This article is freely available to all

Article Abstract

Despite treatment advances over the past decades, schizophrenia remains one of the most severe psychiatric disorders that is associated with a chronic relapsing course and marked functional impairment in a substantial proportion of patients. In this article, a historical overview of the pharmacologic advances in the treatment of schizophrenia over the past 50 years is presented. This is followed by a review of the current developments in optimizing the treatment and outcomes in patients with schizophrenia. Methodological challenges, potential solutions, and areas of particular need for further research are highlighted. Although treatment goals of response, remission, and recovery have been defined more uniformly, a good "effectiveness" measure mapping onto functional outcomes is still lacking. Moreover, the field must advance in transferring measurement-based approaches from research to clinical practice. There is an ongoing debate regarding whether and which first- or second-generation antipsychotics should be used. However, especially when considering individual adverse effect profiles, the differentiation into first- and second-generation antipsychotics as unified classes cannot be upheld, and a more differentiated view and treatment selection are required. The desired, individualized treatment approach needs to consider current symptoms, comorbid conditions, past therapeutic response, and adverse effects, as well as patient choice and expectations. Acute and long-term goals and effects of medication treatment should be balanced. To date, clozapine is the only evidence-based treatment for refractory patients, and the role of antipsychotic polypharmacy and other augmentation strategies remains unclear, at best. To discover novel treatments with enhanced/broader efficacy and improved tolerability, and to enable personalized treatment, the mechanisms underlying illness development and progression, symptomatic improvement, and side effect development need to be elucidated.

J Clin Psychiatry 2010;71(9):1115-1124

Submitted: May 20, 2010; accepted July 9, 2010(doi:10.4088/JCP.10r06264yel).

Corresponding author: John M. Kane, MD, Zucker Hillside Hospital, 75-59 263rd St, Glen Oaks, NY 11004 ([email protected]).

Past and Present Progress in the Pharmacologic Treatment of Schizophrenia

Despite treatment advances over the past decades, schizophrenia remains one of the most severe psychiatric disorders that is associated with a chronic relapsing course and marked functional impairment in a substantial proportion of patients. In this article, a historical overview of the pharmacologic advances in the treatment of schizophrenia over the past 50 years is presented. This is followed by a review of the current developments in optimizing the treatment and outcomes in patients with schizophrenia. Methodological challenges, potential solutions, and areas of particular need for further research are highlighted. Although treatment goals of response, remission, and recovery have been defined more uniformly, a good "effectiveness" measure mapping onto functional outcomes is still lacking. Moreover, the field must advance in transferring measurement-based approaches from research to clinical practice. There is an ongoing debate regarding whether and which first- or second-generation antipsychotics should be used. However, especially when considering individual adverse effect profiles, the differentiation into first- and second-generation antipsychotics as unified classes cannot be upheld, and a more differentiated view and treatment selection are required. The desired, individualized treatment approach needs to consider current symptoms, comorbid conditions, past therapeutic response, and adverse effects, as well as patient choice and expectations. Acute and long-term goals and effects of medication treatment should be balanced. To date, clozapine is the only evidence-based treatment for refractory patients, and the role of antipsychotic polypharmacy and other augmentation strategies remains unclear, at best. To discover novel treatments with enhanced/broader efficacy and improved tolerability, and to enable personalized treatment, the mechanisms underlying illness development and progression, symptomatic improvement, and side effect development need to be elucidated.

J Clin Psychiatry 2010;71(9):1115-1124

Submitted: May 20, 2010; accepted July 9, 2010 (doi:10.4088/JCP.10r06264yel).

Corresponding author: John M. Kane, MD, Zucker Hillside Hospital, 75-59 263rd St, Glen Oaks, NY 11004 ([email protected]).

This article is part of a series honoring the 50th anniversary of the NCDEU Annual Meeting. This meeting has fostered, facilitated, documented, and disseminated a vast growth in our knowledge of clinical psychopharmacology and our ability to apply that knowledge to improving the lives of millions of people. We will try to put some of the historical development in the context of present challenges and future needs and opportunities.

EARLY BEGINNINGS AND THE ROLE OF THE EARLY CLINICAL DRUG EVALUATION UNITS

When the first meeting of the Early Clinical Drug Evaluation Units (later, the New Clinical Drug Evaluation Unit [NCDEU]) took place in 1959, it had been approximately 5 years since the introduction of chlorpromazine in the United States, and clinical trial methodology was in its formative stages.

In 1952, a French surgeon was exploring strategies to reduce surgical shock. He thought that antihistamines might be an effective approach. He noticed, however, that an antihistamine that he was using, chlorpromazine, had a powerful effect on mental state. A psychiatrist, Pierre Deniker, heard about these observations and decided to try chlorpromazine in some of his most difficult-to-manage patients. The results were remarkable. After some reluctance on the part of academic psychiatrists and psychologists in the United States to support testing of the drug, its value was demonstrated among patients in state institutions, and ultimately chlorpromazine was approved by the US Food and Drug Administration (FDA) in 1954. By 1964, approximately 50 million people around the world had been treated with this medication, and a revolution in the management of psychotic disorders was underway.

The somewhat serendipitous observation that chlorpromazine had pronounced "calming" activity even in individuals with psychotic signs and symptoms was one of the great advances in 20th century medicine. Although other drugs (eg, reserpine) had been used with some success to treat psychosis,1 the safety index and overall effectiveness of chlorpromazine, and subsequently other dopamine receptor antagonists, radically changed our ability to treat schizophrenia and other psychotic disorders on a wide scale.

The pace of new discoveries regarding effective psychotropic medications in the 1950s and 1960s was staggering. At the same time, tension remained between the psychodynamic and biologic perspectives regarding the etiology and treatment of the major psychiatric illnesses. Considerable efforts were made to study the impact of psychotropic drugs, and increasingly sophisticated methodologies were brought to bear as clinical trials in medicine underwent rapid development.

In 1949, the World Health Organization published the sixth revision of the International Statistical Classification of Diseases (ICD), which for the first time included a section on mental disorders.2 The first official Diagnostic and Statistical Manual of Mental Disorders was published in 1952 by the American Psychiatric Association (APA).3 Diagnostic criteria were not really specified for discrete disorders until the third edition of DSM,4 which attempted to improve the validity and reliability of psychiatric diagnosis. This, in turn, had enormous implications for clinical practice, clinical research, and drug development.

The ECDEU, which were established by the National Institute of Mental Health (NIMH), served as unique platforms for clinical investigation. They were designed to provide stable funding for investigators studying new drugs. The Psychopharmacology Research Branch, which provided funding and guidance for these units, played a critical role in the advancement of the field.

An example of a seminal contribution by Jerry Levine, William Petrie, and Nina Schooler of NIMH, with colleagues from the Biometric Laboratory at George Washington University, was the first publication of the ECDEU Assessment Manual for Psychopharmacology in 1976.5 The development and testing of assessment instruments that could be demonstrated to be both valid and reliable for the measurement of therapeutic effects on a variety of disease categories was a major advance.

A New Era of Evidence-Based Psychopharmacology and the Role of the NIMH Psychopharmcology Service Center

In 1969, Donald Klein and John Davis published a seminal work entitled Diagnosis and Drug Treatment of Psychiatric Disorders.6 In the introduction, they wrote,

This simple dichotomy between medical and nonmedical practitioners does less than justice to the complicated therapeutic scene. The medical practitioners are divided largely into 2 polar camps: the analytical and psychological versus the organic and directive. The first group developed an ideology that rejects the use of organic treatments and directive methods as usually ineffective, symptomatic at best, and destructive of the growth potential of the patient by fostering pathological dependence. This stand was reinforced by the obvious ineffectiveness of most organic therapies, complicated by the addictive potential and social incapacitation often produced by sedative agents. The directive and organic group, on the other hand, emphasized short-term manipulative and symptom-relieving procedures, deriding or ignoring concern with the resolution of intrapsychic conflict and patient maturity. Unfortunately, the positive contributions of both groups were obscured by their respective biophobic and psychophobic attitudes. One might speculate that the fierce adherence of each group to its methods in the face of the remarkable lack of systematic comparative studies attests to a profound insecurity as to the value of one’s procedures, dealt with by a compensatory evangelism.

