SlideShare a Scribd company logo
1 of 9
Download to read offline
CHRONIC PAIN (R STAUD, SECTION EDITOR)
Biomarkers for Musculoskeletal Pain Conditions: Use of Brain
Imaging and Machine Learning
Jeff Boissoneault1
& Landrew Sevel1
& Janelle Letzen1
& Michael Robinson1
&
Roland Staud2
# Springer Science+Business Media New York 2017
Abstract Chronic musculoskeletal pain condition often
shows poor correlations between tissue abnormalities and
clinical pain. Therefore, classification of pain conditions like
chronic low back pain, osteoarthritis, and fibromyalgia de-
pends mostly on self report and less on objective findings like
X-ray or magnetic resonance imaging (MRI) changes.
However, recent advances in structural and functional brain
imaging have identified brain abnormalities in chronic pain
conditions that can be used for illness classification. Because
the analysis of complex and multivariate brain imaging data is
challenging, machine learning techniques have been increas-
ingly utilized for this purpose. The goal of machine learning is
to train specific classifiers to best identify variables of interest
on brain MRIs (i.e., biomarkers). This report describes classi-
fication techniques capable of separating MRI-based brain
biomarkers of chronic pain patients from healthy controls with
high accuracy (70–92%) using machine learning, as well as
critical scientific, practical, and ethical considerations related
to their potential clinical application. Although self-report re-
mains the gold standard for pain assessment, machine learning
may aid in the classification of chronic pain disorders like
chronic back pain and fibromyalgia as well as provide mech-
anistic information regarding their neural correlates.
Keywords Chronic pain . Musculoskeletal . Magnetic
resonance imaging (MRI) . Machine learning . Classification
Introduction
Chronic pain is a highly prevalent condition associated with
significant disability and societal cost [1]. The etiology of
chronic pain can vary substantially across patients and often
appears to be secondary to biological factors, such as muscu-
loskeletal injury (e.g., osteoarthritis [OA]), nerve injury (neu-
ropathic pain), autoimmunity (rheumatoid arthritis [RA], sys-
temic lupus erythematosus), or substance abuse (alcoholic
neuropathy and chronic pancreatitis). In many cases, however,
chronic pain seems to be the primary illness associated with a
given condition (e.g., fibromyalgia syndrome [FM; 2]). Over
the past several decades, substantial effort has been dedicated
to the development of methodologies for the reliable discrim-
ination or classification of patients with a given chronic pain
condition from people without the condition (i.e., “healthy
controls”), patients with similar but distinct illnesses, or both.
The goal of many studies is to accurately identify individ-
uals with a clinical chronic pain condition in question based
on potential mechanistic underpinnings. Given that some
chronic pain conditions are associated with peripheral tissue
pathophysiology, numerous studies have been aimed at reli-
able discrimination of individuals with clinical pain using pe-
ripheral measures including structural (e.g., knee degenera-
tion, lumbar disc pathology; [3]), functional (e.g., gait abnor-
mality, inflammatory processes in rheumatoid arthritis; [4, 5]),
and genetic data [6]. Because chronic pain often exists as a
primary symptom without remarkable tissue abnormalities,
there is an increasing interest in their neural correlates, i.e.,
mechanistic or structural abnormalities derived from structural
or functional magnetic resonance (MRI) brain imaging studies
This article is part of the Topical Collection on Chronic Pain
* Roland Staud
staudr@ufl.edu
1
Department of Clinical and Health Psychology, University of Florida,
Gainesville, FL, USA
2
Department of Medicine, University of Florida, PO Box 100277,
Gainesville, FL 32610, USA
Curr Rheumatol Rep (2017) 19:5
DOI 10.1007/s11926-017-0629-9
of chronic pain (e.g., grey matter density/volume, white matter
integrity, or functional connectivity between pain-related
brain regions). In addition, such information can be used to
determine whether central nervous system (CNS) abnormali-
ties are suitable to appropriately classify individuals with
chronic pain.
In this regard, a major goal of many MRI-based chronic
pain classification studies is the development of objective bio-
markers for each condition that do not rely on self-report [7,
8•, 9]. Brain-based biomarker studies of chronic pain leverage
the extensive neuroimaging literature describing the critical
role of certain brain regions in the sensory (e.g., thalamus,
primary and secondary somatosensory cortex, posterior
insula, basal ganglia), affective (e.g., anterior insula, hypothal-
amus, anterior cingulate cortex, amygdala, hippocampus), and
cognitive-evaluative (e.g., dorsolateral prefrontal cortex, ante-
rior cingulate cortex, thalamus) aspects of the pain experience
(for review, see [10]). Although our review focuses exclusive-
ly on brain imaging biomarkers, it is worth noting that a sub-
stantial literature has also applied this approach to peripheral
mechanisms. For instance, measurement of exhaled organic
volatiles [11] and joint ultrasound [12] have been suggested as
useful approaches for the automated classification and/or dif-
ferential diagnosis of rheumatoid arthritis.
Some proponents of brain imaging-based approaches to
illness classification claim that brain biomarkers could act as
a “surrogate” for pain self-report [13]. This could result in
greater sensitivity to measure efficacy of analgesic treatments
[9] by avoiding potential pitfalls associated with the use of
subjective patient and provider reports. Furthermore, such
brain biomarkers could serve as targets for novel treatments
[14, 15]. In particular, objective markers of chronic pain may
play an important role in pain classification of patients with
cognitive/psychological dysfunction and in individuals who
are unable to communicate, or in cases of deception [16•,
17], as well as aid in the adjudication of legal claims of
injury-related pain disorders [18]. These purported advantages
have spurred interest in the development of biomarkers, al-
though it is important to note that self-report remains the gold
standard for pain measurement. Numerous studies have indi-
cated that self-report measures of pain and psychosocial fac-
tors have excellent classification accuracy and reliability [8•,
19•, 20]. Indeed, evidence suggests that chronic pain patients
can be distinguished from healthy controls with greater than
90% accuracy based on personality factors [21], perceived
pain and functional disability [22], and simple visual analog
scale (VAS) measures of affect [23••]. Questionnaire-based
approaches have shown similar accuracy for the separation
of FM and RA [24] and FM, RA, and OA [25].
The purpose of this critical review is to examine the devel-
opment of brain biomarkers for chronic musculoskeletal pain
disorders using machine learning (ML) in a manner which
should be useful to physicians and other health care providers.
Therefore, we will restrict our review to musculoskeletal con-
ditions for which chronic pain is a primary complaint. The
conceptual, scientific, and ethical issues related to this ap-
proach, however, will also apply to biomarker studies for other
chronic conditions (e.g., depression).
Machine Learning Algorithms
Existing methods regarding the use of structural and function-
al brain imaging to discriminate patient groups from healthy
controls have largely relied on ML algorithms, which provide
an automated approach to making predictions about previous-
ly unknown data. ML algorithms can be broadly classified as
supervised or unsupervised. Supervised methods develop
models for classifying observations according to a known out-
come (e.g., a clinical diagnosis). In contrast, unsupervised
methods attempt to discern patterns or structure in data with-
out guidance or pre-existing labels. Although unsupervised
ML methods may be of interest for the development of sub-
groups within existing conditions or new diagnostic classifi-
cation criteria based on brain abnormalities, ML studies in
chronic pain to date have mostly relied on supervised
methods. Support vector machines (SVM; [26, 27]), a type
of ML algorithm that uses a training data set composed of
one or more features to determine an optimal boundary sepa-
rating a set of cases, have been most often used [23••, 28, 29].
However, many other ML techniques have been developed
and applied to the automated classification of chronic pain
patients [23••, 30]. Detailed discussions of the strengths and
limitations of these methods are beyond the scope of this re-
view, but basic descriptions of the most common ML algo-
rithms applied in the study of chronic pain, as well as detailed
references, are provided in Table 1.
Scientific and Clinical Utility of Chronic Pain
Biomarkers
In this section, we will discuss theoretical, practical, and eth-
ical issues surrounding pain biomarker development and use,
with a particular emphasis on clinical utility.
Choice of Classification Algorithm
In an ideal scenario, the best algorithm for detecting a chronic
pain condition would be one that matches the underlying mod-
el or structure that distinguishes that condition from healthy
individuals or other conditions (e.g., if a linear decrease of
prefrontal cortical thickness is the underlying mechanism of
a condition, then a linear model would be sufficient).
However, given the complex and often heterogeneous nature
of chronic pain conditions, this information is frequently not
5 Page 2 of 9 Curr Rheumatol Rep (2017) 19:5
available. It is often difficult, if not impossible, to determine a
priori which algorithm might perform best in a certain data set
[37]. This problem frequently necessitates the comparison of
multiple different algorithms for any single classification task
(for examples, see [23••, 38]). As a general rule, simple
models may not perform as well on training data but will
likely generalize better when assessed on testing data, whereas
more complex models will tend to perform well on training
data but will not perform as well on testing data. More com-
plex models or those that blend inputs for many different
algorithms (e.g., ensemble methods) may also lead to chal-
lenges in interpretability, requiring additional pipelines to dis-
till results in ways that are meaningful and useful for clini-
cians. In general, large sample sizes are often needed to in-
crease generalizability and prevent overfitting [39]. A recent
study suggests that accuracy values obtained from smaller
samples are opportunistically biased [40]. Therefore, pro-
posed biomarkers must demonstrate strong performance in
generalizable samples.
Biomarker Reliability and Feasibility of Implementation
A neuroimaging-derived biomarker cannot be reliably used
for scientific or clinical purposes if the measures composing
it do not demonstrate high test-retest reliability (i.e., the repro-
ducibility of results over time). This concern is especially
relevant given recent evidence that the test-retest reliability
of functional connectivity metrics may vary widely depending
on the brain regions examined [19•]. For this reason, the reli-
ability of neuroimaging-based measures should not be taken
for granted. This concern also applies to populations where
typical brain function may be perturbed due to injury or illness
(e.g., post-stroke [41]).
Even a biomarker that is meeting or exceeding criteria for
appropriate use as a diagnostic tool will not be clinically
useful if it cannot be readily assessed in clinic settings. As
noted by Woo and Wager [42], all parameters of the biomarker
and its associated testing procedure should be rigorously stan-
dardized so that all users collect data comparable to the stan-
dard used to derive the measure. Depending on the demands
associated with collecting certain biomarkers (especially those
derived from BOLD activity during task-based fMRI), such
rigor may be difficult to achieve in clinical settings. Often the
additional time, nuanced design, and/or special expertise re-
quired in the acquisition of reliable data will limit some bio-
markers’ applicability and usefulness.
What Constitutes Adequate Biomarker Performance?
A major issue in the application (and reporting) of ML-based
brain biomarkers for chronic pain is determining whether a
given marker’s accuracy is adequate. Developing criteria for
optimal biomarker performance depends on the setting in
which it will be used, as well as the cost of misclassification.
Pure research-oriented use cases (e.g., identifying potential
markers, phenotypic subgroups, or treatment targets; discern-
ing disease mechanisms) may allow for less stringent perfor-
mance criteria. In contrast, clinical tasks (diagnosis, prognosis,
treatment planning) carry risk of harm (misdiagnosis, delaying
treatment, providing inaccurate or unnecessary treatment) and
will therefore require considerably more stringent perfor-
mance criteria for use in patient care. Although the risk of
medical error from decisions based upon ML-derived bio-
markers may be no greater than those commonly accepted in
current practice, the “black-box” nature of these markers
raises liability concerns in the face of medical errors promul-
gated by ML-derived biomarkers [43] and may increase phy-
sicians’ reluctance to use even those that are well validated.
Ultimately, any given biomarker’s practical applicability
will depend on a case-by-case assessment of its cost,
Table 1 Basic descriptions of common supervised machine learning algorithms
Algorithm Basic approach Reference(s)
Logistic regression (LR) Uses linear combination of predictors and regression coefficients to make categorical prediction [31]
Naïve Bayes (NB) Individuals categorized according to probability that they have a certain value for each feature
independently included in prediction model
[32]
k-Nearest Neighbors (kNN) Categorizes individuals based on the most common class of the k closest observations for each
model feature
[33]
Multilayer perceptron (MLP) Determines appropriate weighting for features for multiple layers of decision units (perceptrons)
needed to perfectly classify training data, then applies learned rule to new cases
[34]
Support vector machine (SVM) Determines a linear or non-linear maximum-margin hyperplane (i.e., feature-based cutoff) that
maximizes separation between groups
[26, 27]
Decision tree (e.g., J48/C4.5) Categorizes individuals in a series of decisions based upon optimal feature cutoff values [35]
Least absolute shrinkage and
selection operator (LASSO)
A variant of ordinary least squares regression that constrains the summary of absolute regression
coefficients across features in order to identify the most informative features and optimal cut
points for classification
[36]
Curr Rheumatol Rep (2017) 19:5 Page 3 of 9 5
deployability, and accuracy in the context of its intended use.
For instance, in clinical settings, biomarker performance
should be judged by taking into consideration whether it is
intended to replace physicians in a role they can already per-
form well (e.g., differentially diagnose mechanistically and/or
symptomatically distinct conditions) or, alternatively, provide
novel information that clinicians would not otherwise have.
This approach may help identify mechanistically distinct clin-
ical subgroups, guide treatment decisions, indicate prognosis,
or separate conditions with significant mechanistic or symp-
tomatic overlap. In cases where biomarker application pro-
vides useful clinical information where it would not otherwise
be available, higher cost, more difficult deployability, and/or
relatively poorer performance may be quite tolerable [43].
Impact of Chronic Pain Prevalence on Biomarker
Performance
Ethical and practical issues surrounding potential under- or
overtreatment of pain remain even in cases where biomark-
er sensitivity (i.e., probability of a positive biomarker giv-
en an individual has the condition) and specificity (i.e.,
probability of a negative biomarker given an individual
does not have the condition) reach very high levels, de-
pending on the given use case. This is because biomarker
positive predictive value (PPV) and negative predictive
value (NPV), as previously noted [44•], depend on the
prevalence (i.e., base rate) of the condition of interest in a
particular setting (i.e., Bayes’ theorem). Specifically, bio-
marker PPV increases with prevalence, while NPV in-
creases as prevalence decreases. This relationship is illus-
trated graphically in Fig. 1 using the sensitivity (81%) and
specificity (75%) of a structural brain biomarker for pain
we have previously reported [23••]. As a result, even the
highest performing chronic pain biomarkers previously re-
ported (e.g., 92% sensitivity, 92% specificity [16•]) will
have high rates of false positives in application settings
with low prevalence and high rates of false negatives in
settings with high prevalence. Investigators need to take
use case into account when reporting biomarker perfor-
mance and judging its adequacy.
Finally, self-report of pain symptomatology is ultimately
the basis for diagnosing and assessing the severity of chronic
pain conditions (so-called “Gold Standard”). Therefore, the
predictive utility of biomarkers is necessarily limited by the
accuracy of the existing diagnostic gold standard [8•, 45]. It is
conceivable that subgroups within a diagnostic category could
be identified solely based upon physiological measurements
using unsupervised ML methods and later validated upon self-
report, thereby circumventing this issue. However, to our
knowledge, no previous neuroimaging-based biomarker stud-
ies for chronic pain have used this approach.
Pain Biomarkers Based on Brain Imaging
Structural Biomarkers
Chronic pain brain biomarkers derived from structural MRI
are dependent on the assumption that chronic pain conditions
are associated with abnormalities in brain structure that either
pre-date or are the result of pain chronification. Brain structure
can be characterized in terms of grey matter features, which
reflects integrity of neuronal cell bodies, or white matter fea-
tures that reflect axonal integrity. Note that although white
matter perturbations have been demonstrated in a variety of
chronic pain conditions [46–48], to our knowledge, ML algo-
rithms have yet to be applied to these data in order to generate
chronic pain classifiers. Thus, we will focus on studies utiliz-
ing grey matter features.
Grey matter structure can be assessed in several ways. One
common approach is the use of voxel-based morphometry
(VBM) on high-resolution T1-weighted images. VBM pro-
duces estimates of grey matter density for each voxel and
subject after warping to fit a standardized template brain [49,
50]. Another typical technique is to assess grey matter char-
acteristics in cortical (thickness, volume, surface area, or mean
curvature) or subcortical (volume) structures by assigning a
neuroanatomical label to each brain voxel using a probabilistic
Fig. 1 Illustration of the relationship between chronic pain prevalence
and the positive/negative predictive value of biomarkers using sensitivity
(81%) and specificity (75%) values derived from Robinson et al. [23••].
Positive predictive value (PPV) increases with prevalence, while negative
predictive value (NPV) decreases as prevalence increases. Thus, during
real-world application of brain biomarkers for chronic pain, performance
will depend largely on the expected proportion of individuals with the
condition in question
5 Page 4 of 9 Curr Rheumatol Rep (2017) 19:5
atlas [51, 52]. Though other techniques and measures are
available for assessing grey matter structure, efforts to con-
struct biomarkers for chronic pain conditions based on grey
matter structure have largely relied on grey matter density [29,
53] or thickness/volume/surface area/curvature [23••, 30].
Structural brain biomarkers for chronic pain build on a
significant literature demonstrating both atrophy and hy-
pertrophy in chronic pain patients in numerous pain-
related brain regions. For example, chronic low back pain
(cLBP) has been associated with lower grey matter densi-
ty compared to controls in the dorsolateral prefrontal cor-
tex (DLPFC), a region associated with cognitive/
evaluative function and pain modulation [54]. At the same
time, FM has been associated with both increased grey
matter volume in striatum, orbitofrontal cortex, and cere-
bellum [55], and lower volume in anterior cingulate cor-
tex, amygdala, thalamus, superior temporal gyrus, supple-
mentary motor cortex, and insula [55–58]. Although not
directly contradicting one another, these studies differed
somewhat with regard to regions showing significant dif-
ferences between FM patients and controls, with differ-
ences in anterior cingulate cortex being the most consis-
tently observed [56, 58]. The potential utility of these
measures as features for classification algorithms to dis-
tinguish chronic pain patient groups depends on the as-
sumption that reliable commonalities and differences in
grey matter morphometrics can be detected between
chronic pain conditions. It is also worth noting that the
goal of chronic pain biomarker studies is distinct from
between-group analyses of structural differences because
they are focused specifically on identifying optimal com-
binations of features that best separate patient groups and/
or healthy controls.
Classifier Studies Based on Structural Brain
Abnormalities
ML classifier studies based on structural brain features
have been conducted in several chronic pain conditions,
including CPP, IBS, FM, and cLBP, using samples from
26 to 160 participants. In each case, samples were com-
posed of ∼50% splits of patients to controls. Performance
of these potential biomarkers has differed substantially be-
tween studies, with sensitivity and specificity ranging from
65 to 81%. Although brain regions discriminating patient
groups from normal controls varied, certain areas were
more frequently reported. These included precentral gyrus
(primary motor cortex), postcentral gyrus (primary so-
matosensory cortex), amygdala, and cuneus. In addition,
many discriminating regions were convergent with those
identified in previous studies focused on identifying struc-
tural differences between chronic pain patients and con-
trols (e.g., DLPFC, amygdala, cingulate cortex, insula,
etc.,). For further detail regarding sample size, algorithm(s)
used, classifier performance, and brain structures with the
greatest contribution to performance for studies using
structural brain biomarkers, see Table 2.
Functional Brain Biomarkers
Whereas, high-resolution neuroanatomical data is the basis
for studies proposing structural neuroimaging biomarkers
of chronic pain, brain activity measures are used to derive
functional neuroimaging biomarkers. Functional MRI
(fMRI), which refers to noninvasive methods for measur-
ing brain function, has two common variants. The first
detects changes in cerebral blood oxygenation (BOLD),
and the second, arterial spin labeling (ASL), measures
