336 results on '"Howes, OD"'
Search Results
2. Predicting treatment-resistance from first-episode psychosis using routinely collected clinical information: development and external validation study
- Author
-
Osimo, E, Perry, BI, Mallikarjun, P, Pritchard, M, Lewis, J, Katunda, A, Murray, GK, Perez, J, Jones, PB, Cardinal, RN, Howes, OD, Upthegrove, R, and Khandaker, GM
- Abstract
Around a quarter of people who experience a first episode of psychosis (FEP) will develop treatment-resistant schizophrenia (TRS), but there are currently no established clinically useful methods to predict this from baseline. We aimed to explore the predictive potential for clozapine use as a proxy for TRS of routinely collected, objective biomedical predictors at FEP onset, and to externally validate the model in a separate clinical sample of people with FEP. We developed and externally validated a forced-entry logistic regression risk prediction Model fOr cloZApine tReaTment, or MOZART, to predict up to 8-year risk of clozapine use from FEP using routinely recorded information including age, sex, ethnicity, triglycerides, alkaline phosphatase levels, and lymphocyte counts. We also produced a least-absolute shrinkage and selection operator (LASSO) based model, additionally including neutrophil count, smoking status, body mass index, and random glucose levels. The models were developed using data from two UK psychosis early intervention services (EIS) and externally validated in another UK EIS. Model performance was assessed via discrimination and calibration. We developed the models in 785 patients, and validated externally in 1,110 patients. Both models predicted clozapine use well at internal validation (MOZART: C 0.70; 95%CI 0.63,0.76; LASSO: 0.69; 95%CI 0.63,0.77). At external validation, discrimination performance reduced (MOZART: 0.63; 0.58,0.69; LASSO: 0.64; 0.58,0.69) but recovered after re-estimation of the lymphocyte predictor (C: 0.67; 0.62,0.73). Calibration plots showed good agreement between observed and predicted risk in the forced-entry model. We also present a decision-curve analysis and an online data visualisation tool. The use of routinely collected clinical information including blood-based biomarkers taken at FEP onset can help to predict the individual risk of clozapine use, and should be considered equally alongside other potentially useful information such as symptom scores in large-scale efforts to predict psychiatric outcomes.
- Published
- 2022
3. Reduced mu opioid receptor availability in schizophrenia revealed with [C-11]-carfentanil positron emission tomographic Imaging
- Author
-
Ashok, AH, Myers, J, Marques, TR, Rabiner, EA, and Howes, OD
- Subjects
REWARD ,Science & Technology ,ALCOHOL DEPENDENCE ,NUCLEUS-ACCUMBENS ,C-11 CARFENTANIL ,NEGATIVE SYMPTOMS ,behavioral disciplines and activities ,Multidisciplinary Sciences ,SOCIAL REJECTION ,ANTIPSYCHOTIC-DRUGS ,POSTMORTEM BRAIN ,mental disorders ,ENDOGENOUS OPIOIDS ,Science & Technology - Other Topics ,IN-VIVO - Abstract
Negative symptoms, such as amotivation and anhedonia, are a major cause of functional impairment in schizophrenia. There are currently no licensed treatments for negative symptoms, highlighting the need to understand the molecular mechanisms underlying them. Mu-opioid receptors (MOR) in the striatum play a key role in hedonic processing and reward function and are reduced post-mortem in schizophrenia. However, it is unknown if mu-opioid receptor availability is altered in-vivo or related to negative symptoms in schizophrenia. Using [11 C]-carfentanil positron emission tomography (PET) scans in 19 schizophrenia patients and 20 age-matched healthy controls, here we show a significantly lower MOR availability in patients with schizophrenia in the striatum (Cohen’s d = 0.7), and the hedonic network. In addition, we report a marked global increase in inter-regional covariance of MOR availability in schizophrenia, largely due to increased cortical-subcortical covariance.
- Published
- 2019
4. Determinants of treatment response in first-episode psychosis: an 18F-DOPA PET study
- Author
-
Jauhar, S, Veronese, M, Nour, MM, Rogdaki, M, Hathway, P, Turkheimer, FE, Stone, J, Egerton, A, McGuire, P, Kapur, S, Howes, OD, Jauhar, S, Veronese, M, Nour, MM, Rogdaki, M, Hathway, P, Turkheimer, FE, Stone, J, Egerton, A, McGuire, P, Kapur, S, and Howes, OD
- Abstract
Psychotic illnesses show variable responses to treatment. Determining the neurobiology underlying this is important for precision medicine and the development of better treatments. It has been proposed that dopaminergic differences underlie variation in response, with striatal dopamine synthesis capacity (DSC) elevated in responders and unaltered in non-responders. We therefore aimed to test this in a prospective cohort, with a nested case-control comparison. 40 volunteers (26 patients with first-episode psychosis and 14 controls) received an 18F-DOPA Positron Emission Tomography scan to measure DSC (Kicer) prior to antipsychotic treatment. Clinical assessments (Positive and Negative Syndrome Scale, PANSS, and Global Assessment of Functioning, GAF) occurred at baseline and following antipsychotic treatment for a minimum of 4 weeks. Response was defined using improvement in PANSS Total score of >50%. Patients were followed up for at least 6 months, and remission criteria applied. There was a significant effect of group on Kicer in associative striatum (F(2, 37) = 7.9, p = 0.001). Kicer was significantly higher in responders compared with non-responders (Cohen's d = 1.55, p = 0.01) and controls (Cohen's d = 1.31, p = 0.02). Kicer showed significant positive correlations with improvements in PANSS-positive (r = 0.64, p < 0.01), PANSS negative (rho = 0.51, p = 0.01), and PANSS total (rho = 0.63, p < 0.01) ratings and a negative relationship with change in GAF (r = -0.55, p < 0.01). Clinical response is related to baseline striatal dopaminergic function. Differences in dopaminergic function between responders and non-responders are present at first episode of psychosis, consistent with dopaminergic and non-dopaminergic sub-types in psychosis, and potentially indicating a neurochemical basis to stratify psychosis.
- Published
- 2019
5. Correction: Altered activation and connectivity in a hippocampal-basal ganglia-midbrain circuit during salience processing in subjects at ultra high risk for psychosis.
- Author
-
Winton-Brown, T, Schmidt, A, Roiser, JP, Howes, OD, Egerton, A, Fusar-Poli, P, Bunzeck, N, Grace, AA, Duzel, E, Kapur, S, McGuire, P, Winton-Brown, T, Schmidt, A, Roiser, JP, Howes, OD, Egerton, A, Fusar-Poli, P, Bunzeck, N, Grace, AA, Duzel, E, Kapur, S, and McGuire, P
- Abstract
This Article was originally published under Nature Research's License to Publish, but has now been made available under a CC BY 4.0 license. The PDF and HTML versions of the Article have been modified accordingly.
- Published
- 2018
6. Antipsychotic treatment resistance in first-episode psychosis:prevalence, subtypes and predictors
- Author
-
Demjaha, A, Lappin, JM, Stahl, D, Patel, MX, MacCabe, JH, Howes, OD, Heslin, M, Reininghaus, Ulrich, Donoghue, K, Lomas, B, Charalambides, M, Onyejiaka, A, Fearon, P, Jones, P, Doody, G, Morgan, C, Dazzan, P, and Murray, RM
- Subjects
Treatment-Resistant ,First-Episode- Psychosis ,Schizophrenia ,Clozapine ,Treatment-Response - Abstract
BACKGROUND: We examined longitudinally the course and predictors of treatment resistance in a large cohort of first-episode psychosis (FEP) patients from initiation of antipsychotic treatment. We hypothesized that antipsychotic treatment resistance is: (a) present at illness onset; and (b) differentially associated with clinical and demographic factors.METHOD: The study sample comprised 323 FEP patients who were studied at first contact and at 10-year follow-up. We collated clinical information on severity of symptoms, antipsychotic medication and treatment adherence during the follow-up period to determine the presence, course and predictors of treatment resistance.RESULTS: From the 23% of the patients, who were treatment resistant, 84% were treatment resistant from illness onset. Multivariable regression analysis revealed that diagnosis of schizophrenia, negative symptoms, younger age at onset, and longer duration of untreated psychosis predicted treatment resistance from illness onset.CONCLUSIONS: The striking majority of treatment-resistant patients do not respond to first-line antipsychotic treatment even at time of FEP. Clinicians must be alert to this subgroup of patients and consider clozapine treatment as early as possible during the first presentation of psychosis.
- Published
- 2017
7. The dopamine hypothesis of Bipolar Affective Disorder: the state of the art and implications for treatment
- Author
-
Ashok, A, Reis-Marques, T, Jauhar, S, Nour, M, Goodwin, G, Young, A, and Howes, OD
- Subjects
Psychiatry ,Biochemistry & Molecular Biology ,Science & Technology ,MOOD STABILIZERS ,REWARD ANTICIPATION ,PSYCHOSOCIAL DISABILITY ,NUCLEUS-ACCUMBENS ,PSYCHIATRIC-DISORDERS ,Neurosciences ,MAJOR DEPRESSION ,11 Medical And Health Sciences ,06 Biological Sciences ,NEUROLEPTIC-STABILIZER-NAIVE ,LONG-TERM USE ,17 Psychology And Cognitive Sciences ,I DISORDER ,mental disorders ,TREATMENT-RESISTANT DEPRESSION ,Neurosciences & Neurology ,Life Sciences & Biomedicine ,Bipolar affective disorder, dopamine, imaging, affective disorders, mania, depression, mechanism, etiology, antidopaminergic, mood stabiliser, anti-depressant - Abstract
Bipolar affective disorder is a common neuropsychiatric disorder. Whilst its neurobiological underpinnings are incompletely understood, the dopamine hypothesis has been a key theory of the pathophysiology of both manic and depressive phases of the illness for over four decades. The increased use of antidopaminergics in the treatment of this disorder and new in vivo neuroimaging and post-mortem studies makes it timely to review this theory. To do this, we conducted a systematic search for post-mortem, pharmacological and imaging studies of dopamine function in bipolar disorder. Converging findings from pharmacological and imaging studies support the hypothesis that a state of hyperdopaminergia, specifically elevations in D2/3 receptor availability and a hyperactive reward processing network, underlies mania. In bipolar depression imaging studies show increased dopamine transporter levels, but changes in other aspects of dopaminergic function are inconsistent. Puzzlingly, pharmacological evidence shows that both dopamine agonists and antidopaminergics can improve bipolar depressive symptoms and perhaps actions at other receptors may reconcile these findings. Tentatively, this evidence suggests a model where an elevation in striatal D2/3 receptor availability would lead to increased dopaminergic neurotransmission and mania, whilst increased striatal dopamine transporter (DAT) levels would lead to reduced dopaminergic function and depression. Thus, it can be speculated that a failure of dopamine receptor and transporter homeostasis might underlie the pathophysiology of this disorder. The limitations of this model include its reliance on pharmacological evidence, as these studies could potentially affect other monoamines, and the scarcity of imaging evidence on dopaminergic function. This model, if confirmed, has implications for developing new treatment strategies such as reducing the dopamine synthesis and/or release in mania and DAT blockade in bipolar depression.
- Published
- 2017
8. A test of the transdiagnostic dopamine hypothesis of psychosis using positron emission tomographic imaging in bipolar affective disorder and schizophrenia
- Author
-
Jauhar, S, Nour, MM, Veronese, M, Rogdaki, M, Bonoldi, I, Azis, M, Turkheimer, F, McGuire, P, Young, AH, and Howes, OD
- Subjects
Adult ,Male ,Fluorine Radioisotopes ,Bipolar Disorder ,Dopamine ,Dopamine Agents ,SYNTHESIS CAPACITY ,MENTAL-DISORDERS ,mental disorders ,Humans ,BRAIN ,IN-VIVO ,Psychiatry ,MANIA ,Psychiatric Status Rating Scales ,Science & Technology ,Reproducibility of Results ,MULTIPLE-TREATMENTS METAANALYSIS ,COMPARATIVE EFFICACY ,Dihydroxyphenylalanine ,PET ,Cross-Sectional Studies ,ANTIPSYCHOTIC-DRUGS ,England ,Psychotic Disorders ,ULTRA-HIGH RISK ,Case-Control Studies ,Positron-Emission Tomography ,Schizophrenia ,Female ,Life Sciences & Biomedicine - Abstract
Importance: The dopamine hypothesis suggests that dopamine abnormalities underlie psychosis, irrespective of diagnosis, implicating dopamine dysregulation in bipolar affective disorder and schizophrenia, in line with the research domain criteria approach. However, this hypothesis has not been directly examined in individuals diagnosed with bipolar disorder with psychosis.Objectives: To test whether dopamine synthesis capacity is elevated in bipolar disorder with psychosis and how this compares with schizophrenia and matched controls and to examine whether dopamine synthesis capacity is associated with psychotic symptom severity, irrespective of diagnostic class.Design, Setting, and Participants: This cross-sectional case-control positron emission tomographic study was performed in the setting of first-episode psychosis services in an inner-city area (London, England). Sixty individuals participated in the study (22 with bipolar psychosis [18 antipsychotic naive or free], 16 with schizophrenia [14 antipsychotic naive or free], and 22 matched controls) and underwent fluorodihydroxyphenyl-l-alanine ([18F]-DOPA) positron emission tomography to examine dopamine synthesis capacity. Standardized clinical measures, including the Positive and Negative Syndrome Scale, Young Mania Rating Scale, and Global Assessment of Functioning, were administered. The study dates were March 2013 to November 2016.Main Outcomes and Measures: Dopamine synthesis capacity (Kicer) and clinical measures (Positive and Negative Syndrome Scale, Young Mania Rating Scale, and Global Assessment of Functioning).Results: The mean (SD) ages of participants were 23.6 (3.6) years in 22 individuals with bipolar psychosis (13 male), 26.3 (4.4) years in 16 individuals with schizophrenia (14 male), and 24.5 (4.5) years in controls (14 male). There was a significant group difference in striatal dopamine synthesis capacity (Kicer) (F2,57 = 6.80, P = .002). Kicer was significantly elevated in both the bipolar group (mean [SD], 13.18 [1.08] × 10-3 min-1; P = .002) and the schizophrenia group (mean [SD], 12.94 [0.79] × 10-3 min-1; P = .04) compared with controls (mean [SD], 12.16 [0.92] × 10-3 min-1). There was no significant difference in striatal Kicer between the bipolar and schizophrenia groups. Kicer was significantly positively correlated with positive psychotic symptom severity in the combined bipolar and schizophrenia sample experiencing a current psychotic episode, explaining 27% of the variance in symptom severity (n = 32, r = 0.52, P = .003). There was a significant positive association between Kicer and positive psychotic symptom severity in individuals with bipolar disorder experiencing a current psychotic episode (n = 16, r = 0.60, P = .01), which remained significant after adjusting for manic symptom severity.Conclusions and Relevance: These findings are consistent with a transdiagnostic role for dopamine dysfunction in the pathoetiology of psychosis and suggest dopamine synthesis capacity as a potential novel drug target for bipolar disorder and schizophrenia.