We may be fortunate to be entering a period in which rational comparative study will become standard for therapeutic decision. Although clinical hunches and results of clinical experience are important factors in the termination of proper treatment, the findings of research studies, particularly those which are done with controlled double-blind technique, provide the behavioral scientific data for informed decision. Also important is the evidence available on the interaction of the somatic therapies with other treatment forms, such as psychological and social therapies. This book is an initial effort to organize and present such material to the psychiatric practitioner for his critical review.

With the establishment of The National Institute of Mental Health Psychopharmacology Service Center, a series of cooperative studies led by Jonathan Cole was conducted.7 They included both private and public hospitals and initially compared chlorpromazine, fluphenazine, and thioridazine with placebo. All 3 drugs were found to be equally effective and more efficacious than placebo. A second NIMH Cooperative Study8 compared chlorpromazine, acetophenazine, and fluphenazine. No specific drug showed a consistent pattern of superiority across the 57 dependent variables that were assessed.

By 1969, when Klein and Davis published their review,6 they identified 126 controlled studies comparing antipsychotic drugs and placebo in which the medications were found to be more effective and 26 comparisons in which they were not. The authors also examined the role of dose adequacy and found that most of those studies that found chlorpromazine to be ineffective used very small doses, and all 23 studies that employed doses over 500 mg were positive. Similarly, in all studies, which were judged to be methodologically rigorous, the phenothiazine derivatives (and reserpine) were shown to be more effective than controls.

NEUROMOTOR SIDE EFFECT CONCERN

Shortly after the introduction of the phenothiazines, concerns about adverse neurologic effects—first "parkinsonism" and subsequently tardive dyskinesia—took on considerable saliency. Theories as to minimum effective dosage utilized subtle parkinsonism as a measure of adequate dosing.9 However, both the frequency and potential functional consequences (including attendant stigma) associated with adverse neurologic effects became an important focus of attention.10-12 Given the frequency of extrapyramidal symptoms (EPS) and likelihood of underdiagnosis,13-15 debate ensued as to whether the use of prophylactic antiparkinsonian medication should be routinely recommended. At the same time, antiparkinsonian agents were associated with their own burden of adverse effects.

In the 1980s, the concern about tardive dyskinesia became even more intense with increasing medicolegal issues and the publication of 2 APA Task Force Reports.16,17 Ultimately, prospective studies began to clarify both the incidence of and risk factors for tardive dyskinesia. The incidence was generally found to be 5% per year of cumulative antipsychotic drug exposure with first-generation antipsychotics.18,19 Increasing age, particularly the postmenopausal phase in women, was associated with higher risk, as was vulnerability to early occurring extrapyramidal side effects.20

ANTIPSYCHOTIC DOSE FINDING AND BLOOD LEVELS IN FLUX

Phases in dosage recommendations also ensued over the coming decades with trials of very high doses21 and trials of very low doses,22 with, as usual, mixed results. In general, however, once blood levels of psychotropic drugs became widely available, it became apparent that very high doses provided no added value for the average patient and that measuring blood drug levels might help to some degree in explaining the heterogeneity of response.23 Measurement of blood drug levels never really caught on in routine clinical practice, and even now they play much less of a role in research than they did in the 1980s (for reasons that are not entirely clear). The identification of dopamine as a key neurotransmitter in the mechanism of action of antipsychotic drugs and the discovery of various dopamine receptors in specific brain regions led to renewed enthusiasm about finding more "rational" pharmacologic agents and again setting the stage for further progress in understanding dosage requirements and heterogeneity of response. A number of studies emphasized the feasibility of utilizing substantially lower doses in the maintenance phase of treatment than had been commonly employed.24,25 Interestingly, the most informative studies examining dose-response relationships in maintenance treatment and relapse prevention utilized long-acting injectable formulations. This was particularly important in eliminating the potential confound of poor or partial adherence with dosing requirements.22 Although these studies emphasized the feasibility of utilizing lower than customary doses, they also established thresholds below which relapse rates increased substantially.

Concerns regarding dose-response (and dose-tolerability) relationships were also an important focus in evaluating comparative data between first- and second-generation antipsychotics. Although some reviews and meta-analyses had suggested that some of the apparent superiority of second- versus first-generation antipsychotics was due to unnecessarily high dosages of first-generation medications,26 other reviews have not supported this conclusion.27,28 It has been shown that even low doses of haloperidol, ie, 4 mg/d, in the acute treatment of chronic patients are associated with a significant risk of EPS.29 Moreover, in 2 recent first-episode studies, haloperidol treatment of 3 mg/d was associated with significantly greater relapse rates30 or all-cause discontinuation rates31 than the second-generation comparators.

MAINTENANCE TREATMENT AND RELAPSE PREVENTION

In the late 1950s,32,33 investigators began to systematically examine the consequences of controlled phenothiazine withdrawal. It became increasingly apparent that patients receiving placebo experienced significantly higher rates of rehospitalization than patients continuing to receive medication.34

By 1969, Klein and Davis were already recommending that "all patients who have an acute schizophrenic psychosis should be maintained on phenothiazine, possibly with an adjunctive antidepressant, indefinitely."6(p160) However, others did not share this view, and it took many years to establish a consensus as to the need for maintenance treatment, particularly in the early phases of a schizophrenia illness.

The increasing concern about tardive dyskinesia in the 1980s led to a reevaluation of the overall benefit-to-risk ratio of maintenance or relapse prevention treatment. There were renewed efforts to establish minimum effective dosage and/or the value of "intermittent" or "targeted" treatment, all of which were intended to reduce cumulative medication exposure, with the hope of reducing the incidence of tardive dyskinesia. These results helped to further clarify the need for continuous maintenance treatment for the average patient and confirmed a threshold of drug activity that was necessary to prevent or delay relapse.35

ADDRESSING NONADHERENCE

In the 1970s, long-acting injectable fluphenazine enanthate and fluphenazine decanoate were approved. Fluphenazine decanoate ultimately became the more widely used agent because of better tolerability.36 This provided a strategy to help patients overcome the challenges of consistent medication-taking in the face of a complex illness often resulting in poor insight and impaired cognitive functioning.37,38 Despite the promise of this approach, the use of long-acting injectable medications never became as popular in the United States as it did in many other countries. However, the current availability of more and newer agents in long-acting formulations39 in combination with ever increasing needs to control the costs associated with relapse and rehospitalization might yet impact utilization rates.

TREATMENT-REFRACTORY ILLNESS AND CLOZAPINE

With the development and testin g of clozapine in Europe, early observations suggested a novel compound had been developed with a qualitatively different clinical profile. Most clinicians were impressed with the relative absence of drug-induced extrapyramidal effects, although some debate arose as to the incidence of akathisia. In addition, early observations indicated the potential of clozapine to have some therapeutic benefit among patients who had failed to respond to other agents. At the same time, a series of cases of agranulocytosis associated with clozapine were reported in Finland and elsewhere.40 This led to a delay in the further development of clozapine in the United States. However, once a large clinical trial was conducted demonstrating the clear superiority of clozapine over chlorpromazine in treatment-refractory schizophrenia patients,41 the benefit-to-risk ratio (with the requirement for weekly blood monitoring) was felt to be sufficient to justify FDA approval with the narrow indication for treatment-resistant patients in 1990. Since then, the singular role of clozapine in treatment-refractory patients with schizophrenia has been confirmed.42

To some extent, clozapine served as a prototype and a stimulus for the development of other new drugs with the receptor-binding profiles that might replicate clozapine’s unique clinical attributes. This created a number of challenges, particularly in various domains of drug development as well as in clinical design and methodology. It might be said that a major focus of work in the past decade has been to clarify the extent to which any, some, or all of the second-generation (sometimes referred to as atypical, a term that we believe has outlived its usefulness) medications are superior to any, some, or all of the first-generation antipsychotics.