changes in regional cerebral blood flow (rCBF).
Although neither of these methods directly measure neuro-
nal activity, they are both considered surrogate markers of
such activity (for review of these technologies, see [61]).
These methods excel in their ability to provide spatial in-
formation about where changes in BOLD response or
rCBF occur over the course of time; however, they are
intrinsically limited in temporal resolution, or accuracy in
determining exactly when the neuronal activation oc-
curred, due to the physiological characteristics of blood
oxygenation (BOLD) and/or extended repetition times
(ASL; [62]). Although ASL has several theoretical advan-
tages over BOLD regarding the measurement of brain
function [63], most brain classification studies for chronic
pain to date have relied on BOLD fMRI.
Because functional neuroimaging provides a global mea-
sure of brain activity, participants’ mental state during data
collection is always important to note. For example, brain
activity can be captured during a goal-directed task (e.g.,
participants are asked to continuously rate their levels of
clinical pain or undergo acute painful stimulation), or dur-
ing wakeful rest (i.e., resting state) in which participants are
not instructed to engage in a particular task. Raw data col-
lected during these scans typically undergoes preprocess-
ing, or various image and signal correction techniques, to
improve the ratio of desired signal to undesired noise [64].
Subsequently, preprocessed data are statistically analyzed at
the individual-participant level to determine either 1)
whether a significant change in signal occurred for a given
region or across the whole brain (i.e., activation), or 2) the
coherence in activation among spatially distinct brain re-
gions over time (i.e., functional connectivity, FC).
Statistically analyzed activation or FC values can be select-
ed across the whole brain, or limited to hypothesis-driven
regions of interest (i.e., a priori ROIs). In classification stud-
ies proposing functional neuroimaging biomarkers,
individual-level activation or FC information is then
Curr Rheumatol Rep (2017) 19:5 Page 5 of 9 5
Table2Characteristicsofmachinelearning-basedclassifierstudiesusingbrainimagingresults
StudyPain
condition
Accuracy
(%)
Sensitivity
(%)
Specificity
(%)
ManuscriptNManuscript
baserate(%)
Machine
learning
classifier
Aprioriregions
ofinterest?
Majorregionscontributingtoclassification
Structuralbiomarkers
Bagarinaoetal.[28]CPP7370736650SVMNo>DensityinLSFG,LPCG,RPCG,LPaHcG,R
PaHcG,amygdala,hippocampusinCPP
Labusetal.[30]IBS70657516050sPLS-DANo>ThicknessinLPCG,lesserthicknessinR
insula;>meancurvatureinRPreCG,lesserinL
SCG,Rtemporalsulcus,andLintraparietal
sulcus;and<surfaceareainLfusiformgyrus
inIBS
Robinsonetal.,[23••]FM7681752654LR,MLP,
Bayes,
SVM,J48
No<VolumeinLamygdalainFM
Ungetal.[29]cLBP7676759450SVMNo<GMdensityinRamygdala,LMOG,andR
cuneus;greaterGMdensityinRcerebellum,
R/Lmiddle/superiortemporalgyrus,LPreCG,
LPCG,Rcalcarinesulcus,andRDLPFCincLBP
Dynamicbiomarkers
Callanetal.[16•]cLBP9292922650SLRYes<LPCGactivationand>LIPCincLBPduringpain
Sundermannetal.[59]RA
FM
79(vs.HC)
62(vs.HC)
79(vs.RA)
NRNR5034
32
SVM
SVM
kNN
YesNR
López-Solàetal.[60••]FM9392947251SVM,LRYes>Activityininsula,PCC,precuneus,DMPFC,
parahippocampalgyrus
<ActivityinDLPFC,primary/secondaryvisual
andauditorycortex,lateralcerebellum,basal
ganglia,andhypothalamus
SFGsuperiorfrontalgyrus,PCGpostcentralgyrus,PaHcGparahippocampalgyrus,PreCGprecentralgyrus,SCGsubcollosalgyrus,MOGmedialorbitalgyrus,DMPFCdorsomedialprefrontalcortex,
DLPFCdorsolateralprefrontalcortex,IPCinferiorparietalcortex,RArheumatoidarthritis,FMfibromyalgia,CFSchronicfatiguesyndrome,FMfibromyalgia,IBSirritablebowelsyndrome,cLBPchronic
lowbackpain,LASSOleastabsoluteshrinkageandselectionoperator,OAosteoarthritis,CPPchronicpelvicpain,LRlogisticregressionusingridgeestimator,MLPmultilayerperceptron,J48J48decision
tree,PPVpositivepredictivevalue,NPVnegativepredictivevalue,SLRsparselogisticregression,sPLS-DAsparsepartialleastsquaresfordiscriminantanalysis,IPCinferiorparietalcortex,NRnot
reported
5 Page 6 of 9 Curr Rheumatol Rep (2017) 19:5
entered into computational models, which classify individ-
uals into distinct groups based on these values.
Classifier Studies Based on Functional Brain
Abnormalities
Compared to structural brain abnormalities, fewer studies
have used fMRI activation or functional connectivity metrics
as features in classification models for chronic pain patients.
Using measures of functional brain activation in pain-related
brain regions during repeated 14-s blocks of experimental
electrical pain induction, Callan et al. [16•] achieved 92%
accuracy, sensitivity, and specificity classifying cLBP and
healthy controls. The two most informative regions reported
by Callan et al. [16•] have been strongly implicated in pain
discrimination: (primary somatosensory cortex) and attention
(inferior parietal cortex) [65]. In a separate study using resting
state fMRI [59], RA and FM patients could be discriminated
from HC with 79 and 62% accuracy, respectively, using mea-
sures of functional connectivity between structures associated
with stimulus evaluation (i.e., the “salience network” [SN])
and internal focus/mind wandering (i.e., “default mode net-
work” [DMN]). These networks have been implicated in pain
processing (SN) or reported as perturbed in chronic pain states
(DMN). Interestingly, RA and FM patients could also be dis-
criminated from each other with 79% accuracy, suggesting
that, despite potential commonalities in neuronal plasticity,
the functional neural correlates of certain chronic pain disor-
ders may be sufficiently distinct to enable their separation
[59]. Finally, in a recent fMRI study using ML [60••], the
investigators could discriminate FM patients from HC with
93% accuracy based on their brain activation associated with
painful pressure stimuli [66] and non-painful multisensory
stimulation (e.g., simultaneous auditory, tactile, and visual
stimuli). Overall, the rapid improvement of fMRI-based “pain
signatures” has not only helped our mechanistic understand-
ing of chronic pain but will also benefit the diagnosis of FM
and other chronic pain conditions.
Summary
Taken together, a limited number of studies (see Table 1) have
tested the ability of ML algorithms to discern chronic pain
patients from healthy controls using structural or functional
brain abnormalities. However, interest in ML approaches for
chronic pain diagnosis and classification is growing due to
their purported potential to help elucidate chronic pain mech-
anisms, identify resilient or vulnerable subgroups, improve
clinical decision making, predict treatment outcome, or aug-
ment [29] or even replace self-report (for critical discussion,
see [8•]). However, as previously discussed, certain caveats
apply to the practical application of biomarkers even where
performance metrics may be very high.
Conclusions
Interest in the development of clinical and ML-based bio-
markers for chronic musculoskeletal pain conditions derived
from structural and functional neuroimages has increased sub-
stantially in recent years. Current reports describe novel bio-
markers capable of separating patient groups and healthy con-
trols with accuracies ranging from 70 to 93%. Such studies
provide valuable mechanistic information regarding both the
unique and common neural correlates of these conditions,
with the potential to highlight differences between both mus-
culoskeletal pain patient groups and controls to which tradi-
tional statistical approaches may not be sensitive, or identify
mechanistic subgroups within certain pain conditions.
However, at this time, critical theoretical, practical, and ethical
concerns preclude the replacement of patient self-report for
the diagnosis of chronic musculoskeletal pain with brain
imaging-derived biomarkers, as self-report remains the gold
standard for pain assessment [8•].
Acknowledgements This work was supported by NIH grant R01
NR014049-01
Compliance with Ethical Standards
Conflict of Interest The authors declare that they have no conflict of
interest.
Human and Animal Rights and Informed Consent This article does
not contain any studies with human or animal subjects performed by any
of the authors.
References
Papers of particular interest, published recently, have been
highlighted as:
• Of importance
•• Of major importance
1. Institute of Medicine. Relieving pain in America: a blueprint for
transforming prevention, care, education, and research.
Washington, DC: National Academies Press; 2011.
2. Wolfe F, Clauw DJ, Fitzcharles MA, Goldenberg DL, Katz RS,
Mease P, et al. The American College of Rheumatology preliminary
diagnostic criteria for fibromyalgia and measurement of symptom
severity. Arthritis Care Res. 2010;62(5):600–10. doi:10.1002
/acr.20140.
3. Shamir L, Ling SM, Scott Jr WW, Bos A, Orlov N, Macura TJ,
et al. Knee x-ray image analysis method for automated detection of
osteoarthritis. IEEE Trans Biomed Eng. 2009;56(2):407–15.
doi:10.1109/TBME.2008.2006025.
4. Kirkwood RN, Resende RA, Magalhaes CM, Gomes HA, Mingoti
SA, Sampaio RF. Application of principal component analysis on
gait kinematics in elderly women with knee osteoarthritis. Rev Bras
Fis. 2011;15(1):52–8.
Curr Rheumatol Rep (2017) 19:5 Page 7 of 9 5
5. Frize M, Ogungbemile A. Estimating rheumatoid arthritis activity
with infrared image analysis. Stud Health Technol Inform.
2012;180:594–8.
6. Frampton D, Kerr J, Harrison TJ, Kellam P. Assessment of a 44
gene classifier for the evaluation of chronic fatigue syndrome from
peripheral blood mononuclear cell gene expression. PLoS One.
2011;6(3):e16872. doi:10.1371/journal.pone.0016872.
7. Apkarian AV, Hashmi JA, Baliki MN. Pain and the brain: specific-
ity and plasticity of the brain in clinical chronic pain. Pain.
2011;152(3 Suppl):S49–64. doi:10.1016/j.pain.2010.11.010.
8.• Robinson ME, Staud R, Price DD. Pain measurement and brain
activity: will neuroimages replace pain ratings? J Pain: Off J Am
Pain Soc. 2013;14(4):323–7. doi:10.1016/j.jpain.2012.05.007.
This critical review summarized the limitations of
neuroimaging to replace pain self-report. It also emphasized
that neuroimaging will never replace pain self-report except
in individuals with mental conditions or limited levels of
consciousness.
9. Wortolowska K. How neuroimaging can help us to visualise and
quantify pain? Eur J Pain Suppl. 2011;5(S2):323–7.
10. Bushnell MC, Ceko M, Low LA. Cognitive and emotional control
of pain and its disruption in chronic pain. Nat Rev Neurosci.
2013;14(7):502–11. doi:10.1038/nrn3516.
11. Brekelmans MP, Fens N, Brinkman P, Bos LD, Sterk PJ, Tak PP,
et al. Smelling the diagnosis: the electronic nose as diagnostic tool
in inflammatory arthritis. A case-reference study. PloS One.
2016;11(3):e0151715. doi:10.1371/journal.pone.0151715.
12. Rizzo G, Raffeiner B, Coran A, Ciprian L, Fiocco U, Botsios C,
et al. Pixel-based approach to assess contrast-enhanced ultrasound
kinetics parameters for differential diagnosis of rheumatoid arthritis.
J Med Imaging (Bellingham). 2015;2(3):034503. doi:10.1117/1.
JMI.2.3.034503.
13. Mackey SC. Central neuroimaging of pain. J Pain: Off J Am Pain
Soc. 2013;14(4):328–31. doi:10.1016/j.jpain.2013.01.001.
14. Borsook D, Becerra L, Hargreaves R. Biomarkers for chronic pain
and analgesia. Part 1: the need, reality, challenges, and solutions.
Discov Med. 2011;11(58):197–207.
15. Borsook D, Becerra L, Hargreaves R. Biomarkers for chronic pain
and analgesia. Part 2: how, where, and what to look for using func-
tional imaging. Discov Med. 2011;11(58):209–19.
16.• Callan D, Mills L, Nott C, England R, England S. A tool for clas-
sifying individuals with chronic back pain: using multivariate pat-
tern analysis with functional magnetic resonance imaging data.
PloS One. 2014;9(6):e98007. doi:10.1371/journal.pone.0098007.
This study employed supervised machine learning techniques,
specifically sparse logistic regression, to train a classifier of
chronic back pain based on contrast images using a leave-one-
out cross-validation procedure. It correctly classified 92.3% of
the chronic pain group and 92.3% of the normal controls.
17. Lu HC, Hsieh JC, Lu CL, Niddam DM, Wu YT, Yeh TC, et al.
Neuronal correlates in the modulation of placebo analgesia in
experimentally-induced esophageal pain: a 3T-fMRI study. Pain.
2010;148(1):75–83. doi:10.1016/j.pain.2009.10.012.
18. Pustilnik AC. Imaging brains, changing minds: how pain neuroim-
aging can inform the law. Alabama Law Rev. 2015;66(5).
19.• Letzen JE, Boissoneault J, Sevel LS, Robinson ME. Test-retest
reliability of pain-related functional brain connectivity compared
to pain self-report. Pain. 2016;157(3):546–51. doi:10.1097/j.
pain.0000000000000356. This study examined the test-retest re-
liability for functional connectivity MRI (fcMRI) of pain-
related brain regions and self-reported pain. Intraclass corre-
lations coefficients for fcMRI values varied widely
(range = −.174–.766), whereas intraclass correlations coeffi-
cients for VAS scores ranged from .906 to .947. Overall, self-
reported pain was more reliable than fcMRI data.
20. Nielsen CS, Price DD, Vassend O, Stubhaug A, Harris JR.
Characterizing individual differences in heat-pain sensitivity.
Pain. 2005;119(1–3):65–74. doi:10.1016/j.pain.2005.09.018.
21. Slesinger D, Archer RP, Duane W. MMPI-2 characteristics in a
chronic pain population. Assessment. 2002;9(4):406–14.
22. Takekawa KS, Goncalves JS, Moriguchi CS, Coury HJ, Sato TO.
Can a self-administered questionnaire identify workers with chronic
or recurring low back pain? Ind Health. 2015;53(4):340–5.
doi:10.2486/indhealth.2014-0241.
23.•• Robinson ME, O’Shea AM, Craggs JG, Price DD, Letzen JE, Staud
R. Comparison of machine classification algorithms for fibromyal-
gia: neuroimages versus self-report. J Pain: Off J Am Pain Soc.
2015;16(5):472–7. doi:10.1016/j.jpain.2015.02.002. Separate
models representing brain volumes, mood ratings, and pain
intensity ratings were estimated across several ML
algorithms. Structural magnetic resonance imaging data from
fibromyalgia patients and healthy controls and self-report data
of pain intensity and mood were used. Classification accuracy
of brain volumes ranged from 53 to 76%, whereas mood and
pain intensity ratings ranged from 79 to 96% and 83 to 96%,
respectively. Overall, models derived from self-report data
outperformed neuroimaging models.
24. Friend R, Bennett RM. Distinguishing fibromyalgia from rheuma-
toid arthritis and systemic lupus in clinical questionnaires: an anal-
ysis of the revised Fibromyalgia Impact Questionnaire (FIQR) and
its variant, the Symptom Impact Questionnaire (SIQR), along with
pain locations. Arthritis Res Ther. 2011;13(2):R58. doi:10.1186
/ar3311.
25. Perrot S, Bouhassira D, Fermanian J, Cercle d’Etude de la Douleur
en R. Development and validation of the Fibromyalgia Rapid
Screening Tool (FiRST). Pain. 2010;150(2):250–6. doi:10.1016/j.
pain.2010.03.034.
26. Cortes C, Vapnik V. Support-vector networks. Mach Learn.
1995;20(3):273–97.
27. Pontil M, Verri A. Properties of support vector machines. Neural
Comput. 1998;10(4):955–74.
28. Bagarinao E, Johnson KA, Martucci KT, Ichesco E, Farmer MA,
Labus J, et al. Preliminary structural MRI based brain classification
of chronic pelvic pain: a MAPP network study. Pain. 2014;155(12):
2502–9. doi:10.1016/j.pain.2014.09.002.
29. Ung H, Brown JE, Johnson KA, Younger J, Hush J, Mackey S.
Multivariate classification of structural MRI data detects chronic
low back pain. Cereb Cortex. 2014;24(4):1037–44. doi:10.1093
/cercor/bhs378.
30. Labus JS, Van Horn JD, Gupta A, Alaverdyan M, Torgerson C,
Ashe-McNalley C, et al. Multivariate morphological brain signa-
tures predict patients with chronic abdominal pain from healthy
control subjects. Pain. 2015;156(8):1545–54. doi:10.1097/j.
pain.0000000000000196.
31. Le Cessie S, Van Houwelingen JC. Ridge estimators in logistic
regression. Appl Stat. 1992;41(1):191–201.
32. Langley P, Iba W, Thompson K. An analysis of Bayesian classifiers.
AAAI. 1992;90:223–8.
33. Aha DW, Kibler D, Albert MK. Instance-based learning algorithms.
Mach Learn. 1991;6(1):37–66. doi:10.1023/A:1022689900470.
34. Arora R. Comparative analysis of classification algorithms on differ-
ent datasets using WEKA. Int J Comput Appl. 2012;54(13):21–5.
35. Quinlan JR. C4.5 programs for machine learning. San Mateo:
Morgan Kaufmann; 1992.
36. Tibshirani R. Regression shrinkage and selection via the lasso. J
Roy Stat Soc B Met. 1996;58(1):267–88.
37. Kononenko I. Machine learning for medical diagnosis: history, state
of the art and perspective. Artif Intell Med. 2001;23(1):89–109.
38. Nair SS, French RM, Laroche D, Thomas E. The application of
machine learning algorithms to the analysis of electromyographic
5 Page 8 of 9 Curr Rheumatol Rep (2017) 19:5
patterns from arthritic patients. IEEE Trans Neural Syst Rehabil
Eng. 2010;18(2):174–84. doi:10.1109/TNSRE.2009.2032638.
39. Mamoshina P, Vieira A, Putin E, Zhavoronkov A. Applications of
deep learning in biomedicine. Mol Pharm. 2016;13(5):1445–54.
doi:10.1021/acs.molpharmaceut.5b00982.
40. Schnack HG, Kahn RS. Detecting neuroimaging biomarkers for
psychiatric disorders: sample size matters. Front Psych. 2016;7:
50. doi:10.3389/fpsyt.2016.00050.
41. Siegel JS, Ramsey LE, Snyder AZ, Metcalf NV, Chacko RV,
Weinberger K, et al. Disruptions of network connectivity predict
impairment in multiple behavioral domains after stroke. Proc Natl
Acad Sci U S A. 2016. doi:10.1073/pnas.1521083113.
42. Woo CW, Wager TD. Neuroimaging-based biomarker discovery
and validation. Pain. 2015;156(8):1379–81. doi:10.1097/j.
pain.0000000000000223.
43. Deo RC. Machine learning in medicine. Circulation. 2015;132(20):
1920–30. doi:10.1161/CIRCULATIONAHA.115.001593.
44.• Robinson M, Boissoneault J, Sevel L, Letzen J, Staud R. The effect
of base rate on the predictive value of brain biomarkers. J Pain.
2016;17(6):637–41. doi:10.1016/j.jpain.2016.01.476. Results of
this study strongly suggest that many proposed brain
biomarkers perform quite poorly when realistic illness
prevalence base rates are taken into account.
45. Lueken U, Zierhut KC, Hahn T, Straube B, Kircher T, Reif A, et al.
Neurobiological markers predicting treatment response in anxiety
disorders: a systematic review and implications for clinical applica-
tion. Neurosci Biobehav Rev. 2016;66:143–62. doi:10.1016/j.
neubiorev.2016.04.005.
46. Buckalew N, Haut MW, Aizenstein H, Morrow L, Perera S,
Kuwabara H, et al. Differences in brain structure and function in
older adults with self-reported disabling and nondisabling chronic
low back pain. Pain Med. 2010;11(8):1183–97. doi:10.1111
/j.1526-4637.2010.00899.x.
47. Geha PY, Baliki MN, Harden RN, Bauer WR, Parrish TB, Apkarian
AV. The brain in chronic CRPS pain: abnormal gray-white matter
interactions in emotional and autonomic regions. Neuron.
2008;60(4):570–81. doi:10.1016/j.neuron.2008.08.022.
48. Gustin SM, Peck CC, Cheney LB, Macey PM, Murray GM,
Henderson LA. Pain and plasticity: is chronic pain always associ-
ated with somatosensory cortex activity and reorganization? J
Neurosci: Off J Soc Neurosci. 2012;32(43):14874–84.
doi:10.1523/JNEUROSCI.1733-12.2012.
49. Greve DN. An absolute beginner’s guide to surface- and voxel-
based morphometric analysis. ISMRM 19th Annual Meeting &
Exhibition. 2011;7–13.
50. Ashburner J, Friston K. Morphometry. In: Ashburner J, editor.
Human brain function. 2nd ed. Cambridge: Academic; 2004. p.
707–22.
51. Fischl B, Liu A, Dale AM. Automated manifold surgery: construct-
ing geometrically accurate and topologically correct models of the
human cerebral cortex. IEEE Trans Med Imaging. 2001;20(1):70–
80. doi:10.1109/42.906426.
52. Fischl B, Salat DH, Busa E, Albert M, Dieterich M, Haselgrove C,
et al. Whole brain segmentation: automated labeling of neuroana-
tomical structures in the human brain. Neuron. 2002;33(3):341–55.
53. Baliki MN, Schnitzer TJ, Bauer WR, Apkarian AV. Brain morpho-
logical signatures for chronic pain. PLoS One. 2011;6(10):e26010.
doi:10.1371/journal.pone.0026010.
54. Apkarian AV, Sosa Y, Sonty S, Levy RM, Harden RN, Parrish TB,
et al. Chronic back pain is associated with decreased prefrontal and
thalamic gray matter density. J Neurosci: Off J Soc Neurosci.
2004;24(46):10410–5. doi:10.1523/JNEUROSCI.2541-04.2004.
55. Schmidt-Wilcke T, Luerding R, Weigand T, Jurgens T, Schuierer G,
Leinisch E, et al. Striatal grey matter increase in patients suffering
from fibromyalgia—a voxel-based morphometry study. Pain.
2007;132 Suppl 1:S109–16. doi:10.1016/j.pain.2007.05.010.
56. Burgmer M, Gaubitz M, Konrad C, Wrenger M, Hilgart S, Heuft G,
et al. Decreased gray matter volumes in the cingulo-frontal cortex
and the amygdala in patients with fibromyalgia. Psychosom Med.
2009;71(5):566–73. doi:10.1097/PSY.0b013e3181a32da0.
57. Puri BK, Agour M, Gunatilake KD, Fernando KA, Gurusinghe AI,
Treasaden IH. Reduction in left supplementary motor area grey
matter in adult female fibromyalgia sufferers with marked fatigue
and without affective disorder: a pilot controlled 3-T magnetic res-
onance imaging voxel-based morphometry study. J Int Med Res.
2010;38(4):1468–72.
58. Robinson ME, Craggs JG, Price DD, Perlstein WM, Staud R. Gray
matter volumes of pain-related brain areas are decreased in fibro-
myalgia syndrome. J Pain: Off J Am Pain Soc. 2011;12(4):436–43.
doi:10.1016/j.jpain.2010.10.003.
59. Sundermann B, Burgmer M, Pogatzki-Zahn E, Gaubitz M, Stuber
C, Wessolleck E, et al. Diagnostic classification based on functional
connectivity in chronic pain: model optimization in fibromyalgia
and rheumatoid arthritis. Acad Radiol. 2014;21(3):369–77.
doi:10.1016/j.acra.2013.12.003.
60.•• López-Solà M, Woo CW, Pujol J, Deus J, Harrison BJ, Monfort J,
Wager TD. Towards a neurophysiological signature for fibromyalgia.
Pain. 2016. A ‘Multisensory’ classifier trained on non-painful sen-
sory stimulation revealed a brain signature for FM with high
sensitivity and specificity. This study characterized individual
FM patients based on symptom-related brain features.
61. Detre JA, Wang J. Technical aspects and utility of fMRI using
BOLD and ASL. Clin Neurophysiol. 2002;113(5):621–34.
62. Howseman AM, Bowtell RW. Functional magnetic resonance im-
aging: imaging techniques and contrast mechanisms. Philos Trans
R Soc Lond B Biol Sci. 1999;354(1387):1179–94. doi:10.1098
/rstb.1999.0473.
63. Chen JJ, Jann K, Wang DJ. Characterizing resting-state brain func-
tion using arterial spin labeling. Brain Connect. 2015;5(9):527–42.
doi:10.1089/brain.2015.0344.
64. Smith SM, Jenkinson M, Woolrich MW, Beckmann CF, Behrens
TE, Johansen-Berg H, et al. Advances in functional and structural
MR image analysis and implementation as FSL. NeuroImage.
2004;23 Suppl 1:S208–19. doi:10.1016/j.neuroimage.2004.07.051.
65. Giesecke T, Gracely RH, Grant MA, Nachemson A, Petzke F,
Williams DA, et al. Evidence of augmented central pain processing
in idiopathic chronic low back pain. Arthritis Rheum. 2004;50(2):
613–23. doi:10.1002/art.20063.
66. Wager TD, Atlas LY, Lindquist MA, Roy M, Woo CW, Kross E. An
fMRI-based neurologic signature of physical pain. N Engl J Med.
2013;368(15):1388–97.
Curr Rheumatol Rep (2017) 19:5 Page 9 of 9 5

