence of parkinsonism or worsening of other antipsychotic-induced neurological symptoms (Desmarais et al. 2012). Medications with anticholinergic effects can result in multiple difficulties for patients, including impaired quality of life, impaired cognition, and significant health complications (Salahudeen et al. 2015). Dry mouth due to anticholinergic effects is associated with an increased risk for multiple dental complications (Singh and Papas 2014), and drinking high-calorie fluids in response to dry mouth can contribute to weight gain. Medications with anticholinergic effects can also precipitate acute angle-closure glaucoma (Lachkar and Bouassida 2007), although patients with treated glaucoma seem to be able to tolerate these medications with careful monitoring (Bower et al. 2018).

Other peripheral side effects of anticholinergic medications can include blurred vision, constipation, tachycardia, urinary retention, and effects on thermoregulation (e.g., hyperthermia in hot weather) (Nasrallah and Tandon 2017; Ozbilen and Adams 2009), and central anticholinergic effects can include impaired learning and memory and slowed cognition (Ang et al. 2017; Vinogradov et al. 2009). Older individuals can be particularly sensitive to these anticholinergic effects and can develop problems such as urinary retention, confusion, fecal impaction, and anticholinergic toxicity (with delirium, somnolence, and hallucinations) (Nasrallah and Tandon 2017). In addition, it is important to consider the anticholinergic side effects associated with other medications that a patient is taking, such as antipsychotic medications, some antidepressant medications, urological medications (e.g., oxybutynin), and nonselective antihistamines (e.g., hydroxyzine, diphenhydramine).

Amantadine is an alternative to using an anticholinergic medication to treat medication-induced parkinsonism. Studies of amantadine have had small samples, but the available evidence and clinical experience suggest that amantadine may have comparable or somewhat less benefit than anticholinergic agents in treating medication-induced parkinsonism (Ananth et al. 1975; Borison 1983; DiMascio et al. 1976; Fann and Lake 1976; Greenblatt et al. 1977; Kelly et al. 1974; König et al. 1996; McEvoy 1987; McEvoy et al. 1987; Mindham et al. 1972; Silver et al. 1995). With the absence of anticholinergic properties, side effects, including cognitive impairment, are less prominent with amantadine than with anticholinergic agents. Common adverse effects with amantadine include nausea, dizziness, insomnia, nervousness, impaired concentration, fatigue, and livedo reticularis. Hallucinations and suicidal thoughts have also been reported, as has an increased seizure frequency in individuals with preexisting seizure disorder (Micromedex 2019).

Balancing of Potential Benefits and Harms in Rating the Strength of the Guideline Statement

Benefits

In individuals who have medication-induced parkinsonism, a reduction in signs and symptoms such as rigidity, tremor, and bradykinesia can be of significant benefit, whether such a reduction is achieved by reducing the dose of antipsychotic medication, changing to another antipsychotic medication that has less propensity to cause parkinsonism, or using medications with anticholinergic properties to treat the parkinsonism.

Harms

Reducing the dose of an antipsychotic medication or changing to a different antipsychotic medication can be associated with an increase in psychotic symptoms. The harms of using a medication with anticholinergic properties to treat medication-induced parkinsonism include side effects such as dry mouth, blurred vision, precipitation of acute angle-closure glaucoma, constipation (and in some cases fecal impaction), tachycardia, urinary retention, effects on thermoregulation (e.g., hyperthermia in hot weather), impaired learning and memory, slowed cognition, and anticholinergic toxicity (with delirium, somnolence, and hallucinations). These harms are likely to be greater in older individuals and may be augmented in individuals taking other medications with anticholinergic properties.

Patient Preferences

Clinical experience suggests that most patients are bothered by medication-induced parkinsonism and would like to minimize or eliminate this side effect of antipsychotic medication. However, most patients will also want to minimize the chance that psychotic symptoms will increase. Many patients are also troubled by side effects such as blurred vision, dry mouth, and constipation and may wish to avoid more significant side effects associated with anticholinergic medications. Consequently, the balance of these possible risks and benefits of different approaches to addressing medication-induced parkinsonism is likely to vary for each individual and his or her risk factors and personal preferences.

Balancing of Benefits and Harms

The potential benefits of this guideline statement were viewed as likely to outweigh the potential harms because medication-induced parkinsonism can affect the patient’s quality of life, and patients would prefer to address it, if feasible. However, each of the available options for decreasing or eliminating medication-induced parkinsonism has associated risks and characteristics, and preferences of each patient need to be taken into consideration. In addition, the long-term benefits and harms of anticholinergic medications are less clear, and harms of long-term use may outweigh benefits. For additional discussion of the research evidence, see Appendix C, Statement 12.

Differences of Opinion Among Writing Group Members

Eight writing group members voted to suggest this statement. One writing group member disagreed with this statement, believing that a reduction in antipsychotic medication dose or a change in medication would be preferable to use of an anticholinergic medication.

Review of Available Guidelines From Other Organizations

Statements from other guidelines vary in their approach to medication-induced parkinsonism. The WFSBP guideline notes that use of SGAs or reductions in medication doses should be the primary treatment for medication-induced parkinsonism (Hasan et al. 2013). The BAP guideline notes that decisions about the use of anticholinergic medications for medication-induced parkinsonism should be made on an individual basis, but these medications should not be given prophylactically (Barnes et al. 2011). The PORT guideline notes that prophylactic use of antiparkinsonian agents is not warranted in patients treated with SGAs but may be indicated on an individual basis in patients treated with FGAs (Buchanan et al. 2010).

Quality Measurement Considerations

As a suggestion, this statement is not appropriate for use as a quality measure. It is also not appropriate for incorporation into electronic decision support.

Statement 13: Treatments for Akathisia

APA suggests (2C) the following options for patients who have akathisia associated with antipsychotic therapy: lowering the dosage of the antipsychotic medication, switching to another antipsychotic medication, adding a benzodiazepine medication, or adding a beta-adrenergic blocking agent.

Implementation

Medication-induced acute akathisia is defined in DSM-5 as

[s]ubjective complaints of restlessness, often accompanied by observed excessive movements (e.g., fidgety movements of the legs, rocking from foot to foot, pacing, inability to sit or stand still), developing within a few weeks of starting or raising the dosage of a medication (such as a neuroleptic) or after reducing the dosage of a medication used to treat extrapyramidal symptoms. (American Psychiatric Association 2013a, p. 711)

Akathisia is sometimes difficult to distinguish from psychomotor agitation associated with psychosis, leading to a cycle of increasing doses of antipsychotic medication that lead to further increases in akathisia. Even in mild forms in which the patient is able to control most movements, akathisia is often extremely distressing to patients and is a frequent cause of nonadherence with antipsychotic treatment. If allowed to persist, akathisia can contribute to feelings of dysphoria and, in some instances, suicidal behaviors. The reported rates of akathisia vary from 10%–15% to as many as one-third of patients treated with antipsychotic medication, even when SGAs are used (Juncal-Ruiz et al. 2017; Martino et al. 2018; Mentzel et al. 2017; Miller et al. 2008).

A number of approaches can be taken when a patient is experiencing antipsychotic-induced akathisia. A reduction in the dose of the antipsychotic medication, if feasible, is often helpful in reducing akathisia. In some individuals, it may be appropriate to change the antipsychotic medication to one with a lower likelihood of akathisia (see Statement 4, Table 6). However, before reducing the dose of medication or changing to another antipsychotic medication, the benefits of reduced akathisia should be weighed against the potential for an increase in psychotic symptoms. Careful monitoring for symptom recurrence is always important when making changes or reducing doses of antipsychotic medications, and use of quantitative measures can be helpful in this regard, as described in Statement 2.

Benzodiazepine medications, including lorazepam and clonazepam, can also be helpful in the treatment of akathisia. Among other side effects, somnolence and cognitive difficulties can be associated with benzodiazepine use (Lexicomp 2019; Micromedex 2019). In addition, problems with coordination as a result of benzodiazepines can contribute to falls, particularly in older individuals (Donnelly et al. 2017). Although benzodiazepines are much safer than older sedative agents, respiratory depression can be seen with high doses of a benzodiazepine, particularly in combination with alcohol, other sedating medications, or opioids (Hirschtritt et al. 2017). Caution may also be indicated in prescribing benzodiazepines to individuals with sleep apnea, although few studies are available (Mason et al. 2015). Individuals who are treated with a benzodiazepine may also take them in higher amounts or more frequently than intended. In some patients, a sedative, hypnotic, or anxiolytic use disorder may develop, particularly in individuals with a past or current diagnosis of alcohol use disorder or another substance use disorder.

Another option for treatment of akathisia is the β-adrenergic blocking agent propranolol (Pringsheim et al. 2018), which is typically administered in divided doses, with a total daily dose of 30–120 mg. When using propranolol, it is important to monitor blood pressure with increases in dose and recognize that taking propranolol with protein-rich foods can increase bioavailability by 50%. In addition, propranolol is metabolized by CYP1A2, CYP2D6, CYP2C19, and CYP3A4, which can contribute to drug-drug interactions. Some literature also suggests that mirtazapine may reduce akathisia in some patients (Perry et al. 2018; Poyurovsky and Weizman 2018; Praharaj et al. 2015). In contrast, akathisia tends not to respond to anticholinergic agents (Pringsheim et al. 2018; Rathbone and Soares-Weiser 2006).

Balancing of Potential Benefits and Harms in Rating the Strength of the Guideline Statement

Benefits

In individuals who have akathisia associated with antipsychotic medication, a reduction in symptoms can be of significant benefit, whether such a reduction is achieved by reducing the dose of antipsychotic medication, changing to another antipsychotic medication that has less propensity to cause akathisia, or using a benzodiazepine or a β-adrenergic blocking agent to treat akathisia.

Harms

Reducing the dose of an antipsychotic medication or changing to a different antipsychotic medication can be associated with an increase in psychotic symptoms. The harms of using a benzodiazepine can include somnolence, cognitive difficulties, problems with coordination, and risk of misuse or development of a sedative use disorder. In high doses and particularly in combination with alcohol, other sedating medications, or opioids, respiratory depression may occur. With use of a β-adrenergic blocking agent, such as propranolol, the primary harm relates to lowering of blood pressure.