In this context, a reemphasis on the study of dose-response relationships and dose equivalency between drugs has occurred,43 as did a partial reevaluation of the public health importance of drug-induced parkinsonism and tardive dyskinesia.44

ATTENTION TO FIRST-EPISODE SCHIZOPHRENIA

Beginning in the mid 1980s, the field started to focus on patients with a first episode of schizophrenia.45 The increased attention on first-episode patients seemed warranted in order to evaluate treatment outcomes that were not confounded by the effects of prior treatment, multiple relapses, and chronic illness. Studies revealed cognitive and psychosocial deficits that were present at illness onset,46 a long duration of untreated psychosis prior to first mental health contact and treatment,47 and increased sensitivity to medication side effects,48 but also a better treatment response compared to more chronically ill patients.49 These results, representing a mixture of putative pathophysiologic processes and environmental effects, were greeted with efforts to shorten the duration of untreated psychosis through outreach, which has been associated with some degree of improved outcomes.50

However, despite interventions during the first episode of schizophrenia, the overwhelming majority of patients was found to relapse in the subsequent years,51 with medication discontinuation significantly increasing risk, and the achievement of at least 2 years of concurrent symptomatic and psychosocial recovery has remained as low as 15%.52 The documented low recovery rates revitalized efforts at testing an integrated, personalized, and evidence-based psychopharmacologic and psychosocial intervention package against treatment as usual in first-episode patients in 2 parallel NIMH-funded Recovery After an Initial Schizophrenia Episode (RAISE) projects (http://www.nimh.nih.gov/health/schizophrenia/raise/index.shtml) to evaluate if the functional outcome trajectory can be modified early in the illness phase. In addition, as part of the move toward the early treatment of schizophrenia, and the response to new FDA incentives, the efficacy of antipsychotics has also been tested and validated in recent years in a series of placebo-controlled studies in adolescents with schizophrenia.53

THE PRODROME TO SCHIZOPHRENIA: EARLY RECOGNITION AND PREVENTION EFFORTS

Stimulated by first-episode research and the related recognition of a symptomatic, "prodromal" phase preceding the first full psychotic episode, early identification and intervention even during the prepsychotic illness phase became an area of increasing research attention beginning in the 1990s. The development of specific assessment tools and delineation of criteria for individuals considered at clinical high risk for psychosis54,55 were followed by the examination of conversion rates in at-risk cohorts followed naturalistically. However, despite initially encouraging results concerning the predictive validity of the psychosis risk syndrome criteria, recent studies have reported declining conversion rates,56 highlighting the need for further investigations. Throughout a series of mostly small, randomized, controlled studies, several pharmacologic and nonpharmacologic interventions, involving omega-3 fatty acids, second-generation antipsychotics, and cognitive-behavioral therapy, have been found to delay or prevent the onset of psychosis, at least as long as the active treatment was provided.57 However, recent discussions of potentially including the psychosis risk syndrome in DSM-V have been met with criticism for fear of a high rate of false-positives; an overmedicalization of ill-defined and nonspecific psychopathology; insufficient time and training in clinical settings to utilize complex, research-based instruments for the identification of high-risk individuals; and the resultant risk of stigma and the unnecessary use of treatments with a potential for significant long-term side effects.57

COMPARATIVE EFFICACY AND EFFECTIVENESS OF FIRST-GENERATION AND SECOND-GENERATION ANTIPSYCHOTICS

With the introduction of second-generation antipsychotics, findings of lower EPS burden and tardive dyskinesia risk were coupled with expectations of superior efficacy for positive, negative, and cognitive symptoms. Initial efficacy studies seemed to confirm the superiority of second-generation antipsychotics, but the comparator consisted predominantly of haloperidol, used at moderate to high doses and often without anticholinergic cotreatment, which made early treatment discontinuation and secondary negative symptom presentations more likely in haloperidol-treated patients. Since then, a series of acute phase and longer-term studies have been completed, including large efficacy-effectiveness hybrid trials31,58-60 that compared first- and second-generation antipsychotics. These data have been evaluated and interpreted in a number of different ways. Interpretations include that there is no difference between first- and second-generation antipsychotics, that second-generation antipsychotics are superior to first-generation antipsychotics, that some second-generation antipsychotics are superior to either all or some first-generation antipsychotics, in general, or in certain efficacy and/or side effect domains, or in patient subgroups that are not yet easily identified prior to choosing a specific agent. Because such a number of divergent interpretations have been offered, this indicates that blanket statements do not do justice to the complex clinical situation and database.

Taken together, the evidence seems to suggest that in refractory patients, clozapine is superior to first-generation antipsychotics61-63 and second-generation antipsychotics (although the latter was hardly confirmed by a recent meta-analysis,64 which was attributed to inappropriately low clozapine doses). Compared to first-generation antipsychotics, only 3 second-generation antipsychotics (amisulpride, olanzapine, and risperidone) were superior based on Positive and Negative Syndrome Scale (PANSS) score change differences,61 but these were also the medications studied at a time when first-generation antipsychotics predominated, whereas the newer second-generation antipsychotics were tested mostly at a time of predominant second-generation antipsychotic use. While this could have introduced a cohort sampling bias, the differences between nonclozapine antipsychotics were very modest, with effect sizes as low as 0.1 to 0.3. Similarly, differences between second-generation antipsychotics studied head-to-head were either nonexistent or also marginal, favoring in some comparisons risperidone (vs quetiapine and ziprasidone) or olanzapine (vs aripiprazole, quetiapine, risperidone, and ziprasidone), with the same low effect size difference of only 0.1 to 0.3.59-61 Moreover, the differences between second-generation antipsychotics were even more restricted when not analyzing mean total PANSS score differences, but analyzing discontinuation of medication due to inefficacy.61 Thus, differences in design, including active or placebo control, dosing, and sponsorship,65-67 may have a greater impact on efficacy outcomes than the actual choice of nonclozapine antipsychotics.

The CATIE [Clinical Antipsychotic Trials of Intervention Effectiveness] and CUtLASS [Cost Utility of the Latest Antipsychotic Drugs in Schizophrenia Study] studies seemed to suggest that there are generally no differences between second-generation antipsychotics and first-generation antipsychotics in all-cause discontinuation, especially when analyzing quality of life60 and patients who had switched antipsychotics.68 However, these conclusions have been challenged on the basis of insufficient sample sizes to make noninferiority claims.69 Moreover, in first-episode samples, all-cause discontinuation rates and relapse rates were significantly higher at 1 and 2 years, respectively, with modestly dosed haloperidol compared to the respective second-generation antipsychotic comparators.31 Even in chronically ill samples, relapse rates were also significantly higher in first-generation antipsychotics, although haloperidol doses were higher than currently recommended.70

The clinical effectiveness of first-generation antipsychotics, a measure of objective and subjective outcomes encompassing symptom-based and functional effects, is challenged by increased acute71 and chronic72 extrapyramidal side effects and related symptoms of dysphoria, compared to second-generation (atypical) antipsychotics. Even though at chlorpromazine equivalents below 600 mg/d there was no increased EPS rate with typical versus atypical antipsychotics, at those doses, the efficacy of second-generation antipsychotics was superior.27 Furthermore, while masking of EPS can be achieved with prophylactic anticholinergic treatment,58 the risk of tardive dyskinesia is not reduced, but rather potentially increased,72 and recent data suggest that anticholinergic medication load is associated with decreased efficacy of cognitive remediation treatment.73

Ultimately, we feel that the controversy regarding the most likely oversimplified dichotomy between first- and second-generation antipsychotics has resulted in progress, in that it stimulated the conduct of large trials and examination of effectiveness outcomes beyond symptom reduction. These trials have generated new and important data, but also highlighted methodological challenges. These challenges include the definition of clinically meaningful endpoints, the effect of baseline medication and past treatment history, the limitation of available treatments used at a more chronic illness phase, and the importance of differences in acute and long-term adverse effects. All of these data point to the need for new treatments with novel mechanisms, tailored approaches that map onto the pathophysiology of the disease process (that may vary between patients and between different illness stages), and biologic dissection of patients into meaningful subgroups that can inform a stratified or, even, individualized treatment selection.

