Brain Tumors A Pocket Guide Mohile 1 Ed 2023
Brain Tumors A Pocket Guide Mohile 1 Ed 2023
Brain Tumors A Pocket Guide Mohile 1 Ed 2023
Tumors
A Pocket Guide
Nimish A. Mohile
Alissa A. Thomas
Editors
123
Brain Tumors
Nimish A. Mohile
Alissa A. Thomas
Editors
Brain Tumors
A Pocket Guide
Editors
Nimish A. Mohile Alissa A. Thomas
Department of Neurology Department of Neurological
University of Rochester Medical Sciences
Center University of Vermont Larner
Rochester, NY, USA College of Medicine
Burlington, VT, USA
© The Editor(s) (if applicable) and The Author(s), under exclusive license to
Springer Nature Switzerland AG 2023
This work is subject to copyright. All rights are solely and exclusively licensed by
the Publisher, whether the whole or part of the material is concerned, specifically
the rights of translation, reprinting, reuse of illustrations, recitation, broadcasting,
reproduction on microfilms or in any other physical way, and transmission or
information storage and retrieval, electronic adaptation, computer software, or by
similar or dissimilar methodology now known or hereafter developed.
The use of general descriptive names, registered names, trademarks, service
marks, etc. in this publication does not imply, even in the absence of a specific
statement, that such names are exempt from the relevant protective laws and
regulations and therefore free for general use.
The publisher, the authors, and the editors are safe to assume that the advice and
information in this book are believed to be true and accurate at the date of
publication. Neither the publisher nor the authors or the editors give a warranty,
expressed or implied, with respect to the material contained herein or for any
errors or omissions that may have been made. The publisher remains neutral with
regard to jurisdictional claims in published maps and institutional affiliations.
1 Malignant Glioma���������������������������������������������������������� 3
Shannon Fortin Ensign and Alyx B. Porter
2 Diffuse Astrocytoma������������������������������������������������������ 21
Donna Molaie and Phioanh (Leia) Nghiemphu
3 Oligodendroglioma�������������������������������������������������������� 39
Oluwatosin Akintola
4 BRAF-Mutated Glioma ������������������������������������������������ 51
Karisa C. Schreck and Jean M. Mulcahy Levy
5 Treatment of Meningioma�������������������������������������������� 67
Rimas V. Lukas, Timothy J. Kruser,
and Adam M. Sonabend
6
Treatment of Primary CNS Lymphoma���������������������� 83
Ugur Sener and Lauren Schaff
7 Adult Medulloblastoma ������������������������������������������������103
Tresa McGranahan and Sonia Partap
8 Treatment of Ependymoma������������������������������������������119
Jing Wu and Surabhi Ranjan
v
vi Contents
9 Brain
Edema and Corticosteroid Toxicity ������������������141
Maninder Kaur and Reena Thomas
10 T
umor Related Epilepsy������������������������������������������������153
Thomas Wychowski
11 Treatment
and Prevention of Venous
Thromboembolism ��������������������������������������������������������165
Shiao-Pei Weathers and Alexander Ou
12 Management
of Neurocognitive Symptoms����������������177
Christina Weyer-Jamora and Jennie W. Taylor
13 C
hemotherapy-Related Toxicities
and Management������������������������������������������������������������195
Haroon Ahmad and David Schiff
14 Radiation
Related Toxicities and Management����������211
Sara J. Hardy and Michael T. Milano
15 Neurosurgical
Complications in Brain Tumor
Patients����������������������������������������������������������������������������235
Tyler Schmidt
16 Management
of Older Patients with Brain
Tumors����������������������������������������������������������������������������249
Andrea Wasilewski
17 P
alliative Care in Neuro-oncology��������������������������������267
Young-Bin Song and Lynne P. Taylor
Index����������������������������������������������������������������������������������������283
Part I
Brain Tumor Primer
Malignant Glioma
1
Shannon Fortin Ensign
and Alyx B. Porter
Clinical Scenario
S. F. Ensign
Department of Hematology and Oncology, Mayo Clinic,
Phoenix, AZ, USA
A. B. Porter (*)
Department of Neurology, Mayo Clinic, Phoenix, AZ, USA
e-mail: Porter.alyx@mayo.edu
Gliomas arise from glial cells and neuronal precursors. They con-
stitute 80% of all malignant primary brain and CNS tumors.
Glioblastoma (GBM) is the most invasive, aggressive (grade 4)
and common form. Patients can present with various symptoms
including seizures, headaches, neurological deficits, and altered
mental status. Magnetic Resonance Imaging (MRI) is the diag-
nostic modality of choice when a brain lesion is suspected.
Computed Tomography (CT) scans are appropriate in emergent
situations to evaluate for intracranial hemorrhage or hydrocepha-
lus. While certain imaging characteristics are highly suggestive of
GBM (heterogeneously enhancing expansile lesion), none are
pathognomonic. In fact, many non-neoplastic processes can
mimic gliomas, including multiple sclerosis, granulomatous dis-
1 Malignant Glioma 5
Post-operative Treatment
Radiation
Chemotherapy
Lomustine Lomustine: 110 mg/m2 Ondansetron 8 mg po Monitor PFTs at Monitor for pulmonary
monotherapy q42 days 30 min prior to baseline and periodic toxicity
chemotherapy and then
up to every 8 h as needed
PCV (Procarbazine, Procarbazine: 100 mg/ Ondansetron 16 mg IV, Monitor PFTs as Procarbazine & Vincristine:
Lomustine, m2 PO d1–10 Dexamethasone 12 mg above, & monitor for Hepatic dose adjustment as
Malignant Glioma
Vincristine) Lomustine: 100 mg/m2 IV, Emend 125 mg PO constipation/ileus & indicated
PO d1 peripheral neuropathy
Vincristine: 1.5 mg/m2
IV d1, q42 days
11
12 S. F. Ensign and A. B. Porter
Symptom Management
Surveillance
Treatment at Recurrence
Prognosis/Survivorship
References
1. Brown TJ, Brennan MC, Li M, et al. Association of the extent of resection
with survival in glioblastoma: a systematic review and meta-analysis.
JAMA Oncol. 2016;2(11):1460–9.
2. Golub D, Hyde J, Dogra S, et al. Intraoperative MRI versus 5-ALA in
high-grade glioma resection: a network meta-analysis. J Neurosurg.
2020;1–15:484–98.
3. Stummer W, Pichlmeier U, Meinel T, Wiestler OD, Zanella F, Reulen HJ,
ALA-Glioma Study Group. Fluorescence-guided surgery with
5-aminolevulinic acid for resection of malignant glioma: a randomised
controlled multicentre phase III trial. Lancet Oncol. 2006;7(5):392–401.
https://doi.org/10.1016/S1470-2045(06)70665-9. PMID: 16648043.
4. Louis DN, Perry A, Wesseling P, Brat DJ, Cree IA, Figarella-Branger D,
Hawkins C, Ng HK, Pfister SM, Reifenberger G, Soffietti R, von
Deimling A, Ellison DW. The 2021 WHO classification of tumors of the
central nervous system: a summary. Neuro Oncol. 2021;23(8):1231–51.
https://doi.org/10.1093/neuonc/noab106. PMID: 34185076; PMCID:
PMC8328013.
18 S. F. Ensign and A. B. Porter
5. Corso CD, Bindra RS, Mehta MP. The role of radiation in treating glio-
blastoma: here to stay. J Neurooncol. 2017;134(3):479–85.
6. Laperriere N, Weller M, Stupp R, et al. Optimal management of elderly
patients with glioblastoma. Cancer Treat Rev. 2013;39(4):350–7.
7. Stupp R, Mason WP, van den Bent MJ, et al. Radiotherapy plus con-
comitant and adjuvant temozolomide for glioblastoma. N Engl J Med.
2005;352(10):987–96.
8. Perry JR, Laperriere N, O’Callaghan CJ, et al. Short-course radiation plus
Temozolomide in elderly patients with glioblastoma. N Engl J Med.
2017;376(11):1027–37.
9. van den Bent MJ, Tesileanu CMS, Wick W. Adjuvant and concurrent
temozolomide for 1p/19q non-co-deleted anaplastic glioma (CATNON;
EORTC study 26053-22054): second interim analysis of a randomised,
open-label, phase 3 study. Lancet Oncol. 2021;22(6):813–23. https://doi.
org/10.1016/S1470-2045(21)00090-5. Epub 2021 May 14. PMID:
34000245; PMCID: PMC8191233.
10. Margison GP, Santibanez Koref MF, Povey AC. Mechanisms of carcino-
genicity/chemotherapy by O6-methylguanine. Mutagenesis.
2002;17(6):483–7.
11. Herrlinger U, Tzaridis T, Mack F, et al. Lomustine-temozolomide combi-
nation therapy versus standard temozolomide therapy in patients with
newly diagnosed glioblastoma with methylated MGMT promoter
(CeTeG/NOA-09): a randomised, open-label, phase 3 trial. Lancet.
2019;393(10172):678–88.
12. Kirson ED, Gurvich Z, Schneiderman R, et al. Disruption of cancer cell
replication by alternating electric fields. Cancer Res. 2004;64(9):3288–
95.
13. Stupp R, Taillibert S, Kanner A, et al. Effect of tumor-treating fields plus
maintenance Temozolomide vs maintenance Temozolomide alone on sur-
vival in patients with glioblastoma: a randomized clinical trial. JAMA.
2017;318(23):2306–16.
14. Stupp R, Wong ET, Kanner AA, et al. NovoTTF-100A versus physician’s
choice chemotherapy in recurrent glioblastoma: a randomised phase III
trial of a novel treatment modality. Eur J Cancer. 2012;48(14):2192–202.
15. Taphoorn MJB, Dirven L, Kanner AA, et al. Influence of treatment with
tumor-treating fields on health-related quality of life of patients with
newly diagnosed glioblastoma: a secondary analysis of a randomized
clinical trial. JAMA Oncol. 2018;4(4):495–504.
16. Lote K, Stenwig AE, Skullerud K, Hirschberg H. Prevalence and prog-
nostic significance of epilepsy in patients with gliomas. Eur J Cancer.
1998;34(1):98–102.
17. Happold C, Gorlia T, Chinot O, et al. Does valproic acid or Levetiracetam
improve survival in glioblastoma? A pooled analysis of prospective
clinical trials in newly diagnosed glioblastoma. J Clin Oncol.
2016;34(7):731–9.
1 Malignant Glioma 19
Clinical Vignette
D. Molaie
Department of Neurosurgery, Kaiser Permanente,
Redwood City, CA, USA
P. (L.) Nghiemphu (*)
Department of Neurology, David Geffen School of Medicine at UCLA,
Los Angeles, CA, USA
e-mail: pnghiemphu@mednet.ucla.edu
Clinical Features
Surgical Decision-Making
Pathology
Patients can be stratified by age and EOR into risk groups, which
are helpful for guiding postoperative management. From the pre-
molecular era, age <40 with a GTR is considered low risk, while
age ≥40 and any patient with incomplete resection are considered
high risk. Additional factors to consider when selecting patients
for immediate postoperative chemotherapy are the presence of
risk factors for poor outcome: preoperative neurologic functional
deficit, preoperative tumor size ≥5 cm, and tumor crossing the
corpus callosum [6, 7].
Traditionally, watchful waiting or observation has been accept-
able for low risk patients, while immediate postoperative chemora-
diotherapy is recommended for high risk. Since the recognition of
IDH mutational status’s positive impact on survival, watchful wait-
ing may also be considered in IDH-mutant DA patients <40 with
incomplete resection but small residual tumor in non-eloquent loca-
tions, or ≥40 with GTR who also have other favorable markers.
26 D. Molaie and P. (. Nghiemphu
Post-operative Treatment
MZ
T
1. Regimens:
(a) Concurrent: 75 mg/m2 oral daily concurrent with RT
(b) Adjuvant: 150 mg/m2–200 mg/m2* oral on days 1–5 (D1–
5) of a 28 day cycle for 12 cycles. If no or mild toxicity at
150, escalate to 200 for next cycle. (Table 2.1 describes
toxicities, monitoring and dose modifications for com-
monly used regimens)
2. Symptom management:
(a) Nausea: Ondansetron (or other 5-HT3 serotonin receptor
antagonist) on D1–42 (dose-dense) and D1–5 (adjuvant),
1 h before TMZ
(b) Constipation: Polyethylene glycol
(c) Drug rash: Combination of anti-H1 and anti-H2 histamine
receptor antagonists on D1–5; if severe, add
Methylprednisolone dose pack
(d) Consider Pneumocystis Jiroveci Prophylaxis in patients
with lymphopenia
PCV
This is typically initiated after RT. Cycle length is 42 days, with a
goal of 6 cycles. Note: Average cycle length was 3–4 in RTOG
9802 [13] due to dose-limiting toxicities and still effective in
terms of survival.
1. Regimen:
D1: CCNU 90–110 mg/m2 oral
D8 &D29: Vincristine 1.4 mg/m2 IV (round to nearest
0.1 mg, max 2 mg)
D8–21: Procarbazine 60 mg/m2/day oral (Available in
50 mg tabs which are given as combination of tabs over
14 days to average 60 mg/m2/day or as a compounded
medication)
2. Symptom management:
2
dense absorption NPO 1 h Constipation, Myelosuppression diff, CMP 25% dose reduction
TMZ- before and after (nadir D14–21), Allergic-like rash • D21 CBC w/ • ANC = 1.5–1.0,
adjuvant • Renally excreted (Hives), Liver toxicity, Renal toxicity, diff Plt = 100–75 & count
PCP Pneumoniac, Teratogenic, • D28 CBC w/ recovery <2 weeks stay
Carcinogenic, Gonadotoxic, diff, CMP at 150 mg/m2 daily
Infertility • ANC = 1.5–1.0,
Plt = 100–75 & count
recovery >2 weeks 25%
dose reduction
• ANC = <1.0, Plt = <75
25% dose reduction
(continued)
29
Table 2.1 (continued)
30
dose reduction
• Progressive &/or severe
neuropathic symptoms
discontinue
CBC w/diff Complete blood count with differential, CMP Comprehensive metabolic panel, ANC Absolute neutrophil count, Plt Plate-
let, MAO Monoamine oxidase, TCA Tricyclic anti-depressant
a
Chemotherapies, TMZ, Procarbazine, & CCNU, should not be started until ANC > 1.5, Platelets >100
b
Lab values are based on the lowest count, regardless of day
c
Risk of PCP Pneumonia when TMZ concurrent with radiation, advise appropriate prophylaxis
d
Food rich in tyramine have commonly undergone fermenting and aging, and include cheese, sourdough, smoked/pickled/fermented
meats/fish, fava beans, soy sauce, etc.
31
32 D. Molaie and P. (. Nghiemphu
Surveillance
Treatment at Recurrence
Patient Information
References
1. Kyritsis AP, Bondy ML, Rao JS, Sioka C. Inherited predisposition to
glioma. Neuro Oncol. 2010;12:104–13.
2. Louis DN, et al. The 2016 World Health Organization classification of
tumors of the central nervous system: a summary. Acta Neuropathol.
2016;131:803–20.
3. Baumert BG, et al. Temozolomide chemotherapy versus radiotherapy in
high-risk low-grade glioma: a randomized phase III Intergroup study by
EORTC/NCIC-CTG/TROG/MRC-CTU (EORTC 22033-26033). Lancet
Oncol. 2016;17:1521–32.
4. Hasselblatt M, et al. Diffuse astrocytoma, IDH-wildtype: a dissolving
diagnosis. J Neuropathol Exp Neurol. 2018;77:422–5.
