Anda di halaman 1dari 7

REVIEW

CURRENT
OPINION Neuroimaging of HIV-associated neurocognitive
disorders (HAND)
Beau M. Ances a,b,c and Dima A. Hammoud d

Purpose of review
HIV enters the brain after initial infection, and with time can lead to HIV-associated neurocognitive
disorders (HAND). Although the introduction of combination antiretroviral therapy has reduced the more
severe forms of HAND, milder forms are still highly prevalent. The gold standard for HAND diagnosis
remains detailed neuropsychological performance testing but additional biomarkers (including
neuroimaging) may assist in early detection of HAND.
Recent findings
We review the application of recently developed noninvasive MRI and PET techniques in HIV individuals.
In particular, magnetic resonance spectroscopy may be more sensitive than conventional MRI alone in
detecting HIV associated changes. Diffusion tensor imaging has become increasingly popular for assessing
changes in white matter structural integrity due to HIV. Both functional MRI and PET have been limitedly
performed but could provide keys for characterizing neuropathophysiologic changes due to HIV.
Summary
It is hoped that continued progress will allow novel neuroimaging methods to be included in future HAND
management guidelines.
Keywords
diffusion tensor imaging, functional MRI, HIV, neuroimaging, PET, volumetrics

INTRODUCTION neurocognitive disorder, or HIV-associated demen-


More than 1 million individuals in the USA and over tia (HAD) [12]. Compared to other neurodegenera-
40 million people worldwide are infected with the tive disorders (e.g., Alzheimers disease), additional
HIV. Combination antiretroviral treatment (cART) biomarkers have yet to be added to HAND research
has transformed HIV from a rapidly fatal disease to a criteria.
more manageable chronic condition [13]. As a Neuroimaging could have increased utility in
result, HIV-infected (HIV) individuals receiving the diagnosis and management of HAND. A variety
cART have almost as long a lifespan as HIV-unin- of novel neuroimaging techniques have been devel-
fected (HIV) individuals [4]. A majority of HIV oped and are currently performed in the research
patients will be greater than 50 years old by 2015 [5]. setting. Of note, MRI techniques [including mag-
Despite these advances, HIV cannot be eradi- netic resonance spectroscopy (MRS), volumetrics,
cated from the brain with persistent reservoirs often diffusion tensor imaging (DTI), and functional]
&
remaining [6 ]. The continued presence of HIV-
associated neurocognitive disorders (HAND) despite
a
cART could result from nonmutually exclusive fac- Department of Neurology, School of Medicine, bDepartment of Radi-
tors, including irreversible injury prior to initiating ology, cDepartment of Biomedical Engineering, Washington University in
St Louis, Saint Louis, Missouri and dCenter for Infectious Disease
cART, persistent HIV-1 RNA in the brain [7], anti-
Imaging, Radiology and Imaging Sciences, Clinical Center, National
retroviral treatment toxicities [810], and/or persist- Institutes of Health, Bethesda, Maryland, USA
ent low-level inflammation [11]. Currently, the Correspondence to Beau M. Ances, MD, PhD, Associate Professor in the
diagnosis of HAND requires neuropsychological Department of Neurology, Box 8111, 660 South Euclid Ave, Saint Louis,
performance testing and self-reported assessment MO 63110, USA. Tel: +1 314 747 8423; fax: +1 314 747 8427; e-mail:
of activities of daily living with the following bances@wustl.edu
classifications used: neuropsychologically normal, Curr Opin HIV AIDS 2014, 9:545551
asymptomatic neurocognitive impairment, mild DOI:10.1097/COH.0000000000000112

1746-630X 2014 Wolters Kluwer Health | Lippincott Williams & Wilkins www.co-hivandaids.com

