You are on page 1of 10

REVIEW

CURRENT
OPINION Anesthesia and brain tumor surgery: technical
considerations based on current research evidence
Junichi Saito a,b, Joe Masters b, Kazuyoshi Hirota a, and Daqing Ma b

Purpose of review
Anesthetics may influence cancer recurrence and metastasis following surgery by modulating the
neuroendocrine stress response or by directly affecting cancer cell biology. This review summarizes the
current evidence on whether commonly used anesthetics potentially affect postoperative outcomes following
Downloaded from https://journals.lww.com/co-anesthesiology by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AWnYQp/IlQrHD3hIW04IhZ9AvNyXiDskTJpHrU+NEtsjmqiU3Ukzps0lU= on 09/08/2019

solid organ cancer surgery with particular focus on neurological malignancies.


Recent findings
Despite significant improvement in diagnostic and therapeutic technology over the past decades, mortality
rates after cancer surgery (including brain tumor resection) remains high. With regards to brain tumors,
interaction between microglia/macrophages and tumor cells by multiple biological factors play an
important role in tumor progression and metastasis. Preclinical studies have demonstrated an association
between anesthetics and brain tumor cell biology, and a potential effect on tumor progression and
metastasis has been revealed. However, in the clinical setting, the current evidence is inadequate to draw
firm conclusions on the optimal anesthetic technique for brain tumor surgery.
Summary
Further work at both the basic science and clinical level is urgently needed to evaluate the association
between perioperative factors, including anesthetics/technique, and postoperative brain tumor outcomes.
Keywords
anesthetics, brain tumor, metastasis, recurrence

INTRODUCTION and metastasis after surgery remain common and


leads to poor prognosis.
Epidemiology of brain tumor The potential impact of anesthetics in modulat-
&

Brain tumors are a relatively rare but serious health ing cancer cell biology [4–6,7 ] may either promote
burden in terms of morbidity and mortality. Glioma or inhibit cancer recurrence and metastasis after
is the most common primary brain tumor in adults, surgery. In addition, surgical stress and its associated
and represents about 30% of all brain tumors and changes can massively affect cancer cell behaviors,
80% of malignant brain tumors [1]. Despite signifi- which has been well documented [8,9]. In this
cant improvements in diagnosis and therapy over review, we summarize the current preclinical data
the past decades, the 5-year survival rate of the most on the potential effects of anesthetics on cancer cell
severe form of glioblastoma is less than 5% [2,3]. biology and the clinical evidence that anesthetic
Surgery remains the first-line treatment for
patients with a malignant brain tumor. Complete
surgical resection of brain tumor is generally recom-
a
mended, but care must be taken not to impair the Department of Anesthesiology, Hirosaki University Graduate School of
neurological function with tumors located in Medicine, Hirosaki, Japan and bAnaesthetics, Pain Medicine and Inten-
sive Care, Department of Surgery and Cancer, Faculty of Medicine,
eloquent regions. Hence, adjuvant or neo-adjuvant
Imperial College London, London, UK
therapy is often required to improve survival. How-
Correspondence to Daqing Ma, Anaesthetics, Pain Medicine and Inten-
ever, resistance to radiation and chemotherapy is sive Care, Department of Surgery and Cancer, Faculty of Medicine,
the major problem in the treatment of glioblastoma Imperial College London, Chelsea & Westminster Hospital 369 Fulham
that may be because of resistance to regulated cell Road, London SW10 9NH, UK. Tel: +44 203315 8495;
death, attenuation of cytotoxicity by the microen- fax: +44 203315 5109; e-mail: d.ma@imperial.ac.uk
vironmental changes, and the limited ability to Curr Opin Anesthesiol 2019, 32:553–562
deliver drugs into the tumor. Therefore, recurrence DOI:10.1097/ACO.0000000000000749

0952-7907 Copyright ß 2019 Wolters Kluwer Health, Inc. All rights reserved. www.co-anesthesiology.com

Copyright © 2019 Wolters Kluwer Health, Inc. All rights reserved.


Neuroanesthesia

microglia stimulate the production of pro-matrix


KEY POINTS metalloproteinase (MMP) 2 via TLR2 signaling
&

 Despite significant improvement in diagnostic and [10 ]. Secreted MMP2 activates the membrane type-
therapeutic technology over the past decades, mortality 1 MMP (MT1-MMP) and facilitate glioma cell motility
after surgery for brain tumors remains high. [15]. TGF-b recruits microglia/macrophage to glioma
[16] and stimulates glioma migration. It has been
 Anesthetics may have impact on brain tumor metastasis
suggested that blocking TGF-b abolishes the effects
or recurrence through the effects on tumor cell
signaling, the immune response, and modulation of the of microglia on glioma invasiveness [17] (Fig. 1).
neuro-endocrine stress response. For metastasis, vascular network development is
important to supply enough nutrients and oxygen
 Clinical evidence is inadequate to draw conclusion for to tumor cells. Microglia/macrophages are located at
how to choose the anesthetics during brain
vascular branching points and release vascular
tumor surgery.
endothelial growth factor (VEGF) to stimulate the
 Further clinical study is urgently needed to evaluate the growth of functional vasculature. Microglia-derived
association between the anesthetic management and MMPs have been shown to induce production of
long-term outcomes of brain tumor following surgery. angiogenic factors and stimulate angiogenesis in
&
glioma [10 ]. Hypoxia-inducible factors (HIFs) are
transcription factors that regulate cell proliferation
and survival and have been shown in active path-
technique may influence outcomes following the ways in glioblastomas leading to enhanced invasion
surgical management of brain tumors. [18]. Previous studies revealed that HIFs are modu-
lated by several anesthetics, especially inhalational
agents and propofol [4,19]. In renal and prostate
Brain cancer cell biology and mechanism of cancer cells, isoflurane significantly increases cell
tumor progression and metastasis proliferation and migration caused by the up-regu-
Current research reveals that the interaction between lation of HIFs via the phosphatidylinositide 3-
microglia/macrophages and tumor cells by multiple kinase/Akt/mammalian target of rapamycin path-
factors play an important role in tumor progression way [4,19]. In contrast, propofol inhibits HIF-1a
&
and metastasis in neurological malignancies [10 ]. activation induced by hypoxia or cobalt chloride
Microglia/macrophages are often recruited within in prostate cancer cells. Propofol also prevented
or close to the brain tumor mass. Reactivated micro- isoflurane-induced HIF-1a activation, and reduces
glia are known to lead to local neuronal death cancer cell malignant activities, including cell pro-
through the secretion of pro-inflammatory cytokines liferation, migration, invasion and resistance to the
and also chronic reactive microglia increase produc- chemotherapeutic agent [19].
tion of cytokines and chemokines that escalate tumor Activated microglia cells significantly promote
growth and invasion. Inhibition of microglial activa- colonization of breast tumor tissues in metastasis
tion has been shown to significantly reduce glioma [20]. Wnt pathway is one of the well established
proliferation [11]. Microglia/macrophages are differ- signaling pathways during tumorigenesis and plays
entiated into two types: classical (M1) and alternative an important role in the development of the central
(M2) phenotype. M1 cells have an anti-tumor nervous system [21]. The role of Wnt in vascular
immune function and suppress glioma cell growth development has been found using targeted deletion
[12]. In contrast, M2 cells have a pro-tumor immune mutants of Wnt signaling components in animal
response via producing immunosuppressive media- models. Additionally, in clinical study, Wnt signaling
tors [13]. Both M1 and M2 microglia are found within is elevated in breast cancer patients with brain metas-
brain tumors, but the anti-tumor functions of M1 &&
tasis [22 ]. Wnt signaling pathway is also associated
microglia in the brain tumor are often impaired [14]. with the proliferation of glioma cells. Inhibiting
The balance between the pro-tumor response of M2 microglia function via a Wnt inhibitor significantly
and the anti-tumor response of M1 affects tumor diminished tumor cell invasion whereas treating
growth, invasion and progression. microglia with Wnt increases the production of
IL-6, tumor necrosis factor (TNF)-a and MMPs [23].
Interaction between microglia/ S100 calcium-binding protein B (S100B), a
macrophages and tumor receptor for advanced glycation end products
Soluble factors released from glioma cells are recog- (RAGE) ligand, is secreted by glioma cells and has
nized by Toll-like receptors (TLRs), especially TLR2, been suggested to contribute tumorigenesis by pro-
on microglia. Interleukin (IL)-6 and transforming moting IL-6 expression and signal transducer and
growth factor (TGF)-b secreted by glioma and activator of transcription 3 (STAT3) activation [24].

