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

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

×
Reviews and OverviewsFull Access

Whither TMS: A One-Trick Pony or the Beginning of a Neuroscientific Revolution?

Abstract

Psychiatry has been at the forefront of advancing clinical transcranial magnetic stimulation (TMS) since the mid-1990s, shortly after the invention of modern TMS in 1985 by Barker. Clinical TMS for psychiatric applications is advancing rapidly, with novel methods and innovations for treating depression, as well as a new clinical indication in obsessive-compulsive disorder. This review summarizes the recent findings and peers into the near future of this fertile and rapidly changing field. It is possible that many, perhaps even most, psychiatrists will be incorporating some form of brain stimulation into their practice within the next decade. The author summarizes the reasons for this optimistic view.

This issue of the Journal contains two important articles that report on applications of transcranial magnetic stimulation (TMS), a relatively new technology for psychiatrists. At this stage, one might well wonder: Are these articles the hoof beats of a stampede of new indications using TMS? Will every psychiatrist soon be a brain stimulator of sorts, like most psychiatrists are now trained in psychopharmacology and understand the principles of talking therapies? Or is all this brain stimulation much ado about nothing, and will TMS go the way of insulin therapy for depression (1, 2) or renal dialysis for schizophrenia (3)? I offer here an overview of the issue’s theme with the aim of providing a fundamental understanding of the concepts, methods, and future directions of TMS. The question of where TMS is headed may not yet be fully answerable, but in my view, as I will show, there are ample grounds for optimism about its potential.

Background

Short History

Psychiatry had an early love-hate relationship with TMS soon after Anthony T. Barker developed it in its modern form in 1985 (4). Some, myself included, were eager to use TMS and perfect it as a treatment (57). Other psychiatrists were quite skeptical and resistant (8, 9). TMS involves passing a large but brief electrical current through insulated wires resting on the scalp. The powerful electrical current creates a magnetic field, which penetrates the scalp and skull and then induces electrical currents in the superficial areas of the brain (10). In 1985, Barker was the first to succeed in making a coil and capacitor powerful enough to reach into the spinal cord (his original target) and also the cortex of the brain. In a generous move, Barker then built several machines and helped place them in leading laboratories around the world. One of those laboratories was the National Hospital for Neurology and Neurosurgery, Queen Square, London, where I spent 1989 learning about the entirely new world of functional brain imaging using positron emission tomography (PET), single-photon emission computed tomography (SPECT), and MRI scanning in psychiatry. Luckily, while I was there, I first saw one of Barker’s TMS machines, only 4 years after they were invented.

The early 1990s were important in psychiatry, as we could now finally image our organ of study with the new tools of CT and then MRI scans. Fluorodeoxyglucose (FDG) PET scans were starting to tell us about regional brain activity and behavior in health and disease. The new imaging tools and the imaging revolution were developing initial maps of pathological regions in the brain for psychiatric diseases, much like neurology had developed for Parkinson’s disease (11). Thus the environment could not have been better for a noninvasive stimulating tool like TMS: we had maps of where to stimulate and a new technology that allowed us to explore and test. Many psychiatrists across the globe, particularly those working in imaging, like myself, who were developing these maps, could see the potential for a relatively noninvasive tool like TMS with the ability to stimulate the cortex of an awake, alert person (7, 1217). The optimists among us eagerly sought to stimulate cortical regions in patients and see if we could change disease progression. But how to start? There were and still are a dizzying array of complex questions—where to place the coil, how much electricity to deliver, over how long an interval, in what pattern, and so on. In the 25 years since the first TMS clinical trials in depression (5, 7, 13, 14, 18, 19), we have made remarkable progress in understanding how each of these variables changes what TMS does in the brain. Work in this area is harder than it should be because of certain properties of physics and technological limitations that make it hard to perform preclinical TMS studies. In order to make a TMS coil small enough to proportionately stimulate a mouse or rat brain compared with a human’s, one has to deliver massively larger amounts of electricity through smaller coils—and they explode (2022). Translational neurobiology is not always easy and straightforward!

Why Depression?

Repetitive TMS (rTMS)—a repetitive train of TMS pulses—has had the biggest success as a therapy in treating treatment-resistant depression. Many have wondered why we moved rTMS into the clinic for depression, and not for some other neuropsychiatric illness for which we have a better understanding of the circuits involved, such as stroke recovery, Parkinson’s disease, or tinnitus (23). One reason was that ECT had provided clear evidence that regional electromagnetic stimulation could treat the illness (24, 25). Additionally, beginning in the early 1990s, there was an emerging consensus about key cortical and subcortical regions involved in depression, some of which could be directly stimulated with TMS (19, 26, 27). Activity in some of the regions, moreover, correlated with improvements in symptoms following sleep deprivation (cingulate) (28) or ECT (prefrontal cortex) (24, 29). Ironically, some ECT practitioners and researchers were among the most ardent opponents of TMS. Their incorrect logical concern was that TMS was not causing seizures (true) and that seizures were necessary for the antidepressant effects of ECT (also true), so a non-seizure-producing intervention device could not work (false) (30). (We did not then have the exquisite tools of today, which allow us to show changes in regional functional connectivity that mediate the clinical effects of rTMS [31].) Luckily, depression turned out to be a superb initial choice.

