You are on page 1of 11

J C E M O N L I N E

Hot Topics in Translational Endocrinology—Endocrine Care

Temozolomide Treatment for Aggressive Pituitary


Tumors: Correlation of Clinical Outcome with
O6-Methylguanine Methyltransferase (MGMT)
Promoter Methylation and Expression

Zachary M. Bush, Janina A. Longtine, Tracy Cunningham, David Schiff,

Downloaded from https://academic.oup.com/jcem/article-abstract/95/11/E280/2835354 by guest on 16 June 2020


John A. Jane, Jr., Mary Lee Vance, Michael O. Thorner, Edward R. Laws, Jr.,
and M. Beatriz S. Lopes
Division of Endocrinology and Metabolism (Z.M.B., M.L.V., M.O.T.), University of Virginia, Charlottesville,
Virginia 22908; Department of Pathology (J.A.L.), Brigham and Women’s Hospital, Boston,
Massachusetts 02115; School of Medicine (T.C.), Division of Neurooncology (D.S.), and Department of
Neurosurgery (J.A.J.), University of Virginia, Charlottesville, Virginia 22908; Department of Neurosurgery
(E.R.L.), Brigham and Women’s Hospital, Boston, Massachusetts 02115; and Department of Pathology
(M.B.S.L.), Division of Neuropathology, University of Virginia, Charlottesville, Virginia 22908

Context: The typically indolent behavior of pituitary tumors is juxtaposed with high rates of tumor
cell invasion into adjacent dural structures, and occasional aggressive behavior. Although clinically
significant invasion and malignant transformation remain uncommon, there are limited treatment
options available for the management of these aggressive tumors. Recently, case reports have
described efficacy of temozolomide for the treatment of aggressive pituitary tumors.

Design: Seven patients with aggressive pituitary tumors have been treated with temozolomide. We
compared O6-methylguanine methyltransferase (MGMT) promoter methylation and MGMT ex-
pression in 14 surgical specimens from these seven patients and correlated these molecular features
with the clinical response to temozolomide.

Results: Significant tumor regression was seen in two patients (29%), a 20% reduction in tumor
volume with subsequent stable tumor size was noted in one patient, arrest of tumor growth
occurred in three patients, and progressive metastatic disease developed during treatment in one
patient. The DNA promoter site for MGMT was unmethylated in all 14 adequate specimens, and
variable MGMT expression was seen in all 14 cases. There was no correlation between MGMT
expression and clinical outcomes.

Conclusions: We conclude that medical therapy with temozolomide can be helpful in the man-
agement of life-threatening pituitary tumors that have failed to respond to conventional treat-
ments. The optimal duration of treatment in patients with stabilization or reduction of tumor size
has not been established, and long-term follow up studies are needed. (J Clin Endocrinol Metab 95:
E280 –E290, 2010)

he natural history of pituitary adenomas varies widely. morbidity and mortality are high when aggressive tumor
T The typically indolent growth rate of these tumors is
juxtaposed with high rates of tumor cell invasion into ad-
behavior occurs (2). With the exception of dopamine ag-
onist therapy for prolactinomas, surgery has long been the
jacent dural structures (1). Clinically significant invasion first line treatment for pituitary tumors. Options for med-
and malignant transformation remain uncommon, but ical treatment are limited, and none of the current thera-

ISSN Print 0021-972X ISSN Online 1945-7197 Abbreviations: CN, Cranial nerve; CSF, cerebrospinal fluid; GKR, GammaKnife stereotactic
Printed in U.S.A. radiosurgery; IHC, immunohistochemistry; MGMT, O6-methylguanine methyltransferase;
Copyright © 2010 by The Endocrine Society MRI, magnetic resonance imaging; MSP, methylation-specific PCR.
doi: 10.1210/jc.2010-0441 Received February 23, 2010. Accepted July 1, 2010.
First Published Online July 28, 2010

E280 jcem.endojournals.org J Clin Endocrinol Metab, November 2010, 95(11):E280 –E290