- Published
- 2017
9. Regulation of dopaminergic function: an [18F]-DOPA PET apomorphine challenge study in humans
- Author
-
Jauhar, S, Veronese, M, Rogdaki, M, Bloomfield, M, Natesan, S, Turkheimer, F, Kapur, S, Howes, OD, Jauhar, S, Veronese, M, Rogdaki, M, Bloomfield, M, Natesan, S, Turkheimer, F, Kapur, S, and Howes, OD
- Abstract
Dopaminergic function has a key role in normal brain function, dopaminergic dysfunction being implicated in numerous neuropsychiatric disorders. Animal studies show that dopaminergic stimulation regulates dopaminergic function, but it is not known whether this exists in humans. In the first study (study 1), we measured dopamine synthesis capacity (indexed as Kicer) to identify the relationship between baseline and change in Kicer under resting conditions for comparison with effects of dopaminergic stimulation. In the second study (study 2), we used a within-subjects design to test effects of dopaminergic stimulation on dopamine synthesis capacity. In study 1, eight volunteers received two 18F-DOPA scans on separate days, both at rest. In study 2, 12 healthy male volunteers received two 18F-DOPA positron emission tomographic (PET) scans after treatment with either the dopamine partial agonist apomorphine (0.03 or 0.005 mg kg-1) or placebo. In study 1, no significant correlation was found between baseline and change in dopamine synthesis capacity between scans (r=-0.57, n=8, P=0.17, two-tailed). In study 2, a significant negative correlation was found between baseline dopamine synthesis capacity and percentage change in dopamine synthesis capacity after apomorphine challenge (r=-0.71, n=12, P=0.01, two-tailed). This correlation was significantly different (P<0.01) from the correlation between baseline and change in dopamine synthesis capacity under unstimulated conditions. One-way repeated-measures analysis of variance showed a significant group (study 1/study 2) × time interaction (F(1,18)=11.5, P=0.003). Our findings suggest that regulation of dopamine synthesis capacity by apomorphine depends on baseline dopamine function, consistent with dopamine stimulation stabilizing dopaminergic function. Loss of this autoregulation may contribute to dopaminergic dysfunction in brain disorders such as schizophrenia, substance dependence, and Parkinson's disease.
- Published
- 2017
10. Altered activation and connectivity in a hippocampal-basal ganglia-midbrain circuit during salience processing in subjects at ultra high risk for psychosis
- Author
-
Winton-Brown, T, Schmidt, A, Roiser, JP, Howes, OD, Egerton, A, Fusar-Poli, P, Bunzeck, N, Grace, AA, Duzel, E, Kapur, S, McGuire, P, Winton-Brown, T, Schmidt, A, Roiser, JP, Howes, OD, Egerton, A, Fusar-Poli, P, Bunzeck, N, Grace, AA, Duzel, E, Kapur, S, and McGuire, P
- Abstract
Animal models of psychosis propose that abnormal hippocampal activity drives increased subcortical dopamine function, which is thought to contribute to aberrant salience processing and psychotic symptoms. These effects appear to be mediated through connections between the hippocampus, ventral striatum/pallidum and the midbrain. The aim of the present study was to examine the activity and connectivity in this pathway in people at ultra high risk (UHR) for psychosis. Functional magnetic resonance imaging was used to compare neural responses in a hippocampal-basal ganglia-midbrain network during reward, novelty and aversion processing between 29 UHR subjects and 32 healthy controls. We then investigated whether effective connectivity within this network is perturbed in UHR subjects, using dynamic causal modelling (DCM). Finally, we examined the relationship between alterations in activation and connectivity in the UHR subjects and the severity of their psychotic symptoms. During reward anticipation, UHR subjects showed greater activation than controls in the ventral pallidum bilaterally. There were no differences in activation during novelty or aversion processing. DCM revealed that reward-induced modulation of connectivity from the ventral striatum/pallidum to the midbrain was greater in UHR subjects than controls, and that in UHR subjects, the strength of connectivity in this pathway was correlated with the severity of their abnormal beliefs. In conclusion, ventral striatal/pallidal function is altered in people at UHR for psychosis and this is related to the level of their psychotic symptoms.
- Published
- 2017
11. Salience Attribution and its Relationship to Cannabis-Induced Psychotic Symptoms
- Author
-
Bloomfield, M, Mouchlianitis, E, Morgan, CJ, Freeman, T, Roiser, J, and Howes, OD
- Subjects
Psychiatry ,1117 Public Health And Health Services ,1701 Psychology ,1109 Neurosciences - Abstract
BACKGROUND: Cannabis is a widely used drug associated with increased risk for psychosis. The dopamine hypothesis of psychosis postulates that altered salience processing leads to psychosis. We therefore tested the hypothesis that cannabis users exhibit aberrant salience and explored the relationship between aberrant salience and dopamine synthesis capacity. METHODS: We tested 17 cannabis users and 17 age- and sex-matched non-user controls using the Salience Attribution Test (SAT), a probabilistic reward-learning task. Within users, cannabis-induced psychotic symptoms were measured with the Psychotomimetic States Inventory (PSI). Dopamine synthesis capacity, indexed as the influx rate constant Kicer, was measured in 10 users and 6 controls with 3,4-dihydroxy-6-[18F]-fluoro-l-phenylalanine positron emission tomography. RESULTS: There was no significant difference in aberrant salience between the groups (F1,32 = 1.12, p=.30 [implicit]; F1,32=1.09, p=.30 [explicit]). Within users there was a significant positive relationship between cannabis-induced psychotic symptom severity and explicit aberrant salience scores (r=.61, p=.04) and there was a significant association between cannabis dependency/abuse status and high implicit aberrant salience scores (F1,15= 5.8, p=.03). Within controls, implicit aberrant salience was inversely correlated with whole striatal dopamine synthesis capacity (r=-.91, p=.01), whereas this relationship was non-significant within users (difference between correlations: Z=-2.05, p=.04). CONCLUSIONS: Aberrant salience is positively associated with cannabis-induced psychotic symptom severity, but is not seen in cannabis users overall. This is consistent with the hypothesis that the link between cannabis use and psychosis involves alterations in salience processing. Longitudinal studies are needed to determine whether these cognitive abnormalities are pre-existing or caused by long-term cannabis use.
- Published
- 2016
12. Brain microglia in psychiatric disorders
- Author
-
Mondelli, Valeria, Vernon, Anthony C, Turkheimer, Federico, Dazzan, Paola, Pariante, Carmine M, Frost, JL, Schafer, DP, Banati, RB, Newcombe, J, Gunn, RN, al., et, Tang, Y, Le, W, Estes, ML, McAllister, AK, Ransohoff, RM, Davalos, D, Grutzendler, J, Yang, G, Peferoen, LA, Vogel, DY, Ummenthum, K, Norden, DM, Trojanowski, PJ, Villanueva, E, Navarro, E, Godbout, JP, Kreisel, T, Frank, MG, Licht, T, Wachholz, S, Eßlinger, M, Plümper, J, Manitz, MP, Juckel, G, Friebe, A, Trépanier, MO, Hopperton, KE, Mizrahi, R, Mechawar, N, Bazinet, RP, Torres-Platas, SG, Cruceanu, C, Chen, GG, Turecki, G, Steiner, J, Bielau, H, Brisch, R, Schnieder, TP, Trencevska, I, Rosoklija, G, Fillman, SG, Cloonan, N, Catts, VS, Rupprecht, R, Papadopoulos, V, Rammes, G, Qiu, ZK, Li, MS, He, JL, Hannestad, J, Gallezot, JD, Schafbauer, T, Israel, I, Ohsiek, A, Al-Momani, E, Mirzaei, N, Tang, SP, Ashworth, S, Gulyás, B, Makkai, B, Kása, P, Turkheimer, FE, Rizzo, G, Bloomfield, PS, Owen, DR, Yeo, AJ, DellaGioia, N, Setiawan, E, Wilson, AA, Su, L, Faluyi, YO, Hong, YT, Haarman, BC, Lek, RF Riemersma-Van der, Groot, JC de, Berckel, BN van, Bossong, MG, Boellaard, R, Doorduin, J, Vries, EF de, Willemsen, AT, Dierckx, RA, Klein, HC, Takano, A, Arakawa, R, Ito, H, Kenk, M, Selvanathan, T, Rao, N, Selvaraj, S, Veronese, M, Coughlin, JM, Wang, Y, Ambinder, EB, Doef, TF van der, Witte, LD de, Sutterland, AL, Hafizi, S, Tseng, HH, Holmes, SE, Hinz, R, Drake, RJ, Yaqub, M, Schuitemaker, A, Edison, P, Pavese, N, Lockhart, A, Davis, B, Matthews, JC, Quarantelli, M, Laule, C, Vavasour, IM, Kolind, SH, Pasternak, O, Sochen, N, Gur, Y, Intrator, N, Assaf, Y, Andreasen, NC, Ehrhardt, JC, Swayze, VW, Supprian, T, Hofmann, E, Warmuth-Metz, M, Franzek, E, Becker, T, Pfefferbaum, A, Sullivan, EV, Hedehus, M, Moseley, M, Lim, KO, Mandl, RC, Schnack, HG, Luigjes, J, Cahn, W, Bagary, MS, Symms, MR, Barker, GJ, Mutsatsa, SH, Joyce, EM, Ron, MA, Foong, J, Maier, M, Brocklehurst, S, Miller, DH, Kubicki, M, Park, H, Westin, CF, Bouix, S, Dahlben, B, Oestreich, LK, Shenton, ME, Amato, D, Beasley, CL, Hahn, MK, Vernon, AC, Natesan, S, Modo, M, Kapur, S, Mondelli, V, Reininghaus, U, Kempton, MJ, Valmaggia, L, Baumeister, D, Lightman, SL, Pariante, CM, Danese, A, Moffitt, TE, Ambler, A, Poulton, R, Caspi, A, Akhtar, R, Ciufolini, S, Meyer, U, So, PW, Lythgoe, DJ, Cotel, MC, Lenartowicz, EM, Anacker, C, Calcia, MA, Bonsall, DR, Barichello, T, Howes, OD, Burke, NN, Fan, CY, Trang, T, McMahon, SB, Russa, F La, Bennett, DL, Püntener, U, Booth, SG, Perry, VH, Teeling, JL, Hahn, YK, Podhaizer, EM, Farris, SP, Miles, MF, Hauser, KF, Knapp, PE, Notter, T, Gschwind, T, Varga, B, Markó, K, Hádinger, N, Cattaneo, A, Ferrari, C, Uher, R, Belvederi, Murri M, Sandiego, CM, Pittman, B, Weber, MD, Sheridan, JF, Raison, CL, Rutherford, RE, Woolwine, BJ, Möller, T, Boddeke, HW, O'Connor, JC, Lawson, MA, André, C, Hinwood, M, Morandini, J, Day, TA, Walker, FR, Bard, F, Bhattacharya, A, Pae, CU, Marks, DM, Han, C, Patkar, AA, Oya, K, Kishi, T, Iwata, N, Pathology, NCA - Neuroinflamation, Molecular cell biology and Immunology, Gastroenterology and hepatology, CCA - Disease profiling, ICaR - Heartfailure and pulmonary arterial hypertension, ICaR - Ischemia and repair, NCA - Brain imaging technology, Radiology and nuclear medicine, Amsterdam Neuroscience - Brain Imaging, Otolaryngology / Head & Neck Surgery, EMGO - Quality of care, AII - Infectious diseases, CCA - Imaging, and Neurology
- Subjects
0301 basic medicine ,medicine.medical_specialty ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Humans ,Psychology ,In patient ,Psychiatry ,Biological Psychiatry ,Neuroinflammation ,Inflammation ,Microglia ,business.industry ,Macrophages ,Mental Disorders ,Brain ,Magnetic Resonance Imaging ,Psychiatry and Mental health ,030104 developmental biology ,medicine.anatomical_structure ,Psychosocial stress ,Treatment strategy ,Autopsy ,business ,Neuroscience ,030217 neurology & neurosurgery ,Stress, Psychological ,Immune activation - Abstract
SummaryThe role of immune activation in psychiatric disorders has attracted considerable attention over the past two decades, contributing to the rise of a new era for psychiatry. Microglia, the macrophages of the brain, are progressively becoming the main focus of the research in this field. In this Review, we assess the literature on microglia activation across different psychiatric disorders, including post-mortem and in-vivo studies in humans and experimental studies in animals. Although microglia activation has been noted in all types of psychiatric disorder, no association was seen with specific diagnostic categories. Furthermore, the findings from these studies highlight that not all psychiatric patients have microglial activation. Therefore, the cause of the neuroinflammation in these cohorts and its implications are unclear. We discuss psychosocial stress as one of the main factors determining microglial activation in patients with psychiatric disorders, and explore the relevance of these findings for future treatment strategies.