Copyright Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
HIV neurology

cART can lead to significant improvements, but


KEY POINTS not normalization of MRS metabolite levels [2325].
 The effects of HIV in the brain can be noninvasively Early treatment with cART may be neuroprotective
assessed by structural (e.g., volumetrics and DTI) and and mitigate some of the changes seen soon after
functional (e.g., blood-oxygen-level-dependent imaging) seroconversion. However, certain antiretrovirals
MRI. may cause mitochondrial toxicity and impair
neuronal function [9,26].
 PET of glucose metabolism, neurotransmitter systems
abnormalities, or amyloid deposition could provide HIV patients are living longer and growing
additional understanding of the neuropathophysio- older due to cART. A number of MRS studies
logical changes associated with HIV. have, therefore, investigated the interaction
between HIV and aging [27]. HIV patients have
 Novel neuroimaging methods could be added to
significant changes in brain metabolites with
current criteria for defining HAND. These methods may
also help in evaluating the efficacy of cART regimens. levels measured equivalent to those seen in HIV
controls at least 1015 years older [28]. In general, no
 Neuroimaging studies that are longitudinal; have larger interaction has been observed between HIV and
sample sizes of both HIV infected (HIV) and HIV aging [2].
uninfected (HIV); and include HIV patients of
Overall, MRS changes may be more sensitive
different disease durations are needed.
than conventional MRI alone in detecting changes
associated with HIV. MRS could augment current
neuroimaging protocols but local implementation
and PET have been utilized in HIV individuals. of sequences is required. In the future, MRS could be
This review is not meant to be comprehensive. used to evaluate the efficacy of certain therapeutics.
Instead, it briefly discusses recent results (within However, most MRS studies have been cross-
the past 3 years) using some of these neuroimaging sectional and have primarily focused on specific
methods. regions of interest. Additional longitudinal studies
that focus on HIV patients as they transition across
different disease states are needed [29].
MAGNETIC RESONANCE SPECTROSCOPY
The most common neuroimaging method for
studying neuroHIV in the precART and postcART STRUCTURAL NEUROIMAGING-
eras has been MRS [1316]. MRS detects the signal VOLUMETRICS AND DIFFUSION TENSOR
produced by protons of specific molecules within a IMAGING
volume of brain. Molecules that are typically Volumetric MRI can assess brain structural differ-
measured include: first, N-acetyl-aspartate a ences between HIV and HIV individuals [30].
neuronal marker; second, choline a marker of This technique can concentrate on either specific
cellular proliferation and inflammatory response; brain structures or relatively large brain areas [31].
third, creatine a measure of brain energy meta- In the precART era, significant volume loss was seen
bolism and reference marker; and fourth, myo- in the basal ganglia, posterior cortex, and white
inositol a marker of gliosis. MRS can provide matter of HIV patients compared with HIV
key insights into longitudinal changes in brain controls [3234]. The greatest changes in volumet-
metabolites as an individual progresses from rics were seen in more advanced stages of HAND
primary (1 year since seroconversion) to chronic [35]. In the cART era, both subcortical and cortical
(>1 year since seroconversion) infection. Soon atrophy have been observed in HIV patients
after seroconversion, MRS metabolites are affected [3638] suggesting that brain volume loss can still
[14,1719] with observed neuroimaging changes occur despite the initiation of effective treatment
correlating with inflammation [17] and neuronal [20,30]. More recently, volumetric changes have
injury [19]. Changes seen due to HIV are often been shown to correlate with the degree of cognitive
observed within both subcortical (basal ganglia) impairment and virologic markers. Poorer neuro-
and cortical (frontal grey/white matter and parietal cognitive performance has been associated with
grey matter) compared with HIV controls. HIV
&
smaller brain volumes [22,37,39,40,41 ,4246]
patients with chronic infection have reduced and greater viral burden. In addition, impaired
N-acetyl-aspartate and concomitant increased immune response (nadir CD4 T lymphocyte
choline and myo-inositol [15,2023]. MRS meta- counts) has been associated with greater atrophy
bolite changes in chronically infected HIV patients
& &
[20,38,40,41 ,46,47 ,48,49]. Cortical brain atrophy
vary according to HAND status with HAD patients and expansion of the third ventricle has been
showing the greatest changes [15,21].
&&
observed soon after seroconversion [50 ].

546 www.co-hivandaids.com Volume 9  Number 6  November 2014

Copyright Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Neuroimaging of HAND Ances and Hammoud

Diffusion tensor imaging DTI technique has activation within the left inferior frontal gyrus
become increasingly popular in the assessment of and caudate nucleus compared with HIV controls
&&
white matter structural integrity in the setting of [74 ]. Dysfunction in the fronto-striatal network
HIV. This neuroimaging technique measures the was qualitatively related to degree of neurocognitive
diffusion of water molecules [45,5153]. In the iso- impairment. Differences between HIV and HIV
tropic state water motion is equal in all directions individuals can also be seen at rest using BOLD
(e.g., cerebrospinal fluid). In brain tissues the move- imaging. In particular, functional correlations
ment of water is anisotropic, with diffusion greater between brain networks are significantly reduced
along the length of fiber tracts compared with per- in HIV patients and a signature of the disease
pendicular to them [54]. For each voxel, a tensor is may be present that is different than other neuro-
calculated that describes the three-dimensional degenerative disorders. The effects of HIV and aging
shape of water diffusion. Fiber direction is indicated on BOLD resting state functional correlations were
&
by the tensors main eigenvector. Mean diffusivity shown to be independent of each other [75 ]. How-
reflects the average diffusion in three axes. Frac- ever, the effects of cART have not been assessed
tional anisotropy assesses the general shape of the using BOLD imaging. Additional studies of cerebral
ellipsoid [55]. blood flow have nicely complemented existing
Most studies have shown that HIV leads to an BOLD studies and have demonstrated a reduction
increase in mean diffusivity and a decrease in frac- in resting cerebral blood flow in HIV individuals
tional anisotropy within white matter tracts. How- compared with HIV controls [76,77].
ever, subtle differences may exist as to where Overall, functional neuroimaging studies have
changes are observed depending on the study been performed in a limited number of HIV
&
[5658,59 ]. To date, no studies have assessed DTI patients. Additional longitudinal fMRI studies are
changes soon after seroconversion. Typically, HIV needed to determine if these techniques could be
individuals receiving cART (HIV/cART) and used to follow HIV individuals. Future, BOLD
those nave to cART (HIV/cART) have been studies could evaluate the efficacy of various treat-
merged and compared with HIV controls. Conflict- ment regimens for HAND.
ing results have been observed in the few studies
that have investigated the impact of cART using DTI
&
[60,61,62 ]. POSITRON EMISSION TOMOGRAPHY
Overall, quantitative volumetric changes may Fluorodeoxyglucose (FDG)-PET imaging is a com-
be more sensitive than current conventional MRI monly used technique that measures the metabolic
evaluation. In the future, DTI could be used to activity of various cells/tissues, such as in neoplastic
evaluate the efficacy of certain therapeutics but diseases. In the brain, evaluation of FDG uptake
additional longitudinal studies that focus on HIV by neurons has been utilized in multiple central
patients soon after seroconversion are needed. nervous system (CNS) disorders, such as Alzheimers
disease and Parkinsons disease [78]. In neuroHIV,
early FDG-PET studies demonstrated lower cerebral
FUNCTIONAL MAGNETIC RESONANCE metabolic rate for glucose consumption in HIV
IMAGING patients compared with age-matched HIV
Studies are now starting to utilize blood oxygen level patients, despite the lack of structural abnormalities
dependent (BOLD) functional MRI (fMRI) to inves- on MRI [79]. This probably was the first insight into
tigate the effects of HIV on brain function [63]. early metabolic changes can occur with HIV infec-
Changes in the BOLD response for a particular tion without gross structural volumetric loss. Sub-
stimulus can indirectly reveal the coupling between sequent FDG-PET studies [8082] have described
neuronal activity and cerebral blood flow within two unique metabolic signatures of glucose abnor-
certain brain regions [64]. HIV patients have malities associated with HIV infection. The first was
greater BOLD activity in the parietal lobes for a a hypermetabolic state, particularly in the striatum,
simple attention task and greater frontal and pari- which appeared to provide a disease-specific
etal activation during more complex attention tasks measure of early CNS involvement, despite normal
[65]. These BOLD changes in HIV patients may motor function on neuropsychological perform-
reflect the recruitment of surrounding areas to meet ance testing. This was assumed to reflect abnormal
cognitive requirements [24,28,42,6573]. A recent functional connectivity within subcortical areas
systematic meta-analysis of BOLD fMRI studies [82]. A second pattern was seen during chronic
using various functional tasks in HIV patients stages and was characterized by generalized hypo-
was recently performed using activation likelihood metabolism in both cortical and subcortical regions.
estimation. HIV patients had greater functional These changes correlated with age, cerebral atrophy,