Patient Preferences

Clinical experience suggests that most patients are bothered by akathisia and, in some instances, very distressed by it. Thus, almost all patients would like to minimize or eliminate this side effect of antipsychotic medication. However, most patients will also want to minimize the chance that psychotic symptoms will increase. They may also be concerned about the possible side effects of medications such as benzodiazepines and β-adrenergic blocking agents. Consequently, the balance of these possible risks and benefits of different approaches to addressing akathisia are likely to vary for each individual and his or her risk factors and personal preferences.

Balancing of Benefits and Harms

The potential benefits of this guideline statement were viewed as likely to outweigh the potential harms because akathisia can affect the patient’s quality of life, and patients would prefer to address it, if feasible. However, each of the available options for decreasing or eliminating akathisia has associated risks and characteristics, and the preferences of each patient need to be taken into consideration. For additional discussion of the research evidence, see Appendix C, Statement 13.

Differences of Opinion Among Writing Group Members

There were no differences of opinion. The writing group voted unanimously in favor of this suggestion.

Review of Available Guidelines From Other Organizations

The WFSBP guideline notes that there is some evidence that benzodiazepines are effective in the treatment of akathisia and that there is very limited evidence to support the use of centrally active β-adrenergic blocking agents in the treatment of akathisia (Hasan et al. 2013).

Quality Measurement Considerations

As a suggestion, this statement is not appropriate for use as a quality measure. It is also not appropriate for incorporation into electronic decision support.

Statement 14: VMAT2 Medications for Tardive Dyskinesia

APA recommends (1B) that patients who have moderate to severe or disabling tardive dyskinesia associated with antipsychotic therapy be treated with a reversible inhibitor of the vesicular monoamine transporter 2 (VMAT2).

Implementation

Tardive syndromes are persistent abnormal involuntary movement disorders caused by sustained exposure to antipsychotic medication, the most common of which are tardive dyskinesia, tardive dystonia, and tardive akathisia (Frei et al. 2018). They begin later in treatment than acute dystonia, akathisia, or medication-induced parkinsonism, and they persist and may even increase, despite reduction in dose or discontinuation of the antipsychotic medication. Typically, tardive dyskinesia presents as “[i]nvoluntary athetoid or choreiform movements (lasting at least a few weeks) generally of the tongue, lower face and jaw, and extremities (but sometimes involving the pharyngeal, diaphragmatic, or trunk muscles)” (American Psychiatric Association 2013a, p. 712), whereas tardive dystonia and tardive akathisia resemble their acute counterparts in phenomenology.

Tardive dyskinesia has been reported after exposure to any of the available antipsychotic medications (Carbon et al. 2017, 2018). It occurs at a rate of approximately 4%–8% per year in adult patients treated with FGAs (Carbon et al. 2018; Woods et al. 2010), a risk that appears to be at least three times that observed with SGAs (Carbon et al. 2018; O’Brien 2016; Woods et al. 2010). Various factors are associated with greater vulnerability to tardive dyskinesia, including age greater than 55 years; female sex; white or African race/ethnicity; presence of a mood disorder, intellectual disability, or central nervous system injury; and past or current akathisia, clinically significant parkinsonism, or acute dystonic reactions (Patterson-Lomba et al. 2019; Solmi et al. 2018a).

Evaluation for the presence of tardive syndromes is important in order to identify them, minimize worsening, and institute clinically indicated treatment. However, evaluation of the risk of tardive dyskinesia is complicated by the fact that dyskinetic movements may be observed with a reduction in antipsychotic medication dose, which is termed a withdrawal-emergent dyskinesia (American Psychiatric Association 2013a). Fluctuations in symptoms are also common and may be influenced by such factors as psychosocial stressors. Furthermore, spontaneous dyskinesias, which are clinically indistinguishable from tardive dyskinesia, have been described in elderly patients before the advent of antipsychotic medications and in up to 20% of never-medicated patients with chronic schizophrenia (Blanchet et al. 2004; Crow et al. 1982; Fenton et al. 1997; Saltz et al. 1991).

Regular assessment of patients for tardive syndromes through clinical examination or through the use of a structured evaluative tool can aid in identifying tardive syndromes, clarifying their likely etiology, monitoring their longitudinal course, and determining the effects of medication changes or treatments for tardive dyskinesia (see Statement 1, Table 2). The AIMS and the DISCUS are examples of such tools (Guy 1976; Kalachnik and Sprague 1993; Munetz and Benjamin 1988). When using scales such as the AIMS or the DISCUS, it should be noted that there is no specific score threshold that suggests a need for intervention, although ranges of scores are noted to correspond with mild, moderate, and severe symptoms. In addition, the same total score can be associated with significantly different clinical manifestations and varying impacts on the patient. Patients, family members, and other persons of support may be able to provide information about the onset of movements; their longitudinal course in relation to treatment or other precipitants; and their impact on functioning, health status (including dentition), and quality of life.

Although the majority of patients who develop tardive dyskinesia have mild symptoms, a small proportion will develop symptoms of moderate or severe degree. In such circumstances, assessment for other contributors to a movement disorder is also warranted (Jinnah and Factor 2015; Mehta et al. 2015; Poewe and Djamshidian-Tehrani 2015; Preskorn et al. 2015; Waln and Jankovic 2015). In addition to a neurological examination and complete history of motor symptoms and past and current medications, history and laboratory testing may include liver function tests, thyroid function tests, serum calcium, complete blood count, and antiphospholipid antibodies. Depending on the results of the history and evaluation, additional studies may be indicated (e.g., ceruloplasmin for Wilson’s disease; brain MRI for basal ganglia changes with Huntington’s disease, stroke, or other lesions; lumbar puncture for anti-NMDA receptor encephalitis). If dyskinetic movements have begun or have increased in the context of antipsychotic dose reduction, it is important to assess the longitudinal course of symptoms for up to several months because spontaneous reductions or resolution of the dyskinesia may occur.

If no contributing etiology is identified and moderate to severe or disabling tardive dyskinesia persists, treatment is recommended with a reversible inhibitor of the VMAT2. Treatment with a VMAT2 inhibitor can also be considered for patients with mild tardive dyskinesia on the basis of such factors as patient preference, associated impairment, or effect on psychosocial functioning. Table 11 shows the characteristics of VMAT2 inhibitors that are currently available in the United States.

Reversible inhibitors of human vesicular monoamine transporter type 2a

Deutetrabenazine

Tetrabenazine

Valbenazine

Trade nameb

Austedo

Xenazine

Ingrezza

Available formulations (mg)

Tablet: 6, 9, 12

Tablet: 12.5, 25

Capsule: 40, 80

Typical dose range (mg/day)

12–48

25–75

40–80

Bioavailability

80%

75%

49%

Time to peak level (hours)

3–4

1–2

0.5–1

Protein binding

60%–68% (α-HTBZ)

82%–85%

> 99%

59%–63% (β-HTBZ)

60%–68% (α-HTBZ)

64% α-HTBZ

59%–63% (β-HTBZ)

Metabolism

Hepatic

Hepatic

Hepatic

Metabolic enzymes/transporters

Major substrate of CYP2D6, minor substrate of CYP1A2 and CYP3A4

Major substrate of CYP2D6

Major substrate of CYP3A4, minor substrate of CYP2D6

Metabolites

Deuterated α-HTBZ and β-HTBZ: active

α-HTBZ, β-HTBZ, and O-dealkylated HTBZ: active

α-HTBZ: active

Elimination half-life (hours)

Deuterated α-HTBZ and β-HTBZ: 9–10

α-HTBZ: 4–8

15–22

β-HTBZ: 2–4

Excretion

Urine: ~ 75%–85% changed

Urine: ~ 75% changed

Urine: 60%

Feces: ~ 8%–11%

Feces: ~ 7%–16%

Feces: 30%

Hepatic impairment

Contraindicated

Contraindicated

Maximum dose of 40 mg daily with moderate to severe impairment (Child-Pugh score 7–15)

Renal impairment

No information available

No information available

Use not recommended in severe renal impairment (CrCl< 30 mL/minute)

Common adverse effects

Sedation

Sedation, depression, extrapyramidal effects, insomnia, akathisia, anxiety, nausea, falls

Sedation

Effect of food on bioavailability

Food affects maximal concentration. Administer with food. Swallow tablets whole and do not chew, crush, or break.

Unaffected by food

Can be taken with or without food. High-fat meals decrease the Cmax and AUC for valbenazine, but values for the active metabolite (α-HTBZ) are unchanged.

Commentsc

Give in divided doses; increase from initial dose of 12 mg/day by 6 mg/week to maximum dose of 48 mg/day. Retitrate dose for treatment interruptions of more than 1 week.

Give in divided doses; increase from initial dose of 25–50 mg/day by 12.5 mg/week to maximum of 150–200 mg/day. Retitrate dose for treatment interruptions of more than 5 days.

Initiate at 40 mg/day and increase to 80 mg/day after 1 week. Continuation of 40 mg/day may be considered for some patients.

Follow product labeling if switching from tetrabenazine to deutetrabenazine. Do not exceed total daily dosage of 36 mg/day (18 mg/dose) in poor CYP2D6 metabolizers or patients taking a strong CYP2D6 inhibitor.

Test for CYP2D6 metabolizer status before giving doses > 50 mg/day.

Use is not recommended with strong CYP3A4 inducer. A reduced dose is recommended with concomitant use of strong CYP3A4 or CYP2D6 inhibitors or in poor CYP2D6 metabolizers.

Assess ECG before and after increasing the daily dose above 24 mg in patients at risk for QTc prolongation.

Do not exceed 50 mg/day in poor metabolizers or in patients treated with a strong inhibitor of CYP2D6.

Avoid use in patients with congenital long QT syndrome, with arrhythmias associated with a prolonged QT interval, or with other risks for QTc prolongation (e.g., drugs known to prolong QTc intervals, reduced metabolism via CYP2D6 or CYP3A4).

Avoid use in patients with congenital long QT syndrome, with arrhythmias associated with a prolonged QT interval, or with other risks for QTc prolongation (e.g., drugs known to prolong QTc intervals, reduced metabolism via CYP2D6).