J Clin Endocrinol Metab, November 2010, 95(11):E280 –E290 jcem.endojournals.org E281

pies is tumoricidal (3– 6). When invasive tumors occur, by 0.5. The study was approved by the University of Virginia
repeat surgery and radiation therapy are second-line ther- Institutional Review Board.
The presence of tumor within selected specimen blocks and
apies that have variable efficacy depending on the char-
confirmation of the cell lineage of each tumor were made by
acteristics of the tumor. Extrasellar tumor extension, review of the standard histological, IHC, and ultrastructural
proximity to cranial nerves and critical blood vessels, and studies. MGMT methylation was established by the MSP. The
the delayed therapeutic effects of radiotherapy can all limit intrapatient stability of MGMT promoter methylation was de-
the efficacy of these treatment modalities (7, 8). These termined in surgical specimens obtained before and after radia-
therapeutic shortfalls are most critical in the management tion therapy in patients who underwent multiple operations dur-
ing the course of multimodality treatment; this includes pre- and
of highly invasive adenomas and pituitary carcinomas. postradiation specimens in case 3. IHC was used to determine
Therapeutic efforts have been made with conventional nuclear expression of the MGMT DNA repair enzyme.

Downloaded from https://academic.oup.com/jcem/article-abstract/95/11/E280/2835354 by guest on 16 June 2020


chemotherapeutic agents that cross the blood-brain bar-
rier to treat invasive and malignant pituitary tumors, but Methylation-specific PCR
efficacy has been poor, and partial responses are generally Genomic DNA was isolated from three to five 10-␮m paraffin
short-lived (9 –13). Temozolomide is an oral alkylating tissue sections containing at least 70% tumor (QIAamp DNA
agent that readily crosses the blood-brain barrier and has Mini Kit; QIAGEN, Valencia, CA). DNA methylation patterns
in the CpG island of the MGMT gene (GenBank accession num-
been shown to be efficacious in malignant gliomas. Several
ber AL355531, nucleotides 46931– 47011) was determined by
recent case reports have reported efficacy of temozolo- chemical (bisulfite) modification of unmethylated, but not meth-
mide for aggressive pituitary tumors (14 –18). The mech- ylated, cytosine to uracil (CpGenome Fast DNA Modification
anism by which temozolomide acts on pituitary tumors Kit; from Chemicon, now part of Millipore, Billerica, MA). The
has not been established, but in general, the alkylating bisulfite-treated DNA was precipitated, rehydrated, and ampli-
agents disrupt gene transcription by inducing DNA dam- fied by MSP using primers specific for either the methylated or
the modified unmethylated DNA (18). PCR amplification with
age by attaching a methyl group to the guanine base. Rap- Taq-Gold (Applied Biosystems, Foster City, CA) was performed,
idly dividing tumor cells often lack competent DNA repair and PCR products were analyzed in duplicate parallel runs by
mechanisms, and thus are more prone to cytotoxic DNA capillary gel electrophoresis (ABI 3130xl; Applied Biosystems)
disruption (19, 20). Epigenetic inactivation of the gene for with an expected product of 93 bp for methylated DNA and 80
the DNA repair enzyme O6-methylguanine methyltrans- bp for unmethylated DNA. Bisulfite-treated CPGenome Univer-
sal Unmethylated DNA (Millipore) and SssI methyltransferase
ferase (MGMT) via promoter hypermethylation has been
(New England Biolabs, Ipswich, MA)-treated human DNA were
show to predict therapeutic response to temozolomide in used as negative and positive methylation controls, respectively.
glioblastomas (21, 22), and retrospective analyses of pi- No template controls were performed. The sensitivity of the as-
tuitary tumors have reported MGMT methylation and ex- say based on DNA dilution studies is at least 1:1000. A dichot-
pression by immunohistochemistry (17, 23). omous result of methylated or unmethylated MGMT promoter
We report a series of seven patients with aggressive was reported for each case.
pituitary tumors that have been treated with temozolo-
mide. We compare MGMT promoter methylation status, Immunohistochemistry
IHC for MGMT was performed in formalin-fixed, paraffin-
as established by methylation-specific PCR (MSP), and embedded tissue using the avidin-biotin-peroxidase technique
nuclear MGMT expression by immunohistochemistry (24). Antigen retrieval by boiling in a citrate solution (Target
(IHC) and correlate these molecular features with the clin- Retrieval Solution, pH 6.1; Dako, Carpinteria, CA) was per-
ical response. formed using a pressure cooker (25). Tissue sections were incu-
bated with anti-MGMT monoclonal antibody (clone MT3.1;
1:50 dilution; Chemicon) using the automated staining sys-
tem (DakoCytomation Autostainer Plus; Dako, Glostrup, Den-
Patients and Methods mark). Positive and negative controls were run concomitantly in
From the records of the University of Virginia Pituitary Clinic, we all analyses. Tissue sections from human glioblastomas in which
identified all of the patients with invasive pituitary tumors who MGMT methylation status was known were used to establish
received temozolomide as adjuvant medical therapy. Medical positive IHC control. In addition, normal tissues adjacent to the
records were reviewed to establish duration of treatment, tumor adenoma specimens including blood vessels were used as positive
response, tolerability of temozolomide treatment, and adverse internal controls.
events. Tumor response was established by review of serial mag-
netic resonance imaging (MRI) studies, and in the case of hor- IHC scoring and statistical analysis
monally active tumors, the hormone response to treatment. The IHC specimens were analyzed independently by three review-
chemotherapeutic dose, the calendar regimen employed, and the ers using a semiquantitative method to determine both the pro-
number of cycles administered were established by review of portion of MGMT-positive cells and the intensity of the immu-
medical records. Tumor volume was calculated as elliptical vol- noreactivity. Reviewers were blinded to clinical outcome.
ume—the product of the largest diameter in three planes from Positivity was quantified as negative or 1 for tumors with less
coronal and sagittal postgadolinium T1 MRI images, multiplied than 10% of cells positive for MGMT; 2 for tumors with 10 –
E282
Bush et al.

