Loading

Journal of Addiction Medicine and Therapy

Ketamine and Pharmacological Imaging: Use of Functional Magnetic Resonance Imaging to Evaluate Mechanisms of Action

Review Article | Open Access

  • 1. Yerkes National Primate Research Center, Emory University, USA
  • 2. Department of Radiology and Imaging Sciences, Emory University, USA
  • 3. Department of Psychiatry and Behavioral Sciences, Emory University, USA
+ Show More - Show Less
Corresponding Authors
Leonard Howell, Scientific Programs, Yerkes National Primate Research Center Emory University, 954 Gatewood Rd NE, Atlanta, GA 30329, USA, Tel: (404) 727 7786; Fax: (404) 727; 1266
Abstract

Sub-anesthetic ketamine infusion is the primary pharmacological model used to study schizophrenia and similar administration protocols of the drug are under investigation as a treatment for depression and other psychiatric disorders. However, the mechanisms underlying both the psychotomimetic and therapeutic effects of ketamine remain poorly understood. This review provides an overview of what is known of the neural mechanisms underlying the effects of ketamine and details what functional magnetic resonance imaging studies have revealed at the systems-level. Multiple analysis techniques show that ketamine produces robust and consistent effects at the whole-brain level. These effects are highly conserved across primate species, validating the use of nonhuman primate models for further investigations with ketamine. Regional analysis of functional connectivity suggests that the therapeutic potential of ketamine may be derived from a strengthening of executive control circuitry, making it an intriguing candidate for the treatment of drug abuse. However, there are still many questions about ketamine that can be answered using current functional imaging techniques that have yet to be addressed.

Citation

Maltbie E, Kaundinya G, Howell L (2017) Ketamine and Pharmacological Imaging: Use of Functional Magnetic Resonance Imaging to Evaluate Mechanisms of Action. J Addict Med Ther 5(2): 1031.

Keywords

• Ketamine infusion
• Pharmacological imaging
• Functional connectivity
• Dorsolateral prefrontal cortex

ABBREVIATIONS

NMDA: N-methyl-D-aspartate; fMRI: Functional Magnetic Resonance Imaging; BOLD: Blood Oxygenation Level Dependent; GBC: Global Brain Connectivity; dlPFC: Dorsolateral Prefrontal Cortex; PET: Positron Emission Tomography

INTRODUCTION

Ketamine is a non-competitive N-methyl-D-aspartate (NMDA) glutamate receptor antagonist with a fascinating profile of pharmacological effects that has made it a hot research target in several different fields of medicine and neuroscience. High doses of ketamine have long been used medically as a general anesthetic [1], and the recreational use of ketamine as a psychedelic drug of abuse has a lengthy history as well [2]. For the past twenty years, ketamine has been used as a pharmacological model for schizophrenia as sub-anesthetic infusions have been shown to produce temporary schizophrenia-like symptoms in healthy humans [3,4]. The strength of these models has helped lead to new hypotheses of glutamatergic system dysfunction in schizophrenia [5].

In addition to its utility for modeling schizophrenia, ketamine has emerged as a useful treatment for multiple psychiatric disorders. Sub-anesthetic doses of ketamine in the same range as those used for modeling schizophrenia have shown efficacy for treating postoperative pain [6], neuropathic pain [7], treatment resistant depression [8,9], and suicidal ideation [10]. Indeed, the rapid onset of improvement in suicidal ideation induced by ketamine, reported to emerge as quickly as 40 minutes post-infusion [11], provides a major advantage for treating this psychiatric emergency as other effective treatments are slower acting [12]. Additionally, recent studies have begun to investigate the potential use of ketamine as a treatment for drug addiction [13,14].

NEURAL MECHANISMS UNDERLYING THE EFFECTS OF KETAMINE

The discovery of these remarkable effects of sub-anesthetic ketamine has led to a great deal of research investigating the underlying neural mechanisms. High doses of ketamine result in general suppression of the central nervous system and produce general anesthesia. However, at the lower systemic doses that produce psychotomimetic and rapid antidepressant effects, ketamine administration actually produces enhancement of excitatory glutamatergic transmission [15,16]. There is growing evidence that at the microcircuit level these sub-anesthetic doses of ketamine predominantly block the NMDA receptors on inhibitory interneurons [17,18], resulting in a disinhibition of excitatory projection neurons that is required for the antidepressant effects of ketamine [19]. The reason for ketamine to primarily inhibit interneurons remains unknown. It has been proposed that the tonic firing pattern displayed by many cortical interneurons is likely to persistently remove the magnesium block from NMDA receptors, allowing ketamine to block the channel. Meanwhile, burst firing pyramidal neurons may spend more time with the magnesium block in place, reducing the likelihood that ketamine will block the NMDA receptor channels on these excitatory neurons [20,21].

At the regional level, sub-anesthetic ketamine induces increased levels of both glutamatergic and dopaminergic transmission [15,22,23]. There is evidence suggesting that disinhibition of pyramidal projection neurons in the prefrontal cortex leads to downstream activation of dopaminergic neurons [24-26]. Thus, while sub-anesthetic ketamine induces excitation in many brain areas, its effects on prefrontal circuitry may be of particular importance for the induction of psychotomimetic and antidepressant effects [27-29]. The prominent role of prefrontal circuitry in the effects of sub-anesthetic ketamine may represent a critical limitation for the use of non-primate models [30], in such studies. The greater homology of the human prefrontal cortex to that of other primates [31], might add significant translational value to the use of nonhuman primate models for investigating the effects of ketamine. Indeed, even the micro-circuitry within the prefrontal cortex appears to be well conserved across primate species with NMDA receptors playing an important role in local processing that may not be present in rodents [32].

PHARMACOLOGICAL IMAGING METHODS AND KETAMINE

In order to further investigate the effects of ketamine infusion at the regional and whole-brain level, several different neuroimaging techniques have been used. Among these, functional magnetic resonance imaging (fMRI) has proven invaluable to the study of ketamine. Compared to positron emission tomography (PET), fMRI features higher spatial and temporal resolution [33], which is advantageous for studying ketamine given its relatively short half-life and regionally specific effects in many brain areas. This review will discuss the key findings that fMRI studies have contributed to the mechanistic understanding of the effects ketamine has on the brain and will focus primarily on studies featuring human and nonhuman primate subjects. Multiple fMRI methods have been used to examine the brain activation response to ketamine as well as ketamine-induced changes to functional connectivity.

BRAIN ACTIVATION

Blood oxygenation level dependent (BOLD) fMRI has been used to characterize the regional pattern of changes in neural activity induced by ketamine [34-36]. Importantly, strong evidence that BOLD fMRI reflects neural activity induced by ketamine is available from quantitative PET imaging [37,38]. Taken together, these studies show that cerebral blood flow and oxidative metabolism stay coupled during ketamine infusion (especially at sub-anesthetic doses), thus ensuring that BOLD response to ketamine provides an accurate and quantitative representation of underlying neural activity.