36 D. Molaie and P. (. Nghiemphu
19. Van Den Bent MJ, et al. Long-term efficacy of early versus delayed radio-
therapy for low-grade astrocytoma and oligodendroglioma in adults: the
EORTC 22845 randomised trial. Lancet. 2005;366:985–90.
20. Chang EF, et al. Preoperative prognostic classification system for hemi-
spheric low-grade gliomas in adults: clinical article. J Neurosurg.
2008;109:817–24.
21. Engloti DJ, Chang EG. Epilepsy and brain tumors. Handb Clin Neurol.
2016;134:267–85. https://doi.org/10.1016/j.physbeh.2017.03.040.
22. Ruda R, Bello L, Duffau H, et al. Seizures in low-grade gliomas: natural
history, pathogenesis, and outcome after treatments. Neuro Oncol.
2012;14 Suppl 4(Suppl 4):iv55–64.
23. Boele FW, et al. Health-related quality of life in stable, long-term survi-
vors of low-grade glioma. J Clin Oncol. 2015;33:1023–9.
24. Medical Research Council Antiepileptic Drug Withdrawal Study Group.
Randomised study of antiepileptic drug withdrawal in patients in remis-
sion. Lancet. 1991;337:1175–80.
25. Lossius MI, et al. Consequences of antiepileptic drug withdrawal: a ran-
domized, double-blind study (Akershus study). Epilepsia. 2008;49:455–
63.
26. Vigliani MC, Sichez N, Poisson M, Delattre JY. A prospective study of
cognitive functions following conventional radiotherapy for supratento-
rial gliomas in young adults: 4-year results. Int J Radiat Oncol Biol Phys.
1996;35:527–33.
27. Noll KR, Ziu M, Weinberg JS, Wefel JS, Hospital S. Neurocognitive
functioning in patients with glioma of the left and right temporal lobes. J
Neurooncol. 2016;128:323–31.
28. Meyers CA, Weitzner MA, Valentine AD, Levin VA. Methylphenidate
therapy improves cognition, mood, and function of brain tumor patients.
J Clin Oncol. 1998;16:2522–7.
29. Habets EJJ, et al. Health-related quality of life and cognitive functioning
in long-term anaplastic oligodendroglioma and oligoastrocytoma survi-
vors. J Neurooncol. 2014;116:161–8.
Oligodendroglioma
3
Oluwatosin Akintola
O. Akintola (*)
Department of Neurological Sciences, University of Vermont, Larner
College of Medicine, Burlington, VT, USA
e-mail: Oluwatosin.akintola@uvmhealth.org
anterior aspect of the mass. The case was reviewed by the multi-
disciplinary brain tumor board. Shared decision making was initi-
ated with the patient. The patient proceeded with radiotherapy and
chemotherapy, given the risk of debilitating neurological deficits
if a resection of the mass was attempted.
Post-operative Treatment
Radiation
Chemotherapy
Surveillance
Prognosis
WHO Grade 2, 1p19q co-deleted, IDH WHO Grade 3, 1p19q co-deleted, IDH
Mutant Mutant
• Radiotherapy 54-60Gy
Shared decision making. Proceed to - • Adjuvant PCV x 6 cycles
• Surveillance MRI Head every 3-
6 months for 3-5 years. Expand OR
to 6-12 months if no recurrence.
• Adjuvant temozolomide x 6-12 cycles
OR
Patient Information
References
1. Louis DN, et al. The 2021 WHO classification of tumors of the central
nervous system: a summary. Neuro Oncol. 2021;23(8):1231–51.
2. Morshed RA, et al. Molecular features and clinical outcomes in surgi-
cally treated low-grade diffuse gliomas in patients over the age of 60. J
Neurooncol. 2019;141(2):383–91.
3. Pinkham MB, et al. FISHing tips: what every clinician should know about
1p19q analysis in gliomas using fluorescence in situ hybridisation. Clin
Oncol (R Coll Radiol). 2015;27(8):445–53.
4. Appay R, et al. CDKN2A homozygous deletion is a strong adverse prog-
nosis factor in diffuse malignant IDH-mutant gliomas. Neuro Oncol.
2019;21(12):1519–28.
5. Press RH, et al. Optimal timing of chemoradiotherapy after surgical
resection of glioblastoma: stratification by validated prognostic classifi-
cation. Cancer. 2020;126(14):3255–64.
6. Mohile NA, et al. Therapy for diffuse astrocytic and oligodendroglial
tumors in adults: ASCO-SNO guideline. J Clin Oncol. 2022;40(4):403–
26.
7. Weller M, et al. EANO guidelines on the diagnosis and treatment of dif-
fuse gliomas of adulthood. Nat Rev Clin Oncol. 2021;18(3):170–86.
8. Shaw EG, et al. Randomized trial of radiation therapy plus procarbazine,
lomustine, and vincristine chemotherapy for supratentorial adult low-
grade glioma: initial results of RTOG 9802. J Clin Oncol.
2012;30(25):3065–70.
9. van den Bent MJ, et al. Adjuvant procarbazine, lomustine, and vincristine
chemotherapy in newly diagnosed anaplastic oligodendroglioma: long-
term follow-up of EORTC brain tumor group study 26951. J Clin Oncol.
2013;31(3):344–50.
10. Niyazi M, et al. ESTRO-ACROP guideline “target delineation of glio-
blastomas”. Radiother Oncol. 2016;118(1):35–42.
11. Cairncross G, et al. Phase III trial of chemoradiotherapy for anaplastic
oligodendroglioma: long-term results of RTOG 9402. J Clin Oncol.
2013;31(3):337–43.
12. Nabors LB, et al. NCCN guidelines insights: central nervous system can-
cers, version 1.2017. J Natl Compr Cancer Netw. 2017;15(11):1331–45.
13. Miller JJ. Targeting IDH-mutant glioma. Neurotherapeutics.
2022;19:1724–32.
14. Olson JD, Riedel E, DeAngelis LM. Long-term outcome of low-grade
oligodendroglioma and mixed glioma. Neurology. 2000;54(7):1442–8.
15. Iwadate Y, et al. Eighty percent survival rate at 15 years for 1p/19q co-
deleted oligodendroglioma treated with upfront chemotherapy irrespec-
tive of tumor grade. J Neurooncol. 2019;141(1):205–11.
3 Oligodendroglioma 49
16. Ostrom QT, et al. CBTRUS statistical report: primary brain and other
central nervous system tumors diagnosed in the United States in 2010-
2014. Neuro Oncol. 2017;19(suppl_5):v1–v88.
17. Kerkhof M, et al. Seizures in oligodendroglial tumors. CNS Oncol.
2015;4(5):347–56.
18. Jaeckle KA, et al. CODEL: phase III study of RT, RT + TMZ, or TMZ for
newly diagnosed 1p/19q codeleted oligodendroglioma. Analysis from the
initial study design. Neuro Oncol. 2021;23(3):457–67.
BRAF-Mutated Glioma
4
Karisa C. Schreck
and Jean M. Mulcahy Levy
K. C. Schreck (*)
Johns Hopkins University, Baltimore, MD, USA
e-mail: Ksolt1@jhmi.edu
J. M. Mulcahy Levy
Children’s Hospital Colorado and University of Colorado Anschutz,
Aurora, CO, USA
Fig. 4.1 T1-post gadolinium MRI images from a patient with a BRAF
V600E-mutated epithelioid glioblastoma showing a large solid/cystic enhanc-
ing mass of the right temporal region
BRAF Mutations
Table 4.1 BRAF mutation frequency by tumor type. (Modified with permis-
sion of the authors from Ref. [3])
Tumor type BRAF-SNV (%) BRAF-fusion (%)
Pilocytic astrocytoma 10 60–70
Pediatric low grade astrocytoma 25–35 13
Pediatric high grade astrocytoma 10–20 <1
Adult low grade astrocytoma 5–15 5
Adult high grade astrocytoma 3 <1
Pleomorphic Xanthoastrocytoma 70–80 <1
Ganglioglioma 50 12
Papillary Craniopharyngioma 95 <1
54 K. C. Schreck and J. M. Mulcahy Levy
Post-operative Treatment
High-Grade Glioma
Second Reduce to Reduce to Reduce to Stop Reduce to Reduce to Age ≥ 12 years: Age ≥ 6 years:
dose 75 mg 1 mg once 200 mg binimetinib 480 mg 20 mg once 3 mg/kg/day div 0.015 mg/kg
reduction twice daily daily once daily twice daily daily BID once daily
Age < 12 years: Age < 6 years:
3.75 mg/kg/day 0.015 mg/kg
div BID once daily
(max 2 mg/
day)
BRAF-Mutated Glioma
Third Reduce to Stop Stop Stop Stop Stop dabrafenib Stop trametinib
dose 50 mg trametinib encorafenibc vemurafenib cobimetinib
reduction twice
dailyd
a
Note, if binimetinib is held, reduce encorafenib to a maximum of 300 mg daily until binimetinib is resumed due to increased toxicity
when encorafenib is used as monotherapy
b
Cobimetinib is administered for 21 consecutive days, then held for 7 days during each 28-day cycle
c
If encorafenib or binimetinib is permanently discontinued, discontinue the other targeted therapy (binimetinib or encorafenib) as well
d
Drug is discontinued below this dose level
59
60 K. C. Schreck and J. M. Mulcahy Levy
Treatment at Recurrence
Prognosis/Survivorship
References
1. Ryall S, Arnoldo A, Krishnatry R, Mistry M, Khor K, Sheth J, et al. Mul-
tiplex detection of pediatric low-grade glioma signature fusion transcripts
and duplications using the NanoString nCounter System. J Neuropathol
Exp Neurol. 2017;76(7):562–70.
2. Wen PY, Stein A, van den Bent M, et al. Dabrafenib plus trametinib in
patients with BRAF(V600E)-mutant low-grade and high-grade glioma
(ROAR): a multicentre, open-label, single-arm, phase 2, basket trial. Lan-
cet Oncol. 2022;23:53–64.
3. Schreck KC, Langat P, Bhave VM, et al. Integrated molecular and clinical
analysis of BRAF-mutant glioma in adults. NPJ Precis Oncol. 2023;7:23.
4. Schindler G, Capper D, Meyer J, Janzarik W, Omran H, Herold-Mende C,
et al. Analysis of BRAF V600E mutation in 1320 nervous system tumors
reveals high mutation frequencies in pleomorphic xanthoastrocytoma,
ganglioglioma and extra-cerebellar pilocytic astrocytoma. Acta Neuro-
pathol. 2011;121(3):397–405.
5. Dias-Santagata D, Lam Q, Vernovsky K, Vena N, Lennerz JK, Borger DR,
et al. BRAF V600E mutations are common in pleomorphic xanthoastrocy-
toma: diagnostic and therapeutic implications. PLoS One. 2011;6(3):e17948.
6. Ryall S, Zapotocky M, Fukuoka K, Nobre L, Guerreiro Stucklin A, Ben-
nett J, et al. Integrated molecular and clinical analysis of 1,000 pediatric
low-grade gliomas. Cancer Cell. 2020;37(4):569–583.e5.
7. National Comprehensive Cancer Network. NCCN Guidelines Version
1.2020 Central Nervous Systems Cancer [Internet]. National Comprehen-
sive Cancer Network [cited April 29, 2020].
4 BRAF-Mutated Glioma 65
8. Vuong HG, Altibi AMA, Duong UNP, Ngo HTT, Pham TQ, Fung KM, et
al. BRAF mutation is associated with an improved survival in glioma—a
systematic review and meta-analysis. Mol Neurobiol. 2017;55:3718–24.
9. Lassaletta A, Zapotocky M, Mistry M, Ramaswamy V, Honnorat M,
Krishnatry R, et al. Therapeutic and prognostic implications of BRAF
V600E in pediatric low-grade gliomas. J Clin Oncol.
2017.:JCO2016718726;35:2934–41.
10. Hawkins C, Walker E, Mohamed N, Zhang C, Jacob K, Shirinian M, et al.
BRAF-KIAA1549 fusion predicts better clinical outcome in pediatric
low-grade astrocytoma. Clin Cancer Res. 2011;17(14):4790–8.
11. Chamberlain MC. Salvage therapy with BRAF inhibitors for recurrent
pleomorphic xanthoastrocytoma: a retrospective case series. J Neuroon-
col. 2013;114(2):237–40.
12. Kaley T, Touat M, Subbiah V, Hollebecque A, Rodon J, Lockhart AC, et
al. BRAF inhibition in BRAF(V600)-mutant gliomas: results from the
VE-BASKET study. J Clin Oncol. 2018.:JCO2018789990;36:3477–84.
13. Wen P, Alexander S, van den Bent MJ, De Greve J, Dietrich S, de Vos F,
et al. Updated efficacy and safety of dabrafenib + trametinib in patients
with recurrent or refractory BRAF V60E-mutated high-grade glioma
(HGG) and low-grade glioma (LGG). Neuro Oncol. 2019;21(Suppl
6):vi19–20.
14. Skrypek M, Foreman N, Guillaume D, Moertel C. Pilomyxoid astrocy-
toma treated successfully with vemurafenib. Pediatr Blood Cancer.
2014;61(11):2099–100.
15. del Bufalo F, Carai A, Figa-Talamanca L, Pettorini B, Mallucci C,
Giangaspero F, et al. Response of recurrent BRAFV600E mutated gan-
glioglioma to Vemurafenib as single agent. J Transl Med. 2014;12:356,
014-0356-1.
16. Del Bufalo F, Ceglie G, Cacchione A, Alessi I, Colafati GS, Carai A, et al.
BRAF V600E inhibitor (Vemurafenib) for BRAF V600E mutated low
grade gliomas. Front Oncol. 2018;8:526.
17. Dougherty MJ, Santi M, Brose MS, Ma C, Resnick AC, Sievert AJ, et al.
Activating mutations in BRAF characterize a spectrum of pediatric low-
grade gliomas. Neuro Oncol. 2010;12(7):621–30.
18. Bautista F, Paci A, Minard-Colin V, Dufour C, Grill J, Lacroix L, et al.
Vemurafenib in pediatric patients with BRAFV600E mutated high-grade
gliomas. Pediatr Blood Cancer. 2014;61(6):1101–3.
19. Robinson GW, Orr BA, Gajjar A. Complete clinical regression of a BRAF
V600E-mutant pediatric glioblastoma multiforme after BRAF inhibitor
therapy. BMC Cancer. 2014;14:258, 2407-14-258.
20. Rush S, Foreman N, Liu A. Brainstem ganglioglioma successfully treated
with vemurafenib. J Clin Oncol. 2013;31(10):e159–60.
21. Kieran MW, Bouffet E, Tabori U, Broniscer A, Cohen K, Hansford J, et al.
The first study of dabrafenib in pediatric patients with BRAF V600-
mutant relapsed or refractory low-grade gliomas. Ann Oncol.
2016;27(Supplement 6):vi552–87.
66 K. C. Schreck and J. M. Mulcahy Levy
22. Hargrave DR, Bouffet E, Tabori U, Broniscer A, Cohen KJ, Hansford JR,
et al. Efficacy and safety of Dabrafenib in pediatric patients with BRAF
V600 mutation-positive relapsed or refractory low-grade glioma: results
from a phase I/IIa study. Clin Cancer Res. 2019;25(24):7303–11.
23. Kieran MW, Geoerger B, Dunkel IJ, Broniscer A, Hargrave D, Hingorani
P, et al. A phase I and pharmacokinetic study of oral Dabrafenib in chil-
dren and adolescent patients with recurrent or refractory BRAF V600
mutation-positive solid tumors. Clin Cancer Res. 2019;25(24):7294–302.