More Related Content

What's hot

Instrument-Assisted Soft-Tissue Mobilization
Instrument-Assisted Soft-Tissue MobilizationInstrument-Assisted Soft-Tissue Mobilization
Instrument-Assisted Soft-Tissue MobilizationErnad Fejzić
 
‘Neurodynamics as a therapeutic intervention; the effectiveness and scientifi...
‘Neurodynamics as a therapeutic intervention; the effectiveness and scientifi...‘Neurodynamics as a therapeutic intervention; the effectiveness and scientifi...
‘Neurodynamics as a therapeutic intervention; the effectiveness and scientifi...NVMT-symposium
 
Lecture References and online resources: Tendons: Mechanisms of pathogenicity...
Lecture References and online resources: Tendons: Mechanisms of pathogenicity...Lecture References and online resources: Tendons: Mechanisms of pathogenicity...
Lecture References and online resources: Tendons: Mechanisms of pathogenicity...Mark Sexton
 
IWTC in Portland, USA 2006
IWTC in Portland, USA 2006IWTC in Portland, USA 2006
IWTC in Portland, USA 2006laurenharding
 
Exercise In Rhuematology Lecture
Exercise In Rhuematology LectureExercise In Rhuematology Lecture
Exercise In Rhuematology Lecturedrmomusa
 
PMR Buzz Volume 4.2021
PMR Buzz Volume 4.2021PMR Buzz Volume 4.2021
PMR Buzz Volume 4.2021mrinal joshi
 
BMC-MSK-Coppack et al-2016
BMC-MSK-Coppack et al-2016BMC-MSK-Coppack et al-2016
BMC-MSK-Coppack et al-2016Russ Coppack MBE
 
MSK HQ outcomes from patients attending cognitive behavioural and exercise i...
MSK HQ  outcomes from patients attending cognitive behavioural and exercise i...MSK HQ  outcomes from patients attending cognitive behavioural and exercise i...
MSK HQ outcomes from patients attending cognitive behavioural and exercise i...Andrew Bateman
 
Foot-Ankle-Research-Review-Issue-27
Foot-Ankle-Research-Review-Issue-27Foot-Ankle-Research-Review-Issue-27
Foot-Ankle-Research-Review-Issue-27Dr Matthew Carroll
 
ZMPCZM016000.11.04
ZMPCZM016000.11.04 ZMPCZM016000.11.04
ZMPCZM016000.11.04 painezeeman
 

What's hot (20)

Instrument-Assisted Soft-Tissue Mobilization
Instrument-Assisted Soft-Tissue MobilizationInstrument-Assisted Soft-Tissue Mobilization
Instrument-Assisted Soft-Tissue Mobilization
 
IASTM
IASTMIASTM
IASTM
 
‘Neurodynamics as a therapeutic intervention; the effectiveness and scientifi...
‘Neurodynamics as a therapeutic intervention; the effectiveness and scientifi...‘Neurodynamics as a therapeutic intervention; the effectiveness and scientifi...
‘Neurodynamics as a therapeutic intervention; the effectiveness and scientifi...
 
Lecture References and online resources: Tendons: Mechanisms of pathogenicity...
Lecture References and online resources: Tendons: Mechanisms of pathogenicity...Lecture References and online resources: Tendons: Mechanisms of pathogenicity...
Lecture References and online resources: Tendons: Mechanisms of pathogenicity...
 
watts et al 2016
watts et al 2016watts et al 2016
watts et al 2016
 
IWTC in Portland, USA 2006
IWTC in Portland, USA 2006IWTC in Portland, USA 2006
IWTC in Portland, USA 2006
 
Exercise In Rhuematology Lecture
Exercise In Rhuematology LectureExercise In Rhuematology Lecture
Exercise In Rhuematology Lecture
 
Coccydynia
Coccydynia Coccydynia
Coccydynia
 
UROP Poster
UROP PosterUROP Poster
UROP Poster
 
PMR Buzz Volume 4.2021
PMR Buzz Volume 4.2021PMR Buzz Volume 4.2021
PMR Buzz Volume 4.2021
 
Ricardo santos poster bnm
Ricardo santos poster bnmRicardo santos poster bnm
Ricardo santos poster bnm
 
BMC-MSK-Coppack et al-2016
BMC-MSK-Coppack et al-2016BMC-MSK-Coppack et al-2016
BMC-MSK-Coppack et al-2016
 
MSK HQ outcomes from patients attending cognitive behavioural and exercise i...
MSK HQ  outcomes from patients attending cognitive behavioural and exercise i...MSK HQ  outcomes from patients attending cognitive behavioural and exercise i...
MSK HQ outcomes from patients attending cognitive behavioural and exercise i...
 
