1
|
Furmark T, Wahlstedt K, Faria V. Revisiting the SSRI vs. placebo debate in the treatment of social anxiety disorder: the role of expectancy effects, neural responsivity, and monoamine transporters. Front Psychol 2025; 16:1531725. [PMID: 40420982 PMCID: PMC12104218 DOI: 10.3389/fpsyg.2025.1531725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 04/17/2025] [Indexed: 05/28/2025] Open
Abstract
Selective serotonin reuptake inhibitors (SSRIs), widely used for anxiety and depression, are often criticized for their perceived similarity in efficacy to placebo treatments and the unclear connection between brain serotonin levels, on one hand, and the symptomatology of these disorders, on the other. In this perspective paper we discuss the complex mechanisms behind SSRI and placebo treatments in managing social anxiety disorder (SAD), focusing on both pharmacological and expectancy effects. Through a series of neuroimaging studies using positron emission tomography (PET), we investigated the neural, neurochemical and behavioral changes associated with SSRI and placebo responses in SAD patients. Results from one study revealed that both SSRI and placebo responders showed equal reductions in amygdala activity, a region central to fear processing, as well as comparable improvements in social anxiety symptoms. These findings suggest shared neural pathways between SSRIs and placebos, possibly related to response expectancies. In another study, we manipulated patient expectations using a deception design, showing that overt SSRI treatment yielded greater symptom reduction than covert administration. PET results further underscored the influence of expectation on dopamine signaling. Furthermore, PET data on serotonin transporters indicated that serotonin reuptake inhibition alone does not fully account for SSRIs' clinical efficacy, as serotonin transporter occupancy was not correlated with symptom improvement. In yet another study, combining SSRIs with cognitive-behavioral therapy (CBT) led to more robust and longer-lasting outcomes than placebo combined with CBT, with distinct effects on brain monoamine transporters. Overall, these findings emphasize the intricate interplay between pharmacology, brain mechanisms, and psychological expectations in the treatment of SAD.
Collapse
Affiliation(s)
- Tomas Furmark
- Department of Psychology, Uppsala University, Uppsala, Sweden
| | - Kurt Wahlstedt
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Vanda Faria
- Department of Psychology, Uppsala University, Uppsala, Sweden
- Department of Anesthesia, Critical Care and Pain Medicine, Pain and Affective Neuroscience Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
- Interdisciplinary Pain Center, University Hospital and Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
| |
Collapse
|
2
|
Ozpolat C, Okcay Y, Ulusoy KG, Yildiz O. A narrative review of the placebo effect: historical roots, current applications, and emerging insights. Eur J Clin Pharmacol 2025; 81:625-645. [PMID: 40080139 DOI: 10.1007/s00228-025-03818-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 02/23/2025] [Indexed: 03/15/2025]
Abstract
PURPOSE Placebo is a term to define physiologically inactive compounds used in treatment that causes physical and emotional changes. The placebo effect, driven by expectation and conditioning, plays a significant role in various conditions like pain, depression, and Parkinson's disease, while the nocebo effect can hinder treatment outcomes. Understanding mechanisms such as neuromodulation and genetics has gained importance in modern medicine. This review aims to explore the clinical relevance of placebo responses, particularly in neuropsychiatric disorders, and their potential in personalized medicine. By integrating placebo research into healthcare, it highlights opportunities to enhance treatment efficacy, improve patient well-being, and reduce reliance on pharmacological interventions. METHODS A comprehensive literature search was conducted in PubMed, Scopus, and Google Scholar databases. Recent studies were reviewed to evaluate placebo effects, and the variability of the placebo response in neuropsychiatric disorders was summarized. RESULTS Placebo effects significantly impact treatment outcomes across various conditions, including Parkinson's disease, depression, pain syndromes, and epilepsy. The mechanisms involve neurobiological and psychological factors, with evidence suggesting that placebo interventions can modulate neurotransmitter activity and improve patient well-being. CONCLUSION Integrating placebo research into clinical practice may enhance treatment outcomes, reduce drug dependency, and support personalized medicine by tailoring interventions to individual placebo responsiveness. Understanding placebo and nocebo mechanisms can optimize therapeutic strategies while minimizing unnecessary pharmacological treatments.
Collapse
Affiliation(s)
- Cagri Ozpolat
- Department of Medical Pharmacology, Gulhane Faculty of Medicine, University of Health Sciences, Ankara, Turkey
| | - Yagmur Okcay
- Department of Pharmacology, University of Health Sciences Gulhane Faculty of Pharmacy, Ankara, Turkey
| | - Kemal Gokhan Ulusoy
- Department of Medical Pharmacology, Gulhane Faculty of Medicine, University of Health Sciences, Ankara, Turkey
| | - Oğuzhan Yildiz
- Department of Medical Pharmacology, Gulhane Faculty of Medicine, University of Health Sciences, Ankara, Turkey.
| |
Collapse
|
3
|
Miller CWT, Kozak Z. Therapeutic and Structural Dimensions in Psychiatric Prescribing: Bridging Psychedelics and Antidepressants. Harv Rev Psychiatry 2025; 33:149-157. [PMID: 40095787 DOI: 10.1097/hrp.0000000000000425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
ABSTRACT As practitioners seek more personalized approaches, exploring how patients' environments, relationship templates, and mindsets factor into symptom burden can help broaden understanding of how psychotropic medications facilitate recovery. Despite increasing focus on medications to provide relief, there is an important and undeniable influence the therapeutic environment has on shaping outcomes, particularly for the patient-clinician alliance. While environmental dimensions are relevant for informing possible placebo or nocebo responses, they also build upon the pharmacodynamic and neurobiological effects of medications. By heightening neuroplasticity, some antidepressants may amplify the effects of nonmedication factors in patients' lives, including the patient-prescriber therapeutic relationship. There are important parallels between antidepressants and psychedelics in emerging literature. For instance, the preparatory and integrative work with a provider can be crucial in determining outcomes. This paper will draw from the extant literature to discuss the therapeutic relationship in psychiatric practice, including in acute care settings and instances in which psychotropic prescribing is a key aspect of treatment.
Collapse
Affiliation(s)
- Christopher W T Miller
- From University of Maryland School of Medicine (Dr. Miller) and Sheppard Pratt Health System (Dr. Kozak), Baltimore, MD
| | | |
Collapse
|
4
|
Raghuraman N, White JN, Watson L, Belleï-Rodriguez CÉ, Shafir R, Wang Y, Colloca L. Neuropsychological mechanisms of observational learning in human placebo effects. Psychopharmacology (Berl) 2025; 242:889-900. [PMID: 38743108 PMCID: PMC11561162 DOI: 10.1007/s00213-024-06608-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 05/02/2024] [Indexed: 05/16/2024]
Abstract
Scientific evidence indicates that placebo effects are psychoneurobiological events involving the contribution of distinct central nervous systems and peripheral physiological mechanisms that influence pain perception and other symptoms. Placebo effects can occur without formal conditioning and direct prior experience because crucial information can be acquired through observational learning. Observation of benefits in another person results in placebo effects of a magnitude like those induced by directly experiencing an analgesic benefit. Understanding the psychological mechanisms of observationally induced placebo effects is a complex and multifaceted endeavor. While previous reviews have highlighted various frameworks and models to understand these phenomena, the underlying biological mechanisms have been overlooked. We summarize critically current understanding of its behavioral and neural mechanisms. Understanding the neural mechanisms of hypoalgesia driven by observation can serve as a foundation for future development of novel theoretical and methodological approaches and ultimately, applications.
Collapse
Affiliation(s)
- Nandini Raghuraman
- Department of Pain and Translational Symptom Science, University of Maryland School of Nursing, Baltimore, MD, USA
- Graduate Program in Life Sciences, Program in Epidemiology and Human Genetics, University of Maryland School of Medicine, Baltimore, MD, USA
- Placebo Beyond Opinions Center, University of Maryland School of Nursing, Baltimore, USA
| | - Jewel N White
- Department of Pain and Translational Symptom Science, University of Maryland School of Nursing, Baltimore, MD, USA
- Placebo Beyond Opinions Center, University of Maryland School of Nursing, Baltimore, USA
- Graduate Program in Life Sciences, Program in Neuroscience, University of Maryland School of Medicine, Baltimore, USA
| | - Lakota Watson
- Department of Pain and Translational Symptom Science, University of Maryland School of Nursing, Baltimore, MD, USA
- Graduate Program in Life Sciences, Program in Neuroscience, University of Maryland School of Medicine, Baltimore, USA
| | | | - Roni Shafir
- Department of Pain and Translational Symptom Science, University of Maryland School of Nursing, Baltimore, MD, USA
- Placebo Beyond Opinions Center, University of Maryland School of Nursing, Baltimore, USA
| | - Yang Wang
- Department of Pain and Translational Symptom Science, University of Maryland School of Nursing, Baltimore, MD, USA
- Placebo Beyond Opinions Center, University of Maryland School of Nursing, Baltimore, USA
- Center to Advance Chronic Pain Research, University of Maryland, Baltimore, USA
| | - Luana Colloca
- Department of Pain and Translational Symptom Science, University of Maryland School of Nursing, Baltimore, MD, USA.
- Placebo Beyond Opinions Center, University of Maryland School of Nursing, Baltimore, USA.
- Center to Advance Chronic Pain Research, University of Maryland, Baltimore, USA.
| |
Collapse
|
5
|
Dai F, Wengler K, He X, Wang J, Yang J, Parsey RV, DeLorenzo C. Lack of association between pretreatment glutamate/GABA and major depressive disorder treatment response. Transl Psychiatry 2025; 15:71. [PMID: 40025010 PMCID: PMC11873289 DOI: 10.1038/s41398-025-03292-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 11/21/2024] [Accepted: 02/19/2025] [Indexed: 03/04/2025] Open
Abstract
Studies have shown gamma-amino-butyric acid (GABA) and Glx (a combination of glutamate and glutamine) to be altered in major depressive disorder (MDD). Using proton Magnetic Resonance Spectroscopy (1H-MRS), this study aimed to determine whether lower pretreatment GABA and Glx levels in the medial frontal cortex, a region implicated in MDD pathophysiology, are associated with better antidepressant treatment response. Participants with MDD (N = 74) were antidepressant naïve or medication-free for at least three weeks before imaging. Two MEGA-PRESS 1H-MRS acquisitions were collected, interleaved with a water unsuppressed reference scan. GABA and Glx concentrations were quantified from an average difference spectrum, with preprocessing using Gannet and spectral fitting using TARQUIN. Following imaging, participants were randomized to escitalopram or placebo for 8 weeks in a double-blind design. Multivariable logistic regression models were applied with treatment type and age as covariates. Bayes Factor hypothesis testing was used to interpret the strength of the evidence. No significant association was found between pretreatment Glx, GABA, or Glx/GABA and depression remission status or the continuous outcome, percent change in symptom severity. In an exploratory analysis, no significant correlation was found between pretreatment Glx, GABA or Glx/GABA and days to response. Bayes factor analysis showed strong evidence towards the null hypotheses in all cases. To date, there are no replicated biomarkers in psychiatry. To address this, well-powered, placebo-controlled trials need to be undertaken and reported. The present analysis suggests pretreatment GABA, Glx, or their ratio cannot predict antidepressant treatment response. Future direction including examining glutamate and glutamine separately or examining biological subtypes of MDD separately.Trial Name: Advancing Personalized Antidepressant Treatment Using PET/MRI.Registration Number: NCT02623205 URL: https://clinicaltrials.gov/ct2/show/NCT02623205.
Collapse
Affiliation(s)
| | - Kenneth Wengler
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Xiang He
- Department of Radiology, Northwell Health, New York, NY, USA
| | - Junying Wang
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY, USA
| | - Jie Yang
- Department of Family, Population, and Preventive Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Ramin V Parsey
- Department of Psychiatry, Stony Brook University, Stony Brook, NY, USA
| | - Christine DeLorenzo
- Department of Psychiatry, Stony Brook University, Stony Brook, NY, USA
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA
- Department of Psychiatry, Columbia University, New York, NY, USA
| |
Collapse
|
6
|
Cusin C, Dillon DG, Belleau E, Normandin MD, Petibon Y, El-Fakri G, Dhaynaut M, Hooker J, Kaptchuk T, McKee M, Hayden E, Meyer A, Jahan A, Origlio J, Ang YS, Brunner D, Kang M, Long Y, Fava M, Pizzagalli DA. Novel multi-modal methodology to investigate placebo response in major depressive disorder. J Affect Disord 2025; 368:1-7. [PMID: 39233242 DOI: 10.1016/j.jad.2024.08.226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 08/23/2024] [Accepted: 08/31/2024] [Indexed: 09/06/2024]
Abstract
The neurobiological mechanisms underlying the placebo phenomenon in patients with major depressive disorder (MDD) remain largely unknown. The progressive rise in rates of placebo responses within clinical trials over the past two decades may impede the detection of a true signal and thus present a major obstacle in new treatment development. Understanding the mechanisms would have several important implications, including (1) identifying biomarkers of placebo responders (thereby identifying those individuals who could benefit therapeutically from such interventions), (2) opening new avenues for manipulating such mechanisms to maximize symptom reduction, and (3) refining treatments with approaches that decrease (in clinical trials) or increase (in clinical practice) the placebo response. Here we investigated the research question: is the dopaminergic system one of the neurobiological underpinnings of the placebo response within MDD? Inspired by preclinical and clinical findings that have implicated dopamine in the occurrence, prediction, and expectation of reward, we hypothesized that dopaminergic activity in the mesolimbic system is a critical mediator of placebo response in MDD. To test this hypothesis, we designed a double-blind, placebo-controlled, sequential parallel comparison design clinical trial aimed at maximizing placebo antidepressant response. We integrated behavioral, imaging, and hemodynamic probes of mesocorticolimbic dopaminergic pathways within the context of manipulations of psychological constructs previously linked to placebo responses (e.g., expectation of improvement). The aim of this manuscript is to present the rationale of the study design and to demonstrate how a cross-modal methodology may be utilized to investigate the role of reward circuitry in placebo response in MDD.