1746-630X 2014 Wolters Kluwer Health | Lippincott Williams & Wilkins www.co-hivandaids.com 547

Copyright Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
HIV neurology

and neurocognitive status [81]. A switch from the some to question the reliability of this ligand. Sub-
hypermetabolic to hypometabolic states in subcort- sequent attempts have been made to use second-
ical areas (e.g., basal ganglia) was associated with generation TSPO-PET ligands with higher specific to
changes in functional deficits and progression to nonspecific binding ratios. However, these second
dementia [82]. generation agents have different binding affinities
In the cART era, more subtle FDG-PET changes within HIV controls. In particular, a genetic poly-
&&
have been observed [83,84 ] and can still be seen in morphism of the TSPO receptor has been discovered
optimally treated HIV patients with virologic and could affect the binding potential of these
&&
suppression. At least half of HIV patients on cART various ligands [92 ,93]. This discovery has further
can have varying degrees of hypometabolism in the complicated the analysis of TSPO-PET imaging as
mesial frontal lobes [83]. These abnormalities patients must be stratified as low-affinity binders,
&&
are accentuated by drug abuse and can lead to high-affinity binders, or heterozygous [92 ]. Cross-
extensive cortical hypometabolism [85]. In contrast sectional comparisons, in theory, cannot be done
to some MRI studies, a synergistic interaction has except within the same binding affinity group.
been observed between aging and HIV with changes Despite these limitations, one study used a second-
generation TSPO ligand (11C-DPA713) to compare
&&
primarily seen in the frontal regions [84 ].
&
Ongoing CNS injury observed despite peri- HIV/cART individuals and HIV controls [94 ].
pheral virologic suppression may reflect persistent Adapting a new method of data analysis based on
low-level neuroinflammation. A few studies have assessing the volume-of-distribution ratios relative
attempted to characterize neuroinflammation to overall gray matter, HIV patients were noted to
using PET ligands that specifically target microglia have higher binding in specific brain regions that
activation. The most commonly used ligand has may reflect localized rather than diffuse glial cell
been 11C-PK11195 that binds to the translocator activation. A novel gray matter normalization
protein (TSPO), a mitochondrial receptor known approach was employed that improves test-retest
to be significantly upregulated in activated micro- reproducibility and may uncover abnormal regional
&
glia [86,87]. Early studies showed significantly findings not seen using traditional methods [94 ].
higher 11C-PK11195 binding in HAD patients com- However, additional studies are needed to validate
pared with HIV controls within five out of eight this method for other TSPO ligands.
brain regions of interest. However within a subgroup A third use of PET imaging in the setting of
of nondemented HIV patients, no significant neuroHIV is the evaluation specific neurotransmit-
increases in binding were seen when compared with ter systems. The first system to be evaluated was
HIV controls [88]. A subsequent larger study using the dopaminergic system. Significantly, lower
11
C-PK11195 failed to show increases in ligand dopamine transporters (DAT) availability was
retention in the brain parenchyma of HIV patients seen in the putamen of HIV patients with HAD
compared with HIV controls [89]. One possible compared with nondemented HIV patients. In
explanation for observed differences could be due addition, lower DAT levels were seen in the ventral
to the variability of the HIV patient populations striatum of HIV patients when compared with
recruited for these studies. Although Hammoud HIV patients [95]. These findings suggested dopa-
et al. [88] included cognitively impaired HIV minergic terminal injury occurs in HIV patients
who were not receiving cART, Wiley et al. [89] only with significant cognitive impairment. Within the
evaluated cognitively normal HIV patients receiv- HIV demented group, higher plasma viral load
ing treatment [89]. A subsequent study by Garvey correlated with lower DAT binding in the caudate
et al. [90] compared chronically infected HIV and putamen. This inverse relationship between
patients with and without concomitant HCV infec- plasma viral burden and DAT availability further
tion. No significant differences were noted between support an HIV-mediated neurotoxicity within
these two groups. However, HIV/cART indivi- dopaminergic nerve terminals [95]. A subsequent
duals had lower ligand-binding potentials in the DAT imaging study assessed the cofounding effects
parietal and frontal regions than HIV/cART- of drug abuse (specifically cocaine) and HIV infec-
patients. A follow-up study by the same group com- tion [96]. Although HIV patients showed lower
pared HIV/cART patients with HIV controls and DAT binding in the putamen compared with
showed clusters of increased ligand binding within HIV patients, irrespective of drug abuse, HIV
the corpus callosum, anterior and posterior cingu- patients with a previous history of drug abuse pro-
late gyrus, temporal gyrus, and frontal regions blems had the lowest DAT values in the caudate.
&&
[91 ]. These results further support the theory of drug
Limitations of 11C-PK11195, including its high abuse contributing to CNS injury observed in HIV
nonspecific binding and high lipophilicity, have led patients. Previous cocaine use may increase the