Avoid use in patients with congenital long QT syndrome, with arrhythmias associated with a prolonged QT interval, or with other risks for QTc prolongation (e.g., drugs known to prolong QTc intervals, reduced metabolism via CYP2D6).

aThis table includes information compiled from multiple sources. Detailed information on such issues as dose regimen, dose adjustments, medication administration procedures, handling precautions, and storage can be found in product labeling. It is recommended that readers consult product labeling information for authoritative information on these medications.

bThe most common U.S. trade names are included for reference only. At the time of publication, some of these products may be manufactured only as generic products.

cAll VMAT2 inhibitors are contraindicated within 2 weeks of a monoamine oxidase inhibitor, within 20 days of reserpine, or in patients with active suicidal ideas or untreated depression. Tetrabenazine and deutetrabenazine carry a boxed warning related to depression and suicidal ideation in patients with Huntington’s disease.

Abbreviations. AUC = area under the curve; Cmax = maximum plasma concentration; CrCl = creatinine clearance; CYP = cytochrome P450; ECG = electrocardiogram; HTBZ = dihydrotetrabenazine.

Reversible inhibitors of human vesicular monoamine transporter type 2a

Enlarge table

In general, deutetrabenazine or valbenazine is preferred over tetrabenazine because of the greater evidence base supporting their use. In addition, tetrabenazine has a shorter half-life and greater rates of associated depression when used in the treatment of patients with Huntington’s disease. Other factors that may influence choice of a VMAT2 inhibitor relate to hepatic or renal function; tetrabenazine and deutetrabenazine are contraindicated in individuals with hepatic impairment, whereas valbenazine is not recommended for use in individuals with severe renal impairment. The metabolism of these medications is also somewhat different. Although all of these medications are substrates for CYP2D6 and CYP3A4, tetrabenazine and deutetrabenazine are major substrates for CYP2D6, whereas valbenazine is a major substrate for CYP3A4. Consequently, the patient’s CYP2D6 metabolizer status or use of concomitant medications that influence these metabolic enzymes may affect the choice of a VMAT2 inhibitor. In terms of side effects, these medications are generally well tolerated, with sedation being most common. In initial studies of tetrabenazine in patients with Huntington’s disease, significant rates of depression were noted as well as concerns about suicidal ideas and behaviors (Shen et al. 2013). However, in studies of deutetrabenazine and valbenazine in patients with tardive dyskinesia, there were no apparent increases in depression or suicidal ideas either in the randomized portions of the clinical trials or in longer open-label extension periods (Solmi et al. 2018b). Nevertheless, depression or suicidal ideas could occur during treatment for tardive dyskinesia, and clinicians will want to be alert to this possibility.

Small clinical trials and case series have examined other treatments for tardive dyskinesia. A lower dose of antipsychotic medication can be considered, although evidence for this approach is minimal (Bergman et al. 2017), and the potential for benefit needs to be weighed against the possibility of recurrent symptoms or relapse. Some benefits have been noted with benzodiazepines (Bergman et al. 2018a), although the potential for benefits must be weighed against the potential side effects of these medications, including somnolence, cognitive difficulties, problems with coordination, and risk of misuse or development of a sedative use disorder. In high doses and particularly in combination with alcohol, other sedating medications, or opioids, respiratory depression may occur. A change in antipsychotic therapy to a lower-potency medication (particularly clozapine) may also be associated with a reduction in tardive dyskinesia, particularly for individuals with moderate to severe symptoms (Mentzel et al. 2018). Again, however, the potential benefits of changing medication should be considered in light of the possibility of symptom recurrence.

In general, giving a higher dose of an antipsychotic may suppress movements of tardive dyskinesia in the short term but would be expected to escalate further development of tardive dyskinesia in the long term. Nevertheless, there may be life-threatening circumstances (e.g., patients with constant movement, gagging, or choking) in which rapid suppression of dyskinesia is needed, and judicious use of an antipsychotic may be appropriate. Anticholinergic medications do not improve and may even worsen tardive dyskinesia (Bergman and Soares-Weiser 2018; Cogentin 2013) in addition to producing significant side effects.

For individuals with other tardive syndromes, other approaches may be helpful on an individual basis. For example, depending on the muscle group that is affected, injections of botulinum toxin have been used to treat tardive dystonia (Brashear et al. 1998; Jinnah and Factor 2015; Kiriakakis et al. 1998). In addition, tardive dystonia may respond to β-adrenergic blocking agents (Hatcher-Martin et al. 2016), and in rare cases of severe intractable tardive dystonia, deep brain stimulation might be considered (Paschen and Deuschl 2018). High doses of anticholinergic agents have also been used to treat severe tardive dystonia (Burke et al. 1982; Kang et al. 1986; Wojcik et al. 1991), although these medications are not useful in treating tardive dyskinesia (Bergman and Soares-Weiser 2018; Cogentin 2013). Reserpine, which also depletes monoamines, should not be used to treat tardive syndromes because it has high rates of associated depression and suicidal ideas and lowers blood pressure (Micromedex 2019). Other treatments, such as vitamin B6 or vitamin E, are less likely to be associated with harms but do not appear to be associated with benefits in treating tardive dyskinesia (Adelufosi et al. 2015; Soares-Weiser et al. 2018a).

Balancing of Potential Benefits and Harms in Rating the Strength of the Guideline Statement

Benefits

In individuals with moderate to severe or disabling tardive dyskinesia associated with antipsychotic therapy, VMAT2 inhibitors can be associated with significant reductions in motor signs and symptoms of tardive dyskinesia. These medications may also be effective in other tardive syndromes.

Harms

The harms of treatment with VMAT2 inhibitors include sedation and, with tetrabenazine, extrapyramidal effects, akathisia, insomnia, anxiety, nausea, and falls. Depression and suicidal ideas have been reported in individuals who were administered VMAT2 inhibitors for treatment of Huntington’s disease. Such effects are possible in individuals treated for tardive dyskinesia, although they were not reported in clinical trials.

Patient Preferences

Clinical experience suggests that most patients with moderate to severe or disabling tardive dyskinesia wish to have a diminution of their motor signs and symptoms. Most patients would be willing to take medication to achieve a reduction in motor signs and symptoms, particularly if it is well tolerated.

Balancing of Benefits and Harms

The potential benefits of this guideline statement were viewed as far outweighing the potential harms. The majority of individuals with moderate to severe or disabling tardive dyskinesia would have a greater likelihood of experiencing benefits of a VMAT2 inhibitor than experiencing harms. Patient preferences to reduce motor signs and symptoms are also likely to favor treatment. For additional discussion of the research evidence, see Appendix C, Statement 14.

Differences of Opinion Among Writing Group Members

There were no differences of opinion. The writing group voted unanimously in favor of this recommendation.

Review of Available Guidelines From Other Organizations

The WFSBP guideline notes that tetrabenazine might have positive effects on tardive dyskinesia (Hasan et al. 2013). It also notes that the risk of tardive dyskinesia is less with SGAs than with FGAs and that there is limited evidence of benefit with clozapine for tardive dyskinesia. Information on a range of other treatments is noted to be even less conclusive. The practice guideline of the American Academy of Neurology, which was published before the availability of deutetrabenazine and valbenazine, notes that tetrabenazine might be considered as a treatment for tardive syndromes (Bhidayasiri et al. 2013).

Quality Measurement Considerations

If a quality measure on VMAT2 inhibitor treatment of tardive syndromes is considered at the provider, facility, health plan, integrated delivery system, or population level, testing of feasibility, usability, reliability, and validity would be essential prior to use for purposes of accountability. However, it may be possible and preferable to incorporate this recommendation into internal facility or health plan initiatives focused on enhanced identification and treatment of tardive syndromes.

Electronic decision support using passive alerts may be able to prompt clinicians to consider a VMAT2 inhibitor, although such prompts would depend on accurate and consistent entry of structured information about the presence of a tardive syndrome, its severity, and its associated degree of disability. Nevertheless, in combination with rating scale data (e.g., AIMS), electronic decision support could help identify individuals with a tardive syndrome who may benefit from a trial of a VMAT2 inhibitor. Information from laboratory data, diagnoses, or problem lists would also be helpful to incorporate in terms of potential contraindications to VMAT2 inhibitor treatment (e.g., tetrabenazine and deutetrabenazine are contraindicated in the presence of hepatic impairment; valbenazine is not recommended for use in individuals with severe renal impairment).

Psychosocial Interventions

Statement 15: Coordinated Specialty Care Programs

APA recommends (1B) that patients with schizophrenia who are experiencing a first episode of psychosis be treated in a coordinated specialty care program.*

*This guideline statement should be implemented in the context of a person-centered treatment plan that includes evidence-based nonpharmacological and pharmacological treatments for schizophrenia.

Implementation

For individuals with a first episode of psychosis, coordinated specialty care (CSC) programs have been developed that integrate a number of evidence-based interventions into a comprehensive treatment package. For example, the NAVIGATE program, which was developed for the Recovery After an Initial Schizophrenia Episode (RAISE) Early Treatment research initiative, uses a collaborative, shared decision-making approach that incorporates family involvement and education, individual resiliency training, supported employment and education, and individualized medication treatment (Mueser et al. 2015; National Institute of Mental Health 2019a). Similar CSC programs, which are sometimes referred to as team-based, multicomponent interventions, have also been used in other countries for treatment of early psychosis (Anderson et al. 2018; Craig et al. 2004; Secher et al. 2015). These treatment programs often include individuals with diagnoses other than schizophrenia but have been associated with a number of benefits, including lower mortality (Anderson et al. 2018), lower rates of relapse, better quality of life, better global function, and greater likelihood of working or being in school after receiving up to 2 years of treatment (McDonagh et al. 2017). Patients in such programs may also experience a greater sense of empowerment and support for their autonomy (Browne et al. 2017). Programs are also available that are aimed at early identification and treatment of attenuated psychosis syndrome or related syndromes of high psychosis risk (J. Addington et al. 2017; Cotton et al. 2016); however, these programs are not within the scope of this guideline recommendation because they include individuals who do not have a psychiatric diagnosis or who have diagnoses other than schizophrenia at later follow-up times (Fusar-Poli et al. 2016; Iorfino et al. 2019).

The main barriers to implementing this recommendation in practice relate to the limited availability of first-episode, multicomponent treatment programs. For state health agencies, health systems, or organizations that are implementing these programs, barriers include such issues as funding, training, and implementation support. However, consultation and implementation materials are available to help guide the establishment of new programs with evidence-based approaches (National Institute of Mental Health 2019b; NAVIGATE 2019; OnTrackNY 2019). Tools are also available to assess fidelity of CSC programs to intended implementation principles (Addington et al. 2016, 2018; Durbin et al. 2019; Essock and Addington 2018).