- Published
- 2016
13. The nature of dopamine dysfunction in schizophrenia and what this means for treatment.
- Author
-
Howes OD, Kambeitz J, Kim E, Stahl D, Slifstein M, Abi-Dargham A, and Kapur S
- Published
- 2012
14. Dopamine synthesis capacity before onset of psychosis: a prospective [18F]-DOPA PET imaging study.
- Author
-
Howes OD, Bose SK, Turkheimer F, Valli I, Egerton A, Valmaggia LR, Murray RM, McGuire P, Howes, Oliver D, Bose, Subrata K, Turkheimer, Federico, Valli, Isabel, Egerton, Alice, Valmaggia, Lucia R, Murray, Robin M, and McGuire, Philip
- Abstract
Objective: While there is robust evidence of elevated dopamine synthesis capacity once a psychotic disorder has developed, little is known about whether it is altered prior to the first episode of frank illness. The authors addressed this issue by measuring dopamine synthesis capacity in individuals at ultra-high risk of psychosis and then following them to determine their clinical outcome.Method: This prospective study included 30 patients who met standard criteria for being at ultra-high risk of psychosis and 29 healthy volunteers. Participants were scanned using [(18)F]6-fluoro-L-dopa positron emission tomography. The ultra-high-risk patients were scanned at presentation and followed up for at least 3 years to determine their clinical outcome. Six patients had comorbid schizotypal personality disorder and were excluded from the analysis (data are provided for comparison). Of the remaining patients, nine developed a psychotic disorder (psychotic transition group) and 15 did not (nontransition group).Results: There was a significant effect of group on striatal dopamine synthesis capacity. The psychotic transition group had greater dopamine synthesis capacity in the striatum (effect size=1.18) and its associative subdivision (effect size=1.24) than did the healthy comparison subjects and showed a positive correlation between dopamine synthesis capacity and symptom severity. Dopamine synthesis capacity was also significantly greater in the psychotic transition group than in the nontransition group.Conclusions: These findings provide evidence that the onset of frank psychosis is preceded by presynaptic dopaminergic dysfunction. Further research is needed to determine the specificity of elevated dopamine synthesis capacity to particular psychotic disorders. [ABSTRACT FROM AUTHOR]- Published
- 2011
- Full Text
- View/download PDF
15. Letter to the editor: Mind the translation gap: problems in the implementation of early intervention services.
- Author
-
Howes OD, Lim SJR, and Fusar-Poli P
- Published
- 2010
- Full Text
- View/download PDF
16. Julius Wagner-Jauregg, 1857-1940.
- Author
-
Howes OD, Khambhaita A, Fusar-Poli P, Howes, Oliver D, Khambhaita, Asha, and Fusar-Poli, Paolo
- Published
- 2009
- Full Text
- View/download PDF
17. Compulsions in depression: stalking by text message.
- Author
-
Howes OD and Howes, Oliver D
- Published
- 2006
- Full Text
- View/download PDF
18. Aripiprazole-Associated QT Prolongation in a Healthy Study Volunteer: A Case Report and Literature Review.
- Author
-
Chapman GE, Osugo M, de Marvao A, and Howes OD
- Published
- 2024
- Full Text
- View/download PDF
19. Mesostriatal Dopaminergic Circuit Dysfunction in Schizophrenia: A Multimodal Neuromelanin-Sensitive Magnetic Resonance Imaging and [ 18 F]-DOPA Positron Emission Tomography Study.
- Author
-
Vano LJ, McCutcheon RA, Rutigliano G, Kaar SJ, Finelli V, Nordio G, Wellby G, Sedlacik J, Statton B, Rabiner EA, Ye R, Veronese M, Hopkins SC, Koblan KS, Everall IP, and Howes OD
- Subjects
- Humans, Male, Female, Adult, Middle Aged, Ventral Tegmental Area diagnostic imaging, Ventral Tegmental Area metabolism, Corpus Striatum metabolism, Corpus Striatum diagnostic imaging, Schizophrenia diagnostic imaging, Schizophrenia metabolism, Schizophrenia physiopathology, Positron-Emission Tomography, Melanins metabolism, Magnetic Resonance Imaging, Dopamine metabolism, Substantia Nigra diagnostic imaging, Substantia Nigra metabolism, Dihydroxyphenylalanine analogs & derivatives
- Abstract
Background: Striatal hyperdopaminergia is implicated in the pathoetiology of schizophrenia, but how this relates to dopaminergic midbrain activity is unclear. Neuromelanin (NM)-sensitive magnetic resonance imaging provides a marker of long-term dopamine function. We examined whether midbrain NM-sensitive magnetic resonance imaging contrast-to-noise ratio (NM-CNR) was higher in people with schizophrenia than in healthy control (HC) participants and whether this correlated with dopamine synthesis capacity., Methods: One hundred fifty-four participants (schizophrenia group: n = 74, HC group: n = 80) underwent NM-sensitive magnetic resonance imaging of the substantia nigra and ventral tegmental area (SN-VTA). A subset of the schizophrenia group (n = 38) also received [
18 F]-DOPA positron emission tomography to measure dopamine synthesis capacity (Ki cer ) in the SN-VTA and striatum., Results: SN-VTA NM-CNR was significantly higher in patients with schizophrenia than in HC participants (effect size = 0.38, p = .019). This effect was greatest for voxels in the medial and ventral SN-VTA. In patients, SN-VTA Ki cer positively correlated with SN-VTA NM-CNR (r = 0.44, p = .005) and striatal Ki cer (r = 0.71, p < .001). Voxelwise analysis demonstrated that SN-VTA NM-CNR was positively associated with striatal Ki cer (r = 0.53, p = .005) and that this relationship seemed strongest between the ventral SN-VTA and associative striatum in schizophrenia., Conclusions: Our results suggest that NM levels are higher in patients with schizophrenia than in HC individuals, particularly in midbrain regions that project to parts of the striatum that receive innervation from the limbic and association cortices. The direct relationship between measures of NM and dopamine synthesis suggests that these aspects of schizophrenia pathophysiology are linked. Our findings highlight specific mesostriatal circuits as the loci of dopamine dysfunction in schizophrenia and thus as potential therapeutic targets., (Copyright © 2024 Society of Biological Psychiatry. Published by Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
20. New Drug Treatments for Schizophrenia: A Review of Approaches to Target Circuit Dysfunction.
- Author
-
Howes OD, Dawkins E, Lobo MC, Kaar SJ, and Beck K
- Subjects
- Humans, Animals, Dopamine metabolism, Schizophrenia drug therapy, Antipsychotic Agents pharmacology, Antipsychotic Agents therapeutic use
- Abstract
Schizophrenia is a leading cause of global disease burden. Current drug treatments are associated with significant side effects and have limited efficacy for many patients, highlighting the need to develop new approaches that target other aspects of the neurobiology of schizophrenia. Preclinical, in vivo imaging, postmortem, genetic, and pharmacological studies have highlighted the key role of cortical GABAergic (gamma-aminobutyric acidergic)-glutamatergic microcircuits and their projections to subcortical dopaminergic circuits in the pathoetiology of negative, cognitive, and psychotic symptoms. Antipsychotics primarily act downstream of the dopaminergic component of this circuit. However, multiple drugs are currently in development that could target other elements of this circuit to treat schizophrenia. These include drugs for GABAergic or glutamatergic targets, including glycine transporters, D-amino acid oxidase, sodium channels, or potassium channels. Other drugs in development are likely to primarily act on pathways that regulate the dopaminergic system, such as muscarinic or trace amine receptors or 5-HT
2A receptors, while PDE10A inhibitors are being developed to modulate the downstream consequences of dopaminergic dysfunction. Our review considers where new drugs may act on this circuit and their latest clinical trial evidence in terms of indication, efficacy, and side effects. Limitations of the circuit model, including whether there are neurobiologically distinct subgroups of patients, and future directions are also considered. Several drugs based on the mechanisms reviewed have promising clinical data, with the muscarinic agonist KarXT most advanced. If these drugs are approved for clinical use, they have the potential to revolutionize understanding of the pathophysiology and treatment of schizophrenia., (Copyright © 2024 Society of Biological Psychiatry. Published by Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
21. Understanding variability: the role of meta-analysis of variance.
- Author
-
Howes OD and Chapman GE
- Abstract
Meta-analyses traditionally compare the difference in means between groups for one or more outcomes of interest. However, they do not compare the spread of data (variability), which could mean that important effects and/or subgroups are missed. To address this, methods to compare variability meta-analytically have recently been developed, making it timely to review them and consider their strengths, weaknesses, and implementation. Using published data from trials in major depression, we demonstrate how the spread of data can impact both overall effect size and the frequency of extreme observations within studies, with potentially important implications for conclusions of meta-analyses, such as the clinical significance of findings. We then describe two methods for assessing group differences in variability meta-analytically: the variance ratio (VR) and coefficient of variation ratio (CVR). We consider the reporting and interpretation of these measures and how they differ from the assessment of heterogeneity between studies. We propose general benchmarks as a guideline for interpreting VR and CVR effects as small, medium, or large. Finally, we discuss some important limitations and practical considerations of VR and CVR and consider the value of integrating variability measures into meta-analyses.
- Published
- 2024
- Full Text
- View/download PDF
22. Mortality associated with clozapine: what is the evidence?
- Author
-
Fernandez-Egea E, Flanagan RJ, Taylor D, Gaughran F, Lawrie SM, Jenkins C, Smith S, Howes OD, and MacCabe JH
- Subjects
- Humans, United Kingdom epidemiology, Suicide statistics & numerical data, Pharmacovigilance, Clozapine adverse effects, Antipsychotic Agents adverse effects, Schizophrenia drug therapy, Schizophrenia mortality
- Abstract
While clozapine has risks, relative risk of fatality is overestimated. The UK pharmacovigilance programme is efficient, but comparisons with other drugs can mislead because of reporting variations. Clozapine actually lowers mortality, partly by reducing schizophrenia-related suicides, but preventable deaths still occur. Clozapine should be used earlier and more widely, but there should be better monitoring and better management of toxicity.
- Published
- 2024
- Full Text
- View/download PDF
23. Comparative physiological effects of antipsychotic drugs in children and young people: a network meta-analysis.
- Author
-
Rogdaki M, McCutcheon RA, D'Ambrosio E, Mancini V, Watson CJ, Fanshawe JB, Carr R, Telesia L, Martini MG, Philip A, Gilbert BJ, Salazar-de-Pablo G, Kyriakopoulos M, Siskind D, Correll CU, Cipriani A, Efthimiou O, Howes OD, and Pillinger T
- Subjects
- Humans, Child, Adolescent, Randomized Controlled Trials as Topic, Mental Disorders drug therapy, Heart Rate drug effects, Blood Pressure drug effects, Antipsychotic Agents therapeutic use, Network Meta-Analysis
- Abstract
Background: The degree of physiological responses to individual antipsychotic drugs is unclear in children and adolescents. With network meta-analysis, we aimed to investigate the effects of various antipsychotic medications on physiological variables in children and adolescents with neuropsychiatric and neurodevelopmental conditions., Methods: For this network meta-analysis, we searched Medline, EMBASE, PsycINFO, Web of Science, and Scopus from database inception until Dec 22, 2023, and included randomised controlled trials comparing antipsychotics with placebo in children or adolescents younger than 18 years with any neuropsychiatric and neurodevelopmental condition. Primary outcomes were mean change from baseline to end of acute treatment in bodyweight, BMI, fasting glucose, total cholesterol, LDL cholesterol, HDL cholesterol, triglycerides, prolactin, heart rate, systolic blood pressure (SBP), and QT interval corrected for heart rate (QTc) for patients receiving either active treatment or placebo. For multigroup trials reporting several doses, we calculated a summary value for each physiological variable for all doses. After transitivity assessment, we fitted frequentist random-effects network meta-analyses for all comparisons in the network. A Kilim plot was used to summarise the results for all treatments and outcomes, providing information regarding the strength of the statistical evidence of treatment effects, using p values. Network heterogeneity was assessed with τ, risk of bias of individual trials was assessed with the Cochrane Collaboration's Tool for Assessing Risk of Bias, and the credibility of findings from each network meta-analysis was assessed with the Confidence in Network Meta-Analysis (CINEMA) app. This study is registered on PROSPERO (CRD42021274393)., Findings: Of 6676 studies screened, 47 randomised controlled trials were included, which included 6500 children (mean age 13·29 years, SD 2·14) who received treatment for a median of 7 weeks (IQR 6-8) with either placebo (n=2134) or one of aripiprazole, asenapine, blonanserin, clozapine, haloperidol, lurasidone, molindone, olanzapine, paliperidone, pimozide, quetiapine, risperidone, or ziprasidone (n=4366). Mean differences for bodyweight change gain compared with placebo ranged from -2·00 kg (95% CI -3·61 to -0·39) with molindone to 5·60 kg (0·27 to 10·94) with haloperidol; BMI -0·70 kg/m
2 (-1·21 to -0·19) with molindone to 2·03 kg/m2 (0·51 to 3·55) with quetiapine; total cholesterol -0·04 mmol/L (-0·39 to 0·31) with blonanserin to 0·35 mmol/L (0·17 to 0·53) with quetiapine; LDL cholesterol -0·12 mmol/L (-0·31 to 0·07) with risperidone or paliperidone to 0·17 mmol/L (-0·06 to 0·40) with olanzapine; HDL cholesterol 0·05 mmol/L (-0·19 to 0·30) with quetiapine to 0·48 mmol/L (0·18 to 0·78) with risperidone or paliperidone; triglycerides -0·03 mmol/L (-0·12 to 0·06) with lurasidone to 0·29 mmol/L (0·14 to 0·44) with olanzapine; fasting glucose from -0·09 mmol/L (-1·45 to 1·28) with blonanserin to 0·74 mmol/L (0·04 to 1·43) with quetiapine; prolactin from -2·83 ng/mL (-8·42 to 2·75) with aripiprazole to 26·40 ng/mL (21·13 to 31·67) with risperidone or paliperidone; heart rate from -0·20 bpm (-8·11 to 7·71) with ziprasidone to 12·42 bpm (3·83 to 21·01) with quetiapine; SBP from -3·40 mm Hg (-6·25 to -0·55) with ziprasidone to 10·04 mm Hg (5·56 to 14·51) with quetiapine; QTc from -0·61 ms (-1·47 to 0·26) with pimozide to 0·30 ms (-0·05 to 0·65) with ziprasidone., Interpretation: Children and adolescents show varied but clinically significant physiological responses to individual antipsychotic drugs. Treatment guidelines for children and adolescents with a range of neuropsychiatric and neurodevelopmental conditions should be updated to reflect each antipsychotic drug's distinct profile for associated metabolic changes, alterations in prolactin, and haemodynamic alterations., Funding: UK Academy of Medical Sciences, Brain and Behaviour Research Foundation, UK National Institute of Health Research, Maudsley Charity, the Wellcome Trust, Medical Research Council, National Institute of Health and Care Research Biomedical Centre at King's College London and South London and Maudsley NHS Foundation Trust, the Italian Ministry of University and Research, the Italian National Recovery and Resilience Plan, and Swiss National Science Foundation., Competing Interests: Declaration of interests RAM has participated in speaker meetings for Otsuka, Karuna, and Janssen and in advisory boards for Viatris, Boehringer Ingelheim, and Karuna. ED’A has received lecture fees from Lundbeck. GS-d-P has participated in advisory and speaker meetings for Jansen and Menarini. OE has received honoraria and consulting fees from Biogen, paid to his institution. AC has received research, educational, and consultancy fees from the Italian Network for Paediatric Trials, the Cariplo Foundation, Lundbeck, and Angelini Pharma and is the chief investigator of a randomised controlled trial of seltorexant for adolescents with depression that is sponsored by Janssen. CUC has received consultancy fees as an advisor from Alkermes, Angelini, Boehringer Ingelheim, Cardio Diagnostics, Cerevel, CNX Therapeutics, Compass Pathways, Gedeon Richter, Holmusk, IntraCellular Therapies, Janssen, Johnson & Johnson, Karuna, LB Pharma, Lundbeck, MedAvante–ProPhase, Merck, Mindpax, Mitsubishi Tanabe Pharma, Neurelis, Newron, Noven, Novo Nordisk, Otsuka, Pharmabrain, PPD Biotech, Recordati, Rovi, Seqirus, SK Life Science, Sunovion, Supernus, Takeda, Teva, and Viatris; has received speaker fees from AbbVie, Alkermes, Angelini, Aristo, Boehringer Ingelheim, Cerevel, Darnitsa, Denovo, Gedeon Richter, Hikma, Janssen, Johnson & Johnson, Karuna, Lundbeck, Mylan, Otsuka, Recordati, Rovi, Seqirus, Sunovion, Sun Pharma, Takeda, Teva, and Viatris; has received honoraria from Allergan, Biogen, Relmada, Reviva, and Supernus; has provided expert testimony for Janssen and Otsuka; was on a data safety monitoring board for Compass Pathways, Denovo, Lundbeck, Relmada, Reviva, Rovi, Sage, Supernus, Tolmar, and Teva; has received grant support from Janssen and Takeda; has received royalties from UpToDate; and is a stock option holder of Cardio Diagnostics, Mindpax, LB Pharma, PsiloSterics, and Quantic. ODH has received investigator-initiated research funding from and participated in advisory and speaker meetings for Angellini, Autifony, Biogen, Boehringer Ingelheim, Eli Lilly, Heptares, Global Medical Education, Invicro, Janssen, Lundbeck, Neurocrine, Otsuka, Sunovion, Rand, Recordati, Roche, ROVI Biotech, Viatris, and Mylan. TP has participated in educational speaker meetings for Lundbeck, Otsuka, Sunovion, Janssen, Schwabe Pharma, ROVI Biotech, and Recordati and receives book royalties from Wiley Blackwell. TP and RAM co-direct Pharmatik, which designs digital resources to support treatment of mental illness. All other authors declare no competing interests., (Copyright © 2024 The Author(s). Published by Elsevier Ltd. This is an Open Access article under the CC BY 4.0 license. Published by Elsevier Ltd.. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
24. Overcoming the barriers to identifying and managing treatment-resistant schizophrenia and to improving access to clozapine: A narrative review and recommendation for clinical practice.