Methodically uncovering the details required for clinical use, the community of TMS researchers made initial educated guesses about many issues (coil location, intensity, frequency, pulse width, train length, total number of pulses in a day, dosing schedule, and the number of pulses in a treatment course). We were likely both lucky and relatively clever, and the initial choices proved clinically effective (7, 15, 16). Notably, it took over a decade of work refining these choices in incremental small trials before we were “ready” to launch the first pivotal studies (32, 33). A TMS industry was born, and this initially led to approval from the U.S. Food and Drug Administration (FDA) in 2008 (32, 33) and, not long afterwards, widespread insurance coverage for rTMS to treat acute major depressive episodes.

Depending on your perspective, psychiatrists’ uptake of clinical rTMS has been impressive, or disappointing. Historically, psychiatrists as a group may self-select away from specialties in which procedures are performed. rTMS involves a hands-on procedure by psychiatrists, who hire staff and purchase equipment, or send their patients to colleagues who have developed this expertise (34). The rTMS industry was neither large nor initially well capitalized, so there were no national advertising campaigns until recently. Despite these obstacles, there are now at least seven machines with FDA clearance, and TMS is available clinically across the globe. TMS is now daily producing remissions from depression and saving lives.

TMS thus represents a paradigm shift in psychiatry. It is not a talking therapy, does not involve administration of medications by mouth or intravenously, does not involve seizures, and modulates circuit activity in the brain. Because it is focal and noninvasive, it produces no systemic side effects and no drug-drug interactions. It thus is a good choice in medically complicated patients, and it does not involve anesthesia or have deleterious cognitive effects.

Viewed differently, clinical use of rTMS has been disappointing. Over the past 10 years, rates of suicide and depression have increased. TMS has not had a large public health impact on these. Moreover, the FDA-approved treatment requires daily treatments for 6 weeks, and each treatment takes about 30 minutes to an hour. It is highly inefficient, and thus relatively expensive. Maybe TMS is just a one-trick pony?

What is Happening Now With TMS?

Currently several lines of research with TMS are most promising.

Treatment of Depression

In the area of treating depression, researchers are carefully and appropriately reexamining all the initial choices, and finding that even some simple modifications can improve TMS as an antidepressant treatment.

Whom to treat?

The initial studies involved only patients with treatment-resistant depression, and TMS (like all of our treatments) works less well in patients whose illness is more treatment resistant. Studies are now enrolling patients with less treatment-resistant illness. For reasons of both safety and scientific integrity, the early trials enrolled only patients who were weaned off of their antidepressant medications (32). Now, most patients are treated safely and with good efficacy while staying on antidepressant medications (35, 36).

Where to stimulate?

This is an exciting area of research. I initially proposed the “5 cm rule,” in which the TMS coil was placed 5 cm anterior to the location found to induce a thumb twitch (7, 16, 17). Unfortunately, in perhaps one-third of patients, this does not reach the prefrontal cortex (3739), and most clinicians now place the coil by using an EEG grid system that accounts for differences in head size (40). The search is on for the best cortical location, either at a group level or individually guided. In a fascinating line of research, it may be that the clinical interview actually corresponds with the “proper” TMS coil location (41, 42). Fox and colleagues (43) have performed meta-analyses of many TMS trials with differences in the method of coil placement and a range of clinical effects. Merging this information with data from the human connectome, they found that certain coil locations do better for certain symptoms. More anxious and neurotic patients tend to do better with the 5 cm location, while anhedonic and dysphoric symptoms tend to respond better with a more anterior and medial location (44). What an exciting development this might be for psychiatry if the data hold true in prospective trials! A good clinical examination might be able to parse the depressions into different disease subtypes with differential circuit activity requiring a different coil location. We may now be able to “carve nature at its joints” (as Plato put it in the Phaedrus dialogue), dissecting different depressions using clinical examinations and TMS response.

Conventional TMS can only stimulate the surface of the brain, but a new series of coils can stimulate deeper into the brain, and across broader areas (4548). These H-coils are now approved for the treatment of depression (and obsessive-compulsive disorder [OCD], as discussed below) (49). It is unclear whether they are more effective at treating depression than the other coils. The H-coil manufacturer and others also have developed multiple coils that can be used jointly or independently (50). We now can stimulate multiple regions of the brain with different patterns, exciting some regions and inhibiting others. Remarkable technologies are already here with TMS. What is lagging behind is the translational clinical neuroscience informing us on how best to use these tools.

How to stimulate, in terms of patterns and frequencies?