548 www.co-hivandaids.com Volume 9  Number 6  November 2014

Copyright Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Neuroimaging of HAND Ances and Hammoud

release of dopamine, resulting in microglial acti- of response to treatment and/or neuroprotective


vation, and possibly increases in viral replication measures.
[97]. Apart from the dopaminergic system, only
one study has targeted the serotonergic system in Acknowledgements
HIV patients. Using a serotonin transporter ligand, This work was supported by grants R01NR014449,
11
C-DASB, Hammoud et al. [98] showed dysregu- R01NR012657, R01NR012907, R21MH099979, and
lated serotonergic transmission in HIV patients the Alzheimers Association (B.M.A.).
with depression compared with nondepressed
HIV patients. This observation may reflect Conflicts of interest
increased density of serotonin, leading to increased Drs Ances and Hammoud have no conflicts of interest.
clearance of this neurotransmitter from the syn-
apse, which could subsequently lead to depressive
symptoms [98]. However, additional longitudinal REFERENCES AND RECOMMENDED
studies using these compounds and other recently READING
Papers of particular interest, published within the annual period of review, have
developed ligands are needed in HIV patients. been highlighted as:
A fourth approach to using PET in neuroHIV & of special interest
&& of outstanding interest
has investigated the role of amyloid. Previous patho-
logical studies have shown increases in diffuse 1. Justice AC. HIV and aging: time for a new paradigm. Curr HIV/AIDS Rep
2010; 7:6976.
plaques in HIV patients [99101]. With a greying 2. Holt JL, Kraft-Terry SD, Chang L. Neuroimaging studies of the aging HIV-1-
of the HIV population, increasing concerns have infected brain. J Neurovirol 2012; 18:291302.
3. Aberg JA, Kaplan JE, Libman H, et al. Primary care guidelines for the
arisen as to whether older HIV patients are at management of persons infected with human immunodeficiency virus:
increased risk for developing Alzheimers disease. 2009 update by the HIV medicine Association of the Infectious Diseases
Society of America. Clin Infect Dis 2009; 49:651681.
PET imaging using the amyloid-binding agent 4. Justice AC, Modur SP, Tate JP, et al. Predictive accuracy of the Veterans
11
C-labeled Pittsburgh Compound B (11C-PiB) has Aging Cohort Study index for mortality with HIV infection: a North American
cross cohort analysis. J Acquir Immune Defic Syndr 2013; 62:149163.
demonstrated amyloid deposition in preclinical 5. Luther VP, Wilkin AM. HIV infection in older adults. Clin Geriatr Med 2007;
Alzheimers disease [102]. However, 11C-PiB studies 23:567583; vii.
6. Clifford DB, Ances BM. HIV-associated neurocognitive disorder. Lancet
in HIV individuals failed to show increased amy- & Infect Dis 2013; 13:976986.

loid accumulation, even in symptomatic (HAND) Reviews HAND.