- Author
-
Agid O, Crespo-Facorro B, de Bartolomeis A, Fagiolini A, Howes OD, Seppälä N, and Correll CU
- Subjects
- Humans, Health Services Accessibility, Schizophrenia drug therapy, Clozapine therapeutic use, Clozapine adverse effects, Antipsychotic Agents therapeutic use, Antipsychotic Agents adverse effects, Schizophrenia, Treatment-Resistant drug therapy
- Abstract
Clozapine is the only approved antipsychotic for treatment-resistant schizophrenia (TRS). Although a large body of evidence supports its efficacy and favorable risk-benefit ratio in individuals who have failed two or more antipsychotics, clozapine remains underused. However, variations in clozapine utilization across geographic and clinical settings suggest that it could be possible to improve its use. In this narrative review and expert opinion, we summarized information available in the literature on the mechanisms of action, effectiveness, and potential adverse events of clozapine. We identified barriers leading to discouragement in clozapine prescription internationally, and we proposed practical solutions to overcome each barrier. One of the main obstacles identified to the use of clozapine is the lack of appropriate training for physicians: we highlighted the need to develop specific professional programs to train clinicians, both practicing and in residency, on the relevance and efficacy of clozapine in TRS treatment, initiation, maintenance, and management of potential adverse events. This approach would facilitate physicians to identify eligible patients and offer clozapine as a treatment option in the early stage of the disease. We also noted that increasing awareness of the benefits of clozapine among healthcare professionals, people with TRS, and their caregivers can help promote the use of clozapine. Educational material, such as leaflets or videos, could be developed and distributed to achieve this goal. The information provided in this article may be useful to improve disease burden and support healthcare professionals, patients, and caregivers navigating the complex pathways to TRS management., Competing Interests: Declaration of competing interest BCF declares that has received speaking honoraria (advisory board and educational lectures) and travel expenses from Rovi, Takeda, Menarini, Angelini, Teva, Otsuka, Lundbeck, Boehringer-Ingelheim and Johnson and Johnson. BC-F has also received fundings to support independent research initiatives (i.e., investigator initiated trials) from Lundbeck and also declares that has received funds from Viatris to conduct this study. CUC has been a consultant and/or advisor to or has received honoraria from: AbbVie, Acadia, Adock Ingram, Alkermes, Allergan, Angelini, Aristo, Biogen, Boehringer-Ingelheim, Bristol-Meyers Squibb, Car dio Diagnostics, Cerevel, CNX Therapeutics, Compass Pathways, Darnitsa, Delpor, Denovo, Gedeon Richter, Hikma, Holmusk, IntraCellular Therapies, Jamjoom Pharma, Janssen/J&J, Karuna, LB Pharma, Lundbeck, MedAvante-ProPhase, MedInCell, Merck, Mindpax, Mitsubishi Tanabe Pharma, Mylan, Neurocrine, Neurelis, Newron, Noven, Novo Nordisk, Otsuka, Pharmabrain, PPD Biotech, Recordati, Relmada, Reviva, Rovi, Sage, Seqirus, SK Life Science, Sumitomo Pharma America, Sunovion, Sun Pharma, Supernus, Tabuk, Takeda, Teva, Tolmar, Vertex, and Viatris. He provided expert testimony for Janssen and Otsuka. He served on a Data Safety Monitoring Board for Compass Pathways, Denovo, Lundbeck, Relmada, Reviva, Rovi, Supernus, and Teva. He has received grant support from Janssen and Takeda. He received royalties from UpToDate and is also a stock option holder of Cardio Diagnostics, Kuleon Biosciences, LB Pharma, Mindpax, and Quantic. ODH has received investigator-initiated research funding from and/or participated in advisory/ speaker meetings organised by Angellini, Autifony, Biogen, Boehringer-Ingelheim, Eli Lilly, Elysium, Heptares, Global Medical Education, Invicro, Jansenn, Karuna, Lundbeck, Merck, Neurocrine, Ontrack/ Pangea, Otsuka, Sunovion, Recordati, Roche, Rovi and Viatris/ Mylan. He was previously a part-time employee of Lundbeck A/v. Dr Howes has a patent for the use of dopaminergic imaging. NS is a part time medical consultant for Viatris, Finland. He is also a Medical Advisor for Janssen-Cilag, Finland and Recordati, Sweden, and received lecture fees from Viatris, Finland; Otsuka, Finland. ADB has received unrestricted research support from Janssen, Lundbeck, and Otsuka and lecture honoraria for unrestricted educational meeting from Janssen, Chiesi, Lundbeck, Roche, Sunovion, Mylan, Viatris, Recordati, Angelini and Takeda; he has served on advisory boards for Eli Lilly, Jansen, Lundbeck, Otsuka, Roche, and Takeda, Chiesi, Recordati, Angelini, Viatris, Iqvia, Idorsia. OA acts as advisor and consultant for Janssen-Ortho (Johnson & Johnson); Otsuka; Lundbeck; Allergan/Abbvie; Mylan/Viatris; Teva; he is involved in speaking engagements for Janssen-Ortho (Johnson & Johnson); Lundbeck, Otsuka, Mylan/Viatris; HLS Therapeutics; Allergan/Abbvie; he holds research contracts with Janssen-Ortho (Johnson & Johnson); Lundbeck; Otsuka; Boehringer Ingelheim. AF is a consultant and/or speaker and/or has received research grants from Angelini, Apsen, Biogen, Boheringer Ingelheim, Idorsia Italfarmaco, Lundbeck, Janssen, Medicamenta, Mylan, Otsuka, Pfizer, Recordati, Sunovion, Viatris., (Copyright © 2024. Published by Elsevier B.V.)
- Published
- 2024
- Full Text
- View/download PDF
25. Placebo effects in randomized trials of pharmacological and neurostimulation interventions for mental disorders: An umbrella review.
- Author
-
Huneke NTM, Amin J, Baldwin DS, Bellato A, Brandt V, Chamberlain SR, Correll CU, Eudave L, Garner M, Gosling CJ, Hill CM, Hou R, Howes OD, Ioannidis K, Köhler-Forsberg O, Marzulli L, Reed C, Sinclair JMA, Singh S, Solmi M, and Cortese S
- Abstract
There is a growing literature exploring the placebo response within specific mental disorders, but no overarching quantitative synthesis of this research has analyzed evidence across mental disorders. We carried out an umbrella review of meta-analyses of randomized controlled trials (RCTs) of biological treatments (pharmacotherapy or neurostimulation) for mental disorders. We explored whether placebo effect size differs across distinct disorders, and the correlates of increased placebo effects. Based on a pre-registered protocol, we searched Medline, PsycInfo, EMBASE, and Web of Knowledge up to 23.10.2022 for systematic reviews and/or meta-analyses reporting placebo effect sizes in psychopharmacological or neurostimulation RCTs. Twenty meta-analyses, summarising 1,691 RCTs involving 261,730 patients, were included. Placebo effect size varied, and was large in alcohol use disorder (g = 0.90, 95% CI [0.70, 1.09]), depression (g = 1.10, 95% CI [1.06, 1.15]), restless legs syndrome (g = 1.41, 95% CI [1.25, 1.56]), and generalized anxiety disorder (d = 1.85, 95% CI [1.61, 2.09]). Placebo effect size was small-to-medium in obsessive-compulsive disorder (d = 0.32, 95% CI [0.22, 0.41]), primary insomnia (g = 0.35, 95% CI [0.28, 0.42]), and schizophrenia spectrum disorders (standardized mean change = 0.33, 95% CI [0.22, 0.44]). Correlates of larger placebo response in multiple mental disorders included later publication year (opposite finding for ADHD), younger age, more trial sites, larger sample size, increased baseline severity, and larger active treatment effect size. Most (18 of 20) meta-analyses were judged 'low' quality as per AMSTAR-2. Placebo effect sizes varied substantially across mental disorders. Future research should explore the sources of this variation. We identified important gaps in the literature, with no eligible systematic reviews/meta-analyses of placebo response in stress-related disorders, eating disorders, behavioural addictions, or bipolar mania., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
26. Neuromelanin-Sensitive MRI: A Biomarker for Treatment-Resistant Schizophrenia?
- Author
-
Vano LJ, Veronese M, McCutcheon RA, and Howes OD
- Subjects
- Humans, Antipsychotic Agents therapeutic use, Drug Resistance, Brain diagnostic imaging, Brain metabolism, Schizophrenia metabolism, Schizophrenia diagnostic imaging, Magnetic Resonance Imaging methods, Melanins metabolism, Biomarkers metabolism
- Published
- 2024
- Full Text
- View/download PDF
27. Correction: Sub-Chronic Ketamine Administration Increases Dopamine Synthesis Capacity in the Mouse Midbrain: a Preclinical In Vivo PET Study.
- Author
-
Petty A, Garcia-Hidalgo A, Halff EF, Natesan S, Withers DJ, Irvine EE, Kokkinou M, Wells LA, Bonsall DR, Tang SP, Veronese M, and Howes OD
- Published
- 2024
- Full Text
- View/download PDF
28. Dopamine signaling enriched striatal gene set predicts striatal dopamine synthesis and physiological activity in vivo.
- Author
-
Sportelli L, Eisenberg DP, Passiatore R, D'Ambrosio E, Antonucci LA, Bettina JS, Chen Q, Goldman AL, Gregory MD, Griffiths K, Hyde TM, Kleinman JE, Pardiñas AF, Parihar M, Popolizio T, Rampino A, Shin JH, Veronese M, Ulrich WS, Zink CF, Bertolino A, Howes OD, Berman KF, Weinberger DR, and Pergola G
- Subjects
- Humans, Male, Female, Adult, Caudate Nucleus metabolism, Signal Transduction, Middle Aged, Hippocampus metabolism, Multifactorial Inheritance, Genetic Predisposition to Disease, Dorsolateral Prefrontal Cortex metabolism, Reward, Dopamine metabolism, Dopamine biosynthesis, Schizophrenia genetics, Schizophrenia metabolism, Corpus Striatum metabolism
- Abstract
The polygenic architecture of schizophrenia implicates several molecular pathways involved in synaptic function. However, it is unclear how polygenic risk funnels through these pathways to translate into syndromic illness. Using tensor decomposition, we analyze gene co-expression in the caudate nucleus, hippocampus, and dorsolateral prefrontal cortex of post-mortem brain samples from 358 individuals. We identify a set of genes predominantly expressed in the caudate nucleus and associated with both clinical state and genetic risk for schizophrenia that shows dopaminergic selectivity. A higher polygenic risk score for schizophrenia parsed by this set of genes predicts greater dopamine synthesis in the striatum and greater striatal activation during reward anticipation. These results translate dopamine-linked genetic risk variation into in vivo neurochemical and hemodynamic phenotypes in the striatum that have long been implicated in the pathophysiology of schizophrenia., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
29. Mitochondrial complex I density is associated with IQ and cognition in cognitively healthy adults: an in vivo [ 18 F]BCPP-EF PET study.