1
11
1
 
Foot-Ankle-Research-Review-Issue-27
Foot-Ankle-Research-Review-Issue-27Foot-Ankle-Research-Review-Issue-27
Foot-Ankle-Research-Review-Issue-27
 
Spinal Manipulative Therapy
Spinal Manipulative TherapySpinal Manipulative Therapy
Spinal Manipulative Therapy
 
CURRICULUM VITAE
CURRICULUM  VITAECURRICULUM  VITAE
CURRICULUM VITAE
 
Banu
BanuBanu
Banu
 
Lit Review
Lit ReviewLit Review
Lit Review
 
ZMPCZM016000.11.04
ZMPCZM016000.11.04 ZMPCZM016000.11.04
ZMPCZM016000.11.04
 

Viewers also liked

ICs, birth certificates, Degree scroll and transcript
ICs, birth certificates, Degree scroll and transcriptICs, birth certificates, Degree scroll and transcript
ICs, birth certificates, Degree scroll and transcriptMOHAMAD HANAFI MOHD YATIM
 
Berbagai sumber belajar di sekitar alam lingkungan sekolah 6
Berbagai sumber belajar di sekitar alam lingkungan sekolah 6Berbagai sumber belajar di sekitar alam lingkungan sekolah 6
Berbagai sumber belajar di sekitar alam lingkungan sekolah 6Ismail Fizh
 
Holanda en américa
Holanda en américaHolanda en américa
Holanda en américaMónica Cano
 
Rpp supervisi logaritma sma it assalam
Rpp supervisi logaritma sma it assalamRpp supervisi logaritma sma it assalam
Rpp supervisi logaritma sma it assalamArikha Nida
 
Keselamatan pasien Radiologi
Keselamatan pasien RadiologiKeselamatan pasien Radiologi
Keselamatan pasien RadiologiDwi Adhianto
 
Peraturan menteri-pendidikan-nasional-nomor-24-tahun-2007-tentang-standar-sar...
Peraturan menteri-pendidikan-nasional-nomor-24-tahun-2007-tentang-standar-sar...Peraturan menteri-pendidikan-nasional-nomor-24-tahun-2007-tentang-standar-sar...
Peraturan menteri-pendidikan-nasional-nomor-24-tahun-2007-tentang-standar-sar...Miftahudin Hungkul
 
Digital Class Project - Mountain Dew Digital Plan
Digital Class Project - Mountain Dew Digital Plan Digital Class Project - Mountain Dew Digital Plan
Digital Class Project - Mountain Dew Digital Plan Nghia Le Trong
 
Young marketers 5 - MATCHA
Young marketers 5 - MATCHA Young marketers 5 - MATCHA
Young marketers 5 - MATCHA Nghia Le Trong
 
Nociceptors the sensors of the pain pathway
Nociceptors the sensors of the pain pathway   Nociceptors the sensors of the pain pathway
Nociceptors the sensors of the pain pathway Asmae LGUENSAT
 
CMO 2015 - Moppy Fish Team
CMO 2015 - Moppy Fish Team CMO 2015 - Moppy Fish Team
CMO 2015 - Moppy Fish Team Nghia Le Trong
 
The Top Leadership Qualities Every Manager Needs
The Top Leadership Qualities Every Manager NeedsThe Top Leadership Qualities Every Manager Needs
The Top Leadership Qualities Every Manager NeedsWhen I Work
 
Tututututut 2
Tututututut 2Tututututut 2
Tututututut 2andrecyn
 
Devry card 405 week 4 portfolio draft
Devry card 405 week 4 portfolio draftDevry card 405 week 4 portfolio draft
Devry card 405 week 4 portfolio drafteddie hahn
 
Cociñamos
CociñamosCociñamos
Cociñamoscorgomo
 

Viewers also liked (17)

ICs, birth certificates, Degree scroll and transcript
ICs, birth certificates, Degree scroll and transcriptICs, birth certificates, Degree scroll and transcript
ICs, birth certificates, Degree scroll and transcript
 
Soal logaritma
Soal logaritmaSoal logaritma
Soal logaritma
 
Berbagai sumber belajar di sekitar alam lingkungan sekolah 6
Berbagai sumber belajar di sekitar alam lingkungan sekolah 6Berbagai sumber belajar di sekitar alam lingkungan sekolah 6
Berbagai sumber belajar di sekitar alam lingkungan sekolah 6
 
Holanda en américa
Holanda en américaHolanda en américa
Holanda en américa
 
Rpp supervisi logaritma sma it assalam
Rpp supervisi logaritma sma it assalamRpp supervisi logaritma sma it assalam
Rpp supervisi logaritma sma it assalam
 
Keselamatan pasien Radiologi
Keselamatan pasien RadiologiKeselamatan pasien Radiologi
Keselamatan pasien Radiologi
 
Peraturan menteri-pendidikan-nasional-nomor-24-tahun-2007-tentang-standar-sar...
Peraturan menteri-pendidikan-nasional-nomor-24-tahun-2007-tentang-standar-sar...Peraturan menteri-pendidikan-nasional-nomor-24-tahun-2007-tentang-standar-sar...
Peraturan menteri-pendidikan-nasional-nomor-24-tahun-2007-tentang-standar-sar...
 
Digital Class Project - Mountain Dew Digital Plan
Digital Class Project - Mountain Dew Digital Plan Digital Class Project - Mountain Dew Digital Plan
Digital Class Project - Mountain Dew Digital Plan
 
Young marketers 5 - MATCHA
Young marketers 5 - MATCHA Young marketers 5 - MATCHA
Young marketers 5 - MATCHA
 
Nociceptors the sensors of the pain pathway
Nociceptors the sensors of the pain pathway   Nociceptors the sensors of the pain pathway
Nociceptors the sensors of the pain pathway
 
CMO 2015 - Moppy Fish Team
CMO 2015 - Moppy Fish Team CMO 2015 - Moppy Fish Team
CMO 2015 - Moppy Fish Team
 
The Top Leadership Qualities Every Manager Needs
The Top Leadership Qualities Every Manager NeedsThe Top Leadership Qualities Every Manager Needs
The Top Leadership Qualities Every Manager Needs
 
High Dose Initiation
High Dose InitiationHigh Dose Initiation
High Dose Initiation
 
Slideshare
SlideshareSlideshare
Slideshare
 
Tututututut 2
Tututututut 2Tututututut 2
Tututututut 2
 
Devry card 405 week 4 portfolio draft
Devry card 405 week 4 portfolio draftDevry card 405 week 4 portfolio draft
Devry card 405 week 4 portfolio draft
 
Cociñamos
CociñamosCociñamos
Cociñamos
 

Similar to Chronic Pain Biomarkers Using Brain Imaging and Machine Learning

Crimson Publishers - Assessing Pain Using Morbid Motion Monitor System in the...
Crimson Publishers - Assessing Pain Using Morbid Motion Monitor System in the...Crimson Publishers - Assessing Pain Using Morbid Motion Monitor System in the...
Crimson Publishers - Assessing Pain Using Morbid Motion Monitor System in the...CrimsonpublishersMedical
 
Crimson Publishers - Nursing and Technology Foresight in Futures of a Complex...
Crimson Publishers - Nursing and Technology Foresight in Futures of a Complex...Crimson Publishers - Nursing and Technology Foresight in Futures of a Complex...
Crimson Publishers - Nursing and Technology Foresight in Futures of a Complex...CrimsonpublishersMedical
 
Psychological determinants-of-problematic-outcomes-following-total-knee-arthr...
Psychological determinants-of-problematic-outcomes-following-total-knee-arthr...Psychological determinants-of-problematic-outcomes-following-total-knee-arthr...
Psychological determinants-of-problematic-outcomes-following-total-knee-arthr...Paul Coelho, MD
 
Development of structured orthopedic manual therapy assessment proforma for d...
Development of structured orthopedic manual therapy assessment proforma for d...Development of structured orthopedic manual therapy assessment proforma for d...
Development of structured orthopedic manual therapy assessment proforma for d...BioMedSciDirect Publications
 
Central Sensitization In LBP
Central Sensitization In LBPCentral Sensitization In LBP
Central Sensitization In LBPPaul Coelho, MD
 
Percutaneous image-guided cryoablation of spinal metastases: A systematic review
Percutaneous image-guided cryoablation of spinal metastases: A systematic reviewPercutaneous image-guided cryoablation of spinal metastases: A systematic review
Percutaneous image-guided cryoablation of spinal metastases: A systematic reviewAhmad Ozair
 
Scrambler Therapy: An Innovative Neuromodulation Approach to Complex Regional...
Scrambler Therapy: An Innovative Neuromodulation Approach to Complex Regional...Scrambler Therapy: An Innovative Neuromodulation Approach to Complex Regional...
Scrambler Therapy: An Innovative Neuromodulation Approach to Complex Regional...Calmar Pain Relief Therapy, LLC
 
Neural blockade for persistent pain after breast cancer surgery
Neural blockade for persistent pain after breast cancer surgery Neural blockade for persistent pain after breast cancer surgery
Neural blockade for persistent pain after breast cancer surgery Jason Attaman
 
protocol of shakers swallwing.docx
protocol of shakers swallwing.docxprotocol of shakers swallwing.docx
protocol of shakers swallwing.docxZeinabGamal14
 
Neuromuscular exercise.pdf
Neuromuscular exercise.pdfNeuromuscular exercise.pdf
Neuromuscular exercise.pdfJuanFranCoach1
 
10.1089@photob.2019.4705.pdf photobiomodulation
10.1089@photob.2019.4705.pdf photobiomodulation10.1089@photob.2019.4705.pdf photobiomodulation
10.1089@photob.2019.4705.pdf photobiomodulationcrisendoce
 
Common Brain Mechanisms Between Pain & Addiction
Common Brain Mechanisms Between Pain & AddictionCommon Brain Mechanisms Between Pain & Addiction
Common Brain Mechanisms Between Pain & AddictionPaul Coelho, MD
 
For this assignment activity,  I want you to answer the questions be
For this assignment activity,  I want you to answer the questions beFor this assignment activity,  I want you to answer the questions be
For this assignment activity,  I want you to answer the questions beLilianaJohansen814
 
The emotional brain a a predictor and amplifier of chronic pain.
The emotional brain a a predictor and amplifier of chronic pain.The emotional brain a a predictor and amplifier of chronic pain.
The emotional brain a a predictor and amplifier of chronic pain.Paul Coelho, MD
 
Leysen - Manual Therapy 2014 - SR IPQ-R-2
Leysen - Manual Therapy 2014 - SR IPQ-R-2Leysen - Manual Therapy 2014 - SR IPQ-R-2
Leysen - Manual Therapy 2014 - SR IPQ-R-2Marijke Leysen
 
Effective strategies to monitor clinical risks using biostatistics - Pubrica.pdf
Effective strategies to monitor clinical risks using biostatistics - Pubrica.pdfEffective strategies to monitor clinical risks using biostatistics - Pubrica.pdf
Effective strategies to monitor clinical risks using biostatistics - Pubrica.pdfPubrica
 
Clinical StudyComparison of High-Intensity Laser Therapy and
Clinical StudyComparison of High-Intensity Laser Therapy andClinical StudyComparison of High-Intensity Laser Therapy and
Clinical StudyComparison of High-Intensity Laser Therapy andWilheminaRossi174
 

Similar to Chronic Pain Biomarkers Using Brain Imaging and Machine Learning (20)

Crimson Publishers - Assessing Pain Using Morbid Motion Monitor System in the...
Crimson Publishers - Assessing Pain Using Morbid Motion Monitor System in the...Crimson Publishers - Assessing Pain Using Morbid Motion Monitor System in the...
Crimson Publishers - Assessing Pain Using Morbid Motion Monitor System in the...
 
Crimson Publishers - Nursing and Technology Foresight in Futures of a Complex...
Crimson Publishers - Nursing and Technology Foresight in Futures of a Complex...Crimson Publishers - Nursing and Technology Foresight in Futures of a Complex...
Crimson Publishers - Nursing and Technology Foresight in Futures of a Complex...
 
Psychological determinants-of-problematic-outcomes-following-total-knee-arthr...
Psychological determinants-of-problematic-outcomes-following-total-knee-arthr...Psychological determinants-of-problematic-outcomes-following-total-knee-arthr...
Psychological determinants-of-problematic-outcomes-following-total-knee-arthr...
 
Development of structured orthopedic manual therapy assessment proforma for d...
Development of structured orthopedic manual therapy assessment proforma for d...Development of structured orthopedic manual therapy assessment proforma for d...
Development of structured orthopedic manual therapy assessment proforma for d...
 
Central Sensitization In LBP
Central Sensitization In LBPCentral Sensitization In LBP
Central Sensitization In LBP
 
Percutaneous image-guided cryoablation of spinal metastases: A systematic review
Percutaneous image-guided cryoablation of spinal metastases: A systematic reviewPercutaneous image-guided cryoablation of spinal metastases: A systematic review
Percutaneous image-guided cryoablation of spinal metastases: A systematic review
 
Scrambler Therapy: An Innovative Neuromodulation Approach to Complex Regional...
Scrambler Therapy: An Innovative Neuromodulation Approach to Complex Regional...Scrambler Therapy: An Innovative Neuromodulation Approach to Complex Regional...
Scrambler Therapy: An Innovative Neuromodulation Approach to Complex Regional...
 
Exercícios controle motor e lombalgia
Exercícios controle motor e lombalgiaExercícios controle motor e lombalgia
Exercícios controle motor e lombalgia
 
Neural blockade for persistent pain after breast cancer surgery
Neural blockade for persistent pain after breast cancer surgery Neural blockade for persistent pain after breast cancer surgery
Neural blockade for persistent pain after breast cancer surgery
 
protocol of shakers swallwing.docx
protocol of shakers swallwing.docxprotocol of shakers swallwing.docx
protocol of shakers swallwing.docx
 
Neuromuscular exercise.pdf
Neuromuscular exercise.pdfNeuromuscular exercise.pdf
Neuromuscular exercise.pdf
 
10.1089@photob.2019.4705.pdf photobiomodulation
10.1089@photob.2019.4705.pdf photobiomodulation10.1089@photob.2019.4705.pdf photobiomodulation
10.1089@photob.2019.4705.pdf photobiomodulation
 
Common Brain Mechanisms Between Pain & Addiction
Common Brain Mechanisms Between Pain & AddictionCommon Brain Mechanisms Between Pain & Addiction
Common Brain Mechanisms Between Pain & Addiction
 
For this assignment activity,  I want you to answer the questions be
For this assignment activity,  I want you to answer the questions beFor this assignment activity,  I want you to answer the questions be
For this assignment activity,  I want you to answer the questions be
 
The emotional brain a a predictor and amplifier of chronic pain.
The emotional brain a a predictor and amplifier of chronic pain.The emotional brain a a predictor and amplifier of chronic pain.
The emotional brain a a predictor and amplifier of chronic pain.
 