7. Ellis RJ, Moore DJ, Childers ME, et al. Progression to neuropsychological
patients [103,104]. A more recent study looking at impairment in human immunodeficiency virus infection predicted by elevated
HIV patients across a range of ages also did not cerebrospinal fluid levels of human immunodeficiency virus RNA. Arch Neurol
2002; 59:923928.
show significant increases in amyloid typically seen 8. Liner KJ 2nd, Ro MJ, Robertson KR. HIV, antiretroviral therapies, and the
with Alzheimers disease [105]. Observed differences brain. Curr HIV/AIDS Rep 2010; 7:8591.
9. Marra CM, Zhao Y, Clifford DB, et al. Impact of combination antiretroviral
between HAND and Alzheimers disease could therapy on cerebrospinal fluid HIV RNA and neurocognitive performance.
potentially reflect variances in amyloid metabolism AIDS 2009; 23:13591366.
10. Robertson K, Liner J, Meeker RB. Antiretroviral neurotoxicity. J Neurovirol
between the two disease entities or the lack of 2012; 18:388399.
affinity of current amyloid ligands for more diffuse 11. Hagberg L, Cinque P, Gisslen M, et al. Cerebrospinal fluid neopterin: an
informative biomarker of central nervous system immune activation in HIV-1
amyloid plaques. Additional studies using more infection. AIDS Res Ther 2010; 7:15.
recently developed amyloid agents (e.g., florbetapir) 12. Antinori A, Arendt G, Becker JT, et al. Updated research nosology for HIV-
associated neurocognitive disorders. Neurology 2007; 69:17891799.
are needed. 13. Cysique LA, Moffat K, Moore DM, et al. HIV, vascular and aging injuries in the
brain of clinically stable HIV-infected adults: a (1)H MRS study. PLoS One
2013; 8:e61738.
14. Lentz MR, Kim WK, Kim H, et al. Alterations in brain metabolism during the
CONCLUSION first year of HIV infection. J Neurovirol 2011; 17:220229.
15. Harezlak J, Buchthal S, Taylor M, et al. Persistence of HIV-associated
In conclusion, PET imaging has been used rather cognitive impairment, inflammation, and neuronal injury in era of highly active
limitedly in neuroHIV. However, PET methods antiretroviral treatment. AIDS 2011; 25:625633.
16. Descamps M, Hyare H, Stebbing J, Winston A. Magnetic resonance imaging
have great potential for further characterizing and spectroscopy of the brain in HIV disease. J HIV Ther 2008; 13:5558.
the neuropathophysiological changes associated 17. Valcour V, Chalermchai T, Sailasuta N, et al. Central nervous system viral
invasion and inflammation during acute HIV infection. J Infect Dis 2012;
with HIV, especially in the setting of optimal 206:275282.
cART. Evaluation of various neurotransmitter 18. Sailasuta N, Ross W, Ananworanich J, et al. Change in brain magnetic
resonance spectroscopy after treatment during acute HIV infection. PLoS
systems apart from the dopaminergic and seroto- One 2012; 7:e49272.
nergic systems might shed more light on selective 19. Iannucci G, Rovaris M, Giacomotti L, et al. Correlation of multiple sclerosis
measures derived from T2-weighted, T1-weighted, magnetization transfer,
vulnerability of various neuron subtypes to the and diffusion tensor MR imaging. AJNR Am J Neuroradiol 2001; 22:1462
virus. Evaluation of the effect of drug abuse on 1467.
20. Cardenas VA, Meyerhoff DJ, Studholme C, et al. Evidence for ongoing brain
the course of neuroHIV is needed. Most impor- injury in human immunodeficiency virus-positive patients treated with anti-
tantly, PET imaging might help finding reliable retroviral therapy. J Neurovirol 2009; 15:324333.
21. Mohamed MA, Barker PB, Skolasky RL, et al. Brain metabolism and cognitive
noninvasive biomarkers of neuronal injury in HIV impairment in HIV infection: a 3-T magnetic resonance spectroscopy study.
that could potentially be used in the evaluation Mag Reson Imaging 2010; 28:12511257.

1746-630X 2014 Wolters Kluwer Health | Lippincott Williams & Wilkins www.co-hivandaids.com 549

Copyright Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
HIV neurology

22. Paul RH, Ernst T, Brickman AM, et al. Relative sensitivity of magnetic 48. Cohen RA, Harezlak J, Gongvatana A, et al. Cerebral metabolite abnormal-
resonance spectroscopy and quantitative magnetic resonance imaging to ities in human immunodeficiency virus are associated with cortical and
cognitive function among nondemented individuals infected with HIV. J Int subcortical volumes. J Neurovirol 2010; 16:435444.
Neuropsychol Soc 2008; 14:725733. 49. Ragin AB, DSouza G, Reynolds S, et al. Platelet decline as a predictor of
23. Yiannoutsos CT, Nakas CT, Navia BA. Assessing multiple-group diagnostic brain injury in HIV infection. J Neurovirol 2011; 17:487495.
problems with multidimensional receiver operating characteristic surfaces: 50. Ragin AB, Du H, Ochs R, et al. Structural brain alterations can be detected
application to proton MR spectroscopy (MRS) in HIV-related neurological && early in HIV infection. Neurology 2012; 79:23282334.