554 www.co-anesthesiology.com Volume 32  Number 5  October 2019

Copyright © 2019 Wolters Kluwer Health, Inc. All rights reserved.


Anesthesia and brain tumor surgery Saito et al.

S100B-RAGE-STAT3 signaling stimulates polariza- phosphoinositide 3-kinase/Akt/mammalian target


tion of M2 microglia. M2 microglia polarization is of paramycin pathway [25].
associated with maintaining local immunosuppres-
sion. Tumor-secreted S100B induces STAT3 activa-
tion through the activation of receptor for advanced ANESTHETICS AND IMMUNE
glycation end products. This signaling pathway acti- MODIFICATION, CANCER CELL BIOLOGY
vation results in the suppression of M1 microglia AND POTENTIAL EFFECTS ON TUMOR
function [24]. The unbalance between pro-tumor PROGRESSION AND METASTASIS
response of M2 and anti-tumor response of M1 via Current research suggests that perioperative anes-
S100B-RAGE-STAT3 signaling promotes tumor thetics/technique impacts on metastasis and recur-
growth and invasion. rence through effects on cancer cell signaling, the
immune response, and modulation of the neuroen-
Mitochondria function and brain tumor docrine stress response. Anesthetics and analgesics
growth and development can independently suppress antitumor immune
There are two main signaling pathways that trigger function, even though surgical stress and perioper-
apoptosis for brain tumor cells; the intrinsic mito- ative pain can activate neuroendocrine cascades
chondrial pathway and the extrinsic pathway [25]. that inhibit natural killer cell activity during the
Both pathways cause activating cleavage of caspases perioperative period [9]. This inhibition of natural
3 and 7, which irreversibly instigates an apoptotic killer cell activity leads to immunosuppression and
cascade (Fig. 2). The intrinsic mitochondrial path- promotes proliferation and metastasis of cancer.
way can be triggered by direct damage to the Additionally, anesthetics may have impact on the
cell from reactive oxygen species (ROS) and others. cancer cell biology and modulate gene expression
Protein 53 (p53) accumulates in response to DNA [27]. Changes in serum level of VEGF-C [28], MMPs
damage and oncogene activation. p53 enhances [29] and HIFs [30] by anesthetics may also have an
DNA act as a nuclear transcription factor and enhan- impact on metastasis or recurrence of cancers. Their
ces transcription of pro-apoptotic genes, which effects on molecular targets and/or cell signaling
interact with the B cell lymphoma-2 (BCL-2) family pathway can be seen in Figs 1 and 2.
members, BCL-2-associated X protein (BAX), BCL-2
homologous antagonist/killer and BCL-2-associated
death promoter [25]. The recruitment of pro-apo-
Intravenous agents
ptotic BCL-2 family members facilitates the opening
of a mitochondrial permeability transition pore Propofol
(MPTP). The opening of MPTP is a critical event Propofol has tumor-suppressive effects in various
for apoptosis. Opening the pore allows the rapid types of cancer cells [31,32]. Its effects are because
release of cytochrome C, second mitochondria- of suppression of proliferation and invasion, and
derived activator of caspases (SMAC) and Omi induction of apoptosis in primary brain tumor cells
from the mitochondria. Cytochrome C binds with [33–36].
caspases 9 to form an apoptosome, which cleaves One such study evaluated the role of micro-
caspases 3 and 7 [25]. RNA218 in the effects of propofol on the biological
The association of the anti-apoptotic BCL-2 fam- behavior of glioma cells. Treatment with propofol
ily members with the mitochondrial membranes reduced MMP2 protein expression levels, and over-
contributes to mitochondrial dysfunction in glioma expression of microRNA218 also decreased MMP2
[25]. The anti-apoptotic BCL-2 proteins is highly protein expression levels. The effects of propofol in
over-expressed in human glioma cell and protects U373 cells was reversed by the antimicroRNA218
the glioma cell from undergoing apoptosis after antibody [33]. Propofol suppresses proliferation and
chemically induced DNA damage [26]. invasion, and induces the apoptosis of glioma cells, at
The extrinsic pathway is initiated by the death least partially through upregulation of microRNA218
receptor [25]. Specific ligands bind and, in turn, expression [33]. The other study also revealed that
activate the death receptor, which results in intra- propofol-mediated anti-tumor effects were because of
cellular signal transduction. This signaling causes the regulation of NF-kB pathway, which is associated
activating cleavage of caspases 8 and 10, which with microRNA218 expression [34].
cleave caspases 3 and 7. In glioma cells, the extrinsic Wnt signaling is one of the important pathways
pathway is suppressed, and this is related to tumor in gliomagenesis and b-catenin is a key component
resistance to regulated cell death. Additionally, this of this signaling. Propofol decreases the protein
tumor resistance is related to overactivation of levels of b-catenin in glioma cells and has the

0952-7907 Copyright ß 2019 Wolters Kluwer Health, Inc. All rights reserved. www.co-anesthesiology.com 555

Copyright © 2019 Wolters Kluwer Health, Inc. All rights reserved.


