Jurnal ECT

Download as pdf or txt
Download as pdf or txt
You are on page 1of 25

HHS Public Access

Author manuscript
Harv Rev Psychiatry. Author manuscript; available in PMC 2024 January 01.
Author Manuscript

Published in final edited form as:


Harv Rev Psychiatry. 2023 ; 31(3): 101–113. doi:10.1097/HRP.0000000000000365.

Electroconvulsive Therapy: Mechanisms of Action, Clinical


Considerations, and Future Directions
Michael D. Kritzer, MD, PhD1, Angel V. Peterchev, PhD2,3,4,5, Joan A. Camprodon, MD, PhD,
MPH1
1Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School,
Charlestown, MA, 02129, USA
Author Manuscript

2Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA
3Department of Biomedical Engineering, Duke University, Durham, NC, USA
4Department of Electrical and Computer Engineering, Duke University, Durham, NC, USA
5Department of Neurosurgery, Duke University, Durham, NC, USA

Abstract
Electroconvulsive therapy (ECT) is the most effective treatment for a variety of psychiatric
illnesses, including treatment-resistant depression, bipolar depression, mania, catatonia, and
clozapine-resistant schizophrenia. ECT is a medical and psychiatric procedure whereby electrical
current is delivered to the brain under general anesthesia to induce a generalized seizure. ECT
Author Manuscript

has evolved a great deal since the 1930’s. Though it has been optimized for safety and to
reduce adverse effects on cognition, issues persist. There is a need to understand fundamental
physiologic, cellular, and molecular mechanisms of ECT to devise strategies to optimize
therapeutic outcomes. Clinical trials that set out to adjust parameters, electrode placement,
adjunctive medications and patient selection are critical steps towards the goal of improving
outcomes with ECT. This narrative review provides an overview of ECT, its efficacy in treating
depression, the known effects on cognition, evidence of mechanisms, and future directions.

Keywords
Electroconvulsive Therapy; ECT; Depression; Treatment-Resistant Depression; Electroconvulsive
Stimulation
Author Manuscript

Introduction:
Electroconvulsive therapy (ECT) is a medical procedure whereby electrical current is
administered through the skull under general anesthesia to induce a seizure for the treatment
of a variety of psychiatric illnesses 1–4. For over 80 years, ECT has been the most effective
treatment for severe depression, mania, catatonia and clozapine-resistant schizophrenia.
It has also been shown to help with suicidal ideation and in augmenting clozapine for

Authors have no additional potential conflicts of interest to disclose.


Kritzer et al. Page 2

psychosis 5–9. There is an abundance of evidence supporting the use of ECT for treatment-
Author Manuscript

resistant depression (TRD), and overall, ECT is more effective than pharmacotherapy for
the treatment of depression and other mood disorders, such as bipolar depression and
mania 4. Regardless, given the stigma of its use and adverse effects on memory, it is
widely underutilized 2,4,10. A contributing factor to why patients and their families may
be reluctant to undergo ECT, or clinicians may be reluctant to refer for treatment, is the
impression that little is known about the mechanisms of the therapeutic effect and cognitive
side effects 10,11. While many questions remain open, neuroscience research has made
significant advances to understand the neurobiological effects of ECT. This narrative review
aims to describe the evidence supporting current models that explain how ECT works
therapeutically as well as what causes the cognitive side-effects.

ECT – an ultrabrief history:


Author Manuscript

Electroconvulsive therapy has evolved tremendously since it was first used in the 1930’s
12,13. What began as bilateral stimulation with a sine wave voltage source at the power

line frequency without anesthesia (referred to as “unmodified ECT”), has evolved to its
present form of unilateral (typically right unilateral, RUL) stimulation with individually-
titrated trains of ultrabrief or brief rectangular constant-current pulses under general
anesthesia (termed “modified ECT”, i.e. modified with anesthesia), and bilateral brief pulse
stimulation generally reserved for refractory or emergent cases 14–17. The impetus for these
optimizations has been to prevent physical and cognitive side effects. Despite the robust
antidepressant effect of ECT, some patients still experience adverse effects on memory,
which limits its clinical application.

ECT is a rapid-acting antidepressant:


Author Manuscript

Electroconvulsive therapy begins with a short index course of 6 treatments delivered 3/week,
or 2/week as performed in the UK. Patients typically respond to RUL treatment with > 50%
reduction in their symptoms within that two-week period and then continue with weekly
maintenance treatment for a few weeks and gradually increasing the interval between
maintenance treatments as clinically indicated. If there is little or suboptimal response in
2–3 weeks, particularly if there is concern for physical safety, practitioners often switch to
brief-pulse RUL (change in pulse width) or bilateral treatment (change in montage), which
is understood to be associated with more noticeable cognitive side effects, as we explain
in the following sections. A recent epidemiological study of Medicare data compared
10,460 patients who underwent ECT to 31,160 patient-controls and showed that patients
receiving ECT had lower all-cause mortality for up to 1 year following hospital discharge.
Author Manuscript

Importantly, there was a significant reduction in the rate of suicide following ECT, but with
no difference at 1 year follow-up 18.

Antidepressant efficacy and cognition:


Luccarelli, Henry, and colleagues have recently reviewed large datasets from Massachusetts
General and McLean Hospitals and made some pertinent clinical observations. They have
shown that ECT was associated with a rapid decrease in depression symptoms within

Harv Rev Psychiatry. Author manuscript; available in PMC 2024 January 01.
Kritzer et al. Page 3

the first 10 treatments; which continued for almost two years of maintenance treatment.
Author Manuscript

Importantly, there was no change in cognitive functioning as measured by the Montreal


Cognitive Assessment (MoCA). At their institution, regardless of the initial treatment,
bilateral and brief pulse treatment parameters were more commonly used by treatment
number 50, and most participants either stopped due to remitting or had continued in ECT,
implying side-effects were tolerable enough on average at the end of the study period 19.
Another analysis showed that 37.5% of patients who began with RUL were transitioned to
bilateral treatments 20. Specifically, younger patients and those with bipolar disorder were
more likely to transition from brief pulse RUL to bilateral treatments, but the overall number
of treatments did not differ based on age or primary diagnosis 21.

In a study where cognition was assessed, ECT was associated with a 40% decrease in
depression symptoms (QIDS reduction from 17.1 ± 4.9 to 10.1 ± 5.2), accompanied by
a reduction in MoCA scores that was statistically significant, but likely too small to be
Author Manuscript

clinically significant (from 25.8 ± 3.1 to 25.4 ± 3.1); interestingly, older age was associated
with a milder reduction in MoCA perhaps due to lower baseline at the initiation of treatment
22. Furthermore, in a study of 684 patients that received at least 10 ECT treatments,

investigators tracked QIDS and assessed cognition with the MoCA at baseline and after
treatment. Patients with baseline normal cognition (MoCA ≥ 26; N = 371) had a decrease in
MoCA of 1.44±0.26 points over the course of treatment, while those with baseline impaired
global cognitive function (MoCA < 26; N = 313) experienced an increase in MoCA of
1.72±0.25 points. Importantly, baseline global cognitive function was not associated with a
change in depression after ECT, suggesting that impaired global cognitive function may
not be a contraindication to ECT in geriatric patients 23. It is important to note that
the MoCA may not be the most appropriate assessment tool for the type of cognitive
complaints patients experience with ECT (nuances discussed below) as it is a general
Author Manuscript

multidomain screening test that measures many different cognitive functions and emphasizes
executive function over memory. That said, it is reassuring that there is very little change
in an assessment tool that quickly screens a broad range of cognitive domains, which is
particularly useful in older patients.

Electrical stimulus parameters:


The characteristics of the electrical stimulus influence the therapeutic response as well
as the emergence of side effects. A crucial component to the administration of ECT is
electrode placement, which determines the path of current propagation through the tissue
24,25. There are three commonly used electrode placements: RUL, bitemporal, and bifrontal;

the latter two are types of bilateral placement. Historically, as mentioned above, ECT was
Author Manuscript

administered bilaterally with electrodes placed on the side of the head over the temporal
lobes (bitemporal placement). Because of adverse effects of having current pass directly
through the left temporal lobe where language is generated for most people, practitioners
typically favor beginning treatment with a RUL configuration (right temporal and midline
vertex placement of electrodes). If a patient is left-handed, they may have a right-dominant
hemisphere (as approximately 25% of left-handed individuals do), and steps can be taken
to determine whether the electrodes should be placed left-unilaterally to avoid passing

Harv Rev Psychiatry. Author manuscript; available in PMC 2024 January 01.
Kritzer et al. Page 4

current through their dominant hemisphere in order to minimize cognitive side effects 26,27.
Author Manuscript

Electrodes can also be placed bifrontally across the forehead.

The electrical current parameters also influence the rate of depressive symptom
improvement as well as the emergence of side effects. Conventional dosing of ECT depends
on many factors, but pulse shape and current amplitude are generally fixed; this leaves
changing the pulse width and the pulse train frequency and duration as means of protocol
adjustment. The standard current amplitude used in clinical practice of modern ECT is fixed
at either 800 mA or 900 mA, depending on the device and various practices; the pulse shape
is rectangular with brief (0.5–1 ms) or ultrabrief (0.25–0.3 ms) pulse width; and the pulse
train is comprised of monophasic pulses of alternating polarity. With these parameters fixed,
the electrical dosage is conventionally individualized by increasing the number of pulses in
the stimulus train via titration of the train duration and/or frequency. In ECT practice, the
electrical dosage is often quantified by the total charge or energy of the stimulus train 28.
Author Manuscript

Historically, researchers found that increasing electrical dosage from just above seizure
threshold (ST) to 2.5 times the ST increased the efficacy of brief pulse RUL ECT, although
not to the level of bilateral ECT 29. Sackeim et al. showed that high-dose RUL ECT (6 × ST)
is more effective than lower (1.5 × ST) or moderate (2.5 × ST) RUL ECT 30. Importantly,
high-dosage RUL ECT showed similar therapeutic effects to bilateral ECT (bilateral at 2.5 ×
ST) with less cognitive impairment. Sackeim and colleagues then compared high-dose RUL
(6 × ST) to moderate suprathreshold bilateral ECT (2.5 × ST) for both brief and ultrabrief
pulses, and concluded that high-dose ultrabrief RUL ECT preserved efficacy while reducing
cognitive side effects, whereas ultrabrief bilateral ECT reduced cognitive side effects but
was significantly less effective 15. The authors speculated that at higher dosage relative
to ST, ultrabrief bilateral ECT may also increase its efficacy, although this approach has
Author Manuscript

not been tested systematically 15. Taken together, a clinical approach may be to start with
ultrabrief RUL ECT at a high dosage rather than bilateral ECT because of comparable
efficacy and fewer cognitive adverse effects.

Sienaert and colleagues found both unilateral and bifrontal ultrabrief ECT to improve
cognitive performance with a broad neuropsychological battery at 1 and 6 weeks post-
treatment 31,32. Testing stimuli of 4, 7, and 10 times ST, Quante et al. asserted that stimulus
intensity of ultrabrief RUL ECT in this range does not correlate with symptom improvement
measured by the Hamilton Depression Rating Scale, demonstrating that ultrabrief RUL ECT
at 4 times ST is sufficient for symptom improvement 33. A 2015 meta-analysis showed
that brief versus ultrabrief pulse RUL ECT was significantly more efficacious in treating
depression but also showed significantly more side effects in global cognition, anterograde
learning and recall, as well as retrograde memory 34.
Author Manuscript

In clinical practice, initial electrode placement is typically initiated as RUL or, in more
urgent or severe cases, bitemporal or bifrontal 30. This is because, generally, bilateral
stimulation acts faster with a trade-off of having greater likelihood of cognitive side
effects than unilateral ECT (with either ultrabrief or brief pulse width). The UK ECT
Review Group found that bilateral ECT is generally more effective than unilateral ECT
in improving depression symptoms, based on data from 22 studies comparing bilateral

Harv Rev Psychiatry. Author manuscript; available in PMC 2024 January 01.
Kritzer et al. Page 5

(this meta-analysis combined bifrontal and bitemporal) to unilateral ECT 4. However, two
Author Manuscript

rigorous studies found non-inferiority of brief pulse RUL in comparison with brief pulse
bifrontal and bitemporal ECT, respectively 35,36. A randomized control trial by Kellner
et al comparing bilateral, bitemporal and RUL was consistent with this, with bitemporal
ECT having greater cognitive impairment 37. In a meta-analysis of observational studies
of cognitive function after ECT, Semkovska and McLoughlin showed that both unilateral
and bitemporal ECT are associated with significant decreases in cognitive performance,
mainly executive functioning, and verbal memory, within 3 days post-treatment. Bitemporal
ECT had worse cognitive performance acutely, though long-term follow-up demonstrated
improvements compared to baseline or reattainment of baseline performance (at 14–365
days post-treatment). Although there are concerns about acute ill effects on cognition with
ECT, it is generally accepted that adverse effects on cognition (predominantly memory) will
return to normal within six months of the index course of treatment 4,38. Depending on
Author Manuscript

illness severity, a protocol could begin with bilateral ECT and switch to RUL if cognitive
side effects emerge or perhaps the converse—begin with high-dose RUL ECT for index and
weekly treatments and then use bilateral for monthly maintenance treatments. If cognitive
side effects are of particular concern, high-dose ultrabrief-pulse RUL should be considered
over brief-pulse RUL or bilateral ECT.