24. Jones DTW, Kieran MW, Bouffet E, Alexandrescu S, Bandopadhayay P,
Bornhorst M, et al. Pediatric low-grade gliomas: next biologically driven
steps. Neuro Oncol. 2018;20(2):160–73. 28;11(9):10.3390/cancers11091262.
25. Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ,
et al. Radiotherapy plus concomitant and adjuvant temozolomide for glio-
blastoma. N Engl J Med. 2005;352(10):987–96.
26. Zhang RQ, Shi Z, Chen H, Chung NY, Yin Z, Li KK, et al. Biomarker-
based prognostic stratification of young adult glioblastoma. Oncotarget.
2016;7(4):5030–41.
27. Schreck KC, Guajardo A, Lin DDM, Eberhart CG, Grossman SA. Con-
current BRAF/MEK inhibitors in BRAF V600-mutant high-grade pri-
mary brain tumors. J Natl Compr Cancer Netw. 2018;16(4):343–7.
28. Smith-Cohn M, Davidson C, Colman H, Cohen AL. Challenges of targeting
BRAF V600E mutations in adult primary brain tumor patients: a report of
two cases. CNS Oncol. 2019;8(4):CNS48, 2019-0018. Epub 2019 Dec 10.
29. Marks AM, Bindra RS, DiLuna ML, Huttner A, Jairam V, Kahle KT, et al.
Response to the BRAF/MEK inhibitors dabrafenib/trametinib in an ado-
lescent with a BRAF V600E mutated anaplastic ganglioglioma intolerant
to vemurafenib. Pediatr Blood Cancer. 2018;65(5):e26969.
30. Dummer R, Ascierto PA, Gogas HJ, Arance A, Mandala M, Liszkay G, et
al. Encorafenib plus binimetinib versus vemurafenib or encorafenib in
patients with BRAF-mutant melanoma (COLUMBUS): a multicentre,
openlabel, randomised phase 3 trial. Lancet Oncol. 2018;19(5):603–15.
31. Long GV, Stroyakovskiy D, Gogas H, Levchenko E, de Braud F, Larkin J,
et al. Combined BRAF and MEK inhibition versus BRAF inhibition alone
in melanoma. N Engl J Med. 2014;371(20):1877–88.
32. Brown NF, Carter T, Kitchen N, Mulholland P. Dabrafenib and trametinib
in BRAFV600E mutated glioma. CNS Oncol. 2017;6(4):291–6.
33. Touat M, Gratieux J, Condette Auliac S, Sejean K, Aldea S, Savatovsky J,
et al. Vemurafenib and cobimetinib overcome resistance to vemurafenib
in BRAF-mutant ganglioglioma. Neurology. 2018;91(11):523–5.
Treatment of Meningioma
5
Rimas V. Lukas, Timothy J. Kruser,
and Adam M. Sonabend
Introduction
Clinical Scenario
R. V. Lukas (*)
Department of Neurology, Northwestern University, Chicago, IL, USA
Lou and Jean Malnati Brain Tumor Institute, Northwestern University,
Chicago, IL, USA
e-mail: rimas.lukas@nm.org
T. J. Kruser
Lou and Jean Malnati Brain Tumor Institute, Northwestern University,
Chicago, IL, USA
Department of Radiation Oncology, Northwestern University,
Chicago, IL, USA
SSM Health, Madison, USA
A. M. Sonabend
Lou and Jean Malnati Brain Tumor Institute, Northwestern University,
Chicago, IL, USA
Department of Neurological Surgery, Northwestern University,
Chicago, IL, USA
Diagnostic Evaluation
Imaging
Pathology
Therapeutic Management
Surgery
Radiation
Systemic Therapies
Conclusions
References
1. Buerki RA, Horbinski CM, Kruser T, et al. An overview of meningiomas.
Future Oncol. 2018;14(21):2161–77.
2. Lukas RV, Taylor JW, Kurz SC, Mohile NA. Clinical neuro-oncology for
the neurologist. Neurol Clin Pract. 2020;10(5):458–65. https://doi.
org/10.1212/CPJ.0000000000000765. PMID: 33299675; PMCID:
PMC7717629.
3. Lukas RV, Mrugala MM. Non-malignant brain tumors. Continuum.
2020;26(6):1495–522.
4. Ostrum QT, Gittleman H, Liao P, et al. The CBTRUS statistical report:
primary brain and other central nervous system tumors diagnosed in the
United States in 2011-2015. Neuro Oncol. 2018;20(suppl 4):v1–v86.
5. Sahm F, Toprak UH, Hubschmann D, et al. Meningiomas induced by
low-dose radiation carry structural variants of NF2 and a distinct muta-
tional signature. Acta Neuropathol. 2017;134(1):155–8.
6. Agnihotri S, Suppiah S, Tonge PD, et al. Therapeutic radiation for child-
hood cancer drives structural aberrations of NF2 in meningiomas. Nat
Commun. 2017;8(1):186.
7. Louis DN, Perry A, Wesseling P, et al. The 2021 WHO Classification of
Tumors of the Central Nervous System: a summary. Neuro Oncol.
2021;23(8):1231–51.
8. Louis DN, Perry A, Reifenberger G, et al. The 2016 World Health
Organization classification of tumors of the central nervous system: a
summary. Acta Neuropathol. 2016;131(6):803–20.
9. Olar A, Wani KM, Wilson CD, et al. Global epigenetic profiling identifies
methylation subgroups associated with recurrence-free survival in menin-
gioma. Acta Neuropathol. 2017;133(3):431–44.
10. Sahm F, Schrimpf D, Stichel D, et al. DNA methylation-based classifica-
tion and grading system for meningioma: a multicentre, retrospective
analysis. Lancet Oncol. 2017;18(5):682–94.
11. Brastianos PK, Horowitz PM, Santagata S, et al. Genomic sequencing of
meningiomas identifies oncogenic SMO and AKT1 mutations. Nat
Genet. 2013;45(3):285–9.
12. Aghi MK, Carter BS, Cosgrove GR, et al. Long-term recurrence rates of
atypical meningiomas after gross total resection with or without postop-
erative adjuvant radiation. Neurosurgery. 2009;64(1):56–60.
13. Simpson D. The recurrence of intracranial meningiomas after surgical
treatment. J Neurol Neurosurg Psychiatry. 1957;20(1):22–39.
82 R. V. Lukas et al.
14. Bloch O, Kaur G, Jian BJ, et al. Stereotactic radiosurgery for benign
meningiomas. J Neurooncol. 2012;107(1):13–20.
15. Gondi V, Tome WA, Mehta MP. Fractionated radiotherapy for intracranial
meningiomas. J Neurooncol. 2010;99(3):349–56.
16. Paulsen F, Doerr S, Wilhelm H, et al. Fractionated stereotactic radiother-
apy in patients with optic nerve sheath meningioma. Int J Radiat Oncol
Biol Phys. 2012;82(2):773–8.
17. Rogers L, Zhang P, Vogelbaum MA, et al. Intermediate-risk meningioma:
initial outcomes from NRG oncology RTOG 0539. J Neurosurg.
2018;129(1):35–47.
18. Ji Y, Rankin C, Grunberg S, et al. Double-blind phase III randomized trial
of the antiprogestin agent mifepristone in the treatment of unresectable
meningioma: SWOG S9005. J Clin Oncol. 2015;33:4093–8.
Treatment of Primary CNS
Lymphoma 6
Ugur Sener and Lauren Schaff
Introduction
Clinical Scenario
U. Sener
Department of Neurology, Mayo Clinic, Rochester, MN, USA
L. Schaff (*)
Department of Neurology, Memorial Sloan-Kettering Cancer Center,
New York, NY, USA
e-mail: schaffl@mskcc.org
Epidemiology
Diagnosis
a b c
d e f
Staging
First-Line Treatment
Methotrexate Therapy
Concomitant Medications
Methotrexate also has multiple important drug interactions that
must be considered before starting therapy. When HD-MTX is
administered with proton pump inhibitors (PPIs), serum levels
of methotrexate can be elevated and toxicity may increase.
Severe myelosuppression, aplastic anemia, and gastrointestinal
toxicity may occur when methotrexate is used with nonsteroi-
dal anti-inflammatory drugs (NSAIDs). The antibiotic trime-
thoprim- sulfamethoxazole is a folate antagonist whose
coadministration with methotrexate can potentiate toxicity and
should be avoided [7].
6 Treatment of Primary CNS Lymphoma 91
Methotrexate Toxicity
Despite urine alkalinization and intravenous hydration, acute kid-
ney injury (AKI) with methotrexate administration can occur
through mechanisms such as precipitation in the renal tubules,
vasoconstriction, and direct tubular toxicity. If AKI develops,
intravenous fluids should be increased to ≥3 L/m2 per day to max-
imize urine output [8]. AKI can result in delayed methotrexate
clearance and toxic levels which can lead to systemic effects such
as hepatic injury, pneumonitis, and bone marrow suppression.
Gastrointestinal toxicities including oral mucositis, ulcerative sto-
matitis, and hemorrhagic enteritis have also been reported.
Neurologic toxicities include headache, encephalopathy, leukoen-
cephalopathy, and transient focal neurologic deficits. Rarely tox-
icity can be fatal.
In the event of AKI and methotrexate toxicity, glucarpidase is
FDA approved to rapidly reduce serum methotrexate levels.
Glucarpidase is dosed at 50 units/kg though there is evidence that
lower doses may be as effective [9]. Leucovorin should not be
administered within 2 h of glucarpidase injection since leucovorin
is also a substrate for glucarpidase. If glucarpidase is not avail-
able, hemodialysis can be considered. In the event of delayed
clearance and nephrotoxicity, reduction in methotrexate dose may
be considered for future cycles. Table 6.1 provides a summary of
methotrexate treatment including precautions and supportive
measures that should be undertaken.
R-MVP
R-MVP consists of rituximab, methotrexate (3.5 g/m2), vincris-
tine, and procarbazine. Treatment cycles are 28 days long with
methotrexate administered twice per cycle. The optimal number
of cycles is not known though we favor completion of 4 (8 doses
of methotrexate) with imaging for initial response obtained after
two.
When to incorporate vincristine is a special consideration for
the use of this regimen. Vincristine is a vinca alkaloid that inhibits
microtubule formation, causing arrest of cell division during
metaphase, leading to apoptosis. Toxicities from vincristine
include hair loss, constipation, and most significantly, peripheral
neuropathy. Vincristine is dosed at 1.6 mg/m2, capped at 2.4 mg.
The drug is only administered four times during the induction
regimen to limit toxicity. CNS penetration of vincristine is ques-
tionable, and the drug may be omitted in patients with limited
enhancing disease or pre-existing neuropathy. If neuropathy
symptoms develop while on treatment, early discontinuation of
vincristine should be considered. Vincristine is a vesicant and can
cause tissue damage in the event of extravasation, mandating
careful placement and confirmation of the intravenous line by an
experienced individual prior to treatment.
Procarbazine is an oral chemotherapy agent with unclear
mechanism of action, though likely serves as an alkylator.
Procarbazine can cause hepatotoxicity and dose reduction or dis-
continuation should be considered if this occurs. Patients should
be cautioned that ingestion of alcohol while taking procarbazine
can cause a disulfiram-like reaction. Procarbazine is a weak
monoamine oxidase inhibitor with potential for hypertensive cri-
sis upon ingestion of tyramine-rich foods or coadministration of
sympathomimetic agents.
R-MVP is often followed by use of filgrastim or pegfilgrastim,
a human recombinant form of granulocyte colony-stimulating
factor (G-CSF) which can stimulate production of neutrophils
[14]. G-CSF can be started 24 h after clearance of methotrexate.
Use of G-CSF is designed to prevent infectious complications of
94 U. Sener and L. Schaff
T-R
M
MT-R includes methotrexate, temozolomide, and rituximab.
Temozolomide is an alkylating chemotherapy agent that is typi-
cally well-tolerated. Hepatotoxicity, thrombocytopenia, and lym-
phopenia complicated by Pneumocystis jirovecii pneumonia
(PJP) can occur.
MATRix
MATRix includes methotrexate, cytarabine, thiotepa, and ritux-
imab. Cytarabine is an antimetabolite that inhibits DNA s ynthesis.
Cytarabine and thiotepa can both cause myelosuppression, hepa-
totoxicity, and pulmonary toxicity. Additional toxicities from
cytarabine such as cerebellar ataxia, corneal toxicity, and gastric
ulcers tend to occur with high-dose treatment.
R-MBVP
R-MBVP includes rituximab, methotrexate, BCNU, etoposide,
prednisone. BCNU is an alkylator, which can cause myelosup-
pression and dose-dependent pulmonary toxicity, mandating
monitoring with pulmonary function tests before and during treat-
ment. In order to minimize risk of pulmonary fibrosis, BCNU has
a lifetime dose limit of 1400 mg/m2. Etoposide is a topoisomerase
II inhibitor that prevents DNA replication. Etoposide can cause
myelosuppression, sensitivity reaction, and skin necrosis in the
event of extravasation.
Role of Rituximab
Site-Specific Therapy
Relapsed Disease
Patient Information
What Is My Prognosis?
References
1. Ostrom QT, et al. CBTRUS statistical report: primary brain and other
central nervous system tumors diagnosed in the United States in 2012-
2016. Neuro Oncol. 2019;21(Supplement_5):v1–v100.
2. Villano JL, et al. Age, gender, and racial differences in incidence and
survival in primary CNS lymphoma. Br J Cancer. 2011;105(9):1414–8.
3. Schaff LR, Grommes C. Updates on primary central nervous system
lymphoma. Curr Oncol Rep. 2018;20(2):11.
4. Grommes C, DeAngelis LM. Primary CNS lymphoma. J Clin Oncol.
2017;35(21):2410–8.
5. Borsi JD, Moe PJ. A comparative study on the pharmacokinetics of
methotrexate in a dose range of 0.5 g to 33.6 g/m2 in children with acute
lymphoblastic leukemia. Cancer. 1987;60(1):5–13.
6. Shapiro WR, Young DF, Mehta BM. Methotrexate: distribution in cere-
brospinal fluid after intravenous, ventricular and lumbar injections. N
Engl J Med. 1975;293(4):161–6.
7. Al-Quteimat OM, Al-Badaineh MA. Practical issues with high dose
methotrexate therapy. Saudi Pharma J. 2014;22(4):385–7.
8. Howard SC, et al. Preventing and managing toxicities of high-dose
methotrexate. Oncologist. 2016;21(12):1471–82.
9. Schaff LR, Lobbous M, Carlow D, Schofield R, Gavrilovic IT, Miller
AM, Stone JB, Piotrowski AF, Sener U, Skakodub A, Acosta EP, Ryan
KJ, Mellinghoff IK, DeAngelis LM, Nabors LB, Grommes C. Routine
use of low-dose glucarpidase following high-dose methotrexate in adult
patients with CNS lymphoma: an open-label, multi-center phase I study.
BMC Cancer. 2022;22(1):60. https://doi.org/10.1186/s12885-021-
09164-x. PMID: 35027038; PMCID: PMC8756618.
10. Morris PG, et al. Rituximab, methotrexate, procarbazine, and vincristine
followed by consolidation reduced-dose whole-brain radiotherapy and
cytarabine in newly diagnosed primary CNS lymphoma: final results
and long-term outcome. J Clin Oncol. 2013;31(31):3971–9.
11. Rubenstein JL, et al. Intensive chemotherapy and immunotherapy in
patients with newly diagnosed primary CNS lymphoma: CALGB 50202
(Alliance 50202). J Clin Oncol. 2013;31(25):3061–8.