Balancing of Potential Benefits and Harms in Rating the Strength of the Guideline Statement

Benefits

Use of a CSC program for individuals with a first episode of psychosis can be associated with lower mortality, lower rates of relapse, better quality of life, better global function, and greater likelihood of working or being in school after receiving up to 2 years of treatment (low to moderate strength of research evidence).

Harms

The harms of a CSC program for individuals with a first episode of psychosis are not well delineated but are likely to be small.

Patient Preferences

Clinical experience suggests that many patients with a first episode of psychosis are cooperative with and accepting of a CSC program; however, other patients may not wish to take part in such a program out of a belief that they do not have a condition that requires treatment or because of logistical barriers that influence their ability to access the more intensive treatment provided by such a program.

Balancing of Benefits and Harms

The potential benefits of this guideline statement were viewed as far outweighing the potential harms. CSC treatment programs are generally viewed positively by patients, and they improve a number of patient-oriented outcomes with minimal risk of harms. For additional discussion of the research evidence, see Appendix C, Statement 15.

Differences of Opinion Among Writing Group Members

There were no differences of opinion. The writing group voted unanimously in favor of this recommendation.

Review of Available Guidelines From Other Organizations

Other guidelines did not specifically address the use of CSC programs, but they do endorse many of the individual elements of such programs (e.g., family engagement, psychoeducation, supported employment, medication treatment).

Quality Measurement Considerations

More patients may benefit from CSC programs in the United States than currently receive it. Consequently, state mental health agencies, health plans, and health organizations may wish to implement initiatives to increase the use of a CSC program among individuals with a first episode of psychosis.

This guideline statement would not be appropriate for a performance-based quality measure unless it were tested for feasibility, usability, reliability, and validity. Factors such as geographic variations in treatment availability would need to be considered in testing any quality measures related to CSC program use. It may also be difficult to determine whether a patient is receiving appropriate CSC services.

Electronic decision support using passive alerts may be able to prompt clinicians to consider referral to a CSC program if the presence of a first episode of schizophrenia and the patient’s history of prior treatment were accurately and consistently entered into the electronic record as structured data. The electronic record could also incorporate reference information on the location and referral processes for CSC treatment programs in the local area.

Statement 16: Cognitive-Behavioral Therapy

APA recommends (1B) that patients with schizophrenia be treated with cognitive-behavioral therapy for psychosis (CBTp).*

*This guideline statement should be implemented in the context of a person-centered treatment plan that includes evidence-based nonpharmacological and pharmacological treatments for schizophrenia.

Implementation

The use of cognitive-behavioral therapy (CBT) for individuals with schizophrenia has a number of potential benefits, including improvements in quality of life and global, social, and occupational function and reductions in core symptoms of illness, such as positive symptoms. However, it is important to appreciate that these benefits have been found in studies of CBT that is adapted to use for individuals with psychosis (CBTp), which has some differences from CBT that is focused on other indications. More specifically, CBTp focuses on guiding patients to develop their own alternative explanations for maladaptive cognitive assumptions that are healthier and realistic and do not perpetuate the patient’s convictions regarding the veracity of delusional beliefs or hallucinatory experiences. Thus, the overall approach with CBTp includes developing a collaborative and nonjudgmental therapeutic relationship in which patients can learn to monitor relationships between thoughts, feelings, behaviors, and symptoms and to evaluate the perceptions, beliefs, and thought processes that contribute to symptoms (Beck and Rector 2005; Beck et al. 2009; Beck Institute 2019; Hardy 2019; Kingdon and Turkington 2019; Landa 2019; Lecomte et al. 2016; Morrison 2017; Turkington et al. 2006; Wright et al. 2009). (Videos that demonstrate some of the approaches to CBTp are available at I Can Feel Better, www.icanfeelbetter.org/cbtpskills.) Through this dual focus on monitoring and evaluation, patients can develop beneficial coping strategies and improve functioning, with behavioral self-monitoring serving as a basis for graded task assignments or activity scheduling. In addition, symptoms can be discussed as being within a range of normal experiences (e.g., hearing a loved one’s voice in the context of grief), and alternative explanations for symptoms can be developed that help reduce associated stress (Turkington et al. 2006).

CBTp can be started in any treatment setting, including inpatient settings, and during any phase of illness (Turkington et al. 2006), although some initial reduction in symptoms may be needed for optimal participation (Burns et al. 2014; Valmaggia et al. 2008). It can also be conducted in group as well as in individual formats, either in person or via Web-based delivery platforms. CBTp can also be made available to family members or other persons of support (Turkington and Spencer 2019). Although patient preferences and treatment availability may influence choice of a delivery method, there do not appear to be clear-cut differences in the treatment benefits of group as compared with individual CBTp (McDonagh et al. 2017; Wykes et al. 2008).

The duration of treatment with CBTp has varied in research and clinical practice, with a range from 8 weeks to 5 years of treatment reported in the literature (McDonagh et al. 2017). However, guidelines from other countries recommend a minimum treatment duration of 16 sessions of CBTp (National Institute for Health and Care Excellence 2014; Norman et al. 2017; Scottish Intercollegiate Guidelines Network 2013). Although the available research suggests that treatment benefits are no longer significant when assessed more than 6 months after the end of a CBTp course (McDonagh et al. 2017), it is unclear whether longer durations of treatment with CBTp will result in greater benefits or will help in maintaining treatment-related improvements.

Issues with implementation of CBTp have also been examined. Although the methodological rigor of most studies has been low (Ince et al. 2016), common barriers to CBTp have been identified. For example, some individuals with schizophrenia may be too symptomatic or are experiencing too many side effects (e.g., sedation) to allow effective participation, particularly in inpatient settings. From a patient-centered perspective, CBTp was sometimes viewed as more emotionally challenging and requiring more effort (e.g., homework) than other psychological therapies (Wood et al. 2015). However, engagement strategies, such as motivational interviewing, can be useful in helping patients explore whether CBTp might have potential benefits for them that would offset such concerns.

Attitudinal barriers of staff and organizational management were also found to be common and included a lack of understanding of CBTp and negative expectancies about its value. In addition to inadequate availability of trained staff, staff reported difficulty in identifying patients who were most likely to benefit from CBTp as well as a lack of dedicated time to provide CBTp. Insufficient initial training and insufficient reinforcement of training were also common. Thus, for CBTp to be effective, individuals who are providing CBTp should have appropriate training using established approaches, supervision in CBTp techniques, and experience in treating individuals with schizophrenia. In addition, concerted efforts may be needed to foster positive attitudes and assure adequate time to deliver CBTp. At organizational or health system levels, attention to enhancing the availability of CBTp is also important given the limited availability of CBTp in the United States. Stepped-care approaches, learning collaborative models, and other approaches to best-practice consultations show promise as ways to enhance delivery of CBTp in community settings (Creed et al. 2014; Kopelovich et al. 2019a, 2019b; Stirman et al. 2010).

Balancing of Potential Benefits and Harms in Rating the Strength of the Guideline Statement

Benefits

Use of CBTp in the treatment of schizophrenia can be associated with overall reductions in core illness symptoms, such as positive symptoms (moderate strength of research evidence). CBTp can also be associated with short-term improvements (e.g., for up to 6 months) in quality of life (low strength of research evidence) and global, social, and occupational function (moderate strength of research evidence).

Harms

The harms of CBTp in the treatment of schizophrenia are not well delineated or systematically studied but are likely to be small based on the small number of reported harms in clinical trials.

Patient Preferences

Clinical experience suggests that many patients are cooperative with and accepting of CBTp as part of a treatment plan; however, other patients may not wish to participate in CBTp, may be reluctant to adhere to assignments between sessions, or may experience logistical barriers (e.g., time, access to transportation, childcare, cost) to attending CBTp sessions.

Balancing of Benefits and Harms

The potential benefits of this guideline statement were viewed as far outweighing the potential harms. Specifically, the potential for modest benefits in important patient-centered outcomes during and for periods of up to 6 months after CBTp treatment seemed to outweigh the minimal harms of CBTp treatment. For additional discussion of the research evidence, see Appendix C, Statement 16.

Differences of Opinion Among Writing Group Members

There were no differences of opinion. The writing group voted unanimously in favor of this recommendation.

Review of Available Guidelines From Other Organizations

Statements from other practice guidelines are consistent with this recommendation. The RANZCP and NICE guidelines recommend the use of CBTp for all individuals with schizophrenia (Galletly et al. 2016; National Institute for Health and Care Excellence 2014), whereas the SIGN, CSG, and PORT guidelines recommend CBTp for individuals who have persistent symptoms despite treatment with an antipsychotic medication (Dixon et al. 2010; Norman et al. 2017; Scottish Intercollegiate Guidelines Network 2013). The NICE, SIGN, and CSG guidelines note that CBTp should include at least 16 planned sessions (National Institute for Health and Care Excellence 2014; Norman et al. 2017; Scottish Intercollegiate Guidelines Network 2013).

Quality Measurement Considerations

This guideline statement may not be appropriate for a performance-based quality measure because of the impact of logistical barriers to CBTp, including geographic variations in availability of CBTp, and the difficulty in identifying whether delivered psychotherapy is CBTp. Reminders about CBTp are also not well suited to incorporation into electronic health record clinical decision support. Anecdotal observations suggest that use of CBTp is infrequent in the United States (Kopelovich et al. 2019b). Consequently, health organizations and health plans may wish to implement programs to increase the use of CBTp among individuals with schizophrenia.

Statement 17: Psychoeducation

APA recommends (1B) that patients with schizophrenia receive psychoeducation.*

*This guideline statement should be implemented in the context of a person-centered treatment plan that includes evidence-based nonpharmacological and pharmacological treatments for schizophrenia.

Implementation

Elements of psychoeducation are an integral part of good clinical practice. For example, APA’s Practice Guidelines for the Psychiatric Evaluation of Adults emphasizes the importance of involving patients in treatment-related decision-making and recommends providing the patient with education about the differential diagnosis, risks of untreated illness, treatment options, and benefits and risks of treatment (American Psychiatric Association 2016a). In addition, these informal approaches to psychoeducation have been expanded into formal, systematically delivered programs of psychoeducation that have been evaluated through clinical trials (Pekkala and Merinder 2002; Xia et al. 2011).