- Author
-
Shatalina E, Whitehurst TS, Onwordi EC, Gilbert BJ, Rizzo G, Whittington A, Mansur A, Tsukada H, Marques TR, Natesan S, Rabiner EA, Wall MB, and Howes OD
- Abstract
Background: Mitochondrial function plays a key role in regulating neurotransmission and may contribute to general intelligence. Mitochondrial complex I (MC-I) is the largest enzyme of the respiratory chain. Recently, it has become possible to measure MC-I distribution in vivo, using a novel positron emission tomography tracer [
18 F]BCPP-EF, thus, we set out to investigate the association between MC-I distribution and measures of cognitive function in the living healthy brain., Results: Analyses were performed in a voxel-wise manner and identified significant associations between [18 F]BCPP-EF DVRCS-1 in the precentral gyrus and parietal lobes and WAIS-IV predicted IQ, WAIS-IV arithmetic and WAIS-IV symbol-digit substitution scores (voxel-wise Pearson's correlation coefficients transformed to Z-scores, thresholded at Z = 2.3 family-wise cluster correction at p < 0.05, n = 16). Arithmetic scores were associated with middle frontal and post-central gyri tracer uptake, symbol-digit substitution scores were associated with precentral gyrus tracer uptake. RAVLT recognition scores were associated with [18 F]BCPP-EF DVRCS-1 in the middle frontal gyrus, post-central gyrus, occipital and parietal regions (n = 20)., Conclusions: Taken together, our findings support the theory that mitochondrial function may contribute to general intelligence and indicate that interindividual differences in MC-I should be a key consideration for research into mitochondrial dysfunction in conditions with cognitive impairment., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
30. Trace amine-associated receptor 1 (TAAR1) agonism for psychosis: a living systematic review and meta-analysis of human and non-human data.
- Author
-
Siafis S, Chiocchia V, Macleod MR, Austin C, Homiar A, Tinsdeall F, Friedrich C, Ramage FJ, Kennett J, Nomura N, Maksym O, Rutigliano G, Vano LJ, McCutcheon RA, Gilbert D, Ostinelli EG, Stansfield C, Dehdarirad H, Juma DO, Wright S, Simple O, Elugbadebo O, Tonia T, Mantas I, Howes OD, Furukawa TA, Milligan L, Moreno C, Elliott JH, Hastings J, Thomas J, Michie S, Sena ES, Seedat S, Egger M, Potts J, Cipriani A, Salanti G, and Leucht S
- Abstract
Background: Trace amine-associated receptor 1 (TAAR1) agonism shows promise for treating psychosis, prompting us to synthesise data from human and non-human studies., Methods: We co-produced a living systematic review of controlled studies examining TAAR1 agonists in individuals (with or without psychosis/schizophrenia) and relevant animal models. Two independent reviewers identified studies in multiple electronic databases (until 17.11.2023), extracted data, and assessed risk of bias. Primary outcomes were standardised mean differences (SMD) for overall symptoms in human studies and hyperlocomotion in animal models. We also examined adverse events and neurotransmitter signalling. We synthesised data with random-effects meta-analyses., Results: Nine randomised trials provided data for two TAAR1 agonists (ulotaront and ralmitaront), and 15 animal studies for 10 TAAR1 agonists. Ulotaront and ralmitaront demonstrated few differences compared to placebo in improving overall symptoms in adults with acute schizophrenia (N=4 studies, n=1291 participants; SMD=0.15, 95%CI: -0.05, 0.34), and ralmitaront was less efficacious than risperidone (N=1, n=156, SMD=-0.53, 95%CI: -0.86, -0.20). Large placebo response was observed in ulotaront phase-III trials. Limited evidence suggested a relatively benign side-effect profile for TAAR1 agonists, although nausea and sedation were common after a single dose of ulotaront. In animal studies, TAAR1 agonists improved hyperlocomotion compared to control (N=13 studies, k=41 experiments, SMD=1.01, 95%CI: 0.74, 1.27), but seemed less efficacious compared to dopamine D
2 receptor antagonists (N=4, k=7, SMD=-0.62, 95%CI: -1.32, 0.08). Limited human and animal data indicated that TAAR1 agonists may regulate presynaptic dopaminergic signalling., Conclusions: TAAR1 agonists may be less efficacious than dopamine D2 receptor antagonists already licensed for schizophrenia. The results are preliminary due to the limited number of drugs examined, lack of longer-term data, publication bias, and assay sensitivity concerns in trials associated with large placebo response. Considering their unique mechanism of action, relatively benign side-effect profile and ongoing drug development, further research is warranted., Registration: PROSPERO-ID: CRD42023451628., Competing Interests: Competing interests: Andrea Cipriani received research, educational and consultancy fees from the Italian Network for Paediatric Trials, CARIPLO Foundation, Lundbeck, and Angelini Pharma. Toshi A. Furukawa reports personal fees from Boehringer-Ingelheim, Daiichi Sankyo, DT Axis, Kyoto University Original, Shionogi, and SONY, and a grant from Shionogi outside the submitted work; in addition, TAF has patents 2020-548587 and 2022-082495 pending, and intellectual properties for Kokoro-app licensed to Mitsubishi-Tanabe. Oliver D. Howes has received investigator-initiated research funding from and/or participated in advisory/speaker meetings organised by Angellini, Autifony, Biogen, Boehringer-Ingelheim, Eli Lilly, Elysium, Heptares, Global Medical Education, Invicro, Jansenn, Karuna, Lundbeck, Merck, Neurocrine, Ontrack/ Pangea, Otsuka, Sunovion, Recordati, Roche, Rovi and Viatris/ Mylan. He was previously a part-time employee of Lundbeck A/v. Dr Howes has a patent for the use of dopaminergic imaging. In the last three years Stefan Leucht has received honoraria for advising/consulting and/or for lectures and/or for educational material from Angelini, Boehringer Ingelheim, Eisai, Ekademia, GedeonRichter, Janssen, Karuna, Kynexis, Lundbeck, Medichem, Medscape, Mitsubishi, Otsuka, NovoNordisk, Recordati, Rovi, Teva. Robert A. McCutcheon has received speaker/consultancy fees from Karuna, Janssen, Boehringer Ingelheim, and Otsuka, and is a director of a company that hosts psychotropic prescribing decision tools. Carmen Moreno received honoraria as a consultant and/or advisor and/or for lectures from Angelini, Compass, Esteve, Exeltis Janssen, Lundbeck, Neuraxpharm, Nuvelution, Otsuka, Pfizer, Servier and Sunovion outside the submitted work. Nobuyuki Nomura has received speaker fees from Eisai, Meiji Seika Pharma, Otsuka, and Sumitomo Pharma; and manuscript fees from Sumitomo Pharma. Edoardo G. Ostinelli has received research and consultancy fees from Angelini Pharma. No other competing interests were disclosed., (Copyright: © 2024 Siafis S et al.)- Published
- 2024
- Full Text
- View/download PDF
31. Synaptic Terminal Density Early in the Course of Schizophrenia: An In Vivo UCB-J Positron Emission Tomographic Imaging Study of SV2A.
- Author
-
Onwordi EC, Whitehurst T, Shatalina E, Mansur A, Arumuham A, Osugo M, Marques TR, Jauhar S, Gupta S, Mehrotra R, Rabiner EA, Gunn RN, Natesan S, and Howes OD
- Subjects
- Humans, Presynaptic Terminals metabolism, Electrons, Pyridines, Membrane Glycoproteins metabolism, Positron-Emission Tomography methods, Brain diagnostic imaging, Brain metabolism, Nerve Tissue Proteins metabolism, Pyrrolidinones, Schizophrenia diagnostic imaging
- Abstract
Background: The synaptic hypothesis is an influential theory of the pathoetiology of schizophrenia (SCZ), which is supported by the finding that there is lower uptake of the synaptic terminal density marker [
11 C]UCB-J in patients with chronic SCZ than in control participants. However, it is unclear whether these differences are present early in the illness. To address this, we investigated [11 C]UCB-J volume of distribution (VT ) in antipsychotic-naïve/free patients with SCZ who were recruited from first-episode services compared with healthy volunteers., Methods: Forty-two volunteers (SCZ n = 21, healthy volunteers n = 21) underwent [11 C]UCB-J positron emission tomography to index [11 C]UCB-J VT and distribution volume ratio in the anterior cingulate, frontal, and dorsolateral prefrontal cortices; the temporal, parietal and occipital lobes; and the hippocampus, thalamus, and amygdala. Symptom severity was assessed in the SCZ group using the Positive and Negative Syndrome Scale., Results: We found no significant effects of group on [11 C]UCB-J VT or distribution volume ratio in most regions of interest (effect sizes from d = 0.0-0.7, p > .05), with two exceptions: we found lower distribution volume ratio in the temporal lobe (d = 0.7, uncorrected p < .05) and lower VT /fp in the anterior cingulate cortex in patients (d = 0.7, uncorrected p < .05). The Positive and Negative Syndrome Scale total score was negatively associated with [11 C]UCB-J VT in the hippocampus in the SCZ group (r = -0.48, p = .03)., Conclusions: These findings indicate that large differences in synaptic terminal density are not present early in SCZ, although there may be more subtle effects. When taken together with previous evidence of lower [11 C]UCB-J VT in patients with chronic illness, this may indicate synaptic density changes during the course of SCZ., (Copyright © 2023 Society of Biological Psychiatry. Published by Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
32. Corrigendum: The effect of antipsychotics on glutamate levels in the anterior cingulate cortex and clinical response: a 1 H-MRS study in first-episode psychosis patients.
- Author
-
Zahid U, McCutcheon RA, Borgan F, Jauhar S, Pepper F, Nour MM, Rogdaki M, Osugo M, Murray GK, Hathway P, Murray RM, Egerton A, and Howes OD
- Abstract
[This corrects the article DOI: 10.3389/fpsyt.2022.967941.]., (Copyright © 2024 Zahid, McCutcheon, Borgan, Jauhar, Pepper, Nour, Rogdaki, Osugo, Murray, Hathway, Murray, Egerton and Howes.)
- Published
- 2024
- Full Text
- View/download PDF
33. Gamma Oscillations and Potassium Channel Modulation in Schizophrenia: Targeting GABAergic Dysfunction.
- Author
-
Kaar SJ, Nottage JF, Angelescu I, Marques TR, and Howes OD
- Subjects
- Humans, Electroencephalography, Interneurons physiology, Potassium pharmacology, Potassium Channels pharmacology, Potassium Channels physiology, Schizophrenia
- Abstract
Impairments in gamma-aminobutyric acid (GABAergic) interneuron function lead to gamma power abnormalities and are thought to underlie symptoms in people with schizophrenia. Voltage-gated potassium 3.1 (Kv3.1) and 3.2 (Kv3.2) channels on GABAergic interneurons are critical to the generation of gamma oscillations suggesting that targeting Kv3.1/3.2 could augment GABAergic function and modulate gamma oscillation generation. Here, we studied the effect of a novel potassium Kv3.1/3.2 channel modulator, AUT00206, on resting state frontal gamma power in people with schizophrenia. We found a significant positive correlation between frontal resting gamma (35-45 Hz) power ( n = 22, r = 0.613, P < .002) and positive and negative syndrome scale (PANSS) positive symptom severity. We also found a significant reduction in frontal gamma power ( t
13 = 3.635, P = .003) from baseline in patients who received AUT00206. This provides initial evidence that the Kv3.1/3.2 potassium channel modulator, AUT00206, may address gamma oscillation abnormalities in schizophrenia., Competing Interests: Declaration of Conflicting InterestsThe author(s) declared the following potential conflicts of interest with respect to the research, authorship, and/or publication of this article: SK received travel expenses to attend a scientific meeting arranged by Autifony. IA and JN have nothing to disclose. TRM has received honoraria for speaking and chairing engagements from Lundbeck, Janssen, and Astellas. OH is a part-time employee of H. Lundbeck A/S and has received investigator-initiated research funding from and/or participated in advisory/speaker meetings organised by Angellini, Autifony, Biogen, Boehringer-Ingelheim, Eli Lilly, Heptares, Global Medical Education, Invicro, Jansenn, Lundbeck, Neurocrine, Otsuka, Sunovion, Rand, Recordati, Roche and Viatris/ Mylan. Dr Howes has a patent for the use of dopaminergic imaging.- Published
- 2024
- Full Text
- View/download PDF
34. Histamine-3 Receptor Availability and Glutamate Levels in the Brain: A PET-1H-MRS Study of Patients With Schizophrenia and Healthy Controls.
- Author
-
Arumuham A, Nour MM, Veronese M, Beck K, Onwordi EC, Lythgoe DJ, Jauhar S, Rabiner EA, and Howes OD
- Subjects
- Humans, Histamine, Proton Magnetic Resonance Spectroscopy methods, Cross-Sectional Studies, Brain diagnostic imaging, Positron-Emission Tomography, Gyrus Cinguli, Glutamine, Glutamic Acid, Schizophrenia diagnostic imaging, Schizophrenia drug therapy
- Abstract
Background: The histamine-3 receptor (H3R) may have a role in cognitive processes through its action as a presynaptic heteroreceptor inhibiting the release of glutamate in the brain. To explore this, we examined anterior cingulate cortex (ACC) and striatum H3R availability in patients with schizophrenia and characterized their relationships with glutamate levels in corresponding brain regions., Methods: We employed a cross-sectional study, recruiting 12 patients with schizophrenia and 12 healthy volunteers. Participants underwent positron emission tomography using the H3R-specific radio ligand [11C]MK-8278, followed by proton magnetic resonance spectroscopy to measure glutamate levels, recorded as Glu and Glx. Based on existing literature, the ACC and striatum were selected as regions of interest., Results: We found significant inverse relationships between tracer uptake and Glu (r = -0.66, P = .02) and Glx (r = -0.62, P = .04) levels in the ACC of patients, which were absent in healthy volunteers (Glu: r = -0.19, P = .56, Glx: r = 0.10, P = .75). We also found a significant difference in striatal (F1,20 = 6.00, P = .02) and ACC (F1,19 = 4.75, P = .04) Glx levels between groups., Conclusions: These results provide evidence of a regionally specific relationship between H3Rs and glutamate levels, which builds on existing preclinical literature. Our findings add to a growing literature indicating H3Rs may be a promising treatment target in schizophrenia, particularly for cognitive impairment, which has been associated with altered glutamate signaling., (© The Author(s) 2024. Published by Oxford University Press on behalf of CINP.)
- Published
- 2024
- Full Text
- View/download PDF
35. Immune response to vaccination in people with psychotic disorders relative to healthy controls: prospective study of SARS-CoV-2 vaccination.
- Author
-
O'Brien O, Arumuham A, Mizuno Y, Baxter L, Lobo M, Parmar S, Jolles S, and Howes OD
- Abstract
This prospective study examines the immune response to SARS-CoV-2 vaccination in patients with psychotic disorders compared with healthy volunteers. Participants were recruited naturalistically as part of the UK's COVID-19 vaccination programme. Prior to receiving their first COVID-19 vaccine, blood samples were provided by participants to examine anti-SARS-CoV-2 immunoglobulins (IgG) at baseline, followed by a repeat assay 1 month after receiving their first vaccine to assess vaccine response. The increase of IgG levels from baseline to 1 month post-vaccination was significantly lower in patients compared with controls, supporting evidence of impaired vaccine response in people with psychotic disorders. When excluding patients treated with clozapine from the analysis, this difference was no longer significant, suggesting that effects may be particularly marked in people taking clozapine.