SHIFTING ADVERSE EVENT FOCUS TO PHYSICAL HEALTH

Over the last decade and coinciding with the predominant use of second-generation antipsychotics, there has been a shift in side effect concerns from parkinsonism and tardive dyskinesia to physical health risks and outcomes.74-76 The relevance of antipsychotic-related weight gain was highlighted by data suggesting that severely mentally ill patients die on average 25 years earlier than the general population, and that this is predominantly due to premature cardiovascular and cerebrovascular mortality,77 both of which are related to weight gain, obesity, and associated metabolic abnormalities. Reasons for the increased prevalence of the cardiovascular risk factors, morbidity, and mortality in the mentally ill are complex, but include effects of the psychiatric illness and poor lifestyle behaviors, but also weight gain and metabolic abnormalities conferred by psychiatric treatments, particularly second-generation antipsychotics. For a while, the discussion seemed to focus on having to decide between a higher risk for EPS and tardive dyskinesia with first-generation antipsychotics and a greater risk for weight gain and long-term cardiometabolic consequences with second-generation antipsychotics. Increasingly, however, the field has been moving to a more differentiated view, recognizing that neither second- nor first-generation antipsychotics are homogeneous classes regarding adverse effect risk. Thus, although clozapine and olanzapine, 2 second-generation antipsychotics, are among the most weight gain-producing and metabolically problematic antipsychotics, the low-potency first-generation antipsychotic chlorpromazine is also associated with considerable adverse cardiometabolic effects.78 Furthermore, high- and mid-potency first-generation antipsychotics most likely have a similar cardiometabolic risk potential as low-risk second-generation antipsychotics, such as aripiprazole and ziprasidone, yet, in treatment-naive and first-episode patients, all antipsychotics, even those considered more neutral in chronic patients, are associated with considerable weight gain.79-81

As a result of the antipsychotic-related cardiometabolic effects, the traditional role of psychiatrists as health care providers who have little to do with the somatic well being of their patients has been challenged. The redefinition of the psychotropic medication prescriber and psychiatric health care provider as at least an orchestrator/facilitator of integrated medical care and as the focal point of health care monitoring in patients receiving medications with cardiometabolic impact is still in process.82 Despite the warning of the FDA in 2003 about the diabetes risk associated with antipsychotics, which was shortly followed by monitoring guidelines for weight, blood pressure, and fasting glucose and lipids in antipsychotic-treated patients,83 a series of recent database and audit studies confirmed a concerning low rate of metabolic monitoring that in one study was similar to the background monitoring in a nonpsychiatric control population prescribed albuterol.84,85 In addition to insufficient monitoring, several studies have shown that mentally ill patients receive substandard medical care targeting coronary heart disease risk factors in psychiatric settings86-88 and addressing diabetes or myocardial infarction in medical settings.89,90 While patient nonadherence with medical appointments and interventions might contribute to this problem, the field needs to effectively address the suboptimal monitoring and management behaviors of mental and medical health care providers, as well as systems issues of fragmented care and poor access to care.

RAISING THE BAR FOR OUTCOMES

In addition to a broadened focus on physical health, outcomes other than symptomatic improvement have become standard in the field. These include more standardized approaches to measuring response, remission, and recovery.91-94 In addition, subjective well-being95,96 and quality of life,54 cognition,97-99 and psychosocial performance, including employment,100-102 have become endpoints of interest and goals for patients, families, clinicians, and researchers. To move toward these important goals, it has become clear that the field needs to study and engage in the routine application of measurement-based psychiatry, clinical and shared decision-making, psychoeducation, and adherence management, as well as in the integration of rational psychosocial treatment elements in the often too one-sided pharmacologic treatment planning.103

TARGETING INDIVIDUALIZED TREATMENT

Individualized treatment based on reliable probabilities of outcome for a specific patient is an important treatment goal. Unfortunately, this goal is still largely out of reach, due to the heterogeneity of patients and treatment response, most likely related to mostly unknown genetic, structural, and functional physiologic differences between patients and within patients over time. Efforts at increasing the predictability of outcomes have included clinically driven nosologic and phenomenological approaches, but these have not really succeeded. Current approaches that do not yet have clinical applicability include the use of genetics, neuroimaging, neurocognition, and blood- or tissue-based biomarkers and sets of biomarkers, also called biosignatures.104,105 Similarly, developments are underway to define biomarkers as surrogate endpoints in drug development and approval.106

However, there is a powerful clinical tool that uses the patients’ own response pattern to predict outcomes. This intraindividual test of early response/nonresponse as a predictor of subsequent response107,108 or of dysphoric response109 was studied briefly in the 1980s. As much as 15 to 20 years later, these findings have been revisited and expanded upon, stimulated by analyses showing that, at least at a group level, the majority of antipsychotic response occurs within the first few weeks110,111 and, even, days112 after antipsychotic initiation. Building on these findings, a series of post hoc analyses113-117 plus a recent prospective study118 showed that nonresponse at study endpoint can be predicted with high sensitivity, specificity, and predictive power by presence of less than a minimal response equivalent to less than 20% reduction in the PANSS119 or Brief Psychiatric Rating Scale120 total score at 2 weeks after antipsychotic initiation. However, having identified this general response pattern, questions remain as to whether response patterns are similar in likely more heterogeneous first-episode schizophrenia samples and in treatment-refractory patients, whether a limited set of specific symptom items that could be used in clinical practice is equally valid and reliable, what one can learn from symptom trajectories at an individual patient level, and what alternative treatments are likely to be more successful after early nonresponse has been identified.

CHALLENGES, UNMET NEEDS, AND FUTURE DIRECTIONS

A number of unmet needs and challenges exist in schizophrenia. These include methodological and practical problems, such as the decreasing ability to separate medication effects from placebo, with resultant high rates of "failed" trials and/or the need to increase sample sizes. Unmet needs also include areas of psychopathology that are insufficiently impacted with currently available treatments, such as negative symptoms and cognitive dysfunction, as well as problems with adherence to treatment guidelines and the adoption of best clinical practices, for example by the routine adoption of measurement-based treatment strategies. More work is also needed regarding the conduct of sufficiently large or long-term comparative effectiveness studies; the identification of simple, meaningful, and measurable effectiveness outcome measures; and the best ways to translate evidence into clinical practice. All of these areas seem to be amenable to incremental steps of improvement.

However, to qualitatively change outcomes in schizophrenia, there is a need for the detection of valid biomarkers and biosignatures that map onto the underlying pathophysiology of the disease. In this context, the discovery of mechanisms and predictors of efficacy and tolerability is required to guide the rational treatment selection. Our increasing technological sophistication makes biomarker studies more feasible in an age when clinical classification might be replaced by genomic, proteomic, or metabolomic approaches, to name but a few. The resultant developments are expected to greatly facilitate the much needed discovery of mechanistically novel treatments that either work in a complementary way, enabling also rational combination treatments, or are particularly effective for specific symptom domains and readily separable subgroups of patients. The resultant new treatments will hopefully speed up or increase the magnitude of symptom reduction across all relevant domains of schizophrenia, enhance relapse prevention, and bolster efficacy for nonresponders and currently refractory patients, while reducing the likelihood of developing key adverse effects. Finally, the primary or secondary prevention of psychosis is an important goal that will depend, in part, on uncovering mechanisms underlying the susceptibility for and progression toward psychosis, so that neuroprotective and low-risk agents can be investigated in samples that can be characterized as being at true risk for psychosis in a highly reliable way.49

To discuss the specific agents under development for these various treatment targets is beyond the scope of this review, but compounds are being explored with a variety of putative mechanisms of action. These include metabotropic glutamate agonists, α-nicotinic receptor agonists, muscarinic agonists, histamine-3 receptor antagonists, glycine transporter inhibitors, ampakines, phosphodiesterase-10 inhibitors, D1 agonists, D3 antagonists, 5-HT2A antagonists, and partial dopamine agonists, among others.121-124

SUMMARY AND CONCLUSION

In summary, building on more than 5 decades of pharmacotherapy research and clinical practice in schizophrenia, in which the ECDEU and NCDEU played a major role, the field has finally entered a phase that promises to develop and test the necessary tools that will enable more targeted and, ultimately, individualized treatment approaches. The hope is that a more detailed mechanistic understanding of the factors involved in the development, progression, and amelioration of the disease process will give rise to a number of new treatment approaches and that the focus will shift from symptomatic to disease-modifying and, ideally, curative interventions. Being in the midst of these developments, it is important to realize how far we have come, what role the prior advancements have played in our current state of knowledge, and what still needs to be accomplished to further improve the outcome of patients suffering from schizophrenia.