Collapse
Affiliation(s)
- Cristina Cusin
- Massachusetts General Hospital, Boston, MA, United States of America.
| | | | - Emily Belleau
- McLean Hospital, Belmont, MA, United States of America
| | - Marc D Normandin
- Massachusetts General Hospital, Boston, MA, United States of America
| | - Yoann Petibon
- Massachusetts General Hospital, Boston, MA, United States of America
| | - Georges El-Fakri
- Massachusetts General Hospital, Boston, MA, United States of America
| | - Maeva Dhaynaut
- Massachusetts General Hospital, Boston, MA, United States of America
| | - Jacob Hooker
- Massachusetts General Hospital, Boston, MA, United States of America
| | - Ted Kaptchuk
- Beth Israel Deaconess Medical Center, Boston, MA, United States of America
| | - Madison McKee
- Massachusetts General Hospital, Boston, MA, United States of America
| | - Emma Hayden
- Massachusetts General Hospital, Boston, MA, United States of America
| | - Ashley Meyer
- Massachusetts General Hospital, Boston, MA, United States of America
| | - Aava Jahan
- Massachusetts General Hospital, Boston, MA, United States of America
| | - Julianne Origlio
- Massachusetts General Hospital, Boston, MA, United States of America
| | | | - Devon Brunner
- McLean Hospital, Belmont, MA, United States of America
| | - Min Kang
- McLean Hospital, Belmont, MA, United States of America
| | - Yinru Long
- McLean Hospital, Belmont, MA, United States of America
| | - Maurizio Fava
- Massachusetts General Hospital, Boston, MA, United States of America
| | | |
Collapse
|
7
|
Branchi I. Uncovering the determinants of brain functioning, behavior and their interplay in the light of context. Eur J Neurosci 2024; 60:4687-4706. [PMID: 38558227 DOI: 10.1111/ejn.16331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 03/07/2024] [Indexed: 04/04/2024]
Abstract
Notwithstanding the huge progress in molecular and cellular neuroscience, our ability to understand the brain and develop effective treatments promoting mental health is still limited. This can be partially ascribed to the reductionist, deterministic and mechanistic approaches in neuroscience that struggle with the complexity of the central nervous system. Here, I introduce the Context theory of constrained systems proposing a novel role of contextual factors and genetic, molecular and neural substrates in determining brain functioning and behavior. This theory entails key conceptual implications. First, context is the main driver of behavior and mental states. Second, substrates, from genes to brain areas, have no direct causal link to complex behavioral responses as they can be combined in multiple ways to produce the same response and different responses can impinge on the same substrates. Third, context and biological substrates play distinct roles in determining behavior: context drives behavior, substrates constrain the behavioral repertoire that can be implemented. Fourth, since behavior is the interface between the central nervous system and the environment, it is a privileged level of control and orchestration of brain functioning. Such implications are illustrated through the Kitchen metaphor of the brain. This theoretical framework calls for the revision of key concepts in neuroscience and psychiatry, including causality, specificity and individuality. Moreover, at the clinical level, it proposes treatments inducing behavioral changes through contextual interventions as having the highest impact to reorganize the complexity of the human mind and to achieve a long-lasting improvement in mental health.
Collapse
Affiliation(s)
- Igor Branchi
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
8
|
Mollica A, Ng E, Burke MJ, Nestor SM, Lee H, Rabin JS, Hamani C, Lipsman N, Giacobbe P. Treatment expectations and clinical outcomes following repetitive transcranial magnetic stimulation for treatment-resistant depression. Brain Stimul 2024; 17:752-759. [PMID: 38901565 DOI: 10.1016/j.brs.2024.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 05/28/2024] [Accepted: 06/05/2024] [Indexed: 06/22/2024] Open
Abstract
BACKGROUND Patient expectations, including both positive (placebo) and negative (nocebo) effects, influence treatment outcomes, yet their impact on acute repetitive transcranial magnetic stimulation (rTMS) for treatment-resistant depression (TRD) is unclear. METHODS In this single-center retrospective chart review, 208 TRD patients completed the Stanford Expectation of Treatment Scale (SETS) before starting open-label rTMS treatment. Patients were offered two excitatory rTMS protocols (deep TMS or intermittent theta-burst stimulation), which stimulated the left dorsolateral prefrontal cortex. A minimum of 20 once daily treatments were provided, delivered over 4-6 weeks. Primary outcomes were 1) remission, measured by a post-treatment score of <8 on the Hamilton Depression Rating Scale (HAMD-17), and 2) premature discontinuation. The change in HAMD-17 scores over time was used as a secondary outcome. Physicians were blinded to SETS scores. Logistic and linear regression, adjusting for covariates, assessed SETS and HAMD-17 relationships. RESULTS Of 208 patients, 177 had baseline and covariate data available. The mean positivity bias score (positive expectancy minus negative expectancy subscale averages) was 0.48 ± 2.21, indicating the cohort was neutral regarding the expectations of their treatment on average. Higher positive expectancy scores were significantly associated with greater odds of remission (OR = 1.90, p = 0.003) and greater reduction in HAMD-17 scores (β = 1.30, p = 0.005) at the end of acute treatment, after adjusting for covariates. Negative expectancy was not associated with decreased odds of remission (p = 0.2) or treatment discontinuation (p = 0.8). CONCLUSIONS Higher pre-treatment positive expectations were associated with greater remission rates with open-label rTMS in a naturalistic cohort of patients with TRD.
Collapse
Affiliation(s)
- Adriano Mollica
- Harquail Centre for Neuromodulation and Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada; Department of Psychiatry, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Enoch Ng
- Harquail Centre for Neuromodulation and Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada; Department of Psychiatry, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Matthew J Burke
- Harquail Centre for Neuromodulation and Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada; Department of Psychiatry, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada; Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Sean M Nestor
- Harquail Centre for Neuromodulation and Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada; Department of Psychiatry, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Hyewon Lee
- Harquail Centre for Neuromodulation and Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada; Department of Psychiatry, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Jennifer S Rabin
- Harquail Centre for Neuromodulation and Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada; Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada; Rehabilitation Sciences Institute, University of Toronto, Canada
| | - Clement Hamani
- Harquail Centre for Neuromodulation and Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada; Division of Neurosurgery, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Nir Lipsman
- Harquail Centre for Neuromodulation and Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada; Division of Neurosurgery, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Peter Giacobbe
- Harquail Centre for Neuromodulation and Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada; Department of Psychiatry, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
9
|
Lin K, Sunko D, Wang J, Yang J, Parsey RV, DeLorenzo C. Investigating the relationship between hippocampus/dentate gyrus volume and hypothalamus metabolism in participants with major depressive disorder. Sci Rep 2024; 14:10622. [PMID: 38724691 PMCID: PMC11082185 DOI: 10.1038/s41598-024-61519-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 05/07/2024] [Indexed: 05/12/2024] Open
Abstract
Reduced hippocampal volume occurs in major depressive disorder (MDD), potentially due to elevated glucocorticoids from an overactivated hypothalamus-pituitary-adrenal (HPA) axis. To examine this in humans, hippocampal volume and hypothalamus (HPA axis) metabolism was quantified in participants with MDD before and after antidepressant treatment. 65 participants (n = 24 males, n = 41 females) with MDD were treated in a double-blind, randomized clinical trial of escitalopram. Participants received simultaneous positron emission tomography (PET)/magnetic resonance imaging (MRI) before and after treatment. Linear mixed models examined the relationship between hippocampus/dentate gyrus volume and hypothalamus metabolism. Chi-squared tests and multivariable logistic regression examined the association between hippocampus/dentate gyrus volume change direction and hypothalamus activity change direction with treatment. Multiple linear regression compared these changes between remitter and non-remitter groups. Covariates included age, sex, and treatment type. No significant linear association was found between hippocampus/dentate gyrus volume and hypothalamus metabolism. 62% (38 of 61) of participants experienced a decrease in hypothalamus metabolism, 43% (27 of 63) of participants demonstrated an increase in hippocampus size (51% [32 of 63] for the dentate gyrus) following treatment. No significant association was found between change in hypothalamus activity and change in hippocampus/dentate gyrus volume, and this association did not vary by sex, medication, or remission status. As this multimodal study, in a cohort of participants on standardized treatment, did not find an association between hypothalamus metabolism and hippocampal volume, it supports a more complex pathway between hippocampus neurogenesis and hypothalamus metabolism changes in response to treatment.
Collapse
Affiliation(s)
| | | | - Junying Wang
- Department of Applied Mathematics and Statistics, Stony Brook University, New York, NY, USA
| | - Jie Yang
- Department of Family, Population & Preventive Medicine, Stony Brook University, New York, NY, USA
| | - Ramin V Parsey
- Department of Psychiatry and Behavioral Health, Stony Brook University, Stony Brook, NY, USA
| | - Christine DeLorenzo
- Department of Psychiatry and Behavioral Health, Stony Brook University, Stony Brook, NY, USA.
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA.
| |
Collapse
|
10
|
Goli F. Biosemiotic medicines: Symbolic formulations for placebo enhancements. JOURNAL OF EDUCATION AND HEALTH PROMOTION 2024; 13:156. [PMID: 38784294 PMCID: PMC11114581 DOI: 10.4103/jehp.jehp_1888_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 12/26/2023] [Indexed: 05/25/2024]
Abstract
The healing response is a complex and multiform procedure that involves many physical and symbolic interactions and synchronizations. In the clinical research model, certain factors are abstracted during which contextual elements, such as placebo responses and communicative factors, are excluded to reveal the pieces of evidence that are necessary for the mass production of clinical materials and methods. On the other side, clinical practice is a singular and chaotic communicative action in which we should include contextual and discursive factors for prompting proper biological as well as behavioral responses. Placebo responses, personal history and attitudes, and clinical relationships and communication are some of the contextual and individual factors that can be changed effectively if we can communicate with the symbolic and reflective matrices of clinical practice. In this article, the author introduces a biosemiotic formula for healing responses that include symbolic and reflective factors of healing response aligned with the related biological procedures. Not only are psychological interventions beneficial in mental health problems and symptom control but they could also be used as co-treatments to reinforce placebo responses and improve illness behavior and treatment narratives.
Collapse
Affiliation(s)
- Farzad Goli
- Faculty Bios, Energy Medicine University, California, USA
- Department for Education, Bioethics and Philosophy of Medicine, Iranian Academy of Medical Sciences, Head of Danesh-e Tandorosti Institute, Isfahan, Iran
| |
Collapse
|
11
|
Wang J, Wu DD, DeLorenzo C, Yang J. Examining factors related to low performance of predicting remission in participants with major depressive disorder using neuroimaging data and other clinical features. PLoS One 2024; 19:e0299625. [PMID: 38547128 PMCID: PMC10977765 DOI: 10.1371/journal.pone.0299625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 02/13/2024] [Indexed: 04/02/2024] Open
Abstract
Major depressive disorder (MDD), a prevalent mental health issue, affects more than 8% of the US population, and almost 17% in the young group of 18-25 years old. Since Covid-19, its prevalence has become even more significant. However, the remission (being free of depression) rates of first-line antidepressant treatments on MDD are only about 30%. To improve treatment outcomes, researchers have built various predictive models for treatment responses and yet none of them have been adopted in clinical use. One reason is that most predictive models are based on data from subjective questionnaires, which are less reliable. Neuroimaging data are promising objective prognostic factors, but they are expensive to obtain and hence predictive models using neuroimaging data are limited and such studies were usually in small scale (N<100). In this paper, we proposed an advanced machine learning (ML) pipeline for small training dataset with large number of features. We implemented multiple imputation for missing data and repeated K-fold cross validation (CV) to robustly estimate predictive performances. Different feature selection methods and stacking methods using 6 general ML models including random forest, gradient boosting decision tree, XGBoost, penalized logistic regression, support vector machine (SVM), and neural network were examined to evaluate the model performances. All predictive models were compared using model performance metrics such as accuracy, balanced accuracy, area under ROC curve (AUC), sensitivity and specificity. Our proposed ML pipeline was applied to a training dataset and obtained an accuracy and AUC above 0.80. But such high performance failed while applying our ML pipeline using an external validation dataset from the EMBARC study which is a multi-center study. We further examined the possible reasons especially the site heterogeneity issue.
Collapse
Affiliation(s)
- Junying Wang
- Department of Applied Mathematics and Statistics, Stony Brook University, New York, New York, United states of America
| | - David D. Wu
- School of Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Christine DeLorenzo
- Department of Psychiatry and Behavioral Health, Stony Brook University, Stony Brook, New York, United States of America
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York, United States of America
| | - Jie Yang
- Department of Family, Population & Preventive Medicine, Stony Brook University, Stony Brook, New York, United States of America
| |
Collapse
|
12
|
Shen W, Wang X, Li Q, Ding Q, Zhang H, Qian Z, Sun Z, Chen X, Zhang J, Zhao M, Huang L, Xing W. Research on adults with subthreshold depression after aerobic exercise: a resting-state fMRI study based on regional homogeneity (ReHo). Front Neurosci 2024; 18:1231883. [PMID: 38533447 PMCID: PMC10963409 DOI: 10.3389/fnins.2024.1231883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 01/31/2024] [Indexed: 03/28/2024] Open
Abstract
Objective Subthreshold depression (StD)/subsyndromal depression refers to a threatening precursor to depression. Aerobic exercise is a promising self-supportive adjunctive intervention and an effective measure for StD. Our study utilizes regional homogeneity (ReHo) to investigate the impact of aerobic exercise on resting-state brain function. Methods A total of 78 subjects, aged between 18 and 48 years, (StD group, n = 44; healthy control (HC) group, n = 34) engaged in moderate-intensity aerobic exercise 3-4 times per week for 8 weeks. Resting-state brain function and structural images were acquired before and after the exercise intervention. The ReHo method was employed to analyze abnormal changes in regional brain function, and a correlation analysis was performed using the Patient Health Questionnaire-9 (PHQ-9) and Self-Rating Anxiety Scale (SAS) scores. Results The principal observation reveals synchronous abnormalities in the right anterior cingulate gyrus of the brain in StD subjects compared to HCs at baseline, with these differences dissipating after the implementation of aerobic exercise. After completing the aerobic exercise program, the StD group exhibited a difference in the right middle cingulate gyrus, while the left supplementary motor area (SMA) was altered in the HC group. Conclusion Disparities in neural synchronization are evident between HCs and StD subjects, and the implementation of aerobic exercise intervention can effectively mitigate these distinctions, leading to a significant reduction in depressive symptoms among StD subjects. The primary mechanism of StD symptoms may involve the inhibition of the anterior cingulate gyrus, while the effects of aerobic exercise may be related to the modulation of neural synchronization of emotional reflexes. The discovery of these fMRI evidence findings may offer novel strategies for early detection and intervention in cases of StD.
Collapse
Affiliation(s)
- Wenbin Shen
- Department of Radiology, The Affiliated Changshu Hospital of Nantong University, Changshu No.2 People's Hospital, Changshu, Jiangsu, China
| | - Xiaoxiao Wang
- School of Foreign Studies, China University of Petroleum, Qingdao, Shandong, China
| | - Qin Li
- Department of Radiology, The Affiliated Changshu Hospital of Nantong University, Changshu No.2 People's Hospital, Changshu, Jiangsu, China
| | - Qingguo Ding
- Department of Radiology, The Affiliated Changshu Hospital of Nantong University, Changshu No.2 People's Hospital, Changshu, Jiangsu, China
| | - Hongqiang Zhang
- Department of Radiology, The Affiliated Changshu Hospital of Nantong University, Changshu No.2 People's Hospital, Changshu, Jiangsu, China
| | - Zheng Qian
- Department of Radiology, The Affiliated Changshu Hospital of Nantong University, Changshu No.2 People's Hospital, Changshu, Jiangsu, China
| | - Zhixin Sun
- Department of Radiology, The Affiliated Changshu Hospital of Nantong University, Changshu No.2 People's Hospital, Changshu, Jiangsu, China
| | - Xingyu Chen
- Department of Radiology, The Affiliated Changshu Hospital of Nantong University, Changshu No.2 People's Hospital, Changshu, Jiangsu, China
| | - Jun Zhang
- Department of Psychiatry, Changshu Third People's Hospital, Changshu, Jiangsu, China
| | - Mengqi Zhao
- School of Psychology, Zhejiang Normal University, Jinhua, Zhejiang, China
| | - Lina Huang
- Department of Radiology, The Affiliated Changshu Hospital of Nantong University, Changshu No.2 People's Hospital, Changshu, Jiangsu, China
| | - Wei Xing
- Department of Radiology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| |
Collapse
|
13
|
Phillips ML. Multimodal Predictors of Treatment Response in Major Depressive Disorder: Advancing Personalized Medicine in Psychiatry. Am J Psychiatry 2024; 181:180-182. [PMID: 38425254 DOI: 10.1176/appi.ajp.20231025] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Affiliation(s)
- Mary L Phillips
- Department of Psychiatry, University of Pittsburgh, Pittsburgh
| |
Collapse
|
14
|
Perl O, Shuster A, Heflin M, Na S, Kidwai A, Booker N, Putnam WC, Fiore VG, Gu X. Nicotine-related beliefs induce dose-dependent responses in the human brain. NATURE. MENTAL HEALTH 2024; 2:177-188. [PMID: 39463822 PMCID: PMC11512134 DOI: 10.1038/s44220-023-00188-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 11/21/2023] [Indexed: 10/29/2024]
Abstract
Beliefs have a powerful influence on our behavior, yet their neural mechanisms remain elusive. Here we investigate whether beliefs could impact brain activities in a way akin to pharmacological dose-dependent effects. Nicotine-dependent humans were told that nicotine strength in an electronic cigarette was either 'low', 'medium' or 'high', while nicotine content was held constant. After vaping, participants underwent functional neuroimaging and performed a decision-making task known to engage neural circuits affected by nicotine. Beliefs about nicotine strength induced dose-dependent responses in the thalamus, a key binding site for nicotine, but not in other brain regions such as the striatum. Nicotine-related beliefs also parametrically modulated the connectivity between the thalamus and ventromedial prefrontal cortex, a region important for decision-making. These findings reveal a high level of precision in the way beliefs influence the brain, offering mechanistic insights into humans' heterogeneous responses to drugs and a pivotal role of beliefs in addiction.