Hrqol neuro
Hrqol neuroHrqol neuro
Hrqol neuro
 
Leysen - Manual Therapy 2014 - SR IPQ-R-2
Leysen - Manual Therapy 2014 - SR IPQ-R-2Leysen - Manual Therapy 2014 - SR IPQ-R-2
Leysen - Manual Therapy 2014 - SR IPQ-R-2
 
Effective strategies to monitor clinical risks using biostatistics - Pubrica.pdf
Effective strategies to monitor clinical risks using biostatistics - Pubrica.pdfEffective strategies to monitor clinical risks using biostatistics - Pubrica.pdf
Effective strategies to monitor clinical risks using biostatistics - Pubrica.pdf
 
Pain trials ppt
Pain trials pptPain trials ppt
Pain trials ppt
 
Clinical StudyComparison of High-Intensity Laser Therapy and
Clinical StudyComparison of High-Intensity Laser Therapy andClinical StudyComparison of High-Intensity Laser Therapy and
Clinical StudyComparison of High-Intensity Laser Therapy and
 

More from Paul Coelho, MD

Suicidality Poster References.docx
Suicidality Poster References.docxSuicidality Poster References.docx
Suicidality Poster References.docxPaul Coelho, MD
 
Opioid Milli-equivalence Conversion Factors CDC
Opioid Milli-equivalence Conversion Factors CDCOpioid Milli-equivalence Conversion Factors CDC
Opioid Milli-equivalence Conversion Factors CDCPaul Coelho, MD
 
Labeling Woefulness: The Social Construction of Fibromyalgia
Labeling Woefulness: The Social Construction of FibromyalgiaLabeling Woefulness: The Social Construction of Fibromyalgia
Labeling Woefulness: The Social Construction of FibromyalgiaPaul Coelho, MD
 
Outcomes in Long-term Opioid Tapering and Buprenorphine Transition: A Retrosp...
Outcomes in Long-term Opioid Tapering and Buprenorphine Transition: A Retrosp...Outcomes in Long-term Opioid Tapering and Buprenorphine Transition: A Retrosp...
Outcomes in Long-term Opioid Tapering and Buprenorphine Transition: A Retrosp...Paul Coelho, MD
 
Pain Phenotyping Tool For Primary Care
Pain Phenotyping Tool For Primary CarePain Phenotyping Tool For Primary Care
Pain Phenotyping Tool For Primary CarePaul Coelho, MD
 
Fibromyalgia & Somatic Symptom Disorder Patient Handout
Fibromyalgia & Somatic Symptom Disorder Patient HandoutFibromyalgia & Somatic Symptom Disorder Patient Handout
Fibromyalgia & Somatic Symptom Disorder Patient HandoutPaul Coelho, MD
 
Community Catastrophizing
Community CatastrophizingCommunity Catastrophizing
Community CatastrophizingPaul Coelho, MD
 
Risk-Benefit High-Dose LTOT
Risk-Benefit High-Dose LTOTRisk-Benefit High-Dose LTOT
Risk-Benefit High-Dose LTOTPaul Coelho, MD
 
Mortality quadrupled among opioid-driven hospitalizations notably within lowe...
Mortality quadrupled among opioid-driven hospitalizations notably within lowe...Mortality quadrupled among opioid-driven hospitalizations notably within lowe...
Mortality quadrupled among opioid-driven hospitalizations notably within lowe...Paul Coelho, MD
 
Prescriptions filled following an opioid-related hospitalization.
Prescriptions filled following an opioid-related hospitalization.Prescriptions filled following an opioid-related hospitalization.
Prescriptions filled following an opioid-related hospitalization.Paul Coelho, MD
 
Jamapsychiatry santavirta 2017_oi_170084
Jamapsychiatry santavirta 2017_oi_170084Jamapsychiatry santavirta 2017_oi_170084
Jamapsychiatry santavirta 2017_oi_170084Paul Coelho, MD
 
Correlation of opioid mortality with prescriptions and social determinants -a...
Correlation of opioid mortality with prescriptions and social determinants -a...Correlation of opioid mortality with prescriptions and social determinants -a...
Correlation of opioid mortality with prescriptions and social determinants -a...Paul Coelho, MD
 
Gao recommendations to CMS
Gao recommendations to CMSGao recommendations to CMS
Gao recommendations to CMSPaul Coelho, MD
 
Prescribing by specialty
Prescribing by specialtyPrescribing by specialty
Prescribing by specialtyPaul Coelho, MD
 
The place-of-antipsychotics-in-the-therapy-of-anxiety-disorders-and-obsessive...
The place-of-antipsychotics-in-the-therapy-of-anxiety-disorders-and-obsessive...The place-of-antipsychotics-in-the-therapy-of-anxiety-disorders-and-obsessive...
The place-of-antipsychotics-in-the-therapy-of-anxiety-disorders-and-obsessive...Paul Coelho, MD
 
Structured opioid refill clinic epic smartphrases
Structured opioid refill clinic epic smartphrases Structured opioid refill clinic epic smartphrases
Structured opioid refill clinic epic smartphrases Paul Coelho, MD
 
Opioids for the Treatment of Chronic Pain: Mistakes Made, Lessons Learned, an...
Opioids for the Treatment of Chronic Pain: Mistakes Made, Lessons Learned, an...Opioids for the Treatment of Chronic Pain: Mistakes Made, Lessons Learned, an...
Opioids for the Treatment of Chronic Pain: Mistakes Made, Lessons Learned, an...Paul Coelho, MD
 
The potential adverse influence of physicians’ words.
The potential adverse influence of physicians’ words.The potential adverse influence of physicians’ words.
The potential adverse influence of physicians’ words.Paul Coelho, MD
 
Ctaf chronic pain__evidence_report_100417
Ctaf chronic pain__evidence_report_100417Ctaf chronic pain__evidence_report_100417
Ctaf chronic pain__evidence_report_100417Paul Coelho, MD
 

More from Paul Coelho, MD (20)

Suicidality Poster References.docx
Suicidality Poster References.docxSuicidality Poster References.docx
Suicidality Poster References.docx
 
Opioid Milli-equivalence Conversion Factors CDC
Opioid Milli-equivalence Conversion Factors CDCOpioid Milli-equivalence Conversion Factors CDC
Opioid Milli-equivalence Conversion Factors CDC
 
Labeling Woefulness: The Social Construction of Fibromyalgia
Labeling Woefulness: The Social Construction of FibromyalgiaLabeling Woefulness: The Social Construction of Fibromyalgia
Labeling Woefulness: The Social Construction of Fibromyalgia
 
Outcomes in Long-term Opioid Tapering and Buprenorphine Transition: A Retrosp...
Outcomes in Long-term Opioid Tapering and Buprenorphine Transition: A Retrosp...Outcomes in Long-term Opioid Tapering and Buprenorphine Transition: A Retrosp...
Outcomes in Long-term Opioid Tapering and Buprenorphine Transition: A Retrosp...
 
Pain Phenotyping Tool For Primary Care
Pain Phenotyping Tool For Primary CarePain Phenotyping Tool For Primary Care
Pain Phenotyping Tool For Primary Care
 
Fibromyalgia & Somatic Symptom Disorder Patient Handout
Fibromyalgia & Somatic Symptom Disorder Patient HandoutFibromyalgia & Somatic Symptom Disorder Patient Handout
Fibromyalgia & Somatic Symptom Disorder Patient Handout
 
Community Catastrophizing
Community CatastrophizingCommunity Catastrophizing
Community Catastrophizing
 
Risk-Benefit High-Dose LTOT
Risk-Benefit High-Dose LTOTRisk-Benefit High-Dose LTOT
Risk-Benefit High-Dose LTOT
 
Space Trial
Space TrialSpace Trial
Space Trial
 
Mortality quadrupled among opioid-driven hospitalizations notably within lowe...
Mortality quadrupled among opioid-driven hospitalizations notably within lowe...Mortality quadrupled among opioid-driven hospitalizations notably within lowe...
Mortality quadrupled among opioid-driven hospitalizations notably within lowe...
 
Prescriptions filled following an opioid-related hospitalization.
Prescriptions filled following an opioid-related hospitalization.Prescriptions filled following an opioid-related hospitalization.
Prescriptions filled following an opioid-related hospitalization.
 
Jamapsychiatry santavirta 2017_oi_170084
Jamapsychiatry santavirta 2017_oi_170084Jamapsychiatry santavirta 2017_oi_170084
Jamapsychiatry santavirta 2017_oi_170084
 
Correlation of opioid mortality with prescriptions and social determinants -a...
Correlation of opioid mortality with prescriptions and social determinants -a...Correlation of opioid mortality with prescriptions and social determinants -a...
Correlation of opioid mortality with prescriptions and social determinants -a...
 
Gao recommendations to CMS
Gao recommendations to CMSGao recommendations to CMS
Gao recommendations to CMS
 
Prescribing by specialty
Prescribing by specialtyPrescribing by specialty
Prescribing by specialty
 
The place-of-antipsychotics-in-the-therapy-of-anxiety-disorders-and-obsessive...
The place-of-antipsychotics-in-the-therapy-of-anxiety-disorders-and-obsessive...The place-of-antipsychotics-in-the-therapy-of-anxiety-disorders-and-obsessive...
The place-of-antipsychotics-in-the-therapy-of-anxiety-disorders-and-obsessive...
 
Structured opioid refill clinic epic smartphrases
Structured opioid refill clinic epic smartphrases Structured opioid refill clinic epic smartphrases
Structured opioid refill clinic epic smartphrases
 
Opioids for the Treatment of Chronic Pain: Mistakes Made, Lessons Learned, an...
Opioids for the Treatment of Chronic Pain: Mistakes Made, Lessons Learned, an...Opioids for the Treatment of Chronic Pain: Mistakes Made, Lessons Learned, an...
Opioids for the Treatment of Chronic Pain: Mistakes Made, Lessons Learned, an...
 
The potential adverse influence of physicians’ words.
The potential adverse influence of physicians’ words.The potential adverse influence of physicians’ words.
The potential adverse influence of physicians’ words.
 
Ctaf chronic pain__evidence_report_100417
Ctaf chronic pain__evidence_report_100417Ctaf chronic pain__evidence_report_100417
Ctaf chronic pain__evidence_report_100417
 

Recently uploaded

Call Girls Hyderabad Kirti 9907093804 Independent Escort Service Hyderabad
Call Girls Hyderabad Kirti 9907093804 Independent Escort Service HyderabadCall Girls Hyderabad Kirti 9907093804 Independent Escort Service Hyderabad
Call Girls Hyderabad Kirti 9907093804 Independent Escort Service Hyderabaddelhimodelshub1
 
indian Call Girl Panchkula ❤️🍑 9907093804 Low Rate Call Girls Ludhiana Tulsi
indian Call Girl Panchkula ❤️🍑 9907093804 Low Rate Call Girls Ludhiana Tulsiindian Call Girl Panchkula ❤️🍑 9907093804 Low Rate Call Girls Ludhiana Tulsi
indian Call Girl Panchkula ❤️🍑 9907093804 Low Rate Call Girls Ludhiana TulsiHigh Profile Call Girls Chandigarh Aarushi
 
Call Girls in Mohali Surbhi ❤️🍑 9907093804 👄🫦 Independent Escort Service Mohali
Call Girls in Mohali Surbhi ❤️🍑 9907093804 👄🫦 Independent Escort Service MohaliCall Girls in Mohali Surbhi ❤️🍑 9907093804 👄🫦 Independent Escort Service Mohali
Call Girls in Mohali Surbhi ❤️🍑 9907093804 👄🫦 Independent Escort Service MohaliHigh Profile Call Girls Chandigarh Aarushi
 
Russian Call Girls Hyderabad Indira 9907093804 Independent Escort Service Hyd...
Russian Call Girls Hyderabad Indira 9907093804 Independent Escort Service Hyd...Russian Call Girls Hyderabad Indira 9907093804 Independent Escort Service Hyd...
Russian Call Girls Hyderabad Indira 9907093804 Independent Escort Service Hyd...delhimodelshub1
 
Russian Call Girls in Chandigarh Ojaswi ❤️🍑 9907093804 👄🫦 Independent Escort ...
Russian Call Girls in Chandigarh Ojaswi ❤️🍑 9907093804 👄🫦 Independent Escort ...Russian Call Girls in Chandigarh Ojaswi ❤️🍑 9907093804 👄🫦 Independent Escort ...
Russian Call Girls in Chandigarh Ojaswi ❤️🍑 9907093804 👄🫦 Independent Escort ...High Profile Call Girls Chandigarh Aarushi
 
VIP Call Girl Sector 32 Noida Just Book Me 9711199171
VIP Call Girl Sector 32 Noida Just Book Me 9711199171VIP Call Girl Sector 32 Noida Just Book Me 9711199171
VIP Call Girl Sector 32 Noida Just Book Me 9711199171Call Girls Service Gurgaon
 
Hot Call Girl In Ludhiana 👅🥵 9053'900678 Call Girls Service In Ludhiana
Hot  Call Girl In Ludhiana 👅🥵 9053'900678 Call Girls Service In LudhianaHot  Call Girl In Ludhiana 👅🥵 9053'900678 Call Girls Service In Ludhiana
Hot Call Girl In Ludhiana 👅🥵 9053'900678 Call Girls Service In LudhianaRussian Call Girls in Ludhiana
 
pOOJA sexy Call Girls In Sector 49,9999965857 Young Female Escorts Service In...
pOOJA sexy Call Girls In Sector 49,9999965857 Young Female Escorts Service In...pOOJA sexy Call Girls In Sector 49,9999965857 Young Female Escorts Service In...
pOOJA sexy Call Girls In Sector 49,9999965857 Young Female Escorts Service In...Call Girls Noida
 
Call Girls Service Chandigarh Grishma ❤️🍑 9907093804 👄🫦 Independent Escort Se...
Call Girls Service Chandigarh Grishma ❤️🍑 9907093804 👄🫦 Independent Escort Se...Call Girls Service Chandigarh Grishma ❤️🍑 9907093804 👄🫦 Independent Escort Se...
Call Girls Service Chandigarh Grishma ❤️🍑 9907093804 👄🫦 Independent Escort Se...High Profile Call Girls Chandigarh Aarushi
 
Call Girls in Hyderabad Lavanya 9907093804 Independent Escort Service Hyderabad
Call Girls in Hyderabad Lavanya 9907093804 Independent Escort Service HyderabadCall Girls in Hyderabad Lavanya 9907093804 Independent Escort Service Hyderabad
Call Girls in Hyderabad Lavanya 9907093804 Independent Escort Service Hyderabaddelhimodelshub1
 
Call Girls Service Chandigarh Gori WhatsApp ❤9115573837 VIP Call Girls Chandi...
Call Girls Service Chandigarh Gori WhatsApp ❤9115573837 VIP Call Girls Chandi...Call Girls Service Chandigarh Gori WhatsApp ❤9115573837 VIP Call Girls Chandi...
Call Girls Service Chandigarh Gori WhatsApp ❤9115573837 VIP Call Girls Chandi...Niamh verma
 
Hot Call Girl In Chandigarh 👅🥵 9053'900678 Call Girls Service In Chandigarh
Hot  Call Girl In Chandigarh 👅🥵 9053'900678 Call Girls Service In ChandigarhHot  Call Girl In Chandigarh 👅🥵 9053'900678 Call Girls Service In Chandigarh
Hot Call Girl In Chandigarh 👅🥵 9053'900678 Call Girls Service In ChandigarhVip call girls In Chandigarh
 
Call Girl Hyderabad Madhuri 9907093804 Independent Escort Service Hyderabad
Call Girl Hyderabad Madhuri 9907093804 Independent Escort Service HyderabadCall Girl Hyderabad Madhuri 9907093804 Independent Escort Service Hyderabad
Call Girl Hyderabad Madhuri 9907093804 Independent Escort Service Hyderabaddelhimodelshub1
 
Jalandhar Female Call Girls Contact Number 9053900678 💚Jalandhar Female Call...
Jalandhar  Female Call Girls Contact Number 9053900678 💚Jalandhar Female Call...Jalandhar  Female Call Girls Contact Number 9053900678 💚Jalandhar Female Call...
Jalandhar Female Call Girls Contact Number 9053900678 💚Jalandhar Female Call...Call Girls Service Chandigarh Ayushi
 
Call Girl Raipur 9873940964 Book Hot And Sexy Girls
Call Girl Raipur 9873940964 Book Hot And Sexy GirlsCall Girl Raipur 9873940964 Book Hot And Sexy Girls
Call Girl Raipur 9873940964 Book Hot And Sexy Girlsddev2574
 
Basics of Anatomy- Language of Anatomy.pptx
Basics of Anatomy- Language of Anatomy.pptxBasics of Anatomy- Language of Anatomy.pptx
Basics of Anatomy- Language of Anatomy.pptxAyush Gupta
 
Call Girls LB Nagar 7001305949 all area service COD available Any Time
Call Girls LB Nagar 7001305949 all area service COD available Any TimeCall Girls LB Nagar 7001305949 all area service COD available Any Time
Call Girls LB Nagar 7001305949 all area service COD available Any Timedelhimodelshub1
 
VIP Call Girl Sector 88 Gurgaon Delhi Just Call Me 9899900591
VIP Call Girl Sector 88 Gurgaon Delhi Just Call Me 9899900591VIP Call Girl Sector 88 Gurgaon Delhi Just Call Me 9899900591
VIP Call Girl Sector 88 Gurgaon Delhi Just Call Me 9899900591adityaroy0215
 

Recently uploaded (20)

Call Girl Lucknow Gauri 🔝 8923113531 🔝 🎶 Independent Escort Service Lucknow
Call Girl Lucknow Gauri 🔝 8923113531  🔝 🎶 Independent Escort Service LucknowCall Girl Lucknow Gauri 🔝 8923113531  🔝 🎶 Independent Escort Service Lucknow
Call Girl Lucknow Gauri 🔝 8923113531 🔝 🎶 Independent Escort Service Lucknow
 
Call Girls Hyderabad Kirti 9907093804 Independent Escort Service Hyderabad
Call Girls Hyderabad Kirti 9907093804 Independent Escort Service HyderabadCall Girls Hyderabad Kirti 9907093804 Independent Escort Service Hyderabad
Call Girls Hyderabad Kirti 9907093804 Independent Escort Service Hyderabad
 
indian Call Girl Panchkula ❤️🍑 9907093804 Low Rate Call Girls Ludhiana Tulsi
indian Call Girl Panchkula ❤️🍑 9907093804 Low Rate Call Girls Ludhiana Tulsiindian Call Girl Panchkula ❤️🍑 9907093804 Low Rate Call Girls Ludhiana Tulsi
indian Call Girl Panchkula ❤️🍑 9907093804 Low Rate Call Girls Ludhiana Tulsi
 
Call Girls in Mohali Surbhi ❤️🍑 9907093804 👄🫦 Independent Escort Service Mohali
Call Girls in Mohali Surbhi ❤️🍑 9907093804 👄🫦 Independent Escort Service MohaliCall Girls in Mohali Surbhi ❤️🍑 9907093804 👄🫦 Independent Escort Service Mohali
Call Girls in Mohali Surbhi ❤️🍑 9907093804 👄🫦 Independent Escort Service Mohali
 
Russian Call Girls Hyderabad Indira 9907093804 Independent Escort Service Hyd...
Russian Call Girls Hyderabad Indira 9907093804 Independent Escort Service Hyd...Russian Call Girls Hyderabad Indira 9907093804 Independent Escort Service Hyd...
Russian Call Girls Hyderabad Indira 9907093804 Independent Escort Service Hyd...
 