TABLE 1. Temozolomide treatment cohort


Response
Case Tumor Radiation MGMT
no. Surgery Cell lineage features therapy score Temozolomide Biochemical Imaging Side effects Outcome
1 TSR: 10/08 Null-cell Ki-67 ⬍ 3%; None 3 10 of 12 cycles N/A 20% reduction in Fatigue, headaches Clinically stable
prominent completed tumor volume;
nucleoli subsequent
stable tumor
volume
2 TSR: 07/04; Corticotroph-cell Ki-67 ⫽ 18%; GKRT 10/18/07; 1 11 completed ACTH: 221 pg/ml ⬎80% tumor Sensory neural Clinically improved
craniectomies: with clinical p53 ⫹; 5/02/08 decreased to reduction hearing loss, during
07/07; 06/09 Cushing’s large, 18 ng/ml; fatigue, treatment
disease multinucleated adrenally headaches
cells insufficient at
cycle 8/11
3 TSR: 01/92; 06/ FSH/TSH/␣-SU Ki-67 ⬍ 3%; Conventional 2 13 completed, N/A Stable tumor size Fatigue and Clinically stable
03; 12/06 positive; no atypical fractionated still on tx; 12 headaches,
clinically features radiation cycles of 21/7, lessened with
Temozolomide for Aggressive Pituitary Tumors

nonfunctional 1992 now on 18/10 dose reduction


4 TSR: 07/94; 02/00 Null-cell Ki-67 ⫽ 6%; None 3 10/12 completed N/A Stable tumor size Fatigue, dry mouth Clinically stable
mitotic
figures
present
5 TSR: 08/06; 12/06 Prolactinoma Ki-67 ⬎ 20%; GKRT 02/2007 1 11 completed Prolactin: 5702 ⬎80% tumor Well tolerated Clinically improved
prominent still on tx 21/7 ng/ml reduction during
nucleoli decreased to treatment
121 ng/ml
6 TSR: 06/94; 10/ Pituitary Ki-67 ⬎ 20%; Conventional 3 2 cycles, Apr/ N/A Stable tumor size Fatigue, severe Deceased; cause of
01; 04/03 carcinoma; brisk fractionated May 2005 during 2 dizziness death unrelated
null-cell mitotic radiation temodar/ months of to pituitary
activity 1994; GKRT thalidomide treatment carcinoma
02/05
7 TSR: 01/05; 03/05 Pituitary Ki-67 ⬎ 20%; IMRT 04/05 1 7 cycles N/A Progressive Fatigue, headaches Deceased; death
carcinoma; rare mitotic metastatic attributed to
null-cell figures disease on complications of
treatment pituitary
carcinoma