FUNCTIONAL CONNECTIVITY

The BOLD fMRI signal exhibits spontaneous fluctuations associated with temporal patterns of neural activity. Correlations in these spontaneous signal fluctuations between distant regions are termed functional connectivity and are thought to underlie communication within brain networks [39]. Functional connectivity has been used for many different clinical applications [40], including the study of ketamine infusion. The two types of functional connectivity analysis that have been most commonly used to study the effects of ketamine infusion are regional seed-based analysis and global brain connectivity (GBC).

Regional seed-based functional connectivity analysis utilizes a region-of-interest approach to calculate functional connectivity between specific regions. This technique compares the average time course of the BOLD signal within a specified seed region with the BOLD time course of every brain voxel outside of the seed region (or within a specified target region) usually by means of the cross-correlation coefficient (CC) between respective time courses [41]. This is the most common type of functional connectivity analysis used for determining regionally specific effects.

GBC is a measure of whole-brain functional connectivity [42], that calculates the average correlation between the BOLD time course in a given voxel and the BOLD time course of every other voxel in the brain. Alterations in GBC have been associated with schizophrenia [43]and thus ketamine-induced changes to GBC have been investigated in relation to the psychotomimetic effects of ketamine infusion.

Of note, independent component analysis [44,45] and graph network analysis [46,47] have also been used to investigate the effects of subanesthetic ketamine. However, with the exception of Joules, Doyle [47] (discussed below), these studies were designed to examine either the analgesic [44,45] or non-acute [46] effects of ketamine and employed considerably different scanning or ketamine infusion protocol, and thus are not discussed in detail in this review.

THE BRAIN ACTIVATION RESPONSE TO KETAMINE

Deakin, Lees [34], were the first group to examine the effects of ketamine infusion on BOLD activation. They found an extensive cortical BOLD signal response with peak signal changes occurring 3-5 minutes after the start of infusion in all regions. This timing corresponds very well with the peak ketamine concentration in the blood, and with the onset of behavioral effects. BOLD activation was quite extensive, with multiple frontal, parietal, temporal, and limbic regions showing increased signal. There were also prominent areas of deactivation in the subgenual cingulate and medial orbitofrontal cortex. These regions play an important role in cortico-limbic networks [48], responsible for affective processing and may be important for the antidepressant effects of ketamine [49]. Deakin et al. [34], further found several regions in which changes in BOLD were correlated with ratings of dissociative state or psychotic symptoms, thus establishing the relevance of BOLD activation to the psychotomimetic effects of ketamine.

The test-retest reliability of ketamine-induced BOLD activation was later established by De Simoni et al., [35]. They found the BOLD response to ketamine to be a very robust effect, featuring a consistent magnitude and timecourse across different sessions, and both within and across subjects. Further, De Simoni et al. [35], investigated the dose dependence of ketamine-induced BOLD activation and found that a higher dose (75 ng/mL vs. 50 ng/mL) of ketamine corresponded to greater changes in BOLD signal and greater effect sizes. The full ketamine dose-response function has yet to be established with BOLD activation however, and future work should be focused in this area (as discussed in detail below).

Doyle, De Simoni [50], were the first to test the interaction of an antipsychotic drug with the ketamine-induced BOLD response. They tested the effect of pretreatment with risperidone on ketamine-induced brain activation. Clinically, risperidone is one of the most commonly prescribed antipsychotics, featuring similar efficacy and tolerability to other second-generation (“atypical”) antipsychotics used for the treatment of schizophrenia [51]. Risperidone is an antagonist at both dopamine D2 and serotonin 5-HT2A receptors, but with no affinity for any glutamate receptor [52]. Doyle and De Simoni [50], showed that risperidone attenuated the BOLD response to ketamine globally, blunting signal changes in frontal, insular, striatal, and thalamic regions. However, ketamine still induced significant BOLD activation compared to saline in each of these areas following risperidone pretreatment. The magnitude of the activation was simply reduced. The antagonism of 5-HT2A receptors may play an important role in attenuating the effects of ketamine because the dopamine D2 antagonist haloperidol has been shown to be insufficient to prevent the psychotomimetic effects of ketamine [53]. Neurons expressing 5-HT2A in the prefrontal cortex project to the ventral tegmental area and local antagonism of these receptors blocks dopamine overflow in the prefrontal cortex [54].

Ketamine-induced BOLD activation also appears to be well conserved across species. Chin, Upadhyay [55], found extensive activation in the cortex and hippocampus of awake rats, and indeed, ketamine-induced BOLD activation has been shown in anesthetized rats as well [56]. However, a follow-up paper from the same group provides strong evidence that the isoflourane anesthesia used may confound the effects of sub-anesthetic ketamine [57]. Nonhuman primates provide a more translational model for studying the BOLD response to ketamine [30,32], and methods have been developed that enable rhesus monkeys to undergo MRI scanning without the use of anesthesia and with minimal restraint stress [58]. A recent study has shown that awake rhesus monkeys display ketamine-induced BOLD activation [36] that corresponds closely in both magnitude and extent to what appears in the human literature [34,35,50]. Pretreatment with risperidone attenuated the ketamine-induced changes in BOLD in rhesus monkeys [36], again to a similar extent as in humans [50]. This data suggests that the neurochemical effects of ketamine are well conserved across species and attests to the validity of using ketamine in nonhuman primates as an animal model for schizophrenia, and as a potential model for evaluating novel antipsychotics.

KETAMINE-INDUCED CHANGES TO GLOBAL BRAIN CONNECTIVITY

The first paper to use functional connectivity analysis to study ketamine infusion was published by Driesen, McCarthy [59]. They examined the effects of ketamine on global brain connectivity (GBC). Ketamine infusion was found to increase the GBC of voxels throughout the brain, illustrating a global increase in functional connectivity. This finding is consistent with coherent neural activity across the brain seen during psychosis [60,61]. Additionally, Driesen, McCarthy [59], found that increased GBC correlated with increases to positive schizophrenia symptom scores in a number of regions, implying that increased functional connectivity is associated with the psychotomimetic effects of ketamine.

Anticevic, Corlett [62], measured the effects of ketamine using a variant of GBC in which only voxels within the prefrontal cortex were considered. The restricted GBC was shown to increase after ketamine administration and this prefrontal specific GBC was also found to be significantly elevated in patients who were within one-year of the onset of schizophrenia symptoms. This finding may suggest that elevated functional connectivity in the prefrontal cortex could be a biomarker for schizophrenia and might indicate that the psychotomimetic effects of ketamine can be measured using functional connectivity. However, other than benefitting from decreased processing time, it is unclear why GBC should be restricted to prefrontal regions, because even if prefrontal regions are of primary interest these areas receive inputs from many other brain areas outside the prefrontal cortex and is part of highly integrative brain circuits [27,29]. Thus, it is likely more appropriate to consider changes to connectivity with a greater scope even when considering specific regions. Furthermore, Anticevic, Corlett [62], performed a global signal regression to remove the average brain signal from every voxel in the brain. Given that ketamine increases global signal as reported previously, the effects of this regression will be different for ketamine than for baseline or saline conditions and could confound any between-condition comparisons [63].