Drug names: albuterol (Proventil and others), aripiprazole (Abilify), clozapine (Clozaril, FazaClo, and others), olanzapine (Zyprexa), quetiapine (Seroquel), reserpine (Serpalan and others), risperidone (Risperdal and others), ziprasidone (Geodon).

Author affiliations: The Zucker Hillside Hospital, North Shore—Long Island Jewish Health System, Glen Oaks, New York; and Albert Einstein College of Medicine, Bronx, New York.

Potential conflicts of interest: Dr Kane has been a consultant to AstraZeneca, Boehringer Ingelheim, Cephalon, Dainippon Sumitomo, Eisai, Lundbeck, Johnson & Johnson, Merck, Myriad, Novartis, Wyeth, Janssen, Pfizer, Eli Lilly, Bristol-Myers Squibb, Otsuka, Vanda, Proteus, Takeda, Targacept, Intracellular Therapeutics, and Rules Based Medicine; has received honoraria for lectures from Otsuka, Eli Lilly, Bristol-Myers Squibb, and Janssen; and is a stock shareholder in MedAvante. Dr Correll has been a consultant and/or advisor to Actelion, AstraZeneca, Bristol-Myers Squibb, Cephalon, Eli Lilly, Janssen/Johnson & Johnson, GlaxoSmithKline, Hoffmann-La Roche, Medicure, Otsuka, Pfizer, Schering-Plough, Supernus, Takeda, and Vanda and has received honoraria from Boehringer-Ingelheim, GlaxoSmithKline, Lundbeck, Ortho-McNeil, and Janssen.

Funding/support: Supported by the Zucker Hillside Hospital Advanced Center for Intervention and Services Research for the Study of Schizophrenia (MH074543-01) from the National Institute of Mental Health, Bethesda, Maryland.

REFERENCES

1. Armitage SG, Greenberg PD, Olsen AL, et al. The effects of reserpine on schizophrenic patients. AMA Arch Neurol Psychiatry. 1956;76(2):198-204. PubMed

2. World Health Organization. International Statistical Classification of Diseases, Injuries, and Causes of Death. Sixth Revision. Geneva, Switzerland: World Health Organization; 1949.

3. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders. Washington, DC: American Psychiatric Association; 1952.

4. American Psychiatric Association. Committee on Nomenclature and Statistics. Diagnostic and Statistical Manual of Mental Disorders, Third Edition. Washington, DC: American Psychiatric Association; 1980.

5. Guy W. ECDEU Assessment Manual for Psychopharmacology. Department of Health, Education, and Welfare publication (ADM) 76-388. Rockville, MD: National Institute of Mental Health; 1976.

6. Klein DF, Davis JM. Diagnosis and Drug Treatment of Psychiatric Disorders. Baltimore, MD: The Williams & Wilkins Company; 1969.

7. Guttmacher MS. Phenothiazine treatment in acute schizophrenia; effectiveness: National Institute of Mental Health Psychopharmacology Service Center Collaborative Study Group. Arch Gen Psychiatry. 1964;10(3):246-261. PubMed

8. Clinical effects of three phenothiazines in "acute" schizophrenia. Publication Rep No 6. Bethesda, MD: US Public Health Service; 1966.

9. Haase HJ. Occurrence and interpretation of psychomotor parkinsonism in megaphen or largactil prolonged therapy. Nervenarzt. 1954;25(12):486-492. PubMed

10. Van Putten T. Why do schizophrenic patients refuse to take their drugs? Arch Gen Psychiatry. 1974;31(1):67-72. PubMed

11. Van Putten T, Mutalipassi LR, Malkin MD. Phenothiazine-induced decompensation. Arch Gen Psychiatry. 1974;30(1):102-105. PubMed

12. Van Putten T. The many faces of akathisia. Compr Psychiatry. 1975;16(1):43-47.PubMed doi:10.1016/0010-440X(75)90019-X

13. Rifkin A, Quitkin F, Klein DF. Akinesia: a poorly recognized drug-induced extrapyramidal behavioral disorder. Arch Gen Psychiatry. 1975;32(5):672-674.

14. Rifkin A. The risks of long-term neuroleptic treatment of schizophrenia: especially depression and akinesia. Acta Psychiatr Scand Suppl. 1981;63(S291):129-136.PubMed doi:10.1111/j.1600-0447.1981.tb02465.x

15. Rifkin A, Quitkin F, Kane JM, et al. Are prophylactic antiparkinson drugs necessary? a controlled study of procyclidine withdrawal. Arch Gen Psychiatry. 1978;35(4):483-489. PubMed

16. Baldessarini RJ, Cole JO, Davis JM, et al. Tardive dyskinesia: summary of a task force report of the American Psychiatric Association. Am J Psychiatry. 1980;137(10):1163-1172.

17. Kane JM, Jeste DV, Barnes TRE, et al. American Psychiatric Association Task Force Report on Tardive Dyskinesia. Washington, DC: American Psychiatric Press; 1992.

18. Kane JM, Woerner M, Weinhold P, et al. A prospective study of tardive dyskinesia development: preliminary results. J Clin Psychopharmacol. 1982;2(5):345-349.PubMed doi:10.1097/00004714-198210000-00012

19. Morgenstern H, Glazer WM. Identifying risk factors for tardive dyskinesia among long-term outpatients maintained with neuroleptic medications: results of the Yale Tardive Dyskinesia Study. Arch Gen Psychiatry. 1993;50(9):723-733. PubMed

20. Kane JM, Smith JM. Tardive dyskinesia: prevalence and risk factors, 1959 to 1979. Arch Gen Psychiatry. 1982;39(4):473-481. PubMed

21. Quitkin F, Rifkin A, Klein DF. Very high dosage vs standard dosage fluphenazine in schizophrenia: a double-blind study of nonchronic treatment-refractory patients. Arch Gen Psychiatry. 1975;32(10):1276-1281. PubMed

22. Kane JM, Rifkin A, Woerner M, et al. Low-dose neuroleptic treatment of outpatient schizophrenics. I: preliminary results for relapse rates. Arch Gen Psychiatry. 1983;40(8):893-896. PubMed

23. Kane JM, Rifkin A, Quitkin F, et al. Antipsychotic drug blood levels and clinical outcome. In: Klein DF, Gittelman-Klein R, eds. Progress in Psychiatric Drug Treatment. New York, NY: Brunner Mazel; 1976:399-408.

24. Baldessarini RJ, Davis JM. What is the best maintenance dose of neuroleptics in schizophrenia? Psychiatry Res. 1980;3(2):115-122.PubMed doi:10.1016/0165-1781(80)90028-1

25. Baldessarini RJ, Cohen BM, Teicher MH. Significance of neuroleptic dose and plasma level in the pharmacological treatment of psychoses. Arch Gen Psychiatry. 1988;45(1):79-91. PubMed

26. Geddes J, Freemantle N, Harrison P, et al. Atypical antipsychotics in the treatment of schizophrenia: systematic overview and meta-regression analysis. BMJ. 2000;321(7273):1371-1376.PubMed doi:10.1136/bmj.321.7273.1371

27. Leucht S, Wahlbeck K, Hamann J, et al. New generation antipsychotics versus low-potency conventional antipsychotics: a systematic review and meta-analysis. Lancet. 2003;361(9369):1581-1589.PubMed doi:10.1016/S0140-6736(03)13306-5

28. Leucht S, Corves C, Arbter D, et al. Second-generation versus first-generation antipsychotic drugs for schizophrenia: a meta-analysis. Lancet. 2009;373(9657):31-41.PubMed doi:10.1016/S0140-6736(08)61764-X

29. Zimbroff DL, Kane JM, Tamminga CA, et al; Sertindole Study Group. Controlled, dose-response study of sertindole and haloperidol in the treatment of schizophrenia. Am J Psychiatry. 1997;154(6):782-791. PubMed