Russian Call Girls in Chandigarh Ojaswi ❤️🍑 9907093804 👄🫦 Independent Escort ...
Russian Call Girls in Chandigarh Ojaswi ❤️🍑 9907093804 👄🫦 Independent Escort ...Russian Call Girls in Chandigarh Ojaswi ❤️🍑 9907093804 👄🫦 Independent Escort ...
Russian Call Girls in Chandigarh Ojaswi ❤️🍑 9907093804 👄🫦 Independent Escort ...
 
VIP Call Girl Sector 32 Noida Just Book Me 9711199171
VIP Call Girl Sector 32 Noida Just Book Me 9711199171VIP Call Girl Sector 32 Noida Just Book Me 9711199171
VIP Call Girl Sector 32 Noida Just Book Me 9711199171
 
Hot Call Girl In Ludhiana 👅🥵 9053'900678 Call Girls Service In Ludhiana
Hot  Call Girl In Ludhiana 👅🥵 9053'900678 Call Girls Service In LudhianaHot  Call Girl In Ludhiana 👅🥵 9053'900678 Call Girls Service In Ludhiana
Hot Call Girl In Ludhiana 👅🥵 9053'900678 Call Girls Service In Ludhiana
 
pOOJA sexy Call Girls In Sector 49,9999965857 Young Female Escorts Service In...
pOOJA sexy Call Girls In Sector 49,9999965857 Young Female Escorts Service In...pOOJA sexy Call Girls In Sector 49,9999965857 Young Female Escorts Service In...
pOOJA sexy Call Girls In Sector 49,9999965857 Young Female Escorts Service In...
 
Call Girls Service Chandigarh Grishma ❤️🍑 9907093804 👄🫦 Independent Escort Se...
Call Girls Service Chandigarh Grishma ❤️🍑 9907093804 👄🫦 Independent Escort Se...Call Girls Service Chandigarh Grishma ❤️🍑 9907093804 👄🫦 Independent Escort Se...
Call Girls Service Chandigarh Grishma ❤️🍑 9907093804 👄🫦 Independent Escort Se...
 
#9711199012# African Student Escorts in Delhi 😘 Call Girls Delhi
#9711199012# African Student Escorts in Delhi 😘 Call Girls Delhi#9711199012# African Student Escorts in Delhi 😘 Call Girls Delhi
#9711199012# African Student Escorts in Delhi 😘 Call Girls Delhi
 
Call Girls in Hyderabad Lavanya 9907093804 Independent Escort Service Hyderabad
Call Girls in Hyderabad Lavanya 9907093804 Independent Escort Service HyderabadCall Girls in Hyderabad Lavanya 9907093804 Independent Escort Service Hyderabad
Call Girls in Hyderabad Lavanya 9907093804 Independent Escort Service Hyderabad
 
Call Girls Service Chandigarh Gori WhatsApp ❤9115573837 VIP Call Girls Chandi...
Call Girls Service Chandigarh Gori WhatsApp ❤9115573837 VIP Call Girls Chandi...Call Girls Service Chandigarh Gori WhatsApp ❤9115573837 VIP Call Girls Chandi...
Call Girls Service Chandigarh Gori WhatsApp ❤9115573837 VIP Call Girls Chandi...
 
Hot Call Girl In Chandigarh 👅🥵 9053'900678 Call Girls Service In Chandigarh
Hot  Call Girl In Chandigarh 👅🥵 9053'900678 Call Girls Service In ChandigarhHot  Call Girl In Chandigarh 👅🥵 9053'900678 Call Girls Service In Chandigarh
Hot Call Girl In Chandigarh 👅🥵 9053'900678 Call Girls Service In Chandigarh
 
Call Girl Hyderabad Madhuri 9907093804 Independent Escort Service Hyderabad
Call Girl Hyderabad Madhuri 9907093804 Independent Escort Service HyderabadCall Girl Hyderabad Madhuri 9907093804 Independent Escort Service Hyderabad
Call Girl Hyderabad Madhuri 9907093804 Independent Escort Service Hyderabad
 
Jalandhar Female Call Girls Contact Number 9053900678 💚Jalandhar Female Call...
Jalandhar  Female Call Girls Contact Number 9053900678 💚Jalandhar Female Call...Jalandhar  Female Call Girls Contact Number 9053900678 💚Jalandhar Female Call...
Jalandhar Female Call Girls Contact Number 9053900678 💚Jalandhar Female Call...
 
Call Girl Raipur 9873940964 Book Hot And Sexy Girls
Call Girl Raipur 9873940964 Book Hot And Sexy GirlsCall Girl Raipur 9873940964 Book Hot And Sexy Girls
Call Girl Raipur 9873940964 Book Hot And Sexy Girls
 
Basics of Anatomy- Language of Anatomy.pptx
Basics of Anatomy- Language of Anatomy.pptxBasics of Anatomy- Language of Anatomy.pptx
Basics of Anatomy- Language of Anatomy.pptx
 
Call Girls LB Nagar 7001305949 all area service COD available Any Time
Call Girls LB Nagar 7001305949 all area service COD available Any TimeCall Girls LB Nagar 7001305949 all area service COD available Any Time
Call Girls LB Nagar 7001305949 all area service COD available Any Time
 
VIP Call Girl Sector 88 Gurgaon Delhi Just Call Me 9899900591
VIP Call Girl Sector 88 Gurgaon Delhi Just Call Me 9899900591VIP Call Girl Sector 88 Gurgaon Delhi Just Call Me 9899900591
VIP Call Girl Sector 88 Gurgaon Delhi Just Call Me 9899900591
 