Neuroanesthesia

Anesthetics & Analgesics


Thiopental, Ketamine, Sevoflurane?
?
NMDAR
Brain tumour

Intra-cranial
inflammation Tumour growth
Pro-
MMP2
MMP2 & invasion

S100B

Soluble factors
Midazolam microRNA218
Immunosuppression TGF-β
RAGE
TLR
IL-6

STAT3
Wnt Propofol

VEGF MMPs

Angiogenesis Peripheral cancer cells


(breast cancer)
HIFs

Volatile anesthetics
Tumour metastasis

Propofol

FIGURE 1. Interaction between microglia/macrophage and brain tumor, and possible anesthetic actions. HIF, hypoxia-
inducible factor; IL-6, interleukin6; MMP, matrix metalloproteinase; NMDAR, N-methyl-D-aspartate receptor; RAGE, receptor for
advanced glycation end products; S100B, S100 calcium-binding protein B; TGF-b, transforming growth factor beta; TLR, toll-
like receptors; VEGF, vascular endothelial growth factor.

potential to inhibit Wnt signaling in glioma cells Thiopental


[37]. It has been shown that propofol dose-depen- Thiopental has strong anti-inflammatory effects.
dently inhibits the viability of the glioma cells and Excessive inflammatory response increases the
significantly increases apoptotic glioma cells [37]. secretion of pro-inflammatory cytokines, such as
Propofol showed cytotoxicity in glioblastoma IL-1, IL-6, IL-8 and TNF-a and simultaneously leads
8401 cells but not in DI TNC1 rat normal astrocytes. to increased counter-inflammatory cytokines, such
The cytotoxicity was induced through arrest of cell as IL-4 and IL-10 secretion. NF-kB is an important
cycle progression at the G2/M phase and increased transcription factor that is essential for expression of
intracellular ROS levels only in glioblastoma 8401 pro-inflammatory cytokines. Thiopental suppressed
cells but not normal astrocytes [36]. Another study TNF-a production via inhibition of NF-kB activation
showed that propofol increased intracellular free by lipopolysaccharide in human glioma cells [41].
calcium, leading to cell death that might be through Activation of N-methyl-D-aspartate (NMDA)
ROS-mediated apoptosis [35]. receptor of glutamine increases MMP2 activity in
Another preferable effect of propofol related to glioma cells [42]. Thiopental reduces MMP2 activity
the brain surgery has been reported. Aquaporin 4 and decreases migration of glioma cells [43].
and aquaporin 9 are known as important factors
influencing brain edema after traumatic brain injury Ketamine
[38]. Propofol reduced postischemic cerebral edema Ketamine is an NMDA receptor antagonist. Gluta-
in rats, through the inhibiting aquaporin 4 over- mate is an excitatory neurotransmitter in the brain
expression in the boundary zone of ischemia [39]. that activates glutamate receptors including the
Additionally, propofol inhibited aquaporin 4 NMDA receptor. Glioma cells secrete glutamate
expression compared with sevoflurane in glioma during proliferation, and treatment with the
resection patients, even though the study did not NMDA receptor antagonists MK801 or memantine
evaluate magnitude of edema after the surgery [40]. inhibited the growth of glutamate-secreting tumors

556 www.co-anesthesiology.com Volume 32  Number 5  October 2019

Copyright © 2019 Wolters Kluwer Health, Inc. All rights reserved.


Anesthesia and brain tumor surgery Saito et al.

<Extrinsic Pathway> <Proliferation Pathway> <Intrinsic mitochondrial Pathway>

TNF-α, Fas EDG, CXCL12 Hypoxia, Toxicity,


Ca2+
PDGF Oxidative stress

DR RTX CXCR4
Propofol Propofol
Dexmedetomidine Ca2+ ROS
PI3K
? Midazolam
mTOR Lidocaine
?
Akt δ-opioid
DNA
Midazolam TPRV1 Damage
Caspase 8 p53

Caspase 10 MCL-1
BCL-2 BAX
BCL-xL
BAK

MTPT Cyt C
Caspase 9

Apoptosis!
Midazolam Caspase3

Caspase7
Tumour proliferation & growth

FIGURE 2. Brain tumor growth and apoptotic pathways, and possible anesthetic actions. BAX, BCL-2-associated X protein,
BCL-2, B-cell lymphoma 2, BCL-xL, B-cell lymphoma-extra large, CXCL12, C-X-C motif chemokine 12, CXCR4, C-X-C
chemokine receptor type 4, Cyt C, cytochrome c, DNA, deoxyribonucleic acid, DR, death receptor, EDG, endothelial
differentiation gene, MCL-1, myeloid cell leukemia sequence 1, mTOR, mammalian target of rapamycin, MTPT, mitochondrial
permeability transition pore, PDGF, platelet-derived growth factor, PI3K, phosphatidylinositol-3 kinase, ROS, reactive oxygen
species, RTX, resiniferatoxin, TNF-a, tumor necrosis factor-alfa, TPRV1, transient receptor potential vanilloid 1.

in situ, suggesting that glutamate promoted glioma be also beneficial for non-cancer neurosurgery.
cells proliferation via the activation of NMDA recep- However, dexmedetomidine might have a pro-
tor [44]. Ketamine-induced apoptosis in C6 glioma malignant effect on brain tumors. It has been dem-
cells, resulted in the suppression of C6 cell prolifer- onstrated that dexmedetomidine promoted neuro-
ation [45]. Treatment with the selective NMDA glioma cell proliferation and migration, which was
receptor antagonist D-AP5 significantly suppressed associated with the upregulation of anti-apoptotic
the proliferation of C6 glioma cells and induced proteins (BCL2 and BCL-xL) via the a2-adrenoceptor
apoptosis [45]. On the other hand, ketamine has activation and promoted cancer growth in a xeno-
&
adrenergic activity and is profoundly immunosup- graft cancer model [7 ].
pressive by inhibition of natural killer cell activity.
Accordingly, use of ketamine can lead to increased Midazolam
incidence of metastases showing in non-brain can- Midazolam suppresses neuroglioma cell prolifera-
cer models [46]. The ratio of risk and benefit of tion and migration, induces apoptosis through
ketamine use depends on the physiological status the intrinsic mitochondrial pathway, and increases
of individuals per se. &
intracellular ROS [7 ]. These anti-cancer effects are
mediated by the peripheral benzodiazepine recep-
Dexmedetomidine tor. Midazolam also inhibits IL-1b-stimulated IL-6
The intracranial anti-inflammatory response and release from C6 glioma cells. IL-6 is suppressed by an
neuroprotective effects of dexmedetomidine are inhibitor of phospho-specific inhibitory kappa B
well documented in traumatic brain injury and kinase, an inhibitor of phospho-specific stress-
cerebral ischemia [47–50]. These properties may activated protein kinase/c-Jun N-terminal kinase,

0952-7907 Copyright ß 2019 Wolters Kluwer Health, Inc. All rights reserved. www.co-anesthesiology.com 557

Copyright © 2019 Wolters Kluwer Health, Inc. All rights reserved.