injury. Neuroimage 2008; 40:248255. Observes changes in brain volumetrics soon after seroconversion.
24. Chang L, Ernst T, Leonido-Yee M, et al. Highly active antiretroviral therapy 51. Tate DF, Sampat M, Harezlak J, et al. Regional areas and widths of the
reverses brain metabolite abnormalities in mild HIV dementia. Neurology midsagittal corpus callosum among HIV-infected patients on stable antire-
1999; 53:782789. troviral therapies. J Neurovirol 2011; 17:368379.
25. Tarasow E, Wiercinska-Drapalo A, Jaroszewicz J, et al. Antiretroviral therapy 52. Christensen A, Russ S, Rambaran N, Wright SW. Patient perspectives on
and its influence on the stage of brain damage in patients with HIV - 1H MRS opt-out HIV screening in a Guyanese emergency department. Int Health
evaluation. Med Sci Monit 2004; 10 (Suppl 3):101106. 2012; 4:185191.
26. Robertson KR, Su Z, Margolis DM, et al. Neurocognitive effects of treatment 53. Turner MR, Modo M. Advances in the application of MRI to amyotrophic
interruption in stable HIV-positive patients in an observational cohort. lateral sclerosis. Expert Opin on Med Diagn 2010; 4:483496.
Neurology 2010; 74:12601266. 54. Chanraud S, Zahr N, Sullivan EV, Pfefferbaum A. MR diffusion tensor
27. Dwyer R, Wenhui L, Cysique L, et al. Symptoms of depression and rates of imaging: a window into white matter integrity of the working brain. Neuro-
neurocognitive impairment in HIV positive patients in Beijing, China. J Affect psychol Rev 2010; 20:209225.
Disord 2014; 162:8995. 55. Wycoco V, Shroff M, Sudhakar S, Lee W. White matter anatomy: what the
28. Ernst T, Jiang CS, Nakama H, et al. Lower brain glutamate is associated with radiologist needs to know. Neuroimaging Clin N Am 2013; 23:197216.
cognitive deficits in HIV patients: a new mechanism for HIV-associated 56. Hoare J, Westgarth-Taylor J, Fouche JP, et al. A diffusion tensor imaging and
neurocognitive disorder. J Magn Reson Imaging 2010; 32:10451053. neuropsychological study of prospective memory impairment in South
29. Gongvatana A, Harezlak J, Buchthal S, et al. Progressive cerebral injury in the African HIV positive individuals. Metab Brain Dis 2012; 27:289297.
setting of chronic HIV infection and antiretroviral therapy. J Neurovirol 2013; 57. Stubbe-Drger B, Deppe M, Mohammadi S, et al. Early microstructural white
19:209218. matter changes in patients with HIV: a diffusion tensor imaging study. BMC
30. Ances BM, Ortega M, Vaida F, et al. Independent effects of HIV, aging, and Neurol 2012; 12:23.
HAART on brain volumetric measures. J Acquir Immune Defic Syndr 2012; 58. Du H, Wu Y, Ochs R, et al. A comparative evaluation of quantitative
59:469477. neuroimaging measurements of brain status in HIV infection. Psychiatry
31. Fjell AM, Westlye LT, Amlien I, et al. High consistency of regional cortical Res 2012; 203:9599.
thinning in aging across multiple samples. Cereb Cortex 2009; 19:2001 59. Zhu T, Zhong J, Hu R, et al. Patterns of white matter injury in HIV infection
2012. & after partial immune reconstitution: a DTI tract-based spatial statistics study.
32. Heindel WC, Jernigan TL, Archibald SL, et al. The relationship of quantitative J Neurovirol 2013; 19:1023.
brain magnetic resonance imaging measures to neuropathologic indexes of Observes changes in white matter in HIV patients.
human immunodeficiency virus infection. Arch Neurol 1994; 51:1129 60. Gongvatana A, Cohen RA, Correia S, et al. Clinical contributors to cerebral
1135. white matter integrity in HIV-infected individuals. J Neurovirol 2011; 17:477
33. Aylward EH, Henderer JD, McArthur JC, et al. Reduced basal ganglia volume 486.
in HIV-1-associated dementia: results from quantitative neuroimaging. 61. Masters MC, Ances BM. Role of neuroimaging in HIV-associated neurocog-
Neurology 1993; 43:20992104. nitive disorders. Semin Neurol 2014; 34:89102.
34. Aylward EH, Brettschneider PD, McArthur JC, et al. Magnetic resonance 62. Wright PW, Heaps JM, Shimony JS, et al. The effects of HIV and combination
imaging measurement of gray matter volume reductions in HIV dementia. Am & antiretroviral therapy on white matter integrity. AIDS 2012; 26:15011508.
J Psychiatry 1995; 152:987994. Observes changes in white matter after initiation of cART.
35. Stout JC, Ellis RJ, Jernigan TL, et al. Progressive cerebral volume loss in 63. Rauch A, Rainer G, Logothetis NK. The effect of a serotonin-induced
human immunodeficiency virus infection: a longitudinal volumetric magnetic dissociation between spiking and perisynaptic activity on BOLD functional
resonance imaging study. HIV Neurobehavioral Research Center Group. MRI. Proc Natl Acad Sci U S A 2008; 105:67596764.
Arch Neurol 1998; 55:161168. 64. Zhang D, Raichle ME. Disease and the brains dark energy. Nat Rev Neurol
36. Heaps JM, Joska J, Hoare J, et al. Neuroimaging markers of human immu- 2010; 6:1528.
nodeficiency virus infection in South Africa. J Neurovirol 2012; 18:151156. 65. Chang L, Speck O, Miller EN, et al. Neural correlates of attention and working
37. Thompson PM, Dutton RA, Hayashi KM, et al. Thinning of the cerebral cortex memory deficits in HIV patients. Neurology 2001; 57:10011007.
visualized in HIV/AIDS reflects CD4 T lymphocyte decline. Proc Natl Acad 66. Ances B, Vaida F, Ellis R, Buxton R. Test-retest stability of calibrated
Sci U S A 2005; 102:1564715652. BOLD-fMRI in HIV- and HIV subjects. Neuroimage 2011; 54:21562162.
38. Jernigan TL, Archibald SL, Fennema-Notestine C, et al. Clinical factors 67. Ances BM, Roc AC, Korczykowski M, et al. Combination antiretroviral therapy
related to brain structure in HIV: the CHARTER study. J Neurovirol 2011; modulates the blood oxygen level-dependent amplitude in human immuno-
17:248257. deficiency virus-seropositive patients. J Neurovirol 2008; 14:418424.
39. Patel SH, Kolson DL, Glosser G, et al. Correlation between percentage of 68. Chang L, Tomasi D, Yakupov R, et al. Adaptation of the attention network
brain parenchymal volume and neurocognitive performance in HIV-infected in human immunodeficiency virus brain injury. Ann Neurol 2004; 56:259
patients. AJNR Am J Neuroradiol 2002; 23:543549. 272.
40. Cohen RA, Harezlak J, Schifitto G, et al. Effects of nadir CD4 count and 69. Ernst T, Chang L, Jovicich J, et al. Abnormal brain activation on functional MRI
duration of human immunodeficiency virus infection on brain volumes in the in cognitively asymptomatic HIV patients. Neurology 2002; 59:13431349.
highly active antiretroviral therapy era. J Neurovirol 2010; 16:2532. 70. Ernst T, Yakupov R, Nakama H, et al. Declined neural efficiency in cognitively
41. Pfefferbaum A, Rosenbloom MJ, Sassoon SA, et al. Regional brain structural stable human immunodeficiency virus patients. Ann Neurol 2009; 65:316
& dysmorphology in human immunodeficiency virus infection: effects of 325.
acquired immune deficiency syndrome, alcoholism, and age. Biol Psychiatry 71. Juengst SB, Aizenstein HJ, Figurski J, et al. Alterations in the hemodynamic
2012; 72:361370. response function in cognitively impaired HIV/AIDS subjects. J Neurosci
Investigates the effects of HIV on brain volumetrics. Methods 2007; 163:208212.
42. Castelo JM, Courtney MG, Melrose RJ, Stern CE. Putamen hypertrophy in 72. Maki PM, Cohen MH, Weber K, et al. Impairments in memory and hippo-
nondemented patients with human immunodeficiency virus infection and campal function in HIV-positive vs HIV-negative women: a preliminary study.
cognitive compromise. Arch Neurol 2007; 64:12751280. Neurology 2009; 72:16611668.
43. Thames AD, Foley JM, Wright MJ, et al. Basal ganglia structures differentially 73. Tracey I, Hamberg LM, Guimaraes AR, et al. Increased cerebral blood volume
contribute to verbal fluency: evidence from Human Immunodeficiency Virus in HIV-positive patients detected by functional MRI. Neurology 1998;
(HIV)-infected adults. Neuropsychologia 2012; 50:390395. 50:18211826.
44. Becker JT, Sanders J, Madsen SK, et al. Subcortical brain atrophy persists 74. Plessis SD, Vink M, Joska J, et al. HIV infection and the frontostriatal system:
even in HAART-regulated HIV disease. Brain Imaging Behav 2011; 5:77 && a systematic review and meta-analysis of fMRI studies. AIDS 2014; 28:803