30. Schooler N, Rabinowitz J, Davidson M, et al; Early Psychosis Global Working Group. Risperidone and haloperidol in first-episode psychosis: a long-term randomized trial. Am J Psychiatry. 2005;162(5):947-953.PubMed doi:10.1176/appi.ajp.162.5.947

31. Kahn RS, Fleischhacker WW, Boter H, et al; EUFEST study group. Effectiveness of antipsychotic drugs in first-episode schizophrenia and schizophreniform disorder: an open randomised clinical trial. Lancet. 2008;371(9618):1085-1097.PubMed doi:10.1016/S0140-6736(08)60486-9

32. Good WW, Sterling M, Holtzman WH. Termination of chlorpromazine with schizophrenia patients. Am J Psychol. 1958;115(5):443-448. PubMed

33. Engelhardt DM, Rosen B, Freedman N, et al. Phenothiazines in prevention of psychiatric hospitalization. II: duration of treatment exposure. JAMA. 1963;186:981-983. PubMed

34. Engelhardt DM, Rosen B, Freedman N, et al. Phenothiazines in prevention of psychiatric hospitalization. IV: delay or prevention of hospitalization—a reevaluation. Arch Gen Psychiatry. 1967;16(1):98-101. PubMed

35. Kane JM. Schizophrenia. N Engl J Med. 1996;334(1):34-41.PubMed doi:10.1056/NEJM199601043340109

36. Kane JM, Quitkin F, Rifkin A, et al. Comparison of the incidence and severity of extrapyramidal side effects with fluphenazine enanthate and fluphenazine decanoate. Am J Psychiatry. 1978;135(12):1539-1542. PubMed

37. Adams CE, Fenton MKP, Quraishi S, et al. Systematic meta-review of depot antipsychotic drugs for people with schizophrenia. Br J Psychiatry. 2001;179(4):290-299.PubMed doi:10.1192/bjp.179.4.290

38. Haddad PM, Taylor M, Niaz OS. First-generation antipsychotic long-acting injections v oral antipsychotics in schizophrenia: systematic review of randomised controlled trials and observational studies. Br J Psychiatry Suppl. 2009;52(52):S20-S28.PubMed doi:10.1192/bjp.195.52.s20

39. Fleischhacker WW. Second-generation antipsychotic long-acting injections: systematic review. Br J Psychiatry Suppl. 2009;52(52):S29-S36.PubMed doi:10.1192/bjp.195.52.s29

40. Griffith RW, Saameli K. Clozapine and agranulocytosis [letter]. Lancet. 1975;2(7936):657.PubMed doi:10.1016/S0140-6736(75)90135-X

41. Kane JM, Honigfeld G, Singer J, et al. Clozapine for the treatment-resistant schizophrenic: a double-blind comparison with chlorpromazine. Arch Gen Psychiatry. 1988;45(9):789-796. PubMed

42. Essali A, Al-Haj Haasan N, Li C, et al. Clozapine versus typical neuroleptic medication for schizophrenia. Cochrane Database Syst Rev. 2009; (1)CD000059. PubMed

43. Davis JM, Chen N. Dose response and dose equivalence of antipsychotics. J Clin Psychopharmacol. 2004;24(2):192-208.PubMed doi:10.1097/01.jcp.0000117422.05703.ae

44. Rosenheck RA. Evaluating the cost-effectiveness of reduced tardive dyskinesia with second-generation antipsychotics. Br J Psychiatry. 2007;191(3):238-245.PubMed doi:10.1192/bjp.bp.106.035063

45. Kane JM, Rifkin A, Quitkin F, et al. Fluphenazine vs placebo in patients with remitted, acute first-episode schizophrenia. Arch Gen Psychiatry. 1982;39(1):70-73. PubMed

46. Bilder RM, Goldman RS, Robinson D, et al. Neuropsychology of first-episode schizophrenia: initial characterization and clinical correlates. Am J Psychiatry. 2000;157(4):549-559.PubMed doi:10.1176/appi.ajp.157.4.549

47. Loebel AD, Lieberman JA, Alvir JM, et al. Duration of psychosis and outcome in first-episode schizophrenia. Am J Psychiatry. 1992;149(9):1183-1188. PubMed

48. Robinson DG, Woerner MG, Delman HM, et al. Pharmacological treatments for first-episode schizophrenia. Schizophr Bull. 2005;31(3):705-722.PubMed doi:10.1093/schbul/sbi032

49. Robinson DG, Woerner MG, Alvir JM, et al. Predictors of treatment response from a first episode of schizophrenia or schizoaffective disorder. Am J Psychiatry. 1999;156(4):544-549. PubMed

50. Melle I, Larsen TK, Haahr U, et al. Prevention of negative symptom psychopathologies in first-episode schizophrenia: two-year effects of reducing the duration of untreated psychosis. Arch Gen Psychiatry. 2008;65(6):634-640.PubMed doi:10.1001/archpsyc.65.6.634

51. Robinson DG, Woerner MG, Alvir JM, et al. Predictors of relapse following response from a first episode of schizophrenia or schizoaffective disorder. Arch Gen Psychiatry. 1999;56(3):241-247.PubMed doi:10.1001/archpsyc.56.3.241

52. Robinson DG, Woerner MG, McMeniman M, et al. Symptomatic and functional recovery from a first episode of schizophrenia or schizoaffective disorder. Am J Psychiatry. 2004;161(3):473-479.PubMed doi:10.1176/appi.ajp.161.3.473

53. Kumra S, Oberstar JV, Sikich L, et al. Efficacy and tolerability of second-generation antipsychotics in children and adolescents with schizophrenia. Schizophr Bull. 2008;34(1):60-71.PubMed doi:10.1093/schbul/sbm109

54. Yung AR, McGorry PD. The initial prodrome in psychosis: descriptive and qualitative aspects. Aust N Z J Psychiatry. 1996;30(5):587-599.PubMed doi:10.3109/00048679609062654

55. Miller TJ, McGlashan TH, Woods SW, et al. Symptom assessment in schizophrenic prodromal states. Psychiatr Q. 1999;70(4):273-287.PubMed doi:10.1023/A:1022034115078

56. Yung AR, Stanford C, Cosgrave E, et al. Testing the ultra high risk (prodromal) criteria for the prediction of psychosis in a clinical sample of young people. Schizophr Res. 2006;84(1):57-66.PubMed doi:10.1016/j.schres.2006.03.014

57. Correll CU, Hauser M, Auther AM, et al. Research in people with psychosis risk syndrome: a review of the current evidence and future directions. J Child Psychol Psychiatry. 2010;51(4):390-431.PubMed doi:10.1111/j.1469-7610.2010.02235.x

58. Rosenheck R, Perlick D, Bingham S, et al; Department of Veterans Affairs Cooperative Study Group on the Cost-Effectiveness of Olanzapine. Effectiveness and cost of olanzapine and haloperidol in the treatment of schizophrenia: a randomized controlled trial. JAMA. 2003;290(20):2693-2702.PubMed doi:10.1001/jama.290.20.2693

59. Lieberman JA, Stroup TS, McEvoy JP, et al; Clinical Antipsychotic Trials of Intervention Effectiveness (CATIE) Investigators. Effectiveness of antipsychotic drugs in patients with chronic schizophrenia. N Engl J Med. 2005;353(12):1209-1223.PubMed doi:10.1056/NEJMoa051688

60. Jones PB, Barnes TR, Davies L, et al. Randomized controlled trial of the effect on Quality of Life of second- vs first-generation antipsychotic drugs in schizophrenia: Cost Utility of the Latest Antipsychotic Drugs in Schizophrenia Study (CUtLASS 1). Arch Gen Psychiatry. 2006;63(10):1079-1087.PubMed doi:10.1001/archpsyc.63.10.1079

61. Leucht S, Komossa K, Rummel-Kluge C, et al. A meta-analysis of head-to-head comparisons of second-generation antipsychotics in the treatment of schizophrenia. Am J Psychiatry. 2009;166(2):152-163.PubMed doi:10.1176/appi.ajp.2008.08030368