One of the most remarkable aspects of TMS is that the brain effects are frequency dependent. That is, slow, low-frequency rTMS over time can temporarily inhibit regional brain activity. In contrast, faster, high-frequency patterns tend to be excitatory. This has enabled various studies to use inhibitory patterns to block or “turn down” a region or excitatory patterns to boost a region. A fascinating new development is theta-burst stimulation (TBS), which was long known to basic neuroscientists but only recently rediscovered by the TMS research community (51). Theta burst is a pattern that is intrinsic to the brain, and it is what you might hear if you could listen in on your hippocampal neurons talking to each other (triplets at 50 Hz, which are then repeated at a frequency of 5 Hz, hence the “theta”). Theta bursting is an electrochemical signaling language of the brain. Fascinatingly, theta-burst patterns can also produce diametrically opposite effects solely on the basis of how the pattern is delivered. Intermittent theta burst (iTBS) is excitatory, while continuous theta burst (cTBS) temporarily inhibits brain signaling. Theta-burst TMS is also much more efficient at producing brain changes than is conventional rTMS at 5 or 10 Hz (52). An important study published last year (53) showed that iTBS for 6 minutes was as effective in treating depression as the standard FDA-approved treatment, which takes 30 minutes. One can obviously treat many more patients in a clinic if the time is reduced fivefold, so this may be an important step forward in improving the efficiency of TMS.

What dose to apply?

We have not yet established an upper limit for TMS dose in terms of safety. No one has ever done a comprehensive escalating dosing study with TMS like that required for medications (20, 54, 55). All medications, before they are used in clinical trials, are given to animals first, and then to healthy human volunteers, in ever-larger amounts to determine the doses at which side effects emerge and safety concerns arise. Over the past 25 years, with mounting assurances about TMS safety, there has been a gradual increase in the number of TMS pulses given in a day, or a week, or a treatment course. However, some recent studies have shown that more pulses alone may not be better (56). As in good dancing, the rhythm may be the key. Can one give more treatments in a day and create a more rapid response to TMS? The jury is still out on this, with some case series producing rapid responses (57) and controlled trials using similar approaches and not getting better effects (58). It appears that the initially determined daily treatment pattern is not sacred, and one can deliver TMS treatment sessions in a more creative and flexible pattern than was done in the pivotal clinical trials.

What is the brain doing during treatment?

Another exciting area of research involves manipulating what the brain is doing while TMS is being delivered. This research marries the rich tradition in psychiatry of talk and behavioral therapies with the new technology of brain stimulation. In 1949, Donald Hebb (59) theorized that neurons that fire together, wire together (as paraphrased by others later). Applied to TMS, it suggests that the activity, behavior, or state of the person being treated may matter in terms of whether TMS can induce long-term synaptic changes. Thus, researchers are manipulating brain activity during TMS. In many applications, what the patient is doing during stimulation appears to be important, if not critical (6063). To date, in relation to treating depression, no one has shown that a consistent manipulation of state during TMS treatment produces better outcomes. However, this may be because almost all depressed patients, during a treatment, are likely obsessing about their depression and activating these mood-regulating circuits naturally. That is, during TMS treatments, many patients are likely engaging in “activation” of their internal thoughts and beliefs that are dysphoric or sad. If TMS works through the principles of synaptic plasticity and LTD/LTP (long-term depression/long-term potentiation), it should be possible to add certain medications and boost these effects. There is no convincing evidence to date, however, that any medication enhances or blocks the antidepressant effects of TMS. Some have argued that benzodiazepines may block the antidepressant effect of TMS, but there is a notable confounder in this regard: patients taking benzodiazepines are anxious, and comorbid anxiety is itself a negative response predictor for antidepressant treatment response to TMS and other interventions (64). Interestingly, pretreatment with naloxone blocks the analgesic effects of TMS, suggesting that prefrontal rTMS releases endogenous opioids (65, 66). It is not clear whether this effect is related to its antidepressant actions.

In summary, TMS continues to rapidly evolve as an antidepressant treatment, with research into the translational neurobiological effects and reexamination of the choices of coils, settings, timing, and concomitant treatments to improve its efficiency.

New Neuropsychiatric Indications

Following in the wake of rTMS’s success as an antidepressant, there has been an explosion of research using rTMS as a potential therapy in other neuropsychiatric disorders. For any disease for which there is known regional anatomical dysfunction, rTMS could potentially treat the disorder. A quick search through ClinicalTrials.gov this month shows 1,425 completed and ongoing TMS trials in almost all brain diseases. The listings even include clinical trials in brain diseases without a known specific regional dysfunction, such as the schizophrenia spectrum disorders and autism spectrum disorder. In these diseases, it is common for researchers to focus on a symptom within the overall disorder—say, auditory hallucinations in schizophrenia—and then apply TMS to the regions involved in that one symptom.