Recent work from our lab in awake rhesus monkeys has demonstrated that ketamine-induced changes in functional connectivity are also well conserved across primate species [64]. GBC analysis (Figure 1), shows that ketamine causes global hyperconnectivity in rhesus monkeys that is similar in both magnitude and regional pattern to what Driesen, McCarthy [59], observed in human subjects. Thus, there are data from multiple imaging modalities that nonhuman primates provide a highly translational model for the effects of ketamine.

Finally, Joules, Doyle [47], investigated the effects of ketamine infusion on whole-brain functional connectivity using a graph theory analysis. The measures of whole-brain connectedness they consider are similar to GBC, except instead of being calculated on every voxel they are calculated between anatomical regions within a whole-brain parcellation map. Joules, Doyle [47], found a shift in whole-brain functional connectivity with ketamine infusion that is consistent with the reported increase in GBC [59]. They further showed that a pattern recognition algorithm could be used to consistently classify the pattern of functional connectivity induced by ketamine infusion as different as placebo infusion. This finding demonstrates the robustness of the effects of ketamine infusion on whole-brain functional connectivity.

KETAMINE-INDUCED CHANGES TO REGIONAL SEED-BASED FUNCTIONAL CONNECTIVITY

In addition to their GBC study, Driesen, McCarthy [65], performed an investigation of the effects of ketamine on functional connectivity to a seed in the dorsolateral prefrontal cortex (dlPFC). The dlPFC is a region strongly implicated in schizophrenia because of its important role in working memory [17], and the group hypothesized that ketamine-induced changes in connectivity to the dlPFC would be associated with impaired performance on a working memory task. Unfortunately, for this study they used a global signal regression, which may have confounded their results for the reasons mentioned previously. Speculatively, while they found that ketamine reduced connectivity to the dlPFC seed, this could result from a greater impact of global signal regression under the ketamine condition than the saline condition.

Functional connectivity to the striatum was shown to be enhanced by Dandash, Harrison [66], who used a regional seed-based analysis with seeds placed in the dorsal and ventral putamen, dorsal caudate, and nucleus accumbens. While they did not find differences in connectivity to either of the seeds in the putamen, they found increased connectivity from the thalamus to the dorsal caudate and from the ventromedial prefrontal cortex to the nucleus accumbens. These results may have been limited by the high variability the group observed in ketamine plasma levels (68.6 ± 43.6 ng/ml) producing inconsistent drug effects. Still, their finding of increased connectivity between the medial prefrontal cortex and ventral striatum may inform the study of ketamine as a potential treatment for drug addiction (discussed in detail below); a condition in which fronto-striatal connectivity has been found to be impaired [67,68].

Our lab has recently performed a regional seed-based analysis of changes in functional connectivity induced by ketamine in awake rhesus monkeys [64]. Like Dandash, Harrison [66], we observed that ketamine increased connectivity to a seed in the nucleus accumbens, although the increases we observed were considerably more extensive. In addition to the accumbens seed, our analysis also featured seeds in the amygdala, posterior and sub-genual cingulate, orbitofrontal cortex, and dlPFC. Among these seed regions, the greatest ketamine-induced changes in functional connectivity were seen in dlPFC projections. This may be a key finding for explaining both the psychotomimetic and antidepressant effects of ketamine.

The dlPFC is a region strongly implicated in schizophrenia because of its important role in working memory [17]. The hyperconnectivity induced by ketamine could be related to aberrant processing leading to psychotic symptoms, as hypothesized by Driesen, McCarthy [65]. Further, the dlPFC plays an essential role in the executive control of emotion [69], which has been shown to be dysfunctional in major depression [70]. Direct activation of the dlPFC using repeated transcranial magnetic stimulation is an effective treatment for depression [71], presumably because of a resultant strengthening of network connections responsible for executive control of emotion [72,73]. Thus, the ketamine-induced increases in dlPFC connectivity may be a key indicator of psychotomimetic effects present during ketamine infusion as well as the neuroplastic changes thought to underlie the antidepressant effects that follow ketamine administration.

Another study [74], investigated ketamine-induced changes in functional connectivity specifically between the dlPFC and hippocampus. They found that acute ketamine administration increased dlPFC-hippocampus connectivity in both human subjects and in rats. The breadth of their results is limited because only a single connection was examined. However, this data does support the finding in rhesus monkeys [64], of robust ketamine-induced increases in dlPFC functional connectivity.

FUTURE DIRECTIONS FOR IMPROVING THE UNDERSTANDING OF KETAMINE

While ketamine-induced BOLD activation has been a useful first step for studying the whole-brain effects of ketamine, functional connectivity may prove to be more informative for understanding the mechanism of action of ketamine in the brain. It is important to note that when neural activity increases, metabolic activity (and therefore BOLD signal) is most enhanced at the synapses and not at the cell bodies [33]. Thus, in the case of localized disinhibition of pyramidal neurons, as presumed with ketamine, the downstream areas receiving projections from the disinhibited region(s) may show the greatest enhancement of BOLD signal. On the other hand, a region that becomes disinhibited may increase its functional coupling to downstream areas and hence may show increased functional connectivity even when it does not show as strong of an enhancement in BOLD signal [75]. This may explain why the dlPFC shows the most extensive increases in functional connectivity [64], but only moderately increased BOLD signal during ketamine infusion [35, 36].

Future studies could use fMRI to answer several other questions about ketamine. The dose-response relationship for ketamine remains poorly understood. This is true both for the efficacy of ketamine as an antidepressant and for the acute effects of ketamine on brain activity. De Simoni, Schwarz [35], reported that BOLD activation increased with increasing dose, however no peak dose has been established and no investigation of the dose dependence of changes to functional connectivity has been conducted. Further, investigation into the persistent effects of ketamine on brain activity is also warranted. Most fMRI studies to date have focused on examining the acute effects of ketamine, however the antidepressant effects do not typically begin until at least an hour post-infusion [8,28]. One study [46], investigating the sustained effects of ketamine, has reported significant changes to functional connectivity 24-hours post-infusion, however these results may be confounded by the effects of anesthesia [76], and should be replicated in awake subjects.

Another important question regarding the effects of ketamine is whether they are truly global or if a specific region (such as dlPFC) or subset of regions become disinhibited and drive the excitatory effects elsewhere in the brain. The use of a dynamic functional connectivity analysis [77,78] could potentially be used to determine whether there is a specific regional onset to the effects of ketamine. Currently, there are no published studies employing this type of analysis with any NMDA receptor antagonist.