TSR, Transsphenoidal surgery; GKRT, GammaKnife radiation treatment; IMRT, intensity modulated radiation therapy; N/A, not available; tx, treatment; ␣-SU, ␣-subunit of glycoproteins. Median
MGMT IHC score reported for each case (1 ⫽ ⬍10% MGMT positive cells; 2 ⫽ 10%–50% MGMT positive cells; 3 ⫽ ⬎50% MGMT positive cells).
J Clin Endocrinol Metab, November 2010, 95(11):E280 –E290

Downloaded from https://academic.oup.com/jcem/article-abstract/95/11/E280/2835354 by guest on 16 June 2020


J Clin Endocrinol Metab, November 2010, 95(11):E280 –E290 jcem.endojournals.org E283

Five of the seven patients were


treated with conventional radiation or
GammaKnife stereotactic radiosurgery
(GKR) before temozolomide treatment.
The interval between the last radia-
tion dose and temozolomide treatment
ranged from 5 months to 17 yr (Table
1). The duration of temozolomide treat-
ment ranged from 2 to 18 months. The
initial treatment regimen for all seven pa-

Downloaded from https://academic.oup.com/jcem/article-abstract/95/11/E280/2835354 by guest on 16 June 2020


tients was a 28-d cycle of temozolomide
75 mg/m2䡠d for 21 d followed by 7 d off
therapy. One patient had a dose adjust-
ment after 12 standard cycles to 75 mg/
m2䡠d for 18 d, 10 d off therapy in an effort
to reduce side effects of fatigue and head-
ache. One patient with a null cell tumor
received octreotide concomitant with
seven cycles of temozolomide; progres-
sive metastatic tumor burden in this pa-
tient resulted in progressive clinical de-
cline and death. One patient with an
undifferentiated pituitary carcinoma was
treated with thalidomide concomitant
FIG. 1. Serial T1, Postcontrast MRI images before and after temozolomide treatment. A and with temozolomide; the latter drug was
B, Case 2, an invasive corticotroph tumor pretreatment (A) and after 11 cycles (B) (arrows stopped after two cycles because of debil-
highlight tumor extension). C and D, Case 5, an invasive prolactinoma pretreatment (C) and
after 8 cycles (D) (arrows highlight substantial reduction of tumor). itating dizziness and headaches. There
was no evidence of tumor growth during
2 months of treatment.
50% positive cells; or 3 for tumors with more than 50% positive
cells. Intensity was scored on a scale where 1 ⫽ faint, 2 ⫽ mod-
erate, and 3 ⫽ distinct immunoreactivity equivalent to the in- Clinical response
tensity seen in internal controls. Marginals were not known to
the reviewers a priori; thus, interoperator variability was deter-
Tumor regression of more than 80% tumor volume
mined using Randolph’s multirater variation on Fleiss’ free-mar- resulted in rapid clinical improvement in two patients
ginal ␬ equation (26 –29). The free-marginal ␬ statistic for the (29%; Fig. 1), a 20% reduction in tumor size with subse-
semiquantitative IHC analysis was determined for interoperator quent stable tumor volume was noted in one patient, arrest
agreement for both the whole integer score (IHC positivity score of tumor growth was seen in three patients (Fig. 2), and
0 vs. 1 vs. 2, etc.) and the dichotomous outcome measure of
progressive metastatic disease while on temozolomide oc-
positive (scores of 1, 2, or 3) vs. negative.
curred in one patient with a null cell carcinoma that re-
sulted in death (Table 1).
In two patients, the aggressive pituitary tumor— one
Results
prolactinoma (case 5, Table 1) and one corticotroph
We identified seven cases of invasive pituitary tumors that tumor (case 2, Table 1)— had caused compression of
had been treated with temozolomide between 2005 and cranial nerves and the brain stem. Both tumors had
2009. All seven patients were initially treated with surgery. more than 80% reduction in tumor volume with chronic
Six of the seven patients had repeat surgery before receiving temozolomide therapy, and clinical improvement was
adjuvant treatment. A total of 19 operations were performed evident within the first four cycles; serial MRI studies
on the seven patients during multimodality management. We demonstrated marked reduction in tumor size, but after
eliminated redundant surgical specimens obtained in rapid 11 cycles of treatment some residual tumor remained in
sequence within the same treatment phase (i.e. before or after both cases (Fig. 1). Biochemically, the prolactin levels in
radiation). Fifteen specimens from the seven patients were case 5 decreased from 5702 ng/ml to 121 ng/ml over 11
analyzed by IHC and MSP. One specimen was too necrotic cycles, but have not fully normalized. Symptoms of cra-
to yield results in our molecular studies. nial nerve (CN) dysfunction (partial 6th CN palsy and
E284 Bush et al. Temozolomide for Aggressive Pituitary Tumors J Clin Endocrinol Metab, November 2010, 95(11):E280 –E290