12. Ferreri AJ, et al. Chemoimmunotherapy with methotrexate, cytarabine,
thiotepa, and rituximab (MATRix regimen) in patients with primary
CNS lymphoma: results of the first randomisation of the International
Extranodal Lymphoma Study Group-32 (IELSG32) phase 2 trial. Lan-
cet Haematol. 2016;3(5):e217–27.
13. Bromberg JEC, et al. Rituximab in patients with primary CNS lym-
phoma (HOVON 105/ALLG NHL 24): a randomised, open-label, phase
3 intergroup study. Lancet Oncol. 2019;20(2):216–28.
102 U. Sener and L. Schaff
Clinical Presentation
T. McGranahan
Departments of Neurology and Pediatrics, Stanford University and
Lucile Packard Children’s Hospital, Palo Alto, CA, USA
S. Partap (*)
Departments of Neurology and Neurological Surgery, University of
Washington, Seattle, WA, USA
e-mail: spartap@stanford.edu
Genetic Testing
Radiographic Finding
Staging
The majority of studies have not found the risk stratification used
in children (Table 7.3) to be prognostic in adults. As a result, mul-
tiple definitions have been used to characterize high and average
Surgery
Craniospinal Irradiation
Systemic Therapy
Surveillance
Prognosis
True prognosis and survival data are limited given rarity of dis-
ease, wide variations in treatment and limited prospective studies.
For adults, the SEER and CBTRUS databases estimate 2, 5 and
10 year survival as 85–89%, 74–78% and 67–68% [13, 14].
Molecular subtypes help with stratifying prognosis with WNT
having the best prognosis of 5 year survival of 80%, followed by
SHH with a 5 years survival of 70% and Group 4 of 5 year sur-
vival of less than 50% [3].
Survivorship
Future Directions
References
1. Waszak SM, Northcott PA, Buchhalter I, et al. Spectrum and prevalence
of genetic predisposition in medulloblastoma: a retrospective genetic
study and prospective validation in a clinical trial cohort. Lancet Oncol.
2018;19:785–98.
2. Chang CH, Housepian EM, Herbert C Jr. An operative staging system and
a megavoltage radiotherapeutic technique for cerebellar medulloblasto-
mas. Radiology. 1969;93:1351–9.
3. Zhao F, Ohgaki H, Xu L, et al. Molecular subgroups of adult medullo-
blastoma: a long-term single-institution study. Neuro Oncol.
2016;18:982–90.
4. Louis DN, Perry A, Reifenberger G, et al. The 2016 World Health
Organization classification of tumors of the central nervous system: a
summary. Acta Neuropathol. 2016;131:803–20.
5. Kool M, Korshunov A, Remke M, et al. Molecular subgroups of medul-
loblastoma: an international meta-analysis of transcriptome, genetic
aberrations, and clinical data of WNT, SHH, Group 3, and Group 4
medulloblastomas. Acta Neuropathol. 2012;123:473–84.
6. Remke M, Hielscher T, Northcott PA, et al. Adult medulloblastoma com-
prises three major molecular variants. J Clin Oncol. 2011;29:2717–23.
7. Franceschi E, Hofer S, Brandes AA, et al. EANO–EURACAN clinical
practice guideline for diagnosis, treatment, and follow-up of post-pubertal
and adult patients with medulloblastoma. Lancet Oncol. 2019;20:e715–
28.
8. De B, Beal K, De Braganca KC, et al. Long-term outcomes of adult
medulloblastoma patients treated with radiotherapy. J Neurooncol.
2018;136:95–104.
9. Brown AP, Barney CL, Grosshans DR, et al. Proton beam craniospinal
irradiation reduces acute toxicity for adults with medulloblastoma. Int J
Radiat Oncol Biol Phys. 2013;86:277–84.
118 T. McGranahan and S. Partap
J. Wu (*) · S. Ranjan
Department of Neurology, Cleveland Clinic Florida, Weston, FL, USA
e-mail: Jing.wu3@nih.gov
© The Author(s), under exclusive license to Springer Nature 119
Switzerland AG 2023
N. A. Mohile, A. A. Thomas (eds.), Brain Tumors,
https://doi.org/10.1007/978-3-031-41413-8_8
120 J. Wu and S. Ranjan
a b c
a b
Fig. 8.2 Sagittal T2 sequence (a) and T1 post-contrast sequence (b) of the
thoracic MRI showing sausage-shaped intradural and extramedullary masses
at the T10 and T12 levels
apy to the residual tumor and tumor bed to a total dose of 59.4 Gy
in 33 fractions. Three years later, the patient started complaining
of progressive back pain radiating to the lower anterior abdomen.
An MRI of the spine revealed intradural extramedullary masses at
T10 and T12 level (Fig. 8.2). These tumors were resected and a
pathological exam confirmed a grade 2 ependymoma. No evi-
8 Treatment of Ependymoma 121
Surgical Role
have poor prognosis [16, 17]. PFB are found in children and
adults, have genome-wide polyploidy, low CpG island methylation
and good outcomes. Posterior fossa subependymoma has a bal-
anced genome and a good outcome. Ependymomas occuring in
the spine include are spinal ependymoma, myxopapillary ependy-
moma, and subependymoma. Spinal ependymomas have frequent
NF2 gene mutation and frequently occur in neurofibromatosis
type 2 patients. These have good outcomes. Spinal myxopapillary
ependymomas have genome-wide polyploidy and good progno-
sis. Spinal subependymomas are associated with 6q deletion and
have good prognosis.
Post-operative Treatment
Radiation Therapy
Chemotherapy
Other Modalities
Surveillance
is imaged with contrasted MRI brain and spine every 3 months for
the first 2 years after treatment, then every 4 months for the next
2 years and every 6 months thereafter.
Treatment at Recurrence
References
1. Rodriguez D, Cheung MC, Housri N, Quinones-Hinojosa A, Camphausen
K, Koniaris LG. Outcomes of malignant CNS ependymomas: an exami-
nation of 2408 cases through the Surveillance, Epidemiology, and End
Results (SEER) database (1973-2005). J Surg Res. 2009;156(2):340–51.
https://doi.org/10.1016/j.jss.2009.04.024.
2. Robertson PL, Zeltzer PM, Boyett JM, Rorke LB, Allen JC, Geyer JR,
et al. Survival and prognostic factors following radiation therapy and che-
motherapy for ependymomas in children: a report of the Children’s
Cancer Group. J Neurosurg. 1998;88(4):695–703. https://doi.org/10.3171/
jns.1998.88.4.0695.
3. Healey EA, Barnes PD, Kupsky WJ, Scott RM, Sallan SE, Black PM,
et al. The prognostic significance of postoperative residual tumor in epen-
dymoma. Neurosurgery. 1991;28(5):666–71; discussion 71–2. https://
doi.org/10.1097/00006123-199105000-00005.
4. Duffner PK, Krischer JP, Sanford RA, Horowitz ME, Burger PC, Cohen
ME, et al. Prognostic factors in infants and very young children with
intracranial ependymomas. Pediatr Neurosurg. 1998;28(4):215–22.
https://doi.org/10.1159/000028654.
5. Vera-Bolanos E, Aldape K, Yuan Y, Wu J, Wani K, Necesito-Reyes MJ,
et al. Clinical course and progression-free survival of adult intracranial
and spinal ependymoma patients. Neuro Oncol. 2015;17(3):440–7.
https://doi.org/10.1093/neuonc/nou162.
6. Foreman NK, Love S, Gill SS, Coakham HB. Second-look surgery for
incompletely resected fourth ventricle ependymomas: technical case
136 J. Wu and S. Ranjan
18. Brown DA, Goyal A, Takami H, Graffeo CS, Mahajan A, Krauss WE,
et al. Radiotherapy in addition to surgical resection may not improve
overall survival in WHO grade II spinal ependymomas. Clin Neurol
Neurosurg. 2020;189:105632. https://doi.org/10.1016/j.cli-
neuro.2019.105632.
19. Oh MC, Ivan ME, Sun MZ, Kaur G, Safaee M, Kim JM, et al. Adjuvant
radiotherapy delays recurrence following subtotal resection of spinal cord
ependymomas. Neuro Oncol. 2013;15(2):208–15. https://doi.
org/10.1093/neuonc/nos286.
20. Merchant TE, Bendel AE, Sabin ND, Burger PC, Shaw DW, Chang E,
et al. Conformal radiation therapy for pediatric ependymoma, chemo-
therapy for incompletely resected ependymoma, and observation for
completely resected, supratentorial ependymoma. J Clin Oncol.
2019;37(12):974–83. https://doi.org/10.1200/jco.18.01765.
21. Merchant TE, Li C, Xiong X, Kun LE, Boop FA, Sanford RA. Conformal
radiotherapy after surgery for paediatric ependymoma: a prospective
study. Lancet Oncol. 2009;10(3):258–66. https://doi.org/10.1016/s1470-
2045(08)70342-5.
22. Abdulaziz M, Mallory GW, Bydon M, De la Garza Ramos R, Ellis JA,
Laack NN, et al. Outcomes following myxopapillary ependymoma resec-
tion: the importance of capsule integrity. Neurosurg Focus. 2015;39(2):E8.
https://doi.org/10.3171/2015.5.focus15164.
23. Sonneland PR, Scheithauer BW, Onofrio BM. Myxopapillary ependy-
moma. A clinicopathologic and immunocytochemical study of 77 cases.
Cancer. 1985;56(4):883–93. https://doi.org/10.1002/1097-
0142(19850815)56:4<883::aid-cncr2820560431>3.0.co;2-6.
24. Fassett DR, Pingree J, Kestle JR. The high incidence of tumor dissemina-
tion in myxopapillary ependymoma in pediatric patients. Report of five
cases and review of the literature. J Neurosurg. 2005;102(1 Suppl):59–
64. https://doi.org/10.3171/ped.2005.102.1.0059.
25. Kukreja S, Ambekar S, Sin AH, Nanda A. Cumulative survival analysis of
patients with spinal myxopapillary ependymomas in the first 2 decades of
life. J Neurosurg Pediatr. 2014;13(4):400–7. https://doi.org/10.3171/201
4.1.peds13532.
26. Grill J, Le Deley M-C, Gambarelli D, Raquin M-A, Couanet D, Pierre-
Kahn A, et al. Postoperative chemotherapy without irradiation for epen-
dymoma in children under 5 years of age: a multicenter trial of the French
Society of Pediatric Oncology. J Clin Oncol. 2001;19(5):1288–96.
27. Grundy RG, Wilne SA, Weston CL, Robinson K, Lashford LS, Ironside
J, et al. Primary postoperative chemotherapy without radiotherapy for
intracranial ependymoma in children: the UKCCSG/SIOP prospective
study. Lancet Oncol. 2007;8(8):696–705. https://doi.org/10.1016/s1470-
2045(07)70208-5.
28. Garvin JH Jr, Selch MT, Holmes E, Berger MS, Finlay JL, Flannery A,
et al. Phase II study of pre-irradiation chemotherapy for childhood intra-
138 J. Wu and S. Ranjan
Principles of Treatment
M. Kaur
Loma Linda University Health, Loma Linda, CA, USA
R. Thomas (*)
Stanford University, Palo Alto, CA, USA
e-mail: Reenat@stanford.edu
ing any guidance [5]. Below are the recommendations from our
general practice. For patients requiring chronic dosing of steroids,
doses should rarely exceed 8 mg per day. Twice daily dosing is
ideal, with the second dose given in the early afternoon to prevent
insomnia.
Patients who are started on steroids but otherwise have mini-
mal or no symptoms, should be tapered off rapidly. For patients
who have been on steroids for a short period, the rapid taper
should not cause any steroid withdrawal symptoms and they can
be tapered off within a few days. The effects of a taper are typi-
cally noticeable 72 h after a dose change and tapers should involve
dose adjustments every 3 days. If they have been on steroids for
more than 2 weeks, then doses should by dropped by 2 mg every
3 days. For some patients who are symptomatic during their treat-
ment course, tapering off of steroids can be very difficult. As in
other cases, we recommend starting a taper by decreasing dexa-
methasone by 1–2 mg every 3–5 days. If patients experience neu-
rologic symptoms due to the taper, we recommend going back to
the dose they previously tolerated. The taper should then be
slower, generally 0.5–1 mg every 5–7 days. In patients that can
tolerate a taper and become steroid-dependent, alternative
approaches should be considered [5, 6].
Steroid Toxicities
Treatment Treatment
Take second dose early in • Oral hypoglycemics
the afternoon (before • Attempt to keep
2 p.m.) glucose before 150 mg/dL
Good bedtime hygiene (no
screen time at least 2 h
prior to bed)
(continued)
147
Table 9.2 (continued)
148
Preventative Measures
Patient Instructions
Timing: If giving twice a day dosing, please take the first dose
early in the morning (by 8 a.m.) and second dose no later than
2 p.m. to prevent insomnia and well as nocturia which can further
lead to sleep deprivation.
Stomach protection: Continue to take medications to protect
your stomach from gastritis especially if you are on a higher dose
(>2 mg/day) of dexamethasone. We recommend famotidine or
omeprazole to be taken twice a day.
Glucose: Many patients develop elevated glucose levels, even
if they had no concerns with pre- diabetes or diabetes prior to
starting steroids. We recommend a close monitor on food intake
as increased appetite due to steroids with potential for insulin
resistance and quickly lead to elevated glucose levels that can be
detrimental.
Mood: Please be mindful that steroids can cause many differ-
ent mood disorders including depression, anxiety as well as frank
psychosis. If you feel these are affecting your daily functioning
and relationship, we advise to bring this to the attention of your
prescriber.
Myopathy: To prevent weakness, especially if you are antici-
pated to be on steroids for a long time, we strongly advice daily
152 M. Kaur and R. Thomas
References
1. Posner JB, DeAngelis LM, Deangelis LM. Neurologic complications of
cancer. Oxford: Oxford University Press; 2009.
2. Nag S, Manias JL, Stewart DJ. Pathology and new players in the patho-
genesis of brain edema. Acta Neuropathol. 2009;118:197–217.
3. Kim H, Lee JM, Park JS, Jo SA, Kim Y-O, Kim C-W, Jo I. Dexamethasone
coordinately regulates angiopoietin-1 and VEGF: a mechanism of gluco-
corticoid- induced stabilization of blood–brain barrier. Biochem Biophys
Res Commun. 2008;372(1):243–8. ISSN 0006-291X. https://doi.
org/10.1016/j.bbrc.2008.05.025.
4. Drappatz J. Management of vasogenic edema in patients with primary and
metastatic brain tumors. In: Wen P, editor. Uptodate. 2020.
5. Vecht CJ, Hovestadt A, Verbiest HB, et al. Dose-effect relationship of
dexamethasone on Karnofsky performance in metastatic brain tumors: a
randomized study of doses of 4, 8, and 16 mg per day. Neurology.
1994;44(4):675–80.
6. Roth P, Happold C, Weller M. Corticosteroid use in neuro-oncology: an
update. Neurooncol Pract. 2015;2(1):6–12. https://doi.org/10.1093/nop/
npu029. Advance Access date 9 October 2014.
7. Fardet L, Petersen I, Nazareth I. Monitoring of patients on long-term glu-
cocorticoid therapy: a population-based cohort study. Medicine
(Baltimore). 2015;94(15):e647.
8. Ajlan A, Thomas P, Albakr A, Nagpal S, Recht L. Optimizing bevaci-
zumab dosing in glioblastoma: less is more. J Neurooncol.
2017;135(1):99–105. https://doi.org/10.1007/s11060-017-2553-2.