The psychoeducational programs that have been studied have varied in their format, duration, and scope. Some psychoeducational programs are delivered on an individual basis, whereas others are delivered in a group format, often in conjunction with family members or other individuals who are involved in the patient’s life. In clinical trials, a 12-session program of psychoeducation is the norm; however, briefer psychoeducation programs of 10 sessions or fewer have also been studied (Pekkala and Merinder 2002; Xia et al. 2011). Typically, psychoeducation is conducted on an outpatient basis, but elements of formal psychoeducation programs can also be incorporated into care in inpatient settings.

Information that is commonly conveyed in a psychoeducation program includes key information about diagnosis, symptoms, psychosocial interventions, medications, and side effects as well as information about stress and coping, crisis plans, early warning signs, and suicide and relapse prevention (Bäuml et al. 2006). Teaching of illness management or self-management strategies (Substance Abuse and Mental Health Services Administration 2010a), as discussed in Statement 21, is often incorporated into psychoeducation. In addition to conveying empathy and respect for the individual, psychoeducation is delivered in a manner that aims to stimulate hope, reassurance, resilience, and empowerment. Typically, psychoeducation incorporates multiple educational modalities, such as workbooks (McCrary et al. 2019), pamphlets, videos, and individual or group discussions in achieving the goals of psychoeducation. Information that may be useful to patients and families as a part of psychoeducation is available through SMI Adviser (https://smiadviser.org). Barriers to providing psychoeducation as a part of the treatment plan relate primarily to program availability. Online delivery of psychoeducation may be one approach to enhancing availability.

Balancing of Potential Benefits and Harms in Rating the Strength of the Guideline Statement

Benefits

Use of psychoeducation in the treatment of schizophrenia can be associated with a number of potential benefits, including improvements in global function (low strength of research evidence) and reductions in relapse rates (moderate strength of research evidence). Enhancements in treatment adherence and improved satisfaction with mental health services have also been noted in some studies.

Harms

The harms of psychoeducation are likely to be minimal on the basis of results from clinical trials that show no differences in the rate of harms experienced by individuals treated with psychoeducation as compared with usual care (low strength of research evidence).

Patient Preferences

Clinical experience suggests that most patients are interested in receiving information about their diagnosis and potential treatments as part of their care. In addition, most patients are accepting of more formal and systematic approaches to psychoeducation. However, some patients may not wish to participate in psychoeducation or may experience logistical barriers (e.g., time, access to transportation, childcare, costs) in attending psychoeducation sessions.

Balancing of Benefits and Harms

The potential benefits of this guideline statement were viewed as far outweighing the potential harms. Specifically, any minimal harms of psychoeducation seem to be outweighed by the potential for modest benefits in important patient-centered outcomes such as improvements in global function and reductions in relapse rates. For additional discussion of the research evidence, see Appendix C, Statement 17.

Differences of Opinion Among Writing Group Members

There were no differences of opinion. The writing group voted unanimously in favor of this recommendation.

Review of Available Guidelines From Other Organizations

Guidelines from other organizations, including CSG, RANZCP, and SIGN, note the value of psychoeducation for individuals with schizophrenia, including information about diagnosis (Galletly et al. 2016; Norman et al. 2017; Scottish Intercollegiate Guidelines Network 2013).

Quality Measurement Considerations

This guideline statement may not be appropriate for a performance-based quality measure because of the diversity of psychoeducational approaches and services and uncertainty regarding linking specific patient needs for psychoeducation with markers of delivery of psychoeducation. Reminders about psychoeducation are also not well suited to incorporation into electronic health record clinical decision support. However, health organizations and health plans may wish to implement quality improvement efforts to increase the use of formal psychoeducational programs among individuals with schizophrenia.

Statement 18: Supported Employment Services

APA recommends (1B) that patients with schizophrenia receive supported employment services.*

*This guideline statement should be implemented in the context of a person-centered treatment plan that includes evidence-based nonpharmacological and pharmacological treatments for schizophrenia.

Implementation

Supported employment differs from other vocational rehabilitation services in providing assistance in searching for and maintaining competitive employment concurrently with job training, embedded job support, and mental health treatment (Becker and Drake 2003; Frederick and VanderWeele 2019; Substance Abuse and Mental Health Services Administration 2010b). In contrast, other vocational rehabilitation approaches focus on training before placement and put greater emphasis on placement in sheltered and transitional employment rather than in a competitive employment setting (Marino and Dixon 2014). For individuals whose goals are related to educational advancement prior to pursuit of employment, supported educational services may also be pursued (Substance Abuse and Mental Health Services Administration 2012b).

Of approaches to supported employment, the bulk of studies involve individual placement and support (IPS; Becker and Drake 2003; Frederick and VanderWeele 2019; McDonagh et al. 2017; Substance Abuse and Mental Health Services Administration 2010b). In addition to a focus on rapid attainment of competitive employment, IPS emphasizes patient preferences in the types of jobs sought, the nature of the services that are delivered, and the outreach that occurs with potential employers (Marino and Dixon 2014). Patient preferences also guide whether to disclose the presence of a psychiatric illness to the employer, with more than half of individuals choosing to disclose this information (DeTore et al. 2019). Services are offered to anyone who is interested, with no exclusion criteria for participation. Additional principles of IPS include individualized long-term job support and integration of employment specialists with the clinical team. Employment specialists also develop relationships with community employers and provide personalized benefits counseling to participants.

Evidence consistently shows that supported employment is associated with greater rates of competitive employment than transitional employment or prevocational training, although prevocational training is superior to no vocational intervention at all (Marshall et al. 2014; McDonagh et al. 2017; Metcalfe et al. 2018; Modini et al. 2016; Richter and Hoffmann 2019; Suijkerbuijk et al. 2017). Augmenting supported employment with symptom-related skills training, training in workplace fundamentals, or cognitive training may assist in gaining and maintaining competitive employment (Dewa et al. 2018; Suijkerbuijk et al. 2017). Other benefits of supported employment include greater number of hours worked per week, a longer duration of each job, a longer duration of total employment, and an increase in earnings (McDonagh et al. 2017). Individuals receiving supported employment are also more likely to obtain job-related accommodations than individuals with mental illness who are not receiving supported employment (McDowell and Fossey 2015). Such accommodations typically relate to assistance from the supported employment coach but may also include flexible scheduling, reduced hours, modified job duties, and modified training and supervision. In addition to job-related benefits of supported employment, there is some evidence of modest reductions in symptoms and a reduced risk of hospitalization associated with obtaining a job (Bouwmans et al. 2015; Burns et al. 2007; Charzyńska et al. 2015; Hoffmann et al. 2014; Luciano et al. 2014).

Among individuals who receive supported employment, factors that may be associated with a greater likelihood of success include lower levels of symptoms, higher levels of cognitive functioning (e.g., attention, memory, executive functioning, psychomotor speed), greater work success in the past, higher levels of educational attainment, and greater interest in obtaining employment (Kirsh 2016). Peer support and support from families and others in the patient’s social network may also be associated with better outcomes, although these factors have been less well studied (Kirsh 2016). Even when individuals do not experience initial success with supported employment, addition of cognitive remediation may improve vocational outcomes (McGurk et al. 2015, 2016).

There are a number of barriers to supported employment, including economic and regulatory factors (Kirsh 2016; Metcalfe et al. 2018; Modini et al. 2016) and the limited number of available programs (Marshall et al. 2014; Sherman et al. 2017). Although data are limited, employers may be reluctant to participate in supported employment out of concern about the impact of providing work-related accommodations and because of discrimination and bias toward individuals with serious mental illness (Kirsh 2016). Treating clinicians may also serve as a barrier by having inappropriately limited expectations (Kirsh 2016) and being unaware that some individuals with schizophrenia are able to function at high levels of occupational achievement (Cohen et al. 2017). In addition, concerns about losing disability benefits or health insurance may lead some individuals to forgo supported employment opportunities (Kirsh 2016). Such concerns are not entirely unrealistic because many of the competitive jobs that individuals do obtain are entry-level and/or part-time positions without health insurance benefits (Kirsh 2016). Within supported employment programs, organizational barriers to success have included poor fidelity to supported employment principles (Marshall et al. 2014); insufficient time devoted to leading and management of the programs; and insufficient training, skills, and business and public relations knowledge of program staff (Kirsh 2016; Swanson et al. 2013). Each of these barriers is important to address at individual, systems, and policy levels so that more patients can benefit from supported employment interventions.

For clinicians and organizations wishing to learn more about supported employment or develop supported employment programs, additional information is available through SMI Adviser (https://smiadviser.org), NAVIGATE (https://navigateconsultants.org/manuals), and the Boston University Center for Psychiatric Rehabilitation (https://cpr.bu.edu).

Balancing of Potential Benefits and Harms in Rating the Strength of the Guideline Statement

Benefits

Use of supported employment as part of the treatment of schizophrenia can be associated with significantly better employment outcomes, including a significantly greater likelihood of obtaining competitive employment, a significantly greater likelihood of working more than 20 hours per week, more weeks of employment, and greater earnings relative to vocational training or no vocational interventions (moderate strength of research evidence).

Harms

The harms of supported employment in the treatment of schizophrenia are not well delineated or systematically reported but are likely to be small.

Patient Preferences

Clinical experience suggests that few patients are currently receiving supported employment, but a significant number of individuals may be interested in supported employment if it were readily available and offered to them. However, some individuals may be in school, have responsibilities at home, or already be employed. Others would rather not seek employment or may have concerns about losses of benefits or health insurance if they did pursue competitive employment. Logistical barriers (e.g., access to transportation, childcare) may also affect patient preferences related to supported employment.

Balancing of Benefits and Harms

The potential benefits of this guideline statement were viewed as far outweighing the potential harms. Specifically, the potential for benefits in important patient-centered outcomes related to employment seemed to outweigh the minimal harms of supported employment programs. For additional discussion of the research evidence, see Appendix C, Statement 18.

Differences of Opinion Among Writing Group Members

There were no differences of opinion. The writing group voted unanimously in favor of this recommendation.