Neuroanesthesia

and an inhibitor of Janus family of tyrosine kinase 1, time-dependent manner [58]. The inhibition of the d-
2 and 3. Midazolam markedly suppresses IL-1b-stim- opioid receptors results in brain glioma cell apoptosis
ulated STAT3 phosphorylation without affecting and is associated with the mitochondrial pathways
the phosphorylation of p38 mitogen-activated pro- [58]. One clinical retrospective study in patients who
tein kinase, phospho-specific stress-activated pro- underwent brain tumor resection showed that
tein kinase/c-Jun N-terminal kinase or phospho- patients who received remifentanil during surgery
specific inhibitory kappa B. Midazolam inhibits had significantly lower in-hospital mortality
IL-1b-induced IL-6 release in rat C6 glioma cells (1.5 vs. 3.0%; P ¼ 0.029) compared with patients
via suppression of STAT3 activation [51]. who received fentanyl during surgery. The odds ratio
for use of remifentanil for in-hospital mortality was
0.47 (95% confidence interval, 0.25–0.91; P ¼ 0.025)
Volatile agents [59]. Further studies are needed to verify these bene-
Exposure of human glioblastoma stem cells to 1.2% fits because of its retrospective nature.
isoflurane for 6 h results in increased proliferation
and decreased apoptosis when compared with the Local anesthetics
control group. In addition, isoflurane exposure Local anesthetic agents may reduce the incidence of
causes increased migration capacity in vitro and cancer recurrence through systemic anti-inflamma-
the distance migrated is increased in vivo [52]. This tory effect in addition to direct effects on the pro-
may be because isoflurane increases the expression liferation and migration of cancer cells [60,61].
of HIFs [4], which are transcription factors that Local anesthetics and regional anesthetic techni-
regulate cell proliferation and survival and have ques are associated with a decrease of stress response
been found as an active pathway in glioblastomas [62]. This allows for a reduction in opioid usage as
leading to enhanced invasion. Similarly, exposure of well as decreased release of endogenous opioids [63].
human primary glioma stem cells to 2% sevoflurane Local anesthetics can reduce activation of intraop-
for 6 h induced a larger number of proliferated cells. erative and postoperative neuroendocrine stress
The levels of CD133, VEGF, HIF-1a, HIF-2a, and response, which inhibits natural killer cell function
phospho-Akt are up-regulated by sevoflurane in a in several cancer surgeries.
time and/or concentration related manner [53]. Transient receptor potential (TRP) channels are a
group of ion channels that can be activated by several
factors [64]. TRP channels regulate cell differentia-
Opioids tion, proliferation and death [65]. Suppression of
Intraoperative and postoperative pain has been TRP melastatin 7 (TRPM7) inhibits proliferation,
shown to activate the stress response and induce migration and invasion of glioma cells [66].
immunosuppression postoperatively shown in both Lidocaine has the anti-proliferation effects on glioma
animal and human studies [54,55]. Perioperative pain cells and its effect is mediated by inhibiting
control is important, but drugs used for perioperative TRPM7 [67]. Lidocaine also increases TRP
pain control also have an impact on the immune subfamily V, member 1 (TRPV1) gene expression
system and on cancer progression and metastasis. and induces the increase of intracellular calcium
Indeed, opioids themselves have been shown to ion concentration, mitochondrial potential and
reduce natural killer cell function and stimulate can- apoptosis in a dose-dependent manner in U87-MG
cer cell proliferation through effects on angiogenesis glioma cells. Lidocaine increased TPRV1 gene
and tumor cell signaling pathways [9,56]. increase mitochondrial membrane potential, leading
Differences in affinity for the opioid receptors to U87-MG glioma cell apoptosis [68]. Additionally,
cause different effects on glioblastoma cells. recent study reveals that lidocaine induced protective
Morphine stimulates human glioblastoma T98G cell autophagy in rat C6 glioma cells [69].
proliferation in vitro in a dose-dependent manner
but biphalin, an enkephalin analog, shows inhibi-
tory effect on T98G cell proliferation. The inhibitory CLINICAL SIGNIFICANCE
effect of biphalin is associated with suppression of Retrospective studies have shown that general anes-
Ki-67 expression [57]. thesia combined with regional anesthesia or regional
The downregulation of the d-opioid receptor anesthesia alone has been associated with a reduction
promotes changes in BAX and BCL-2 protein expres- of recurrence rate and improve survival in breast
&
sion in glioma cells. The BAX activates the caspase [70 ], prostate [71–73], ovarian [74], melanoma
family to cause apoptosis of glioma cells. The [75] and colorectal cancer [76–78] when compared
specific inhibitor of d-opioid receptors inhibits with general anesthesia alone. Additionally, a retro-
glioma cell proliferation in a dose-dependent and spective study reveals that propofol-based total

558 www.co-anesthesiology.com Volume 32  Number 5  October 2019

Copyright © 2019 Wolters Kluwer Health, Inc. All rights reserved.


Anesthesia and brain tumor surgery Saito et al.

Table 1. Anesthetics and prognosis of brain tumor patients

Author Journal Year Model Protocol Main finding

Dong et al. [82 ] J Neurosurg 2019 A single-center Propofol (n ¼ 154) Median PFS and median OS were
&

Anesthesiol retrospective study vs. Sevoflurane not significantly different between


(n ¼ 140) propofol anesthesia and
sevoflurane anesthesia
Cata et al. [83] J Clin Neurosci 2017 A single-center Isoflurane (n ¼ 117) Isoflurane or desflurane were not
retrospective study vs. Desflurane independent prognostic factors
(n ¼ 261) for PFS (HR, 95% CI: 1.07,
0.85–1.37, P ¼ 0.531) and OS
(HR, 95% CI: 1.13, 0.86–1.48,
P ¼ 0.531).
Uchida et al. [59] J Anesth 2012 A retrospective study Remifentanil Patients who received remifentanil
using the Japanese (n ¼ 936) vs. during surgery had significantly
Diagnosis Procedure Fentanyl lower in-hospital mortality (1.5 vs.
Combination (nonremifentanil) 3.0%; P ¼ 0.029). The odds ratio
Database (n ¼ 936) for use of remifentanil for in-
hospital mortality was 0.47 (95%
CI, 0.25–0.91; P ¼ 0.025).
Y. Peng et al. Unpublished - A single-center, Propofol vs. -
(NCT02756312) observation randomized, parallel Sevoflurane
group controlled
clinical trial

CI, confidence interval; HR, hazard ratio; OS, overall survival; PFS, progression-free survival.