More recently, a novel approach to electrode placement, focal electrically administered


seizure therapy (FEAST), has been explored as a means of reducing the adverse cognitive
effects from seizure therapy. FEAST utilizes unidirectional stimulation and an asymmetrical
frontal electrode placement with a large posterior electrode (cathode) in front of the right
motor cortex and a small anterior electrode (anode) above the center of the right eyebrow,
over the right orbitofrontal cortex, with the aim of focusing the seizure induction in the right
prefrontal cortex 39–42.
Author Manuscript

Another experimental approach aiming to reduce the side effects of ECT is magnetic
seizure therapy (MST), which utilizes high intensity and frequency transcranial magnetic
stimulation (TMS) under general anesthesia to induce a seizure 43. Compared to
conventional ECT, MST induces a weaker and more superficial electric field 24,44. Like
in ECT, optimal MST stimulation targeting and parameters are being investigated. For
example, prefrontal stimulation at higher frequencies may increase depression remission
rates, whereas lower frequencies appear advantageous for suicidality remission 45–47.

Computational, animal, and clinical studies have suggested that the electric field generated
by standard ECT in the brain is markedly higher than the levels needed to depolarize
neurons and induce a seizure 48,49. The current amplitude is a key factor driving the electric
Author Manuscript

field into deeper brain regions, such as the hippocampus and temporal lobe, which may
contribute to adverse effects on cognition and memory. Reducing the current amplitude
makes the electric field weaker and more focal 50. Therefore, decreasing the stimulus current
amplitude could be a potential means of reducing cognitive side effects 28. A recent proof
of concept non-randomized clinical trial suggested that ultrabrief RUL ECT with a lower
current amplitude (as low as 500 mA to 600 mA) may improve cognitive outcomes by
reducing reorientation time, a measure correlated with worse cognitive outcomes from ECT.
Intriguingly, there was no difference in treatment response in depressive symptoms when

Harv Rev Psychiatry. Author manuscript; available in PMC 2024 January 01.
Kritzer et al. Page 6

compared to higher current ultrabrief pulse RUL 51,52. There may be, however, an individual
Author Manuscript

tradeoff between the efficacy of treating depression and preservation of cognition controlled
by the current amplitude. Abbott and colleagues investigated ultrabrief RUL ECT delivered
at three current amplitudes and found that 700 and 800 mA were more effective at reducing
depression symptoms, while 600 mA had less cognitive side effects as measured by the
Hopkins Verbal Learning Test and aspects of the Delis Kaplan Executive Function System,
which together assess frontal-temporal cognitive functions (e.g., verbal fluency, cognitive
flexibility, inhibition, and processing speed) 53. Notably, current strength of 500 mA was
not included in this trial based on evidence of insufficient therapeutic efficacy during pilot
testing. Additionally, subjects enrolled in the trial were switched from ultrabrief to brief
pulse stimulation, due to concerns about low efficacy of the 600 mA arm. Thus, reducing
the current amplitude may diminish therapeutic effects, at least for some subjects and ECT
parameter settings.
Author Manuscript

The relationship between ECT current amplitude and clinical outcomes may vary
individually due to differences in the electric field delivered to the brain stemming from
anatomical variability. The electric field in the brain can be modeled computationally based
on individual MRI scans 24. The few studies to date that modeled the ECT electric field in
large groups of patients have arrived at intriguing though diverse relationships. The electric
field in the left hippocampus and left amygdala were found to correlate with increases of the
volume of these structures after RUL ECT, but neither the volume changes nor the electric
field strength were associated with antidepressant response 54. The latter finding contradicts
other studies (often using data from a single or two centers, with smaller sample sizes
but greater sample homogeneity), which demonstrated a significant relationship between
increases in hippocampal and amygdala volume primarily (but also other limbic structures)
and symptom improvement 55–57. One line of investigation tried to combine genetic
Author Manuscript

differences in hippocampal size after ECT and found that a single nucleotide polymorphism
(SNP) in the promoter of vascular endothelial growth factor (VEGF) attenuated hippocampal
growth whereas a SNP in the brain derived neurotrophic factor (BDNF) gene did not 58.

The study of current amplitude including RUL and bitemporal electrode placement found
improved antidepressant outcomes to correlate with volume increases of the right, but not
left, hippocampus, which, in turn, was affected by the local electric field. In contrast to this
indirect effect, stronger electric field in the right hippocampus worsened the antidepressant
effect and cognitive performance 59. A similar effect of worse antidepressant effect with
a stronger electric field in the temporal lobes was observed for bilateral but not RUL
ECT in another study 60. Other studies have also observed lateralized effects, in which
RUL ECT leads to right-sided volumetric changes while bilateral ECT leads to bilateral
Author Manuscript

volumetric changes 61. While these limited data do not afford firm conclusions, they suggest
adverse effects of direct electric field exposure in the temporal lobes, while also indicating
potential beneficial indirect effect on hippocampus, perhaps mediated by the seizure and/or
transsynaptic activation.

These results raise the question of the relative contribution to ECT outcomes of the electrical
stimulation versus the seizure itself. The strong electrical stimulation delivered by ECT
produces synchronized rhythmic firing over broad populations of neurons, akin to the brain

Harv Rev Psychiatry. Author manuscript; available in PMC 2024 January 01.
Kritzer et al. Page 7

activity during a seizure. Thus, it is possible that some of the effects of ECT are conferred
Author Manuscript

or modulated by the electrical stimulus rather than the seizure. This may be a reason
why, to be effective, RUL ECT has to be delivered with six times the number of pulses
needed to induce a seizure. Moreover, a pilot study of intensive bifrontal subconvulsive
stimulation with an ECT device reported significant therapeutic effects in the absence of a
seizure 62. Finally, accelerated repetitive transcranial TMS, which delivers a large number of
subconvulsive stimuli over a short period of time, has recently demonstrated remarkably
high remission rates in treatment resistant depression 63,64. These observations may
point to using targeted stimulation to induce seizures while avoiding direct hippocampal
stimulation, combined with more stimulation pulses and sessions for cumulative therapeutic
effects 50. Lower current amplitude produces somewhat less cognitive side effects, but
there is substantial individual variability in efficacy versus current amplitude. Therefore,
individualization of the current amplitude may be appropriate, by seizure or motor threshold
titration in the amplitude domain or individual electric field modeling 25,59,65. Broadly,
Author Manuscript

ECT parameters could be explored further as pathways for improving outcomes. Optimized
and individualized stimulus delivery may become an important option to lower the risk
of cognitive side effects, especially in patients at higher risk or with a previous adverse
outcome.

Cognition Considerations:
Studying cognition in response to ECT has a few pitfalls. The first is that depression,
particularly TRD, has been shown to negatively impact many aspects cognition, mainly
attention and aspects of memory 66. Patients often report adverse effects of ECT on memory
67. These subjective reports have been challenging to characterize 68. This can be explained

partly because studies of cognition are confounded by a few factors: First, multiple cognitive
Author Manuscript

domains improve after ECT as depression negatively affects attention, executive function,
and working memory; Secondly, studies typically compare MoCA, Mini-Mental Status
Exam (MMSE) or the Cognitive Failures Questionnaire (CFQ) before and after ECT 69.
MMSE and MoCA are tools to assess mild to severe cognitive impairment and do not appear
to capture the subtleties of subjective cognitive complaints. These screening tests do not
examine retrograde episodic memory – the type of memory impairment primarily reported
by patients. Certainly, if ECT caused major neurocognitive impairment, we would have
stopped administering it decades ago.

The effects of ECT on memory is a major concern for patients. Many studies have evaluated
cognition broadly and found non-memory cognition to improve substantially. Importantly,
studies at long-term follow-up after ECT generally show improvements in memory using
Author Manuscript

objective measures 70. In the subacute period following ECT (0–3 days), tests of executive
function and processing speed were impaired compared to baseline 70. However, when
measured between 4 and 14 days, most tests showed significantly improved outcomes
compared with baseline, and none were below baseline 70. This improvement is likely to
have been a result of successful treatment as studies suggest that a high percentage of
patients with severe depression have substantial cognitive impairment at baseline 71. Longer
term, with a wide range of follow-up intervals (2 weeks to 2 years), there was no evidence of
impairment in tests of non-memory cognition 70.

Harv Rev Psychiatry. Author manuscript; available in PMC 2024 January 01.
Kritzer et al. Page 8

The effects on memory have been variable and proven more challenging to assess. Studies of
Author Manuscript

subjective report of cognitive side-effects show varying percentages of patients complaining


of ‘memory loss’, depending on many factors, including the interviews were conducted 67.
When patients report cognitive side-effects, it is often loss of retrograde autobiographical
memory concerning them, i.e., the loss of detailed memory for events that the person
has experienced. However, both episodic memory (experiential memory) and semantic
memory (factual memory) may be affected 72. Retrograde amnesia (memory loss) for
non-autobiographical information has also been shown 73. Furthermore, when objectively
measured, this retrograde amnesia could persist for over one year. Objective testing
of autobiographical memory, typically using the Columbia University Autobiographical
Memory Interview (CUAMI), has shown that ECT memory loss exceeds the normal
decay of memories over time in healthy matched participants and the degree of loss
differs depending on the treatment ECT parameters used; though importantly, many of
Author Manuscript

these comparisons were not to that of depressed. Importantly, subjective impairment often
reduces throughout the course of treatment, correlating with clinical response, and perhaps
representing negative bias associated with depressive states 74,75. Brief assessments of
cognitive side effects of ECT were reviewed by Martin et al, and an emerging tool,
the Electroconvulsive Therapy Cognitive Assessment (ECCA) is gaining interest, though
widespread adoption is lacking, as is thorough validation of it for autobiographical memory
loss 76,77.

Interestingly, ECT does not appear to adversely affect verbal memory long-term, even with
bilateral brief pulse, with current passing directly through the left frontotemporal lobe. More
appropriate tests would be those that test non-verbal and visuospatial memory, working
memory, reasoning and executive function 69. It is possible that the brain regions undergoing
the most dramatic changes during a course of ECT, i.e., mesial temporal lobes and limbic
Author Manuscript

structures, are the ones responsible for the efficacy of ECT, but also harbor the source of
some cognitive changes – be it objective or perceived.

Ketamine has been studied as a potential augmentation strategy co-administered with ECT
to alleviate side effects and/or increase efficacy. It is important to note that anesthetic doses
of ketamine used for ECT seem to have a different effects on mood when compared to
subanesthetic doses used to treat depression 78. Importantly, ketamine for the treatment of
depression is used at a subanesthetic dose in awake patients, so findings in anesthetized
patients with higher doses of ketamine may not share the identical mechanism of action.
A recent meta-analysis of add-on ketamine to ECT has shown that ketamine enhances
ECT’s therapeutic effects but unfortunately also exacerbates side effects 79,80. Ketamine is
known to inhibit N-methyl-D-aspartate (NMDA) receptors and, interestingly, endogenous
Author Manuscript

NMDA receptor agonist quinolinic acid (QUIN), is decreased following successful ECT –
suggesting a role of NMDA receptor biology in ECT’s mechanisms. Ill-effects on cognition
resulting from ECT seems to depend on many factors, including a patient’s baseline
cognitive status and underlying cognitive reserve prior to obtaining ECT 81,82. That said,
there are parameters of ECT delivery that seem to directly increase the likelihood of adverse
cognitive outcomes including bilateral electrode placement, current, and stimulus type 83.