Review of Available Guidelines From Other Organizations

Guidelines from other organizations are generally consistent with this recommendation. NICE and PORT recommend that supported employment be offered to individuals with schizophrenia who wish to find or return to work (Dixon et al. 2010; National Institute for Health and Care Excellence 2014), and RANZCP recommends IPS services for individuals with first-episode psychosis (Galletly et al. 2016). RANZCP, NICE, and CSG also emphasize the appropriateness of other occupational or educational activities for individuals with schizophrenia (Galletly et al. 2016; National Institute for Health and Care Excellence 2014; Norman et al. 2017).

Quality Measurement Considerations

This guideline statement may not be appropriate for a performance-based quality measure because of the barriers to supported employment, including variations in availability, and difficulty identifying when patients desire competitive employment. Reminders about supported employment are also not well suited to incorporation into electronic health record clinical decision support. However, given the infrequent availability of supported employment in the United States, health organizations and health plans may wish to implement programs to increase the use of supported employment among individuals with schizophrenia.

Statement 19: Assertive Community Treatment

APA recommends (1B) that patients with schizophrenia receive assertive community treatment if there is a history of poor engagement with services leading to frequent relapse or social disruption (e.g., homelessness; legal difficulties, including imprisonment).*

*This guideline statement should be implemented in the context of a person-centered treatment plan that includes evidence-based nonpharmacological and pharmacological treatments for schizophrenia.

Implementation

ACT, sometimes referred to as programs of assertive community treatment, is a multidisciplinary, team-based approach in which patients receive individualized care outside a formal clinical setting. Thus, individuals may be engaged in their homes, workplaces, or other community locations. Continuity of care is enhanced because individuals work with an assigned team, which has 24/7 availability, rather than being assigned to a designated clinician for care. Team members typically include a psychiatrist, nurse, and social worker or case manager. Peer specialists, vocational specialists, and clinicians with expertise in substance use treatment are often part of the team as well.

Other features of ACT include its provision of personalized and flexible care that addresses the patient’s needs and preferences without time limits or other constraints on services. Particularly in rural areas, some ACT teams are augmenting face-to-face visits with telepsychiatry visits, although research will be needed to determine whether such an approach alters the benefits of ACT (Swanson and Trestman 2018). ACT teams also work with a smaller number of individuals than traditional outpatient clinicians or case managers do, which contributes to the ability to provide frequent visits and a more personalized and comprehensive approach to care. For these reasons, ACT is often used in individuals who have a history of poor engagement with services that leads to frequent relapse or social disruption (e.g., homelessness; legal difficulties, including imprisonment), although it can also be used for individuals who are better engaged but still have high rates of service utilization. Many individuals who are referred for ACT are also at risk for poor adherence and may benefit from consideration of an LAI antipsychotic medication (see Statement 10).

Studies of ACT suggest that it is associated with symptom improvement comparable to other treatment delivery approaches and that individuals who receive ACT are more likely to be domiciled, living independently, and working and less likely to be hospitalized as compared with individuals who receive treatment as usual (McDonagh et al. 2017). Although ACT has multiple strengths that would make it an attractive approach in individuals with co-occurring disorders and schizophrenia, the impact of ACT on physical health has not been well studied (Vanderlip et al. 2017). Also, in individuals with a concomitant substance use disorder, research to date has not shown associated improvements in functioning, mortality, or substance use as compared with usual care (McDonagh et al. 2017).

In terms of implementation barriers, there is often limited availability of ACT programs. Funding these programs can be challenging because the comprehensive and multidisciplinary nature of ACT services is not well aligned with payment models in the U.S. health care delivery system (Monroe-DeVita et al. 2012). Effective delivery of ACT services is also dependent on having a high level of fidelity to ACT program standards (Monroe-DeVita et al. 2012; Thorning et al. 2016), and this requires considerable training as well as ongoing mentoring, collaboration, and consultation with individuals who are skilled in ACT implementation. Attention to outcomes and organizational culture is also important in providing a team-based approach that is warm, flexible, pragmatic, collaborative, and supportive of patients’ recovery (Monroe-DeVita et al. 2012). For organizations or state mental health systems that are implementing ACT programs, a number of resources are available (Center for Evidence-Based Practices 2019; Substance Abuse and Mental Health Services Administration 2008; Thorning et al. 2016).

Balancing of Potential Benefits and Harms in Rating the Strength of the Guideline Statement

Benefits

Use of ACT in the treatment of schizophrenia can be associated with a number of benefits as compared with treatment as usual, including a greater likelihood of being domiciled, living independently, or working and a lower likelihood of being hospitalized (low to moderate strength of research evidence).

Harms

The harms of ACT in the treatment of schizophrenia are not well delineated but are likely to be small.

Patient Preferences

Clinical experience suggests that most patients are cooperative with and accepting of ACT, particularly once they have engaged with treatment. In some circumstances, patients may be reluctant to accept ACT services because of impaired awareness of a need for treatment. In this context, ACT may be used as one component of court-mandated care (e.g., assisted outpatient treatment, community treatment order, outpatient commitment). However, in the few studies that have examined patient perceptions, ACT is generally viewed as supporting patients and building relationships in a recovery-oriented fashion (Appelbaum and Le Melle 2008; Morse et al. 2016).

Balancing of Benefits and Harms

The potential benefits of this guideline statement were viewed as far outweighing the potential harms. ACT is generally viewed positively by patients, and it improves a number of patient-oriented outcomes with minimal risk of harms. For additional discussion of the research evidence, see Appendix C, Statement 19.

Differences of Opinion Among Writing Group Members

There were no differences of opinion. The writing group voted unanimously in favor of this recommendation.

Review of Available Guidelines From Other Organizations

This guideline recommendation is consistent with the SIGN recommendation to offer assertive outreach to individuals with schizophrenia who “make high use of inpatient services, who show residual psychotic symptoms and who have a history of poor engagement with services leading to frequent relapse and/or social breakdown (for example homelessness)” (Scottish Intercollegiate Guidelines Network 2013, p. 8). The PORT guideline also notes that ACT should be included in systems of care that serve individuals with schizophrenia and that it “should be provided to individuals who are at risk for repeated hospitalizations or have recent homelessness” (Dixon et al. 2010, p. 49). In addition, RANZCP recommends the use of ACT “after initial contact, during crises and after discharge from hospital” in individuals with schizophrenia (Galletly et al. 2016, p. 29).

Quality Measurement Considerations

This guideline statement may not be appropriate for a performance-based quality measure because of the impact of logistical barriers to ACT, including geographic variations in availability. Reminders about ACT are also not well suited to incorporation into electronic health record clinical decision support because of the multiple patient-specific factors that may contribute to a decision to recommend ACT. However, anecdotal observations suggest that more patients may benefit from ACT in the United States than currently receive it. Consequently, state mental health agencies, health plans, and health organizations may wish to implement programs to increase the use of ACT among individuals with schizophrenia who have had a history of poor engagement with services, leading to frequent relapse or social disruption (e.g., homelessness; legal difficulties, including imprisonment).

Statement 20: Family Interventions

APA suggests (2B) that patients with schizophrenia who have ongoing contact with family receive family interventions.*

*This guideline statement should be implemented in the context of a person-centered treatment plan that includes evidence-based nonpharmacological and pharmacological treatments for schizophrenia.

Implementation

An important aspect of good psychiatric treatment is involvement of family members, person(s) of support, and other individuals who play a key role in the patient’s life. In addition to spouses, parents, children, or other biological or nonbiological relatives, such individuals may include people who reside with the patient, intimate partners, and close friends who are an integral part of the patient’s support network. Such individuals benefit from discussion of such topics as diagnosis and management of schizophrenia, types of support that are available, and ways to plan for and access help in a crisis. Other goals include helping individuals repair or strengthen their connections with family members and other members of their support system.

Family interventions are systematically delivered, extend beyond conveying of information, and focus on the future rather than on past events (Mueser et al. 2013). The family interventions that are suggested in this guideline statement go beyond the basics of family involvement and illness education that are important for good clinical care. They may include structured approaches to problem-solving, training in how to cope with illness symptoms, assistance with improving family communication, provision of emotional support, and strategies for reducing stress and enhancing social support networks (McDonagh et al. 2017; McFarlane 2016; Mueser et al. 2013). Family interventions can be particularly important early in the course of schizophrenia (McFarlane 2016) but can also be helpful during any phase of treatment.

Most patients want family to be involved in their treatment (Cohen et al. 2013). Nevertheless, even when a patient does not wish for a specific person to be involved in his or her care, the clinician may listen to information provided by that individual, as long as confidential information is not provided to the informant (American Psychiatric Association 2016a). Although some health professionals may be unsure about legal or regulatory aspects of sharing information, general information that is not specific to the patient can be provided (e.g., common approaches to treatment, general information about medications and their side effects, available support and emergency assistance). Also, to prevent or lessen a serious and imminent threat to the health or safety of the patient or others, the “Principles of Medical Ethics” (American Psychiatric Association 2013f) and HIPAA (Office for Civil Rights 2017a, 2017b) permit clinicians to disclose necessary information about a patient to family members, caregivers, law enforcement, or other persons involved with the patient. HIPAA also permits health care providers to disclose necessary information to the patient’s family, friends, or other persons involved in the patient’s care or payment for care when such disclosure is judged to be in the best interests of the patient and the patient is not present or is unable to agree or object to a disclosure because of incapacity or emergency circumstances (Office for Civil Rights 2017b).

The family interventions that have been studied include a variety of formats and approaches (McDonagh et al. 2017; McFarlane 2016). Interventions may or may not include the patient and can be conducted with a single family or a multifamily group. In many early studies, family interventions included the patient and were led by a member of the patient’s clinical care team. This approach allowed a liaison to develop among the care team, the patient, members of the family, and other person(s) of support. Other studies have been conducted independent of the patient’s care team. In terms of approach, some family interventions focus on psychoeducation, whereas other interventions incorporate other treatment elements (e.g., motivational interviewing, goal setting, cognitive-behavioral intervention, behavioral family therapy, support groups, social network development, communication training, role-playing, stress management, relaxation training). Given the diversity of options for family interventions, the selection of a specific approach should consider the preferences of the patient and family in collaboration with the clinician.

Benefits of family interventions include reductions in core symptoms of illness and reductions in relapses, including rehospitalization (McDonagh et al. 2017). Some studies have also shown benefits for family members such as reductions in levels of burden and distress or improvements in relationships among family members (McFarlane 2016; Sin et al. 2017a). Evidence suggests that benefits of family interventions are greatest when more than 10 treatment sessions are delivered over a period of at least 7 months (McDonagh et al. 2017). However, the Family-to-Family intervention available through the National Alliance on Mental Illness has shown significant benefits using a 12-week program consisting of weekly sessions of 2–3 hours each (Dixon et al. 2011; Lucksted et al. 2013; Marcus et al. 2013; Toohey et al. 2016).