The independent contributions of the individual pharmacological components of ketamine remain largely unknown. Ketamine is a chiral compound consisting of a pair of (R,S) enantiomers and there is some evidence suggesting that R-ketamine may have greater antidepressant efficacy [79] while also producing fewer psychotomimetic effects [80]. Further, there is one report that found a specific metabolite of ketamine to be sufficient for producing antidepressant effects [81]. Imaging these independent components of ketamine separately may lead to important new insights into how the effects of ketamine are mediated. Such experiments may also help to determine whether the psychotomimetic effects of ketamine can truly be segregated from the antidepressant effects.

FUTURE DIRECTIONS FOR INVESTIGATING THE THERAPEUTIC USE OF KETAMINE

Ketamine has already shown tremendous therapeutic value in the treatment of major depression, and the results from functional imaging experiments provide tantalizing evidence of further therapeutic uses for ketamine. The finding that ketamine enhances connectivity to the dlPFC [64], and potentially causes neuroplastic changes that strengthen executive control, has major implications for the potential use of ketamine in treating other psychiatric disorders. Impaired executive control has been associated with a number of disorders and is a particularly common finding in drug addiction [82-84]. Indeed, acute administration of cocaine has been shown to significantly reduce functional connectivity between dlPFC and nucleus accumbens in awake nonhuman primates, and the connectivity between these regions is negatively correlated with cocaine intake during self-administration [68]. There is already some evidence to indicate that sub-anesthetic ketamine infusion may be an effective treatment for cocaine abuse [13,14], and as there are currently no FDA approved medications for the treatment of psychostimulant abuse, further investigation is certainly warranted. Nonhuman primate self-administration represents the gold-standard for modeling addiction, andfMRI studies provide strong evidence that ketamine induces highly translational effects on BOLD signal and functional connectivity in nonhuman primates. An elegantly designed study could investigate the efficacy of ketamine for reducing drug self-administration, as well as the predictive value of dlPFC functional connectivity as a biomarker for drug addiction.

CONCLUSIONS

Acute administration of sub-anesthetic ketamine produces a robust, global increase in BOLD signal that is correlated with the psychotomimetic effects of ketamine. Functional connectivity also undergoes robust, global increases during acute ketamine administration that correlate with the psychotomimetic effects of ketamine. These effects are very well conserved across primate species and could be used to create a translational pharmacological model of schizophrenia in nonhuman primates. Ketamine shows exciting potential as a therapeutic and the results from fMRI experiments suggest it may bolster executive control circuits, making it a particularly good candidate for investigation in the treatment of drug abuse. Future fMRI studies should be able to elucidate many of the questions that still remain unanswered about the mechanisms mediating the effects of ketamine.

ACKNOWLEDGEMENTS

This research was supported by P51OD11132 (Yerkes National Primate Research Center), Sunovion Pharmaceuticals, Ltd (LLH) and DA031246 (LLH). Special thanks to the Yerkes Imaging Center, technicians Marisa Olsen and Juliet Brown, and to Christopher Muly, MD, PhD, Naoko Urushino, PhD, and Doty Kempf, DVM.

REFERENCES

1. Rowland LM. Subanesthetic ketamine: how it alters physiology and behavior in humans. Aviat Space Environ Med. 2005; 76: 52-58.

2. Morgan CJ, Curran HV. Acute and chronic effects of ketamine upon human memory: a review. Psychopharmacology (Berl). 2006; 188: 408-424.

3. Krystal JH, Karper LP, Seibyl JP, Freeman GK, Delaney R, Bremner JD, et al. Subanesthetic effects of the noncompetitive NMDA antagonist, ketamine, in humans. Psychotomimetic, perceptual, cognitive, and neuroendocrine responses. Arch Gen Psychiatry. 1994; 51: 199-214.

4. Olney JW, Farber NB. Glutamate receptor dysfunction and schizophrenia. Arch Gen Psychiatry. 1995; 52: 998-1007.

5. Frohlich J, Van Horn JD. Reviewing the ketamine model for schizophrenia. J Psychopharmacol. 2014; 28: 287-302.

6. Schmid RL, Sandler AN, Katz J. Use and efficacy of low-dose ketamine in the management of acute postoperative pain: a review of current techniques and outcomes. Pain. 1999; 82: 111-125.

7. Schwartzman RJ, Alexander GM, Grothusen JR, Paylor T, Reichenberger E, Perreault M. Outpatient intravenous ketamine for the treatment of complex regional pain syndrome: a double-blind placebo controlled study. Pain. 2009; 147: 107-115.

8. Berman RM, Cappiello A, Anand A, Oren DA, Heninger GR, Charney DS, et al. Antidepressant effects of ketamine in depressed patients. Biol Psychiatry. 2000; 47: 351-354.

9. Krystal JH, Sanacora G, Duman RS. Rapid-acting glutamatergic antidepressants: the path to ketamine and beyond. Biol Psychiatry. 2013; 73: 1133-1141.

10. Price RB, Mathew SJ. Does ketamine have anti-suicidal properties? Current status and future directions. CNS Drugs. 2015; 29: 181-188.

11. DiazGranados N, Ibrahim LA, Brutsche NE, Ameli R, Henter ID, Luckenbaugh DA, et al. Rapid resolution of suicidal ideation after a single infusion of an N-methyl-D-aspartate antagonist in patients with treatment-resistant major depressive disorder. J Clin Psychiatry. 2010; 71: 1605-1611.

12. Reinstatler L, Youssef NA. Ketamine as a potential treatment for suicidal ideation: a systematic review of the literature. Drugs RD. 2015; 15: 37-43.

13. Dakwar E, Hart CL, Levin FR, Nunes EV, Foltin RW. Cocaine self-administration disrupted by the N-methyl-D-aspartate receptor antagonist ketamine: a randomized, crossover trial. Mol Psychiatry. 2017; 22: 76-81.

14. Dakwar E, Levin F, Foltin RW, Nunes EV, Hart CL. The effects of subanesthetic ketamine infusions on motivation to quit and cue-induced craving in cocaine-dependent research volunteers. Biol Psychiatry. 2014; 76: 40-46.

15. Moghaddam B, Adams B, Verma A, Daly D. Activation of glutamatergic neurotransmission by ketamine: a novel step in the pathway from NMDA receptor blockade to dopaminergic and cognitive disruptions associated with the prefrontal cortex. J Neurosci. 1997; 17: 2921- 2927.

16. Duncan GE, Moy SS, Knapp DJ, Mueller RA, Breese GR. Metabolic mapping of the rat brain after subanesthetic doses of ketamine: potential relevance to schizophrenia. Brain Res. 1998; 787: 181-190.