Harv Rev Psychiatry. Author manuscript; available in PMC 2024 January 01.
Kritzer et al. Page 9

Additionally, general anesthetics have been shown to have varying effects on cognition and
likely worse outcomes in older patients 84. The cognitive effects of the interaction between
Author Manuscript

ECT and anesthesia are beyond the scope of this review; however, it is important to highlight
the fact that even though generalized anesthesia for ECT is limited to approximately 10
minutes for each session, a patient may undergo dozens of ECT treatments with the potential
for adverse effects due to the accumulation of anesthetic events 85.

Mechanisms of ECT:
γ-Aminobutyric acid and Glutamate:
γ-Aminobutyric acid (GABA) is an inhibitory neurotransmitter expressed in interneurons
distributed across the brain to modulate excitatory neural circuits. It is intriguing to consider
the observation that ECT appears to effectively treat a variety of psychiatric illnesses
that are also effectively treated with GABAergic medications such as benzodiazepines.
Author Manuscript

These include unipolar and bipolar depression, bipolar mania, and catatonia 86–88. Low
GABAergic function may be a feature in a subset of mood disorder patients, representing
a genetic susceptibility, consequence of chronic stress, or more likely a combination of
the two. There is evidence supporting the notion that ECT has significant anticonvulsant
properties and results in decreased neural metabolic activity over the course of treatment
89–91.

Typically, a repeated seizures throughout a course of ECT reduces seizure duration and
an increases in intracortical inhibition, both of which have been correlated with clinical
improvement 91,92. In a very large retrospective study of over 3000 patients receiving
an acute course of ECT there was a decrease in the mean ECT seizure duration, with
the greatest change within the first 3 treatments. Older age, higher ECT dose and higher
Author Manuscript

treatment number were all associated with shorter seizures 93. Interestingly, this notion has
been evaluated with TMS. Cortical excitability in neurocircuitry has been assessed with
single and paired pulse TMS to investigate these anticonvulsant effects of ECT. After 10
sessions of RUL ECT, researchers observed an enhanced activity of inhibitory circuits in
human motor cortex, as measured by both increased intracortical inhibition and cortical
silent period duration, while intracortical facilitation and resting motor threshold did not
change 92.

There is evidence suggesting GABAergic involvement in ECT’s mechanism of


antidepressant action 94. Postictal suppression of seizure activity, largely thought to be
driven by GABAergic neurons, has been correlated with treatment outcomes and there
may be ways to alter seizure suppression with pharmacological augmentation during ECT
Author Manuscript

administration 95–103. Compared to non-responders, ECT responders appear to have higher


GABA levels at baseline and after a course of ECT. Interestingly, immediately after ECT,
free plasma GABA was decreased for up to 1 hour and another study found that occipital
cortex GABA levels doubled 104,105. Two hours after a single session of ECT, GABA and
amino acids plasma levels were decreased in depressed patients possibly due to shifts into
the brain 106. A neurophysiological explanation of an ECT mechanism is that repeated
electrically-induced seizures increase the ST, and therefore mood stability, by increasing the
activity of GABAergic neurons that regulate the function of neurocircuits, thereby relieving

Harv Rev Psychiatry. Author manuscript; available in PMC 2024 January 01.
Kritzer et al. Page 10

aspects of neurovegetative depression such as rumination, anxious distress and even mania
or catatonia 107. There are other potential mechanisms, recently reviewed by Leaver, et al,
Author Manuscript

suggesting a role in the thalamus and cerebellum in the attenuation of seizure activity after
ECT as the data supporting the GABA hypothesis has not been completely consistent 108.

Hormonal changes:
In humans, serum prolactin levels increase in response to seizures, with a 10- to 50-fold
increase observed after ECT 109. This has been observed in severely depressed patients,
with ECT resulting in increases of prolactin and estrogen stimulating neurophysin (ESN)
for at least 6 minutes post-ECT 110. This was also observed for a shorter time course
for nicotine stimulating neurophysin (NSN) but no other stress/hypothalamic hormones
increased this dramatically, indicating that ECT has specific hormonal effects that cannot be
generalized to the release of any or all pituitary or hypothalamic stress-induced hormones
Author Manuscript

110. Another important marker of stress, corticotropin-releasing factor (CRF), has been

studied, though with mixed results. Nemeroff and colleagues found that cerebrospinal
fluid (CSF) concentrations of CRF in depressed patients decreased after ECT 111. Kling
and colleagues also found that CRF levels decrease after a successful course of ECT
112. However, other studies have found no significant change in CSF-CRF following ECT

despite clinical improvement 113,114. Interestingly, Banki and colleagues noted that in the 15
patients who did not relapse within 6 months of ECT treatment, the CSF-CRF concentration
was indeed reduced post-ECT 113. Cortisol levels have also been reported to be increased
following ECT treatment, potentially due to increased arginine vasopressin stimulation of
CRF-induced acetylcholine release 114,115. Lastly, β-endorphin was shown to be transiently
increased following ECT administration in depressed patients 115,116.

Some studies have shown that growth hormone is decreased in depressed patients following
Author Manuscript

ECT, though other studies have found no statistical difference 115,117. Generally, stress
hormone and neuropeptide changes have been documented, though given the heterogeneity
of sample populations, sizes, and parameters, there has been a failure to establish a
consensus. While the immediate release of stress hormones such as prolactin and cortisol
may be important in the mechanism of ECT, it may also simply be an epiphenomenon and
marker of seizure activity with little reliable insights into the therapeutic effects of ECT 118.

A few other studies have attempted to identify molecular substrates that may be involved
in the therapeutic effect of ECT in human subjects. The kynurenine pathway and its
metabolites have emerged as potential mediators in the pathophysiology of depression and
the response to ECT 119. Tryptophan, the amino acid basis for serotonin, degrades along
the kynurenine pathway. Also, tryptophan consists of, among other compounds, QUIN,
Author Manuscript

an NMDA-receptor agonist, and kynurenic acid (KYNA), an NMDA-receptor antagonist.


QUIN upregulation and increased QUIN/KYNA plasma ratios have been observed in
depressed patients. Schwieler et al. found that after ECT, QUIN plasma levels and QUIN/
KYNA ratios were decreased in 80% of patients 119. The authors suggest that the capacity
of ECT to decrease the neurotoxic branch of the kynurenine pathway may be of clinical
importance. However, they mention that plasma QUIN levels correlate with ECT treatment
but not with the response to ECT as measured by the Montgomery-Asberg Depression

Harv Rev Psychiatry. Author manuscript; available in PMC 2024 January 01.
Kritzer et al. Page 11

Scale (MADRS), consistent with another finding that QUIN levels correlate suicidality but
not depression generally 120. Furthermore, QUIN and KYNA, as the authors acknowledge,
Author Manuscript

do not easily pass the blood-brain barrier (BBB), and may not reflect significant changes
in the CSF. Considering ECT may disrupt the integrity of the BBB these biomarkers
may consequently have relevance to depression phenotyping and may potentially become
predictive of ECT response, though determining the appropriate time to assay them is of
high importance 121.

Serotonin:
Neurotransmitters and their receptors have been studied extensively in depression though, to
a lesser degree, in response to ECT. Serotonin (5-hydroxytryptophan, 5-HT), its receptors
(5-HT1–7), and its transporters have been investigated thoroughly in the pathogenesis of
depression and the use of antidepressants, specifically selective serotonin reuptake inhibitors
Author Manuscript

(SSRIs). The theory of low serotonin levels and depression has been popular, though not
clearly substantiated and perhaps even untenable 122.

Serotonin receptor 5-HT1A postsynaptic density was found to be decreased following ECT
in depressed patients 123. Ishihara and colleagues found that 5-HT1A postsynaptic receptors
are sensitized to 5-HT following ECT treatment 124. Widespread reduction of brain 5-HT2
receptors has been observed in depressed patients following ECT, similarly to what has been
observed with antidepressants 125. Interestingly, peripheral platelet 5-HT2 receptor density
has been reported to be increased after ECT in depressed patients 126. Plein and Berk
found that platelet 5-HT2A receptors are desensitized to serotonin following ECT, and they
hypothesized that this decreased sensitivity may be a mechanism of ECT’s antidepressant
action 127. Ishihara and Sasa noted that the 5-HT1A increased sensitivity and perhaps
subsequent decrease in 5-HT2A receptors explain the effects of ECT; they also point out
Author Manuscript

that 5-HT3 sensitization may play an important role 124,128. Additionally, they have shown
that ECT increases the release of glutamate and GABA and decrease the receptor function
of noradrenergic and dopaminergic neurons in the locus coeruleus and substantia nigra
respectively, thereby increasing noradrenaline and dopamine. These effects may also be
mediated through the 5-HT3 receptor sensitization.

Magnetic resonance spectroscopy:


Investigators have utilized proton magnetic resonance spectroscopic (1H-MRS) to measure
neurotransmitter levels and their metabolites in the brains of patients with major depression
before and after ECT – and details can be found in recent thorough reviews 129,130. The
results have been heterogeneous, but trends are emerging. One study found no significant
differences in GABA, Glutamate (Glu), glutamine (Gln), choline (Cho) or glutathione
Author Manuscript

(GSH) between pre- and post-ECT, though there was a decrease in N-acetylaspartate (NAA)
levels after ECT that correlated with the number of ECT sessions 131. A study in RUL ECT
in patients with TRD showed that Gln concentrations were anti-correlated with their degree
of depression; and after a successful ECT course, Glx (Glu + Gln) increased significantly
to that of age-matched controls 132. Another study found increased levels Glx in depressed
patient that responded to ECT 133. One group found significantly reduced Glx levels in the

Harv Rev Psychiatry. Author manuscript; available in PMC 2024 January 01.
Kritzer et al. Page 12

patients’ left cingulum at baseline compared to healthy controls that normalized after ECT
134.
Author Manuscript

A detailed anatomical 1H-MRS study measured changes in Glx, Creatinine (Cre), Cho and
NAA in the dorsal (dACC) and subgenual anterior cingulate cortex (sgACC) as well as
the bilateral hippocampus in patients receiving ECT for depression. At baseline, patients
showed less Glx in the sgACC, less NAA in the left hippocampus and more Glx in the left
hippocampus compared to controls. After ECT they observed significant increases in Cre in
the dACC and sgACC and decreases in NAA within the dACC and right hippocampus.
Both ECT and symptom improvement were associated with decreased Glx in the left
hippocampus and increased Glx in the sgACC. Lower NAA levels in the dACC at baseline
predicted ECT-induced reductions in depressive symptoms. Changes in Glx in the sgACC
and hippocampus approached that if controls after ECT. This may reflect and imbalance
between underactive cortical and overactive subcortical limbic circuitry in patients with
Author Manuscript

depression 135. Severe depression appears to be associated with decreased Glx and therefore
increasing Glx may be important to the mechanism and a potential biomarker of ECT
response 136.

Neuroanatomical changes:
Magnetic resonance imaging (MRI) studies after ECT consistently show volumetric changes
across cortical and subcortical brain regions 137. Anatomical changes correlate with number
of treatments, likely with the strength of the electric field 54, and the placement of electrodes
61. Recent mega-analysis corroborated this by illustrating approximately 5% increase in

the gray matter subfields of the hippocampus and the basolateral nuclei of the amygdala.
Beyond the subcortical mesial temporal lobe structures, volumetric increases have been
observed in the anterior cingulate, postcentral, fusiform gyri, the medial prefrontal cortex,
Author Manuscript

supplementary motor cortex, insula, and striatum 138. The Global ECT-MRI Research
Collaboration (GEMRIC) performed a multisite analysis of individual-level subject data
and showed that, except for the cerebellum, the volumetric expansion of all subcortical
regions of interest negatively correlated with ventricle size, indicating that the increases
was likely at the expense of cerebrospinal fluid spaces 137. These mega-analysis findings
support widespread brain changes regardless of known relation to neurocircuits implicated in
depression (i.e., default mode network, cortico-striatal-thalamic, and cortico-hypothalamic
circuits) 139. Large-scale data consistently point to changes in mesial temporal lobe
(e.g., hippocampus and amygdala), far from the ECT electrodes, undergoing the greatest
volumetric changes. Smaller studies did not consistently find volume changes outside of the
mesial temporal lobe where effect sizes seem to be larger, possibly due to the lower sample
sizes and associated reduced statistical power.140. Understanding how these changes support
Author Manuscript

response to ECT (therapeutic and iatrogenic), and the neurocircuits involved, remains an
area of exploration 108.