A common barrier to implementing family interventions relates to program availability. However, guidance is available on developing family intervention programs focused on psychoeducation (Glynn et al. 2014; Substance Abuse and Mental Health Services Administration 2009). In addition, the National Alliance on Mental Illness has reduced this barrier through its Family-to-Family program, which has led to a significant expansion in the availability of family interventions (National Alliance on Mental Illness 2019b). Additional barriers include constraints of family members (e.g., work schedules, access to transportation, childcare, health issues) that may limit their ability to be involved in frequent family sessions. Similar logistical barriers can exist for patients when family interventions incorporate patient participation. Other implementation barriers include organizational and clinician-focused barriers such as time and cost constraints and insufficient understanding of the potential benefits of family intervention (Ince et al. 2016).

Balancing of Potential Benefits and Harms in Rating the Strength of the Guideline Statement

Benefits

Use of family interventions in the treatment of schizophrenia can reduce the likelihood of relapse (low to moderate strength of research evidence) and reduce core illness symptoms (low strength of research evidence).

Harms

The harms of family interventions in the treatment of schizophrenia are not well documented but appear to be minimal.

Patient Preferences

Clinical experience suggests that many patients are cooperative with and accepting of family interventions as part of a treatment plan; however, other patients may have had difficulties in relationships with family members in the past and may not want family members to be involved in their treatment.

Balancing of Benefits and Harms

The potential benefits of this guideline statement were viewed as likely to outweigh the potential harms. For patients who have ongoing contact with their families, including relatives and significant others, there are distinct benefits to family interventions. However, some patients may not be in favor of family involvement even when they do have some ongoing contact with family members, and, for this reason, the statement was suggested rather than being recommended for all individuals. For additional discussion of the research evidence, see Appendix C, Statement 20.

Differences of Opinion Among Writing Group Members

Eight writing group members voted in favor of this suggestion. One writing group member disagreed with this statement as worded and felt that it would be preferable for the guideline statement to make specific mention of other persons of support who may be involved with the patient and are commonly included in such interventions in addition to family members.

Review of Available Guidelines From Other Organizations

This guideline statement is consistent with guidelines from other organizations. CSG, NICE, RANZCP, SIGN, and PORT guidelines all recommend offering family interventions when an individual with schizophrenia reside with or are in close contact with family (Dixon et al. 2010; Galletly et al. 2016; National Institute for Health and Care Excellence 2014; Norman et al. 2017; Scottish Intercollegiate Guidelines Network 2013). These guidelines also emphasize the importance of providing information to family and others involved in the patient’s care on such topics as diagnosis and management of schizophrenia, types of support that are available, and ways to access help in a crisis.

Quality Measurement Considerations

As a suggestion, this guideline statement is not appropriate for use as a performance-based quality measure or for incorporation into electronic decision support. Nevertheless, given the potential benefits of this approach, health care organizations and health plans may wish to track the availability and utilization of family interventions.

Statement 21: Self-Management Skills and Recovery-Focused Interventions

APA suggests (2C) that patients with schizophrenia receive interventions aimed at developing self-management skills and enhancing person-oriented recovery.*

*This guideline statement should be implemented in the context of a person-centered treatment plan that includes evidence-based nonpharmacological and pharmacological treatments for schizophrenia.

Implementation

Illness self-management training programs have been applied to help address many chronic conditions and are designed to improve knowledge about one’s illness and management of symptoms (Grady and Gough 2014). Goals include reducing the risk of relapse, recognizing signs of relapse, developing a relapse prevention plan, and enhancing coping skills to address persistent symptoms, with the aim of improving quality of life and social and occupational functioning. In the studies included in the AHRQ review (McDonagh et al. 2017), self-management training was generally delivered in a group setting with sessions of 45–90 minutes each, and the number of intervention sessions ranged from 7 to 48 sessions. However, the evidence suggested better outcomes in patients who participated in at least 10 self-management intervention sessions. Self-management sessions were typically facilitated by clinicians, although peer-facilitated sessions have also been used. In addition, some studies have used individually targeted interventions, either face-to-face or via computer-based formats (Lean et al. 2019). Self-management approaches have also been used to address co-occurring medical conditions in individuals with serious mental illness, including schizophrenia, with benefits that included increased patient activation and improved health-related quality of life (Druss et al. 2010; Goldberg et al. 2013; Muralidharan et al. 2019).

Recovery-focused interventions have also been developed that focus on fostering self-determination in relation to a patient’s personal goals, needs, and strengths. Such approaches may include elements of self-management skill development, psychoeducation, and peer-based interventions but also include components and activities that allow participants to share experiences and receive support, learn and practice strategies for success, and identify and take steps toward reaching personal goals. Studies of peer-based interventions are limited (Castelein et al. 2015; Chien et al. 2019), and studies of recovery-focused interventions have also been small in number. Nevertheless, the available information suggests that these interventions may promote increased recovery, hope, and empowerment among individuals with serious mental illnesses (Le Boutillier et al. 2011; Mueser et al. 2013; Thomas et al. 2018).

The most common barrier to implementing this guideline statement is the availability of programs for developing self-management skills and enhancing person-oriented recovery. However, a toolkit for developing illness management and recovery-based programs in mental health is available through the Substance Abuse and Mental Health Services Administration (https://store.samhsa.gov/product/Illness-Management-and-Recovery-Evidence-Based-Practices-EBP-KIT/sma09-4463). Other resources are available through the Boston University Center for Psychiatric Rehabilitation (https://cpr.bu.edu), the Center on Integrated Health Care and Self-Directed Recovery (www.center4healthandsdc.org), Digital Opportunities for Outcomes in Recovery Services (https://skills.digitalpsych.org), Mental Health America (www.mhanational.org/self-help-tools), the National Alliance on Mental Illness (www.nami.org), NAVIGATE (https://navigateconsultants.org/manuals), SMI Adviser (https://smiadviser.org/individuals-families), and the Temple University Collaborative on Community Inclusion (www.tucollaborative.org).

Balancing of Potential Benefits and Harms in Rating the Strength of the Guideline Statement

Benefits

Use of interventions aimed at developing self-management skills and enhancing person-oriented recovery in individuals with schizophrenia can be associated with reductions in symptom severity and risk of relapse and an increased sense of hope and empowerment (low to moderate strength of research evidence). Self-management approaches that are aimed at addressing co-occurring medical conditions in individuals with serious mental illness also have benefits that include increased patient activation and improved health-related quality of life.

Harms

The harms of interventions aimed at developing self-management skills and enhancing person-oriented recovery in the treatment of schizophrenia are not well studied but are likely to be minimal.

Patient Preferences

Clinical experience suggests that most patients are cooperative with and accepting of interventions aimed at developing self-management skills and enhancing person-oriented recovery. However, some patients may not wish to take part in such interventions because of personal preferences or logistical barriers to attending group sessions (e.g., access to transportation, childcare).

Balancing of Benefits and Harms

The potential benefits of this guideline statement in terms of patient engagement, empowerment, and beneficial outcomes were viewed as likely to outweigh the potential harms, which were viewed as minimal. For additional discussion of the research evidence, see Appendix C, Statement 21.

Differences of Opinion Among Writing Group Members

There were no differences of opinion. The writing group voted unanimously in favor of this suggestion.

Review of Available Guidelines From Other Organizations

This guideline statement is consistent with recommendations of other guidelines (RANZCP, NICE) that support the use of self-management and peer-support programs in the treatment of individuals with schizophrenia (Galletly et al. 2016; National Institute for Health and Care Excellence 2014).

Quality Measurement Considerations

As a suggestion, this guideline statement is not appropriate for use as a performance-based quality measure or for incorporation into electronic decision support. Nevertheless, given the potential benefits of such interventions, health care organizations and health plans may wish to track the availability and utilization of programs to develop self-management skills and enhance person-oriented recovery.

Statement 22: Cognitive Remediation

APA suggests (2C) that patients with schizophrenia receive cognitive remediation.*

*This guideline statement should be implemented in the context of a person-centered treatment plan that includes evidence-based nonpharmacological and pharmacological treatments for schizophrenia.

Implementation

Cognitive remediation approaches are intended to address cognitive difficulties that can accompany schizophrenia, with the aim of enhancing function and quality of life. A number of different cognitive remediation approaches have been used, typically in group or computer-based formats, in an effort to enhance cognitive processes such as attention, memory, executive function, social cognition, or meta-cognition (Delahunty and Morice 1996; Medalia et al. 2018; Reeder et al. 2016; Wykes et al. 2011). Some programs have focused on improving cognitive flexibility (e.g., shifting cognitive sets), working memory (e.g., sequencing, multitasking, delayed recall), and planning (e.g., sequencing and chunking, active coding), whereas meta-cognitive approaches have attempted to teach patients how and when particular strategies that bypass specific cognitive limitations can be used. Some programs add aspects of social and communication skills to neurocognitive elements of remediation (Pentaraki et al. 2017).

Although this variability in program format and content confounds interpretation of the evidence, cognitive remediation does seem to result in improvements in cognition, symptoms, and function in individuals with schizophrenia, at least on a short-term basis (Harvey et al. 2018; McDonagh et al. 2017; Revell et al. 2015). Although long-term follow-up studies of cognitive remediation are not available in individuals with schizophrenia, data from healthy older individuals show long-term improvements as a result of cognitive training (Rebok et al. 2014). Beneficial effects on psychosocial outcomes seem particularly robust when cognitive remediation is used as a component of or adjunct to other forms of psychiatric rehabilitation rather than being delivered as a stand-alone intervention (McGurk et al. 2007; Revell et al. 2015; van Duin et al. 2019; Wykes et al. 2011). However, some apparent improvements in cognitive performance may result from practicing specific tasks and may not produce generalizable changes in other contexts. Furthermore, the specific elements of a particular cognitive remediation program may influence the benefits that are observed (Cella and Wykes 2019).