Tumor Related Epilepsy
10
Thomas Wychowski
T. Wychowski (*)
University of Rochester, Rochester, NY, USA
e-mail: Thomas_wychowski@urmc.rochester.edu
References
1. Cascino GD. Epilepsy and brain tumors: implications for treatment.
Epilepsia. 1990;31(Suppl 3):S37–44.
2. van Breemen MSMW, Erik B, Vecht CJ. Epilepsy in patients with brain
tumours: epidemiology, mechanisms, and management. Lancet Neurol.
2007;6(5):421–30.
3. Wychowski T, Wang H, Buniak L, Henry JC, Mohile N. Considerations
in prophylaxis for tumor-associated epilepsy: prevention of status epilep-
ticus and tolerability of newer generation ASMs. Clin Neurol Neurosurg.
2013;115(11):2365–9.
4. Hildebrand J, Lecaille C, Perennes J, Delattre JY. Epileptic seizures dur-
ing follow-up of patients treated for primary brain tumors. Neurology.
2005;65(2):212–5.
5. Fisher RS, Acevedo C, Arzimanoglou A, et al. ILAE official report: a
practical clinical definition of epilepsy. Epilepsia. 2014;55(4):475–82.
6. Moots PL, Maciunas RJ, Eisert DR, Parker RA, Laporte K, Abou-Khalil
B. The course of seizure disorders in patients with malignant gliomas.
Arch Neurol. 1995;52(7):717–24.
7. Sutter R, Marsch S, Fuhr P, Ruegg S. Mortality and recovery from refrac-
tory status epilepticus in the intensive care unit: a 7-year observational
study. Epilepsia. 2013;54(3):502–11.
8. Wasilewski A, Serventi J, Kamalyan L, Wychowski T, Mohile N. Acute
care in glioblastoma: the burden and the consequences. Neurooncol
Pract. 2017;4(4):248–54.
9. van Breemen MS, Wilms EB, Vecht CJ. Epilepsy in patients with brain
tumours: epidemiology, mechanisms, and management. Lancet Neurol.
2007;6(5):421–30.
10. Köhling R, Senner V, Paulus W, Speckmann EJ. Epileptiform activity
preferentially arises outside tumor invasion zone in glioma xenotrans-
plants. Neurobiol Dis. 2006;22(1):64–75.
11. Wychowski T, James Fessler A, Nadler J, et al. A feasibility study of elec-
trocorticography during routine craniotomy to predict tumor-related epi-
lepsy. Interdiscipl Neurosurg. 2020;21:100772.
12. Zhong Z, Wang Z, Wang Y, You G, Jiang T. IDH1/2 mutation is associated
with seizure as an initial symptom in low-grade glioma: a report of 311
Chinese adult glioma patients. Epilepsy Res. 2015;109:100–5.
13. Stockhammer F, Misch M, Helms HJ, et al. IDH1/2 mutations in WHO
grade II astrocytomas associated with localization and seizure as the ini-
tial symptom. Seizure. 2012;21(3):194–7.
14. Buckingham SC, Campbell SL, Haas BR, et al. Glutamate release by pri-
mary brain tumors induces epileptic activity. Nat Med. 2011;17(10):1269–
74.
164 T. Wychowski
Prevention of VTE
Patient Instructions
References
1. Olson NC, Cushman M, Judd SE, et al. American Heart Association’s
Life’s simple 7 and risk of venous thromboembolism: the reasons for
geographic and racial differences in stroke (REGARDS) study. J Am
Heart Assoc. 2015;4:e001494.
2. Sartori MT, Della Puppa A, Ballin A, Saggiorato G, Bernardi D, Padoan
A, Scienza R, d’Avella D, Cella G. Prothrombotic state in glioblastoma
multiforme: an evaluation of the procoagulant activity of circulating mic-
roparticles. J Neurooncol. 2011;104(1):225–31.
3. Riedl J, Preusser M, Nazari PM, Posch F, Panzer S, Marosi C, Birner P,
Thaler J, Brostjan C, Lötsch D, Berger W, Hainfellner JA, Pabinger I, Ay
C. Podoplanin expression in primary brain tumors induces platelet aggre-
gation and increases risk of venous thromboembolism. Blood.
2017;129(13):1831–9. https://doi.org/10.1182/blood-2016-06-720714.
Epub 2017 Jan 10.
4. Nabi S, Kahlon P, Borzognia F, Arshad A, Mikkelsen T, Donthireddy
V. Predictors of venous thromboembolism in patients with glioblastoma.
Pathol Oncol Res. 2016;22:311–6.
5. Khorana AA, Kuderer NM, Culakova E, Lyman GH, Francis
CW. Development and validation of a predictive model for chemother-
apy-associated thrombosis. Blood. 2008;111:4902–7.
6. Helmi A, Chan A, Towfighi S, Kapadia A, Perry J, Ironside S,
Machnowska M, Symons SP, Fox AJ, Sahgal A, Maralani PJ. Incidence
of dural venous sinus thrombosis in patients with glioblastoma and its
implications. World Neurosurg. 2019;125:e189–97.
7. Semrad TJ, O’Donnell R, Wun T, et al. Epidemiology of venous throm-
boembolism in 9489 patients with malignant glioma. J Neurosurg.
2007;106(4):601–8.
8. Duman T, Uluduz D, Midi I, Bektas H, Kablan Y, Goksel BK, et al. A
multicenter study of 1144 patients with cerebral venous thrombosis: the
VENOST study. J Stroke Cerebrovasc Dis. 2017;26(8):1848–57.
9. Zuurbier SM, Hiltunen S, Lindgren E, Silvis SM, Jood K, Devasagayam
S, et al. Cerebral venous thrombosis in older patients. Stroke.
2018;49(1):197–200.
10. Perry JR, Julian JA, LaPerriere NJ, Geerts W, Agnelli G, Roges LR,
Malkin MG, Sawaya R, Baker R, Falanga A, Parpia S, Finch T, Levine
MN. PRODIGE: a randomized placebo-controlled trial of dalteparin low-
molecular-weight heparin thromboprophylaxis in patients with newly
diagnosed malignant glioma. J Thromb Haemost. 2010;8:1959–65.
11. Carrier M, Abou-Nassar K, Mallick R, Tagalakis V, Shivakumar S,
Schattner A, Kuruvilla P, Hill D, Spadafora S, Marquis K, Trinkaus M,
Tomiak A, Lee AYY, Gross PL, Lazo-Langner A, El-Maraghi R, Goss G,
Le Gal G, Stewart D, Ramsay T, Rodger M, Witham D, Wells
PS. Apixaban to prevent venous thromboembolism in patients with can-
cer. N Engl J Med. 2019;380(8):711–9.
176 S.-P. Weathers and A. Ou
12. Key NS, Khorana AA, Kuderer NM, Bohlke K, Lee AYY, Arcelus JI, Wong
SL, Balaban EP, Flowers CR, Francis CW, Gates LE, Kakkar AK, Levine
MN, Liebman HA, Tempero MA, Lyman GH, Falanga A. Venous thrombo-
embolism prophylaxis and treatment in patients with cancer: ASCO clinical
practice guideline update. J Clin Oncol. 2020;38(5):496–520.
13. National Comprehensive Cancer Network. Cancer-associated venous
thromboembolic disease (version 1.2019). https://www.nccn.org/profes-
sionals/physician_gls/pdf/vte.pdf. Accessed 6 Feb 2020.
14. Agnelli G, Piovella F, Buoncristiani P, Severi P, Pini M, D’Angelo A,
Beltrametti C, Damiani M, Andrioli GC, Pugliese R, Iorio A, Brambilla
G. Enoxaparin plus compression stockings compared with compression
stockings alone in the prevention of venous thromboembolism after elec-
tive neurosurgery. N Engl J Med. 1998;339:80–5.
15. Lee AYY, Levine MN, Baker RI, Bowden C, Kakkar AK, Prins M,
Rickles FR, Julian JA, Haley S, Kovacs MJ, Gent M. Low-molecular-
weight heparin versus a Coumarin for the prevention of recurrent venous
thromboembolism in patients with cancer. N Engl J Med. 2003;349:146–
53.
16. McBane R, Wysokinski W, Le-Rademacher J, Zemla T, Ashrani A, Tafur
A, Perepu U, Anderson D, Gundabolu K, Kuzma C, Botero JP, Ferre
RAL, Henkin S, Lenz CJ, Houghton DE, Vishnu P, Loprinzi CL. Apixaban
and dalteparin in active malignancy-associated venous thromboembo-
lism: the ADAM VTE trial. J Thromb Haemost. 2020;18:411–21.
17. Einstein Investigators, Bauersachs R, Berkowitz SD, et al. Oral rivaroxa-
ban for symptomatic venous thromboembolism. N Engl J Med.
2010;363:2499–510.
18. Bannow BTS, Lee A, Khorana AA, Zwicker JI, Noble S, Ay C, Carrier
M. Management of cancer-associated thrombosis in patients with throm-
bocytopenia: guidance from the SSC of the ISTH. J Thromb Haemost.
2018;16(6):1246–9.
19. Mantia C, Uhlmann EJ, Puligandla M, Weber GM, Neuberg D, Zwicker
JI. Predicting the higher rate of intracranial hemorrhage in glioma patients
receiving therapeutic enoxaparin. Blood. 2017;129(25):3379–85.
20. Carney BJ, Uhlmann EJ, Pligandla M, Mantia C, Weber GM, Neuberg
DS, Zwicker JI. Intracranial hemorrhage with direct oral anticoagulants
in patients with brain tumors. J Thromb Haemost. 2019;17(1):72–6.
Management
of Neurocognitive 12
Symptoms
Christina Weyer-Jamora
and Jennie W. Taylor
C. Weyer-Jamora
Department of Neurological Surgery, University of California, San
Francisco, San Francisco, CA, USA
Department of Psychiatry, Zuckerberg San Francisco General Hospital,
San Francisco, CA, USA
J. W. Taylor (*)
Department of Neurology, University of California, San Francisco,
San Francisco, CA, USA
e-mail: Jennie.taylor@ucsf.edu
Language Impairment
Non-pharmacological Interventions
Exercise
Pharmacologic Interventions
Methylphenidate
Modafenil
Donepezil
Memantine
dropout rate was high in this clinical trial. Additional studies sup-
port preserving cognition by combining memantine with whole
brain radiation with hippocampal avoidance.
Individuals with brain tumors who meet criteria for clinical
depression and/or anxiety may consider antidepressants such as
selective serotonin reuptake inhibitors (SSRIs). Though SSRIs
may not directly improve cognition, effective mood management
can lead to improvement in cognitive symptoms, as discussed
above. Similarly, antipsychotics may improve HROL in patients
with significant behavioral problems.
As patients near the end of their life, issues such as acute confu-
sion, reduced communication, and diminished memory can be
quite distressing for both patients and caregivers. In general,
reducing environmental stimulation (e.g. low light and noise, lim-
ited visitation), planning activities when the patient is most alert,
and providing calm reassurance (as needed) are helpful strategies.
Rapid changes can magnify confusion, and consistency in care
providers, talking through tasks with simple instructions (such as
helping with ADLs), and maintaining a consistent daily routine
help to minimize confusion. Other strategies include ensuring
sensory adaptation for hearing and vision, keeping instructions
simple with frequent repeating, and using diversional activities
(e.g. folding items, looking at pictures, etc.). Antipsychotics such
as olanzapine, haloperidol, or quetiapine may be helpful toward
the end of life for effective behavior management for treatment of
agitation and disorientation.
In conclusion, patients with CNS involvement of their malig-
nancy commonly experience cognitive impairment from direct
injury from tumor or radiotherapy/chemotherapy, and may be
exacerbated by seizures, medications, mood and sleep distur-
bances. These symptoms can significantly impair their quality
of life and are important to identify and treat with a combina-
tion of both non-pharmacologic and pharmacologic interven-
tions.
192 C. Weyer-Jamora and J. W. Taylor
Suggested Reading
Coomans MB, van der Linden SD, Gehring K, Taphoorn MJB. Treatment of
cognitive deficits in brain tumour patients: current status and future direc-
tions. Curr Opin Oncol. 2019;31(6):540–7.
Cormie P, Nowak AK, Chambers SK, Galvão DA, Newton RU. The potential
role of exercise in neuro-oncology. Front Oncol. 2015;5:85.
Correa DD, DeAngelis LM, Shi W, Thaler HT, Lin M, Abrey LE. Cognitive
functions in low-grade gliomas: disease and treatment effects. J Neuroon-
col. 2007;81(2):175–84.
Johnstone B, Stonnington HH, editors. Rehabilitation of neuropsychological
disorders: a practical guide for rehabilitation professionals. 2nd ed.
New York: Psychology Press; 2009.
Karschnia P, Parsons MW, Dietrich J. Pharmacologic management of cogni-
tive impairment induced by cancer therapy. Lancet Oncol. 2019;20(2):e92–
e102.
van Lonkhuizen PJC, Klaver KM, Wefel JS, Sitskoorn MM, Schagen SB,
Gehring K. Interventions for cognitive problems in adults with brain can-
cer: a narrative review. Eur J Cancer Care. 2019;28(3):e13088.
Madhusoodanan S, Ting MB, Farah T, Ugur U. Psychiatric aspects of brain
tumors: a review. World J Psychiatry. 2015;5(3):273.
Makale MT, McDonald CR, Hattangadi-Gluth JA, Kesari S. Mechanisms of
radiotherapy-associated cognitive disability in patients with brain
tumours. Nat Rev Neurol. 2017;13(1):52–64.
National Institute for Clinical Excellence, Great Britain, and National Health
Service. Improving supportive and palliative care for adults with cancer:
the manual. London: National Institute for Clinical Excellence; 2004.
Ng JCH, See AAQ, Ang TY, Tan LYR, Ang BT, King NKK. Effects of surgery
on neurocognitive function in patients with glioma: a meta-analysis of
immediate post-operative and long-term follow-up neurocognitive out-
comes. J Neurooncol. 2019;141(1):167–82.
Parsons MW, Hammeke TA, editors. Clinical neuropsychology: a pocket
handbook for assessment. 3rd ed. American Psychological Association;
2014.
Robinson GA, Biggs V, Walker DG. Cognitive screening in brain tumors:
short but sensitive enough? Front Oncol. 2015;5:60.
Taphoorn MJB, Klein M. Cognitive deficits in adult patients with brain
tumours. Lancet Neurol. 2004;3(3):159–68.
van den Bent MJ, Wefel JS, Schiff D, Taphoorn MJB, Jaeckle K, Junck L,
Armstrong T, et al. Response assessment in neuro-oncology (a report of
the RANO group): assessment of outcome in trials of diffuse low-grade
gliomas. Lancet Oncol. 2011;12(6):583–93.
12 Management of Neurocognitive Symptoms 193
H. Ahmad (*)
Department of Neurology, University of Virginia, Charlottesville,
VA, USA
e-mail: hahmad@som.umaryland.edu
D. Schiff
Department of Neurology, University of Maryland, School of Medicine,
Baltimore, MD, USA
e-mail: s4jd@hscmail.mcc.virginia.edu
Temozolomide
selves have potential side effects. PJP prophylaxis choices are dis-
cussed in the following section. Thrombocytopenia with platelet
values less than 100,000 should delay the next cycle of therapy.
Exceptions can be made based on which way a platelet value is
trending. TMZ dose should be titrated down by 25–50 mg/m2 for
grade 3–4 neutropenia or thrombocytopenia. Specific manage-
ment of neutropenia, thrombocytopenia, and lymphopenia is also
discussed in the next section.
TMZ has no specific drug-drug interactions, though adverse
effects can be additive with other medications, such as thrombo-
cytopenia induced by certain AEDs.