Neurogenesis and Brain-Derived Neurotropic Factor:


Smaller hippocampi have been observed in patients with depression in MRI as well as
post-mortem studies 141,142. Intriguingly, hippocampal size is increased after ECT and may
be dose- and parameter-dependent 143. A recent post-mortem human study of patients that

Harv Rev Psychiatry. Author manuscript; available in PMC 2024 January 01.
Kritzer et al. Page 13

underwent ECT displayed an increase in doublecortin, a marker of neurogenesis, in the


dentate gyrus 144. Whether adult neurogenesis occurs in humans remains controversial
Author Manuscript

145,146. Regardless, it occurs in rodents and is increased by electroconvulsive stimulation

(ECS) 147,148. The neuroplasticity hypothesis is based on work in animal models suggesting
a dose-dependent increase of neurogenesis in the dentate gyrus of the hippocampus after
ECS, the preclinical approach to studying mechanisms of ECT. Additionally, in mice, the
behaviors associated with ECT, were shown to be reliant on neurogenesis 149–151.

There is evidence supporting the role of neurotrophic factors in human affective disorders.
Decreased brain-derived neurotrophic factor (BDNF) levels have been observed in humans
with depression 152. BDNF has received considerable attention in ECS research as well
as depression research broadly 153. BDNF mediates long-term neural and behavioral
plasticity during social stress. In addition, BDNF expression has been implicated as a
potential mediator in depression as decreased levels may atrophy limbic structures, while
Author Manuscript

its upregulation plays a role in antidepressant response 154. Karege and colleagues found
that depressed patients had significantly lower serum levels of BDNF, which is increased
in individuals responsive to ketamine 155,156. Additionally, patients with TRD have lower
baseline levels of plasma BDNF which are significantly increased by ECT. Furthermore,
Piccinni and colleagues found that BDNF levels increase at the same time of observed
clinical improvement, suggesting that changes of BDNF levels in response to ECT may
represent a biological, state-dependent marker of remission following ECT 157. This is in
line with the belief that BDNF levels are associated with clinical changes in depression and
that neuroplastic changes may underlie these clinical changes 158.

Gliogenesis and Angiogenesis:


Cell proliferation induced by ECS includes glial (astrocytes, oligodendrocytes, and
Author Manuscript

microglia) and endothelial cells in addition to neurons. Wennstrom and colleagues reported
glial cell proliferation in the adult rat hippocampus following ECS, and that inhibition
of gliogenesis by corticosterone administration is mitigated by ECS 159,160. Their group
also notes that glial cell proliferation increases in the rat amygdala following ECS 161.
As corticosterone inhibition of gliogenesis in the rat hippocampus is counteracted by
ECS, the mechanism of ECS’s antidepressant effect may be influenced by glial cell
proliferation. Interestingly, glial cell proliferation after ECS has also been reported in the
rat frontal cortex 162. The increase in number of these glial cells may exert a neuroprotective
effect and counteract the effects of stress, thereby implementing an antidepressant effect.
Additionally, endothelial cell proliferation in response to ECS has been observed in the rat
prefrontal cortex 163, hypothalamic regions including the paraventricular nucleus, supraoptic
nucleus, and ventromedial nucleus in response to ECS 164. Endothelial cell proliferation
Author Manuscript

and increased angiogenesis has also been observed in the adult rat hippocampus following
ECS and this increased vascularization may be needed to support hippocampal neurogenesis
165,166. These observed endothelial cell inductions support angiogenesis as a potentially

important component of ECS’s mechanism.

Microglial cells are resident macrophages that enter the brain early in development, and
while they have homeostatic roles in brain function, they can cause neuroinflammation

Harv Rev Psychiatry. Author manuscript; available in PMC 2024 January 01.
Kritzer et al. Page 14

167.One study showed that mice subjected to chronic social defeat stress and subsequent
ECS had activated hippocampal microglial cells 168, while a study comparing ECS to a
Author Manuscript

model of status epilepticus showed no changes in hippocampal CA1 microglial density 169.
Taking these findings together, it is possible that the antidepressant effects of ECS may be
collectively mediated by neuro-, glio-, and angiogenesis, mainly in the hippocampus and
associated brain structures. Overall, given the brain regions involved, the rodent literature
appears to support changes involving the hippocampus, amygdala, hypothalamus, and
frontal cortical regions, though no direct interrogations of the circuits connecting these
regions have been conducted.

Perspective and Conclusion:


ECT remains one of the most effective treatments for several neuropsychiatric conditions,
particularly depression. Understanding the neurobiological mechanisms that underlie its
Author Manuscript

effects could provide important opportunities to optimize ECT as well as reveal response
biomarkers and treatment targets to engaged with novel, safe, and efficacious therapeutic
approaches 170. Animal models have revealed molecular and cellular insights into the
effects of ECT despite their translational limitations. There are many caveats to modeling
depression in rodents, including the fact that the evaluation of antidepressant treatment
efficacy is state-dependent 171–174 and cannot easily be reproduced in animals, especially in
rodents. For example, ECT is frequently used in geriatric patients with severe depression,
particularly with psychotic features 175. Rodent studies are almost entirely performed around
12 weeks of age, corresponding to young adult in humans 176,177. Therefore, the findings,
while informative, are not likely translatable to older populations, which have a much
higher risk and rate of cognitive deficits and delayed recovery times 178,179. Additionally,
while recent data suggests multiple brain region changes after ECT, we are just starting to
Author Manuscript

understand the neurocircuitry of ECT effects and how to optimize a circuit-based approach,
such as pairing electrode placement with specific and intentional stimulus parameter
selection. For example, a recent study has shown an increase in frontal lobe activity and
the executive control network with potential fMRI resting-state biomarkers that include
the dorsomedial and anterior cingulate cortex after ECT 180–182. Another study indicated
that connectivity between the dorsal prefrontal region and the limbic and default-mode
networks serves as a significant predictor of response to ECT 183. Is the future of ECT
one that uses fMRI-guided electrode placement to optimize antidepressant and cognitive
outcomes? Or perhaps, current-amplitude-titrated or electric-field-informed dosing based on
hippocampal changes? In this context, it may be relevant to disentangle the direct effects of
the ECT electrical stimulus versus the seizure it induces, including drawing parallels with
subconvulsive stimulation methods such as TMS.
Author Manuscript

Our understanding of the mechanisms of ECT efficacy and adverse effects on cognition are
far from sufficient. Is it a coincidence that the hippocampus and amygdala are consistently
implicated in the mechanisms of ECT and yet these are the same brain regions responsible
for episodic/autobiographical memory, loss of which is the most concerning side effect
of ECT? In addition to research of fundamental mechanisms, evaluation of mitigation
approaches for optimization of outcomes is paramount. We can consider undertaking
large retrospective studies on concomitant medication and comorbidities so that adjunctive

Harv Rev Psychiatry. Author manuscript; available in PMC 2024 January 01.
Kritzer et al. Page 15

treatments and screening can be implemented. Additional longitudinal studies looking at


these data are necessary to begin to determine predictors or outcomes and side effects 57.
Author Manuscript

As these data emerge, meta- and mega-analyses of ECT responders and non-responders may
help us triage treatments for those at risk of cognitive side effects. Further evaluation into
electrode placement and stimulation parameters is needed to determine clinical approaches
to improving outcomes. Additionally, there may be inflammatory and/or hormonal profiles
that are relatively indicated or contraindicated. While there is no “unified theory” of
ECT mechanism and how it relates to seemingly divergent psychiatric illness from mood
disorders to catatonia, lines of evidence point to the modulation of dysregulated neurocircuit
function in disease states by repeated application of electric fields and/or seizure activity.
This is likely achieved by enhancing the regulation of aberrant neurocircuit activity at
various levels of biological function (e.g. molecular, cellular, anatomical, physiological),
which results in more adaptive circuit dynamics, in turn leading to euthymia. A better
Author Manuscript

understanding of neurocircuits dynamics and molecular mechanisms associated with ECT


efficacy and side effects would inform both research on treatment development and clinical
treatment planning and would reduce stigma.

Disclosures:
J. A. Camprodon, MD, MPH, PhD, serves on the scientific advisory board of Hyka Therapeutics and Feelmore
Labs; he has also been a consultant for Neuronetics. A. V. Peterchev has received research funding, travel
support, patent royalties, consulting fees, equity options, equipment loans, hardware donations, and/or patent
application support from Rogue Research, Magstim, MagVenture, Neuronetics, BTL Industries, Advise Connect
Inspire, Soterix, and Ampa. M. D. Kritzer, MD, PhD, receives funding from MGH Training Grant Translational
Neuroscience Training for Clinicians (T32MH112485) and the Harvard Medical School Department of Psychiatry
Dupont Warren Grant. This work was also supported in part by the National Institute of Mental Health of the
National Institutes of Health under award number R01MH091083 (AVP) and R01MH112737 (JAC). The content
is solely the responsibility of the authors and does not necessarily represent the official views of the National
Institutes of Health.
Author Manuscript

References:
1. Fink M, Abrams R. ECT has proved effective in treating depression. Nature. 2000.
doi:10.1038/35002776
2. Kumar S, Mulsant BH, Liu AY, Blumberger DM, Daskalakis ZJ, Rajji TK. Systematic Review of
Cognitive Effects of Electroconvulsive Therapy in Late-Life Depression. Am J Geriatr Psychiatry.
2016. doi:10.1016/j.jagp.2016.02.053
3. Medda P, Perugi G, Zanello S, Ciuffa M, Cassano GB. Response to ECT in bipolar I, bipolar II and
unipolar depression. J Affect Disord. 2009. doi:10.1016/j.jad.2009.01.014
4. UK ECT Review Group. Efficacy and safety of electroconvulsive therapy in depressive disorders:
a systematic review and meta-analysis. Lancet (London, England). 2003;361(9360):799–808.
doi:10.1016/S0140-6736(03)12705-5 [PubMed: 12642045]
5. Pagnin D, De Queiroz V, Pini S, Cassano GB. Efficacy of ECT in Depression: A Meta-Analytic
Author Manuscript

Review. J ECT. 2004. doi:10.1097/00124509-200403000-00004


6. Medda P, Toni C, Perugi G. The Mood-stabilizing effects of electroconvulsive therapy. J ECT. 2014.
doi:10.1097/YCT.0000000000000160
7. Luchini F, Medda P, Mariani MG, Mauri M, Toni C, Perugi G. Electroconvulsive therapy in
catatonic patients: Efficacy and predictors of response. World J Psychiatry. 2015. doi:10.5498/
wjp.v5.i2.182
8. Fink M, Kellner CH, McCall WV. The role of ECT in suicide prevention. In: Journal of ECT.; 2014.
doi:10.1097/YCT.0b013e3182a6ad0d

Harv Rev Psychiatry. Author manuscript; available in PMC 2024 January 01.
Kritzer et al. Page 16

9. Havaki-Kontaxaki BJ, Ferentinos PP, Kontaxakis VP, Paplos KG, Soldatos CR. Concurrent
administration of clozapine and electroconvulsive therapy in clozapine-resistant schizophrenia. Clin
Author Manuscript