Chronic Pain Biomarkers Using Brain Imaging and Machine Learning

  • 1. CHRONIC PAIN (R STAUD, SECTION EDITOR) Biomarkers for Musculoskeletal Pain Conditions: Use of Brain Imaging and Machine Learning Jeff Boissoneault1 & Landrew Sevel1 & Janelle Letzen1 & Michael Robinson1 & Roland Staud2 # Springer Science+Business Media New York 2017 Abstract Chronic musculoskeletal pain condition often shows poor correlations between tissue abnormalities and clinical pain. Therefore, classification of pain conditions like chronic low back pain, osteoarthritis, and fibromyalgia de- pends mostly on self report and less on objective findings like X-ray or magnetic resonance imaging (MRI) changes. However, recent advances in structural and functional brain imaging have identified brain abnormalities in chronic pain conditions that can be used for illness classification. Because the analysis of complex and multivariate brain imaging data is challenging, machine learning techniques have been increas- ingly utilized for this purpose. The goal of machine learning is to train specific classifiers to best identify variables of interest on brain MRIs (i.e., biomarkers). This report describes classi- fication techniques capable of separating MRI-based brain biomarkers of chronic pain patients from healthy controls with high accuracy (70–92%) using machine learning, as well as critical scientific, practical, and ethical considerations related to their potential clinical application. Although self-report re- mains the gold standard for pain assessment, machine learning may aid in the classification of chronic pain disorders like chronic back pain and fibromyalgia as well as provide mech- anistic information regarding their neural correlates. Keywords Chronic pain . Musculoskeletal . Magnetic resonance imaging (MRI) . Machine learning . Classification Introduction Chronic pain is a highly prevalent condition associated with significant disability and societal cost [1]. The etiology of chronic pain can vary substantially across patients and often appears to be secondary to biological factors, such as muscu- loskeletal injury (e.g., osteoarthritis [OA]), nerve injury (neu- ropathic pain), autoimmunity (rheumatoid arthritis [RA], sys- temic lupus erythematosus), or substance abuse (alcoholic neuropathy and chronic pancreatitis). In many cases, however, chronic pain seems to be the primary illness associated with a given condition (e.g., fibromyalgia syndrome [FM; 2]). Over the past several decades, substantial effort has been dedicated to the development of methodologies for the reliable discrim- ination or classification of patients with a given chronic pain condition from people without the condition (i.e., “healthy controls”), patients with similar but distinct illnesses, or both. The goal of many studies is to accurately identify individ- uals with a clinical chronic pain condition in question based on potential mechanistic underpinnings. Given that some chronic pain conditions are associated with peripheral tissue pathophysiology, numerous studies have been aimed at reli- able discrimination of individuals with clinical pain using pe- ripheral measures including structural (e.g., knee degenera- tion, lumbar disc pathology; [3]), functional (e.g., gait abnor- mality, inflammatory processes in rheumatoid arthritis; [4, 5]), and genetic data [6]. Because chronic pain often exists as a primary symptom without remarkable tissue abnormalities, there is an increasing interest in their neural correlates, i.e., mechanistic or structural abnormalities derived from structural or functional magnetic resonance (MRI) brain imaging studies This article is part of the Topical Collection on Chronic Pain * Roland Staud staudr@ufl.edu 1 Department of Clinical and Health Psychology, University of Florida, Gainesville, FL, USA 2 Department of Medicine, University of Florida, PO Box 100277, Gainesville, FL 32610, USA Curr Rheumatol Rep (2017) 19:5 DOI 10.1007/s11926-017-0629-9
  • 2. of chronic pain (e.g., grey matter density/volume, white matter integrity, or functional connectivity between pain-related brain regions). In addition, such information can be used to determine whether central nervous system (CNS) abnormali- ties are suitable to appropriately classify individuals with chronic pain. In this regard, a major goal of many MRI-based chronic pain classification studies is the development of objective bio- markers for each condition that do not rely on self-report [7, 8•, 9]. Brain-based biomarker studies of chronic pain leverage the extensive neuroimaging literature describing the critical role of certain brain regions in the sensory (e.g., thalamus, primary and secondary somatosensory cortex, posterior insula, basal ganglia), affective (e.g., anterior insula, hypothal- amus, anterior cingulate cortex, amygdala, hippocampus), and cognitive-evaluative (e.g., dorsolateral prefrontal cortex, ante- rior cingulate cortex, thalamus) aspects of the pain experience (for review, see [10]). Although our review focuses exclusive- ly on brain imaging biomarkers, it is worth noting that a sub- stantial literature has also applied this approach to peripheral mechanisms. For instance, measurement of exhaled organic volatiles [11] and joint ultrasound [12] have been suggested as useful approaches for the automated classification and/or dif- ferential diagnosis of rheumatoid arthritis. Some proponents of brain imaging-based approaches to illness classification claim that brain biomarkers could act as a “surrogate” for pain self-report [13]. This could result in greater sensitivity to measure efficacy of analgesic treatments [9] by avoiding potential pitfalls associated with the use of subjective patient and provider reports. Furthermore, such brain biomarkers could serve as targets for novel treatments [14, 15]. In particular, objective markers of chronic pain may play an important role in pain classification of patients with cognitive/psychological dysfunction and in individuals who are unable to communicate, or in cases of deception [16•, 17], as well as aid in the adjudication of legal claims of injury-related pain disorders [18]. These purported advantages have spurred interest in the development of biomarkers, al- though it is important to note that self-report remains the gold standard for pain measurement. Numerous studies have indi- cated that self-report measures of pain and psychosocial fac- tors have excellent classification accuracy and reliability [8•, 19•, 20]. Indeed, evidence suggests that chronic pain patients can be distinguished from healthy controls with greater than 90% accuracy based on personality factors [21], perceived pain and functional disability [22], and simple visual analog scale (VAS) measures of affect [23••]. Questionnaire-based approaches have shown similar accuracy for the separation of FM and RA [24] and FM, RA, and OA [25]. The purpose of this critical review is to examine the devel- opment of brain biomarkers for chronic musculoskeletal pain disorders using machine learning (ML) in a manner which should be useful to physicians and other health care providers. Therefore, we will restrict our review to musculoskeletal con- ditions for which chronic pain is a primary complaint. The conceptual, scientific, and ethical issues related to this ap- proach, however, will also apply to biomarker studies for other chronic conditions (e.g., depression). Machine Learning Algorithms Existing methods regarding the use of structural and function- al brain imaging to discriminate patient groups from healthy controls have largely relied on ML algorithms, which provide an automated approach to making predictions about previous- ly unknown data. ML algorithms can be broadly classified as supervised or unsupervised. Supervised methods develop models for classifying observations according to a known out- come (e.g., a clinical diagnosis). In contrast, unsupervised methods attempt to discern patterns or structure in data with- out guidance or pre-existing labels. Although unsupervised ML methods may be of interest for the development of sub- groups within existing conditions or new diagnostic classifi- cation criteria based on brain abnormalities, ML studies in chronic pain to date have mostly relied on supervised methods. Support vector machines (SVM; [26, 27]), a type of ML algorithm that uses a training data set composed of one or more features to determine an optimal boundary sepa- rating a set of cases, have been most often used [23••, 28, 29]. However, many other ML techniques have been developed and applied to the automated classification of chronic pain patients [23••, 30]. Detailed discussions of the strengths and limitations of these methods are beyond the scope of this re- view, but basic descriptions of the most common ML algo- rithms applied in the study of chronic pain, as well as detailed references, are provided in Table 1. Scientific and Clinical Utility of Chronic Pain Biomarkers In this section, we will discuss theoretical, practical, and eth- ical issues surrounding pain biomarker development and use, with a particular emphasis on clinical utility. Choice of Classification Algorithm In an ideal scenario, the best algorithm for detecting a chronic pain condition would be one that matches the underlying mod- el or structure that distinguishes that condition from healthy individuals or other conditions (e.g., if a linear decrease of prefrontal cortical thickness is the underlying mechanism of a condition, then a linear model would be sufficient). However, given the complex and often heterogeneous nature of chronic pain conditions, this information is frequently not 5 Page 2 of 9 Curr Rheumatol Rep (2017) 19:5
  • 3. available. It is often difficult, if not impossible, to determine a priori which algorithm might perform best in a certain data set [37]. This problem frequently necessitates the comparison of multiple different algorithms for any single classification task (for examples, see [23••, 38]). As a general rule, simple models may not perform as well on training data but will likely generalize better when assessed on testing data, whereas more complex models will tend to perform well on training data but will not perform as well on testing data. More com- plex models or those that blend inputs for many different algorithms (e.g., ensemble methods) may also lead to chal- lenges in interpretability, requiring additional pipelines to dis- till results in ways that are meaningful and useful for clini- cians. In general, large sample sizes are often needed to in- crease generalizability and prevent overfitting [39]. A recent study suggests that accuracy values obtained from smaller samples are opportunistically biased [40]. Therefore, pro- posed biomarkers must demonstrate strong performance in generalizable samples. Biomarker Reliability and Feasibility of Implementation A neuroimaging-derived biomarker cannot be reliably used for scientific or clinical purposes if the measures composing it do not demonstrate high test-retest reliability (i.e., the repro- ducibility of results over time). This concern is especially relevant given recent evidence that the test-retest reliability of functional connectivity metrics may vary widely depending on the brain regions examined [19•]. For this reason, the reli- ability of neuroimaging-based measures should not be taken for granted. This concern also applies to populations where typical brain function may be perturbed due to injury or illness (e.g., post-stroke [41]). Even a biomarker that is meeting or exceeding criteria for appropriate use as a diagnostic tool will not be clinically useful if it cannot be readily assessed in clinic settings. As noted by Woo and Wager [42], all parameters of the biomarker and its associated testing procedure should be rigorously stan- dardized so that all users collect data comparable to the stan- dard used to derive the measure. Depending on the demands associated with collecting certain biomarkers (especially those derived from BOLD activity during task-based fMRI), such rigor may be difficult to achieve in clinical settings. Often the additional time, nuanced design, and/or special expertise re- quired in the acquisition of reliable data will limit some bio- markers’ applicability and usefulness. What Constitutes Adequate Biomarker Performance? A major issue in the application (and reporting) of ML-based brain biomarkers for chronic pain is determining whether a given marker’s accuracy is adequate. Developing criteria for optimal biomarker performance depends on the setting in which it will be used, as well as the cost of misclassification. Pure research-oriented use cases (e.g., identifying potential markers, phenotypic subgroups, or treatment targets; discern- ing disease mechanisms) may allow for less stringent perfor- mance criteria. In contrast, clinical tasks (diagnosis, prognosis, treatment planning) carry risk of harm (misdiagnosis, delaying treatment, providing inaccurate or unnecessary treatment) and will therefore require considerably more stringent perfor- mance criteria for use in patient care. Although the risk of medical error from decisions based upon ML-derived bio- markers may be no greater than those commonly accepted in current practice, the “black-box” nature of these markers raises liability concerns in the face of medical errors promul- gated by ML-derived biomarkers [43] and may increase phy- sicians’ reluctance to use even those that are well validated. Ultimately, any given biomarker’s practical applicability will depend on a case-by-case assessment of its cost, Table 1 Basic descriptions of common supervised machine learning algorithms Algorithm Basic approach Reference(s) Logistic regression (LR) Uses linear combination of predictors and regression coefficients to make categorical prediction [31] Naïve Bayes (NB) Individuals categorized according to probability that they have a certain value for each feature independently included in prediction model [32] k-Nearest Neighbors (kNN) Categorizes individuals based on the most common class of the k closest observations for each model feature [33] Multilayer perceptron (MLP) Determines appropriate weighting for features for multiple layers of decision units (perceptrons) needed to perfectly classify training data, then applies learned rule to new cases [34] Support vector machine (SVM) Determines a linear or non-linear maximum-margin hyperplane (i.e., feature-based cutoff) that maximizes separation between groups [26, 27] Decision tree (e.g., J48/C4.5) Categorizes individuals in a series of decisions based upon optimal feature cutoff values [35] Least absolute shrinkage and selection operator (LASSO) A variant of ordinary least squares regression that constrains the summary of absolute regression coefficients across features in order to identify the most informative features and optimal cut points for classification [36] Curr Rheumatol Rep (2017) 19:5 Page 3 of 9 5
  • 4. deployability, and accuracy in the context of its intended use. For instance, in clinical settings, biomarker performance should be judged by taking into consideration whether it is intended to replace physicians in a role they can already per- form well (e.g., differentially diagnose mechanistically and/or symptomatically distinct conditions) or, alternatively, provide novel information that clinicians would not otherwise have. This approach may help identify mechanistically distinct clin- ical subgroups, guide treatment decisions, indicate prognosis, or separate conditions with significant mechanistic or symp- tomatic overlap. In cases where biomarker application pro- vides useful clinical information where it would not otherwise be available, higher cost, more difficult deployability, and/or relatively poorer performance may be quite tolerable [43]. Impact of Chronic Pain Prevalence on Biomarker Performance Ethical and practical issues surrounding potential under- or overtreatment of pain remain even in cases where biomark- er sensitivity (i.e., probability of a positive biomarker giv- en an individual has the condition) and specificity (i.e., probability of a negative biomarker given an individual does not have the condition) reach very high levels, de- pending on the given use case. This is because biomarker positive predictive value (PPV) and negative predictive value (NPV), as previously noted [44•], depend on the prevalence (i.e., base rate) of the condition of interest in a particular setting (i.e., Bayes’ theorem). Specifically, bio- marker PPV increases with prevalence, while NPV in- creases as prevalence decreases. This relationship is illus- trated graphically in Fig. 1 using the sensitivity (81%) and specificity (75%) of a structural brain biomarker for pain we have previously reported [23••]. As a result, even the highest performing chronic pain biomarkers previously re- ported (e.g., 92% sensitivity, 92% specificity [16•]) will have high rates of false positives in application settings with low prevalence and high rates of false negatives in settings with high prevalence. Investigators need to take use case into account when reporting biomarker perfor- mance and judging its adequacy. Finally, self-report of pain symptomatology is ultimately the basis for diagnosing and assessing the severity of chronic pain conditions (so-called “Gold Standard”). Therefore, the predictive utility of biomarkers is necessarily limited by the accuracy of the existing diagnostic gold standard [8•, 45]. It is conceivable that subgroups within a diagnostic category could be identified solely based upon physiological measurements using unsupervised ML methods and later validated upon self- report, thereby circumventing this issue. However, to our knowledge, no previous neuroimaging-based biomarker stud- ies for chronic pain have used this approach. Pain Biomarkers Based on Brain Imaging Structural Biomarkers Chronic pain brain biomarkers derived from structural MRI are dependent on the assumption that chronic pain conditions are associated with abnormalities in brain structure that either pre-date or are the result of pain chronification. Brain structure can be characterized in terms of grey matter features, which reflects integrity of neuronal cell bodies, or white matter fea- tures that reflect axonal integrity. Note that although white matter perturbations have been demonstrated in a variety of chronic pain conditions [46–48], to our knowledge, ML algo- rithms have yet to be applied to these data in order to generate chronic pain classifiers. Thus, we will focus on studies utiliz- ing grey matter features. Grey matter structure can be assessed in several ways. One common approach is the use of voxel-based morphometry (VBM) on high-resolution T1-weighted images. VBM pro- duces estimates of grey matter density for each voxel and subject after warping to fit a standardized template brain [49, 50]. Another typical technique is to assess grey matter char- acteristics in cortical (thickness, volume, surface area, or mean curvature) or subcortical (volume) structures by assigning a neuroanatomical label to each brain voxel using a probabilistic Fig. 1 Illustration of the relationship between chronic pain prevalence and the positive/negative predictive value of biomarkers using sensitivity (81%) and specificity (75%) values derived from Robinson et al. [23••]. Positive predictive value (PPV) increases with prevalence, while negative predictive value (NPV) decreases as prevalence increases. Thus, during real-world application of brain biomarkers for chronic pain, performance will depend largely on the expected proportion of individuals with the condition in question 5 Page 4 of 9 Curr Rheumatol Rep (2017) 19:5
  • 5. atlas [51, 52]. Though other techniques and measures are available for assessing grey matter structure, efforts to con- struct biomarkers for chronic pain conditions based on grey matter structure have largely relied on grey matter density [29, 53] or thickness/volume/surface area/curvature [23••, 30]. Structural brain biomarkers for chronic pain build on a significant literature demonstrating both atrophy and hy- pertrophy in chronic pain patients in numerous pain- related brain regions. For example, chronic low back pain (cLBP) has been associated with lower grey matter densi- ty compared to controls in the dorsolateral prefrontal cor- tex (DLPFC), a region associated with cognitive/ evaluative function and pain modulation [54]. At the same time, FM has been associated with both increased grey matter volume in striatum, orbitofrontal cortex, and cere- bellum [55], and lower volume in anterior cingulate cor- tex, amygdala, thalamus, superior temporal gyrus, supple- mentary motor cortex, and insula [55–58]. Although not directly contradicting one another, these studies differed somewhat with regard to regions showing significant dif- ferences between FM patients and controls, with differ- ences in anterior cingulate cortex being the most consis- tently observed [56, 58]. The potential utility of these measures as features for classification algorithms to dis- tinguish chronic pain patient groups depends on the as- sumption that reliable commonalities and differences in grey matter morphometrics can be detected between chronic pain conditions. It is also worth noting that the goal of chronic pain biomarker studies is distinct from between-group analyses of structural differences because they are focused specifically on identifying optimal com- binations of features that best separate patient groups and/ or healthy controls. Classifier Studies Based on Structural Brain Abnormalities ML classifier studies based on structural brain features have been conducted in several chronic pain conditions, including CPP, IBS, FM, and cLBP, using samples from 26 to 160 participants. In each case, samples were com- posed of ∼50% splits of patients to controls. Performance of these potential biomarkers has differed substantially be- tween studies, with sensitivity and specificity ranging from 65 to 81%. Although brain regions discriminating patient groups from normal controls varied, certain areas were more frequently reported. These included precentral gyrus (primary motor cortex), postcentral gyrus (primary so- matosensory cortex), amygdala, and cuneus. In addition, many discriminating regions were convergent with those identified in previous studies focused on identifying struc- tural differences between chronic pain patients and con- trols (e.g., DLPFC, amygdala, cingulate cortex, insula, etc.,). For further detail regarding sample size, algorithm(s) used, classifier performance, and brain structures with the greatest contribution to performance for studies using structural brain biomarkers, see Table 2. Functional Brain Biomarkers Whereas, high-resolution neuroanatomical data is the basis for studies proposing structural neuroimaging biomarkers of chronic pain, brain activity measures are used to derive functional neuroimaging biomarkers. Functional MRI (fMRI), which refers to noninvasive methods for measur- ing brain function, has two common variants. The first detects changes in cerebral blood oxygenation (BOLD), and the second, arterial spin labeling (ASL), measures changes in regional cerebral blood flow (rCBF). Although neither of these methods directly measure neuro- nal activity, they are both considered surrogate markers of such activity (for review of these technologies, see [61]). These methods excel in their ability to provide spatial in- formation about where changes in BOLD response or rCBF occur over the course of time; however, they are intrinsically limited in temporal resolution, or accuracy in determining exactly when the neuronal activation oc- curred, due to the physiological characteristics of blood oxygenation (BOLD) and/or extended repetition times (ASL; [62]). Although ASL has several theoretical advan- tages over BOLD regarding the measurement of brain function [63], most brain classification studies for chronic pain to date have relied on BOLD fMRI. Because functional neuroimaging provides a global mea- sure of brain activity, participants’ mental state during data collection is always important to note. For example, brain activity can be captured during a goal-directed task (e.g., participants are asked to continuously rate their levels of clinical pain or undergo acute painful stimulation), or dur- ing wakeful rest (i.e., resting state) in which participants are not instructed to engage in a particular task. Raw data col- lected during these scans typically undergoes preprocess- ing, or various image and signal correction techniques, to improve the ratio of desired signal to undesired noise [64]. Subsequently, preprocessed data are statistically analyzed at the individual-participant level to determine either 1) whether a significant change in signal occurred for a given region or across the whole brain (i.e., activation), or 2) the coherence in activation among spatially distinct brain re- gions over time (i.e., functional connectivity, FC). Statistically analyzed activation or FC values can be select- ed across the whole brain, or limited to hypothesis-driven regions of interest (i.e., a priori ROIs). In classification stud- ies proposing functional neuroimaging biomarkers, individual-level activation or FC information is then Curr Rheumatol Rep (2017) 19:5 Page 5 of 9 5