85. 811.
45. Sullivan EV, Rosenbloom MJ, Rohlfing T, et al. Pontocerebellar contribution Reviews functional neuroimaging studies in HIV patients.
to postural instability and psychomotor slowing in HIV infection without 75. Thomas JB, Brier MR, Snyder AZ, et al. Pathways to neurodegeneration:
dementia. Brain Imaging Behav 2011; 5:1224. & effects of HIV and aging on resting-state functional connectivity. Neurology
46. Li C, Zhang X, Komery A, et al. Longitudinal diffusion tensor imaging and 2013; 80:11861193.
perfusion MRI investigation in a macaque model of neuro-AIDS: a preliminary Observes changes in functional MRI in HIV patients.
study. Neuroimage 2011; 58:286292. 76. Ances BM, Sisti D, Vaida F, et al. Resting cerebral blood flow: a potential
47. Kallianpur KJ, Shikuma C, Kirk GR, et al. Peripheral blood HIV DNA is biomarker of the effects of HIV in the brain. Neurology 2009; 73:702708.
& associated with atrophy of cerebellar and subcortical gray matter. Neurology 77. Chang L, Ernst T, Leonido-Yee M, Speck O. Perfusion MRI detects rCBF
2013; 80:17921799. abnormalities in early stages of HIV-cognitive motor complex. Neurology
Compares neuroimaging measures to blood biomarkers. 2000; 54:389396.

550 www.co-hivandaids.com Volume 9  Number 6  November 2014

Copyright Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Neuroimaging of HAND Ances and Hammoud