Neuropharmacol. 2006. doi:10.1097/00002826-200601000-00012


10. Semkovska M, McLoughlin DM. Objective cognitive performance associated with
electroconvulsive therapy for depression: A systematic review and meta-analysis. Biol Psychiatry.
2010. doi:10.1016/j.biopsych.2010.06.009
11. Fosse R, Read J. Electroconvulsive treatment: Hypotheses about mechanisms of action. Front
Psychiatry. 2013. doi:10.3389/fpsyt.2013.00094
12. Zhu A, Phuong M, Giacobbe P. The Story of ECT: Behind the Scenes of a Controversial yet
Effective Treatment. Comics Grid J Comics Scholarsh. 2018. doi:10.16995/cg.129
13. Taylor S Electroconvulsive therapy: A review of history, patient selection, technique, and
medication management. South Med J. 2007. doi:10.1097/SMJ.0b013e318038fce0
14. Payne NA, Prudic J. Electroconvulsive therapy: Part II: A biopsychosocial perspective. J Psychiatr
Pract. 2009. doi:10.1097/01.pra.0000361278.73092.85
15. Sackeim HA, Prudic J, Nobler MS, et al. Effects of pulse width and electrode placement on
the efficacy and cognitive effects of electroconvulsive therapy. Brain Stimul. 2008. doi:10.1016/
Author Manuscript

j.brs.2008.03.001
16. Tor PC, Bautovich A, Wang MJ, Martin D, Harvey SB, Loo C. A Systematic review and meta-
analysis of brief versus ultrabrief right unilateral electroconvulsive therapy for depression. ?. 2015.
doi:10.4088/JCP.14r09145
17. Squire LR, Zouzounis JA. ECT and memory: Brief pulse versus sine wave. Am J Psychiatry. 1986.
doi:10.1176/ajp.143.5.596
18. Rhee TG, Sint K, Olfson M, Gerhard T, H Busch S, Wilkinson ST. Association of ECT With
Risks of All-Cause Mortality and Suicide in Older Medicare Patients. https://doi.org/101176/
appi.ajp202121040351. 2021;178(12):1089–1097. doi:10.1176/APPI.AJP.2021.21040351
19. Luccarelli J, McCoy TH, Seiner SJ, Henry ME. Maintenance ECT is associated with sustained
improvement in depression symptoms without adverse cognitive effects in a retrospective cohort
of 100 patients each receiving 50 or more ECT treatments. J Affect Disord. 2020;271:109–114.
doi:10.1016/J.JAD.2020.03.152 [PubMed: 32479305]
20. Luccarelli J, McCoy TH, Shannon AP, Forester BP, Seiner SJ, Henry ME. Rate of continuing
Author Manuscript

acute course treatment using right unilateral ultrabrief pulse electroconvulsive therapy at a large
academic medical center. Eur Arch Psychiatry Clin Neurosci. 2021;271(1):191–197. doi:10.1007/
S00406-020-01202-2 [PubMed: 33196856]
21. Luccarelli J, McCoy TH, Shannon AP, Forester BP, Seiner SJ, Henry ME. Duration of Treatment
in Electroconvulsive Therapy Among Patients Beginning With Acute Course Right Unilateral
Brief Pulse Stimuli. J ECT. 2021;37(4):238–242. doi:10.1097/YCT.0000000000000768 [PubMed:
33840804]
22. Luccarelli J, McCoy TH, Seiner SJ, Henry ME. Real-world evidence of age-independent
electroconvulsive therapy efficacy: A retrospective cohort study. Acta Psychiatr Scand.
2022;145(1):100–108. doi:10.1111/ACPS.13378 [PubMed: 34662429]
23. Luccarelli J, Forester BP, Dooley M, et al. The Effects of Baseline Impaired Global Cognitive
Function on the Efficacy and Cognitive Effects of Electroconvulsive Therapy in Geriatric Patients:
A Retrospective Cohort Study. Am J Geriatr Psychiatry. 2022;30(7):790–798. doi:10.1016/
j.jagp.2021.12.008 [PubMed: 34996701]
24. Lee WH, Lisanby SH, Laine AF, Peterchev AV. Comparison of electric field strength and spatial
Author Manuscript

distribution of electroconvulsive therapy and magnetic seizure therapy in a realistic human head
model. Eur Psychiatry. 2016;36:55–64. doi:10.1016/J.EURPSY.2016.03.003 [PubMed: 27318858]
25. Unal G, Swami JK, Canela C, et al. Adaptive current-flow models of ECT: Explaining individual
static impedance, dynamic impedance, and brain current density. Brain Stimul. 2021;14(5):1154–
1168. doi:10.1016/J.BRS.2021.07.012 [PubMed: 34332156]
26. Dragovic M, Allet L, Janca A. Electroconvulsive therapy and determination of cerebral dominance.
Ann Gen Hosp Psychiatry. 2004;3:14. doi:10.1186/1475-2832-3-14 [PubMed: 15306035]
27. Pratt RT, Warrington EK. The Assessment of Cerebral Dominance with Unilateral ECT. Br J
Psychiatry. 1972;121(562):327–328. doi:10.1192/BJP.121.3.327 [PubMed: 5073786]

Harv Rev Psychiatry. Author manuscript; available in PMC 2024 January 01.
Kritzer et al. Page 17

28. Peterchev AV, Rosa MA, Deng Z De, Prudic J, Lisanby SH. Electroconvulsive therapy stimulus
parameters: rethinking dosage. J ECT. 2010;26(3):159–174. doi:10.1097/yct.0b013e3181e48165
Author Manuscript

[PubMed: 20805726]
29. Sackeim HA, Prudic J, Devanand DP, et al. Effects of Stimulus Intensity and Electrode Placement
on the Efficacy and Cognitive Effects of Electroconvulsive Therapy. N Engl J Med. 1993.
doi:10.1056/NEJM199303253281204
30. Sackeim HA, Prudic J, Devanand DP, et al. A prospective, randomized, double-blind comparison
of bilateral and right unilateral electroconvulsive therapy at different stimulus intensities. Arch
Gen Psychiatry. 2000. doi:10.1001/archpsyc.57.5.425
31. Sienaert P, Vansteelandt K, Demyttenaere K, Peuskens J. Randomized comparison of ultra-brief
bifrontal and unilateral electroconvulsive therapy for major depression: cognitive side-effects. J
Affect Disord. 2010. doi:10.1016/j.jad.2009.06.011
32. Sienaert P, Vansteelandt K, Demyttenaere K, Peuskens J. Randomized comparison of ultra-brief
bifrontal and unilateral electroconvulsive therapy for major depression: Clinical efficacy. J Affect
Disord. 2009;116(1–2):106–112. doi:10.1016/J.JAD.2008.11.001 [PubMed: 19081638]
33. Quante A, Luborzewski A, Brakemeier EL, Merkl A, Danker-Hopfe H, Bajbouj M. Effects of
Author Manuscript

3 different stimulus intensities of ultrabrief stimuli in right unilateral electroconvulsive therapy


in major depression: A randomized, double-blind pilot study. J Psychiatr Res. 2011. doi:10.1016/
j.jpsychires.2010.08.006
34. Tor PC, Bautovich A, Wang MJ, Martin D, Harvey SB, Loo C. A Systematic review and meta-
analysis of brief versus ultrabrief right unilateral electroconvulsive therapy for depression. J Clin
Psychiatry. 2015;76(9):e1092–e1098. doi:10.4088/JCP.14r09145 [PubMed: 26213985]
35. Eschweiler GW, Vonthein R, Bode R, et al. Clinical efficacy and cognitive side effects of bifrontal
versus right unilateral electroconvulsive therapy (ECT): A short-term randomised controlled trial
in pharmaco-resistant major depression. J Affect Disord. 2007. doi:10.1016/j.jad.2006.11.012
36. Semkovska M, Landau S, Dunne R, et al. Bitemporal Versus High-Dose Unilateral Twice-Weekly
Electroconvulsive Therapy for Depression (EFFECT-Dep): A Pragmatic, Randomized, Non-
Inferiority Trial. Am J Psychiatry. 2016;173(4):408–417. doi:10.1176/APPI.AJP.2015.15030372
[PubMed: 26892939]
37. Kellner CH, Knapp R, Husain MM, et al. Bifrontal, bitemporal and right unilateral electrode
placement in ECT: randomised trial. Br J Psychiatry. 2010;196(3):226–234. doi:10.1192/
Author Manuscript

BJP.BP.109.066183 [PubMed: 20194546]


38. Glynn K, Therese O, Lyons D. Cognitive deficits post electroconvulsive therapy: Latest research
and future directions. In: Advances in Psychology Research.; 2015.
39. Sahlem GL, Short EB, Kerns S, et al. Expanded Safety and Efficacy Data for a New Method of
Performing Electroconvulsive Therapy: Focal Electrically Administered Seizure Therapy. J ECT.
2016;32(3):197–203. doi:10.1097/YCT.0000000000000328 [PubMed: 27379790]
40. Sahlem GL, McCall WV., Short EB, et al. A two-site, open-label, non-randomized trial comparing
Focal Electrically-Administered Seizure Therapy (FEAST) and right unilateral ultrabrief pulse
electroconvulsive therapy (RUL-UBP ECT). Brain Stimul. 2020;13(5):1416–1425. doi:10.1016/
j.brs.2020.07.015 [PubMed: 32735987]
41. Spellman T, Peterchev AV., Lisanby SH. Focal electrically administered seizure therapy: a novel
form of ECT illustrates the roles of current directionality, polarity, and electrode configuration in
seizure induction. Neuropsychopharmacology. 2009;34(8):2002–2010. doi:10.1038/npp.2009.12
[PubMed: 19225453]
Author Manuscript

42. Youssef NA, George MS, McCall WV., et al. The Effects of Focal Electrically Administered
Seizure Therapy Compared With Ultrabrief Pulse Right Unilateral Electroconvulsive Therapy
on Suicidal Ideation: A 2-Site Clinical Trial. J ECT. 2021;37(4):256–262. doi:10.1097/
YCT.0000000000000776 [PubMed: 34015791]
43. Lisanby SH, Luber B, Schlaepfer TE, Sackeim HA. Safety and feasibility of magnetic
seizure therapy (MST) in major depression: randomized within-subject comparison with
electroconvulsive therapy. Neuropsychopharmacology. 2003;28(10):1852–1865. doi:10.1038/
sj.npp.1300229 [PubMed: 12865903]

Harv Rev Psychiatry. Author manuscript; available in PMC 2024 January 01.
Kritzer et al. Page 18

44. Deng Z De Lisanby SH, Peterchev AV. Controlling stimulation strength and focality
in electroconvulsive therapy via current amplitude and electrode size and spacing:
Author Manuscript

comparison with magnetic seizure therapy. J ECT. 2013;29(4):325. doi:10.1097/YCT.10.1097/


YCT.0B013E3182A4B4A7 [PubMed: 24263276]
45. Daskalakis ZJ, Dimitrova J, McClintock SM, et al. Magnetic seizure therapy (MST) for
major depressive disorder. Neuropsychopharmacol 2019 452. 2019;45(2):276–282. doi:10.1038/
s41386-019-0515-4
46. Weissman CR, Blumberger DM, Dimitrova J, et al. Magnetic Seizure Therapy for
Suicidality in Treatment-Resistant Depression. JAMA Netw open. 2020;3(8). doi:10.1001/
JAMANETWORKOPEN.2020.7434
47. Tang VM, Blumberger DM, Hill AT, et al. Magnetic Seizure Therapy for the Treatment
of Suicidality in Bipolar Depression. Biol Psychiatry. 2021;90(10):e51–e53. doi:10.1016/
j.biopsych.2020.09.020 [PubMed: 33172609]
48. Lee WH, Lisanby SH, Laine AF, Peterchev AV. Minimum Electric Field Exposure
for Seizure Induction with Electroconvulsive Therapy and Magnetic Seizure Therapy.
Neuropsychopharmacology. 2017;42(6):1192–1200. doi:10.1038/NPP.2016.276 [PubMed:
Author Manuscript