Exploring the use of TMS in other indications is starting to bear fruit. TMS is now FDA cleared for OCD, largely on the basis of the clinical trial published in this issue of the Journal (67). OCD is notoriously difficult to treat, although exposure therapy and the selective serotonin reuptake inhibitors are partly effective. In the Carmi et al. article in this issue (67), the researchers took a page from Hebb’s ideas and performed a 3- to 5-minute individually prepared exposure therapy immediately before TMS: The clinicians made sure that these patients with treatment-resistant OCD were definitely obsessing during the treatment. They then stimulated the medial prefrontal cortex and cingulate gyrus at high frequency, with a deep and broad coil. An earlier pilot study suggested that only high-frequency stimulation worked at this target (68). Other OCD studies have examined the premotor cortex, or the supplemental motor area, reasoning that OCD is related to abnormal motor patterns (69). The important double-blind results reported in this issue showed improvements that were statistically significant and clinically meaningful in a group with treatment-resistant illness. This FDA-approved approach requires a collaboration between providers who understand OCD exposure therapy and those who know how to deliver TMS and manage medications. It is not TMS or exposure therapy or medication management, but rather a weaving together of all these therapeutic threads. As with the development of TMS for depression, researchers are now examining response predictors, durability of response, and other important clinical questions and outcomes of TMS for OCD.

While the study in this issue by Philip et al. on TMS in the treatment of PTSD (70) has not opened up a new FDA-approved indication, it is an important study along that path. Here, the researchers used the new frequency, theta burst, intermittently (iTBS), thus attempting to excite the right prefrontal cortex. They did not formally manipulate PTSD symptoms, although others have shown that this is possible (71). With a relatively small sample of 50 patients, they found improvements in active and not sham treatment over just 2 weeks, which is a short time for treating PTSD. Larger effects emerged when the treatment continued another 2 weeks, although this second phase was not blinded. Again, larger clinical trials are needed before this approach can be translated into another FDA-approved treatment.

Clearly, rTMS is thus not a one-trick pony, and good clinical rTMS research is under way in many other brain disorders. Depending on the outcomes of these large trials, our field may soon be using rTMS to treat pain, substance use disorders, anxiety disorders, autism spectrum disorder, and some symptoms of schizophrenia. Thus, any brain disease with a known regional anatomical dysfunction might be treatable with skilled application of rTMS, combined in many instances with behavioral challenges and adjunctive medications that help with disease symptoms or actually promote synaptic plasticity and therapeutic change. The future is indeed bright for rTMS, and this treatment tool will only get better as we develop our understanding of the translational effects, identify keys to inducing plasticity, and decipher genetic modifiers.

The Likely and Exciting Next Few Years

From my vantage point, rTMS is the first wave of an entire new approach to treatment in neuropsychiatry. The rich and fertile field of brain stimulation is rapidly changing and moving forward with many other technologies and overlaps both psychiatry and bioengineering (72). Technology is advancing rapidly, particularly body sensors and the ability to interact through wearable devices. TMS technology itself continues to evolve, as we have seen with the development of new broad and deep TMS coils. However, other competing technologies may develop into clinical applications that are as important as TMS, or even eventually replace TMS. Two candidate therapies are transcranial direct current stimulation (tDCS) (which is less expensive than TMS and likely works through different mechanisms) and transcranial pulsed ultrasound (low-intensity focused ultrasound pulsation).

tDCS involves passing relatively weak direct current through the brain for about 20 minutes per session. It is inexpensive and relatively safe (73). While TMS might accidentally cause a seizure, and is powerful enough to induce a thumb twitch, tDCS cannot cause a seizure and is weak. Thus, most researchers always manipulate behavior while applying tDCS. So far, the clinical outcomes with tDCS have not been impressive and have not resulted in FDA clearance. Perhaps the most significant clinical study to date was a noninferiority trial of tDCS combined with speech training in patients with aphasia following a stroke (74, 75). Interestingly, a brain-derived neurotrophic factor (BDNF) neuroplasticity gene has been found to be correlated with treatment response (76). As in the early days with TMS, it is still not clear how to dose tDCS, and we currently lack the ability to easily individualize the tDCS dose for each patient and make sure that we are delivering the appropriate amount to interact with their brain.

The holy grail of brain stimulation treatment is a tool that is cheap, portable, and painless, can be applied in awake, alert humans, and can penetrate deep in the brain and focally modulate a specific region and only that region. Low-intensity focused ultrasound pulsation (LIFUP) may be the next big thing in clinical brain stimulation, as it may have many or all of these attributes (77). For reasons that are unclear, neurons will fire if they receive pulsed ultrasound signals (not the constant ultrasound signal used in ultrasound imaging). This is not due to heating or damage. Clever researchers have figured out ways to deliver ultrasound through the intact skull, focus it deep in the brain, and not cause tissue damage as with the ablative ultrasound used in neurosurgery (78, 79). Currently, LIFUP must be performed in an MRI scanner, but it will likely be able to be moved into the clinic, with image guidance systems. It may or may not be necessary to stimulate focally and deep. There may be intrinsic homeostatic and circuit-regulating behaviors that are best suited to modification by cortical stimulation, as with TMS. But it will be important to have the tools to figure out which brain diseases are best treated with our various approaches—broad and cortical, as with TMS, versus focal and deep, as with ultrasound.