intravenous anesthesia for colon cancer surgery was to isolate respective contributions of each drug.
associated with better survival compared with iso- Interaction between the different drugs and techni-
&
flurane [79] and desflurane anesthesia [80 ]. How- ques is also inevitable and the impact of this is
ever, a randomized control trial did not show the largely unknown. Similarly, randomized control tri-
significant difference of the duration of recurrence- als (RCTs) are lacking, although one prospective
free survival between general anesthesia alone and randomized controlled trial (NCT02756312) is
general anesthesia combined with epidural anesthe- underway to evaluate the association between anes-
sia in patients after major abdominal oncologic thetic management and postoperative malignant
surgery [81]. Further prospective studies are needed glioma progression. However, it will be a few years
to define the impact of anesthetics and anesthetic before meaningful conclusions can be drawn
technique on the prognosis of cancer patients. from this, and so other forms of investigation must
With regards to brain tumors, the impact of continue in the meantime including basic
anesthesia on patient prognosis has not been well science studies.
evaluated (Table 1). A recent retrospective clinical
study revealed that sevoflurane (n ¼ 140) did not
worsen progression-free survival and overall survival FURTHER CONSIDERATION
in the high-grade glioma patients who underwent The surgical procedure itself affects cancer recur-
tumor resection when compared with propofol rence and metastasis after excision. Among several
(n ¼ 154) [82 ]. However, subgroup analysis revealed
&
theories to explain this phenomenon, the adverse
that sevoflurane shortened the overall survival in impact of surgical stress on immunity [54,55] and
patients with Karnofsky performance status less unintended seeding of tumor cells during surgery
than 80 when compared with propofol. Another [84] may be most considerable. Raised adrenocorti-
retrospective study shows that the type of volatile cotropic hormone because of surgical stress induces
anesthetic used, isoflurane vs. desflurane, was not excess cortisol release and leads to insulin resistance
associated with a difference in prognosis [83]. The and raising blood glucose concentration. All these
extent to which anesthetic agents and anesthetic can have a negative impact on postoperative patient
technique may affect short-term and long-term out- recovery. For example, postoperative hyperglycemia
comes in patients with brain tumor remains unclear. increases surgical site infection [85] and surgical
Balanced anesthesia and multimodal analgesia are stress has also been shown to have an immunosup-
used clinically and many drugs are therefore, used pressive effect by reducing natural killer cell func-
during the perioperative period. Thus, it is difficult tion and T-cell responses [86].

0952-7907 Copyright ß 2019 Wolters Kluwer Health, Inc. All rights reserved. www.co-anesthesiology.com 559

Copyright © 2019 Wolters Kluwer Health, Inc. All rights reserved.


Neuroanesthesia

Maximal surgical resection in patients with a Acknowledgements


malignant brain tumor has been shown to have a J.S. drafted the manuscript. D.M. conceived the study
positive effect on the duration of progression-free idea. All authors participated to write the manuscript.
survival [87,88]. As neuroanesthesia improved dra- D.M. is the archival author.
matically, awake craniotomy with brain mapping
has become popular all over the world [89], espe- Financial support and sponsorship
cially for intra-axial tumors in eloquent areas. The work was supported by a BOC Chair grant, RCoA,
Compared with conventional craniotomy under London, UK. J.S. as a research fellow was supported by
general anesthesia, awake craniotomy has shown Hirosaki University Graduate School of Medicine, Hir-
several advantages [90]. A systematic review reveals osaki, Japan.
that patients who underwent awake craniotomy
have shorter hospital stay and fewer postoperative Conflicts of interest
neurological deficits compared with those who
There are no conflicts of interest.
underwent conventional craniotomy under general
anesthesia [91]. A more recent systematic review and
meta-analysis also shows that the advantages of REFERENCES AND RECOMMENDED
awake craniotomy include a lower rate of postoper- READING
ative nausea and vomiting and shorter hospital stay Papers of particular interest, published within the annual period of review, have
been highlighted as:
[92]. Additional potential benefits include, higher & of special interest
rate of complete tumor resection (100%) [93], better && of outstanding interest

neurological outcomes [94], less postoperative pain


1. Goodenberger ML, Jenkins RB. Genetics of adult glioma. Cancer Genet
[95], less opioid usage [96] and less intra-operative 2012; 205:613–621.
vasoactive drug usage [96], which have been dem- 2. McNeill KA. Epidemiology of brain tumors. Neurol Clin 2016; 34:981–998.
3. Bondy ML, Scheurer ME, Malmer B, et al., Brain Tumor Epidemiology
onstrated in the small retrospective studies. Another Consortium. Brain tumor epidemiology: consensus from the Brain Tumor
meta-analysis revealed that intra-operative brain Epidemiology Consortium. Cancer 2008; 113:1953–1968.
4. Benzonana LL, Perry NJ, Watts HR, et al. Isoflurane, a commonly used volatile
sensorimotor mapping was associated with a lower anesthetic, enhances renal cancer growth and malignant potential via the
rate of late, severe neurological deficits and greater hypoxia-inducible factor cellular signaling pathway in vitro. Anesthesiology
2013; 119:593–605.
extent resection of tumor [97]. However, the long- 5. Iwasaki M, Zhao H, Jaffer T, et al. Volatile anaesthetics enhance the metastasis
term outcomes of conventional vs. awake surgical related cellular signalling including CXCR2 of ovarian cancer cells. Onco-
target 2016; 7:26042–26056.
resection for brain tumor are unknown and sub- 6. Freeman J, Crowley PD, Foley AG, et al. Effect of perioperative lidocaine and
jected to further studies. cisplatin on metastasis in a murine model of breast cancer surgery. Anticancer
Res 2018; 38:5599–5606.
7. Wang C, Datoo T, Zhao H, et al. Midazolam and dexmedetomidine affect
& neuroglioma and lung carcinoma cell biology in vitro and in vivo. Anesthesiol-
CONCLUSION ogy 2018; 129:1000–1014.
This article describes that dexmedetomidine enhances cancer cell proliferation
Current evidence does not conclusively support the and migration, on the contrary, midazolam suppresses cancer cell proliferation and
migration. The data suggests that commonly used anesthetics has impact on
superiority of a specific anesthetic technique for tumour cell growth.
&
brain tumor surgery [82 ,83]. However, the use of 8. Buggy DJ, Hemmings HC. Special issue on anaesthesia and cancer. Br J
Anaesth 2014; 113(Suppl 1):i1–i3.
local anesthesia seems to be particularly appealing 9. Tavare AN, Perry NJ, Benzonana LL, et al. Cancer recurrence after surgery: direct
for patients with brain tumors and has a number of and indirect effects of anesthetic agents. Int J Cancer 2012; 130:1237–1250.
10. Wu SY, Watabe K. The roles of microglia/macrophages in tumor progression
theoretical benefits. Local anesthetics can reduce & of brain cancer and metastatic disease. Front Biosci (Landmark Ed) 2017;
surgical stress and the amount of general anesthetic 22:1805–1829.
This review article focuses on the roles of microglia/macrophages in brain tumour
use, leading to decreased immunosuppression and progression and metastasis. This article helps us to understand the biology of brain
hence reduced risk of tumor progression and metas- tumour.
11. Graeber MB, Scheithauer BW, Kreutzberg GW. Microglia in brain tumors.
tasis. Compared with volatile anesthetics, a propo- Glia 2002; 40:252–259.
fol-based technique may also have some advantages 12. Sarkar S, Doring A, Zemp FJ, et al. Therapeutic activation of macrophages and
microglia to suppress brain tumor-initiating cells. Nat Neurosci 2014;
in brain tumor patients; a prospective RCT 17:46–55.
(NCT02756312) is ongoing. Although current evi- 13. Wei J, Gabrusiewicz K, Heimberger A. The controversial role of microglia in
malignant gliomas. Clin Dev Immunol 2013; 2013:285246.
dence does not yet support the need for a change in 14. Schartner JM, Hagar AR, Van Handel M, et al. Impaired capacity for upregula-
clinical practice, anesthesiologists should consider tion of MHC class II in tumor-associated microglia. Glia 2005; 51:279–285.
15. Mayes DA, Hu Y, Teng Y, et al. PAX6 suppresses the invasiveness of
anesthetic technique as a potentially modifiable risk glioblastoma cells and the expression of the matrix metalloproteinase-2 gene.
factor for brain cancer recurrence. Further clinical Cancer Res 2006; 66:9809–9817.
16. Ye XZ, Xu SL, Xin YH, et al. Tumor-associated microglia/macrophages
trials to evaluate the association between periopera- enhance the invasion of glioma stem-like cells via TGF-beta1 signaling
tive factors (including anesthetic technique) and pathway. J Immunol 2012; 189:444–453.
17. Merzak A, McCrea S, Koocheckpour S, Pilkington GJ. Control of human
long-term outcomes following brain tumour surgery glioma cell growth, migration and invasion in vitro by transforming growth
are urgently needed. factor beta 1. Br J Cancer 1994; 70:199–203.