27934961]
49. Rosa MA, Abdo GL, Lisanby SH, Peterchev A. Seizure induction with low-amplitude-current (0.5
A) electroconvulsive therapy. J ECT. 2011;27(4):341–342. doi:10.1097/YCT.0B013E31822149DB
50. Deng Z De Lisanby SH, Peterchev AV. Controlling stimulation strength and focality
in electroconvulsive therapy via current amplitude and electrode size and spacing:
comparison with magnetic seizure therapy. J ECT. 2013;29(4):325–335. doi:10.1097/yct.10.1097/
yct.0b013e3182a4b4a7 [PubMed: 24263276]
51. Youssef NA, Sidhom E. Feasibility, safety, and preliminary efficacy of Low Amplitude Seizure
Therapy (LAP-ST): A proof of concept clinical trial in man. J Affect Disord. 2017;222:1–6.
doi:10.1016/J.JAD.2017.06.022 [PubMed: 28667887]
52. Youssef N, Ravilla D, McCall W, et al. Magnitude of Reduction & Speed of Remission of
Suicidality for Low AmPlitude Seizure Therapy (LAP-ST) Compared to Standard Right Unilateral
ECT. Brain Stimul Basic, Transl Clin Res Neuromodulation. 2019;12(2):439. doi:10.1016/
J.BRS.2018.12.422
53. Abbott CC, Quinn D, Miller J, et al. Electroconvulsive Therapy Pulse Amplitude and
Author Manuscript

Clinical Outcomes. Am J Geriatr Psychiatry. 2021;29(2):166–178. doi:10.1016/j.jagp.2020.06.008


[PubMed: 32651051]
54. Argyelan M, Oltedal L, Deng Z De, et al. Electric field causes volumetric changes in the human
brain. Elife. 2019;8. doi:10.7554/ELIFE.49115
55. Joshi SH, Espinoza RT, Pirnia T, et al. Structural plasticity of the hippocampus and amygdala
induced by electroconvulsive therapy in major depression. Biol Psychiatry. 2016. doi:10.1016/
j.biopsych.2015.02.029
56. Cano M, Lee E, Worthley A, et al. Electroconvulsive therapy effects on anhedonia and
reward circuitry anatomy: A dimensional structural neuroimaging approach. J Affect Disord.
2022;313:243–250. doi:10.1016/J.JAD.2022.06.062 [PubMed: 35764228]
57. Cano M, Martínez-Zalacaín I, Bernabéu-Sanz, et al. Brain volumetric and metabolic correlates of
electroconvulsive therapy for treatment-resistant depression: a longitudinal neuroimaging study.
Transl Psychiatry. 2017;7(2):e1023. doi:10.1038/TP.2016.267 [PubMed: 28170003]
58. Van Den Bossche MJA, Emsell L, Dols A, et al. Hippocampal volume change following ECT is
Author Manuscript

mediated by rs699947 in the promotor region of VEGF. Transl Psychiatry. 2019;9(1). doi:10.1038/
S41398-019-0530-6
59. Deng Z De, Argyelan M, Miller J, et al. Electroconvulsive therapy, electric field, neuroplasticity,
and clinical outcomes. Mol Psychiatry. 2022;27(3):1676. doi:10.1038/S41380-021-01380-Y
[PubMed: 34853404]
60. Fridgeirsson EA, Deng Z De, Denys D, van Waarde JA, van Wingen GA. Electric field strength
induced by electroconvulsive therapy is associated with clinical outcome. NeuroImage Clin.
2021;30. doi:10.1016/j.nicl.2021.102581

Harv Rev Psychiatry. Author manuscript; available in PMC 2024 January 01.
Kritzer et al. Page 19

61. Cano M, Lee E, Cardoner N, et al. Brain Volumetric Correlates of Right Unilateral Versus
Bitemporal Electroconvulsive Therapy for Treatment-Resistant Depression. J Neuropsychiatry
Author Manuscript

Clin Neurosci. 2019;31(2):152–158. doi:10.1176/APPI.NEUROPSYCH.18080177 [PubMed:


30458664]
62. Regenold WT, Noorani RJ, Piez D, Patel P. Nonconvulsive Electrotherapy for Treatment Resistant
Unipolar and Bipolar Major Depressive Disorder: A Proof-of-concept Trial. Brain Stimul Basic,
Transl Clin Res Neuromodulation. 2015;8(5):855–861. doi:10.1016/J.BRS.2015.06.011
63. Cole EJ, Stimpson KH, Bentzley BS, et al. Stanford accelerated
intelligent neuromodulation therapy for treatment-resistant depression. Am J
Psychiatry. 2020;177(8):716–726. doi:10.1176/APPI.AJP.2019.19070720/ASSET/IMAGES/
LARGE/APPI.AJP.2019.19070720F3.JPEG [PubMed: 32252538]
64. Cole EJ, Phillips AL, Bentzley BS, et al. Stanford Neuromodulation Therapy (SNT): A
Double-Blind Randomized Controlled Trial. 10.1176/appi.ajp202120101429. 2021;179(2):132–
141. doi:10.1176/APPI.AJP.2021.20101429
65. Peterchev A V, Krystal AD, Rosa MA, Lisanby SH. Individualized low-amplitude
seizure therapy: minimizing current for electroconvulsive therapy and magnetic seizure
Author Manuscript

therapy. Neuropsychopharmacology. 2015;40(9):2076–2084. doi:10.1038/npp.2015.122 [PubMed:


25920013]
66. Ahern E, Semkovska M. Cognitive functioning in the first-episode of major depressive disorder:
A systematic review and meta-analysis. Neuropsychology. 2017;31(1):52–72. doi:10.1037/
NEU0000319 [PubMed: 27732039]
67. Rose D, Wykes T, Leese M, Bindman J, Fleischmann P. Patients’ perspectives on electroconvulsive
therapy: Systematic review. Br Med J. 2003. doi:10.1136/bmj.326.7403.1363
68. Brus O, Nordanskog P, Båve U, et al. Subjective Memory Immediately Following
Electroconvulsive Therapy. J ECT. 2017;33(2):96–103. doi:10.1097/YCT.0000000000000377
[PubMed: 27930429]
69. Robertson H, Pryor R. Memory and cognitive effects of ECT: informing and assessing patients.
Adv Psychiatr Treat. 2006;12(3):228–237. doi:10.1192/APT.12.3.228
70. Semkovska M, McLoughlin DM. Objective cognitive performance associated with
electroconvulsive therapy for depression: a systematic review and meta-analysis. Biol Psychiatry.
2010;68(6):568–577. doi:10.1016/j.biopsych.2010.06.009 [PubMed: 20673880]
Author Manuscript

71. Mohn C, Rund BR. Neurocognitive profile in major depressive disorders: relationship to
symptom level and subjective memory complaints. BMC Psychiatry. 2016;16(1). doi:10.1186/
S12888-016-0815-8
72. Porter RJ, Baune BT, Morris G, et al. Cognitive side-effects of electroconvulsive therapy: what
are they, how to monitor them and what to tell patients. BJPsych Open. 2020;6(3). doi:10.1192/
BJO.2020.17
73. Semkovska M, McLoughlin DM. Measuring retrograde autobiographical amnesia following
electroconvulsive therapy historical perspective and current issues. J ECT. 2013;29(2):127–133.
doi:10.1097/YCT.0B013E318279C2C9 [PubMed: 23303426]
74. Disner SG, Beevers CG, Haigh EAP, Beck AT. Neural mechanisms of the cognitive model of
depression. Nat Rev Neurosci. 2011;12(8):467–477. doi:10.1038/NRN3027 [PubMed: 21731066]
75. Beevers CG, Mullarkey MC, Dainer-Best J, et al. Association between negative cognitive bias and
depression: A symptom-level approach. J Abnorm Psychol. 2019;128(3):212–227. doi:10.1037/
ABN0000405 [PubMed: 30652884]
Author Manuscript

76. Martin DM, McClintock SM, Loo CK. Brief cognitive screening instruments for
electroconvulsive therapy: Which one should I use? 10.1177/0004867420924093. 2020;54(9):867–
873. doi:10.1177/0004867420924093
77. Hermida AP, Goldstein FC, Loring DW, et al. ElectroConvulsive therapy Cognitive Assessment
(ECCA) tool: A new instrument to monitor cognitive function in patients undergoing ECT. J
Affect Disord. 2020;269:36–42. doi:10.1016/J.JAD.2020.03.010 [PubMed: 32217341]
78. Kritzer M, Mischel N, Young J, et al. Ketamine for treatment of mood disorders and suicidality: A
narrative review of recent progress. Ann Clin Psychiatry. 2021;33(4). doi:10.12788/ACP.0048

Harv Rev Psychiatry. Author manuscript; available in PMC 2024 January 01.
Kritzer et al. Page 20

79. Ren L, Deng J, Min S, Peng L, Chen Q. Ketamine in electroconvulsive therapy for depressive
disorder: A systematic review and meta-analysis. J Psychiatr Res. 2018;104:144–156. doi:10.1016/
Author Manuscript

J.JPSYCHIRES.2018.07.003 [PubMed: 30077114]


80. Anderson IM, Blamire A, Branton T, et al. Ketamine augmentation of electroconvulsive
therapy to improve neuropsychological and clinical outcomes in depression (Ketamine-ECT): a
multicentre, double-blind, randomised, parallel-group, superiority trial. The Lancet Psychiatry.
2017. doi:10.1016/S2215-0366(17)30077-9
81. Tielkes CEM, Comijs HC, Verwijk E, Stek ML. The effects of ECT on cognitive functioning in the
elderly: A review. Int J Geriatr Psychiatry. 2008. doi:10.1002/gps.1989
82. Sackeim HA, Prudic J, Fuller R, Keilp J, Lavori PW, Olfson M. The cognitive effects of
electroconvulsive therapy in community settings. Neuropsychopharmacology. 2007. doi:10.1038/
sj.npp.1301180
83. Brakemeier EL, Berman R, Prudic J, Zwillenberg K, Sackeim HA. Self-evaluation of the cognitive
effects of electroconvulsive therapy. J ECT. 2011. doi:10.1097/YCT.0b013e3181d77656
84. Wu L, Zhao H, Weng H, Ma D. Lasting effects of general anesthetics on the brain in the young
and elderly: “mixed picture” of neurotoxicity, neuroprotection and cognitive impairment. J Anesth.
Author Manuscript

2019;33(2):321. doi:10.1007/S00540-019-02623-7 [PubMed: 30859366]


85. Avramov MN, Husain MM, White PF. The comparative effects of methohexital,
propofol, and etomidate for electroconvulsive therapy. Anesth Analg. 1995.
doi:10.1097/00000539-199509000-00031
86. Dunlop BW, Davis PG. Combination treatment with benzodiazepines and SSRIs for comorbid
anxiety and depression: A review. Prim Care Companion J Clin Psychiatry. 2008. doi:10.4088/
pcc.v10n0307
87. Chouinard G The use of benzodiazepines in the treatment of manic-depressive illness. J Clin
Psychiatry. 1988.
88. Pelzer ACM, van der Heijden FMMA, den Boer E. Systematic review of catatonia treatment.
Neuropsychiatr Dis Treat. 2018. doi:10.2147/NDT.S147897
89. Sackeim H, Decina P, Prohovnik I, Malitz S. Seizure Threshold in Electroconvulsive Therapy:
Effects of Sex, Age, Electrode Placement, and Number of Treatments. Arch Gen Psychiatry. 1987.
doi:10.1001/archpsyc.1987.01800160067009
Author Manuscript

90. Sackeim HA, Decina P, Portnoy S, Neeley P, Malitz S. Studies of dosage, seizure threshold, and
seizure duration in ECT. Biol Psychiatry. 1987. doi:10.1016/0006-3223(87)90144-2
91. Duthie AC, Perrin JS, Bennett DM, Currie J, Reid IC. Anticonvulsant Mechanisms of
Electroconvulsive Therapy and Relation to Therapeutic Efficacy. J ECT. 2015. doi:10.1097/
YCT.0000000000000210
92. Bajbouj M, Lang UE, Niehaus L, Hellen FE, Heuser I, Neu P. Effects of right unilateral
electroconvulsive therapy on motor cortical excitability in depressive patients. J Psychiatr Res.
2006. doi:10.1016/j.jpsychires.2005.07.002
93. Luccarelli J, McCoy TH, Seiner SJ, Henry ME. Changes in seizure duration during acute course
electroconvulsive therapy. Brain Stimul. 2021;14(4):941–946. doi:10.1016/J.BRS.2021.05.016
[PubMed: 34119670]
94. Esel E, Kose K, Hacimusalar Y, et al. The effects of electroconvulsive therapy on gabaergic
function in major depressive patients. J ECT. 2008. doi:10.1097/YCT.0b013e31815cbaa1
95. Moulier V, Guehl J, Evêque-Mourroux E, Quesada P, Rothärmel M. A Retrospective Study
of Postictal Suppression during Electroconvulsive Therapy. J Clin Med. 2022;11(5):1440.
Author Manuscript

doi:10.3390/JCM11051440 [PubMed: 35268529]