  • 6. Table2Characteristicsofmachinelearning-basedclassifierstudiesusingbrainimagingresults StudyPain condition Accuracy (%) Sensitivity (%) Specificity (%) ManuscriptNManuscript baserate(%) Machine learning classifier Aprioriregions ofinterest? Majorregionscontributingtoclassification Structuralbiomarkers Bagarinaoetal.[28]CPP7370736650SVMNo>DensityinLSFG,LPCG,RPCG,LPaHcG,R PaHcG,amygdala,hippocampusinCPP Labusetal.[30]IBS70657516050sPLS-DANo>ThicknessinLPCG,lesserthicknessinR insula;>meancurvatureinRPreCG,lesserinL SCG,Rtemporalsulcus,andLintraparietal sulcus;and<surfaceareainLfusiformgyrus inIBS Robinsonetal.,[23••]FM7681752654LR,MLP, Bayes, SVM,J48 No<VolumeinLamygdalainFM Ungetal.[29]cLBP7676759450SVMNo<GMdensityinRamygdala,LMOG,andR cuneus;greaterGMdensityinRcerebellum, R/Lmiddle/superiortemporalgyrus,LPreCG, LPCG,Rcalcarinesulcus,andRDLPFCincLBP Dynamicbiomarkers Callanetal.[16•]cLBP9292922650SLRYes<LPCGactivationand>LIPCincLBPduringpain Sundermannetal.[59]RA FM 79(vs.HC) 62(vs.HC) 79(vs.RA) NRNR5034 32 SVM SVM kNN YesNR López-Solàetal.[60••]FM9392947251SVM,LRYes>Activityininsula,PCC,precuneus,DMPFC, parahippocampalgyrus <ActivityinDLPFC,primary/secondaryvisual andauditorycortex,lateralcerebellum,basal ganglia,andhypothalamus SFGsuperiorfrontalgyrus,PCGpostcentralgyrus,PaHcGparahippocampalgyrus,PreCGprecentralgyrus,SCGsubcollosalgyrus,MOGmedialorbitalgyrus,DMPFCdorsomedialprefrontalcortex, DLPFCdorsolateralprefrontalcortex,IPCinferiorparietalcortex,RArheumatoidarthritis,FMfibromyalgia,CFSchronicfatiguesyndrome,FMfibromyalgia,IBSirritablebowelsyndrome,cLBPchronic lowbackpain,LASSOleastabsoluteshrinkageandselectionoperator,OAosteoarthritis,CPPchronicpelvicpain,LRlogisticregressionusingridgeestimator,MLPmultilayerperceptron,J48J48decision tree,PPVpositivepredictivevalue,NPVnegativepredictivevalue,SLRsparselogisticregression,sPLS-DAsparsepartialleastsquaresfordiscriminantanalysis,IPCinferiorparietalcortex,NRnot reported 5 Page 6 of 9 Curr Rheumatol Rep (2017) 19:5
  • 7. entered into computational models, which classify individ- uals into distinct groups based on these values. Classifier Studies Based on Functional Brain Abnormalities Compared to structural brain abnormalities, fewer studies have used fMRI activation or functional connectivity metrics as features in classification models for chronic pain patients. Using measures of functional brain activation in pain-related brain regions during repeated 14-s blocks of experimental electrical pain induction, Callan et al. [16•] achieved 92% accuracy, sensitivity, and specificity classifying cLBP and healthy controls. The two most informative regions reported by Callan et al. [16•] have been strongly implicated in pain discrimination: (primary somatosensory cortex) and attention (inferior parietal cortex) [65]. In a separate study using resting state fMRI [59], RA and FM patients could be discriminated from HC with 79 and 62% accuracy, respectively, using mea- sures of functional connectivity between structures associated with stimulus evaluation (i.e., the “salience network” [SN]) and internal focus/mind wandering (i.e., “default mode net- work” [DMN]). These networks have been implicated in pain processing (SN) or reported as perturbed in chronic pain states (DMN). Interestingly, RA and FM patients could also be dis- criminated from each other with 79% accuracy, suggesting that, despite potential commonalities in neuronal plasticity, the functional neural correlates of certain chronic pain disor- ders may be sufficiently distinct to enable their separation [59]. Finally, in a recent fMRI study using ML [60••], the investigators could discriminate FM patients from HC with 93% accuracy based on their brain activation associated with painful pressure stimuli [66] and non-painful multisensory stimulation (e.g., simultaneous auditory, tactile, and visual stimuli). Overall, the rapid improvement of fMRI-based “pain signatures” has not only helped our mechanistic understand- ing of chronic pain but will also benefit the diagnosis of FM and other chronic pain conditions. Summary Taken together, a limited number of studies (see Table 1) have tested the ability of ML algorithms to discern chronic pain patients from healthy controls using structural or functional brain abnormalities. However, interest in ML approaches for chronic pain diagnosis and classification is growing due to their purported potential to help elucidate chronic pain mech- anisms, identify resilient or vulnerable subgroups, improve clinical decision making, predict treatment outcome, or aug- ment [29] or even replace self-report (for critical discussion, see [8•]). However, as previously discussed, certain caveats apply to the practical application of biomarkers even where performance metrics may be very high. Conclusions Interest in the development of clinical and ML-based bio- markers for chronic musculoskeletal pain conditions derived from structural and functional neuroimages has increased sub- stantially in recent years. Current reports describe novel bio- markers capable of separating patient groups and healthy con- trols with accuracies ranging from 70 to 93%. Such studies provide valuable mechanistic information regarding both the unique and common neural correlates of these conditions, with the potential to highlight differences between both mus- culoskeletal pain patient groups and controls to which tradi- tional statistical approaches may not be sensitive, or identify mechanistic subgroups within certain pain conditions. However, at this time, critical theoretical, practical, and ethical concerns preclude the replacement of patient self-report for the diagnosis of chronic musculoskeletal pain with brain imaging-derived biomarkers, as self-report remains the gold standard for pain assessment [8•]. Acknowledgements This work was supported by NIH grant R01 NR014049-01 Compliance with Ethical Standards Conflict of Interest The authors declare that they have no conflict of interest. Human and Animal Rights and Informed Consent This article does not contain any studies with human or animal subjects performed by any of the authors. References Papers of particular interest, published recently, have been highlighted as: • Of importance •• Of major importance 1. Institute of Medicine. Relieving pain in America: a blueprint for transforming prevention, care, education, and research. Washington, DC: National Academies Press; 2011. 2. Wolfe F, Clauw DJ, Fitzcharles MA, Goldenberg DL, Katz RS, Mease P, et al. The American College of Rheumatology preliminary diagnostic criteria for fibromyalgia and measurement of symptom severity. Arthritis Care Res. 2010;62(5):600–10. doi:10.1002 /acr.20140. 3. Shamir L, Ling SM, Scott Jr WW, Bos A, Orlov N, Macura TJ, et al. Knee x-ray image analysis method for automated detection of osteoarthritis. IEEE Trans Biomed Eng. 2009;56(2):407–15. doi:10.1109/TBME.2008.2006025. 4. Kirkwood RN, Resende RA, Magalhaes CM, Gomes HA, Mingoti SA, Sampaio RF. Application of principal component analysis on gait kinematics in elderly women with knee osteoarthritis. Rev Bras Fis. 2011;15(1):52–8. Curr Rheumatol Rep (2017) 19:5 Page 7 of 9 5
  • 8. 5. Frize M, Ogungbemile A. Estimating rheumatoid arthritis activity with infrared image analysis. Stud Health Technol Inform. 2012;180:594–8. 6. Frampton D, Kerr J, Harrison TJ, Kellam P. Assessment of a 44 gene classifier for the evaluation of chronic fatigue syndrome from peripheral blood mononuclear cell gene expression. PLoS One. 2011;6(3):e16872. doi:10.1371/journal.pone.0016872. 7. Apkarian AV, Hashmi JA, Baliki MN. Pain and the brain: specific- ity and plasticity of the brain in clinical chronic pain. Pain. 2011;152(3 Suppl):S49–64. doi:10.1016/j.pain.2010.11.010. 8.• Robinson ME, Staud R, Price DD. Pain measurement and brain activity: will neuroimages replace pain ratings? J Pain: Off J Am Pain Soc. 2013;14(4):323–7. doi:10.1016/j.jpain.2012.05.007. This critical review summarized the limitations of neuroimaging to replace pain self-report. It also emphasized that neuroimaging will never replace pain self-report except in individuals with mental conditions or limited levels of consciousness. 9. Wortolowska K. How neuroimaging can help us to visualise and quantify pain? Eur J Pain Suppl. 2011;5(S2):323–7. 10. Bushnell MC, Ceko M, Low LA. Cognitive and emotional control of pain and its disruption in chronic pain. Nat Rev Neurosci. 2013;14(7):502–11. doi:10.1038/nrn3516. 11. Brekelmans MP, Fens N, Brinkman P, Bos LD, Sterk PJ, Tak PP, et al. Smelling the diagnosis: the electronic nose as diagnostic tool in inflammatory arthritis. A case-reference study. PloS One. 2016;11(3):e0151715. doi:10.1371/journal.pone.0151715. 12. Rizzo G, Raffeiner B, Coran A, Ciprian L, Fiocco U, Botsios C, et al. Pixel-based approach to assess contrast-enhanced ultrasound kinetics parameters for differential diagnosis of rheumatoid arthritis. J Med Imaging (Bellingham). 2015;2(3):034503. doi:10.1117/1. JMI.2.3.034503. 13. Mackey SC. Central neuroimaging of pain. J Pain: Off J Am Pain Soc. 2013;14(4):328–31. doi:10.1016/j.jpain.2013.01.001. 14. Borsook D, Becerra L, Hargreaves R. Biomarkers for chronic pain and analgesia. Part 1: the need, reality, challenges, and solutions. Discov Med. 2011;11(58):197–207. 15. Borsook D, Becerra L, Hargreaves R. Biomarkers for chronic pain and analgesia. Part 2: how, where, and what to look for using func- tional imaging. Discov Med. 2011;11(58):209–19. 16.• Callan D, Mills L, Nott C, England R, England S. A tool for clas- sifying individuals with chronic back pain: using multivariate pat- tern analysis with functional magnetic resonance imaging data. PloS One. 2014;9(6):e98007. doi:10.1371/journal.pone.0098007. This study employed supervised machine learning techniques, specifically sparse logistic regression, to train a classifier of chronic back pain based on contrast images using a leave-one- out cross-validation procedure. It correctly classified 92.3% of the chronic pain group and 92.3% of the normal controls. 17. Lu HC, Hsieh JC, Lu CL, Niddam DM, Wu YT, Yeh TC, et al. Neuronal correlates in the modulation of placebo analgesia in experimentally-induced esophageal pain: a 3T-fMRI study. Pain. 2010;148(1):75–83. doi:10.1016/j.pain.2009.10.012. 18. Pustilnik AC. Imaging brains, changing minds: how pain neuroim- aging can inform the law. Alabama Law Rev. 2015;66(5). 19.• Letzen JE, Boissoneault J, Sevel LS, Robinson ME. Test-retest reliability of pain-related functional brain connectivity compared to pain self-report. Pain. 2016;157(3):546–51. doi:10.1097/j. pain.0000000000000356. This study examined the test-retest re- liability for functional connectivity MRI (fcMRI) of pain- related brain regions and self-reported pain. Intraclass corre- lations coefficients for fcMRI values varied widely (range = −.174–.766), whereas intraclass correlations coeffi- cients for VAS scores ranged from .906 to .947. Overall, self- reported pain was more reliable than fcMRI data. 20. Nielsen CS, Price DD, Vassend O, Stubhaug A, Harris JR. Characterizing individual differences in heat-pain sensitivity. Pain. 2005;119(1–3):65–74. doi:10.1016/j.pain.2005.09.018. 21. Slesinger D, Archer RP, Duane W. MMPI-2 characteristics in a chronic pain population. Assessment. 2002;9(4):406–14. 22. Takekawa KS, Goncalves JS, Moriguchi CS, Coury HJ, Sato TO. Can a self-administered questionnaire identify workers with chronic or recurring low back pain? Ind Health. 2015;53(4):340–5. doi:10.2486/indhealth.2014-0241. 23.•• Robinson ME, O’Shea AM, Craggs JG, Price DD, Letzen JE, Staud R. Comparison of machine classification algorithms for fibromyal- gia: neuroimages versus self-report. J Pain: Off J Am Pain Soc. 2015;16(5):472–7. doi:10.1016/j.jpain.2015.02.002. Separate models representing brain volumes, mood ratings, and pain intensity ratings were estimated across several ML algorithms. Structural magnetic resonance imaging data from fibromyalgia patients and healthy controls and self-report data of pain intensity and mood were used. Classification accuracy of brain volumes ranged from 53 to 76%, whereas mood and pain intensity ratings ranged from 79 to 96% and 83 to 96%, respectively. Overall, models derived from self-report data outperformed neuroimaging models. 24. Friend R, Bennett RM. Distinguishing fibromyalgia from rheuma- toid arthritis and systemic lupus in clinical questionnaires: an anal- ysis of the revised Fibromyalgia Impact Questionnaire (FIQR) and its variant, the Symptom Impact Questionnaire (SIQR), along with pain locations. Arthritis Res Ther. 2011;13(2):R58. doi:10.1186 /ar3311. 25. Perrot S, Bouhassira D, Fermanian J, Cercle d’Etude de la Douleur en R. Development and validation of the Fibromyalgia Rapid Screening Tool (FiRST). Pain. 2010;150(2):250–6. doi:10.1016/j. pain.2010.03.034. 26. Cortes C, Vapnik V. Support-vector networks. Mach Learn. 1995;20(3):273–97. 27. Pontil M, Verri A. Properties of support vector machines. Neural Comput. 1998;10(4):955–74. 28. Bagarinao E, Johnson KA, Martucci KT, Ichesco E, Farmer MA, Labus J, et al. Preliminary structural MRI based brain classification of chronic pelvic pain: a MAPP network study. Pain. 2014;155(12): 2502–9. doi:10.1016/j.pain.2014.09.002. 29. Ung H, Brown JE, Johnson KA, Younger J, Hush J, Mackey S. Multivariate classification of structural MRI data detects chronic low back pain. Cereb Cortex. 2014;24(4):1037–44. doi:10.1093 /cercor/bhs378. 30. Labus JS, Van Horn JD, Gupta A, Alaverdyan M, Torgerson C, Ashe-McNalley C, et al. Multivariate morphological brain signa- tures predict patients with chronic abdominal pain from healthy control subjects. Pain. 2015;156(8):1545–54. doi:10.1097/j. pain.0000000000000196. 31. Le Cessie S, Van Houwelingen JC. Ridge estimators in logistic regression. Appl Stat. 1992;41(1):191–201. 32. Langley P, Iba W, Thompson K. An analysis of Bayesian classifiers. AAAI. 1992;90:223–8. 33. Aha DW, Kibler D, Albert MK. Instance-based learning algorithms. Mach Learn. 1991;6(1):37–66. doi:10.1023/A:1022689900470. 34. Arora R. Comparative analysis of classification algorithms on differ- ent datasets using WEKA. Int J Comput Appl. 2012;54(13):21–5. 35. Quinlan JR. C4.5 programs for machine learning. San Mateo: Morgan Kaufmann; 1992. 36. Tibshirani R. Regression shrinkage and selection via the lasso. J Roy Stat Soc B Met. 1996;58(1):267–88. 37. Kononenko I. Machine learning for medical diagnosis: history, state of the art and perspective. Artif Intell Med. 2001;23(1):89–109. 38. Nair SS, French RM, Laroche D, Thomas E. The application of machine learning algorithms to the analysis of electromyographic 5 Page 8 of 9 Curr Rheumatol Rep (2017) 19:5
  • 9. patterns from arthritic patients. IEEE Trans Neural Syst Rehabil Eng. 2010;18(2):174–84. doi:10.1109/TNSRE.2009.2032638. 39. Mamoshina P, Vieira A, Putin E, Zhavoronkov A. Applications of deep learning in biomedicine. Mol Pharm. 2016;13(5):1445–54. doi:10.1021/acs.molpharmaceut.5b00982. 40. Schnack HG, Kahn RS. Detecting neuroimaging biomarkers for psychiatric disorders: sample size matters. Front Psych. 2016;7: 50. doi:10.3389/fpsyt.2016.00050. 41. Siegel JS, Ramsey LE, Snyder AZ, Metcalf NV, Chacko RV, Weinberger K, et al. Disruptions of network connectivity predict impairment in multiple behavioral domains after stroke. Proc Natl Acad Sci U S A. 2016. doi:10.1073/pnas.1521083113. 42. Woo CW, Wager TD. Neuroimaging-based biomarker discovery and validation. Pain. 2015;156(8):1379–81. doi:10.1097/j. pain.0000000000000223. 43. Deo RC. Machine learning in medicine. Circulation. 2015;132(20): 1920–30. doi:10.1161/CIRCULATIONAHA.115.001593. 44.• Robinson M, Boissoneault J, Sevel L, Letzen J, Staud R. The effect of base rate on the predictive value of brain biomarkers. J Pain. 2016;17(6):637–41. doi:10.1016/j.jpain.2016.01.476. Results of this study strongly suggest that many proposed brain biomarkers perform quite poorly when realistic illness prevalence base rates are taken into account. 45. Lueken U, Zierhut KC, Hahn T, Straube B, Kircher T, Reif A, et al. Neurobiological markers predicting treatment response in anxiety disorders: a systematic review and implications for clinical applica- tion. Neurosci Biobehav Rev. 2016;66:143–62. doi:10.1016/j. neubiorev.2016.04.005. 46. Buckalew N, Haut MW, Aizenstein H, Morrow L, Perera S, Kuwabara H, et al. Differences in brain structure and function in older adults with self-reported disabling and nondisabling chronic low back pain. Pain Med. 2010;11(8):1183–97. doi:10.1111 /j.1526-4637.2010.00899.x. 47. Geha PY, Baliki MN, Harden RN, Bauer WR, Parrish TB, Apkarian AV. The brain in chronic CRPS pain: abnormal gray-white matter interactions in emotional and autonomic regions. Neuron. 2008;60(4):570–81. doi:10.1016/j.neuron.2008.08.022. 48. Gustin SM, Peck CC, Cheney LB, Macey PM, Murray GM, Henderson LA. Pain and plasticity: is chronic pain always associ- ated with somatosensory cortex activity and reorganization? J Neurosci: Off J Soc Neurosci. 2012;32(43):14874–84. doi:10.1523/JNEUROSCI.1733-12.2012. 49. Greve DN. An absolute beginner’s guide to surface- and voxel- based morphometric analysis. ISMRM 19th Annual Meeting & Exhibition. 2011;7–13. 50. Ashburner J, Friston K. Morphometry. In: Ashburner J, editor. Human brain function. 2nd ed. Cambridge: Academic; 2004. p. 707–22. 51. Fischl B, Liu A, Dale AM. Automated manifold surgery: construct- ing geometrically accurate and topologically correct models of the human cerebral cortex. IEEE Trans Med Imaging. 2001;20(1):70– 80. doi:10.1109/42.906426. 52. Fischl B, Salat DH, Busa E, Albert M, Dieterich M, Haselgrove C, et al. Whole brain segmentation: automated labeling of neuroana- tomical structures in the human brain. Neuron. 2002;33(3):341–55. 53. Baliki MN, Schnitzer TJ, Bauer WR, Apkarian AV. Brain morpho- logical signatures for chronic pain. PLoS One. 2011;6(10):e26010. doi:10.1371/journal.pone.0026010. 54. Apkarian AV, Sosa Y, Sonty S, Levy RM, Harden RN, Parrish TB, et al. Chronic back pain is associated with decreased prefrontal and thalamic gray matter density. J Neurosci: Off J Soc Neurosci. 2004;24(46):10410–5. doi:10.1523/JNEUROSCI.2541-04.2004. 55. Schmidt-Wilcke T, Luerding R, Weigand T, Jurgens T, Schuierer G, Leinisch E, et al. Striatal grey matter increase in patients suffering from fibromyalgia—a voxel-based morphometry study. Pain. 2007;132 Suppl 1:S109–16. doi:10.1016/j.pain.2007.05.010. 56. Burgmer M, Gaubitz M, Konrad C, Wrenger M, Hilgart S, Heuft G, et al. Decreased gray matter volumes in the cingulo-frontal cortex and the amygdala in patients with fibromyalgia. Psychosom Med. 2009;71(5):566–73. doi:10.1097/PSY.0b013e3181a32da0. 57. Puri BK, Agour M, Gunatilake KD, Fernando KA, Gurusinghe AI, Treasaden IH. Reduction in left supplementary motor area grey matter in adult female fibromyalgia sufferers with marked fatigue and without affective disorder: a pilot controlled 3-T magnetic res- onance imaging voxel-based morphometry study. J Int Med Res. 2010;38(4):1468–72. 58. Robinson ME, Craggs JG, Price DD, Perlstein WM, Staud R. Gray matter volumes of pain-related brain areas are decreased in fibro- myalgia syndrome. J Pain: Off J Am Pain Soc. 2011;12(4):436–43. doi:10.1016/j.jpain.2010.10.003. 59. Sundermann B, Burgmer M, Pogatzki-Zahn E, Gaubitz M, Stuber C, Wessolleck E, et al. Diagnostic classification based on functional connectivity in chronic pain: model optimization in fibromyalgia and rheumatoid arthritis. Acad Radiol. 2014;21(3):369–77. doi:10.1016/j.acra.2013.12.003. 60.•• López-Solà M, Woo CW, Pujol J, Deus J, Harrison BJ, Monfort J, Wager TD. Towards a neurophysiological signature for fibromyalgia. Pain. 2016. A ‘Multisensory’ classifier trained on non-painful sen- sory stimulation revealed a brain signature for FM with high sensitivity and specificity. This study characterized individual FM patients based on symptom-related brain features. 61. Detre JA, Wang J. Technical aspects and utility of fMRI using BOLD and ASL. Clin Neurophysiol. 2002;113(5):621–34. 62. Howseman AM, Bowtell RW. Functional magnetic resonance im- aging: imaging techniques and contrast mechanisms. Philos Trans R Soc Lond B Biol Sci. 1999;354(1387):1179–94. doi:10.1098 /rstb.1999.0473. 63. Chen JJ, Jann K, Wang DJ. Characterizing resting-state brain func- tion using arterial spin labeling. Brain Connect. 2015;5(9):527–42. doi:10.1089/brain.2015.0344. 64. Smith SM, Jenkinson M, Woolrich MW, Beckmann CF, Behrens TE, Johansen-Berg H, et al. Advances in functional and structural MR image analysis and implementation as FSL. NeuroImage. 2004;23 Suppl 1:S208–19. doi:10.1016/j.neuroimage.2004.07.051. 65. Giesecke T, Gracely RH, Grant MA, Nachemson A, Petzke F, Williams DA, et al. Evidence of augmented central pain processing in idiopathic chronic low back pain. Arthritis Rheum. 2004;50(2): 613–23. doi:10.1002/art.20063. 66. Wager TD, Atlas LY, Lindquist MA, Roy M, Woo CW, Kross E. An fMRI-based neurologic signature of physical pain. N Engl J Med. 2013;368(15):1388–97. Curr Rheumatol Rep (2017) 19:5 Page 9 of 9 5