Collapse
Affiliation(s)
- Ofer Perl
- Center for Computational Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anastasia Shuster
- Center for Computational Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthew Heflin
- Center for Computational Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Soojung Na
- Center for Computational Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ambereen Kidwai
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Dallas, TX, USA
| | - Natalie Booker
- Department of Pharmacy Practice, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Dallas, TX, USA
| | - William C. Putnam
- Department of Pharmacy Practice, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Dallas, TX, USA
| | - Vincenzo G. Fiore
- Center for Computational Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Xiaosi Gu
- Center for Computational Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
15
|
Downar J, Siddiqi SH, Mitra A, Williams N, Liston C. Mechanisms of Action of TMS in the Treatment of Depression. Curr Top Behav Neurosci 2024; 66:233-277. [PMID: 38844713 DOI: 10.1007/7854_2024_483] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/26/2024]
Abstract
Transcranial magnetic stimulation (TMS) is entering increasingly widespread use in treating depression. The most common stimulation target, in the dorsolateral prefrontal cortex (DLPFC), emerged from early neuroimaging studies in depression. Recently, more rigorous casual methods have revealed whole-brain target networks and anti-networks based on the effects of focal brain lesions and focal brain stimulation on depression symptoms. Symptom improvement during therapeutic DLPFC-TMS appears to involve directional changes in signaling between the DLPFC, subgenual and dorsal anterior cingulate cortex, and salience-network regions. However, different networks may be involved in the therapeutic mechanisms for other TMS targets in depression, such as dorsomedial prefrontal cortex or orbitofrontal cortex. The durability of therapeutic effects for TMS involves synaptic neuroplasticity, and specifically may depend upon dopamine acting at the D1 receptor family, as well as NMDA-receptor-dependent synaptic plasticity mechanisms. Although TMS protocols are classically considered 'excitatory' or 'inhibitory', the actual effects in individuals appear quite variable, and might be better understood at the level of populations of synapses rather than individual synapses. Synaptic meta-plasticity may provide a built-in protective mechanism to avoid runaway facilitation or inhibition during treatment, and may account for the relatively small number of patients who worsen rather than improve with TMS. From an ethological perspective, the antidepressant effects of TMS may involve promoting a whole-brain attractor state associated with foraging/hunting behaviors, centered on the rostrolateral periaqueductal gray and salience network, and suppressing an attractor state associated with passive threat defense, centered on the ventrolateral periaqueductal gray and default-mode network.
Collapse
Affiliation(s)
- Jonathan Downar
- Department of Psychiatry, Faculty of Medicine, Institute of Medical Science, University of Toronto, Toronto, ON, Canada.
- Temerty Centre for Therapeutic Brain Intervention, Centre for Addiction and Mental Health, Toronto, ON, Canada.
| | - Shan H Siddiqi
- Center for Brain Circuit Therapeutics, Brigham & Women's Hospital, Boston, MA, USA
- Department of Psychiatry, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Anish Mitra
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Nolan Williams
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Conor Liston
- Department of Psychiatry, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
16
|
Donnelly BM, Hsu DT, Gardus J, Wang J, Yang J, Parsey RV, DeLorenzo C. Orbitofrontal and striatal metabolism, volume, thickness and structural connectivity in relation to social anhedonia in depression: A multimodal study. Neuroimage Clin 2023; 41:103553. [PMID: 38134743 PMCID: PMC10777107 DOI: 10.1016/j.nicl.2023.103553] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 11/10/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023]
Abstract
BACKGROUND Social anhedonia is common within major depressive disorder (MDD) and associated with worse treatment outcomes. The orbitofrontal cortex (OFC) is implicated in both reward (medial OFC) and punishment (lateral OFC) in social decision making. Therefore, to understand the biology of social anhedonia in MDD, medial/lateral OFC metabolism, volume, and thickness, as well as structural connectivity to the striatum, amygdala, and ventral tegmental area/nucleus accumbens were examined. A positive relationship between social anhedonia and these neurobiological outcomes in the lateral OFC was hypothesized, whereas an inverse relationship was hypothesized for the medial OFC. The association between treatment-induced changes in OFC neurobiology and depression improvement were also examined. METHODS 85 medication-free participants diagnosed with MDD were assessed with Wisconsin Schizotypy Scales to assess social anhedonia and received pretreatment simultaneous fluorodeoxyglucose positron emission tomography (FDG-PET) and magnetic resonance imaging (MRI), including structural and diffusion. Participants were then treated in an 8-week randomized placebo-controlled double-blind course of escitalopram. PET/MRI were repeated following treatment. Metabolic rate of glucose uptake was quantified from dynamic FDG-PET frames using Patlak graphical analysis. Structure (volume and cortical thickness) was quantified from structural MRI using Freesurfer. To assess structural connectivity, probabilistic tractography was performed on diffusion MRI and average FA was calculated within the derived tracts. Linear mixed models with Bonferroni correction were used to examine the relationships between variables. RESULTS A significantly negative linear relationship between pretreatment social anhedonia score and structural connectivity between the medial OFC and the amygdala (estimated coefficient: -0.006, 95 % CI: -0.0108 - -0.0012, p-value = 0.0154) was observed. However, this finding would not survive multiple comparisons correction. No strong evidence existed to show a significant linear relationship between pretreatment social anhedonia score and metabolism, volume, thickness, or structural connectivity to any of the regions examined. There was also no strong evidence to suggest significant linear relationships between improvement in depression and percent change in these variables. CONCLUSIONS Based on these multimodal findings, the OFC likely does not underlie social anhedonia in isolation and therefore should not be the sole target of treatment for social anhedonia. This is consistent with previous reports that other areas of the brain such as the amygdala and the striatum are highly involved in this behavior. Relatedly, amygdala-medial OFC structural connectivity could be a future target. The results of this study are crucial as, to our knowledge, they are the first to relate structure/function of the OFC with social anhedonia severity in MDD. Future work may need to involve a whole brain approach in order to develop therapeutics for social anhedonia.
Collapse
Affiliation(s)
| | - David T Hsu
- Department of Psychiatry and Behavioral Health, Stony Brook University, 100 Nicolls Rd, Stony Brook, NY 11794, USA
| | - John Gardus
- Department of Psychiatry and Behavioral Health, Stony Brook University, 100 Nicolls Rd, Stony Brook, NY 11794, USA
| | - Junying Wang
- Department of Applied Mathematics and Statistics, Stony Brook University, 100 Nicolls Rd, Stony Brook, NY 11794, USA
| | - Jie Yang
- Department of Family, Population & Preventive Medicine, Stony Brook University, 100 Nicolls Rd, Stony Brook, NY 11794, USA
| | - Ramin V Parsey
- Department of Psychiatry and Behavioral Health, Stony Brook University, 100 Nicolls Rd, Stony Brook, NY 11794, USA
| | - Christine DeLorenzo
- Department of Psychiatry and Behavioral Health, Stony Brook University, 100 Nicolls Rd, Stony Brook, NY 11794, USA; Department of Biomedical Engineering, Stony Brook University, 100 Nicolls Rd, Stony Brook, NY 11794, USA.
| |
Collapse
|
17
|
Michel K, Lutz-Beck D, Engeroff S. Improving the Therapeutic Relationship When Prescribing Antidepressants: A Pilot Study. Healthcare (Basel) 2023; 11:2825. [PMID: 37957970 PMCID: PMC10650839 DOI: 10.3390/healthcare11212825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 10/22/2023] [Accepted: 10/24/2023] [Indexed: 11/15/2023] Open
Abstract
Studies indicate that the quality of the doctor-patient relationship moderates the effect of pharmacotherapy. To enhance the quality of the therapeutic relationship in the pharmacotherapy of depression, we developed a brief manual with interactive materials for residents in psychiatry and their patients. In a pilot study at a psychiatric university hospital's outpatient department, we compared patient-centered treatment parameters of a first patient group treated as usual and a second patient group treated using the manual. The study had no influence on the choice of medication. In the manual group, patient satisfaction with the doctor-patient relationship increased significantly at the three-month follow-up. Depression parameters declined in both groups, without group differences. Continuation of antidepressant medication at six months was higher in the manual group. In conclusion, a simple intervention using written materials for doctors prescribing antidepressants improved doctors' and patients' satisfaction with treatment.
Collapse
Affiliation(s)
- Konrad Michel
- University Hospital of Psychiatry, University of Bern, CH-3008 Bern, Switzerland
| | | | | |
Collapse
|
18
|
Cheng CM, Li CT, Jeng JS, Chang WH, Lin WC, Chen MH, Bai YM, Tsai SJ, Su TP. Antidepressant effects of prolonged intermittent theta-burst stimulation monotherapy at the bilateral dorsomedial prefrontal cortex for medication and standard transcranial magnetic stimulation-resistant major depression: a three arm, randomized, double blind, sham-controlled pilot study. Eur Arch Psychiatry Clin Neurosci 2023; 273:1433-1442. [PMID: 36484844 PMCID: PMC9735131 DOI: 10.1007/s00406-022-01523-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 11/15/2022] [Indexed: 12/13/2022]
Abstract
The dorsomedial prefrontal cortex (DMPFC) plays a pivotal role in depression and anxiosomatic symptom modulation. However, DMPFC stimulation using a double-cone coil has demonstrated inconsistent results for antidepressant efficacy. No study thus far has investigated the antidepressant and anti-anxiosomatic effects of prolonged intermittent theta-burst stimulation (piTBS) bilaterally over DMPFC. Furthermore, head-to-head comparisons of antidepressant effects between standard iTBS and piTBS warrant investigation. This double-blind, randomized, sham-controlled trial recruited 34 patients with highly treatment-resistant depression (TRD) unresponsive to antidepressants and standard repetitive transcranial magnetic stimulation (rTMS)/piTBS. These patients were randomly assigned to one of three monotherapy groups (standard iTBS, piTBS, or sham), with treatment administered bilaterally over the DMPFC twice per day for 3 weeks. The primary outcome was the overall changes of 17-item Hamilton Depression Rating Scale (HDRS-17) over 3-weeks intervention. The changes in Depression and Somatic Symptoms Scale (DSSS) as the secondary outcome and the anxiosomatic cluster symptoms as rated by HDRS-17 as the post-hoc outcome were measured. Multivariable generalized estimating equation analysis was performed. Although no differences in overall HDRS-17 changes between three groups were found, the antidepressant efficacy based on DSSS was higher in piTBS than in iTBS and sham at week 3 (group effect,p = 0.003, post-hoc: piTBS > iTBS, p = 0.002; piTBS > sham, p = 0.038). In post-hoc analyses, piTBS had more alleviation in anxiosomatic symptoms than iTBS (group effect, p = 0.002; post-hoc, p = 0.001). This first randomized sham-controlled study directly compared piTBS and iTBS targeting the DMPFC using a figure-of-8 coil and found piTBS may fail to demonstrate a significant antidepressant effect on overall depressive symptoms, but piTBS seems superior in alleviating anxiosomatic symptoms, even in depressed patients with high treatment resistance. This Trial registration (Registration number: NCT04037592). URL: https://clinicaltrials.gov/ct2/show/NCT04037592 .
Collapse
Affiliation(s)
- Chih-Ming Cheng
- Institute of Brain Science, School of Medicine, National Yang-Ming Chiao-Tung University, Taipei, Taiwan
- Division of Psychiatry, School of Medicine, National Yang-Ming Chiao-Tung University, Taipei, Taiwan
- Department of Psychiatry, Taipei Veterans General Hospital, Beitou District, No. 201, Sec. 2, Shih-Pai Road, Taipei, 112, Taiwan
| | - Cheng-Ta Li
- Institute of Brain Science, School of Medicine, National Yang-Ming Chiao-Tung University, Taipei, Taiwan.
- Division of Psychiatry, School of Medicine, National Yang-Ming Chiao-Tung University, Taipei, Taiwan.
- Department of Psychiatry, Taipei Veterans General Hospital, Beitou District, No. 201, Sec. 2, Shih-Pai Road, Taipei, 112, Taiwan.