17. Wang M, Yang Y, Wang CJ, Gamo NJ, Jin LE, Mazer JA, et al. NMDA receptors subserve persistent neuronal firing during working memory in dorsolateral prefrontal cortex. Neuron. 2013; 77: 736-749.

18. Homayoun H, Moghaddam B. NMDA receptor hypofunction produces opposite effects on prefrontal cortex interneurons and pyramidal neurons. J Neurosci. 2007; 27: 11496-11500.

19. Maeng, S, Zarate CA Jr, Du J, Schloesser RJ, McCammon J, Chen G, et al. Cellular mechanisms underlying the antidepressant effects of ketamine: role of alpha-amino-3-hydroxy-5-methylisoxazole-4- propionic acid receptors. Biol Psychiatry. 2008; 63: 349-352.

20. Wang HX, Gao WJ. Cell type-specific development of NMDA receptors in the interneurons of rat prefrontal cortex. Neuropsychopharmacology. 2009; 34: 2028-2040.

21. Wang HX, Gao WJ. Prolonged exposure to NMDAR antagonist induces cell-type specific changes of glutamatergic receptors in rat prefrontal cortex. Neuropharmacology. 2012; 62: 1808-1822.

22. Lorrain DS, Baccei CS, Bristow LJ, Anderson JJ, Varney MA. Effects of ketamine and N-methyl-D-aspartate on glutamate and dopamine release in the rat prefrontal cortex: modulation by a group II selective metabotropic glutamate receptor agonist LY379268. Neuroscience. 2003; 117: 697-706.

23. Vollenweider FX, et al. Effects of (S)-Ketamine on Striatal Dopamine: A [11C]raclopride PET Study of a Model Psychosis in Humans. J Psychiatr Res. 2000; 34: 35-43.

24. Del Arco A, Segovia G, Mora F. Blockade of NMDA receptors in the prefrontal cortex increases dopamine and acetylcholine release in the nucleus accumbens and motor activity. Psychopharmacology(Berl). 2008; 201: 325-338.

25. Takahata R, Moghaddam B. Activation of glutamate neurotransmission in the prefrontal cortex sustains the motoric and dopaminergic effects of phencyclidine. Neuropsychopharmacology. 2003; 28: 1117-11124.

26. Carr DB, Sesack SR. Dopamine terminals synapse on callosal projection neurons in the rat prefrontal cortex. J Comp Neurol. 2000; 425: 275- 283.

27. Arnsten AF, Wang MJ, Paspalas CD, Neuromodulation of thought: flexibilities and vulnerabilities in prefrontal cortical network synapses. Neuron. 2012; 76: 223-239.

28. Opler LA, Opler MG, Arnsten AF. Ameliorating treatment-refractory depression with intranasal ketamine: potential NMDA receptor actions in the pain circuitry representing mental anguish. CNS Spectr. 2016; 21: 12-22

29. Del Arco A, Mora F, Neurotransmitters and prefrontal cortex-limbic system interactions: implications for plasticity and psychiatric disorders. J Neural Transm (Vienna). 2009; 116: 941-952.

30. Preuss TM. Do rats have prefrontal cortex? The rose-woolsey-akert program reconsidered. J Cogn Neurosci. 1995; 7: 1-24.

31. Phillips KA, Bales KL, Capitanio JP, Conley A, Czoty PW, ‘t Hart BA, et al. Why primate models matter. Am J Primatol. 2014; 76: 801-827.

32. Wang M, Arnsten AF. Contribution of NMDA receptors to dorsolateral prefrontal cortical networks in primates. Neurosci Bull. 2015; 31: 191-197.

33. Buxton RB, Introduction to functional magnetic resonance imaging: principles and techniques. 2002, Cambridge, UK ; New York: Cambridge University Press. xi, 523.

34. Deakin JF, Lees J, McKie S, Hallak JE, Williams SR, Dursun SM. Glutamate and the neural basis of the subjective effects of ketamine: a pharmaco-magnetic resonance imaging study. Arch Gen Psychiatry. 2008; 65: 154-164.

35. De Simoni S, Schwarz AJ, O’Daly OG, Marquand AF, Brittain C, Gonzales C, et al. Test-retest reliability of the BOLD pharmacological MRI response to ketamine in healthy volunteers. Neuroimage. 2013; 64: 75-90.

36. Maltbie E, Gopinath K, Urushino N, Kempf D, Howell L. Ketamine-induced brain activation in awake female nonhuman primates: a translational functional imaging model. Psychopharmacology (Berl). 2016. 233: 961-972.

37. Långsjö JW, Salmi E, Kaisti KK, Aalto S, Hinkka S, Aantaa R, et al. Effects of subanesthetic ketamine on regional cerebral glucose metabolism in humans. Anesthesiology. 2004; 100: p. 1065-1071.

38. Långsjö JW, Kaisti KK, Aalto S, Hinkka S, Aantaa R, Oikonen V, et al. Effects of subanesthetic doses of ketamine on regional cerebral blood flow, oxygen consumption, and blood volume in humans. Anesthesiology. 2003; 99: 614-623.

39. Fox MD, Snyder AZ, Vincent JL, Corbetta M, Van Essen D, Raichle M. The human brain is intrinsically organized into dynamic, anticorrelated functional networks. Proc Natl Acad Sci U S A. 2005; 102: 9673-9678.

40. Fox MD, Greicius M. Clinical applications of resting state functional connectivity. Front Syst Neurosci. 2010; 4: 19.

41. Fox MD, Raichle ME. Spontaneous fluctuations in brain activity observed with functional magnetic resonance imaging. Nat Rev Neurosci. 2007; 8: 700-711.

42. Cole MW, Pathak S, Schneider W. Identifying the brain’s most globally connected regions. Neuroimage, 2010; 49: 3132-3148.

43. Cole MW, Anticevic A, Repovs G, Barch D. Variable global dysconnectivity and individual differences in schizophrenia. Biol Psychiatry. 2011; 70: 43-50.

44. Niesters M, Khalili-Mahani N, Martini C, Aarts L, van Gerven J, Mark A. van, et al. Effect of subanesthetic ketamine on intrinsic functional brain connectivity: a placebo-controlled functional magnetic resonance imaging study in healthy male volunteers. Anesthesiology. 2012; 117: 868-877.

45. Kleinloog D, Serge R, Remco Z, Linda K, Marieke N, Khalili-Mahani Najmeh, et al. Subjective Effects of Ethanol, Morphine, Delta(9)- Tetrahydrocannabinol, and Ketamine Following a Pharmacological Challenge Are Related to Functional Brain Connectivity. Brain Connect. 2015; 5: 641-648.

46. Lv Q, Liqin Yang, Guoliang Li, Zhiwei Wang, Zhuangming Shen, Wenwen Yu, et al. Large-Scale Persistent Network Reconfiguration Induced by Ketamine in Anesthetized Monkeys: Relevance to Mood Disorders. Biol Psychiatry. 2016; 79: 765-775.