62. McEvoy JP, Lieberman JA, Stroup TS, et al; CATIE Investigators. Effectiveness of clozapine versus olanzapine, quetiapine, and risperidone in patients with chronic schizophrenia who did not respond to prior atypical antipsychotic treatment. Am J Psychiatry. 2006;163(4):600-610.PubMed doi:10.1176/appi.ajp.163.4.600

63. Lewis SW, Barnes TR, Davies L, et al. Randomized controlled trial of effect of prescription of clozapine versus other second-generation antipsychotic drugs in resistant schizophrenia. Schizophr Bull. 2006;32(4):715-723.PubMed doi:10.1093/schbul/sbj067

64. Leucht S, Arbter D, Engel RR, et al. How effective are second-generation antipsychotic drugs? a meta-analysis of placebo-controlled trials. Mol Psychiatry. 2009;14(4):429-447.PubMed doi:10.1038/sj.mp.4002136

65. Woods SW, Gueorguieva RV, Baker CB, et al. Control group bias in randomized atypical antipsychotic medication trials for schizophrenia. Arch Gen Psychiatry. 2005;62(9):961-970.PubMed doi:10.1001/archpsyc.62.9.961

66. Kemmler G, Hummer M, Widschwendter C, et al. Dropout rates in placebo-controlled and active-control clinical trials of antipsychotic drugs: a meta-analysis. Arch Gen Psychiatry. 2005;62(12):1305-1312.PubMed doi:10.1001/archpsyc.62.12.1305

67. Heres S, Davis J, Maino K, et al. Why olanzapine beats risperidone, risperidone beats quetiapine, and quetiapine beats olanzapine: an exploratory analysis of head-to-head comparison studies of second-generation antipsychotics. Am J Psychiatry. 2006;163(2):185-194.PubMed doi:10.1176/appi.ajp.163.2.185

68. Essock SM, Covell NH, Davis SM, et al. Effectiveness of switching antipsychotic medications. Am J Psychiatry. 2006;163(12):2090-2095.PubMed doi:10.1176/appi.ajp.163.12.2090

69. Kraemer HC, Glick ID, Klein DF. Clinical trials design lessons from the CATIE study. Am J Psychiatry. 2009;166(11):1222-1228.PubMed doi:10.1176/appi.ajp.2009.08121809

70. Leucht S, Barnes TR, Kissling W, et al. Relapse prevention in schizophrenia with new-generation antipsychotics: a systematic review and exploratory meta-analysis of randomized, controlled trials. Am J Psychiatry. 2003;160(7):1209-1222.PubMed doi:10.1176/appi.ajp.160.7.1209

71. Fischer-Barnicol D, Lanquillon S, Haen E, et al; Working Group Drugs in Psychiatry. Typical and atypical antipsychotics—the misleading dichotomy: results from the Working Group Drugs in Psychiatry (AGATE). Neuropsychobiology. 2008;57(1-2):80-87.PubMed doi:10.1159/000135641

72. Correll CU, Leucht S, Kane JM. Lower risk for tardive dyskinesia associated with second-generation antipsychotics: a systematic review of 1-year studies. Am J Psychiatry. 2004;161(3):414-425.PubMed doi:10.1176/appi.ajp.161.3.414

73. Vinogradov S, Fisher M, Warm H, et al. The cognitive cost of anticholinergic burden: decreased response to cognitive training in schizophrenia. Am J Psychiatry. 2009;166(9):1055-1062.PubMed doi:10.1176/appi.ajp.2009.09010017

74. Newcomer JW. Second-generation (atypical) antipsychotics and metabolic effects: a comprehensive literature review. CNS Drugs. 2005;19(suppl 1):1-93.PubMed doi:10.2165/00023210-200519001-00001

75. Fleischhacker WW, Cetkovich-Bakmas M, De Hert M, et al. Comorbid somatic illnesses in patients with severe mental disorders: clinical, policy, and research challenges. J Clin Psychiatry. 2008;69(4):514-519.PubMed doi:10.4088/JCP.v69n0401

76. Leucht S, Burkard T, Henderson J, et al. Physical illness and schizophrenia: a review of the literature. Acta Psychiatr Scand. 2007;116(5):317-333.PubMed doi:10.1111/j.1600-0447.2007.01095.x

77. Colton CW, Manderscheid RW. Congruencies in increased mortality rates, years of potential life lost, and causes of death among public mental health clients in eight states. Prev Chronic Dis. 2006;3(2):A42. PubMed

78. Lieberman JA, Phillips M, Gu H, et al. Atypical and conventional antipsychotic drugs in treatment-naive first-episode schizophrenia: a 52-week randomized trial of clozapine vs chlorpromazine. Neuropsychopharmacology. 2003;28(5):995-1003. PubMed

79. Zipursky RB, Gu H, Green AI, et al. Course and predictors of weight gain in people with first-episode psychosis treated with olanzapine or haloperidol. Br J Psychiatry. 2005;187(6):537-543.PubMed doi:10.1192/bjp.187.6.537

80. Perez-Iglesias R, Crespo-Facorro B, Martinez-Garcia O, et al. Weight gain induced by haloperidol, risperidone and olanzapine after 1 year: findings of a randomized clinical trial in a drug-naׯve population. Schizophr Res. 2008;99(1-3):13-22.PubMed doi:10.1016/j.schres.2007.10.022

81. Correll CU, Manu P, Olshanskiy V, et al. Cardiometabolic risk of second-generation antipsychotic medications during first-time use in children and adolescents. JAMA. 2009;302(16):1765-1773.PubMed doi:10.1001/jama.2009.1549

82. Lambert TJ, Newcomer JW. Are the cardiometabolic complications of schizophrenia still neglected? barriers to care. Med J Aust. 2009;190(suppl):S39-S42. PubMed

83. American Diabetes Association; American Psychiatric Association; American Association of Clinical Endocrinologists; et al. Consensus development conference on antipsychotic drugs and obesity and diabetes. Diabetes Care. 2004;27(2):596-601.PubMed doi:10.2337/diacare.27.2.596

84. Morrato EH, Druss B, Hartung DM, et al. Metabolic testing rates in 3 state Medicaid programs after FDA warnings and ADA/APA recommendations for second-generation antipsychotic drugs. Arch Gen Psychiatry. 2010;67(1):17-24.PubMed doi:10.1001/archgenpsychiatry.2009.179

85. Morrato EH, Nicol GE, Maahs D, et al. Metabolic screening in children receiving antipsychotic drug treatment. Arch Pediatr Adolesc Med. 2010;164(4):344-351. PubMed

86. Nasrallah HA, Meyer JM, Goff DC, et al. Low rates of treatment for hypertension, dyslipidemia and diabetes in schizophrenia: data from the CATIE schizophrenia trial sample at baseline. Schizophr Res. 2006;86(1-3):15-22.PubMed doi:10.1016/j.schres.2006.06.026

87. Correll CU, Harris JL, Pantaleon Moya RA, et al. Low-density lipoprotein cholesterol in patients treated with atypical antipsychotics: missed targets and lost opportunities. Schizophr Res. 2007;92(1-3):103-107.PubMed doi:10.1016/j.schres.2007.01.004

88. Correll CU, Druss BG, Lombardo I, et al. Findings of a national cardiometabolic screening program in 10,084 psychiatric outpatients. Psychiatr Serv. In press.