- Institute of Cognitive Neuroscience, National Central University, Jhongli, Taiwan.
| | - Jia-Shyun Jeng
- Department of Psychiatry, Taipei Veterans General Hospital, Beitou District, No. 201, Sec. 2, Shih-Pai Road, Taipei, 112, Taiwan
- Department of Psychiatry, Kaohsiung Veterans General Hospital, Pingtung Branch, Pingtung, Taiwan
| | - Wen-Han Chang
- Department of Psychiatry, Taipei Veterans General Hospital, Beitou District, No. 201, Sec. 2, Shih-Pai Road, Taipei, 112, Taiwan
- Graduate Institute of Statistics National Central University, Taoyuan, Taiwan
| | - Wei-Chen Lin
- Institute of Brain Science, School of Medicine, National Yang-Ming Chiao-Tung University, Taipei, Taiwan
- Division of Psychiatry, School of Medicine, National Yang-Ming Chiao-Tung University, Taipei, Taiwan
- Department of Psychiatry, Taipei Veterans General Hospital, Beitou District, No. 201, Sec. 2, Shih-Pai Road, Taipei, 112, Taiwan
| | - Mu-Hong Chen
- Institute of Brain Science, School of Medicine, National Yang-Ming Chiao-Tung University, Taipei, Taiwan
- Division of Psychiatry, School of Medicine, National Yang-Ming Chiao-Tung University, Taipei, Taiwan
- Department of Psychiatry, Taipei Veterans General Hospital, Beitou District, No. 201, Sec. 2, Shih-Pai Road, Taipei, 112, Taiwan
| | - Ya-Mei Bai
- Institute of Brain Science, School of Medicine, National Yang-Ming Chiao-Tung University, Taipei, Taiwan
- Division of Psychiatry, School of Medicine, National Yang-Ming Chiao-Tung University, Taipei, Taiwan
- Department of Psychiatry, Taipei Veterans General Hospital, Beitou District, No. 201, Sec. 2, Shih-Pai Road, Taipei, 112, Taiwan
| | - Shih-Jen Tsai
- Institute of Brain Science, School of Medicine, National Yang-Ming Chiao-Tung University, Taipei, Taiwan
- Division of Psychiatry, School of Medicine, National Yang-Ming Chiao-Tung University, Taipei, Taiwan
- Department of Psychiatry, Taipei Veterans General Hospital, Beitou District, No. 201, Sec. 2, Shih-Pai Road, Taipei, 112, Taiwan
| | - Tung-Ping Su
- Institute of Brain Science, School of Medicine, National Yang-Ming Chiao-Tung University, Taipei, Taiwan
- Division of Psychiatry, School of Medicine, National Yang-Ming Chiao-Tung University, Taipei, Taiwan
- Department of Psychiatry, Taipei Veterans General Hospital, Beitou District, No. 201, Sec. 2, Shih-Pai Road, Taipei, 112, Taiwan
- Department of Psychiatry, Cheng Hsin General Hospital, Taipei, Taiwan
| |
Collapse
|
19
|
Chatterjee SA, Seidler RD, Skinner JW, Lysne PE, Sumonthee C, Wu SS, Cohen RA, Rose DK, Woods AJ, Clark DJ. Effects of Prefrontal Transcranial Direct Current Stimulation on Retention of Performance Gains on an Obstacle Negotiation Task in Older Adults. Neuromodulation 2023; 26:829-839. [PMID: 35410769 PMCID: PMC9547038 DOI: 10.1016/j.neurom.2022.02.231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 02/16/2022] [Accepted: 02/18/2022] [Indexed: 11/28/2022]
Abstract
OBJECTIVES Complex walking in older adults can be improved with task practice and might be further enhanced by pairing transcranial direct current stimulation (tDCS) to the dorsolateral prefrontal cortex. We tested the hypothesis that a single session of practice of a complex obstacle negotiation task paired with active tDCS in older adults would produce greater within-session improvements in walking performance and retention of gains, compared to sham tDCS and no tDCS conditions. MATERIALS AND METHODS A total of 50 older adults (mean age = 74.46 years ± 6.49) with self-reported walking difficulty were randomized to receive either active tDCS (active-tDCS group) or sham tDCS (sham-tDCS group) bilaterally to the dorsolateral prefrontal cortex or no tDCS (no-tDCS group). Each group performed ten practice trials of an obstacle negotiation task at their fastest safe speed. Retention of gains in walking performance was assessed with three trials conducted one week later. Within-session effects of practice and between-session retention effects on obstacle negotiation speed were examined. RESULTS At the practice session, all three groups exhibited significant within-session gains in walking speed (p ≤ 0.005). However, the gains were significantly greater in the sham-tDCS group than in the active-tDCS and no-tDCS groups (p ≤ 0.03) and were comparable between the active-tDCS and no-tDCS groups (p = 0.89). At one-week follow-up, the active-tDCS group exhibited significant between-session retention of gains and continued "offline" improvement in walking speed (p = 0.005). The active-tDCS group showed significantly greater retention of gains than the no-tDCS (p = 0.02) but not the sham-tDCS group (p = 0.24). CONCLUSIONS Pairing prefrontal active tDCS with a single session of obstacle negotiation practice may enhance one-week retention of gains in walking performance compared to no tDCS. However, the evidence is insufficient to suggest a benefit of active tDCS over sham tDCS for enhancing the gains in walking performance. Additional studies with a multisession intervention design and larger sample size are needed to further investigate these findings. CLINICAL TRIAL REGISTRATION The Clinicaltrials.gov registration number for the study is NCT03122236.
Collapse
Affiliation(s)
- Sudeshna A Chatterjee
- Department of Aging and Geriatric Research, University of Florida, Gainesville, FL, USA; Brain Rehabilitation Research Center, Malcom Randall VA Medical Center, Gainesville, FL, USA.
| | - Rachael D Seidler
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA
| | - Jared W Skinner
- Geriatric Research, Education, and Clinical Center, Malcom Randall VA Medical Center, Gainesville, FL, USA
| | - Paige E Lysne
- Department of Aging and Geriatric Research, University of Florida, Gainesville, FL, USA
| | - Chanoan Sumonthee
- College of Public Health and Health Professions, University of Florida, Gainesville, FL, USA
| | - Samuel S Wu
- Department of Biostatistics, University of Florida, Gainesville, FL, USA
| | - Ronald A Cohen
- Department of Clinical and Health Psychology, Center for Cognitive Aging and Memory, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Dorian K Rose
- Brain Rehabilitation Research Center, Malcom Randall VA Medical Center, Gainesville, FL, USA; Department of Physical Therapy, University of Florida, Gainesville, FL, USA; Brooks Rehabilitation, Jacksonville, FL, USA
| | - Adam J Woods
- Department of Clinical and Health Psychology, Center for Cognitive Aging and Memory, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - David J Clark
- Department of Aging and Geriatric Research, University of Florida, Gainesville, FL, USA; Brain Rehabilitation Research Center, Malcom Randall VA Medical Center, Gainesville, FL, USA
| |
Collapse
|
20
|
Zhao K, Xie H, Fonzo GA, Tong X, Carlisle N, Chidharom M, Etkin A, Zhang Y. Individualized fMRI connectivity defines signatures of antidepressant and placebo responses in major depression. Mol Psychiatry 2023; 28:2490-2499. [PMID: 36732585 DOI: 10.1038/s41380-023-01958-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 01/04/2023] [Accepted: 01/11/2023] [Indexed: 02/04/2023]
Abstract
Though sertraline is commonly prescribed in patients with major depressive disorder (MDD), its superiority over placebo is only marginal. This is in part due to the neurobiological heterogeneity of the individuals. Characterizing individual-unique functional architecture of the brain may help better dissect the heterogeneity, thereby defining treatment-predictive signatures to guide personalized medication. In this study, we investigate whether individualized brain functional connectivity (FC) can define more predictable signatures of antidepressant and placebo treatment in MDD. The data used in the present work were collected by the Establishing Moderators and Biosignatures of Antidepressant Response in Clinical Care (EMBARC) study. Patients (N = 296) were randomly assigned to antidepressant sertraline or placebo double-blind treatment for 8 weeks. The whole-brain FC networks were constructed from pre-treatment resting-state functional magnetic resonance imaging (rs-fMRI). Then, FC was individualized by removing the common components extracted from the raw baseline FC to train regression-based connectivity predictive models. With individualized FC features, the established prediction models successfully identified signatures that explained 22% variance for the sertraline group and 31% variance for the placebo group in predicting HAMD17 change. Compared with the raw FC-based models, the individualized FC-defined signatures significantly improved the prediction performance, as confirmed by cross-validation. For sertraline treatment, predictive FC metrics were predominantly located in the left middle temporal cortex and right insula. For placebo, predictive FC metrics were primarily located in the bilateral cingulate cortex and left superior temporal cortex. Our findings demonstrated that through the removal of common FC components, individualization of FC metrics enhanced the prediction performance compared to raw FC. Associated with previous MDD clinical studies, our identified predictive biomarkers provided new insights into the neuropathology of antidepressant and placebo treatment.
Collapse
Affiliation(s)
- Kanhao Zhao
- Department of Bioengineering, Lehigh University, Bethlehem, PA, USA
| | - Hua Xie
- Center for Neuroscience Research, Children's National Hospital, Washington, DC, USA
| | - Gregory A Fonzo
- Center for Psychedelic Research and Therapy, Department of Psychiatry and Behavioral Sciences, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - Xiaoyu Tong
- Department of Bioengineering, Lehigh University, Bethlehem, PA, USA
| | - Nancy Carlisle
- Department of Psychology, Lehigh University, Bethlehem, PA, USA
| | | | - Amit Etkin
- Alto Neuroscience, Inc, Los Altos, CA, USA
| | - Yu Zhang
- Department of Bioengineering, Lehigh University, Bethlehem, PA, USA.
- Department of Electrical and Computer Engineering, Lehigh University, Bethlehem, PA, USA.
| |
Collapse
|
21
|
Affiliation(s)
- Shan H Siddiqi
- Center for Brain Circuit Therapeutics, Brigham and Women's Hospital, and Department of Psychiatry, Harvard Medical School, Boston (Siddiqi); Center for Neurorestoration and Neurotechnology, Providence VA Healthcare System, and Department of Psychiatry and Human Behavior, Alpert Medical School of Brown University, Providence, R.I. (Philip)
| | - Noah S Philip
- Center for Brain Circuit Therapeutics, Brigham and Women's Hospital, and Department of Psychiatry, Harvard Medical School, Boston (Siddiqi); Center for Neurorestoration and Neurotechnology, Providence VA Healthcare System, and Department of Psychiatry and Human Behavior, Alpert Medical School of Brown University, Providence, R.I. (Philip)
| |
Collapse
|
22
|
Kang H, Miksche MS, Ellingsen DM. Association between personality traits and placebo effects: a preregistered systematic review and meta-analysis. Pain 2023; 164:494-508. [PMID: 35947877 DOI: 10.1097/j.pain.0000000000002753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 08/02/2022] [Indexed: 11/26/2022]
Abstract
ABSTRACT Placebo effects are ubiquitous yet highly variable between individuals and therefore strongly affect clinical trial outcomes such as pain relief. It is unclear whether dispositional psychological traits influence responsiveness to placebo. This preregistered meta-analysis and systematic review synthesized the literature investigating the association between personality traits and placebo effects. Based on 21 studies with 798 participants, we performed formal meta-analyses for 10 different personality traits, including behavioral inhibition, fun seeking, goal-drive persistence, reward responsiveness, empathic concern, empathic fantasy, perspective-taking, personal distress, optimism, and anxiety. We did not find evidence of associations between any of these traits and magnitude of placebo effects, which was supported by equivalence tests. Furthermore, we did not find evidence for moderating factors such as placebo manipulation type (conditioning or nonconditioning) or condition (pain or nonpain). These findings challenge the notion that personality influences responsiveness to placebos and contradict its utility for identifying placebo "responders" and "nonresponders."
Collapse
Affiliation(s)
- Heemin Kang
- Department of Psychology, University of Oslo, Norway
- Norwegian Centre for Mental Disorders Research, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | | | - Dan-Mikael Ellingsen
- Department of Psychology, University of Oslo, Norway
- Division of Radiology and Nuclear Medicine, Department of Physics and Computational Radiology, Oslo University Hospital, Oslo, Norway
- School of Health Sciences, Kristiania University College, Oslo, Norway
| |
Collapse
|
23
|
Deng Y, Li W, Zhang B. Functional Activity in the Effect of Transcranial Magnetic Stimulation Therapy for Patients with Depression: A Meta-Analysis. J Pers Med 2023; 13:405. [PMID: 36983590 PMCID: PMC10051603 DOI: 10.3390/jpm13030405] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 02/22/2023] [Accepted: 02/22/2023] [Indexed: 03/03/2023] Open
Abstract
Depression is a long-lasting mental disorder that affects more than 264 million people worldwide. Transcranial magnetic stimulation (TMS) can be a safe and effective choice for the treatment of depression. Functional neuroimaging provides unique insights into the neuropsychiatric effects of antidepressant TMS. In this meta-analysis, we aimed to assess the functional activity of brain regions caused by TMS for depression. A literature search was conducted from inception to 5 January 2022. Studies were then selected according to predetermined inclusion and exclusion criteria. Activation likelihood estimation was applied to analyze functional activation. Five articles were ultimately included after selection. The main analysis results indicated that TMS treatment for depression can alter the activity in the right precentral gyrus, right posterior cingulate, left inferior frontal gyrus and left middle frontal gyrus. In resting-state studies, increased activation was shown in the right precentral gyrus, right posterior cingulate, left inferior frontal gyrus and left superior frontal gyrus associated with TMS treatment. In task-related studies, clusters in the right middle frontal gyrus, left sub-gyrus, left middle frontal gyrus and left posterior cingulate were hyperactivated post-treatment. Our study offers an overview of brain activity changes in patients with depression after TMS treatment.
Collapse
Affiliation(s)
- Yongyan Deng
- The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou 510370, China
- Peking University Sixth Hospital, Beijing 100191, China
| | - Wenyue Li
- The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou 510370, China
| | - Bin Zhang
- Institute of Mental Health, Tianjin Anding Hospital, Tianjin Medical University, Tianjin 300222, China
| |
Collapse
|
24
|
Nowak M, Schindler S, Storch M, Geyer S, Schönknecht P. Mammillary body and hypothalamic volumes in mood disorders. J Psychiatr Res 2023; 158:216-225. [PMID: 36603316 DOI: 10.1016/j.jpsychires.2022.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 10/20/2022] [Accepted: 12/10/2022] [Indexed: 12/15/2022]
Abstract
We have previously reported an in vivo enlargement of the left hypothalamus in mood disorders using 7 T magnetic resonance imaging. The aim of this follow-up study was to find out whether the hypothalamic volume difference may be located in the mammillary bodies (MB) rather than being widespread across the hypothalamus. We developed and evaluated a detailed segmentation algorithm that allowed a reliable segmentation of the MBs, and applied it to 20 unmedicated (MDDu) and 20 medicated patients with major depressive disorder, 21 medicated patients with bipolar disorder, and 23 controls. 20 out of 23 healthy controls were matched to the MDDu. We tested for group differences in MB and hypothalamus without MB (HTh) volumes using analyses of covariance. Associations between both volumes of interest were analysed using bivariate and partial correlations. In contrast to postmortem findings, we found no statistically significant differences of the MB volumes between the study groups. Left HTh volumes differed significantly across the study groups after correction for intracranial volume (ICV) and for ICV and sex. Our result of an HTh enlargement in mood disorders was confirmed by a paired t-test between the matched pairs of MDDu and healthy controls using the native MB and HTh volumes. In the whole sample, MB volumes correlated significantly with the ipsilateral HTh volumes. Our results indicate a structural relationship between both volumes, and that our previous in vivo finding of a hypothalamus enlargement does not extend to the MB, but is limited to the HTh. The enlargement is more likely related to the dysregulation of the HPA axis than to cognitive dysfunctions accompanying mood disorders.