47. Joules R, Doyle OM, Schwarz AJ, O’Daly OG, Brammer M, Williams SC, et al. Ketamine induces a robust whole-brain connectivity pattern that can be differentially modulated by drugs of different mechanism and clinical profile. Psychopharmacol. 2015; 232: 21-22.

48. Mayberg HS. Modulating dysfunctional limbic-cortical circuits in depression: towards development of brain-based algorithms for diagnosis and optimised treatment. Br Med Bull. 2003; 65: 193-207.

49. Johansen-Berg H, Gutman DA, Behrens TE, Matthews PM, Rushworth MF, Katz E, et al. Anatomical connectivity of the subgenual cingulate region targeted with deep brain stimulation for treatment-resistant depression. Cereb Cortex. 2008; 18: 1374-1383.

50. Doyle OM, De Simoni S, Schwarz AJ, Brittain C, O’Daly OG, Williams SC et al. Quantifying the attenuation of the ketamine pharmacological magnetic resonance imaging response in humans: a validation using antipsychotic and glutamatergic agents. J Pharmacol Exp Ther. 2013; 345: 151-160.

51. Komossa K, Rummel-Kluge C, Schwarz S, Schmid F, Hunger H, Kissling W, et al., Risperidone versus other atypical antipsychotics for schizophrenia. Cochrane Database Syst Rev. 2011; CD006626.

52. Muly EC, Votaw JR, Ritchie J, Howell LL. Relationship between dose, drug levels, and D2 receptor occupancy for the atypical antipsychotics risperidone and paliperidone. J Pharmacol Exp Ther. 2012; 341: 81- 89.

53. Krystal JH, D’Souza DC, Karper LP, Bennett A, Abi-Dargham A, Abi-Saab D, et al. Interactive effects of subanesthetic ketamine and haloperidol in healthy humans. Psychopharmacol. 1999; 193-204.

54. Bortolozzi A, Díaz-Mataix L, Scorza MC, Celada P, Artigas F. The activation of 5-HT receptors in prefrontal cortex enhances dopaminergic activity. J Neurochem. 2005; 95: 1597-1607.

55. Chin CL, Upadhyay J, Marek GJ, Baker SJ, Zhang M, Mezler M, et al. Awake rat pharmacological magnetic resonance imaging as a translational pharmacodynamic biomarker: metabotropic glutamate 2/3 agonist modulation of ketamine-induced blood oxygenation level dependence signals. J Pharmacol Exp Ther. 2011; 336: 709-715.

56. Littlewood CL, Jones N, O’Neill MJ, Mitchell SN, Tricklebank M, Williams SC. Mapping the central effects of ketamine in the rat using pharmacological MRI. Psychopharmacol. 2006; 64-81.

57. Hodkinson DJ, de Groote C, McKie S, Deakin JF, Williams SR. Differential Effects of Anaesthesia on the phMRI Response to Acute Ketamine Challenge. Br J Med Med Res. 2012; 2: 373-385.

58. Murnane KS, Howell LL. Development of an apparatus and methodology for conducting functional magnetic resonance imaging (fMRI) with pharmacological stimuli in conscious rhesus monkeys. J Neurosci Methods. 2010; 191: 11-20.

59. Driesen NR, McCarthy G, Bhagwagar Z, Bloch M, Calhoun V, Deepak CD, et al. Relationship of resting brain hyperconnectivity and schizophrenia-like symptoms produced by the NMDA receptor antagonist ketamine in humans. Mol Psychiatry. 2013; 18: 1199-1204.

60. Hakami T, Nigel CJ, Elena AT, Julien G, Joseph C, Michael S, et al. NMDA receptor hypofunction leads to generalized and persistent aberrant gamma oscillations independent of hyperlocomotion and the state of consciousness. PLoS One. 2009; 4: e6755.

61. Wood J, Kim Y, Moghaddam B. Disruption of prefrontal cortex large scale neuronal activity by different classes of psychotomimetic drugs. J Neurosci. 2012; 32: 3022-3031.

62. Anticevic A, Corlett PR, Cole MW, Savic A, Gancsos M, Tang Y, Repovs G, et al. N-methyl-D-aspartate receptor antagonist effects on prefrontal cortical connectivity better model early than chronic schizophrenia. Biol Psychiatry. 2015; 77: 569-580.

63. Saad ZS, Stephen JG, Kevin Murphy, Gang Chen, Hang JJ, Alex Martin, et al. Trouble at rest: how correlation patterns and group differences become distorted after global signal regression. Brain Connect. 2012; 2: 25-32.

64. Gopinath K, Kaundinya G, Naoko U, Doty K, Leonard H. Ketamineinduced changes in connectivity of functional brain networks in awake female nonhuman primates: a translational functional imaging model. Psychopharmacol. 2016; 233: 3673-3684.

65. Driesen NR, McCarthy G, Bhagwagar Z, Michael HB, Vincent DC. The impact of NMDA receptor blockade on human working memory-related prefrontal function and connectivity. Neuropsychopharmacol. 2013; 38: 2613-2622.

66. Dandash O, Ben JH, Ram A, Raphael G, Francesco G, Stephen JW, et al. Selective augmentation of striatal functional connectivity following NMDA receptor antagonism: implications for psychosis. Neuropsychopharmacol. 2015; 40: 622-631.

67. Hu Y, Jo Salmeron B, Gu H, Elliot AS, Yang Y. Impaired functional connectivity within and between frontostriatal circuits and its association with compulsive drug use and trait impulsivity in cocaine addiction. JAMA Psychiatry. 2015; 72: 584-592.

68. Murnane KS, Gopinath KS, Maltbie E, Daunais JB, Telesford QK, Howell LL. Functional connectivity in frontal-striatal brain networks and cocaine self-administration in female rhesus monkeys. Psychopharmacol. 2015; 232: 745-754.

69. Ochsner KN, Gross JJ. The cognitive control of emotion. Trends Cogn Sci. 2005; 9: 242-249.

70. Fales CL, Barch DM, Rundle MM, Mintun MA, Snyder AZ, Cohen JD, et al. Altered emotional interference processing in affective and cognitive-control brain circuitry in major depression. Biol Psychiatry. 2008; 63: 377-384.

71. Concerto, Lanza G, Cantone M, Ferri R, Pennisi G, Bella RC. Repetitive transcranial magnetic stimulation in patients with drug-resistant major depression: A six-month clinical follow-up study. Int J Psychiatry Clin Pract. 2015; 19: 252-258.

72. Fox MD, Buckner RL, White MP, Greicius MD, Pascual-Leone A. Efficacy of transcranial magnetic stimulation targets for depression is related to intrinsic functional connectivity with the subgenual cingulate. Biol Psychiatry. 2012; 72: 595-603.