89. Druss BG, Bradford WD, Rosenheck RA, et al. Quality of medical care and excess mortality in older patients with mental disorders. Arch Gen Psychiatry. 2001;58(6):565-572.PubMed doi:10.1001/archpsyc.58.6.565

90. Frye RL. Optimal care of patients with type 2 diabetes mellitus and coronary artery disease. Am J Med. 2003;115(suppl 8A):93S-98S.PubMed doi:10.1016/j.amjmed.2003.09.015

91. Andreasen NC, Carpenter WT Jr, Kane JM, et al. Remission in schizophrenia: proposed criteria and rationale for consensus. Am J Psychiatry. 2005;162(3):441-449.PubMed doi:10.1176/appi.ajp.162.3.441

92. Kane JM, Leucht S. Unanswered questions in schizophrenia clinical trials. Schizophr Bull. 2008;34(2):302-309.PubMed doi:10.1093/schbul/sbm143

93. Leucht S, Heres S, Hamann J, et al. Methodological issues in current antipsychotic drug trials. Schizophr Bull. 2008;34(2):275-285.PubMed doi:10.1093/schbul/sbm159

94. Liberman RP, Kopelowicz A, Ventura J, et al. Operational criteria and factors related to recovery from schizophrenia. Int Rev Psychiatry. 2002;14(4):256-272. doi:10.1080/0954026021000016905

95. Naber D, Karow A, Lambert M. Subjective well-being under the neuroleptic treatment and its relevance for compliance. Acta Psychiatr Scand suppl. 2005;111(427):29-34. doi:10.1111/j.1600-0447.2005.00542.x

96. Lambert M, Schimmelmann BG, Naber D, et al. Prediction of remission as a combination of symptomatic and functional remission and adequate subjective well-being in 2960 patients with schizophrenia. J Clin Psychiatry. 2006;67(11):1690-1697.PubMed doi:10.4088/JCP.v67n1104

97. Green MF, Penn DL, Bentall R, et al. Social cognition in schizophrenia: an NIMH workshop on definitions, assessment, and research opportunities. Schizophr Bull. 2008;34(6):1211-1220.PubMed doi:10.1093/schbul/sbm145

98. Harvey PD, Green MF, Bowie C, et al. The dimensions of clinical and cognitive change in schizophrenia: evidence for independence of improvements. Psychopharmacology (Berl). 2006;187(3):356-363.PubMed doi:10.1007/s00213-006-0432-1

99. Keefe RS, Vinogradov S, Medalia A, et al. Report from the Working Group Conference on Multisite Trial Design for Cognitive Remediation in Schizophrenia [published online ahead of print March 1, 2010]. Schizophr Bull. doi:10.1093/schbul/sbq010 PubMed

100. Lehman AF, Goldberg R, Dixon LB, et al. Improving employment outcomes for persons with severe mental illnesses. Arch Gen Psychiatry. 2002;59(2):165-172.PubMed doi:10.1001/archpsyc.59.2.165

101. McGurk SR, Schiano D, Mueser KT, et al. Implementation of the thinking skills for work program in a psychosocial clubhouse. Psychiatr Rehabil J. 2010;33(3):190-199.PubMed doi:10.2975/33.3.2010.190.199

102. Evans JD, Bond GR, Meyer PS, et al. Cognitive and clinical predictors of success in vocational rehabilitation in schizophrenia. Schizophr Res. 2004;70(2-3):331-342.PubMed doi:10.1016/j.schres.2004.01.011

103. Dixon LB, Dickerson F, Bellack AS, et al; Schizophrenia Patient Outcomes Research Team (PORT). The 2009 schizophrenia PORT psychosocial treatment recommendations and summary statements. Schizophr Bull. 2010;36(1):48-70.PubMed doi:10.1093/schbul/sbp115

104. Nasrallah HA, Keshavan MS, Benes FM, et al. Proceedings and data from The Schizophrenia Summit: a critical appraisal to improve the management of Schizophrenia. J Clin Psychiatry. 2009;70(suppl 1):4-46. PubMed

105. Falkai P, Mike O, Inez MG, et al; MEOS Consortium. A roadmap to disentangle the molecular etiology of schizophrenia. Eur Psychiatry. 2008;23(4):224-232.PubMed doi:10.1016/j.eurpsy.2008.02.006

106. Lathia CD, Amakye D, Dai W, et al. The value, qualification, and regulatory use of surrogate end points in drug development. Clin Pharmacol Ther. 2009;86(1):32-43.PubMed doi:10.1038/clpt.2009.69

107. Nedopil N, Pflieger R, Rüther E. The prediction of acute response, remission and general outcome of neuroleptic treatment in acute schizophrenic patients. Pharmacopsychiatria. 1983;16(6):201-205.PubMed doi:10.1055/s-2007-1019499

108. Bartkó G, Herczeg I, Békésy M. Predicting outcome of neuroleptic treatment on the basis of subjective response and early clinical improvement. J Clin Psychiatry. 1987;48(9):363-365. PubMed

109. Awad AG, Voruganti LN. Neuroleptic dysphoria: revisiting the concept 50 years later. Acta Psychiatr Scand suppl. 2005;111(s427):6-13. doi:10.1111/j.1600-0447.2005.00539.x PubMed

110. Agid O, Kapur S, Arenovich T, et al. Delayed-onset hypothesis of antipsychotic action: a hypothesis tested and rejected. Arch Gen Psychiatry. 2003;60(12):1228-1235.PubMed doi:10.1001/archpsyc.60.12.1228

111. Leucht S, Busch R, Hamann J, et al. Early-onset hypothesis of antipsychotic drug action: a hypothesis tested, confirmed and extended. Biol Psychiatry. 2005;57(12):1543-1549.PubMed doi:10.1016/j.biopsych.2005.02.023

112. Agid O, Kapur S, Warrington L, et al. Early onset of antipsychotic response in the treatment of acutely agitated patients with psychotic disorders. Schizophr Res. 2008;102(1-3):241-248.PubMed doi:10.1016/j.schres.2008.03.016

113. Correll CU, Malhotra AK, Kaushik S, et al. Early prediction of antipsychotic response in schizophrenia. Am J Psychiatry. 2003;160(11):2063-2065.PubMed doi:10.1176/appi.ajp.160.11.2063

114. Leucht S, Busch R, Kissling W, et al. Early prediction of antipsychotic nonresponse among patients with schizophrenia. J Clin Psychiatry. 2007;68(3):352-360.PubMed doi:10.4088/JCP.v68n0301

115. Chang YC, Lane HY, Yang KH, et al. Optimizing early prediction for antipsychotic response in schizophrenia. J Clin Psychopharmacol. 2006;26(6):554-559.PubMed doi:10.1097/01.jcp.0000246211.95905.8c

116. Ascher-Svanum H, Nyhuis AW, Faries DE, et al. Clinical, functional, and economic ramifications of early nonresponse to antipsychotics in the naturalistic treatment of schizophrenia. Schizophr Bull. 2008;34(6):1163-1171.PubMed doi:10.1093/schbul/sbm134

117. Kinon BJ, Chen L, Ascher-Svanum H, et al. Predicting response to atypical antipsychotics based on early response in the treatment of schizophrenia. Schizophr Res. 2008;102(1-3):230-240.PubMed doi:10.1016/j.schres.2008.02.021

118. Kinon BJ, Chen L, Ascher-Svanum H, et al. Early response to antipsychotic drug therapy as a clinical marker of subsequent response in the treatment of schizophrenia. Neuropsychopharmacology. 2010;35(2):581-590.PubMed doi:10.1038/npp.2009.164

119. Kay SR, Fiszbein A, Opler LA. The Positive and Negative Syndrome Scale (PANSS) for schizophrenia. Schizophr Bull. 1987;13(2):261-276. PubMed

120. Overall JE, Gorham DR. The Brief Psychiatric Rating Scale. Psychol Rep. 1962;10(3):799-812. doi:10.2466/PR0.10.3.799-812

121. Guidotti A, Auta J, Davis JM, et al. GABAergic dysfunction in schizophrenia: new treatment strategies on the horizon. Psychopharmacology (Berl). 2005;180(2):191-205.PubMed doi:10.1007/s00213-005-2212-8

122. Mansvelder HD, van Aerde KI, Couey JJ, et al. Nicotinic modulation of neuronal networks: from receptors to cognition. Psychopharmacology (Berl). 2006;184(3-4):292-305.PubMed doi:10.1007/s00213-005-0070-z

123. Conn PJ, Tamminga C, Schoepp DD, et al. Schizophrenia: moving beyond monoamine antagonists. Mol Interv. 2008;8(2):99-107.PubMed doi:10.1124/mi.8.2.7

124. Sellin AK, Shad M, Tamminga C. Muscarinic agonists for the treatment of cognition in schizophrenia. CNS Spectr. 2008;13(11):985-996. PubMed