Collapse
Affiliation(s)
- Markus Nowak
- University Hospital Leipzig, Department of Psychiatry and Psychotherapy, Semmelweisstraße 10, 04103, Leipzig, Germany; Charité University of Medicine, Department of Psychiatry and Psychotherapy and St. Hedwig Hospital Berlin, Große Hamburger Straße 5-11, 10115, Berlin, Germany.
| | - Stephanie Schindler
- University Hospital Leipzig, Department of Psychiatry and Psychotherapy, Semmelweisstraße 10, 04103, Leipzig, Germany
| | - Melanie Storch
- University Hospital Leipzig, Department of Psychiatry and Psychotherapy, Semmelweisstraße 10, 04103, Leipzig, Germany
| | - Stefan Geyer
- Max Planck Institute for Human Cognitive and Brain Sciences, Department of Neurophysics, Stephanstraße 1a, 04103, Leipzig, Germany
| | - Peter Schönknecht
- University Hospital Leipzig, Department of Psychiatry and Psychotherapy, Semmelweisstraße 10, 04103, Leipzig, Germany; University Hospital Leipzig, Out-patient Department for Sexual-therapeutic Prevention and Forensic Psychiatry, Semmelweisstraße 10, 04103, Leipzig, Germany; Academic State Hospital Arnsdorf, Hufelandstraße 15, 01477, Arnsdorf, Germany
| |
Collapse
|
25
|
Nie J, Wei Q, Bai T, Zhang T, Lv H, Zhang L, Ji G, Yu F, Tian Y, Wang K. Electroconvulsive therapy changes temporal dynamics of intrinsic brain activity in depressed patients. Psychiatry Res 2022; 316:114732. [PMID: 35926361 DOI: 10.1016/j.psychres.2022.114732] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 07/14/2022] [Accepted: 07/16/2022] [Indexed: 11/24/2022]
Abstract
Electroconvulsive therapy (ECT) has been demonstrated to be effective in treating depressed patients. Previous neuroimaging studies have focused mainly on alterations in static brain activity and connectivity to study the effects of ECT in depressed patients. However, it remains unclear whether the temporal dynamics of brain activity are associated with mechanisms of ECT in depressed patients. We measured the dynamics of spontaneous brain activity using dynamic amplitude of low-frequency fluctuation (dALFF) in healthy controls (n = 40) and patients diagnosed with unipolar depression (UD, n = 36) or bipolar disorder (BD, n = 9) before and after ECT. Furthermore, the temporal variability of intrinsic brain activity (iBA) was quantified as the variance of dALFF across sliding window. In addition, correlation analysis was performed to investigate the relationships among dALFF, depressive symptoms, and cognitive function in depressed patients. We lack second resting-state functional magnetic resonance imaging (rs-fMRI) data for healthy controls. After ECT, patients showed decreased brain dynamics (less temporal variability) in the right dorsal anterior cingulate cortex (dACC) and the right precuneus, whereas they showed increased brain dynamics in the bilateral superior medial frontal cortex (mSFC). No significant correlation was found between the dALFF and clinical variables in depressed patients. Our findings suggest that right dACC, right precuneus, and bilateral mSFC play an important role in response to ECT depressed patients from the perspective of dynamic local brain activity, indicating that the dALFF variability may be useful in further understanding the mechanisms of ECT's antidepressant effects.
Collapse
Affiliation(s)
- Jiajia Nie
- Department of Psychology and Sleep Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui, China; Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, Anhui, China.
| | - Qiang Wei
- Department of Neurology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui, China; Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, Anhui, China
| | - Tongjian Bai
- Department of Neurology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui, China; Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, Anhui, China
| | - Ting Zhang
- Department of Neurology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui, China; Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, Anhui, China
| | - Huaming Lv
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui, China
| | - Li Zhang
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui, China
| | - Gongjun Ji
- Department of Neurology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui, China; Department of medical psychology, Anhui Medical University, Hefei, Anhui, China; Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, Anhui, China
| | - Fengqiong Yu
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui, China; Department of medical psychology, Anhui Medical University, Hefei, Anhui, China; Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, Anhui, China
| | - Yanghua Tian
- Department of Psychology and Sleep Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Department of Neurology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui, China; Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, Anhui, China.
| | - Kai Wang
- Department of Neurology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui, China; Department of medical psychology, Anhui Medical University, Hefei, Anhui, China; Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, Anhui, China.
| |
Collapse
|
26
|
Ang YS, Bruder GE, Keilp JG, Rutherford A, Alschuler DM, Pechtel P, Webb CA, Carmody T, Fava M, Cusin C, McGrath PJ, Weissman M, Parsey R, Oquendo MA, McInnis MG, Cooper CM, Deldin P, Trivedi MH, Pizzagalli DA. Exploration of baseline and early changes in neurocognitive characteristics as predictors of treatment response to bupropion, sertraline, and placebo in the EMBARC clinical trial. Psychol Med 2022; 52:2441-2449. [PMID: 33213541 PMCID: PMC7613805 DOI: 10.1017/s0033291720004286] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Treatment for major depressive disorder (MDD) is imprecise and often involves trial-and-error to determine the most effective approach. To facilitate optimal treatment selection and inform timely adjustment, the current study investigated whether neurocognitive variables could predict an antidepressant response in a treatment-specific manner. METHODS In the two-stage Establishing Moderators and Biosignatures of Antidepressant Response for Clinical Care (EMBARC) trial, outpatients with non-psychotic recurrent MDD were first randomized to an 8-week course of sertraline selective serotonin reuptake inhibitor or placebo. Behavioral measures of reward responsiveness, cognitive control, verbal fluency, psychomotor, and cognitive processing speeds were collected at baseline and week 1. Treatment responders then continued on another 8-week course of the same medication, whereas non-responders to sertraline or placebo were crossed-over under double-blinded conditions to bupropion noradrenaline/dopamine reuptake inhibitor or sertraline, respectively. Hamilton Rating for Depression scores were also assessed at baseline, weeks 8, and 16. RESULTS Greater improvements in psychomotor and cognitive processing speeds within the first week, as well as better pretreatment performance in these domains, were specifically associated with higher likelihood of response to placebo. Moreover, better reward responsiveness, poorer cognitive control and greater verbal fluency were associated with greater likelihood of response to bupropion in patients who previously failed to respond to sertraline. CONCLUSION These exploratory results warrant further scrutiny, but demonstrate that quick and non-invasive behavioral tests may have substantial clinical value in predicting antidepressant treatment response.
Collapse
Affiliation(s)
- Yuen-Siang Ang
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, USA
- Center for Depression, Anxiety and Stress Research, McLean Hospital, Belmont, Massachusetts, USA
| | - Gerard E. Bruder
- Department of Psychiatry, New York State Psychiatric Institute and Columbia University Vagelos College of Physicians and Surgeons, New York, USA
| | - John G. Keilp
- Department of Psychiatry, New York State Psychiatric Institute and Columbia University Vagelos College of Physicians and Surgeons, New York, USA
| | - Ashleigh Rutherford
- Center for Depression, Anxiety and Stress Research, McLean Hospital, Belmont, Massachusetts, USA
| | - Daniel M. Alschuler
- Department of Psychiatry, New York State Psychiatric Institute and Columbia University Vagelos College of Physicians and Surgeons, New York, USA
| | - Pia Pechtel
- Center for Depression, Anxiety and Stress Research, McLean Hospital, Belmont, Massachusetts, USA
| | - Christian A. Webb
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, USA
- Center for Depression, Anxiety and Stress Research, McLean Hospital, Belmont, Massachusetts, USA
| | - Thomas Carmody
- Department of Psychiatry, University of Texas, Southwestern Medical Center, Dallas, Texas, USA
| | - Maurizio Fava
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, USA
- Department of Psychiatry, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Cristina Cusin
- Department of Psychiatry, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Patrick J. McGrath
- Department of Psychiatry, New York State Psychiatric Institute and Columbia University Vagelos College of Physicians and Surgeons, New York, USA
| | - Myrna Weissman
- Department of Psychiatry, New York State Psychiatric Institute and Columbia University Vagelos College of Physicians and Surgeons, New York, USA
| | - Ramin Parsey
- Department of Psychiatry, Stony Brook University, Stony Brook, New York, USA
| | - Maria A. Oquendo
- Department of Psychiatry, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Melvin G. McInnis
- Department of Psychiatry, University of Michigan, Ann Arbor, Michigan, USA
| | - Crystal M. Cooper
- Department of Psychiatry, University of Texas, Southwestern Medical Center, Dallas, Texas, USA
| | - Patricia Deldin
- Department of Psychiatry, University of Michigan, Ann Arbor, Michigan, USA
| | - Madhukar H. Trivedi
- Department of Psychiatry, University of Texas, Southwestern Medical Center, Dallas, Texas, USA
| | - Diego A. Pizzagalli
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, USA
- Center for Depression, Anxiety and Stress Research, McLean Hospital, Belmont, Massachusetts, USA
- McLean Imaging Center, McLean Hospital, Belmont, Massachusetts, USA
| |
Collapse
|
27
|
Butler M, Jelen L, Rucker J. Expectancy in placebo-controlled trials of psychedelics: if so, so what? Psychopharmacology (Berl) 2022; 239:3047-3055. [PMID: 36063208 PMCID: PMC9481484 DOI: 10.1007/s00213-022-06221-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 08/23/2022] [Indexed: 11/30/2022]
Abstract
Modern psychedelic research remains in an early phase, and the eventual introduction of psychedelics into clinical practice remains in doubt. In this piece, we discuss the role of blinding and expectancy in psychedelic trials, and place this in a broader historical and contemporary context of blinding in trials across the rest of healthcare. We suggest that premature and uncritical promotion ('hype') of psychedelics as medicines is not only misleading, but also directly influences participant expectancy in ongoing psychedelic trials. We argue that although psychedelic trials are likely to significantly overestimate treatment effects by design due to unblinding and expectancy effects, this is not a unique situation. Placebo-controlled RCTs are not a perfect fit for all therapeutics, and problems in blinding should not automatically disqualify medications from licencing decisions. We suggest that simple practical measures may be (and indeed already are) taken in psychedelic trials to partially mitigate the effects of expectancy and unblinding, such as independent raters and active placebos. We briefly suggest other alternative trial methodologies which could be used to bolster RCT results, such as naturalistic studies. We conclude that the results of contemporary placebo-controlled RCTs of psychedelics should neither be dismissed due to imperfections in design, nor should early data be taken as firm evidence of effectiveness.
Collapse
Affiliation(s)
- Matt Butler
- Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK.
| | - Luke Jelen
- Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
| | - James Rucker
- Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
| |
Collapse
|
28
|
Pardo-Cabello AJ, Manzano-Gamero V, Puche-Cañas E. Placebo: a brief updated review. Naunyn Schmiedebergs Arch Pharmacol 2022; 395:1343-1356. [PMID: 35943515 PMCID: PMC9361274 DOI: 10.1007/s00210-022-02280-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 08/03/2022] [Indexed: 11/04/2022]
Abstract
Our aims were to provide updated information on placebo/nocebo effect and the potential use of placebo in clinical practice. This article can only provide a rough overview on the placebo and nocebo effect and is intended to serve as a starting point for the reader to go deeper into the corresponding literature. The placebo effect has been observed in multiple medical conditions, after oral administration, with manual therapies as well as with surgery and invasive procedures. The use of placebo in clinical trials is fundamental, although the ethics of its use is under discussion. The placebo may behave like a drug from the pharmacokinetic and pharmacodynamic point of view and can also be associated with adverse events (nocebo effect). Placebo can modify treatment by increasing or decreasing the effects of drugs. The factors associated with the occurrence of placebo effect are multiple, but in addition to those that depend on the placebo itself, the doctor-patient relationship would be the most important. As a result of findings that were published in the last two decades, the psycho-neurobiological basis of placebo is becoming better understood, although further studies are needed. In conclusion, the placebo effect in the clinic exhibits weak to moderate intensity. Placebo, in addition to its use in the clinical trial, should be considered another therapeutic remedy either as stand alone or in association with treatment, and could be useful in certain circumstances. The use of placebo should be regulated by the European health authorities through a guide in clinical practice that will improve patient care.
Collapse
Affiliation(s)
- Alfredo Jose Pardo-Cabello
- Department of Internal Medicine, Hospital Universitario San Cecilio, Avda. de La Innovación, s/n, 18016, Granada, Spain.
| | - Victoria Manzano-Gamero
- Department of Internal Medicine, Hospital Universitario Virgen de Las Nieves, Granada, Spain
| | - Emilio Puche-Cañas
- Department of Pharmacology, School of Medicine, University of Granada, Granada, Spain
| |
Collapse
|
29
|
Ohnishi T, Wakamatsu A, Kobayashi H. Different symptomatic improvement pattern revealed by factor analysis between placebo response and response to Esketamine in treatment resistant depression. Psychiatry Clin Neurosci 2022; 76:377-383. [PMID: 35596932 DOI: 10.1111/pcn.13379] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 04/14/2022] [Accepted: 05/08/2022] [Indexed: 11/28/2022]
Abstract
AIMS The aim of this study is to determine whether there is difference in the change in each symptom of depression and in symptomatic improvement pattern between placebo and antidepressant responses. METHODS Using data from a randomized, double-blind (DB), placebo-controlled trial of esketamine (ESK) in patients with treatment-resistant depression (TRD), we conducted exploratory analyses. To determine differences in the change in each depressive symptom on the MADRS subscale between placebo and antidepressant responses, a two-way factorial analysis was conducted using the amount of change on Day 2 and 28 of treatment. In addition, exploratory and confirmatory factor analyses were conducted on the MADRS subtotal variables on Day 2 and 28 of treatment to determine symptomatic improvement pattern between placebo response and antidepressant responses. RESULTS We found that as well as MADRS total score, each subscale of MADRS score did not significantly differ between esketamine and placebo at Day 2 and 28. On the other hand, factor analysis revealed that the factor structure of the response was different between esketamine and placebo at the 2nd day. There was no difference in the factor structure between esketamine and placebo in response on Day 28 of treatment. CONCLUSION Factor analysis revealed different patterns of symptom improvement in the early phase of the intervention between esketamine and placebo. This finding suggests that a data driven approach may provide detailed efficacy information in clinical trials for antidepressants. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT02918318. Registered: 28 September 2016.
Collapse
Affiliation(s)
- Takashi Ohnishi
- Medical Affairs Division, Janssen Pharmaceutical K.K., Tokyo, Japan
| | | | - Hisanori Kobayashi
- Research and Development, Clinical Science Division, Janssen Pharmaceutical K.K., Tokyo, Japan
| |
Collapse
|
30
|
Wunram HL, Hamacher S, Oberste M, Neufang S, Belke L, Jänicke F, Graf C, Schönau E, Bender S, Fricke O. Influence of motivational placebo-related factors on the effects of exercise treatment in depressive adolescents. Eur Child Adolesc Psychiatry 2022; 31:1-14. [PMID: 33709258 PMCID: PMC9343287 DOI: 10.1007/s00787-021-01742-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 02/08/2021] [Indexed: 11/26/2022]
Abstract
Recent meta-analyses reveal a moderate effect of physical activity (PA) in the treatment of adolescent depression. However, not only the underlying neurobiological mechanisms, also the influences of placebo-related motivational factors (beliefs and expectancies in sporting, enjoyment and prior sports experiences), are still unclear. Based on the data of our prior study "Mood Vibes", we hypothesized that placebo-inherent factors like positive prior sports experiences and motivational factors, (positive beliefs, expectancies, and enjoyment related to PA), would increase the effects of an add-on exercise-therapy in juvenile depression. From 64 included depressed adolescents, 41 underwent an intensive add-on PA-therapy. Motivational factors were assessed using sport-specific scales. The changes in depression scores under treatment were rated by self-rating scale (German "Childhood Depression Inventory", (DIKJ)). A mixed model for repeated measures (MMRM) was used to analyze the effects of the different motivational variates on DIKJ. While prior sports experiences had no impact, motivational factors showed a significant effect on PA-induced changes in DIKJ scores (p = 0.002). The demotivated participants improved less, whereas it was sufficient to be neutral towards sporting to benefit significantly more. Motivational placebo-related factors (beliefs, expectancies and enjoyment regarding PA) affected the outcomes of an exercise treatment in depressed adolescents. Yet, a neutral mindset was sufficient to profit more from PA. Prior sporting in the sense of positive conditioning and as a protective factor did not play a role. Knowledge about these influences could in a second step help to develop tailored therapies.