- Published
- 2024
- Full Text
- View/download PDF
36. How to classify antipsychotics: time to ditch dichotomies?
- Author
-
McCutcheon RA, Cannon A, Parmer S, and Howes OD
- Subjects
- Humans, Antipsychotic Agents therapeutic use, Schizophrenia drug therapy
- Abstract
The dichotomies of 'typical/atypical' or 'first/second generation' have been employed for several decades to classify antipsychotics, but justification for their use is not clear. In the current analysis we argue that this classification is flawed from both clinical and pharmacological perspectives. We then consider what approach should ideally be employed in both clinical and research settings.
- Published
- 2024
- Full Text
- View/download PDF
37. Schizophrenia: from neurochemistry to circuits, symptoms and treatments.
- Author
-
Howes OD, Bukala BR, and Beck K
- Subjects
- Humans, Dopamine therapeutic use, Glutamic Acid, Schizophrenia therapy, Neurochemistry, Psychotic Disorders etiology, Psychotic Disorders therapy
- Abstract
Schizophrenia is a leading cause of global disability. Current pharmacotherapy for the disease predominantly uses one mechanism - dopamine D2 receptor blockade - but often shows limited efficacy and poor tolerability. These limitations highlight the need to better understand the aetiology of the disease to aid the development of alternative therapeutic approaches. Here, we review the latest meta-analyses and other findings on the neurobiology of prodromal, first-episode and chronic schizophrenia, and the link to psychotic symptoms, focusing on imaging evidence from people with the disorder. This evidence demonstrates regionally specific neurotransmitter alterations, including higher glutamate and dopamine measures in the basal ganglia, and lower glutamate, dopamine and γ-aminobutyric acid (GABA) levels in cortical regions, particularly the frontal cortex, relative to healthy individuals. We consider how dysfunction in cortico-thalamo-striatal-midbrain circuits might alter brain information processing to underlie psychotic symptoms. Finally, we discuss the implications of these findings for developing new, mechanistically based treatments and precision medicine for psychotic symptoms, as well as negative and cognitive symptoms., (© 2023. Springer Nature Limited.)
- Published
- 2024
- Full Text
- View/download PDF
38. Relapse in clinically stable adult patients with schizophrenia or schizoaffective disorder: evidence-based criteria derived by equipercentile linking and diagnostic test accuracy meta-analysis.
- Author
-
Siafis S, Brandt L, McCutcheon RA, Gutwinski S, Schneider-Thoma J, Bighelli I, Kane JM, Arango C, Kahn RS, Fleischhacker WW, McGorry P, Carpenter WT, Falkai P, Hasan A, Marder SR, Schooler N, Engel RR, Honer WG, Buchanan RW, Davidson M, Weiser M, Priller J, Davis JM, Howes OD, Correll CU, and Leucht S
- Subjects
- Adult, Male, Female, Humans, Psychiatric Status Rating Scales, Diagnostic Tests, Routine, Antipsychotic Agents therapeutic use, Schizophrenia diagnosis, Schizophrenia drug therapy, Psychotic Disorders psychology
- Abstract
Background: There is no consensus on defining relapse in schizophrenia, and scale-derived criteria with unclear clinical relevance are widely used. We aimed to develop an evidence-based scale-derived set of criteria to define relapse in patients with schizophrenia or schizoaffective disorder., Methods: We searched the Yale University Open Data Access (YODA) for randomised controlled trials (RCTs) in clinically stable adults with schizophrenia or schizoaffective disorder, and obtained individual participant data on Positive and Negative Syndrome Scale (PANSS), Clinical Global Impression Severity (CGI-S), Personal and Social Performance (PSP), and Social and Occupational Functioning Assessment Scale (SOFAS). Our main outcomes were PANSS-derived criteria based on worsening in PANSS total score. We examined their relevance using equipercentile linking with CGI-S and functioning scales, and their test-performance in defining relapse with diagnostic test accuracy meta-analysis against CGI-S worsening (≥1-point increase together with a score ≥4 points) and psychiatric hospitalisation., Findings: Based on data from seven RCTs (2354 participants; 1348 men [57·3%] and 1006 women [42·7%], mean age of 39·5 years [SD 12·0, range 17-89]; 303 Asian [12.9%], 255 Black [10.8%], 1665 White [70.7%], and other or unspecified 131 [5.6%]), an increase of 12 points or more in PANSS total (range 30-210 points) corresponded to clinically important deterioration in global severity of illness (≥1 point increase in CGI-S, range 1-7) and functioning (≥10 points decline in PSP or SOFAS, range 1-100). The interpretation of percentage changes varied importantly across different baseline scores. An increase of 12 points or more in PANSS total had good sensitivity and specificity using CGI-S as reference standard (sensitivity 82·1% [95% CI 77·1-86·4], specificity 86·9% [82·9-90·3]), as well as good sensitivity but lower specificity compared to hospitalisation (sensitivity 81·7% [74·1-87·7], specificity 69·2% [60·5-76·9]). Requiring either an increase in PANSS total or in specific items for positive and disorganization symptoms further improved test-performance. Cutoffs for situations where high sensitivity or specificity is needed are presented., Interpretation: An increase of either 12 points or more in the PANSS total score, or worsening of specific positive and disorganisation symptom items could be a reasonable evidence-based definition of relapse in schizophrenia, potentially linking symptoms used to define remission and relapse. Percentage changes should not be used to define relapse because their interpretation depends on baseline scores., Funding: German Research Foundation (grant number: 428509362)., Competing Interests: Declaration of interests RAM has received speaker or consultancy fees from Karuna, Janssen, Boehringer Ingelheim, and Otsuka, and co-directs a company that designs digital resources to support treatment of mental illness. JMK has been a consultant or advisor for, or has received honoraria from, Alkermes, Allergan, LB Pharmaceuticals, H Lundbeck, Intracellular Therapies, Janssen Pharmaceuticals, Johnson & Johnson, Merck, Minerva, Neurocrine, Newron, Otsuka, Pierre Fabre, Reviva, Roche, Sumitomo Dainippon, Sunovion, Takeda, Teva, and UpToDate, and is a shareholder in LB Pharmaceuticals and Vanguard Research Group. CA has been a consultant to, or has received honoraria or grants from, Acadia, Abbot, Ambrosetti, AMGEN, AstraZeneca, Bristol-Myers Squibb, Dainippon Sumitomo Pharma, Forum, Gedeon Richter, Janssen Cilag, Lundbeck, Merck, Otsuka, Pfizer, Roche, Servier, Shire, Schering Plough, Sunovion, and Takeda. WWF has received consulting fees from Boehringer Ingelheim, Angelini, Richter, and Recordati, and grant support from Lundbeck and Otsuka. PF received paid speakership from Otsuka, Recordati, Richter, Rovi, and Janssen, and is a member of advisory boards of Richter, Rovi, and Janssen. AH has received paid speakerships from AbbVie, Janssen, Otsuka, Rercordati, and Lundbeck. He was member of Rovi, Recordati, Otsuka, Lundbeck, and Janssen advisory boards. SRM has received consulting fees from Boehringer-Ingelheim, H. Lundbeck, Otsuka, Takeda, Teva, Roche, Genentech, Targacept, Forum, AbbVie, Allergan, and Neurocrine. He has received research support from Boehringer-Ingelheim, Takeda, and Neurocrine. NS has been a consultant or advisor to, or has received honoraria, from Alkermes, Lundbeck, Otsuka, Otsuka Canada, and Teva. WGH has received consulting fees or sat on paid advisory boards for In Silico Biosciences, Translational Life Sciences, Newron, Otsuka, and AbbVie. RWB is a data and safety monitoring board member for Merck, Newron, and Roche; on the advisory board for Acadia, Boehringer Ingelheim, Karuna, Neurocrine, and Roche; and consultant for Boehringer Ingelheim. MD is an employee of Minerva Neurosciences. MW He has received advisory board, speaker's, or consultant fees from, and owns stock, with Acadia, Dexcel, Janssen, Lundbeck, Minerva, Pfizer, Roche, and Teva. OH was a part-time employee of H. Lundbeck, and has received investigator-initiated research funding from, or participated in, advisory or speaker meetings organised by Angellini, Autifony, Biogen, Boehringer-Ingelheim, Eli Lilly, Heptares, Global Medical Education, Invicro, Jansenn, Lundbeck, Neurocrine, Otsuka, Sunovion, Rand, Recordati, Roche, and Viatris/Mylan. Neither he nor his family have holdings/a financial stake in any pharmaceutical company. He has a patent for the use of dopaminergic imaging. CUC has been a consultant or advisor to, or has received honoraria from, AbbVie, Acadia, Alkermes, Allergan, Angelini, Aristo, Axsome, Cardio Diagnostics, Compass, Damitsa, Gedeon Richter, Hikma, Holmusk, IntraCellular Therapies, Janssen/J&J, Karuna, LB Pharma, Lundbeck, MedAvante-ProPhase, MedInCell, Medscape, Merck, Mindpax, Mitsubishi Tanabe Pharma, Mylan, Neurocrine, Noven, Otsuka, Pfizer, Recordati, Relmada, Reviva, Rovi, Seqirus, Servier, SK Life Science, Sumitomo Dainippon, Sunovion, Supernus, Takeda, Teva, and Viatris. He provided expert testimony for Janssen and Otsuka. He served on a data safety monitoring board for Lundbeck, Relmada, Reviva, Rovi, and Teva. He has received grant support from Janssen and Takeda. He received royalties from UpToDate and is also a stock option holder of Cardio Diagnostics, Mindpax, LB Pharma and Quantics. In the past 3 years, SL has received honoraria as a consultant, adviser, or lecturer from Alkermes, Angelini, Eisai, Gedeon Richter, Janssen, Lundbeck, Lundbeck Institute, Merck Sharpp and Dome, Otsuka, Recordati, Rovi, Sanofi Aventis, TEVA, Medichem, and Mitsubishi. All other authors declare no competing interests., (Copyright © 2024 Elsevier Ltd. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
39. Dopaminergic-related Anatomical Pattern of Dorsal Striatum in Schizophrenia.
- Author
-
Xie C, Xiang S, Zheng Y, Shen C, Peng X, Li Y, Cheng W, Chang X, Cheng J, Cui LB, Huang CC, Kuang N, Li C, Lin CP, Luo C, Tang Y, Wang J, Wu X, Yao D, Zhang J, Zhang T, Heinz A, Robbins TW, Howes OD, Schumann G, Jia T, and Feng J
- Abstract
Striatal dopaminergic overactivity was hypothesized as the core pathophysiology of schizophrenia. However, morphological alterations of striatum in schizophrenia remains exclusive, largely because brain regional heterogeneity limited traditional group-mean based approach. Leveraging third-party brain maps of neurotransmitter and cognition behaviours, we developed a pattern-based representation feature score (ReFS) to investigate structural spatial pattern variation in schizophrenia. Structural ReFS of subcortical regions, particularly the striatum, were linked to schizophrenia diagnosis, symptom severity, and genetic susceptibility. Dopaminergic-ReFS of striatum was increased in schizophrenia patients and reliably reproduced across 13 datasets. The pattern-based ReFS effectively captured the shared genetic pathways underlying both schizophrenia and striatum. The results provide convergent, multimodal suggest the central role of striatal spatial patterns in schizophrenia psychopathologies and and open new avenues to develop individualized treatments for psychotic disorders.
- Published
- 2023
- Full Text
- View/download PDF
40. Sub-Chronic Ketamine Administration Increases Dopamine Synthesis Capacity in the Mouse Midbrain: a Preclinical In Vivo PET Study.
- Author
-
Petty A, Garcia-Hidalgo A, Halff EF, Natesan S, Withers DJ, Irvine EE, Kokkinou M, Wells LA, Bonsall DR, Tang SP, Veronese M, and Howes OD
- Subjects
- Humans, Adult, Male, Animals, Mice, Positron Emission Tomography Computed Tomography, Mice, Inbred C57BL, Dihydroxyphenylalanine, Positron-Emission Tomography methods, Corpus Striatum, Mesencephalon diagnostic imaging, Dopamine, Ketamine pharmacology
- Abstract
Purpose: There is robust evidence that people with schizophrenia show elevated dopamine (DA) synthesis capacity in the striatum. This finding comes from positron emission tomography (PET) studies using radiolabelled l-3,4-dihydroxyphenylalanine (
18 F-DOPA). DA synthesis capacity also appears to be elevated in the midbrain of people with schizophrenia compared to healthy controls. We therefore aimed to optimise a method to quantify18 F-DOPA uptake in the midbrain of mice, and to utilise this method to quantify DA synthesis capacity in the midbrain of the sub-chronic ketamine model of schizophrenia-relevant hyperdopaminergia., Procedures: Adult male C57Bl6 mice were treated daily with either ketamine (30 mg/kg, i.p.) or vehicle (saline) for 5 days. On day 7, animals were administered18 F-DOPA (i.p.) and scanned in an Inveon PET/CT scanner. Data from the saline-treated group were used to optimise an atlas-based template to position the midbrain region of interest and to determine the analysis parameters which resulted in the greatest intra-group consistency. These parameters were then used to compare midbrain DA synthesis capacity (Ki Mod ) between ketamine- and saline-treated animals., Results: Using an atlas-based template to position the 3.7 mm3 midbrain ROI with a T*-Tend window of 15-140 min to estimate Ki Mod resulted in the lowest intra-group variability and moderate test-retest agreement. Using these parameters, we found that Ki Mod was elevated in the midbrain of ketamine-treated animals in comparison to saline-treated animals (t(22) = 2.19, p = 0.048). A positive correlation between DA synthesis capacity in the striatum and the midbrain was also evident in the saline-treated animals (r2 = 0.59, p = 0.005) but was absent in ketamine-treated animals (r2 = 0.004, p = 0.83)., Conclusions: Using this optimised method for quantifying18 F-DOPA uptake in the midbrain, we found that elevated striatal DA synthesis capacity in the sub-chronic ketamine model extends to the midbrain. Interestingly, the dysconnectivity between the midbrain and striatum seen in this model is also evident in the clinical population. This model may therefore be ideal for assessing novel compounds which are designed to modulate pre-synaptic DA synthesis capacity., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