560 www.co-anesthesiology.com Volume 32  Number 5  October 2019

Copyright © 2019 Wolters Kluwer Health, Inc. All rights reserved.


Anesthesia and brain tumor surgery Saito et al.

18. Wang G, Wang JJ, Fu XL, et al. Advances in the targeting of HIF-1alpha and 46. Melamed R, Bar-Yosef S, Shakhar G, et al. Suppression of natural killer cell
future therapeutic strategies for glioblastoma multiforme (review). Oncol Rep activity and promotion of tumor metastasis by ketamine, thiopental, and
2017; 37:657–670. halothane, but not by propofol: mediating mechanisms and prophylactic
19. Huang H, Benzonana LL, Zhao H, et al. Prostate cancer cell malignancy via measures. Anesth Analg 2003; 97:1331–1339.
modulation of HIF-1alpha pathway with isoflurane and propofol alone and in 47. Ma D, Hossain M, Rajakumaraswamy N, et al. Dexmedetomidine produces its
combination. Br J Cancer 2014; 111:1338–1349. neuroprotective effect via the alpha 2A-adrenoceptor subtype. Eur J Pharma-
20. Pukrop T, Dehghani F, Chuang HN, et al. Microglia promote colonization of col 2004; 502:87–97.
brain tissue by breast cancer cells in a Wnt-dependent way. Glia 2010; 48. Ma D, Rajakumaraswamy N, Maze M. alpha2-Adrenoceptor agonists: shed-
58:1477–1489. ding light on neuroprotection? Br Med Bull 2004; 71:77–92.
21. Gao L, Chen B, Li J, et al. Wnt/beta-catenin signaling pathway inhibits the 49. Rajakumaraswamy N, Ma D, Hossain M, et al. Neuroprotective interaction
proliferation and apoptosis of U87 glioma cells via different mechanisms. produced by xenon and dexmedetomidine on in vitro and in vivo neuronal
PLoS One 2017; 12:e0181346. injury models. Neurosci Lett 2006; 409:128–133.
22. Smid M, Wang Y, Zhang Y, et al. Subtypes of breast cancer show preferential 50. Gu J, Sun P, Zhao H, et al. Dexmedetomidine provides renoprotection against
&& site of relapse. Cancer Res 2008; 68:3108–3114. ischemia-reperfusion injury in mice. Crit Care 2011; 15:R153.
This article highlights the importance of WNT/b-catenin signaling to contribute to 51. Tanabe K, Kozawa O, Iida H. Midazolam suppresses interleukin-1beta-induced
breast tumors metastasizing to brain. This observation is important to understand interleukin-6 release from rat glial cells. J Neuroinflammation 2011; 8:68.
the mechanism of breast cancer and metastasis. 52. Zhu M, Li M, Zhou Y, et al. Isoflurane enhances the malignant potential of
23. Pukrop T, Klemm F, Hagemann T, et al. Wnt 5a signaling is critical for glioblastoma stem cells by promoting their viability, mobility in vitro and
macrophage-induced invasion of breast cancer cell lines. Proc Natl Acad migratory capacity in vivo. Br J Anaesth 2016; 116:870–877.
Sci U S A 2006; 103:5454–5459. 53. Shi QY, Zhang SJ, Liu L, et al. Sevoflurane promotes the expansion of glioma
24. Zhang L, Liu W, Alizadeh D, et al. S100B attenuates microglia activation in stem cells through activation of hypoxia-inducible factors in vitro. Br J Anaesth
gliomas: possible role of STAT3 pathway. Glia 2011; 59:486–498. 2015; 114:825–830.
25. Ordys BB, Launay S, Deighton RF, et al. The role of mitochondria in glioma 54. Tsuchiya Y, Sawada S, Yoshioka I, et al. Increased surgical stress promotes
pathophysiology. Mol Neurobiol 2010; 42:64–75. tumor metastasis. Surgery 2003; 133:547–555.
26. Lopez J, Tait SW. Mitochondrial apoptosis: killing cancer using the enemy 55. Boomsma MF, Garssen B, Slot E, et al. Breast cancer surgery-induced
within. Br J Cancer 2015; 112:957–962. immunomodulation. J Surg Oncol 2010; 102:640–648.
27. Huitink JM, Heimerikxs M, Nieuwland M, et al. Volatile anesthetics modulate 56. Kim R. Effects of surgery and anesthetic choice on immunosuppression and
gene expression in breast and brain tumor cells. Anesth Analg 2010; cancer recurrence. J Transl Med 2018; 16:8.
111:1411–1415. 57. Lazarczyk M, Matyja E, Lipkowski AW. A comparative study of morphine
28. Looney M, Doran P, Buggy DJ. Effect of anesthetic technique on serum stimulation and biphalin inhibition of human glioblastoma T98G cell prolifera-
vascular endothelial growth factor C and transforming growth factor beta in tion in vitro. Peptides 2010; 31:1606–1612.
women undergoing anesthesia and surgery for breast cancer. Anesthesiology 58. Zhou L, Guo X, Chen M, et al. Inhibition of delta-opioid receptors induces
2010; 113:1118–1125. brain glioma cell apoptosis through the mitochondrial and protein kinase C
29. Deegan CA, Murray D, Doran P, et al. Anesthetic technique and the cytokine pathways. Oncol Lett 2013; 6:1351–1357.
and matrix metalloproteinase response to primary breast cancer surgery. Reg 59. Uchida K, Yasunaga H, Miyata H, et al. Impact of remifentanil use on early
Anesth Pain Med 2010; 35:490–495. postoperative outcomes following brain tumor resection or rectal cancer
30. Ma D, Lim T, Xu J, et al. Xenon preconditioning protects against renal surgery. J Anesth 2012; 26:711–720.
ischemic-reperfusion injury via HIF-1alpha activation. J Am Soc Nephrol 60. Xuan W, Zhao H, Hankin J, et al. Local anesthetic bupivacaine induced ovarian
2009; 20:713–720. and prostate cancer apoptotic cell death and underlying mechanisms in vitro.
31. Altenburg JD, Harvey KA, McCray S, et al. A novel 2,6-diisopropylphenyl- Sci Rep 2016; 6:26277.