78. Singhal T. Positron emission tomography applications in clinical neurology. 92. Kreisl WC, Jenko KJ, Hines CS, et al. A genetic polymorphism for translo-
Semin Neurol 2012; 32:421431. && cator protein 18 kDa affects both in vitro and in vivo radioligand binding in
79. Pascal S, Resnick L, Barker WW, et al. Metabolic asymmetries in asympto- human brain to this putative biomarker of neuroinflammation. J Cereb Blood
matic HIV-1 seropositive subjects: relationship to disease onset and MRI Flow Metab 2013; 33:5358.
findings. J Nucl Med 1991; 32:17251729. Observes changes in HIV patients using new PET technique.
80. Hinkin CH, van Gorp WG, Mandelkern MA, et al. Cerebral metabolic change 93. Owen DR, Gunn RN, Rabiner EA, et al. Mixed-affinity binding in humans with
in patients with AIDS: report of a six-month follow-up using positron-emission 18-kDa translocator protein ligands. J Nucl Med 2011; 52:2432.
tomography. J Neuropsychiatry Clin Neurosci 1995; 7:180187. 94. Coughlin JM, Wang Y, Ma S, et al. Regional brain distribution of translocator
81. Rottenberg DA, Sidtis JJ, Strother SC, et al. Abnormal cerebral glucose & protein using [(11)C]DPA-713 PET in individuals infected with HIV.
metabolism in HIV-1 seropositive subjects with and without dementia. J Nucl J Neurovirol 2014; 20:219232.
Med 1996; 37:11331141. Observes changes in HIV patients using new PET technique.
82. von Giesen HJ, Antke C, Hefter H, et al. Potential time course of human 95. Wang GJ, Chang L, Volkow ND, et al. Decreased brain dopaminergic trans-
immunodeficiency virus type 1-associated minor motor deficits: electrophy- porters in HIV-associated dementia patients. Brain 2004; 127:2452
siologic and positron emission tomography findings. Arch Neurol 2000; 2458.
57:16011607. 96. Chang L, Wang GJ, Volkow ND, et al. Decreased brain dopamine transpor-
83. Andersen AB, Law I, Ostrowski SR, et al. Self-reported fatigue common ters are related to cognitive deficits in HIV patients with or without cocaine
among optimally treated HIV patients: no correlation with cerebral FDG-PET abuse. Neuroimage 2008; 42:869878.
scanning abnormalities. Neuroimmunomodulation 2006; 13:6975. 97. Purohit V, Rapaka R, Shurtleff D. Drugs of abuse, dopamine, and HIV-
84. Towgood KJ, Pitkanen M, Kulasegaram R, et al. Regional cerebral blood flow associated neurocognitive disorders/HIV-associated dementia. Mol Neuro-
&& and FDG uptake in asymptomatic HIV-1 men. Hum Brain Mapp 2013; biol 2011; 44:102110.
34:24842493. 98. Hammoud DA, Endres CJ, Hammond E, et al. Imaging serotonergic trans-
Analyzes changes in PET in HIV patients. mission with [11C]DASB-PET in depressed and nondepressed patients
85. Georgiou MF, Gonenc A, Waldrop-Valverde D, et al. Analysis of the effects of infected with HIV. Neuroimage 2010; 49:25882595.
injecting drug use and HIV-1 infection on 18F-FDG PET brain metabolism. 99. An SF, Giometto B, Groves M, et al. Axonal damage revealed by accumula-
J Nucl Med 2008; 49:19992005. tion of beta-APP in HIV-positive individuals without AIDS. J Neuropathol Exp
86. Chen MK, Guilarte TR. Translocator protein 18 kDa (TSPO): molecular Neurol 1997; 56:12621268.
sensor of brain injury and repair. Pharmacol Ther 2008; 118:117. 100. Esiri MM, Biddolph SC, Morris CS. Prevalence of Alzheimer plaques in AIDS.
87. Venneti S, Lopresti BJ, Wiley CA. The peripheral benzodiazepine receptor J Neurol Neurosurg Psychiatry 1998; 65:2933.
(Translocator protein 18kDa) in microglia: from pathology to imaging. Prog 101. Vehmas A, Lieu J, Pardo CA, et al. Amyloid precursor protein expression
Neurobiol 2006; 80:308322. in circulating monocytes and brain macrophages from patients with
88. Hammoud DA, Endres CJ, Chander AR, et al. Imaging glial cell activation with HIV-associated cognitive impairment. J Neuroimmunol 2004; 157:99
[11C]-R-PK11195 in patients with AIDS. J Neurovirol 2005; 11:346355. 110.
89. Wiley CA, Lopresti BJ, Becker JT, et al. Positron emission tomography 102. Cohen AD, Klunk WE. Early detection of Alzheimers disease using PiB and
imaging of peripheral benzodiazepine receptor binding in human immuno- FDG PET. Neurobiol Dis 2014; doi: 10.1016/j.nbd.2014.05.001. [Epub
deficiency virus-infected subjects with and without cognitive impairment. ahead of print]
J Neurovirol 2006; 12:262271. 103. Ances BM, Benzinger TL, Christensen JJ, et al. 11C-PiB imaging of human
90. Garvey LJ, Pavese N, Ramlackhansingh A, et al. Acute HCV/HIV coinfection immunodeficiency virus-associated neurocognitive disorder. Arch Neurol
is associated with cognitive dysfunction and cerebral metabolite disturbance, 2012; 69:7277.
but not increased microglial cell activation. PLoS One 2012; 7:e38980. 104. Ances BM, Christensen JJ, Teshome M, et al. Cognitively unimpaired HIV-
91. Garvey LJ, Pavese N, Politis M, et al. Increased microglia activation in positive subjects do not have increased 11C-PiB: a case-control study.
&& neurologically asymptomatic HIV-infected patients receiving effective ART. Neurology 2010; 75:111115.
AIDS 2014; 28:6772. 105. Ortega M, Ances BM. Role of HIV in amyloid metabolism. J Neuroimmune
Observes changes in HIV patients using new PET technique. Pharmacol 2014; 9:483491.

1746-630X 2014 Wolters Kluwer Health | Lippincott Williams & Wilkins www.co-hivandaids.com 551

Copyright Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

Anda mungkin juga menyukai