96. Magloire V, Mercier MS, Kullmann DM, Pavlov I. GABAergic Interneurons
in Seizures: Investigating Causality With Optogenetics. Neuroscientist. 2019;25(4):344–
358. doi:10.1177/1073858418805002/ASSET/IMAGES/10.1177_1073858418805002-IMG1.PNG
[PubMed: 30317911]
97. Ye H, Kaszuba S. Inhibitory or excitatory? Optogenetic interrogation of the functional
roles of GABAergic interneurons in epileptogenesis. J Biomed Sci. 2017;24(1). doi:10.1186/
S12929-017-0399-8/FIGURES/5

Harv Rev Psychiatry. Author manuscript; available in PMC 2024 January 01.
Kritzer et al. Page 21

98. Geretsegger C, Nickel M, Judendorfer B, Rochowanski E, Novak E, Aichhorn W. Propofol


and methohexital as anesthetic agents for electroconvulsive therapy: A randomized, double-blind
Author Manuscript

comparison of electroconvulsive therapy seizure quality, therapeutic efficacy, and cognitive


performance. J ECT. 2007. doi:10.1097/0b013e31814da971
99. Perera TD, Luber B, Nobler MS, Prudic J, Anderson C, Sackeim HA. Seizure expression
during electroconvulsive therapy: Relationships with clinical outcome and cognitive side effects.
Neuropsychopharmacology. 2004. doi:10.1038/sj.npp.1300377
100. Mischel N, Rakesh G, Falcone-Gunderson G, et al. ECT seizure parameter modulation with
bupropion: a pilot study. Brain Stimul. 2019;12(2):500–501. doi:10.1016/j.brs.2018.12.638
101. Krystal AD, Weiner RD, Coffey CE. The ictal EEG as a marker of adequate stimulus
intensity with unilateral ECT. J Neuropsychiatry Clin Neurosci. 1995;7(3):295–303. doi:10.1176/
JNP.7.3.295 [PubMed: 7580187]
102. Hoyer C, Kranaster L, Janke C, Sartorius A. Impact of the anesthetic agents ketamine, etomidate,
thiopental, and propofol on seizure parameters and seizure quality in electroconvulsive therapy:
A retrospective study. Eur Arch Psychiatry Clin Neurosci. 2014;264(3):255–261. doi:10.1007/
s00406-013-0420-5 [PubMed: 23835527]
Author Manuscript

103. Barry JJ, Ettinger AB, Friel P, et al. Consensus statement: The evaluation and treatment of people
with epilepsy and affective disorders. Epilepsy Behav. 2008. doi:10.1016/j.yebeh.2008.04.005
104. Devanand DP, Shapira B, Petty F, et al. Effects of electroconvulsive therapy on plasma GABA.
Convuls Ther. 1995.
105. Sanacora G, Mason GF, Rothman DL, et al. Increased cortical GABA concentrations in depressed
patients receiving ECT. Am J Psychiatry. 2003. doi:10.1176/appi.ajp.160.3.577
106. Palmio J, Huuhka M, Saransaari P, et al. Changes in plasma amino acids after electroconvulsive
therapy of depressed patients. Psychiatry Res. 2005. doi:10.1016/j.psychres.2005.07.010
107. Sackeim HA, Decina P, Prohovnik I, Malitz S, Resor SR. Anticonvulsant and antidepressant
properties of electroconvulsive therapy: A proposed mechanism of action. Biol Psychiatry. 1983.
108. Leaver AM, Espinoza R, Wade B, Narr KL. Parsing the Network Mechanisms
of Electroconvulsive Therapy. Biol Psychiatry. 2022;92(3):193–203. doi:10.1016/
J.BIOPSYCH.2021.11.016 [PubMed: 35120710]
109. Ohman R, Balldin J, Walinder J, Wallin L, Abrahamsson L. PROLACTIN RESPONSE TO
Author Manuscript

ELECTROCONVULSIVE THERAPY. Lancet. 1976. doi:10.1016/S0140-6736(76)90895-3


110. Whalley LJ, Dick H, Watts AG, et al. IMMEDIATE INCREASES IN PLASMA PROLACTIN
AND NEUROPHYSIN BUT NOT OTHER HORMONES AFTER ELECTROCONVULSIVE
THERAPY. Lancet. 1982. doi:10.1016/S0140-6736(82)90004-6
111. Nemeroff CB, Bissette G, Akil H, Fink M. Neuropeptide concentrations in the cerebrospinal
fluid of depressed patients treated with electroconvulsive therapy. Corticotrophin-releasing factor,
β-endorphin and somatostatin. Br J Psychiatry. 1991. doi:10.1192/bjp.158.1.59
112. Kling MA, Geracioti TD, Licinio J, Michelson D, Oldfield EH, Gold PW. Effects of
electroconvulsive therapy on the CRH-ACTH-cortisol system in melancholic depression:
Preliminary findings. In: Psychopharmacology Bulletin.; 1994.
113. Banki CM, Karmacsi L, Bissette G, Nemeroff CB. CSF corticotropin-releasing hormone
and somatostatin in major depression: response to antidepressant treatment and relapse. Eur
Neuropsychopharmacol. 1992. doi:10.1016/0924-977X(92)90019-5
114. Widerlöv E, Ekman R, Jensen L, Borglund L, Nyman K. Arginine vasopressin, but not
corticotropin releasing factor, is a potent stimulator of adrenocorticotropic hormone following
Author Manuscript

electroconvulsive treatment. J Neural Transm. 1989. doi:10.1007/BF01677423


115. Weizman A, Gil-Ad I, Grupper D, Tyano S, Laron Z. The effect of acute and repeated
electroconvulsive treatment on plasma β-endorphin, growth hormone, prolactin and cortisol
secretion in depressed patients. Psychopharmacology (Berl). 1987. doi:10.1007/BF02439598
116. Alexopoulos GS, Inturrisi CE, Lipman R, et al. Plasma Immunoreactive β-Endorphin Levels
in Depression: Effect of Electroconvulsive Therapy. Arch Gen Psychiatry. 1983. doi:10.1001/
archpsyc.1983.01790020079007

Harv Rev Psychiatry. Author manuscript; available in PMC 2024 January 01.
Kritzer et al. Page 22

117. Kronfol Z, Hamdan-Allen G, Goel K, Hill EM. Effects of single and repeated electroconvulsive
therapy sessions on plasma ACTH, prolactin, growth hormone and cortisol concentrations.
Author Manuscript

Psychoneuroendocrinology. 1991. doi:10.1016/0306-4530(91)90020-T


118. Wahlund B, von Rosen D. ECT of major depressed patients in relation to biological and clinical
variables: A brief overview. Neuropsychopharmacology. 2003. doi:10.1038/sj.npp.1300135
119. Schwieler L, Samuelsson M, Frye MA, et al. Electroconvulsive therapy suppresses the
neurotoxic branch of the kynurenine pathway in treatment-resistant depressed patients. J
Neuroinflammation. 2016. doi:10.1186/s12974-016-0517-7
120. Erhardt S, Lim CK, Linderholm KR, et al. Connecting inflammation with glutamate agonism in
suicidality. Neuropsychopharmacology. 2013. doi:10.1038/npp.2012.248
121. BOLWIG TG, HERTZ MM, PAULSON OB, SPOTOFT H, RAFAELSEN OJ. The permeability
of the blood-brain barrier during electrically induced seizures in man. Eur J Clin Invest.
1977;7(2):87–93. doi:10.1111/J.1365-2362.1977.TB01578.X [PubMed: 404164]
122. Cowen PJ, Browning M. What has serotonin to do with depression? World Psychiatry. 2015.
doi:10.1002/wps.20229
123. Lanzenberger R, Baldinger P, Hahn A, et al. Global decrease of serotonin-1A receptor binding
Author Manuscript

after electroconvulsive therapy in major depression measured by PET. Mol Psychiatry. 2013.
doi:10.1038/mp.2012.93
124. Ishihara K, Sasa M. Mechanism underlying the therapeutic effects of electroconvulsive therapy
(ECT) on depression. Jpn J Pharmacol. 1999. doi:10.1254/jjp.80.185
125. Yatham LN, Liddle PF, Lam RW, et al. Effect of electroconvulsive therapy on brain 5-HT2
receptors in major depression. Br J Psychiatry. 2010. doi:10.1192/bjp.bp.109.069567
126. Stain-Malmgren R, Tham A, Åberg-Wistedt A. Increased platelet 5-HT2 receptor binding after
electroconvulsive therapy in depression. J ECT. 1998. doi:10.1097/00124509-199803000-00003
127. Plein H, Berk M. Changes in the platelet intracellular calcium response to serotonin in patients
with major depression treated with electroconvulsive therapy: State or trait marker status. Int Clin
Psychopharmacol. 2000. doi:10.1097/00004850-200015020-00005
128. Ishihara K, Sasa M. Potentiation of 5-HT3 receptor functions in the hippocampal CA1 region
of rats following repeated electroconvulsive shock treatments. Neurosci Lett. 2001. doi:10.1016/
S0304-3940(01)01902-4
Author Manuscript

129. Ousdal OT, Brancati GE, Kessler U, et al. The Neurobiological Effects of Electroconvulsive
Therapy Studied Through Magnetic Resonance: What Have We Learned, and Where Do We
Go? Biol Psychiatry. 2022;91(6):540–549. doi:10.1016/J.BIOPSYCH.2021.05.023 [PubMed:
34274106]
130. Kraus C, Cabrera Abreu C, Lai C-H, et al. Magnetic Resonance Spectroscopy in
Depressed Subjects Treated With Electroconvulsive Therapy-A Systematic Review of Literature.
2021;12:608857. doi:10.3389/fpsyt.2021.608857
131. Knudsen MK, Near J, Blicher AB, Videbech P, Blicher JU. Magnetic resonance (MR)
spectroscopic measurement of γ-aminobutyric acid (GABA) in major depression before and
after electroconvulsive therapy. Acta Neuropsychiatr. 2019. doi:10.1017/neu.2018.22
132. Michael N, Erfurth A, Ohrmann P, Arolt V, Heindel W, Pfleiderer B. Metabolic changes within
the left dorsolateral prefrontal cortex occurring with electroconvulsive therapy in patients with
treatment resistant unipolar depression. Psychol Med. 2003. doi:10.1017/S0033291703007931
133. Michael N, Erfurth A, Ohrmann P, Arolt V, Heindel W, Pfleiderer B. Neurotrophic effects
of electroconvulsive therapy: A proton magnetic resonance study of the left amygdalar region
Author Manuscript

in patients with treatment-resistant depression. Neuropsychopharmacology. 2003. doi:10.1038/


sj.npp.1300085
134. Pfleiderer B, Michael N, Erfurth A, et al. Effective electroconvulsive therapy reverses glutamate/
glutamine deficit in the left anterior cingulum of unipolar depressed patients. Psychiatry Res -
Neuroimaging. 2003. doi:10.1016/S0925-4927(03)00003-9
135. Njau S, Joshi SH, Espinoza R, et al. Neurochemical correlates of rapid treatment response
to electroconvulsive therapy in patients with major depression. J Psychiatry Neurosci. 2017.
doi:10.1503/jpn.150177

Harv Rev Psychiatry. Author manuscript; available in PMC 2024 January 01.
Kritzer et al. Page 23