TMZ is a teratogen; both men and women of child-bearing age
should be strongly advised to use birth control during the therapy.
TMZ can cause long term azoospermia and infertility. Patients
should consider spermatocyte banking or oocyte retrieval prior to
the initiation of treatment. It is critical that these topics be dis-
cussed with appropriate patients at the time of consent.
Carmustine (BCNU)
Bevacizumab
Irinotecan
Checkpoint Inhibitors
Nausea/Vomiting
Weight Loss
Fatigue
Peripheral Neuropathies
Conclusion
Vincristine Peripheral Constipation, ileus, None 1.4 mg/m2 Pregnancy Metabolized [3]
neuropathy skin necrosis significant IV, dose cap category D by and
(vesicant) of 2 mg weakly
induces
CYP3A4
enzyme
Lomustine Neutropenia, Pulmonary WBC 80–110 mg/ Pregnancy None [7]
lymphopenia, toxicity, renal 5–6 weeks, m2 each category significant
nausea, vomiting toxicity Platelets cycle. D. Affected male
4 weeks Reduce to fertility in
70% for preclinical
hematologic studies
toxicity
Carmustine Nausea, vomiting, Pulmonary WBC 200 mg/m2 Pregnancy None [6]
(intravenous) facial flushing, toxicity, hepatic 5–6 weeks, IV each category significant
neutropenia, toxicity Platelets cycle D. Affected male
lymphopenia, 4 weeks fertility in
thrombocytopenia preclinical
studies
Chemotherapy-Related Toxicities and Management
(continued)
207
208
References
1. Merck IWS, NJ, USA. Temodar (Temozolomide) [Package Insert]. US
Food and Drug Administration. Revised 2015.
2. Sigma-Tau Pharmaceuticals IG, MD. Matulane (Procarbazine) [Package
Insert]. US Food and Drug Administration. Revised 2008.
3. Hospira ILF, IL. Vincristine sulfate [Package Insert]. US Food and Drug
Administration. Revised 2013.
4. Jutras G, Belanger K, Letarte N, et al. Procarbazine, lomustine and vin-
cristine toxicity in low-grade gliomas. Curr Oncol. 2018;25(1):e33–9.
5. O’Driscoll BR, Hasleton PS, Taylor PM, Poulter LW, Gattameneni HR,
Woodcock AA. Active lung fibrosis up to 17 years after chemotherapy
with carmustine (BCNU) in childhood. N Engl J Med. 1990;323(6):378–
82.
6. Bristol-Myers Squibb Company P, NJ. BiCNU (Carmustine) [Package
Insert]. US Food and Drug Administration. Revised 2007.
7. NextSource Biotechnology M, FL. Gleostine (Lomustine) [Package
Insert]. US Food and Drug Administration. Revised 2014.
8. Genentech ISSF, CA. Avastin (Bevacizumab) [Package Insert]. US Food
and Drug Administration. Revised 2009.
9. Pfizer Injectables. NY N. Camptosar (Irinotecan) [Package Insert]. US
Food and Drug Administration. Revised 2014.
10. Smith EM, Pang H, Cirrincione C, et al. Effect of duloxetine on pain,
function, and quality of life among patients with chemotherapy-induced
painful peripheral neuropathy: a randomized clinical trial. JAMA.
2013;309(13):1359–67.
11. Momota H, Narita Y, Miyakita Y, Shibui S. Secondary hematological
malignancies associated with temozolomide in patients with glioma.
Neuro Oncol. 2013;15(10):1445–50.
Radiation Related Toxicities
and Management 14
Sara J. Hardy and Michael T. Milano
Acute side effects occur days to weeks after irradiation [4]. Aside
from fatigue, the majority of acute side effects from cranial radio-
therapy can be predicted from the dose distribution. Worsening
intracranial edema can occur in the high dose region; this can
cause headaches, nausea/vomiting, altered mental status, or wors-
ening of focal neurologic symptoms. Less frequent symptoms
include vertigo or seizures. Symptoms are typically transient and
managed with corticosteroids [5]. If the acoustic structures are in
the radiation field, patients can experience serous otitis media due
to eustachian tube dysfunction, occasionally requiring over-the-
counter decongestants. This will usually resolve several weeks
after completion of radiotherapy, but if persistent, can be treated
using myringotomy [6]. Alopecia can occur over the whole scalp
with WBRT or in patches with partial brain radiotherapy.
Permanent hair loss is more common with higher radiation doses
[7]. If the parotid gland is irradiated, patients can experience
parotid swelling, dry mouth, and tenderness consistent with par-
otitis [8].
Skin may be exposed to radiation incidentally during radio-
therapy of intracranial lesions, particularly superficial tumors,
14 Radiation Related Toxicities and Management 213
a b
c d
Fig. 14.1 MRI brain for patient with radiation necrosis after radiotherapy (a,
b) before and (c, d) after treatment with bevacizumab
216 S. J. Hardy and M. T. Milano
Pseudoprogression
Radiation can impact multiple structures within the eyes and optic
pathways including the optic nerves.
Optic Neuropathy
Ocular Neuromyotonia
Central Hypopituitarism
a b
Fig. 14.2 Patient with frontal cavernomas on imaging 11 years after treat-
ment with radiotherapy for a glioma shown on the (a) T1 + gadolinium
sequence (b) susceptibility weighted imaging sequence
222 S. J. Hardy and M. T. Milano
SMART Syndrome
to lens post
radiotherapy
Dry eyes 30 Gy to meibomian Eye exam n/a Years after Artificial tears,
gland, 50–60 Gy to radiotherapy anti-inflammatory
lacrimal gland eye drops
(continued)
227
Table 14.2 (continued)
228
Telangiectasias Children and adults MRI with T1, T2, Hypo- to isointense Years after Conservative
after radiotherapy susceptibility weighted on T1, iso- to radiotherapy management
imaging slightly
hyperintense on T2,
faint enhancement
on T1 postcontrast,
dark on SWI
Stroke-like Received a dose of MRI brain with Focal gyral 1–5 years Aspirin,
migraine attacks >50 Gy and received contrast thickening and after propranolol,
after radiation radiation to the gyriform contrast radiotherapy verapamil,
therapy (SMART) posterior fossa enhancement most anticonvulsants
syndrome commonly in the
parieto-occipital
region
Accelerated Child and adults, MRA, CTA, cerebral Focal stenosis, >5 years Statins, aspirin,
atherosclerosis particularly if angiography vessel antiplatelets
radiation dose to circle collateralization
Radiation Related Toxicities and Management
of Willis
Cognitive change Children and adults Metabolic work-up, n/a 4 months to Memantine,
who receive partial or vitamin B12, MRI brain, years after investigational
whole brain radiation neuropsychological radiotherapy strategies
therapy testing, depression/
anxiety screen
229
230 S. J. Hardy and M. T. Milano
Patient Advice
Whole brain radiation: This is typically given over
10 days (2 weeks). During the initial planning session, the
patient will be fitted for a mask to give the radiation treat-
ment safely. Patients will typically feel fine for the first
week, but will start to have some tiredness starting the sec-
ond week. This should resolve about 2 weeks after complet-
ing treatment. Less common symptoms include headache,
poor appetite, nausea, vomiting, and worsening of neuro-
logic symptoms. Most patients will have hair loss, but it will
typically grow back. Some patients will have hearing
changes due to an issue with the middle ear, which should
improve on its own. Studies show that many patients have
changes in memory months to years after completing radia-
tion treatment. Patients may receive a medication called
memantine and/or treated using a technique called hippo-
campal sparing to decrease these side effects.
Radiosurgery: Patients will receive radiation treatment
over 1–5 days. During the initial planning session, the
patient will be fitted for a tight mask to give radiotherapy
safely. Some patients will have mild tiredness the week
after completing this treatment. About one-third of patients
have immediate mild to moderate side effects after radiosur-
gery. They typically resolve on their own, but may be treated
with a short course of corticosteroids.
Partial brain radiotherapy for glioma: Most patients
are treated with 27–30 radiation treatments given over
5–6 weeks. It is possible that the radiation oncologist will
recommend a shorter treatment based on the specific case.
During the initial planning treatment, the patient will be fit-
ted for a mask to give the radiotherapy safely. The radiation
oncologist will coordinate with a neuro-oncologist (or med-
ical oncologist) so that chemotherapy will be started appro-
priately. Most patients will experience tiredness starting in
the second week of radiotherapy. This will gradually worsen
during treatment. There is usually patchy hair loss. Hair
14 Radiation Related Toxicities and Management 231
may not grow back even after treatment ends. Patients may
experience headaches, nausea, vomiting, seizures, or
changes in neurologic symptoms. Patients will start to feel
more normal about 2 weeks after radiotherapy ends and
should be mostly recovered by 4–6 weeks after the end of
treatment.
References
1. Baskar R, et al. Cancer and radiation therapy: current advances and future
directions. Int J Med Sci. 2012;9(3):193–9.
2. Mitin TM. Radiation therapy techniques in cancer treatment. UptoDate.
3. Marks LB, et al. Use of normal tissue complication probability models in
the clinic. Int J Radiat Oncol Biol Phys. 2010;76(3 Suppl):S10–9.
4. Marks PRLSCL. ALERT—adverse late effects of cancer treatment.
Springer. 2014.
5. Lawrence YR, et al. Radiation dose-volume effects in the brain. Int J
Radiat Oncol Biol Phys. 2010;76(3 Suppl):S20–7.
6. Jereczek-Fossa BA, et al. Radiotherapy-induced ear toxicity. Cancer
Treat Rev. 2003;29(5):417–30.
7. Lawenda BD, et al. Permanent alopecia after cranial irradiation: dose-
response relationship. Int J Radiat Oncol Biol Phys. 2004;60(3):879–87.
8. Cairncross JG, et al. Acute parotitis and hyperamylasemia following
whole-brain radiation therapy. Ann Neurol. 1980;7(4):385–7.
9. Kandil AO, et al. Multifocal Stevens-Johnson syndrome after concurrent
phenytoin and cranial and thoracic radiation treatment, a case report.
Radiat Oncol. 2010;5:49.
10. Anker CJ, et al. Avoiding severe toxicity from combined BRAF inhibitor
and radiation treatment: consensus guidelines from the Eastern
Cooperative Oncology Group (ECOG). Int J Radiat Oncol Biol Phys.
2016;95(2):632–46.
11. Meijer OW, et al. Single-fraction vs. fractionated linac-based stereotactic
radiosurgery for vestibular schwannoma: a single-institution study. Int J
Radiat Oncol Biol Phys. 2003;56(5):1390–6.
12. Han MS, et al. Hemifacial spasm caused by small epidermoid tumor, mis-
interpreted as delayed secondary hemifacial spasm caused by vestibular
schwannoma treated with gamma knife surgery. World Neurosurg.
2019;130:410–4.
13. Yang HC, et al. Gamma knife radiosurgery for larger-volume vestibular
schwannomas. Clinical article. J Neurosurg. 2011;114(3):801–7.
232 S. J. Hardy and M. T. Milano
14. Hayman JA, et al. Distribution of proliferating bone marrow in adult can-
cer patients determined using FLT-PET imaging. Int J Radiat Oncol Biol
Phys. 2011;79(3):847–52.
15. Petersson K, et al. Haematological toxicity in adult patients receiving cra-
niospinal irradiation—indication of a dose-bath effect. Radiother Oncol.
2014;111(1):47–51.
16. Werner-Wasik M, et al. Immediate side effects of stereotactic radiother-
apy and radiosurgery. Int J Radiat Oncol Biol Phys. 1999;43(2):299–304.
17. Kim JM, et al. The risk of radiation necrosis following stereotactic radio-
surgery with concurrent systemic therapies. J Neurooncol.
2017;133(2):357–68.
18. Minniti G, et al. Single-fraction versus multifraction (3 × 9 Gy) stereotac-
tic radiosurgery for large (>2 cm) brain metastases: a comparative analy-
sis of local control and risk of radiation-induced brain necrosis. Int J
Radiat Oncol Biol Phys. 2016;95(4):1142–8.
19. Lehrer EJ, et al. Treatment of brain metastases with stereotactic radiosur-
gery and immune checkpoint inhibitors: an international meta-analysis of
individual patient data. Radiother Oncol. 2019;130:104–12.
20. Shah R, et al. Radiation necrosis in the brain: imaging features and dif-
ferentiation from tumor recurrence. Radiographics. 2012;32(5):1343–59.
21. Levin VA, et al. Randomized double-blind placebo-controlled trial of
bevacizumab therapy for radiation necrosis of the central nervous system.
Int J Radiat Oncol Biol Phys. 2011;79(5):1487–95.
22. Xu Y, et al. Bevacizumab monotherapy reduces radiation-induced brain
necrosis in nasopharyngeal carcinoma patients: a randomized controlled
trial. Int J Radiat Oncol Biol Phys. 2018;101(5):1087–95.
23. Suh JH, et al. Current approaches to the management of brain metastases.
Nat Rev Clin Oncol. 2020;17:279–99.
24. Wen PY, et al. Updated response assessment criteria for high-grade glio-
mas: response assessment in neuro-oncology working group. J Clin
Oncol. 2010;28(11):1963–72.
25. Herrlinger U, et al. Lomustine-temozolomide combination therapy ver-
sus standard temozolomide therapy in patients with newly diagnosed
glioblastoma with methylated MGMT promoter (CeTeG/NOA-09): a
randomised, open-label, phase 3 trial. Lancet. 2019;393(10172):678–88.
26. Kelly PJ, et al. Unexpected late radiation neurotoxicity following bevaci-
zumab use: a case series. J Neurooncol. 2011;102(3):485–90.
27. Biousse VN. Nancy, neuro-ophthalmology illustrated. New York:
Thieme; 2009.
28. Roper-Hall G, Chung SM, Cruz OA. Ocular neuromyotonia: differential
diagnosis and treatment. Strabismus. 2013;21(2):131–6.
29. Sara Hardy IS, Milano M, Vates E, Constine L. Hypopituitarism. In:
PDB, Chang EL, Lo SS, Sahgal A, Suh JH, editor. Adult CNS radiation
oncology. Springer. 2018.
14 Radiation Related Toxicities and Management 233
30. Wahl M, et al. Relationship between radiation dose and microbleed for-
mation in patients with malignant glioma. Radiat Oncol. 2017;12(1):126.
31. Murphy ES, et al. Review of cranial radiotherapy-induced vasculopathy.
J Neurooncol. 2015;122(3):421–9.
32. Nordstrom M, et al. Large vessel arteriopathy after cranial radiation ther-
apy in pediatric brain tumor survivors. J Child Neurol. 2018;33(5):359–
66.
33. Bowers DC, et al. Late-occurring stroke among long-term survivors of
childhood leukemia and brain tumors: a report from the Childhood
Cancer Survivor Study. J Clin Oncol. 2006;24(33):5277–82.
34. El-Fayech C, et al. Cerebrovascular diseases in childhood cancer survi-
vors: role of the radiation dose to Willis circle arteries. Int J Radiat Oncol
Biol Phys. 2017;97(2):278–86.
35. Boulet J, et al. Statin use and risk of vascular events among cancer
patients after radiotherapy to the thorax, head, and neck. J Am Heart
Assoc. 2019;8(13):e005996.
36. Zheng Q, et al. Stroke-like migraine attacks after radiation therapy syn-
drome. Chin Med J (Engl). 2015;128(15):2097–101.
37. Makar GS, Siegal T, Syrow L. Stroke-like migraine attacks after radiation
therapy (SMART) syndrome: a rare, reversible complication of radiation
therapy. Headache. 2020;60:998–9.