Collapse
Affiliation(s)
- Heidrun Lioba Wunram
- Department of Child and Adolescent Psychiatry Psychosomatic and Psychotherapy, University Hospital of Cologne, Cologne, Germany
| | - Stefanie Hamacher
- Department of Medical Statistics, Informatics and Epidemiology (IMSIE), University of Cologne, Cologne, Germany
| | - Max Oberste
- Department of Medical Statistics, Informatics and Epidemiology (IMSIE), University of Cologne, Cologne, Germany
- Department for Molecular and Cellular Sports Medicine, German Sport University Cologne, Cologne, Germany
| | - Susanne Neufang
- Department of Psychiatry and Psychotherapy, Medical Faculty Heinrich-Heine University, Düsseldorf, Germany
| | - Luisa Belke
- Children’s Hospital Amsterdam Street, Cologne, Germany
| | | | - Christine Graf
- Institute of Movement and Neuroscience, German Sport University Cologne, Cologne, Germany
| | - Eckhard Schönau
- Children’s Hospital, University Hospital of Cologne and UniReha®, University Hospital of Cologne, Cologne, Germany
| | - Stephan Bender
- Department of Child and Adolescent Psychiatry Psychosomatic and Psychotherapy, University Hospital of Cologne, Cologne, Germany
| | - Oliver Fricke
- Department of Child and Adolescent Psychiatry, Psychotherapy and Child Neurology, Gemeinschaftskrankenhaus Herdecke and Chairs of Child and Adolescent Psychiatry, Witten/Herdecke University, Witten, Germany
| |
Collapse
|
31
|
Sheldon RS, Raj SR. Treating Syncope Without Drugs: Standing Still, Exercising Hard, or Simply the “Expert's Touch”? J Cardiovasc Electrophysiol 2022; 33:1871-1873. [DOI: 10.1111/jce.15576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 05/26/2022] [Indexed: 11/26/2022]
Affiliation(s)
- Robert S Sheldon
- Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of MedicineUniversity of CalgaryCalgaryABCanada
| | - Satish R Raj
- Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of MedicineUniversity of CalgaryCalgaryABCanada
| |
Collapse
|
32
|
Burke MJ, Romanella SM, Mencarelli L, Greben R, Fox MD, Kaptchuk TJ, Pascual-Leone A, Santarnecchi E. Placebo effects and neuromodulation for depression: a meta-analysis and evaluation of shared mechanisms. Mol Psychiatry 2022; 27:1658-1666. [PMID: 34903861 DOI: 10.1038/s41380-021-01397-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 10/31/2021] [Accepted: 11/23/2021] [Indexed: 12/20/2022]
Abstract
There is growing evidence that placebo effects can meaningfully modulate the brain. However, there has been little consideration of whether these changes may overlap with regions/circuits targeted by depression treatments and what the implications of this overlap would be on measuring efficacy in placebo-controlled clinical trials. In this systematic review and meta-analysis, we searched PubMed/Medline and Google Scholar for functional MRI and PET neuroimaging studies of placebo effects. Studies recruiting both healthy subjects and patient populations were included. Neuroimaging coordinates were extracted and included for Activation Likelihood Estimation (ALE) meta-analysis. We then searched for interventional studies of transcranial magnetic stimulation (TMS) and deep brain stimulation (DBS) for depression and extracted target coordinates for comparative spatial analysis with the placebo effects maps. Of 1169 articles identified, 34 neuroimaging studies of placebo effects were included. There were three significant clusters of activation: left dorsolateral prefrontal cortex (DLPFC) (x = -41, y = 16, z = 34), left sub-genual anterior cingulate cortex (sgACC)/ventral striatum (x = -8, y = 18, z = -15) and the right rostral anterior cingulate cortex (rACC) (x = 4, y = 42, z = 10). There were two significant deactivation clusters: right basal ganglia (x = 20, y = 2, z = 7) and right dorsal anterior cingulate cortex (dACC) (x = 1, y = -5, z = 45). TMS and DBS targets for depression treatment overlapped with the left DLPFC cluster and sgACC cluster, respectively. Our findings identify a common set of brain regions implicated in placebo effects across healthy individuals and patient populations, and provide evidence that these regions overlap with depression treatment targets. We model the statistical impacts of this overlap and demonstrate critical implications on measurements of clinical trial efficacy for this field.
Collapse
Affiliation(s)
- Matthew J Burke
- Neuropsychiatry Program, Department of Psychiatry and Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada. .,Harquail Centre for Neuromodulation and Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada. .,Berenson-Allen Center for Noninvasive Brain Stimulation, Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| | - Sara M Romanella
- Berenson-Allen Center for Noninvasive Brain Stimulation, Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.,Siena Brain Investigation & Neuromodulation Lab (Si-BIN Lab), Department of Medicine, Surgery and Neuroscience, Neurology and Clinical Neurophysiology Section, University of Siena, Siena, Italy
| | - Lucia Mencarelli
- Berenson-Allen Center for Noninvasive Brain Stimulation, Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.,Siena Brain Investigation & Neuromodulation Lab (Si-BIN Lab), Department of Medicine, Surgery and Neuroscience, Neurology and Clinical Neurophysiology Section, University of Siena, Siena, Italy
| | - Rachel Greben
- Harquail Centre for Neuromodulation and Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Michael D Fox
- Berenson-Allen Center for Noninvasive Brain Stimulation, Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.,Center for Brain Circuit Therapeutics, Departments of Neurology, Psychiatry, and Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Athinoula A. Martinos Centre for Biomedical Imaging, Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Ted J Kaptchuk
- Program in Placebo Studies, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Alvaro Pascual-Leone
- Department of Neurology, Harvard Medical School, Boston, MA, USA.,Hinda and Arthur Marcus Institute for Aging Research and Center for Memory Health, Hebrew SeniorLife, Boston, MA, USA.,Guttmann Brain Health Institut, Guttmann Institut, Universitat Autonoma, Barcelona, Spain
| | - Emiliano Santarnecchi
- Berenson-Allen Center for Noninvasive Brain Stimulation, Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA. .,Precision Neuroscience & Neuromodulation Program, Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
33
|
Jeong H, Song IU, Chung YA, Park JS, Na SH, Im JJ, Bikson M, Lee W, Yoo SS. Short-Term Efficacy of Transcranial Focused Ultrasound to the Hippocampus in Alzheimer’s Disease: A Preliminary Study. J Pers Med 2022; 12:jpm12020250. [PMID: 35207738 PMCID: PMC8878180 DOI: 10.3390/jpm12020250] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/27/2022] [Accepted: 02/03/2022] [Indexed: 11/17/2022] Open
Abstract
Preclinical studies have suggested that low-intensity transcranial focused ultrasound (tFUS) may have therapeutic potential for Alzheimer’s disease (AD) by opening the blood–brain barrier (BBB), reducing amyloid pathology, and improving cognition. This study investigated the effects of tFUS on BBB opening, regional cerebral metabolic rate of glucose (rCMRglu), and cognitive function in AD patients. Eight patients with AD received image-guided tFUS to the right hippocampus immediately after intravenous injection of microbubble ultrasound contrast agents. Patients completed magnetic resonance imaging (MRI), 18F-fluoro-2-deoxyglucose positron emission tomography (PET), and cognitive assessments before and after the sonication. No evidence of transient BBB opening was found on T1 dynamic contrast-enhanced MRI. However, immediate recall (p = 0.03) and recognition memory (p = 0.02) were significantly improved on the verbal learning test. PET image analysis demonstrated increased rCMRglu in the right hippocampus (p = 0.001). In addition, increases of hippocampal rCMRglu were correlated with improvement in recognition memory (Spearman’s ρ = 0.77, p = 0.02). No adverse event was observed. Our results suggest that tFUS to the hippocampus of AD patients may improve rCMRglu of the target area and memory in the short term, even without BBB opening. Further larger sham-controlled trials with loger follow-up are warranted to evaluate the efficacy and safety of tFUS in patients with AD.
Collapse
Affiliation(s)
- Hyeonseok Jeong
- Department of Nuclear Medicine, Incheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 21431, Korea; (H.J.); (J.J.I.)
- Department of Radiology, Incheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 21431, Korea
| | - In-Uk Song
- Department of Neurology, Incheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 21431, Korea; (J.-S.P.); (S.-H.N.)
- Correspondence: (I.-U.S.); (Y.-A.C.); Tel.: +82-32-280-5010 (I.-U.S.); +82-32-280-5242 (Y.-A.C.); Fax: +82-32-280-5244 (I.-U.S. & Y.-A.C.)
| | - Yong-An Chung
- Department of Nuclear Medicine, Incheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 21431, Korea; (H.J.); (J.J.I.)
- Department of Radiology, Incheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 21431, Korea
- Correspondence: (I.-U.S.); (Y.-A.C.); Tel.: +82-32-280-5010 (I.-U.S.); +82-32-280-5242 (Y.-A.C.); Fax: +82-32-280-5244 (I.-U.S. & Y.-A.C.)
| | - Jong-Sik Park
- Department of Neurology, Incheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 21431, Korea; (J.-S.P.); (S.-H.N.)
| | - Seung-Hee Na
- Department of Neurology, Incheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 21431, Korea; (J.-S.P.); (S.-H.N.)
| | - Jooyeon Jamie Im
- Department of Nuclear Medicine, Incheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 21431, Korea; (H.J.); (J.J.I.)
| | - Marom Bikson
- Department of Biomedical Engineering, The City College of New York, New York, NY 10031, USA;
| | - Wonhye Lee
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (W.L.); (S.-S.Y.)
| | - Seung-Schik Yoo
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (W.L.); (S.-S.Y.)
| |
Collapse
|
34
|
Zhang T, Hou Q, Bai T, Ji G, Lv H, Xie W, Jin S, Yang J, Qiu B, Tian Y, Wang K. Functional and structural alterations in the pain-related circuit in major depressive disorder induced by electroconvulsive therapy. J Neurosci Res 2022; 100:477-489. [PMID: 34825381 DOI: 10.1002/jnr.24979] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/08/2021] [Accepted: 09/25/2021] [Indexed: 12/12/2022]
Abstract
Approximately two-thirds of major depressive disorder (MDD) patients have pain, which exacerbates the severity of depression. Electroconvulsive therapy (ECT) is an efficacious treatment that can alleviate depressive symptoms; however, treatment for pain and the underlying neural substrate is elusive. We enrolled 34 patients with MDD and 33 matched healthy controls to complete clinical assessments and neuroimaging scans. MDD patients underwent second assessments and scans after ECT. We defined a pain-related network with a published meta-analysis and calculated topological patterns to reveal topologic alterations induced by ECT. Using the amplitude of low-frequency fluctuations (ALFFs), we probed local function aberrations of pain-related circuits in MDD patients. Subsequently, we applied gray matter volume (GMV) to reveal structural alterations of ECT relieving pain. The relationships between functional and structural aberrations and pain were determined. ECT significantly alleviated pain. The neural mechanism based on pain-related circuits indicated that ECT weakened the circuit function (ALFF: left amygdala and right supplementary motor area), while augmenting the structure (GMV: bilateral amygdala/insula/hippocampus and anterior cingulate cortex). The topologic patterns became less efficient after ECT. Correlation analysis between the change in pain and GMV had negative results in bilateral amygdala/insula/hippocampus. Similarity, there was a positive correlation between a change in ALFF in the left amygdala and improved clinical symptoms. ECT improved pain by decreasing brain local function and global network patterns, while increasing structure in pain-related circuits. Functional and structural alterations were associated with improvement in pain.
Collapse
Affiliation(s)
- Ting Zhang
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, China
| | - Qiangqiang Hou
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, China
| | - Tongjian Bai
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, China
| | - Gongjun Ji
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, China
| | - Huaming Lv
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, China
| | - Wen Xie
- Anhui Mental Health Center, Hefei, China
| | | | - Jinying Yang
- Center for Biomedical Engineering, University of Science and Technology of China, Hefei, China
| | - Bensheng Qiu
- Center for Biomedical Engineering, University of Science and Technology of China, Hefei, China
| | - Yanghua Tian
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, China
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China
| | - Kai Wang
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, China
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China
- School of Mental Health and Psychological Sciences, Anhui Medical University, Hefei, China
| |
Collapse
|
35
|
Huneke NTM, Aslan IH, Fagan H, Phillips N, Tanna R, Cortese S, Garner M, Baldwin DS. Functional Neuroimaging Correlates of Placebo Response in Patients With Depressive or Anxiety Disorders: A Systematic Review. Int J Neuropsychopharmacol 2022; 25:433-447. [PMID: 35078210 PMCID: PMC9211006 DOI: 10.1093/ijnp/pyac009] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/23/2021] [Accepted: 01/24/2022] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND The mechanisms underlying placebo effects of psychotropic drugs remain poorly understood. We carried out the first, to our knowledge, systematic review of functional neuroimaging correlates of placebo response in adults with anxiety/depressive disorders. METHODS We systematically searched a large set of databases up to February 2021 based on a pre-registered protocol (PROSPERO CRD42019156911). We extracted neuroimaging data related to clinical improvement following placebo or related to placebo mechanisms. We did not perform a meta-analysis due to the small number of included studies and significant heterogeneity in study design and outcome measures. RESULTS We found 12 relevant studies for depressive disorders and 4 for anxiety disorders. Activity in the ventral striatum, rostral anterior cingulate cortex and other default mode network regions, orbitofrontal cortex, and dorsolateral prefrontal cortex correlated with placebo antidepressant responses. Activity in regions of the default mode network, including posterior cingulate cortex, was associated with placebo anxiolysis. There was also evidence for possible involvement of the endogenous opioid, dopamine, and serotonin systems in placebo antidepressant and anxiolytic effects. CONCLUSIONS Several brain regions and molecular systems may be involved in these placebo effects. Further adequately powered studies exploring causality and controlling for confounders are required.
Collapse
Affiliation(s)
- Nathan T M Huneke
- Correspondence: Nathan T. M. Huneke, University Department of Psychiatry, Academic Centre, College Keep, 4-12 Terminus Terrace, Southampton, SO14 3DT, UK ()
| | - Ibrahim H Aslan
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK,University Department of Psychiatry, Academic Centre, Southampton, UK
| | - Harry Fagan
- Southern Health National Health Service Foundation Trust, Southampton, UK,University Department of Psychiatry, Academic Centre, Southampton, UK
| | | | - Rhea Tanna
- Southern Health National Health Service Foundation Trust, Southampton, UK
| | - Samuele Cortese
- Solent National Health Service Trust, Southampton, UK,Center for Innovation in Mental Health, School of Psychology, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, UK,Hassenfeld Children’s Hospital at NYU Langone, New York University Child Study Center, New York City, New York, USA,Division of Psychiatry and Applied Psychology, School of Medicine, University of Nottingham, Nottingham, UK
| | - Matthew Garner
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK,School of Psychology, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, UK
| | - David S Baldwin
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK,Southern Health National Health Service Foundation Trust, Southampton, UK,University Department of Psychiatry, Academic Centre, Southampton, UK,University Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
36
|
Cherkasova MV, Fu JF, Jarrett M, Johnson P, Abel S, Tam R, Rauscher A, Sossi V, Kolind S, Li DKB, Sadovnick AD, Machan L, Girard JM, Emond F, Vosoughi R, Traboulsee A, Stoessl AJ. Cortical morphology predicts placebo response in multiple sclerosis. Sci Rep 2022; 12:732. [PMID: 35031632 PMCID: PMC8760243 DOI: 10.1038/s41598-021-04462-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 12/22/2021] [Indexed: 11/27/2022] Open
Abstract
Despite significant insights into the neural mechanisms of acute placebo responses, less is known about longer-term placebo responses, such as those seen in clinical trials, or their interactions with brain disease. We examined brain correlates of placebo responses in a randomized trial of a then controversial and now disproved endovascular treatment for multiple sclerosis. Patients received either balloon or sham extracranial venoplasty and were followed for 48 weeks. Venoplasty had no therapeutic effect, but a subset of both venoplasty- and sham-treated patients reported a transient improvement in health-related quality of life, suggesting a placebo response. Placebo responders did not differ from non-responders in total MRI T2 lesion load, count or location, nor were there differences in normalized brain volume, regional grey or white matter volume or cortical thickness (CT). However, responders had higher lesion activity. Graph theoretical analysis of CT covariance showed that non-responders had a more small-world-like CT architecture. In non-responders, lesion load was inversely associated with CT in somatosensory, motor and association areas, precuneus, and insula, primarily in the right hemisphere. In responders, lesion load was unrelated to CT. The neuropathological process in MS may produce in some a cortical configuration less capable of generating sustained placebo responses.