The primary barriers to use of cognitive remediation are related to program availability. Online delivery of cognitive remediation may be one way to overcome these barriers. Information and training on developing cognitive remediation programs are available (Medalia 2019; Medalia et al. 2018). In addition, Web-based programs have been used in clinical trials and may provide options for patients without access to in-person cognitive remediation programs (Jahshan et al. 2019; Kukla et al. 2018).

Balancing of Potential Benefits and Harms in Rating the Strength of the Guideline Statement

Benefits

Use of cognitive remediation is associated with moderate improvements in specific aspects of cognition (Harvey et al. 2018; Wykes et al. 2011) as well as small positive effects on social, occupational, and global function (low strength of research evidence); core illness symptoms (low strength of research evidence); and negative symptoms (moderate strength of research evidence) compared with usual care over approximately 16 weeks of treatment (McDonagh et al. 2017).

Harms

The harms of cognitive remediation in the treatment of schizophrenia are not well studied but are likely to be small.

Patient Preferences

Evidence from research trials suggests that patients are likely to be cooperative with and accepting of cognitive remediation as part of a treatment plan (Reeder et al. 2016); however, other patients may not wish to participate because of logistical barriers (e.g., time, cost, access to transportation, childcare).

Balancing of Benefits and Harms

The potential benefits of this guideline statement were viewed as likely to outweigh the potential harms, which were viewed as minimal. Differences in patient preferences, variability in the appropriateness of cognitive remediation for individuals with schizophrenia, and the unclear durability of benefits led to suggesting cognitive remediation rather than recommending it. For additional discussion of the research evidence, see Appendix C, Statement 22.

Differences of Opinion Among Writing Group Members

There were no differences of opinion. The writing group voted unanimously in favor of this suggestion.

Review of Available Guidelines From Other Organizations

The RANZCP guideline recommends that cognitive remediation be available to individuals with schizophrenia if cognitive impairment is present and should be specifically “offered when cognitive deficits are affecting recovery and function” (Galletly et al. 2016, p. 20). The SIGN and CSG guidelines note that cognitive remediation “may be considered for individuals diagnosed with schizophrenia who have persisting problems associated with cognitive difficulties” (Norman et al. 2017, p. 621; Scottish Intercollegiate Guidelines Network 2013, p. 28).

Quality Measurement Considerations

As a suggestion, this guideline statement is not appropriate for use as a performance-based quality measure or for incorporation into electronic decision support.

Statement 23: Social Skills Training

APA suggests (2C) that patients with schizophrenia who have a therapeutic goal of enhanced social functioning receive social skills training.*

*This guideline statement should be implemented in the context of a person-centered treatment plan that includes evidence-based nonpharmacological and pharmacological treatments for schizophrenia.

Implementation

Social skills training in the treatment of schizophrenia can improve social function, core illness symptoms, and negative symptoms more than usual care can (McDonagh et al. 2017). Reductions in relapse rates, including rehospitalization rates, have also been noted in some studies (McDonagh et al. 2017).

Social skills training has an overarching goal of improving interpersonal and social skills but can be delivered using a number of approaches (Almerie et al. 2015; Kopelowicz et al. 2006; Turner et al. 2018). These include cognitive-behavioral, social-cognitive, interpersonal, and functional adaptive skills training. Social skills training is delivered in a group format and includes homework assignments to facilitate skill acquisition. Other specific elements of the intervention will vary with the theoretical emphasis of the training. However, examples of techniques that can be used in social skills training include role-playing, modeling, and feedback approaches to enhance interpersonal interactions; behaviorally oriented exercises in assertiveness, appropriate contextual responses, and verbal and nonverbal communication; and instruction and practice with social and emotional perceptions (Almerie et al. 2015; Kopelowicz et al. 2006; Turner et al. 2018). These techniques are aimed at generating improvements in typical social behaviors such as making eye contact, smiling at appropriate times, actively listening to others, and sustaining conversations. In some social skills training programs, group sessions are augmented with video or technologically based interventions, in vivo community trips to practice social skills (Glynn et al. 2002; Mueser et al. 2010), and involvement of support people who are accessible, pleasant, and knowledgeable about the local environments’ resources and limitations (Mueser et al. 2010; Tauber et al. 2000).

As with other psychosocial interventions, availability of social skills training is a common barrier to its incorporation into treatment. However, information about social skills training is available for organizations that wish to develop such programs (Bellack and Goldberg 2019; Bellack et al. 2004; Granholm et al. 2016).

Balancing of Potential Benefits and Harms in Rating the Strength of the Guideline Statement

Benefits

Social skills training in the treatment of schizophrenia can improve social function, core illness symptoms, and negative symptoms more than usual care can (low strength of research evidence).

Harms

The harms of social skills training in the treatment of schizophrenia have not been well documented but appear to be minimal.

Patient Preferences

Clinical experience suggests that many patients are cooperative with and accepting of social skills training as part of a treatment plan; however, other patients may not wish to take part in social skills training because of logistical barriers (e.g., time, cost, access to transportation, childcare) or having goals for treatment that are unrelated to social skills.

Balancing of Benefits and Harms

The potential benefits of this guideline statement were viewed as likely to outweigh the potential harms. Although the harms appear to be minimal, there is a low strength of research evidence for benefits, and patient preferences may differ in terms of desiring to focus on social skills as a part of treatment. Consequently, this guideline statement was rated as a suggestion. For additional discussion of the research evidence, see Appendix C, Statement 23.

Differences of Opinion Among Writing Group Members

There were no differences of opinion. The writing group voted unanimously in favor of this suggestion.

Review of Available Guidelines From Other Organizations

Other guidelines are generally consistent with this guideline statement. The PORT, RANZCP, CSG, and SIGN guidelines suggest offering social skills training to individuals with schizophrenia who have deficits in social skills (Buchanan et al. 2010; Galletly et al. 2016; Norman et al. 2017; Scottish Intercollegiate Guidelines Network 2013). However, the NICE guideline notes that social skills training should not be routinely offered to individuals with schizophrenia (National Institute for Health and Care Excellence 2014).

Quality Measurement Considerations

As a suggestion, this guideline statement is not appropriate for use as a performance-based quality measure or for incorporation into electronic decision support. Nevertheless, given the potential benefits of such an approach for some patients, health care organizations and health plans may wish to track the availability and utilization of social skills training for individuals with schizophrenia.

Statement 24: Supportive Psychotherapy

APA suggests (2C) that patients with schizophrenia be treated with supportive psychotherapy.*

*This guideline statement should be implemented in the context of a person-centered treatment plan that includes evidence-based nonpharmacological and pharmacological treatments for schizophrenia.

Implementation

Supportive psychotherapy is commonly a part of the treatment plan in individuals with schizophrenia who are not receiving other modes of psychotherapy (e.g., CBTp). Because the evidence related to its benefits is limited, supportive psychotherapy should not take precedence over other evidence-based psychosocial treatments (e.g., CSC, CBTp, psychoeducation). When compared with treatment as usual, no advantage was seen for supportive psychotherapy in terms of global or social function (L. A. Buckley et al. 2015; McDonagh et al. 2017); however, these findings are difficult to interpret given the frequent use of supportive psychotherapy techniques as part of usual care. When compared with insight-oriented psychotherapies, a small number of early studies suggested that supportive psychotherapy might be associated with better outcomes in coping skills, adherence, and relapse (Fenton 2000; Hogarty et al. 1997; Stanton et al. 1984).

The focus of supportive psychotherapy is reality based and present centered (Kates and Rockland 1994; Novalis et al. 1993; Winston 2014; Winston et al. 2012). It commonly aims to help patients cope with symptoms, improve adaptive skills, and enhance self-esteem, although descriptions of the goals of supportive psychotherapy have varied. Examples of techniques used to foster these goals include reassurance; praise; encouragement; explanation; clarification; reframing; guidance; suggestion; and use of a conversational, nonconfrontational style of communication. Many of the common elements that have been identified in effective psychotherapies, including a positive therapeutic alliance, are also integral to supportive psychotherapy (Frank and Frank 1991; Wampold 2015). Typically, supportive psychotherapy is conducted in conjunction with medication management at a frequency that can vary from weekly to every few months depending on the needs of the individual patient. Other psychosocial treatments can also be used as part of the treatment plan in conjunction with these modalities.

Balancing of Potential Benefits and Harms in Rating the Strength of the Guideline Statement

Benefits

Use of supportive psychotherapy in the treatment of schizophrenia was not associated with relative benefits in global or social function as compared with treatment as usual (low strength of research evidence). However, treatment as usual already incorporates supportive psychotherapy under most circumstances. In addition, clinical experience suggests that supportive psychotherapy may be associated with such benefits as strengthening the therapeutic alliance, reducing demoralization, and developing practical coping strategies in the treatment of individuals with schizophrenia.

Harms

The harms of using supportive psychotherapy in the treatment of schizophrenia appear to be small, although evidence is limited. However, if supportive psychotherapy is used preferentially instead of a treatment that is associated with more robust evidence of benefit, there may be indirect negative effects.

Patient Preferences

Clinical experience suggests that most patients are cooperative with and accepting of supportive psychotherapy as part of a treatment plan, even when they are reluctant to engage in other psychosocial interventions. However, some patients may not wish to engage in psychotherapy or may have logistical barriers (e.g., time, access to transportation, financial considerations) that make it difficult to attend psychotherapy sessions.

Balancing of Benefits and Harms

The potential benefits of this guideline statement were viewed as likely to outweigh the potential harms. In clinical practice, the use of supportive psychotherapy is commonplace as part of the treatment of schizophrenia, which makes it challenging to interpret the research comparisons of supportive psychotherapy versus treatment as usual. Given the limited evidence of any harms of supportive psychotherapy, the potential benefits of supportive psychotherapy appear to be greater than the harms. For additional discussion of the research evidence, see Appendix C, Statement 24.

Differences of Opinion Among Writing Group Members

There were no differences of opinion. The writing group voted unanimously in favor of this suggestion.

Review of Available Guidelines From Other Organizations

The NICE guideline notes that supportive psychotherapy should not be offered routinely to individuals with schizophrenia if other psychosocial treatments that have greater efficacy are available (National Institute for Health and Care Excellence 2014). However, the NICE guideline also notes that patient preferences should be taken into account, particularly if other psychosocial interventions are not available locally.

Quality Measurement Considerations

As a suggestion, this guideline statement is not appropriate for use as a performance-based quality measure or for incorporation into electronic decision support.