38. Hardy SJ, et al. Cognitive changes in cancer survivors. Am Soc Clin
Oncol Educ Book. 2018;38:795–806.
39. DeAngelis LM, Delattre JY, Posner JB. Radiation-induced dementia in
patients cured of brain metastases. Neurology. 1989;39(6):789–96.
40. Chang EL, et al. Neurocognition in patients with brain metastases treated
with radiosurgery or radiosurgery plus whole-brain irradiation: a ran-
domised controlled trial. Lancet Oncol. 2009;10(11):1037–44.
41. Brown PD, et al. Hippocampal avoidance during whole-brain radiother-
apy plus memantine for patients with brain metastases: phase III trial
NRG oncology CC001. J Clin Oncol. 2020:Jco1902767.
42. Jalali R, et al. Efficacy of stereotactic conformal radiotherapy vs conven-
tional radiotherapy on benign and low-grade brain tumors: a randomized
clinical trial. JAMA Oncol. 2017;3(10):1368–76.
43. https://clinicaltrials.gov/ct2/show/NCT03180502
44. Brown PD, et al. Memantine for the prevention of cognitive dysfunction
in patients receiving whole-brain radiotherapy: a randomized, double-
blind, placebo-controlled trial. Neuro Oncol. 2013;15(10):1429–37.
45. Rapp SR, et al. Donepezil for irradiated brain tumor survivors: a phase III
randomized placebo-controlled clinical trial. J Clin Oncol.
2015;33(15):1653–9.
Neurosurgical
Complications in Brain 15
Tumor Patients
Tyler Schmidt
T. Schmidt (*)
University of Rochester, Rochester, NY, USA
e-mail: Tyler_Schmidt@URMC.Rochester.edu
Preoperative Management
Steroids
Hyperglycemia
Preoperative Imaging
Intraoperative Management
Patient Positioning
Closure
Hemorrhage
Seizures
Neurologic Deficit
Hydrocephalus
Infection
Venous Thromboembolism
References
1. Sawaya R, Hammoud M, Schoppa D, et al. Neurosurgical outcomes in a
modern series of 400 craniotomies for treatment of parenchymal tumors.
Neurosurgery. 1998;42(5):1044–55; discussion 1055–6. https://doi.
org/10.1097/00006123-199805000-00054.
2. Greenhalgh J, Weston J, Dundar Y, Nevitt SJ, Marson AG. Antiepileptic
drugs as prophylaxis for postcraniotomy seizures. Cochrane Database
Syst Rev. 2020;4(4):CD007286. https://doi.org/10.1002/14651858.
CD007286.pub5.
3. Chen CC, Rennert RC, Olson JJ. Congress of Neurological Surgeons sys-
tematic review and evidence-based guidelines on the role of prophylactic
anticonvulsants in the treatment of adults with metastatic brain tumors.
Neurosurgery. 2019;84(3):E195–7. https://doi.org/10.1093/neuros/
nyy545.
4. Hagan K, Bhavsar S, Arunkumar R, et al. Association between periopera-
tive hyperglycemia and survival in patients with glioblastoma. J
Neurosurg Anesthesiol. 2017;29(1):21–9. https://doi.org/10.1097/
ANA.0000000000000339.
5. Jenkinson MD, Barone DG, Bryant A, et al. Intraoperative imaging tech-
nology to maximise extent of resection for glioma. Cochrane Database
Syst Rev. 2018;1(1):CD012788. https://doi.org/10.1002/14651858.
CD012788.pub2.
6. Li P, Qian R, Niu C, Fu X. Impact of intraoperative MRI-guided resection
on resection and survival in patients with gliomas: a meta-analysis. Curr
Med Res Opin. 2017;33(4):621–30. https://doi.org/10.1080/03007995.20
16.1275935.
7. Caras A, Mugge L, Miller WK, Mansour TR, Schroeder J, Medhkour
A. Usefulness and impact of intraoperative imaging for glioma resection
on patient outcome and extent of resection: a systematic review and meta-
analysis. World Neurosurg. 2020;134:98–110. https://doi.org/10.1016/j.
wneu.2019.10.072.
8. Papadopoulos G, Kuhly P, Brock M, Rudolph KH, Link J, Eyrich
K. Venous and paradoxical air embolism in the sitting position. A pro-
spective study with transoesophageal echocardiography. Acta Neurochir.
1994;126(2–4):140–3. https://doi.org/10.1007/BF01476424.
9. Eyüpoglu IY, Hore N, Savaskan NE, et al. Improving the extent of malig-
nant glioma resection by dual intraoperative visualization approach.
PLoS One. 2012;7(9):e44885. https://doi.org/10.1371/journal.
pone.0044885.
15 Neurosurgical Complications in Brain Tumor Patients 247
A. Wasilewski (*)
Givens Brain Tumor Center, Allina Health, Minneapolis, MN, USA
e-mail: andrea.wasilewski@allina.com
GBM
23 AA
(continued)
251
Table 16.1 (continued)
252
Surgical Treatment
Radiotherapy
Adjuvant Therapy
Disease Recurrence
While all brain tumor patients are at risk of treatment- and tumor-
related complications, these issues are of particular importance
for older patients. This population is at increased risk of polyphar-
macy, adverse effects from supportive care medications, falls,
mood issues and sleep dysregulation which impact quality of life
and survival. Supportive care interventions should begin early in
the course of treatment and should be continually addressed and
reassessed.
258 A. Wasilewski
Corticosteroids
Tumor-Associated Epilepsy
Fall Prevention
Cognition Dysfunction
Mood
Polypharmacy
References
1. Yabroff KR, Lamont EB, Mariotto A, et al. Cost of care for elderly cancer
patients in the United States. J Natl Cancer Inst. 2008;100(9):630–41.
2. Iwamoto FM, et al. Patterns of care in elderly glioblastoma patients. Ann
Neurol. 2008;64(6):628–34.
3. Dressler EV, Liu M, Garcia CR, et al. Patterns and disparities of care in
glioblastoma. Neurooncol Pract. 2019;6(1):37–46.
4. Stupp R, et al. Radiotherapy plus concomitant and adjuvant temozolo-
mide for glioblastoma. N Engl J Med. 2005;352:987–96.
5. Stupp R, Wong E, Scott C, et al. Interim analysis of the EF-14 trial: a
prospective, multi-center trial of Novo-TTF-100A together with temo-
zolomide compared to temozolomide alone in newly diagnosed
GBM. Neuro Oncol. 2014;16:167.
6. Keime-Guibert F. Radiotherapy for glioblastoma in the elderly. N Engl J
Med. 2007;356(15):1527–35.
7. Roa W, Brasher PM, Bauman G, et al. Abbreviated course of radiation
therapy in older patients with glioblastoma multiforme: a prospective ran-
domized clinical trial. J Clin Oncol. 2004;22(9):1583–8.
8. Roa W, Kepka L, Kumar N, et al. International Atomic Energy Agency
randomized phase III study of radiation therapy in elderly and/or frail
patients with newly diagnosed glioblastoma multiforme. J Clin Oncol.
2015;33(35):4145–50.
9. Wick W, Platten M, Meisner C. Temozolomide chemotherapy alone ver-
sus radiotherapy alone from malignant astrocytoma in the elderly: the
NOA-08 randomised phase 3 trial. Lancet Oncol. 2012;13(7):707–15.
10. Perry JR, et al. Short-course radiation plus temozolomide in elderly
patients with glioblastoma. N Engl J Med. 2017;376(11):1027–37.
11. Malmstrom A, Gronberg BH, Marosi C, et al. Temozolomide versus stan-
dard 6-week radiotherapy versus hypofractionated radiotherapy in
patients older than 60 years with glioblastoma: the Nordic randomized,
phase 3 trial. Lancet Oncol. 2012;13(9):916–26.
12. Chaichana KI, Chaichana KK, Olivi A, et al. Surgical outcomes for older
patients with glioblastoma multiforme: preoperative factors associated
with decreased survival. Clinical article. J Neurosurg. 2011;114(3):587–
94.
13. Hanna C, Lawrie TA, Rogozinska E, et al. Treatment of newly diagnosed
glioblastoma in the elderly: a network meta-analysis. Cochrane Database
Syst Rev. 2020;3:CD013261.
14. Friedman HS, et al. Bevacizumab alone and in combination with irinote-
can. J Clin Oncol. 2009;27(28):4733–40.
15. Gilbert MR, et al. A randomized trial of bevacizumab for newly diag-
nosed glioblastoma. N Engl J Med. 2014;370(8):699–708.
16 Management of Older Patients with Brain Tumors 265
16. Stupp R, Wong ET, Kanner AA, et al. NovoTTF-100A versus physician’s
choice chemotherapy in recurrent glioblastoma: a randomised phase III
trial of a novel treatment modality. Eur J Cancer. 2012;48(14):2192–202.
17. Stark AM, Stepper W, Mehdorn HM. Outcome evaluation in glioblas-
toma patients using different ranking scores: KPS, GOC, mRS and
MRC. Eur J Cancer Care. 2010;19:39–44.
18. Roa AV, Hsieh F, et al. Geriatric evaluation and management units the
care of the frail elderly cancer patient. J Gerontol A Biol Sci Med Sci.
2005;60:798–803.
19. Rodin MB, Mohile SG. A practical approach to geriatric assessment in
oncology. J Clin Oncol. 2007;25:1936–44.
20. NCCN Clinical practice guidelines in oncology. n.d.. https://www.nccn.
org/professionals/physician_gls/default.aspx#age. Accessed 21 Mar
2020.
21. Wildiers H, Heeren P, Puts M, et al. International Society of Geriatric
Oncology consensus on geriatric assessment in older patients with can-
cer. J Clin Oncol. 2014;32:2595–603.
22. Puts MT, Hardt J, Monette J, et al. Use of geriatric assessment for older
adults in the oncology setting: a systematic review. J Natl Cancer Inst.
2012;104:1133–64.
23. Deluche E, Leobon S, Lamarche F, et al. First validation of the G-8 geri-
atric screening tool in older patients with glioblastoma. J Geriatr Oncol.
2019;10:159–63.
24. Wychowski T, Wang H, Buniak L, et al. Considerations in prophylaxis for
tumor-associated epilepsy: prevention of status epilepticus and tolerabil-
ity of newer generation of AEDs. Clin Neurol Neurosurg.
2013;115(11):2365–9.
25. Delaunois A, Colomar A, Depelchin BO, et al. Cardiac safety of lacos-
amide: the non-clinical perspective. Acta Neurol Scand. 2015;132(5):337–
45.
26. Wadley VG, Okonkwo O, et al. Mild cognitive impairment and everyday
function: evidence of reduced speed in performing instrumental activities
of daily living. Am J Geriatr Psychiatry. 2008;16:416–24.
27. Rooney AG, Carson A, Grant R. Depression in cerebral glioma patients:
a systematic review of observational studies. J Natl Cancer Inst.
2011;103(1):61–76.
28. Armstong TS, Gilbert MR. Practical strategies for management of fatigue
and sleep disorders in people with brain tumors. Neuro Oncol.
2012;13(Suppl 4):iv65–72.
29. Cramp F, Byron-Daniel J. Exercise for the management of cancer-
associated fatigue in adults. Cochrane Database Syst Rev.
2012;11:CD006145.
30. National Comprehensive Cancer Network. Cancer-related fatigue
(Version 2.2015). 2015. https://jnccn.org/abstract/journals/jnccn/13/8/
article-p1012.xml. Accessed 28 Mar 2020.
266 A. Wasilewski
31. American Geriatrics Society 2015 Beers Criteria Update Expert Panel.
American Geriatrics Society 2015 updated Beers criteria for potentially
inappropriate medication use in older adults. J Am Geriatr Soc.
2015;63(11):2227.
32. Maione P, Perrone F, Gallo C, et al. Pre-treatment quality of life and func-
tional status assessment significantly predict survival of elderly patients
with advanced non-small-cell lung cancer receiving chemotherapy: a
prognostic analysis of the multicenter Italian lung cancer in the elderly
study. J Clin Oncol. 2005;23:6865–72.
Palliative Care
in Neuro-oncology 17
Young-Bin Song and Lynne P. Taylor
Introduction
Palliative care is the active, total care of patients and families who
are dealing with serious, life-threatening, illness which is not
expected to be responsive to curative treatment. Palliate, derived
from the Latin root palliare, or “to cloak,” means to protect and
comfort with the goal of preserving the best possible quality of
life. Because the hospice movement began in the United States in
1985 with the establishment of the Medicare hospice benefit, and
the National Hospice Organization renamed as the National
Hospice and Palliative Care Organization (NHPCO) in 2000, the
average American has always assumed that palliative care and
hospice services are one and the same. It is our hope that palliative
care becomes a normative part of the early care of our patients
with primary brain tumors to allow adequate exploration of goals
of care, and to improve communication and maximize treatment
of all symptoms. The goal is to acknowledge dying as a natural
process and to provide compassionate care by relieving physical,
social, psychological, and spiritual suffering.
Communication Skills
Communication within a neuro-oncology practice presents
numerous challenges brought about by our patient’s symptoms
and varying degrees of caregiver support. Many of our patients
suffer from slowed cognitive processing, aphasia, and difficulties
with insight and judgment, and might not be fully able to partici-
pate in discussions about prognostication, goals of care, or
17 Palliative Care in Neuro-oncology 269
Responding to Emotions
The NURSE acronym is a tool that can be used to help us identify
emotions and respond in ways to help us understand the patient
perspective more completely. See Table 17.2.
Prognostication
Discussion of prognosis should begin by first asking permission
from the patient and family if they are ready to have the conversa-
tion while adjusting it in the moment to their emotional responses
[8]. When symptom progression leads to re-addressing treatment
plans or goals of care, prognostic discussions are vital. To help
decision making, if asked about the likelihood of overall survival,
instead of using a specific period, it is recommended to use terms
like “months to years,” “days to weeks,” or “hours to days” [8].
Cognitive Dysfunction
Cognitive dysfunction is common among brain tumor patients,
including deficits in memory, attention, and executive function-
ing, and can be caused by the tumor, seizures, and side effects of
treatment [8] as well as energy levels, sleep quality, and medica-
tion side effects.
Managing cognitive dysfunction can be difficult. Setting short
and long-term goals can be helpful for those who exhibit disorga-
nization; cognitive rehabilitation can improve visual attention and
verbal memory. Patients with attentional deficits might benefit
from pharmacologic interventions such as methylphenidate
(Ritalin) or modafinil (Provigil) though proof of efficacy in large
studies does not exist. See Table 17.3.
272 Y.-B. Song and L. P. Taylor
Fatigue
Fatigue occurs commonly in our brain tumor patients and is mul-
tifactorial in etiology. It affects more than 80% of patients who are
undergoing radiation therapy [12]. Fatigue can also result from
chemotherapy, both directly as a drug side effect, and indirectly
by anemia and metabolic deficiencies. Other contributing factors
include anxiety and depression, seizure medications, steroid use,
and decreased mobility.
Management strategies for fatigue should be specific and tar-
geted. Fatigue due to medications should lead to a discussion of
17 Palliative Care in Neuro-oncology 273
Insomnia
Insomnia is a common symptom among primary glioma patients.
In one study by Robertson et al. (2016) [17], 46.8% of 340 recur-
rent glioma patients had insomnia and 20% required the use of
sleep medications. Causes of insomnia are multifactorial, includ-
ing mood issues, disruption to circadian rhythm, and steroid use.
The same study [17] found that use of corticosteroids was associ-
ated with insomnia. Treatments for insomnia include sleep
hygiene education, evaluation for sleep apnea, and sleep aid med-
ications. Pharmacologic sleep aids include melatonin, trazodone,
and for those experiencing nighttime agitation or delirium, con-
sider atypical psychotics, such as quetiapine or risperidone
(Table 17.3).