73. Koenigs M, Grafman J. The functional neuroanatomy of depression: distinct roles for ventromedial and dorsolateral prefrontal cortex. Behav Brain Res. 2009; 201: 239-243.

74. Grimm O, Gass N, Weber-Fahr W, Sartorius A, Schenker E, Spedding M, et al. Acute ketamine challenge increases resting state prefrontalhippocampal connectivity in both humans and rats. Psychopharmacol. 2015; 232: 4231-4241.

75. Gusnard DA, Raichle ME. Searching for a baseline: functional imaging and the resting human brain. Nat Rev Neurosci. 2001; 2: 685-694.

76. Hudetz AG. General anesthesia and human brain connectivity. Brain Connect. 2012; 2: 291-302.

77. Hutchison RM,Womelsdorf T, Allen EA, Bandettini PA, Calhoun VD, Corbetta M, et al. Dynamic functional connectivity: promise, issues, and interpretations. Neuroimage. 2013; 80: 360-378.

78. Chang C, Leopold DA, Schölvinck ML, Mandelkow H, Picchioni D, Liu X, et al. Tracking brain arousal fluctuations with fMRI. Proc Natl Acad Sci USA. 2016; 113: 4518-4523.

79. Zhang JC, Li SX, Hashimoto K. R (-)-ketamine shows greater potency and longer lasting antidepressant effects than S (+)-ketamine. Pharmacol Biochem Behav. 2014; 116: 137-141.

80. Yang C, Shirayama Y, Zhang JC, Ren Q, Yao W, Ma M, et al. R-ketamine: a rapid-onset and sustained antidepressant without psychotomimetic side effects. Transl Psychiatry. 2015; 5: e632.

81. Zanos P, Moaddel R, Morris PJ, Georgiou P, Fischell J, Elmer GI, et al. NMDAR inhibition-independent antidepressant actions of ketamine metabolites. Nature. 2016; 533: 481-486.

82. Goldstein RZ, Volkow ND. Dysfunction of the prefrontal cortex in addiction: neuroimaging findings and clinical implications. Nat Rev Neurosci. 2011; 12: 652-669.

83. Volkow ND, Tomasi D, Wang GJ, Fowler JS, Telang F, Goldstein RZ, et al. Reduced metabolism in brain “control networks” following cocainecues exposure in female cocaine abusers. PLoS One. 2011; 6: e16573.

84. Jentsch JD, Taylor JR. Impulsivity resulting from frontostriatal dysfunction in drug abuse: implications for the control of behavior by reward-related stimuli. Psychopharmacol. 1999; 146: 373-390.

Maltbie E, Kaundinya G, Howell L (2017) Ketamine and Pharmacological Imaging: Use of Functional Magnetic Resonance Imaging to Evaluate Mechanisms of Action. J Addict Med Ther 5(2): 1031.