docosahexaenoamide conjugate induces apoptosis in T cell acute lympho- 61. Xuan W, Hankin J, Zhao H, et al. The potential benefits of the use of regional
blastic leukemia cell lines. Biochem Biophys Res Commun 2011; anesthesia in cancer patients. Int J Cancer 2015; 137:2774–2784.
411:427–432. 62. O’Riain SC, Buggy DJ, Kerin MJ, et al. Inhibition of the stress response to
32. Mammoto T, Mukai M, Mammoto A, et al. Intravenous anesthetic, propofol breast cancer surgery by regional anesthesia and analgesia does not affect
inhibits invasion of cancer cells. Cancer Lett 2002; 184:165–170. vascular endothelial growth factor and prostaglandin E2. Anesth Analg 2005;
33. Xu J, Xu W, Zhu J. Propofol suppresses proliferation and invasion of glioma 100:244–249.
cells by upregulating microRNA-218 expression. Mol Med Rep 2015; 63. Chae BK, Lee HW, Sun K, et al. The effect of combined epidural and light
12:4815–4820. general anesthesia on stress hormones in open heart surgery patients. Surg
34. Zheng Y, Liu H, Liang Y. Genistein exerts potent antitumour effects alongside Today 1998; 28:727–731.
anaesthetic, propofol, by suppressing cell proliferation and nuclear factor- 64. Clapham DE. TRP channels as cellular sensors. Nature 2003; 426:517–524.
kappaB-mediated signalling and through upregulating microRNA-218 expres- 65. Dadon D, Minke B. Cellular functions of transient receptor potential channels.
sion in an intracranial rat brain tumour model. J Pharm Pharmacol 2017; Int J Biochem Cell Biol 2010; 42:1430–1445.
69:1565–1577. 66. Leng TD, Li MH, Shen JF, et al. Suppression of TRPM7 inhibits proliferation,
35. Liang WZ, Jan CR, Lu CH. Investigation of 2,6-diisopropylphenol (propofol)- migration, and invasion of malignant human glioma cells. CNS Neurosci Ther
evoked Ca2þ movement and cell death in human glioblastoma cells. Toxicol 2015; 21:252–261.
In Vitro 2012; 26:862–871. 67. Leng T, Lin S, Xiong Z, Lin J. Lidocaine suppresses glioma cell proliferation by
36. Hsu SS, Jan CR, Liang WZ. Evaluation of cytotoxicity of propofol and its inhibiting TRPM7 channels. Int J Physiol Pathophysiol Pharmacol 2017;
related mechanism in glioblastoma cells and astrocytes. Environ Toxicol 9:8–15.
2017; 32:2440–2454. 68. Lu J, Ju YT, Li C, et al. Effect of TRPV1 combined with lidocaine on cell state
37. Xu W, Zheng J, Bie S, et al. Propofol inhibits Wnt signaling and exerts and apoptosis of U87-MG glioma cell lines. Asian Pac J Trop Med 2016;
anticancer activity in glioma cells. Oncol Lett 2018; 16:402–408. 9:288–292.
38. Cartagena CM, Phillips KL, Tortella FC, et al. Temporal alterations in aqua- 69. Izdebska M, Halas-Wisniewska M, Zielinska W, et al. Lidocaine induces
porin and transcription factor HIF1alpha expression following penetrating protective autophagy in rat C6 glioma cell line. Int J Oncol 2018;
ballistic-like brain injury (PBBI). Mol Cell Neurosci 2014; 60:81–87. 54:1099–1111.
39. Zheng YY, Lan YP, Tang HF, et al. Propofol pretreatment attenuates aqua- 70. Exadaktylos AK, Buggy DJ, Moriarty DC, et al. Can anesthetic technique for
porin-4 over-expression and alleviates cerebral edema after transient focal & primary breast cancer surgery affect recurrence or metastasis? Anesthesiol-
brain ischemia reperfusion in rats. Anesth Analg 2008; 107:2009–2016. ogy 2006; 105:660–664.
40. Yang WC, Zhou LJ, Zhang R, et al. Effects of propofol and sevoflurane on This article is the first clinical study that shows that anesthetic technique has
aquaporin-4 and aquaporin-9 expression in patients performed gliomas potential to affect the prognosis of cancer patients.
resection. Brain Res 2015; 1622:1–6. 71. Biki B, Mascha E, Moriarty DC, et al. Anesthetic technique for radical
41. Ichiyama T, Nishikawa M, Lipton JM, et al. Thiopental inhibits NF-kappaB prostatectomy surgery affects cancer recurrence: a retrospective analysis.
activation in human glioma cells and experimental brain inflammation. Brain Anesthesiology 2008; 109:180–187.
Res 2001; 911:56–61. 72. Tsui BC, Rashiq S, Schopflocher D, et al. Epidural anesthesia and cancer
42. Ramaswamy P, Aditi Devi N, Hurmath Fathima K, et al. Activation of NMDA recurrence rates after radical prostatectomy. Can J Anaesth 2010; 57:
receptor of glutamate influences MMP-2 activity and proliferation of glioma 107–112.
cells. Neurol Sci 2014; 35:823–829. 73. Scavonetto F, Yeoh TY, Umbreit EC, et al. Association between neuraxial
43. Hurmath FK, Mittal M, Ramaswamy P, et al. Sevoflurane and thiopental analgesia, cancer progression, and mortality after radical prostatectomy: a
preconditioning attenuates the migration and activity of MMP-2 in U87MG large, retrospective matched cohort study. Br J Anaesth 2014; 113(Suppl
glioma cells. Neurochem Int 2016; 94:32–38. 1):i95–i102.
44. Takano T, Lin JH, Arcuino G, et al. Glutamate release promotes growth of 74. de Oliveira GS Jr, Ahmad S, Schink JC, et al. Intraoperative neuraxial
malignant gliomas. Nat Med 2001; 7:1010–1015. anesthesia but not postoperative neuraxial analgesia is associated with
45. Niwa H, Furukawa KI, Seya K, et al. Ketamine suppresses the proliferation of increased relapse-free survival in ovarian cancer patients after primary cytor-
rat C6 glioma cells. Oncol Lett 2017; 14:4911–4917. eductive surgery. Reg Anesth Pain Med 2011; 36:271–277.