It is clear that we are well on the way to understanding the recipes of brain activation during stimulation, behavior, dosing and timing, and local pharmacology, and to being able to sculpt and change regional brain activity. Keep watching the brain stimulation revolution as it unfolds. We are just at the beginning, with TMS as the vanguard. Work in this area will advance our field and the treatment of our patients. TMS is not a one-trick pony, and the best is yet to come.

Department of Psychiatry and Behavioral Sciences and Institute of Psychiatry, Medical University of South Carolina, Charleston; and Ralph H. Johnson VA Medical Center, Charleston.
Send correspondence to Dr. George ().

Dr. George has served as a consultant for PureTech Ventures; he has served as an unpaid consultant and received research support and/or has received donated equipment and/or served on data safety monitoring boards for Brainsway, Magstim, Mecta, Microtransponder, Neuronetics, and NeoSync; and he serves as journal editor for Brain Stimulation (Elsevier Press).

Dr. George is funded by the Layton McCurdy Endowed Chair Fund, NIH grant P20 GM109040 (“South Carolina Research Center for Recovery from Stroke”), National Institute of Child Health and Human Development grant P2CHD086844 (National Center of Neuromodulation for Rehabilitation), the Tiny Blue Dot Foundation, BRASH, and the Translational Research Institute for Space Health.

Dr. George thanks Dr. Tony Barker for inventing modern TMS, Dr. John Rothwell for first showing him a coil, Dr. Robert Post at NIH, who first allowed him to do TMS research, and Drs. Mark Hallett and Eric Wasserman at NIH for allowing his work in their TMS lab.

References

1 Neduva AA, Dorokhov VV, Sulukhiia SV: [Forced insulin coma therapy and its efficacy]. Zh Nevropatol Psikhiatr Im S S Korsakova 1991; 91:75–77 (Russian)MedlineGoogle Scholar

2 Cramond WA: Lessons from the insulin story in psychiatry. Aust N Z J Psychiatry 1987; 21:320–326MedlineGoogle Scholar

3 Carpenter WT Jr, Sadler JH, Light PD, et al.: The therapeutic efficacy of hemodialysis in schizophrenia. N Engl J Med 1983; 308:669–675MedlineGoogle Scholar

4 Barker AT, Jalinous R, Freeston IL: Non-invasive magnetic stimulation of human motor cortex. Lancet 1985; 1:1106–1107Crossref, MedlineGoogle Scholar

5 George MS, Wassermann EM: Rapid-rate transcranial magnetic stimulation and ECT. Convuls Ther 1994; 10:251–254, discussion 255–258MedlineGoogle Scholar

6 George MS: An introduction to the emerging neuroanatomy of depression. Psychiatr Ann 1994; 24:635–636Google Scholar

7 George MS, Wassermann EM, Williams WA, et al.: Daily repetitive transcranial magnetic stimulation (rTMS) improves mood in depression. Neuroreport 1995; 6:1853–1856Crossref, MedlineGoogle Scholar

8 Kellner CH, Husain M, Petrides G, et al.: Comment on “Repetitive transcranial magnetic stimulation versus electroconvulsive therapy for major depression: preliminary results of a randomized trial” (letter). Biol Psychiatry 2002; 52:1032–1033MedlineGoogle Scholar

9 Fink M: The mode of action of ECT. Psychopharmacol Bull 1994; 30:309–312MedlineGoogle Scholar

10 Higgins ES, George MS: Brain Stimulation Therapies for Clinicians, 2nd ed. Washington, DC, American Psychiatric Association Publishing, 2020Google Scholar

11 Alexander GE, DeLong MR, Strick PL: Parallel organization of functionally segregated circuits linking basal ganglia and cortex. Annu Rev Neurosci 1986; 9:357–381Crossref, MedlineGoogle Scholar

12 Belmaker RH, Fleischmann A: Transcranial magnetic stimulation: a potential new frontier in psychiatry. Biol Psychiatry 1995; 38:419–421Crossref, MedlineGoogle Scholar

13 Grisaru N, Yarovslavsky U, Abarbanel J, et al.: Transcranial magnetic stimulation in depression and schizophrenia. Eur Neuropsychopharmacol 1994; 4:287–288Crossref, MedlineGoogle Scholar

14 Kolbinger HM, Hoflich G, Hufnagel A, et al.: Transcranial magnetic stimulation (TMS) in the treatment of major depression: a pilot study. Hum Psychopharmacol 1995; 10:305–310CrossrefGoogle Scholar

15 George MS, Wassermann EM, Kimbrell TA, et al.: Mood improvement following daily left prefrontal repetitive transcranial magnetic stimulation in patients with depression: a placebo-controlled crossover trial. Am J Psychiatry 1997; 154:1752–1756LinkGoogle Scholar

16 George MS, Wassermann EM, Williams WA, et al.: Changes in mood and hormone levels after rapid-rate transcranial magnetic stimulation (rTMS) of the prefrontal cortex. J Neuropsychiatry Clin Neurosci 1996; 8:172–180Crossref, MedlineGoogle Scholar