41. Placebo effects in mental health disorders: protocol for an umbrella review.
- Author
-
Huneke NT, Amin J, Baldwin DS, Chamberlain SR, Correll CU, Garner M, Hill CM, Hou R, Howes OD, Sinclair JM, Solmi M, and Cortese S
- Subjects
- Humans, Placebo Effect, Systematic Reviews as Topic, Review Literature as Topic, Mental Disorders drug therapy, Mental Health
- Abstract
Introduction: Given the high prevalence of mental health disorders and their significant socioeconomic burden, there is a need to develop improved treatments, and to evaluate them through placebo-controlled trials. However, the magnitude of the placebo response in randomised controlled trials to test medications may be substantial, affecting their interpretation. Therefore, improved understanding of the patient, trial and mental disorder factors that influence placebo responses would inform clinical trial design to better detect active treatment effects. There is a growing literature exploring the placebo response within specific mental health disorders, but no overarching synthesis of this research has been produced to date. We present a protocol for an umbrella review of systematic reviews and/or meta-analyses in which we aim to understand the effect size and potential predictors of placebo response within, and across, mental health disorders., Methods and Analysis: We will systematically search databases (Medline, PsycINFO, EMBASE+EMBASE Classic, Web of Knowledge) for systematic reviews and/or meta-analyses that report placebo effect size in clinical trials in patients with mental health disorders (initial search date 23 October 2022). Screening of abstracts and full texts will be done in pairs. We will extract data to qualitatively examine how placebo effect size varies across mental health disorders. We also plan to qualitatively summarise predictors of increased placebo response identified either quantitatively (eg, through meta-regression) or qualitatively. Risk of bias will be assessed using the AMSTAR-2 tool. We aim to not only summarise the current literature but also to identify gaps in knowledge and generate further hypotheses., Ethics and Dissemination: We do not believe there are any specific ethical considerations relevant to this study. We will publish the results in a peer-reviewed journal., Competing Interests: Competing interests: DSB is President of the British Association for Psychopharmacology (BAP) and Editor of Human Psychopharmacology journal (for which he receives an Editor’s honorarium). CMH has acted on an expert advisory board for Neurim pharmaceuticals. ODH is a part-time employee and stock holder of Lundbeck A/s. He has received investigator-initiated research funding from and/or participated in advisory/speaker meetings organised by Angellini, Autifony, Biogen, Boehringer-Ingelheim, Eli Lilly, Heptares, Global Medical Education, Invicro, Jansenn, Lundbeck, Neurocrine, Otsuka, Sunovion, Recordati, Roche and Viatris/Mylan. ODH has a patent for the use of dopaminergic imaging. All other authors declare no competing interests. MS has received honoraria/has been a consultant for Angelini, Lundbeck, Otsuka. SRC receives an honorarium from Elsevier for associate editor work at two academic journals. SCo has received honoraria from non-profit associations (BAP, ACAMH, CADDRA) for educational activities. CUC has been a consultant and/or advisor to or has received honoraria from: AbbVie, Acadia, Adock Ingram, Alkermes, Allergan, Angelini, Aristo, Biogen, Boehringer-Ingelheim, Bristol-Meyers Squibb, Cardio Diagnostics, Cerevel, CNX Therapeutics, Compass Pathways, Darnitsa, Denovo, Gedeon Richter, Hikma, Holmusk, IntraCellular Therapies, Jamjoom Pharma, Janssen/J&J, Karuna, LB Pharma, Lundbeck, MedAvante-ProPhase, MedInCell, Merck, Mindpax, Mitsubishi Tanabe Pharma, Mylan, Neurocrine, Neurelis, Newron, Noven, Novo Nordisk, Otsuka, Pharmabrain, PPD Biotech, Recordati, Relmada, Reviva, Rovi, Sage, Seqirus, SK Life Science, Sumitomo Pharma America, Sunovion, Sun Pharma, Supernus, Takeda, Teva, Tolmar, Vertex and Viatris. He provided expert testimony for Janssen and Otsuka. He served on a Data Safety Monitoring Board for Compass Pathways, Denovo, Lundbeck, Relmada, Reviva, Rovi, Supernus and Teva. He has received grant support from Janssen and Takeda. He received royalties from UpToDate and is also a stock option holder of Cardio Diagnostics, Kuleon Biosciences, LB Pharma, Mindpax and Quantic., (© Author(s) (or their employer(s)) 2023. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2023
- Full Text
- View/download PDF
42. Antidepressant and antipsychotic side-effects and personalised prescribing: a systematic review and digital tool development.
- Author
-
Pillinger T, Howes OD, Correll CU, Leucht S, Huhn M, Schneider-Thoma J, Gaughran F, Jauhar S, McGuire PK, Taylor DM, Young AH, and McCutcheon RA
- Subjects
- Humans, Depressive Disorder, Major drug therapy, Precision Medicine, Drug-Related Side Effects and Adverse Reactions, Antipsychotic Agents adverse effects, Antipsychotic Agents therapeutic use, Antidepressive Agents adverse effects, Antidepressive Agents therapeutic use, Schizophrenia drug therapy
- Abstract
Background: Side-effects of psychiatric medication impair quality of life and functioning. Furthermore, they contribute to morbidity, mortality, stigma, and poor treatment concordance resulting in relapse of psychiatric illness. Guidelines recommend discussing side-effects with patients when making treatment decisions, but a synthesis of antidepressant and antipsychotic side-effects to guide this process is missing, and considering all side-effects is a complex, multidimensional process. We aimed to create comprehensive databases of antipsychotic and antidepressant side-effects, and a digital tool to support database navigation., Methods: To create the databases, we did an umbrella review of Embase, PsycINFO, and MEDLINE from database inception to June 26, 2023. We included meta-analyses of randomised controlled trials examining antipsychotic monotherapy in the treatment of schizophrenia or antidepressant monotherapy in the treatment of major depressive disorder. We included meta-analyses in adults (aged ≥18 years) that assessed drugs with a common comparator. The search was complemented by a review of national and international guidelines and consensus statements for the treatment of major depressive disorder and schizophrenia in adults. Effect sizes for antipsychotic and antidepressant side-effects were extracted from meta-analyses examining the largest number of drugs. In cases of incomplete meta-analytic coverage, data were imputed on the basis of guideline-derived ordinal rankings or, if imputation was not possible, ordinal scores were extracted. Both meta-analytic and ordinal outcomes were normalised to provide values between 0 and 1. We then constructed a digital tool, the Psymatik Treatment Optimizer, to combine the side-effect databases with side-effect concerns of an individual user, to enable users to select side-effects of concern and the relative degree of concern for each side-effect. Concern weightings and the side-effect databases are synthesised via a multicriteria decision analysis method (technique for order of preference by similarity to ideal situation, or TOPSIS)., Findings: Of 3724 citations, 14 articles containing 68 meta-analyses of individual side-effects met inclusion criteria. After review of 19 guidelines, seven provided ordinal data. Antipsychotic data were extracted from five studies (11 meta-analyses, n=65 594 patients) and four guidelines, and antidepressant data were extracted from three guidelines. The resultant databases included data on 32 antipsychotics (14 side-effects) and 37 antidepressants (nine side-effects). The databases highlighted the clinical dilemma associated with balancing side-effects, with avoidance of one side-effect (eg, weight gain for antipsychotics) increasing the risk of others (eg, akathisia). To aid with this dilemma, the Psymatik Treatment Optimizer synthesises the side-effect databases with individual user-defined concern weights. After computing up to 5851 pairwise comparisons for antidepressants and 5142 pairwise comparisons for antipsychotics, Psymatik ranks treatments in order of preference for the individual user, with the output presented in a heatmap., Interpretation: By facilitating collaborative, personalised, and evidence-based prescribing decisions, the side-effect databases and digital application supports care delivery that is consistent with international regulatory guidance for the treatment of schizophrenia and depression, and it therefore has promise for informing psychiatric practice and improving outcomes., Funding: National Institute for Health and Care Research, Maudsley Charity, Wellcome Trust, Medical Research Council., Competing Interests: Declaration of interests TP has participated in educational speaker meetings organised by Lundbeck, Otsuka, Sunovion, Janssen, Schwabe Pharma, ROVI Biotech, and Recordati. RAMcC has participated in advisory or speaker meetings organised by Otsuka, Karuna, Boehringer Ingelheim, and Janssen. TP and RAMcC are directors of Pharmatik that funds website hosting for Psymatik. Neither TP nor RAMcC have holdings or financial stakes in any pharmaceutical company. MH has received honoraria for advisory boards and lectures from Recordati. SJ has participated in educational speaker meetings organised by Lundbeck, Otsuka, Sunovion, Janssen, and Boehringer Ingelheim. FG has received honoraria from Lundbeck, Otsuka, Sunovion, and Boehringer Ingelheim. ODH has received investigator-initiated research funding from or participated in advisory or speaker meetings organised by Angellini, Autifony, Biogen, Boehringer Ingelheim, Eli Lilly, Heptares, Global Medical Education, Invicro, Janssen, Lundbeck, Neurocrine, Otsuka, Sunovion, Rand, Recordati, Roche, Viatris (formerly Mylan), and ROVI Biotech. AHY has delivered paid lectures and advisory boards for the following companies: AstraZeneca, Boehringer Ingelheim, Eli Lilly, LivaNova, Lundbeck, Sunovion, Servier, Livanova, Janssen, Allegan, Bionomics, Sumitomo Dainippon Pharma, COMPASS Pathways, Sage, Novartis, and Neurocentrx. CUC has been a consultant or advisor to or has received honoraria from AbbVie, Acadia, Alkermes, Allergan, Angelini, Aristo, Boehringer Ingelheim, Cardio Diagnostics, Cerevel, CNX Therapeutics, COMPASS Pathways, Darnitsa, Gedeon Richter, Hikma, Holmusk, IntraCellular Therapies, Janssen, Johnson & Johnson, Karuna, LB Pharma, Lundbeck, MedAvante-ProPhase, MedInCell, Merck, Mindpax, Mitsubishi Tanabe Pharma, Mylan, Neurocrine, Newron, Noven, Novo Nordisk, Otsuka, Pharmabrain, PPD Biotech, Recordati, Relmada, Reviva, Rovi, Seqirus, SK Life Science, Sunovion, Sun Pharma, Supernus, Takeda, Teva, and Viatris; has provided expert testimony for Janssen and Otsuka; has served on a data safety monitoring board for COMPASS Pathways, Lundbeck, Relmada, Reviva, Rovi, Supernus, and Teva; has received grant support from Janssen and Takeda; has received royalties from UpToDate; and is a stock option holder of Cardio Diagnostics, MindPax, LB Pharma, and Quantic. All other authors declare no competing interests., (Copyright © 2023 The Author(s). Published by Elsevier Ltd. This is an Open Access article under the CC BY 4.0 license. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
43. Dopamine in major depressive disorder: A systematic review and meta-analysis of in vivo imaging studies.
- Author
-
Mizuno Y, Ashok AH, Bhat BB, Jauhar S, and Howes OD
- Subjects
- Humans, Tomography, Emission-Computed, Single-Photon, Positron-Emission Tomography, Receptors, Dopamine D2 metabolism, Dopamine Plasma Membrane Transport Proteins, Dopamine, Depressive Disorder, Major diagnostic imaging
- Abstract
Background: Major depressive disorder (MDD) is a leading cause of global disability. Several lines of evidence implicate the dopamine system in its pathophysiology. However, the magnitude and consistency of the findings are unknown. We address this by systematically reviewing in vivo imaging evidence for dopamine measures in MDD and meta-analysing these where there are sufficient studies., Methods: Studies investigating the dopaminergic system using positron emission tomography or single photon emission computed tomography in MDD and a control group were included. Demographic, clinical and imaging measures were extracted from each study, and meta-analyses and sensitivity analyses were conducted., Results: We identified 43 studies including 662 patients and 801 controls. Meta-analysis of 38 studies showed no difference in mean or mean variability of striatal D
2/3 receptor availability ( g = 0.06, p = 0.620), or combined dopamine synthesis and release capacity ( g = 0.19, p = 0.309). Dopamine transporter (DAT) availability was lower in the MDD group in studies using DAT selective tracers ( g = -0.56, p = 0.006), but not when tracers with an affinity for serotonin transporters were included ( g = -0.21, p = 0.420). Subgroup analysis showed greater dopamine release ( g = 0.49, p = 0.030), but no difference in dopamine synthesis capacity ( g = -0.21, p = 0.434) in the MDD group. Striatal D1 receptor availability was lower in patients with MDD in two studies., Conclusions: The meta-analysis indicates striatal DAT availability is lower, but D2/3 receptor availability is not altered in people with MDD compared to healthy controls. There may be greater dopamine release and lower striatal D1 receptors in MDD, although further studies are warranted. We discuss factors associated with these findings, discrepancies with preclinical literature and implications for future research., Competing Interests: Declaration of conflicting interestsThe author(s) declared the following potential conflicts of interest with respect to the research, authorship and/or publication of this article: Dr Mizuno has received fellowship grants from Canon Foundation in Europe, manuscript fees from Sumitomo Dainippon Pharma, and consultant fees from WCG Clinical and Signant Health within the past 3 years. Dr Ashok has conducted research funded by the National Institute of Health Research (Reference number: NIHR ACF-2019-14-004). Dr Bhat has no conflicting interests to declare. Dr Jauhar has received honoraria for educational talks given for Boehringer Ingelheim, Lundbeck, Janssen and Sunovion, has advised on antipsychotics to LB Pharmaceuticals, and has sat on a Wellcome Funding Panel on backwards translation in mental health. Professor Howes has received investigator-initiated research funding from and/or participated in advisory/speaker meetings organised by Angelini, Autifony, Biogen, Boehringer Ingelheim, Eli Lilly, Heptares, Global Medical Education, Invicro, Janssen, Lundbeck, Neurocrine, Otsuka, Sunovion, Rand, Recordati, Roche and Viatris/Mylan, and was a part-time employee of H Lundbeck A/S. Professor Howes has a patent for the use of dopaminergic imaging.- Published
- 2023
- Full Text
- View/download PDF
44. The histamine system and cognitive function: An in vivo H3 receptor PET imaging study in healthy volunteers and patients with schizophrenia.