Mood Issues
Mood issues are common in neuro-oncology patients and are
caused by many varied factors, including past psychiatric illness
and medication side effects (dexamethasone and antiepileptic
drugs (AEDs). Among patients with primary brain tumors, the
rates of depression can vary from 13 to 47% and rates of anxiety
from 35 to 48% [18]. These negatively affect our patient’s quality
of life through decreased emotional well-being, which should be
screened and addressed during clinic visits.
A meta-analysis [19] confirmed that physician-based assess-
ment tools are reliable and consistent for diagnosing depression in
a clinical setting. Different treatment options (see Table 17.4) for
depression include medication, counseling, and therapy. SSRIs
and SNRIs are generally well tolerated with minimal drug-to-
drug interactions. Gabapentin, lamotrigine and valproic acid can
all be used as mood stabilizers. Pseudobulbar affect (PBA) can be
a very difficult problem in patients with either deep bihemispheric
274 Y.-B. Song and L. P. Taylor
Headache
Headaches in brain tumor patients often present in a similar fash-
ion to migraines and will respond if treated as such, if not due to
increased intra-cranial pressure [20]. Otherwise, of course, ste-
roid therapy is the mainstay of treatment.
Steroid Use
Steroids are used to treat vasogenic edema and can rapidly allevi-
ate focal neurologic symptoms. Dexamethasone is the steroid of
choice, due to its long half-life and lack of mineralocorticoid
activity. It has many side effects, however, including insomnia,
irritability, psychosis, delirium, and anxiety, in addition to hyper-
glycemia and proximal muscle weakness. While dexamethasone
is given 3–4 times a day in the hospital, it should be given only
once or twice daily in the outpatient setting as it has an exception-
17 Palliative Care in Neuro-oncology 275
ally long serum half-life of 36–72 h, with the second dose early in
the afternoon to mitigate against insomnia. Look for opportunities
for dose tapering and give a clear schedule to patients and caregiv-
ers. See Table 17.5 for guidelines. Dexamethasone can usually be
tapered quickly down to 2 mg daily, followed by a slower tapering
plan.
Seizures: See Table 17.7. Most of our patients will have a well-
defined anti-convulsant regimen by the time they are near the end
of their lives. In an urgent situation, it is helpful to know that both
midazolam and diazepam can be given intra-nasally. Both now
have a pre-packaged device for administration but a small syringe,
fitted with a mucosal atomization device (MAD) is much less
expensive. One to two drops can also be dripped into the nasal
cavity with seizure cessation at 4 min [23].
276 Y.-B. Song and L. P. Taylor
Family/Caregivers
References
1. Taylor LP, Besbris JM, Graf WD, Rubin MA, Cruz-Flores S, Epstein LG,
et al. Clinical Guidance in Neuropalliative Care: An AAN Position
Statement. Neurology. 2022;98(10):409–16. https://doi.org/10.1212/
wnl.0000000000200063.
2. Faithfull S, Cook K, Lucas C. Palliative care of patients with a primary
malignant brain tumour: care review of service use and support provided.
Palliat Med. 2005;19(7):545–50. https://doi.org/10.1191/0269216305pm
1068oa.
3. McFarlin JM, Barclay JS. Communicating effectively. In: Creutzfeldt CJ,
Kluger BM, Holloway RG, editors. Neuropalliative care. A guide to
improving the lives of patients and families affected by neurologic dis-
ease. Cham: Springer; 2019. p. 151–60.
4. Gofton TE, Graber J, Carver A. Identifying the palliative care needs of
patients living with cerebral tumors and metastases: a retrospective analy-
sis. J Neurooncol. 2012;108(3):527–34. https://doi.org/10.1007/s11060-
012-0855-y. Epub 2012 Mar 31.
280 Y.-B. Song and L. P. Taylor
17. Robertson ME, McSherry F, Herndon JE, Peters KB. Insomnia and its
associations in patients with recurrent glial neoplasms. Springerplus.
2016;5(1):823. https://doi.org/10.1186/s40064-016-2578-6. eCollection
2016.
18. Ford E, Catt S, Chalmers A, Fallowfield L. Systematic review of support-
ive care needs in patients with primary malignant brain tumors. Neuro
Oncol. 2012;14(4):392–404. https://doi.org/10.1093/neuonc/nor229.
Epub 2012 Feb 3.
19. Huang J, Zeng C, Xiao J, Zhao D, Tang H, Wu H, et al. Association
between depression and brain tumor: a systematic review and meta-
analysis. Oncotarget. 2017;8(55):94932–43. https://doi.org/10.18632/
oncotarget.19843.
20. Nelson S, Taylor LP. Headaches in brain tumor patients: primary or sec-
ondary? Headache. 2014;54(4):776–85. https://doi.org/10.1111/
head.12326.
21. Shimmer BP, Funder JW. Adrenocorticotropic hormone, adrenal steroids,
and the adrenal cortex. In: Brunton LL, Knollmann BC, Hilal-Dandan R,
editors. Goodman & Gilman’s: the pharmacological basis of therapeutics,
13th ed. McGraw-Hill. 2017. https://accessmedicine-mhmedical-com.
offcampus.lib.washington.edu/content.aspx?bookid=2189§io
nid=172482605. Accessed 24 Jan 2021.
22. National POLST Paradigm Task Force. POLST legislative guide. 2014.
http://www.polst.org. Accessed 24 Jan 2021.
23. Mahmoudian T, Mohammadi Zadah M. Comparison of intranasal mid-
azolam with intravenous diazepam for treating acute seizures in children.
Epilepsy Behav. 2004;5:253–5.
24. Back AL, Arnold RM, Baile WF, Tulsky JA, Fryer-Edwards
K. Approaching difficult communication tasks in oncology. CA Cancer J
Clin. 2005;55(3):164–77. https://doi.org/10.3322/canjclin.55.3.164.
25. Sizoo EM, Braam L, Postma TJ, Pasman HRW, Heiman J, Klein M, et al.
Symptoms and problems in the end-of-life phase of high-grade glioma
patients. Neuro Oncol. 2010;12(11):1162–6. https://doi.org/10.1093/neu-
onc/nop045. Epub 2010 Jan 27.
26. Krouwer HG, Pallagi JL, Graves NM. Management of seizures in brain
tumor patients at the end of life. J Palliat Med. 2000;3(4):465–75. https://
doi.org/10.1089/jpm.2000.3.4.465.
27. Drumm MR, Dixit KS, Grimm S, Kumthekar P, Lukas R, Raizer JJ, et al.
Extensive brainstem infiltration, not mass effect, is a common feature of
end-stage cerebral glioblastomas. Neuro Oncol. 2020;22(4):470–9.
https://doi.org/10.1093/neuonc/noz216.
Index
A
Absolute neutrophil count (ANC), 197
Advance care planning, 262
5-Aminolevulinic acid (5-ALA), 6
Anaplastic oligodendrogliomas, 41
Antibiotic trimethoprim-sulfamethoxazole, 90
Anticoagulation medications, 237, 238
Anti-edema effects, 144
Anti-epileptic therapy, 12
Antiplatelet medications, 237, 238
Ask, Tell, Ask Method, 276
Aspartate aminotransferase (AST), 89
Asymptomatic meningiomas, 75
Atherosclerosis, and ischemic stroke, 222, 223
AVERT trial, 168
B
B-cells, 99
Bevacizumab, 200, 201
Bone marrow suppression, 204, 205
BRAFi/MEKi therapy, 55
BRAF-mutated glioma
clinical scenario, 51
clinical trials, 64
high-grade glioma, 55, 56
low grade glioma, 54, 55
mutations, 52, 53
prognosis/survivorship, 63
surveillance, 57, 60
targeted therapy and dosing, 56, 57
toxicities management, 60, 63
© The Editor(s) (if applicable) and The Author(s), under exclusive 283
license to Springer Nature Switzerland AG 2023
N. A. Mohile, A. A. Thomas (eds.), Brain Tumors,
https://doi.org/10.1007/978-3-031-41413-8
284 Index
Brain edema
alternative approaches to steroids, 150, 151
chronic steroid use, 144, 145
intracranial pressure management, 144
patient instructions, 151
preventative measures, 150
steroid toxicities, 145, 150
tapering, 144, 145
treatment, principles of, 141, 144
Brain tumor patients
antiplatelet and anticoagulation medications, 237, 238
closure, 242
gross total resection (GTR), 241
hydrocephalus, 244
hyperglycemia, 238
infections, 244
intraoperative neurophysiological monitoring, 242
neurologic deficit, 243, 244
optimizing other medical conditions, 239
other systemic complications, 245, 246
patient positioning, 240
perioperative antiepileptic therapy, 236, 237
postoperative intracranial hemorrhage, 242, 243
preoperative imaging, 239
seizures, 243
skin incision and tissue handling, 241
steroids, 237
venous air embolism, 240
venous thromboembolism (VTE), 245
wound healing complications, 245
Bruton tyrosine kinase (BTK), 97
C
Carmustine (BCNU), 200
Central hypopituitarism, 219, 221
Checkpoint inhibitors, 202
Chemo-induced side effects
cytopenia and bone marrow suppression, 204, 205
fatigue and malaise, 203
nausea/vomiting, 202, 203
peripheral neuropathies, 204
teratogenic effects and decreased fertility, 205
weight loss, 203
Index 285
D
Deep venous thrombosis (DVT), 166
Dexamethasone, 144
Diffuse astrocytoma (DA)
awake vs open craniotomy, 23
clinical features, 22
clinical vignette, 21, 22
fertility preservation (FP), 28
pathology, 24
patient information, 31, 35
286 Index
E
Ependymoma treatment
chemotherapy, 125, 127–129
clinical scenario, 119, 120
other modalities, 129
pathologic and molecular, 122, 124
patient information, 133–135
prognosis, 130, 132
proton therapy, 125
radiation therapy, 124, 125
recurrence, 130
RELA fusion position, 122
surgical resection, 121, 122
surveillance guidelines, 129, 130
survivorship, 130, 132
External beam radiotherapy (EBRT), 211
F
Fall prevention, 259
Fatigue
palliative care, 272, 273
and sleep disturbances, 260, 262
Fertility preservation (FP), 28
Fluorescence in situ hybridization (FISH), 41
Folinic acid, 90
Index 287
G
Gastritis, 150
Gene sequencing, 41
Glutamate pathways, 154
Granulocyte colony-stimulating factor (GCSF), 93, 197
Gross total resection (GTR), 75, 121, 241
H
Headache, palliative care, 274
High-dose myeloablative chemotherapy followed by autologous stem cell
transplantation (HDC-ASCT), 96
High-grade glioma (HGG), 55, 56
Hospice and end of life care, 277, 278
Hydrocephalus, 244
Hyperglycemia, 149, 238
Hypertension (HTN), 201
Hypofractionated radiotherapy, 8
I
IDH-wildtype, 24
Immunohistochemistry staining, 41
Insomnia, 149, 261
palliative care, 272, 273
Integrated histopathologic-molecular diagnosis, 41, 42
Irinotecan, 201, 202
Ischemic stroke, 222, 223
Isocitrate dehydrogenase (IDH), 154
IDH1, 22
mutation, 7
L
Leucovorin, 90, 99
Levetiracetam (LEV), 156
LMWH, 173
Lomustine (CCNU), 198, 199
Low grade glioma (LGG), 54, 55
M
Malignant glioma
adjuvant therapy, 254
advance care planning, 262
anti-epileptic therapy, 12
288 Index
N
Next generation sequencing (NGS), 73
Non-pharmacological interventions, cognitive symptoms
exercise, 185, 186
meditation/mindfulness stress reduction, 186
mood and behavior management, 186, 187
sleep and fatigue management, 187, 188
training and rehabilitation, 185
290 Index
O
Ocular neuromyotonia, 217, 218
Oligodendroglioma
chemotherapy, 43, 46–47
clinical scenario, 39, 40
integrated histopathologic-molecular diagnosis, 41, 42
patient information, 47
prognosis, 44, 45
radiation therapy, 42
surgery, role of, 40, 41
surveillance, 43, 44
treatment, 44
treatment flowchart, 46
Optic neuropathy, 217
Osteoporosis, 149
P
Palliative care, 268, 270
advanced care planning, 276, 277
cognitive dysfunction, 271
fatigue, 272, 273
headaches, 274
hospice and end of life care, 277, 278
insomnia, 272, 273
mood issues, 273, 274
need, 270, 271
steroids, 274, 275
Partial brain radiotherapy, 230
PCV (procarbazine, lomustine, and vincristine), 27, 28
Perioperative antiepileptic therapy, 236, 237
Pharmacologic interventions, cognitive symptoms
donepezil, 190
memanatine, 190, 191
methylphenidate, 188, 189
modafenil, 190
Polypharmacy, 262, 263
Positron emission tomography (PET) imaging, 86
Primary CNS lymphoma (PCNSL)
clinical scenario, 83, 84
common consolidation regimens, 96, 97
common induction regimens, 92
concomitant medications, 90
diagnosis of, 85
epidemiology, 84
high-dose methotrexate treatment, 87–88
Index 291
MATRix, 94
methotrexate therapy, 87, 89
methotrexate toxicity, 90, 91
MT-R, 93
patient information, 86, 98
relapsed disease, 97, 98
rituximab, 94, 95
R-MBVP, 94
R-MVP, 92, 93
site-specific therapy, 95
staging of, 86
supportive medication and monitoring, 89, 90
treatment expectations, 98
Procarbazine, 93, 198, 199
Proton Pump Inhibitors (PPIs), 150
Pseudoprogression, 216
Pulmonary embolism (PE), 166
R
Radiation-related cognitive decline, 224, 226–229
Radiation therapy (RT)
chemoradiotherapy, 26, 27
chemotherapy, 26, 27
doses, 26
timing of, 25, 26
toxicities, 26
Radionecrosis, 214, 216
Radiosurgery, 230
Relapsed disease, 97, 98
Rituximab, 94, 95
R-MBVP (rituximab, methotrexate, carmustine [BCNU], etoposide,
prednisone), 94
R-MVP (rituximab, methotrexate, vincristine, procarbazine), 92, 93
S
Sensorineural hearing loss, 218, 219
Serum methotrexate, 90
Signaling aberrations, 16
Simpson grade of resection, 76
Single nucleotide variants (SNVs), 52
Site-specific therapy, 95
Sleep dysregulation, 261
SMART syndrome, 223, 224
Sonic-hedgehog (SHH) pathway, 107
292 Index
T
Tapering protocol, 144
T-cells, 99
Temozolomide (TMZ), 8, 27, 196, 198
monotherapy, 8
Transcriptome profiling, 123
Tumor-associated epilepsy, 258, 259
Tumor-related epilepsy (TRE)
epidemiology, 153, 154
pathophysiology, 153, 154
pharmacologic management of, 156–157
status epilepticus (SE), 154, 155
Tumor treating fields (TTF) therapy, 9, 12–13
V
Valproic acid (VPA), 156
Vascular malformations, 221, 222
Vasogenic edema management, 13
Venous air embolism (VAE), 240
Venous thromboembolism (VTE), 245
anticoagulation, 173
and clinical diagnosis, 166, 168
contraindications to anticoagulation, 172
inpatient primary prevention, 169
medical risks, 172, 173
outpatient primary prevention, 168, 169
patient instructions, 174
prophylactic and therapeutic anticoagulant dosing, 170–171
treatment, 171, 173
Vincristine, 198, 199
Vision, blurring of, 149
W
Whole brain radiation therapy (WBRT), 211, 230