17 George MS, Wassermann EM, Post RM: Transcranial magnetic stimulation: a neuropsychiatric tool for the 21st century. J Neuropsychiatry Clin Neurosci 1996; 8:373–382Crossref, MedlineGoogle Scholar

18 Hoflich G, Kasper S, Hufnagel A, et al.: Application of transcranial magnetic stimulation in the treatment of drug-resistant major depression. Hum Psychopharmacol 1993; 8:361–365CrossrefGoogle Scholar

19 George MS, Ketter TA, Post RM: Prefrontal cortex dysfunction in clinical depression. Depression 1994; 2:59–72CrossrefGoogle Scholar

20 Rossini PM, Burke D, Chen R, et al.: Non-invasive electrical and magnetic stimulation of the brain, spinal cord, roots and peripheral nerves: basic principles and procedures for routine clinical and research application: an updated report from an IFCN Committee. Clin Neurophysiol 2015; 126:1071–1107Crossref, MedlineGoogle Scholar

21 Vahabzadeh-Hagh AM, Muller PA, Gersner R, et al.: Translational neuromodulation: approximating human transcranial magnetic stimulation protocols in rats. Neuromodulation 2012; 15:296–305MedlineGoogle Scholar

22 Lisanby SH, Belmaker RH: Animal models of the mechanisms of action of repetitive transcranial magnetic stimulation (rTMS): comparisons with electroconvulsive shock (ECS). Depress Anxiety 2000; 12:178–187Crossref, MedlineGoogle Scholar

23 Ridding MC, Rothwell JC: Is there a future for therapeutic use of transcranial magnetic stimulation? Nat Rev Neurosci 2007; 8:559–567Crossref, MedlineGoogle Scholar

24 Nobler MS, Sackeim HA, Prohovnik I, et al.: Regional cerebral blood flow in mood disorders, III: treatment and clinical response. Arch Gen Psychiatry 1994; 51:884–897Crossref, MedlineGoogle Scholar

25 Nobler MS, Sackeim HA, Solomou M, et al.: EEG manifestations during ECT: effects of electrode placement and stimulus intensity. Biol Psychiatry 1993; 34:321–330Crossref, MedlineGoogle Scholar

26 George MS, Nahas Z, Lomarov M, et al.: How knowledge of regional brain dysfunction in depression will enable new somatic treatments in the next millennium. CNS Spectr 1999; 4:53–61MedlineGoogle Scholar

27 George MS, Ketter TA, Parekh PI, et al.: Brain activity during transient sadness and happiness in healthy women. Am J Psychiatry 1995; 152:341–351LinkGoogle Scholar

28 Wu JC, Gillin JC, Buchsbaum MS, et al.: Effect of sleep deprivation on brain metabolism of depressed patients. Am J Psychiatry 1992; 149:538–543LinkGoogle Scholar

29 Nobler MS, Oquendo MA, Kegeles LS, et al.: Decreased regional brain metabolism after ECT. Am J Psychiatry 2001; 158:305–308LinkGoogle Scholar

30 Fink M: How does convulsive therapy work? Neuropsychopharmacology 1990; 3:73–82MedlineGoogle Scholar

31 Avissar M, Powell F, Ilieva I, et al.: Functional connectivity of the left DLPFC to striatum predicts treatment response of depression to TMS. Brain Stimul 2017; 10:919–925Crossref, MedlineGoogle Scholar

32 George MS, Lisanby SH, Avery D, et al.: Daily left prefrontal transcranial magnetic stimulation therapy for major depressive disorder: a sham-controlled randomized trial. Arch Gen Psychiatry 2010; 67:507–516Crossref, MedlineGoogle Scholar

33 O’Reardon JP, Solvason HB, Janicak PG, et al.: Efficacy and safety of transcranial magnetic stimulation in the acute treatment of major depression: a multisite randomized controlled trial. Biol Psychiatry 2007; 62:1208–1216Crossref, MedlineGoogle Scholar

34 Perera T, George MS, Grammer G, et al.: The Clinical TMS Society consensus review and treatment recommendations for TMS therapy for major depressive disorder. Brain Stimul 2016; 9:336–346Crossref, MedlineGoogle Scholar

35 Yesavage JA, Fairchild JK, Mi Z, et al.: Effect of repetitive transcranial magnetic stimulation on treatment-resistant major depression in US veterans: a randomized clinical trial. JAMA Psychiatry 2018; 75:884–893Crossref, MedlineGoogle Scholar

36 Carpenter LL, Janicak PG, Aaronson ST, et al.: Transcranial magnetic stimulation (TMS) for major depression: a multisite, naturalistic, observational study of acute treatment outcomes in clinical practice. Depress Anxiety 2012; 29:587–596Crossref, MedlineGoogle Scholar

37 Herbsman T, Avery D, Ramsey D, et al.: More lateral and anterior prefrontal coil location is associated with better repetitive transcranial magnetic stimulation antidepressant response. Biol Psychiatry 2009; 66:509–515Crossref, MedlineGoogle Scholar