- Author
-
Arumuham A, Nour MM, Veronese M, Onwordi EC, Rabiner EA, and Howes OD
- Subjects
- Humans, Histamine metabolism, Healthy Volunteers, Cognition, Positron-Emission Tomography methods, Schizophrenia, Receptors, Histamine H3 metabolism
- Abstract
Background: The histamine-3 receptor (H3R) is an auto- and heteroreceptor that inhibits the release of histamine and other neurotransmitters. Post-mortem evidence has found altered H3R expression in patients with psychotic disorders, which may underlie cognitive impairment associated with schizophrenia (CIAS)., Aims: We used positron emission tomography (PET) imaging to compare brain uptake of an H3R selective tracer between patients with schizophrenia and matched controls (healthy individuals). Regions of interest included the dorsolateral prefrontal cortex (DLPFC) and striatum. We explored correlations between tracer uptake and symptoms, including cognitive domains., Methods: A total of 12 patients and 12 matched controls were recruited to the study and were assessed with psychiatric and cognitive rating scales. They received a PET scan using the H3R-specific radioligand [
11 C]MK-8278 to determine H3R availability., Results: There was no statistically significant difference in tracer uptake between patients and controls in the DLPFC ( t19 = 0.79, p = 0.44) or striatum (t21 = 1.18, p = 0.25). An exploratory analysis found evidence for lower volume of distribution in the left cuneus (pFWE-corrected = 0.01). DLPFC tracer uptake was strongly correlated with cognition in controls (trail making test (TMT) A: r = 0.77, p = 0.006; TMT B: rho = 0.74, p = 0.01), but not in patients (TMT A: r = -0.18, p = 0.62; TMT B: rho = -0.06, p = 0.81)., Conclusions: These findings indicate H3R in the DLPFC might play a role in executive function and this is disrupted in schizophrenia in the absence of major alterations in H3R availability as assessed using a selective radiotracer for H3R. This provides further evidence for the role of H3R in CIAS., Competing Interests: Declaration of conflicting interestsThe author(s) declared the following potential conflicts of interest with respect to the research, authorship, and/or publication of this article: AA, MMN, MV and ECO have no conflicting interests to declare. EAR is a full-time employee of Invicro, an imaging centre conducting contract studies for pharmaceutical and biotech partners. As part of his role, EAR has consulted for numerous industry and academic entities. ODH is a part-time employee and stock holder of Lundbeck A/s. He has received investigator-initiated research funding from and/or participated in advisory/speaker meetings organised by Angellini, Autifony, Biogen, Boehringer-Ingelheim, Eli Lilly, Heptares, Global Medical Education, Invicro, Jansenn, Lundbeck, Neurocrine, Otsuka, Sunovion, Recordati, Roche and Viatris/Mylan. ODH has a patent for the use of dopaminergic imaging.- Published
- 2023
- Full Text
- View/download PDF
45. Dopamine and schizophrenia from bench to bedside: Discovery of a striatal co-expression risk gene set that predicts in vivo measures of striatal function.
- Author
-
Sportelli L, Eisenberg DP, Passiatore R, D'Ambrosio E, Antonucci LA, Chen Q, Czarapata J, Goldman AL, Gregory M, Griffiths K, Hyde TM, Kleinman JE, Pardiñas AF, Parihar M, Popolizio T, Rampino A, Shin JH, Veronese M, Ulrich WS, Zink CF, Bertolino A, Howes OD, Berman KF, Weinberger DR, and Pergola G
- Abstract
Schizophrenia (SCZ) is characterized by a polygenic risk architecture implicating diverse molecular pathways important for synaptic function. However, how polygenic risk funnels through these pathways to translate into syndromic illness is unanswered. To evaluate biologically meaningful pathways of risk, we used tensor decomposition to characterize gene co-expression in post-mortem brain (of neurotypicals: N=154; patients with SCZ: N=84; and GTEX samples N=120) from caudate nucleus (CN), hippocampus (HP), and dorsolateral prefrontal cortex (DLPFC). We identified a CN-predominant gene set showing dopaminergic selectivity that was enriched for genes associated with clinical state and for genes associated with SCZ risk. Parsing polygenic risk score for SCZ based on this specific gene set (parsed-PRS), we found that greater pathway-specific SCZ risk predicted greater in vivo striatal dopamine synthesis capacity measured by [
18 F]-FDOPA PET in three independent cohorts of neurotypicals and patients (total N=235) and greater fMRI striatal activation during reward anticipation in two additional independent neurotypical cohorts (total N=141). These results reveal a 'bench to bedside' translation of dopamine-linked genetic risk variation in driving in vivo striatal neurochemical and hemodynamic phenotypes that have long been implicated in the pathophysiology of SCZ.- Published
- 2023
- Full Text
- View/download PDF
46. Trace amine-associated receptor 1 (TAAR1) agonists for psychosis: protocol for a living systematic review and meta-analysis of human and non-human studies.
- Author
-
Siafis S, McCutcheon R, Chiocchia V, Ostinelli EG, Wright S, Stansfield C, Juma DO, Mantas I, Howes OD, Rutigliano G, Ramage F, Tinsdeall F, Friedrich C, Milligan L, Moreno C, Elliott JH, Thomas J, Macleod MR, Sena ES, Seedat S, Salanti G, Potts J, Cipriani A, and Leucht S
- Abstract
Background: There is an urgent need to develop more effective and safer antipsychotics beyond dopamine 2 receptor antagonists. An emerging and promising approach is TAAR1 agonism. Therefore, we will conduct a living systematic review and meta-analysis to synthesize and triangulate the evidence from preclinical animal experiments and clinical studies on the efficacy, safety, and underlying mechanism of action of TAAR1 agonism for psychosis., Methods: Independent searches will be conducted in multiple electronic databases to identify clinical and animal experimental studies comparing TAAR1 agonists with licensed antipsychotics or other control conditions in individuals with psychosis or animal models for psychosis, respectively. The primary outcomes will be overall psychotic symptoms and their behavioural proxies in animals. Secondary outcomes will include side effects and neurobiological measures. Two independent reviewers will conduct study selection, data extraction using predefined forms, and risk of bias assessment using suitable tools based on the study design. Ontologies will be developed to facilitate study identification and data extraction. Data from clinical and animal studies will be synthesized separately using random-effects meta-analysis if appropriate, or synthesis without meta-analysis. Study characteristics will be investigated as potential sources of heterogeneity. Confidence in the evidence for each outcome and source of evidence will be evaluated, considering the summary of the association, potential concerns regarding internal and external validity, and reporting biases. When multiple sources of evidence are available for an outcome, an overall conclusion will be drawn in a triangulation meeting involving a multidisciplinary team of experts. We plan trimonthly updates of the review, and any modifications in the protocol will be documented. The review will be co-produced by multiple stakeholders aiming to produce impactful and relevant results and bridge the gap between preclinical and clinical research on psychosis., Protocol Registration: PROSPERO-ID: CRD42023451628., Competing Interests: Competing interests: Spyridon Siafis: None Robert McCutcheon: RM received speaker/consultancy fees from Karuna, Janssen, Boehringer Ingelheim and Otsuka, and is director of a company that hosts psychotropic prescribing decision tools. Virginia Chiocchia: None. Edoardo G. Ostinelli: EGO has received research and consultancy fees from Angelini Pharma. Simonne Wright: None. Claire Stansfield: None. Damian Omari Juma: None. Ioannis Mantas: None. Oliver D. Howes: ODH is a part-time employee of H. Lundbeck A/S and has received investigator-initiated research funding from and/or participated in advisory/speaker meetings organized by Angellini, Autifony, Biogen, Boehringer-Ingelheim, Eli Lilly, Heptares, Global Medical Education, Invicro, Jansenn, Lundbeck, Neurocrine, Otsuka, Sunovion, Rand, Recordati, Roche and Viatris/Mylan. Neither Howes or his family have holdings/a financial stake in any pharmaceutical company Grazia Rutigliano: None. Fiona Ramage: None. Francesca Tinsdeall: None. Claire Friedrich: None. Lea Milligan: None. Carmen Moreno: CM received honoraria as a consultant and/or advisor and/or for lectures from Angelini, Compass, Esteve, Exeltis Janssen, Lundbeck, Neuraxpharm, Nuvelution, Otsuka, Pfizer, Servier and Sunovion outside the submitted work. Julian H Elliott: None. James Thomas: None. Emily Sena: None. Malcolm R. MacLeod: None. Soraya Seedat: None Georgia Salanti: None. Jennifer Potts: None. Andrea Cipriani: AC received research, educational and consultancy fees from the Italian Network for Paediatric Trials, CARIPLO Foundation, Lundbeck, and Angelini Pharma. Stefan Leucht: SL has received honoraria as advisor and/or for lectures and/or for educational material from Alkermes, Angelini, Apsen, Eisai, Gedeon Richter, Janssen, Karuna, Kynexis, Lundbeck, Medichem, Medscape, Merck Sharpp and Dome, Mitshubishi, Neurotorium, NovoNordisk, Otsuka, Recordati, Roche, Rovi, Sanofi Aventis, TEVA, (Copyright: © 2023 Siafis S et al.)
- Published
- 2023
- Full Text
- View/download PDF
47. A leaky umbrella has little value: evidence clearly indicates the serotonin system is implicated in depression.
- Author
-
Jauhar S, Arnone D, Baldwin DS, Bloomfield M, Browning M, Cleare AJ, Corlett P, Deakin JFW, Erritzoe D, Fu C, Fusar-Poli P, Goodwin GM, Hayes J, Howard R, Howes OD, Juruena MF, Lam RW, Lawrie SM, McAllister-Williams H, Marwaha S, Matuskey D, McCutcheon RA, Nutt DJ, Pariante C, Pillinger T, Radhakrishnan R, Rucker J, Selvaraj S, Stokes P, Upthegrove R, Yalin N, Yatham L, Young AH, Zahn R, and Cowen PJ
- Subjects
- Serotonin, Depression
- Published
- 2023
- Full Text
- View/download PDF
48. Reduced cortical cerebral blood flow in antipsychotic-free first-episode psychosis and relationship to treatment response.
- Author
-
Selvaggi P, Jauhar S, Kotoula V, Pepper F, Veronese M, Santangelo B, Zelaya F, Turkheimer FE, Mehta MA, and Howes OD
- Subjects
- Humans, Follow-Up Studies, Cerebrovascular Circulation physiology, Magnetic Resonance Imaging, Antipsychotic Agents pharmacology, Antipsychotic Agents therapeutic use, Psychotic Disorders diagnostic imaging, Psychotic Disorders drug therapy, Psychotic Disorders pathology, Schizophrenia diagnostic imaging, Schizophrenia drug therapy, Schizophrenia pathology
- Abstract
Background: Altered cerebral blood flow (CBF) has been found in people at risk for psychosis, with first-episode psychosis (FEP) and with chronic schizophrenia (SCZ). Studies using arterial spin labelling (ASL) have shown reduction of cortical CBF and increased subcortical CBF in SCZ. Previous studies have investigated CBF using ASL in FEP, reporting increased CBF in striatum and reduced CBF in frontal cortex. However, as these people were taking antipsychotics, it is unclear whether these changes are related to the disorder or antipsychotic treatment and how they relate to treatment response., Methods: We examined CBF in FEP free from antipsychotic medication ( N = 21), compared to healthy controls ( N = 22). Both absolute and relative-to-global CBF were assessed. We also investigated the association between baseline CBF and treatment response in a partially nested follow-up study ( N = 14)., Results: There was significantly lower absolute CBF in frontal cortex (Cohen's d = 0.84, p = 0.009) and no differences in striatum or hippocampus. Whole brain voxel-wise analysis revealed widespread cortical reductions in absolute CBF in large cortical clusters that encompassed occipital, parietal and frontal cortices (Threshold-Free Cluster Enhancement (TFCE)-corrected <0.05). No differences were found in relative-to-global CBF in the selected region of interests and in voxel-wise analysis. Relative-to-global frontal CBF was correlated with percentage change in total Positive and Negative Syndrome Scale after antipsychotic treatment ( r = 0.67, p = 0.008)., Conclusions: These results show lower cortical absolute perfusion in FEP prior to starting antipsychotic treatment and suggest relative-to-global frontal CBF as assessed with magnetic resonance imaging could potentially serve as a biomarker for antipsychotic response.
- Published
- 2023
- Full Text
- View/download PDF
49. The association between peripheral inflammation, brain glutamate and antipsychotic response in Schizophrenia: Data from the STRATA collaboration.
- Author
-
Fenn-Moltu S, Deakin B, Drake R, Howes OD, Lawrie SM, Lewis S, Nikkheslat N, Walters JTR, MacCabe JH, Mondelli V, and Egerton A
- Subjects
- Humans, Glutamic Acid metabolism, Interleukin-8, Brain metabolism, Inflammation metabolism, Schizophrenia drug therapy, Schizophrenia metabolism, Antipsychotic Agents therapeutic use, Encephalitis metabolism
- Abstract
Glutamate and increased inflammation have been separately implicated in the pathophysiology of schizophrenia and the extent of clinical response to antipsychotic treatment. Despite the mechanistic links between pro-inflammatory and glutamatergic pathways, the relationships between peripheral inflammatory markers and brain glutamate in schizophrenia have not yet been investigated. In this study, we tested the hypothesis that peripheral levels of pro-inflammatory cytokines would be positively associated with brain glutamate levels in schizophrenia. Secondary analyses determined whether this relationship differed according to antipsychotic treatment response. The sample consisted of 79 patients with schizophrenia, of whom 40 were rated as antipsychotic responders and 39 as antipsychotic non-responders. Brain glutamate levels were assessed in the anterior cingulate cortex (ACC) and caudate using proton magnetic resonance spectroscopy (
1 H-MRS) and blood samples were collected for cytokine assay on the same study visit (IL-6, IL-8, IL-10, TNF- α and IFN-γ). Across the whole patient sample, there was a positive relationship between interferon-gamma (IFN-γ) and caudate glutamate levels (r = 0.31, p = 0.02). In the antipsychotic non-responsive group only, there was a positive relationship between interleukin-8 (IL-8) and caudate glutamate (r = 0.46, p = 0.01). These findings provide evidence to link specific peripheral inflammatory markers and caudate glutamate in schizophrenia and may suggest that this relationship is most marked in patients who show a poor response to antipsychotic treatment., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
50. Low-frequency monitoring for community clozapine initiations: A comparative study relative to standard frequency assessments.
- Author
-
Mizuno Y, Bridglal DL, Coumbe J, McGrath H, Adhikari A, Butler E, Bonoldi I, Taylor D, and Howes OD
- Subjects
- Cognition, Clozapine adverse effects, Antipsychotic Agents adverse effects
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.