136. Merkl A, Schubert F, Quante A, et al. Abnormal cingulate and prefrontal cortical neurochemistry
in major depression after electroconvulsive therapy. Biol Psychiatry. 2011. doi:10.1016/
Author Manuscript

j.biopsych.2010.08.009
137. Ousdal OT, Brancati GE, Kessler U, et al. The Neurobiological Effects of Electroconvulsive
Therapy Studied Through Magnetic Resonance: What Have We Learned, and Where Do We Go?
Biol Psychiatry. 2022;91(6):540–549. doi:10.1016/j.biopsych.2021.05.023 [PubMed: 34274106]
138. Ousdal OT, Argyelan M, Narr KL, et al. Brain Changes Induced by Electroconvulsive
Therapy Are Broadly Distributed. Biol Psychiatry. 2020;87(5):451–461. doi:10.1016/
j.biopsych.2019.07.010 [PubMed: 31561859]
139. Price JL, Drevets WC. Neurocircuitry of Mood Disorders. 2010. doi:10.1038/npp.2009.104
140. Oltedal L, Bartsch H, Sørhaug OJE, et al. The Global ECT-MRI Research Collaboration
(GEMRIC): Establishing a multi-site investigation of the neural mechanisms underlying response
to electroconvulsive therapy. Neuroimage (Amst). 2017;14:422. doi:10.1016/J.NICL.2017.02.009
141. Malykhin N V, Carter R, Seres P, Coupland NJ. Structural changes in the hippocampus in
major depressive disorder: Contributions of disease and treatment. J Psychiatry Neurosci. 2010.
doi:10.1503/jpn.100002
Author Manuscript

142. Stockmeier CA, Mahajan GJ, Konick LC, et al. Cellular changes in the postmortem hippocampus
in major depression. Biol Psychiatry. 2004. doi:10.1016/j.biopsych.2004.08.022
143. Oltedal L, Narr KL, Abbott C, et al. Volume of the Human Hippocampus and Clinical Response
Following Electroconvulsive Therapy. Biol Psychiatry. 2018. doi:10.1016/j.biopsych.2018.05.017
144. Dols A, Loef D, Eijndhoven P van, et al. Upregulation of Neuroplasticity Marker
Doublecortin in the Human Post-Mortem Brain After Electroconvulsive Therapy. Biol Psychiatry.
2022;91(9):S56. doi:10.1016/J.BIOPSYCH.2022.02.159
145. Sorrells SF, Paredes MF, Cebrian-Silla A, et al. Human hippocampal neurogenesis drops sharply
in children to undetectable levels in adults. Nature. 2018. doi:10.1038/nature25975
146. Lima SMA, Gomes-Leal W. Neurogenesis in the hippocampus of adult humans: Controversy
“fixed” at last. Neural Regen Res. 2019. doi:10.4103/1673-5374.259616
147. Ito M, Seki T, Liu J, et al. Effects of repeated electroconvulsive seizure on cell proliferation in the
rat hippocampus. Synapse. 2010. doi:10.1002/syn.20796
148. Zhao C, Warner-Schmidt J, Duman RS, Gage FH. Electroconvulsive seizure promotes spine
Author Manuscript

maturation in newborn dentate granule cells in adult rat. Dev Neurobiol. 2012;72(6):937–942.
doi:10.1002/dneu.20986 [PubMed: 21976455]
149. Scott BW, Wojtowicz JM, Burnham WM. Neurogenesis in the Dentate Gyrus of the Rat
Following Electroconvulsive Shock Seizures. Exp Neurol. 2000;165(2):231–236. doi:10.1006/
exnr.2000.7458 [PubMed: 10993683]
150. Smitha JSM, Roopa R, Sagar BC, Kutty BM, Andrade C. Images in electroconvulsive
therapy: ECS dose-dependently increases cell proliferation in the subgranular region of the
rat hippocampus. J ECT. 2014;30(3):193–194. doi:10.1097/YCT.0000000000000076 [PubMed:
24901429]
151. Schloesser RJ, Orvoen S, Jimenez DV., et al. Antidepressant-like Effects of Electroconvulsive
Seizures Require Adult Neurogenesis in a Neuroendocrine Model of Depression. Brain Stimul.
2015. doi:10.1016/j.brs.2015.05.011
152. Martinowich K, Manji H, Lu B. New insights into BDNF function in depression and anxiety. Nat
Neurosci. 2007. doi:10.1038/nn1971
153. Berton O, Mcclung CA, DiLeone RJ, et al. Essential role of BDNF in the mesolimbic dopamine
Author Manuscript

pathway in social defeat stress. Science (80- ). 2006;311(February):864–868. doi:10.1126/


science.1120972
154. Duman RS, Monteggia LM. A Neurotrophic Model for Stress-Related Mood Disorders. Biol
Psychiatry. 2006. doi:10.1016/j.biopsych.2006.02.013
155. Karege F, Perret G, Bondolfi G, Schwald M, Bertschy G, Aubry JM. Decreased serum
brain-derived neurotrophic factor levels in major depressed patients. Psychiatry Res. 2002.
doi:10.1016/S0165-1781(02)00005-7

Harv Rev Psychiatry. Author manuscript; available in PMC 2024 January 01.
Kritzer et al. Page 24

156. Haile CN, Murrough JW, Iosifescu DV., et al. Plasma brain derived neurotrophic factor (BDNF)
and response to ketamine in treatment-resistant depression. Int J Neuropsychopharmacol. 2014.
Author Manuscript

doi:10.1017/S1461145713001119
157. Piccinni A, Del Debbio A, Medda P, et al. Plasma Brain-Derived Neurotrophic
Factor in treatment-resistant depressed patients receiving electroconvulsive therapy. Eur
Neuropsychopharmacol. 2009. doi:10.1016/j.euroneuro.2009.01.002
158. Brunoni AR, Lopes M, Fregni F. A systematic review and meta-analysis of clinical studies on
major depression and BDNF levels: Implications for the role of neuroplasticity in depression. Int
J Neuropsychopharmacol. 2008. doi:10.1017/S1461145708009309
159. Wennström M, Hellsten J, Ekdahl CT, Tingström A. Electroconvulsive seizures induce
proliferation of NG2-expressing glial cells in adult rat hippocampus. Biol Psychiatry. 2003.
doi:10.1016/S0006-3223(03)00693-0
160. Wennström M, Hellsten J, Ekstrand J, Lindgren H, Tingström A. Corticosterone-induced
inhibition of gliogenesis in rat hippocampus is counteracted by electroconvulsive seizures. Biol
Psychiatry. 2006;59(2):178–186. doi:10.1016/j.biopsych.2005.08.032 [PubMed: 16431219]
161. Wennström M, Hellsten J, Tingström A. Electroconvulsive seizures induce proliferation of NG2-
Author Manuscript

expressing glial cells in adult rat amygdala. Biol Psychiatry. 2004;55(5):464–471. doi:10.1016/
j.biopsych.2003.11.011 [PubMed: 15023573]
162. Öngür D, Pohlman J, Dow AL, et al. Electroconvulsive Seizures Stimulate Glial Proliferation
and Reduce Expression of Sprouty2 within the Prefrontal Cortex of Rats. Biol Psychiatry. 2007.
doi:10.1016/j.biopsych.2006.11.014
163. Madsen TM, Yeh DD, Valentine GW, Duman RS. Electroconvulsive seizure treatment
increases cell proliferation in rat frontal cortex. Neuropsychopharmacology. 2005. doi:10.1038/
sj.npp.1300565
164. Jansson L, Hellsten J, Tingström A. Region Specific Hypothalamic Neuronal Activation and
Endothelial Cell Proliferation in Response to Electroconvulsive Seizures. Biol Psychiatry. 2006.
doi:10.1016/j.biopsych.2005.11.019
165. Hellsten J, Wennström M, Bengzon J, Mohapel P, Tingström A. Electroconvulsive seizures induce
endothelial cell proliferation in adult rat hippocampus. Biol Psychiatry. 2004. doi:10.1016/
j.biopsych.2003.08.013
166. Hellsten J, West MJ, Arvidsson A, et al. Electroconvulsive seizures induce angiogenesis in adult
Author Manuscript

rat hippocampus. Biol Psychiatry. 2005. doi:10.1016/j.biopsych.2005.05.023


167. Helmut K, Hanisch UK, Noda M, Verkhratsky A. Physiology of microglia. Physiol Rev. 2011.
doi:10.1152/physrev.00011.2010
168. Van Buel EM, Sigrist H, Seifritz E, et al. Mouse repeated electroconvulsive seizure (ECS) does
not reverse social stress effects but does induce behavioral and hippocampal changes relevant
to electroconvulsive therapy (ECT) side-effects in the treatment of depression. PLoS One. 2017.
doi:10.1371/journal.pone.0184603
169. Sepulveda-Rodriguez A, Li P, Khan T, et al. Electroconvulsive Shock Enhances Responsive
Motility and Purinergic Currents in Microglia in the Mouse Hippocampus. eNeuro. 2019.
doi:10.1523/ENEURO.0056-19.2019
170. Ferrari AJ, Charlson FJ, Norman RE, et al. Burden of Depressive Disorders by Country,
Sex, Age, and Year: Findings from the Global Burden of Disease Study 2010. PLoS Med.
2013;10(11):e1001547. doi:10.1371/journal.pmed.1001547 [PubMed: 24223526]
171. O’Neil MF, Moore NA. Animal models of depression: Are there any? Hum Psychopharmacol.
Author Manuscript

2003. doi:10.1002/hup.496
172. Arnone D, Mckie S, Elliott R, et al. State-dependent changes in hippocampal grey matter in
depression. Mol Psychiatry. 2013. doi:10.1038/mp.2012.150
173. Ming Q, Zhong X, Zhang X, et al. State-independent and dependent neural responses to
psychosocial stress in current and remitted depression. Am J Psychiatry. 2017. doi:10.1176/
appi.ajp.2017.16080974
174. Tik M, Hoffmann A, Sladky R, et al. Towards understanding rTMS mechanism of action:
Stimulation of the DLPFC causes network-specific increase in functional connectivity.
Neuroimage. 2017. doi:10.1016/j.neuroimage.2017.09.022

Harv Rev Psychiatry. Author manuscript; available in PMC 2024 January 01.
Kritzer et al. Page 25

175. Van Diermen L, Van Den Ameele S, Kamperman AM, et al. Prediction of electroconvulsive
therapy response and remission in major depression: Meta-analysis. Br J Psychiatry. 2018.
Author Manuscript

doi:10.1192/bjp.2017.28
176. Jackson SJ, Andrews N, Ball D, et al. Does age matter? The impact of rodent age on study
outcomes. Lab Anim. 2017. doi:10.1177/0023677216653984
177. Ohmura Y, Kuniyoshi Y. A translational model to determine rodent’s age from human foetal age.
Sci Rep. 2017. doi:10.1038/s41598-017-17571-z
178. Westoby CJ, Mallen CD, Thomas E. Cognitive complaints in a general population of older adults:
Prevalence, association with pain and the influence of concurrent affective disorders. Eur J Pain.
2009. doi:10.1016/j.ejpain.2008.11.011
179. Murman DL. The Impact of Age on Cognition. Semin Hear. 2015. doi:10.1055/s-0035-1555115
180. Porta-Casteràs D, Cano M, Camprodon JA, et al. A multimetric systematic review of fMRI
findings in patients with MDD receiving ECT. Prog Neuropsychopharmacol Biol Psychiatry.
2021;108. doi:10.1016/J.PNPBP.2020.110178
181. Wang J, Wei Q, Wang L, et al. Functional reorganization of intra- and internetwork connectivity
in major depressive disorder after electroconvulsive therapy. Hum Brain Mapp. 2018;39(3):1403–
Author Manuscript

1411. doi:10.1002/hbm.23928 [PubMed: 29266749]


182. Van Waarde JA, Scholte HS, Van Oudheusden LJB, Verwey B, Denys D, Van Wingen GA.
A functional MRI marker may predict the outcome of electroconvulsive therapy in severe and
treatment-resistant depression. Mol Psychiatry. 2015. doi:10.1038/mp.2014.78
183. Moreno-Ortega M, Prudic J, Rowny S, et al. Resting state functional connectivity predictors of
treatment response to electroconvulsive therapy in depression. Sci Reports 2019 91. 2019;9(1):1–
19. doi:10.1038/s41598-019-41175-4
Author Manuscript
Author Manuscript

Harv Rev Psychiatry. Author manuscript; available in PMC 2024 January 01.

You might also like