38 Herwig U, Satrapi P, Schönfeldt-Lecuona C: Using the international 10-20 EEG system for positioning of transcranial magnetic stimulation. Brain Topogr 2003; 16:95–99Crossref, MedlineGoogle Scholar

39 Herwig U, Padberg F, Unger J, et al.: Transcranial magnetic stimulation in therapy studies: examination of the reliability of “standard” coil positioning by neuronavigation. Biol Psychiatry 2001; 50:58–61Crossref, MedlineGoogle Scholar

40 Beam W, Borckardt JJ, Reeves ST, et al.: An efficient and accurate new method for locating the F3 position for prefrontal TMS applications. Brain Stimul 2009; 2:50–54Crossref, MedlineGoogle Scholar

41 Weigand A, Horn A, Caballero R, et al.: Prospective validation that subgenual connectivity predicts antidepressant efficacy of transcranial magnetic stimulation sites. Biol Psychiatry 2018; 84:28–37Crossref, MedlineGoogle Scholar

42 Drysdale AT, Grosenick L, Downar J, et al.: Resting-state connectivity biomarkers define neurophysiological subtypes of depression. Nat Med 2017; 23:28–38Crossref, MedlineGoogle Scholar

43 Fox MD: Mapping symptoms to brain networks with the human connectome. N Engl J Med 2018; 379:2237–2245Crossref, MedlineGoogle Scholar

44 Siddiqi S, Taylor S, Cooke D, et al.: Distinct symptom-specific targets for circuit-based neuromodulation (abstract O25). Biol Psychiatry 2019; 85:S115–S116Google Scholar

45 Deng ZD, Lisanby SH, Peterchev AV: On the characterization of coils for deep transcranial magnetic stimulation. Clin Neurophysiol 2015; 126:1456–1457MedlineGoogle Scholar

46 Deng ZD, Lisanby SH, Peterchev AV: Coil design considerations for deep transcranial magnetic stimulation. Clin Neurophysiol 2014; 125:1202–1212Crossref, MedlineGoogle Scholar

47 Deng ZD, Lisanby SH, Peterchev AV: Electric field depth-focality tradeoff in transcranial magnetic stimulation: simulation comparison of 50 coil designs. Brain Stimul 2013; 6:1–13Crossref, MedlineGoogle Scholar

48 Roth Y, Zangen A, Hallett M: A coil design for transcranial magnetic stimulation of deep brain regions. J Clin Neurophysiol 2002; 19:361–370Crossref, MedlineGoogle Scholar

49 Levkovitz Y, Isserles M, Padberg F, et al.: Efficacy and safety of deep transcranial magnetic stimulation for major depression: a prospective multicenter randomized controlled trial. World Psychiatry 2015; 14:64–73Crossref, MedlineGoogle Scholar

50 Roth Y, Levkovitz Y, Pell GS, et al.: Safety and characterization of a novel multi-channel TMS stimulator. Brain Stimul 2014; 7:194–205MedlineGoogle Scholar

51 Di Lazzaro V, Pilato F, Saturno E, et al.: Theta-burst repetitive transcranial magnetic stimulation suppresses specific excitatory circuits in the human motor cortex. J Physiol 2005; 565:945–950Crossref, MedlineGoogle Scholar

52 Huang YZ, Sommer M, Thickbroom G, et al.: Consensus: new methodologies for brain stimulation. Brain Stimul 2009; 2:2–13Crossref, MedlineGoogle Scholar

53 Blumberger DM, Vila-Rodriguez F, Thorpe KE, et al.: Effectiveness of theta burst versus high-frequency repetitive transcranial magnetic stimulation in patients with depression (THREE-D): a randomised non-inferiority trial. Lancet 2018; 391:1683–1692Crossref, MedlineGoogle Scholar

54 Anderson B, Mishory A, Nahas Z, et al.: Tolerability and safety of high daily doses of repetitive transcranial magnetic stimulation in healthy young men. J ECT 2006; 22:49–53Crossref, MedlineGoogle Scholar

55 Rossi S, Hallett M, Rossini PM, et al.: Safety, ethical considerations, and application guidelines for the use of transcranial magnetic stimulation in clinical practice and research. Clin Neurophysiol 2009; 120:2008–2039Crossref, MedlineGoogle Scholar

56 Fitzgerald PB, Hoy KE, Reynolds J, et al.: A pragmatic randomized controlled trial exploring the relationship between pulse number and response to repetitive transcranial magnetic stimulation treatment in depression. Brain Stimul (Epub ahead of print, September 6, 2019)Google Scholar

57 Williams NR, Sudheimer KD, Bentzley BS, et al.: High-dose spaced theta-burst TMS as a rapid-acting antidepressant in highly refractory depression. Brain 2018; 141:e18Crossref, MedlineGoogle Scholar

58 Fitzgerald PB, Chen L, Richardson K, et al.: A pilot investigation of an intensive theta burst stimulation protocol for patients with treatment resistant depression. Brain Stimul (Epub ahead of print, August 22, 2019)Google Scholar