Collapse
Affiliation(s)
- Mariya V Cherkasova
- Department of Psychology, University of British Columbia, Vancouver, Canada. .,Department of Psychology, West Virginia University, 2128 Life Science Building, Morgantown, WV, 26506, USA.
| | - Jessie F Fu
- Department of Physics and Astronomy, University of British Columbia, Vancouver, BC, Canada
| | - Michael Jarrett
- Population Data BC, University of British Columbia, Vancouver, BC, Canada
| | - Poljanka Johnson
- Department of Medicine (Division of Neurology), Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Shawna Abel
- Department of Medicine (Division of Neurology), Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Roger Tam
- Department of Radiology, University of British Columbia, Vancouver, BC, Canada.,School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Alexander Rauscher
- Depatment of Pediatrics (Division of Neurology), University of British Columbia, Vancouver, BC, Canada
| | - Vesna Sossi
- Department of Physics and Astronomy, University of British Columbia, Vancouver, BC, Canada
| | - Shannon Kolind
- Department of Medicine (Division of Neurology), Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - David K B Li
- Department of Medicine (Division of Neurology), Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.,Department of Radiology, University of British Columbia, Vancouver, BC, Canada
| | - A Dessa Sadovnick
- Department of Medicine (Division of Neurology), Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Lindsay Machan
- Department of Radiology, University of British Columbia, Vancouver, BC, Canada
| | - J Marc Girard
- Centre Hospitalier de L'Université de Montréal, Montréal, QC, Canada
| | - Francois Emond
- CHU de Québec-Université Laval, Hôpital de L'Enfant-Jésus, Québec, Canada
| | - Reza Vosoughi
- Department of Internal Medicine (Neurology), University of Manitoba, Winnipeg, Canada
| | - Anthony Traboulsee
- Department of Medicine (Division of Neurology), Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - A Jon Stoessl
- Department of Medicine (Division of Neurology), Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
37
|
Frisaldi E, Shaibani A, Trucco M, Milano E, Benedetti F. What is the role of placebo in neurotherapeutics? Expert Rev Neurother 2021; 22:15-25. [PMID: 34845956 DOI: 10.1080/14737175.2022.2012156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION The widespread use of the word 'placebo' in the medical literature emphasizes the importance of this phenomenon in modern biomedical sciences. Neuroscientific research over the past thirty years shows that placebo effects are genuine psychobiological events attributable to the overall therapeutic context, and can be robust in both laboratory and clinical settings. AREAS COVERED Here the authors describe the biological mechanisms and the clinical implications of placebo effects with particular emphasis on neurology and psychiatry, for example in pain, movement disorders, depression. In these conditions, a number of endogenous systems have been identified, such as endogenous opioids, endocannabinoids, and dopamine, which contribute to the placebo-induced benefit. EXPERT OPINION Every effort should be made to maximize the placebo effect and reduce its evil twin, the nocebo effect, in medical practice. This does not require the administration of a placebo, but rather the enhancement of the effects of pharmacological and nonpharmacological treatments through a good doctor-patient interaction.
Collapse
Affiliation(s)
- Elisa Frisaldi
- Neuroscience Department, University of Turin Medical School, Turin, Italy
| | - Aziz Shaibani
- Nerve & Muscle Center of Texas, Houston, Texas, USA.,Baylor College of Medicine, Houston, Texas, USA
| | - Marco Trucco
- Division of Physical and Rehabilitation Medicine, San Camillo Medical Center, Turin, Italy
| | - Edoardo Milano
- Division of Physical and Rehabilitation Medicine, San Camillo Medical Center, Turin, Italy
| | - Fabrizio Benedetti
- Neuroscience Department, University of Turin Medical School, Turin, Italy.,Medicine and Physiology of Hypoxia, Plateau Rosà, Switzerland
| |
Collapse
|
38
|
Differential power of placebo across major psychiatric disorders: a preliminary meta-analysis and machine learning study. Sci Rep 2021; 11:21301. [PMID: 34716400 PMCID: PMC8556377 DOI: 10.1038/s41598-021-99534-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 09/09/2021] [Indexed: 11/09/2022] Open
Abstract
The placebo effect across psychiatric disorders is still not well understood. In the present study, we conducted meta-analyses including meta-regression, and machine learning analyses to investigate whether the power of placebo effect depends on the types of psychiatric disorders. We included 108 clinical trials (32,035 participants) investigating pharmacological intervention effects on major depressive disorder (MDD), bipolar disorder (BD) and schizophrenia (SCZ). We developed measures based on clinical rating scales and Clinical Global Impression scores to compare placebo effects across these disorders. We performed meta-analysis including meta-regression using sample-size weighted bootstrapping techniques, and machine learning analysis to identify the disorder type included in a trial based on the placebo response. Consistently through multiple measures and analyses, we found differential placebo effects across the three disorders, and found lower placebo effect in SCZ compared to mood disorders. The differential placebo effects could also distinguish the condition involved in each trial between SCZ and mood disorders with machine learning. Our study indicates differential placebo effect across MDD, BD, and SCZ, which is important for future neurobiological studies of placebo effects across psychiatric disorders and may lead to potential therapeutic applications of placebo on disorders more responsive to placebo compared to other conditions.
Collapse
|
39
|
Peciña M, Chen J, Lyew T, Karp JF, Dombrovski AY. μ Opioid Antagonist Naltrexone Partially Abolishes the Antidepressant Placebo Effect and Reduces Orbitofrontal Cortex Encoding of Reinforcement. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2021; 6:1002-1012. [PMID: 33684624 PMCID: PMC8419202 DOI: 10.1016/j.bpsc.2021.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 02/24/2021] [Accepted: 02/24/2021] [Indexed: 10/22/2022]
Abstract
BACKGROUND Like placebo analgesia, the antidepressant placebo effect appears to involve cortical and subcortical endogenous opioid signaling, yet the mechanism through which opioid release affects mood remains unclear. The orbitofrontal cortex (OFC)-which integrates various attributes of a stimulus to predict associated outcomes-has been implicated in placebo effects and is rich in μ opioid receptors. We hypothesized that naltrexone blockade of μ opioid receptors would blunt OFC-dependent antidepressant placebo effects. METHODS Twenty psychotropic-free patients with major depressive disorder completed a randomized, double-blind, placebo-controlled crossover study of 1 oral dose of 50 mg of naltrexone or matching placebo immediately before completing 2 sessions of the antidepressant placebo functional magnetic resonance imaging task. This task manipulates placebo-associated expectancies and their reinforcement while assessing expected and actual mood improvement. RESULTS Behaviorally, manipulations of antidepressant placebo expectancies and their reinforcement had positive, interactive effects on participants' expectancy and mood ratings. The high-expectancy condition recruited the dorsolateral and ventrolateral prefrontal cortex, as well as dorsal attention stream regions. Interestingly, increased dorsolateral and ventrolateral prefrontal cortex brain responses appeared to attenuate the antidepressant placebo effect. The administration of 1 oral dose of naltrexone, compared with placebo, partially abolished the interaction of the expectancy and reinforcement manipulation on mood and blocked reinforcement-induced responses in the right central OFC. CONCLUSIONS Our results show preliminary evidence for the role of μ opioid central OFC modulation in antidepressant placebo effects by positively biasing the value of placebo based on reinforcement and enhancing subsequent hedonic experiences.
Collapse
Affiliation(s)
- Marta Peciña
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania.
| | - Jiazhou Chen
- National Institutes of Health, Bethesda, Maryland; The Faculty of Brain Sciences, Division of Psychology and Language Sciences, University College London, London, United Kingdom
| | - Thandi Lyew
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jordan F Karp
- Department of Psychiatry, University of Arizona, Tucson, Arizona
| | | |
Collapse
|
40
|
Schiena G, Franco G, Boscutti A, Delvecchio G, Maggioni E, Brambilla P. Connectivity changes in major depressive disorder after rTMS: a review of functional and structural connectivity data. Epidemiol Psychiatr Sci 2021; 30:e59. [PMCID: PMC8444152 DOI: 10.1017/s2045796021000482] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Aims In the search for effective therapeutic strategies for depression, repetitive transcranial magnetic stimulation (rTMS) emerged as a non-invasive, promising treatment. This is because the antidepressant effect of rTMS might be related to neuronal plasticity mechanisms possibly reverting connectivity alterations often observed in depression. Therefore, in this review, we aimed at providing an overview of the findings reported by studies investigating functional and structural connectivity changes after rTMS in depression. Methods A bibliographic search was conducted on PubMed, including studies that used unilateral, excitatory (⩾10 Hz) rTMS treatment targeted on the left dorsolateral prefrontal cortex (DLPFC) in unipolar depressed patients. Results The majority of the results showed significant TMS-induced changes in functional connectivity (FC) between areas important for emotion regulation, including the DLPFC and the subgenual anterior cingulate cortex, and among regions that are part of the major resting-state networks, such as the Default Mode Network, the Salience Networks and the Central Executive Network. Finally, in diffusion tensor imaging studies, it has been reported that rTMS appeared to increase fractional anisotropy in the frontal lobe. Limitations The small sample size, the heterogeneity of the rTMS stimulation parameters, the concomitant use of psychotropic drugs might have limited the generalisability of the results. Conclusions Overall, rTMS treatment induces structural and FC changes in brain regions and networks implicated in the pathogenesis of unipolar depression. However, whether these changes underlie the antidepressant effect of rTMS still needs to be clarified.
Collapse
Affiliation(s)
- G. Schiena
- Department of Neurosciences and Mental Health, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - G. Franco
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - A. Boscutti
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - G. Delvecchio
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
- Author for correspondence: G. Delvecchio, E-mail:
| | - E. Maggioni
- Department of Neurosciences and Mental Health, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - P. Brambilla
- Department of Neurosciences and Mental Health, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| |
Collapse
|
41
|
Kraemer S. Rebuilding trust after misdiagnosis: where is the liaison between mental and medical? BMJ 2021; 374:n1671. [PMID: 34210670 DOI: 10.1136/bmj.n1671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
42
|
Sretavan Wong K, Migó M, Dougherty DD, Ghaemi SN. Neural correlates of citalopram and placebo response in acute bipolar depression: A randomized trial. J Psychiatr Res 2021; 138:463-466. [PMID: 33965734 PMCID: PMC8192448 DOI: 10.1016/j.jpsychires.2021.04.035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 04/14/2021] [Accepted: 04/25/2021] [Indexed: 10/21/2022]
Abstract
While serotonin reuptake inhibitors are sometimes used in clinical practice to treat acute bipolar depression, the neurophysiological substrates underlying their efficacy are little studied. In the context of a larger clinical efficacy trial, the present study explored neural mechanisms associated with citalopram versus placebo treatment for bipolar depression. FDG-PET imaging examined whole-brain metabolic changes before and after treatment. Clinical efficacy was similar for citalopram versus placebo. Neuroimaging results demonstrated greater glucose metabolism in the left orbitofrontal cortex (OFC) before treatment (combined citalopram and placebo subjects) relative to after treatment, but did not correlate with clinical recovery. Glucose metabolism in the left OFC was also a predictor of depression severity when baseline scans were regressed with baseline MADRS scores. Despite of our small sample size and possibly underpowered whole-brain analysis approach, these preliminary results suggest the OFC, a key region involved in reward circuity, may be a neural substrate for depressive symptom improvement in bipolar depression, regardless of whether due to active treatment or placebo.
Collapse
Affiliation(s)
- Karianne Sretavan Wong
- Division of Neurotherapeutics, Department of Psychiatry, Massachusetts General Hospital / Harvard Medical School, Boston, MA, USA.
| | - Marta Migó
- Division of Neurotherapeutics, Department of Psychiatry, Massachusetts General Hospital / Harvard Medical School, Boston, Massachusetts
| | - Darin D. Dougherty
- Division of Neurotherapeutics, Department of Psychiatry, Massachusetts General Hospital / Harvard Medical School, Boston, Massachusetts
| | - S. Nassir Ghaemi
- Department of Psychiatry, Tufts University School of Medicine, Boston, Massachusetts,Department of Psychiatry, Cambridge Health Alliance, Harvard Medical School, Cambridge, Massachusetts
| |
Collapse
|
43
|
De Pascalis V, Scacchia P, Vecchio A. Influences of hypnotic suggestibility, contextual factors, and EEG alpha on placebo analgesia. AMERICAN JOURNAL OF CLINICAL HYPNOSIS 2021; 63:302-328. [PMID: 33999775 DOI: 10.1080/00029157.2020.1863182] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
We tested the role of hypnotic suggestibility, involuntariness, pain expectation, and subjective hypnotic depth in the prediction of placebo analgesia (PA) responsiveness. We also tested the link of lower and upper alpha sub-band (i.e., 'alpha1' and 'alpha2') power changes with tonic PA responding during waking and hypnosis conditions. Following an initial PA manipulation condition, we recorded EEG activity during waking and hypnosis under two treatments: (1) painful stimulation (Pain); (2) painful stimulation after application of a PA cream. Alpha1 and alpha2 power were derived using the individual alpha frequency method. We found that (1) PA in both waking and hypnosis conditions significantly reduced relative pain perception; (2) during waking, all the above mentioned contextual measures were associated with pain reduction, while involuntariness alone was associated with pain reduction within hypnosis. Enhanced alpha2 power at the left-parietal lead was solely associated with pain reduction in waking, but not in hypnosis condition. Using multiple regression and mediation analyses we found that: (i) during waking, the enhancement of relative left-parietal alpha2 power, directly influenced the enhancement in pain reduction, and, indirectly, through the mediating positive effect of involuntariness; (j) during hypnosis, the enhancement of left-temporoparietal alpha2 power, through the mediation of involuntariness, influenced pain reduction. Current findings obtained during waking suggest that enhanced alpha2 power may serve as a direct-objective measure of the subjective reduction of tonic pain in response to PA treatment. Overall, our findings suggest that placebo analgesia during waking and hypnosis involves different processes of top-down regulation.
Collapse
|
44
|
Schienle A, Jurinec N. Combined Cognitive-Behavioral Therapy and Placebo Treatment for Patients with Depression: A Follow-Up Assessment. Psychol Res Behav Manag 2021; 14:233-238. [PMID: 33654440 PMCID: PMC7912085 DOI: 10.2147/prbm.s294940] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 01/19/2021] [Indexed: 11/23/2022] Open
Abstract
Introduction A previous study revealed that patients with depression who received a combination of cognitive-behavioral therapy (CBT) and placebo treatment (CBT+placebo) showed greater symptom reduction than a CBT group without a placebo. Moreover, the CBT+placebo group practiced relaxation training more frequently. We conducted a 3-month follow-up assessment to investigate the temporal stability of the placebo effects. Methods Eighty-two outpatients with a diagnosis of major depressive disorder who had participated in a 4-week CBT course (CBT: n = 40; CBT with daily placebo treatment: n = 42) returned to a 3-month follow-up assessment. The participants of the CBT+placebo group had been debriefed directly after the course. Results Compared to the CBT group, the CBT+placebo group had lower scores on the Beck Depression Inventory-II (BDI-II) at follow-up and more participants were below the clinical cut-off score of the BDI-II. Additionally, the CBT+placebo group continued to practice relaxation more frequently. Discussion This study demonstrates that placebo effects are not short-lived and continue to be present after the debriefing.