Received : 15 Mar 2017
Accepted : 29 Jun 2017
Published : 30 Jun 2017
Journals
Annals of Otolaryngology and Rhinology
ISSN : 2379-948X
Launched : 2014
JSM Schizophrenia
Launched : 2016
Journal of Nausea
Launched : 2020
JSM Internal Medicine
Launched : 2016
JSM Hepatitis
Launched : 2016
JSM Oro Facial Surgeries
ISSN : 2578-3211
Launched : 2016
Journal of Human Nutrition and Food Science
ISSN : 2333-6706
Launched : 2013
JSM Regenerative Medicine and Bioengineering
ISSN : 2379-0490
Launched : 2013
JSM Spine
ISSN : 2578-3181
Launched : 2016
Archives of Palliative Care
ISSN : 2573-1165
Launched : 2016
JSM Nutritional Disorders
ISSN : 2578-3203
Launched : 2017
Annals of Neurodegenerative Disorders
ISSN : 2476-2032
Launched : 2016
Journal of Fever
ISSN : 2641-7782
Launched : 2017
JSM Bone Marrow Research
ISSN : 2578-3351
Launched : 2016
JSM Mathematics and Statistics
ISSN : 2578-3173
Launched : 2014
Journal of Autoimmunity and Research
ISSN : 2573-1173
Launched : 2014
JSM Arthritis
ISSN : 2475-9155
Launched : 2016
JSM Head and Neck Cancer-Cases and Reviews
ISSN : 2573-1610
Launched : 2016
JSM General Surgery Cases and Images
ISSN : 2573-1564
Launched : 2016
JSM Anatomy and Physiology
ISSN : 2573-1262
Launched : 2016
JSM Dental Surgery
ISSN : 2573-1548
Launched : 2016
Annals of Emergency Surgery
ISSN : 2573-1017
Launched : 2016
Annals of Mens Health and Wellness
ISSN : 2641-7707
Launched : 2017
Journal of Preventive Medicine and Health Care
ISSN : 2576-0084
Launched : 2018
Journal of Chronic Diseases and Management
ISSN : 2573-1300
Launched : 2016
Annals of Vaccines and Immunization
ISSN : 2378-9379
Launched : 2014
JSM Heart Surgery Cases and Images
ISSN : 2578-3157
Launched : 2016
Annals of Reproductive Medicine and Treatment
ISSN : 2573-1092
Launched : 2016
JSM Brain Science
ISSN : 2573-1289
Launched : 2016
JSM Biomarkers
ISSN : 2578-3815
Launched : 2014
JSM Biology
ISSN : 2475-9392
Launched : 2016
Archives of Stem Cell and Research
ISSN : 2578-3580
Launched : 2014
Annals of Clinical and Medical Microbiology
ISSN : 2578-3629
Launched : 2014
JSM Pediatric Surgery
ISSN : 2578-3149
Launched : 2017
Journal of Memory Disorder and Rehabilitation
ISSN : 2578-319X
Launched : 2016
JSM Tropical Medicine and Research
ISSN : 2578-3165
Launched : 2016
JSM Head and Face Medicine
ISSN : 2578-3793
Launched : 2016
JSM Cardiothoracic Surgery
ISSN : 2573-1297
Launched : 2016
JSM Bone and Joint Diseases
ISSN : 2578-3351
Launched : 2017
JSM Bioavailability and Bioequivalence
ISSN : 2641-7812
Launched : 2017
JSM Atherosclerosis
ISSN : 2573-1270
Launched : 2016
Journal of Genitourinary Disorders
ISSN : 2641-7790
Launched : 2017
Journal of Fractures and Sprains
ISSN : 2578-3831
Launched : 2016
Journal of Autism and Epilepsy
ISSN : 2641-7774
Launched : 2016
Annals of Marine Biology and Research
ISSN : 2573-105X
Launched : 2014
JSM Health Education & Primary Health Care
ISSN : 2578-3777
Launched : 2016
JSM Communication Disorders
ISSN : 2578-3807
Launched : 2016
Annals of Musculoskeletal Disorders
ISSN : 2578-3599
Launched : 2016
Annals of Virology and Research
ISSN : 2573-1122
Launched : 2014
JSM Renal Medicine
ISSN : 2573-1637
Launched : 2016
Journal of Muscle Health
ISSN : 2578-3823
Launched : 2016
JSM Genetics and Genomics
ISSN : 2334-1823
Launched : 2013
JSM Anxiety and Depression
ISSN : 2475-9139
Launched : 2016
Clinical Journal of Heart Diseases
ISSN : 2641-7766
Launched : 2016
Annals of Medicinal Chemistry and Research
ISSN : 2378-9336
Launched : 2014
JSM Pain and Management
ISSN : 2578-3378
Launched : 2016
JSM Women's Health
ISSN : 2578-3696
Launched : 2016
Clinical Research in HIV or AIDS
ISSN : 2374-0094
Launched : 2013
Journal of Endocrinology, Diabetes and Obesity
ISSN : 2333-6692
Launched : 2013
Journal of Substance Abuse and Alcoholism
ISSN : 2373-9363
Launched : 2013
JSM Neurosurgery and Spine
ISSN : 2373-9479
Launched : 2013
Journal of Liver and Clinical Research
ISSN : 2379-0830
Launched : 2014
Journal of Drug Design and Research
ISSN : 2379-089X
Launched : 2014
JSM Clinical Oncology and Research
ISSN : 2373-938X
Launched : 2013
JSM Bioinformatics, Genomics and Proteomics
ISSN : 2576-1102
Launched : 2014
JSM Chemistry
ISSN : 2334-1831
Launched : 2013
Journal of Trauma and Care
ISSN : 2573-1246
Launched : 2014
JSM Surgical Oncology and Research
ISSN : 2578-3688
Launched : 2016
Annals of Food Processing and Preservation
ISSN : 2573-1033
Launched : 2016
Journal of Radiology and Radiation Therapy
ISSN : 2333-7095
Launched : 2013
JSM Physical Medicine and Rehabilitation
ISSN : 2578-3572
Launched : 2016
Annals of Clinical Pathology
ISSN : 2373-9282
Launched : 2013
Annals of Cardiovascular Diseases
ISSN : 2641-7731
Launched : 2016
Journal of Behavior
ISSN : 2576-0076
Launched : 2016
Annals of Clinical and Experimental Metabolism
ISSN : 2572-2492
Launched : 2016
Clinical Research in Infectious Diseases
ISSN : 2379-0636
Launched : 2013
JSM Microbiology
ISSN : 2333-6455
Launched : 2013
Journal of Urology and Research
ISSN : 2379-951X
Launched : 2014
Journal of Family Medicine and Community Health
ISSN : 2379-0547
Launched : 2013
Annals of Pregnancy and Care
ISSN : 2578-336X
Launched : 2017
JSM Cell and Developmental Biology
ISSN : 2379-061X
Launched : 2013
Annals of Aquaculture and Research
ISSN : 2379-0881
Launched : 2014
Clinical Research in Pulmonology
ISSN : 2333-6625
Launched : 2013
Journal of Immunology and Clinical Research
ISSN : 2333-6714
Launched : 2013
Annals of Forensic Research and Analysis
ISSN : 2378-9476
Launched : 2014
JSM Biochemistry and Molecular Biology
ISSN : 2333-7109
Launched : 2013
Annals of Breast Cancer Research
ISSN : 2641-7685
Launched : 2016
Annals of Gerontology and Geriatric Research
ISSN : 2378-9409
Launched : 2014
Journal of Sleep Medicine and Disorders
ISSN : 2379-0822
Launched : 2014
JSM Burns and Trauma
ISSN : 2475-9406
Launched : 2016
Chemical Engineering and Process Techniques
ISSN : 2333-6633
Launched : 2013
Annals of Clinical Cytology and Pathology
ISSN : 2475-9430
Launched : 2014
JSM Allergy and Asthma
ISSN : 2573-1254
Launched : 2016
Journal of Neurological Disorders and Stroke
ISSN : 2334-2307
Launched : 2013
Annals of Sports Medicine and Research
ISSN : 2379-0571
Launched : 2014
JSM Sexual Medicine
ISSN : 2578-3718
Launched : 2016
Annals of Vascular Medicine and Research
ISSN : 2378-9344
Launched : 2014
JSM Biotechnology and Biomedical Engineering
ISSN : 2333-7117
Launched : 2013
Journal of Hematology and Transfusion
ISSN : 2333-6684
Launched : 2013
JSM Environmental Science and Ecology
ISSN : 2333-7141
Launched : 2013
Journal of Cardiology and Clinical Research
ISSN : 2333-6676
Launched : 2013
JSM Nanotechnology and Nanomedicine
ISSN : 2334-1815
Launched : 2013
Journal of Ear, Nose and Throat Disorders
ISSN : 2475-9473
Launched : 2016
JSM Ophthalmology
ISSN : 2333-6447
Launched : 2013
Journal of Pharmacology and Clinical Toxicology
ISSN : 2333-7079
Launched : 2013
Annals of Psychiatry and Mental Health
ISSN : 2374-0124
Launched : 2013
Medical Journal of Obstetrics and Gynecology
ISSN : 2333-6439
Launched : 2013
Annals of Pediatrics and Child Health
ISSN : 2373-9312
Launched : 2013
JSM Clinical Pharmaceutics
ISSN : 2379-9498
Launched : 2014
JSM Foot and Ankle
ISSN : 2475-9112
Launched : 2016
JSM Alzheimer's Disease and Related Dementia
ISSN : 2378-9565
Launched : 2014
Journal of Veterinary Medicine and Research
ISSN : 2378-931X
Launched : 2013
Annals of Public Health and Research
ISSN : 2378-9328
Launched : 2014
Annals of Orthopedics and Rheumatology
ISSN : 2373-9290
Launched : 2013
Journal of Clinical Nephrology and Research
ISSN : 2379-0652
Launched : 2014
Annals of Community Medicine and Practice
ISSN : 2475-9465
Launched : 2014
Annals of Biometrics and Biostatistics
ISSN : 2374-0116
Launched : 2013
JSM Clinical Case Reports
ISSN : 2373-9819
Launched : 2013
Journal of Cancer Biology and Research
ISSN : 2373-9436
Launched : 2013
Journal of Surgery and Transplantation Science
ISSN : 2379-0911
Launched : 2013
Journal of Dermatology and Clinical Research
ISSN : 2373-9371
Launched : 2013
JSM Gastroenterology and Hepatology
ISSN : 2373-9487
Launched : 2013
Annals of Nursing and Practice
ISSN : 2379-9501
Launched : 2014
JSM Dentistry
ISSN : 2333-7133
Launched : 2013
Author Information X