59 Hebb DO: The Organization of Behavior: A Neuropsychological Theory. New York, John Wiley & Sons, 1949Google Scholar

60 Gorelick DA, Zangen A, George MS: Transcranial magnetic stimulation in the treatment of substance addiction. Ann N Y Acad Sci 2014; 1327:79–93MedlineGoogle Scholar

61 Dinur-Klein L, Dannon P, Hadar A, et al.: Smoking cessation induced by deep repetitive transcranial magnetic stimulation of the prefrontal and insular cortices: a prospective, randomized controlled trial. Biol Psychiatry 2014; 76:742–749Crossref, MedlineGoogle Scholar

62 Amiaz R, Levy D, Vainiger D, et al.: Repeated high-frequency transcranial magnetic stimulation over the dorsolateral prefrontal cortex reduces cigarette craving and consumption. Addiction 2009; 104:653–660Crossref, MedlineGoogle Scholar

63 Li X, Hartwell KJ, Owens M, et al.: Repetitive transcranial magnetic stimulation of the dorsolateral prefrontal cortex reduces nicotine cue craving. Biol Psychiatry 2013; 73:714–720Crossref, MedlineGoogle Scholar

64 Lisanby SH, Husain MM, Rosenquist PB, et al.: Daily left prefrontal repetitive transcranial magnetic stimulation in the acute treatment of major depression: clinical predictors of outcome in a multisite, randomized controlled clinical trial. Neuropsychopharmacology 2009; 34:522–534Crossref, MedlineGoogle Scholar

65 Taylor JJ, Borckardt JJ, Canterberry M, et al.: Naloxone-reversible modulation of pain circuitry by left prefrontal rTMS. Neuropsychopharmacology 2013; 38:1189–1197Crossref, MedlineGoogle Scholar

66 Taylor JJ, Borckardt JJ, George MS: Endogenous opioids mediate left dorsolateral prefrontal cortex rTMS-induced analgesia. Pain 2012; 153:1219–1225MedlineGoogle Scholar

67 Carmi L, Tendler A, Bystritsky A, et al.: Efficacy and safety of deep transcranial magnetic stimulation for obsessive-compulsive disorder: a prospective multicenter randomized double-blind placebo-controlled trial. Am J Psychiatry 2019; 176:931–938LinkGoogle Scholar

68 Carmi L, Alyagon U, Barnea-Ygael N, et al.: Clinical and electrophysiological outcomes of deep TMS over the medial prefrontal and anterior cingulate cortices in OCD patients. Brain Stimul 2018; 11:158–165Crossref, MedlineGoogle Scholar

69 Adams TG Jr, Badran BW, George MS: Integration of cortical brain stimulation and exposure and response prevention for obsessive-compulsive disorder (OCD). Brain Stimul 2014; 7:764–765Crossref, MedlineGoogle Scholar

70 Philip NS, Barredo J, Aiken E, et al.: Theta-burst transcranial magnetic stimulation for posttraumatic stress disorder. Am J Psychiatry 2019; 176:939–948LinkGoogle Scholar

71 Fryml LD, Pelic CG, Acierno R, et al.: Exposure therapy and simultaneous repetitive transcranial magnetic stimulation: a controlled pilot trial for the treatment of posttraumatic stress disorder. J ECT 2019; 35:53–60Crossref, MedlineGoogle Scholar

72 Sackeim HA, George MS: Brain stimulation-basic, translational, and clinical research in neuromodulation: why a new journal? Brain Stimul 2008; 1:4–6MedlineGoogle Scholar

73 Antal A, Alekseichuk I, Bikson M, et al.: Low intensity transcranial electric stimulation: safety, ethical, legal regulatory and application guidelines. Clin Neurophysiol 2017; 128:1774–1809Crossref, MedlineGoogle Scholar

74 Fridriksson J, Basilakos A, Stark BC, et al.: Transcranial direct current stimulation to treat aphasia: longitudinal analysis of a randomized controlled trial. Brain Stimul 2019; 12:190–191MedlineGoogle Scholar

75 Fridriksson J, Rorden C, Elm J, et al.: Transcranial direct current stimulation vs sham stimulation to treat aphasia after stroke: a randomized clinical trial. JAMA Neurol 2018; 75:1470–1476MedlineGoogle Scholar

76 Fridriksson J, Elm J, Stark BC, et al.: BDNF genotype and tDCS interaction in aphasia treatment. Brain Stimul 2018; 11:1276–1281MedlineGoogle Scholar

77 Bystritsky A, Korb AS, Douglas PK, et al.: A review of low-intensity focused ultrasound pulsation. Brain Stimul 2011; 4:125–136MedlineGoogle Scholar

78 Mueller J, Legon W, Opitz A, et al.: Transcranial focused ultrasound modulates intrinsic and evoked EEG dynamics. Brain Stimul 2014; 7:900–908MedlineGoogle Scholar

79 Legon W, Sato TF, Opitz A, et al.: Transcranial focused ultrasound modulates the activity of primary somatosensory cortex in humans. Nat Neurosci 2014; 17:322–329MedlineGoogle Scholar