0952-7907 Copyright ß 2019 Wolters Kluwer Health, Inc. All rights reserved. www.co-anesthesiology.com 561

Copyright © 2019 Wolters Kluwer Health, Inc. All rights reserved.


Neuroanesthesia

75. Schlagenhauff B, Ellwanger U, Breuninger H, et al. Prognostic impact of the 85. Ata A, Lee J, Bestle SL, et al. Postoperative hyperglycemia and surgical site
type of anaesthesia used during the excision of primary cutaneous melanoma. infection in general surgery patients. Arch Surg 2010; 145:858–864.
Melanoma Res 2000; 10:165–169. 86. Leaver HA, Craig SR, Yap PL, Walker WS. Lymphocyte responses following
76. Gupta A, Bjornsson A, Fredriksson M, et al. Reduction in mortality after open and minimally invasive thoracic surgery. Eur J Clin Invest 2000;
epidural anaesthesia and analgesia in patients undergoing rectal but not 30:230–238.
colonic cancer surgery: a retrospective analysis of data from 655 patients in 87. Smith JS, Chang EF, Lamborn KR, et al. Role of extent of resection in the long-
central Sweden. Br J Anaesth 2011; 107:164–170. term outcome of low-grade hemispheric gliomas. J Clin Oncol 2008;
77. Gottschalk A, Ford JG, Regelin CC, et al. Association between epidural 26:1338–1345.
analgesia and cancer recurrence after colorectal cancer surgery. Anesthe- 88. Hervey-Jumper SL, Li J, Lau D, et al. Awake craniotomy to maximize glioma
siology 2010; 113:27–34. resection: methods and technical nuances over a 27-year period. J Neurosurg
78. Zimmitti G, Soliz J, Aloia TA, et al. Positive impact of epidural analgesia on 2015; 123:325–339.
oncologic outcomes in patients undergoing resection of colorectal liver 89. Dziedzic T, Bernstein M. Awake craniotomy for brain tumor: indications,
metastases. Ann Surg Oncol 2016; 23:1003–1011. technique and benefits. Expert Rev Neurother 2014; 14:1405–1415.
79. Wigmore TJ, Mohammed K, Jhanji S. Long-term survival for patients under- 90. Meng L, Berger MS, Gelb AW. The potential benefits of awake craniotomy for
going volatile versus IV anesthesia for cancer surgery: a retrospective brain tumor resection: an anesthesiologist’s perspective. J Neurosurg An-
analysis. Anesthesiology 2016; 124:69–79. esthesiol 2015; 27:310–317.
80. Wu ZF, Lee MS, Wong CS, et al. Propofol-based total intravenous anesthesia 91. Brown T, Shah AH, Bregy A, et al. Awake craniotomy for brain tumor
& is associated with better survival than desflurane anesthesia in colon cancer resection: the rule rather than the exception? J Neurosurg Anesthesiol
surgery. Anesthesiology 2018; 129:932–941. 2013; 25:240–247.
This article evaluates the association between anesthetics and patient survival 92. Lu VM, Phan K, Rovin RA. Comparison of operative outcomes of eloquent
after elective colon cancer surgery. The results suggest that patients anesthetized glioma resection performed under awake versus general anesthesia: a sys-
with propofol had better overall survival compared with those with desflurane. tematic review and meta-analysis. Clin Neurol Neurosurg 2018;
81. Myles PS, Peyton P, Silbert B, et al., ANZCA Trials Group Investigators. 169:121–127.
Perioperative epidural analgesia for major abdominal surgery for cancer and 93. Eseonu CI, Rincon-Torroella J, ReFaey K, et al. Awake craniotomy vs cra-
recurrence-free survival: randomised trial. BMJ 2011; 342:d1491. niotomy under general anesthesia for perirolandic gliomas: evaluating peri-
82. Dong J, Zeng M, Ji N, et al. Impact of anesthesia on long-term outcomes in operative complications and extent of resection. Neurosurgery 2017;
& patients with supratentorial high-grade glioma undergoing tumor resection: a 81:481–489.
retrospective cohort study. J Neurosurg Anesthesiol 2019. [Epub ahead of print] 94. Sacko O, Lauwers-Cances V, Brauge D, et al. Awake craniotomy vs surgery
This retrospective study evaluates the impact of anesthetic use during surgery on under general anesthesia for resection of supratentorial lesions. Neurosurgery
the patients who underwent brain tumour resection. Subgroup analysis suggests 2011; 68:1192–1198.
that propofol might be beneficial in patients with poor preoperative Karnofsky 95. Wrede KH, Stieglitz LH, Fiferna A, et al. Patient acceptance of awake
performance status. craniotomy. Clin Neurol Neurosurg 2011; 113:880–884.
83. Cata JP, Hagan KB, Bhavsar SD, et al. The use of isoflurane and desflurane as 96. Rajan S, Cata JP, Nada E, et al. Asleep-awake-asleep craniotomy: a compar-
inhalational agents for glioblastoma surgery. A survival analysis. J Clin Neu- ison with general anesthesia for resection of supratentorial tumors. J Clin
rosci 2017; 35:82–87. Neurosci 2013; 20:1068–1073.
84. Camara O, Kavallaris A, Noschel H, et al. Seeding of epithelial cells into 97. De Witt Hamer PC, Robles SG, Zwinderman AH, et al. Impact of intraopera-
circulation during surgery for breast cancer: the fate of malignant and benign tive stimulation brain mapping on glioma surgery outcome: a meta-analysis. J
mobilized cells. World J Surg Oncol 2006; 4:67. Clin Oncol 2012; 30:2559–2565.

562 www.co-anesthesiology.com Volume 32  Number 5  October 2019

Copyright © 2019 Wolters Kluwer Health, Inc. All rights reserved.

You might also like