Collapse
Affiliation(s)
- Anne Schienle
- Instiute of Psychology, University of Graz, Graz, Austria
| | - Nina Jurinec
- Instiute of Psychology, University of Graz, Graz, Austria.,Community Health Center Gornja Radgona, Gornja Radgona, Slovenia
| |
Collapse
|
45
|
Theodosis-Nobelos P, Filotheidou A, Triantis C. The placebo phenomenon and the underlying mechanisms. Hormones (Athens) 2021; 20:61-71. [PMID: 32940864 DOI: 10.1007/s42000-020-00243-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 09/02/2020] [Indexed: 12/17/2022]
Abstract
The clinical role of the placebo effect is a topic of increasing interest for the scientific community. Focus is shifting from the inert role of placebos in randomized controlled trials (RCTs) to potential effects in clinical applications, since the phenomenon is thought to be inherent in routine clinical practice, affecting therapy success rates. Mediation of the mind-brain-body relationship involves both psychosocial and neurobiological factors, the interaction of which comprises the placebo mechanisms. Psychosocial factors include environmentally induced expectations, reward expectations, and even conditioned responses to certain stimuli. Expectations also depend on previous experience of the patient with a similar procedure and can affect future responses. Moreover, the supportive bedside behavior of the clinician and the positive framing of information provided to the patient have proven to be of great importance, setting the foundations for reconsideration of standardized practices. Neurobiological mechanisms mediate these effects through neurotransmitter and neuromodulator pathways. The best understood mechanisms are those regulating non-opioid- and opioid-mediated analgesic responses that implicate specific brain regions of pain control and activation of endogenous opioids. Other responses concern, among others, hormonal control, motor performance, and antidepressant responses. Although mechanisms underlying placebo responses are not as yet completely elucidated, there is substantial evidence suggesting that placebo effects are indicative of healthy functioning of intact brain structures and occur through actual functional changes, and are not simply subjective symptom reports. These effects can be utilized in treatment optimization while maintaining an ethical and respectful manner toward the patient and the standardized disclosure procedures.
Collapse
Affiliation(s)
| | - A Filotheidou
- Department of Pharmacy, Frederick University, Nicosia, Cyprus
| | - C Triantis
- Department of Pharmacy, Frederick University, Nicosia, Cyprus.
| |
Collapse
|
46
|
Li YY, Ni XK, You YF, Qing YH, Wang PR, Yao JS, Ren KM, Zhang L, Liu ZW, Song TJ, Wang J, Zang YF, Shen YD, Chen W. Common and Specific Alterations of Amygdala Subregions in Major Depressive Disorder With and Without Anxiety: A Combined Structural and Resting-State Functional MRI Study. Front Hum Neurosci 2021; 15:634113. [PMID: 33658914 PMCID: PMC7917186 DOI: 10.3389/fnhum.2021.634113] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 01/22/2021] [Indexed: 12/25/2022] Open
Abstract
Anxious major depressive disorder is a common subtype of major depressive disorder; however, its unique neural mechanism is not well-understood currently. Using multimodal MRI data, this study examined common and specific alterations of amygdala subregions between patients with and without anxiety. No alterations were observed in the gray matter volume or intra-region functional integration in either patient group. Compared with the controls, both patient groups showed decreased functional connectivity between the left superficial amygdala and the left putamen, and between the right superficial amygdala and the bilateral anterior cingulate cortex and medial orbitofrontal cortex, while only patients with anxiety exhibited decreased activity in the bilateral laterobasal and superficial amygdala. Moreover, the decreased activity correlated negatively with the Hamilton depression scale scores in the patients with anxiety. These findings provided insights into the pathophysiologic processes of anxious major depressive disorder and may help to develop new and effective treatment programs.
Collapse
Affiliation(s)
- Yao Yao Li
- Department of Psychiatry, Hangzhou Seventh People's Hospital, Hangzhou, China
| | - Xiao Kang Ni
- Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ya Feng You
- Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yan Hua Qing
- Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Pei Rong Wang
- Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, China
| | - Jia Shu Yao
- Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ke Ming Ren
- Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lei Zhang
- Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhi Wei Liu
- Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tie Jun Song
- Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jinhui Wang
- Guangdong Key Laboratory of Mental Health and Cognitive Science, Center for Studies of Psychological Application, Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, China
| | - Yu-Feng Zang
- Zhejiang Key Laboratory for Research in Assessment of Cognitive Impairments, Hangzhou, China
| | - Yue di Shen
- Department of Diagnostics, School of Medicine, Hangzhou Normal University, Hangzhou, China
| | - Wei Chen
- Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Department of Psychology and Behavioral Sciences, Zhejiang University, Hangzhou, China.,Key Laboratory of Medical Neurobiology of Zhejiang Province, Hangzhou, China
| |
Collapse
|
47
|
Peciña M, Dombrovski AY, Price R, Karim HT. Understanding the Neurocomputational Mechanisms of Antidepressant Placebo Effects. JOURNAL OF PSYCHIATRY AND BRAIN SCIENCE 2021; 6:e210001. [PMID: 33732892 PMCID: PMC7963355 DOI: 10.20900/jpbs.20210001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Over the last two decades, neuroscientists have used antidepressant placebo probes to examine the biological mechanisms implicated in antidepressant placebo effects. However, findings from these studies have not yet elucidated a model-based theory that would explain the mechanism through which antidepressant expectancies evolve to induce persistent mood changes. Emerging evidence suggests that antidepressant placebo effects may be informed by models of reinforcement learning (RL). Such that an individual's expectation of improvement is updated with the arrival of new sensory evidence, by incorporating a reward prediction error (RPE), which signals the mismatch between the expected (expected value) and perceived improvement. Consistent with this framework, neuroimaging studies of antidepressant placebo effects have demonstrated placebo-induced μ-opioid activation and increased blood-oxygen-level dependent (BOLD) responses in regions tracking expected values (e.g., ventromedial prefrontal cortex (vmPFC)) and RPEs (e.g., ventral striatum (VS)). In this study, we will demonstrate the causal contribution of reward learning signals (expected values and RPEs) to antidepressant placebo effects by experimentally manipulating expected values using transcranial magnetic stimulation (TMS) targeting the vmPFC and μ-opioid striatal RPE signal using pharmacological approaches. We hypothesized that antidepressant placebo expectancies are represented in the vmPFC (expected value) and updated by means of μ-opioid-modulated striatal learning signal. In a 3 × 3 factorial double-blind design, we will randomize 120 antidepressant-free individuals with depressive symptoms to one of three between-subject opioid conditions: the μ-opioid agonist buprenorphine, the μ-opioid antagonist naltrexone, or an inert pill. Within each arm, individuals will be assigned to receive three within-subject counterbalanced forms of TMS targeting the vmPFC-intermittent Theta Burst Stimulation (TBS) expected to potentiate the vmPFC, continuous TBS expected to de-potentiate the vmPFC, or sham TBS. These experimental manipulations will be used to modulate trial-by-trial reward learning signals and related brain activity during the Antidepressant Placebo functional MRI (fMRI) Task to address the following aims: (1) investigate the relationship between reward learning signals within the vmPFC-VS circuit and antidepressant placebo effects; (2) examine the causal contribution of vmPFC expected value computations to antidepressant placebo effects; and (3) investigate the causal contribution of μ-opioid-modulated striatal RPEs to antidepressant placebo effects. The proposed study will be the first to investigate the causal contribution of μ-opioid-modulated vmPFC-VS learning signals to antidepressant placebo responses, paving the way for developing novel treatments modulating learning processes and objective means of quantifying and potentially reducing placebo effects during drug development. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT04276259.
Collapse
Affiliation(s)
- Marta Peciña
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | | | - Rebecca Price
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Helmet T. Karim
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| |
Collapse
|
48
|
Hippocampal cerebral blood flow increased following low-pressure hyperbaric oxygenation in firefighters with mild traumatic brain injury and emotional distress. Neurol Sci 2021; 42:4131-4138. [PMID: 33532950 DOI: 10.1007/s10072-021-05094-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 01/27/2021] [Indexed: 01/19/2023]
Abstract
BACKGROUND Recent evidence suggests that hyperbaric oxygenation (HBO), which has been used as an effective treatment for certain types of tissue injury, may change neural activities in the human brain and subsequently improve symptoms of psychiatric disorders. To scrutinize the neural mechanism of HBO in the human brain, we investigated whether 20 sessions of HBO changed regional cerebral blood flow (rCBF) of the limbic system in firefighters with mild traumatic brain injury (mTBI) and subjective emotional distress. METHODS Twenty firefighters with mTBI and mild emotional distress were treated with HBO at a relatively low pressure of 1.3 atmospheres absolute for 45 min a day for 20 consecutive days (the mild emotional distress group). The rCBF of the limbic system was measured using an arterial spin labeling perfusion magnetic resonance imaging before and after the HBO. Analyses were performed on the data from fourteen individuals who completed the study and 14 age- and sex-matched healthy firefighters (the comparison group). RESULTS Firefighters in the mild emotional distress group showed increase rCBF following HBO in a cluster encompassing the right hippocampal and parahippocampal regions (peak t = 4.31; cluster size = 248 mm3)(post-hoc analysis, z = 5.92, p < 0.001) that had lower rCBF relative to the comparison group at baseline (post-hoc analysis, t = -2.20, p = 0.04). CONCLUSION The current study demonstrated that low-pressure HBO might increase rCBF of the hippocampal and parahippocampal regions, suggesting a potential underpinning mechanism of HBO in the human brain.
Collapse
|
49
|
Lu L, Li H, Mills JA, Schroeder H, Mossman SA, Varney ST, Cecil KM, Huang X, Gong Q, Levine A, DelBello MP, Sweeny JA, Strawn JR. Greater Dynamic and Lower Static Functional Brain Connectivity Prospectively Predict Placebo Response in Pediatric Generalized Anxiety Disorder. J Child Adolesc Psychopharmacol 2020; 30:606-616. [PMID: 32721213 PMCID: PMC7864114 DOI: 10.1089/cap.2020.0024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Objectives: Placebo response is one of the most significant barriers to detecting treatment effects in pediatric (and adult) clinical trials focusing on affective and anxiety disorders. We sought to identify neurofunctional predictors of placebo response in adolescents with generalized anxiety disorder (GAD) by examining dynamic and static functional brain connectivity. Methods: Before randomization to blinded placebo, adolescents, aged 12-17 years, with GAD (N = 25) underwent resting state functional magnetic resonance imaging. Whole brain voxelwise correlation analyses were used to determine the relationship between change in anxiety symptoms from baseline to week 8 and seed-based dynamic and static functional connectivity maps of regions in the salience and ventral attention networks (amygdala, dorsal anterior cingulate cortex [dACC], and ventrolateral prefrontal cortex [VLPFC]). Results: Greater dynamic functional connectivity variability in amygdala, dACC, VLPFC, and regions within salience, default mode, and frontoparietal networks was associated with greater placebo response. Lower static functional connectivity between amygdala and dorsolateral prefrontal cortex, amygdala and medial prefrontal cortex, dACC and posterior cingulate cortex and greater static functional connectivity between VLPFC and inferior parietal lobule were associated with greater placebo response. Conclusion: Placebo response is associated with a distinct dynamic and static connectivity fingerprint characterized by "variable" dynamic but "weak" static connectivity in the salience, default mode, frontoparietal, and ventral attention networks. These data provide granular evidence of how circuit-based biotypes mechanistically relate to placebo response. Finding biosignatures that predict placebo response is critically important in clinical psychopharmacology and to improve our ability to detect medication-placebo differences in clinical trials.
Collapse
Affiliation(s)
- Lu Lu
- Department of Radiology, Huaxi MR Research Center (HMRRC), West China Hospital of Sichuan University, Chengdu, China.,Department of Psychiatry, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Hailong Li
- Department of Radiology, Huaxi MR Research Center (HMRRC), West China Hospital of Sichuan University, Chengdu, China
| | - Jeffrey A. Mills
- Department of Economics, Lindner College of Business, University of Cincinnati, Cincinnati, Ohio, USA
| | - Heidi Schroeder
- Department of Psychiatry, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Sarah A. Mossman
- Department of Psychiatry, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Sara T. Varney
- Department of Psychiatry, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Kim M. Cecil
- Department of Radiology, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA,Imaging Research Center, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Xiaoqi Huang
- Department of Radiology, Huaxi MR Research Center (HMRRC), West China Hospital of Sichuan University, Chengdu, China.,Psychoradiology Research Unit of Chinese Academy of Medical Sciences, West China Hospital of Sichuan University, Chengdu, China
| | - Qiyong Gong
- Department of Radiology, Huaxi MR Research Center (HMRRC), West China Hospital of Sichuan University, Chengdu, China.,Psychoradiology Research Unit of Chinese Academy of Medical Sciences, West China Hospital of Sichuan University, Chengdu, China.,Address correspondence to: Qiyong Gong, MD, PhD, Department of Radiology, Huaxi MR Research Center (HMRRC), West China Hospital of Sichuan University, No. 37 Guo Xue Xiang, Chengdu, Sichuan 610041, China
| | - Amir Levine
- Department of Psychiatry, Columbia University and New York State Psychiatric Institute, New York City, New York, USA
| | - Melissa P. DelBello
- Department of Psychiatry, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - John A. Sweeny
- Department of Radiology, Huaxi MR Research Center (HMRRC), West China Hospital of Sichuan University, Chengdu, China.,Department of Psychiatry, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Jeffrey R. Strawn
- Department of Psychiatry, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
50
|
McCarter G. Harnessing Placebo Responses to Improve Health Outcomes. AMERICAN JOURNAL OF PHARMACEUTICAL EDUCATION 2020; 84:8184. [PMID: 34283778 PMCID: PMC7779879 DOI: 10.5688/ajpe8184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 08/12/2020] [Indexed: 06/13/2023]
Abstract
Variations in the psychosocial aspects of the provision of health care treatments can measurably affect the health outcomes resulting from the use of such treatments. These benefits (or harms) in outcomes result from processes beyond the specific physiological mechanisms induced by the treatments. Such phenomena can be most clearly seen when physiological improvements are induced by administering inert placebo medications in the same manner as if they were actual medications. By logic, these physiological improvements should also occur during the provision of actual medications and potentiate the latter's effectiveness. There are likely many manipulations of the patient-clinician interaction that can positively or negatively affect therapeutic outcomes for many conditions. Clinicians should thus be able to make choices in their behavior that optimize any possible increases in drug effectiveness resulting from placebo responses. This commentary makes the assertion that pharmacists are ethically obligated to learn and practice techniques that maximize placebo responses and that it is incumbent upon the Academy to explore and understand such techniques and effectively teach them to students.
Collapse
Affiliation(s)
- Gordon McCarter
- Touro University California College of Pharmacy, Vallejo, California
| |
Collapse
|