Side effects and adverse events


Side effects were common in our cohort. Fatigue was
experienced by six patients and prompted dose reduction
after 12 cycles in one patient with a stable tumor burden
(case 3, Table 1). Case 2 experienced fatigue throughout
treatment, and once biochemical remission was achieved
temozolomide was discontinued to relieve the persistent
side effect, given the unknown benefit of continued ther-
apy. Headache occurred in four patients, but was not
dose-limiting. Severe dizziness occurred in one patient

Downloaded from https://academic.oup.com/jcem/article-abstract/95/11/E280/2835354 by guest on 16 June 2020


(case 6) who was on concomitant thalidomide; the temo-
zolomide was stopped after two cycles, and symptoms
were relieved (thalidomide is also know to cause dizziness
and may have contributed to the severity of this side ef-
fect). One adverse event occurred during the first four
cycles of temozolomide treatment in case 2; the patient
developed progressive bilateral sensorineural hearing loss,
which necessitated hearing aids that were unable to cor-
rect the sensory deficit. We have not seen hearing loss in
the hundreds of patients receiving temozolomide mono-
therapy for malignant gliomas at our center, but we cannot
FIG. 2. Serial MRI images from case 4. A–E, Progressive tumor growth
exclude temozolomide as the causative agent in this case.
in a patient with no history of radiation treatment. E, Immediately
before temozolomide; F, arrest of tumor growth during eight cycles of Despite the adverse event, the patient and treating physi-
temozolomide. cians elected to continue treatment because of clinical and
radiological evidence of tumor response. There has been
a painful 5th CN neuropathy) improved during the first
no improvement in the patient’s hearing loss over the 6
6 wk of treatment and resolved within four cycles of
months since temozolomide was stopped.
temozolomide. This patient received GKR treatment 26
months before temozolomide, with evidence of rapidly
progressive disease up to the point of initiating temo- MSP and IHC
zolomide treatment. We attribute her treatment re- The MGMT promoter was unmethylated in all 14 ad-
sponse to temozolomide. Case 2 had an ACTH-secret- equate specimens. The unmethylated state was stable over
ing macroadenoma with clinical Cushing’s disease. The time in case 3 in which both pre- and postradiation sur-
marked reduction in tumor volume led to a cerebrospi- gical specimens were available (Table 2). The IHC analysis
nal fluid (CSF) leak during cycle 11 of temozolomide (a demonstrated variable immunoreactivity for MGMT in a
phenomenon seen when prolactinomas rapidly respond nuclear pattern in all 14 adequate specimens (Table 2 and
to dopamine agonist therapy). The ACTH level pre- Fig. 3). Four specimens were scored as negative for
treatment was 221 pg/ml, which steadily declined dur- MGMT immunoreactivity by at least one reviewer, but
ing 13 cycles, ultimately normalizing at 18 pg/ml, with there was not a consensus for negative MGMT immuno-
clinical adrenal insufficiency developing near the end of reactivity in any of the 14 specimens (Table 2). Median
the treatment period. Tissue specimens obtained during MGMT scores for each case are reported in Table 1. Three
transsphenoidal surgery to repair the CSF leak revealed cases had less than 10% of cells immunoreactive for
histological evidence of invasive corticotroph tumor MGMT; one case had 10 –50% MGMT immunoreactive
cells in dural structures; nonetheless, the patient has cells; three cases had MGMT immunoreactivity in more
been off treatment for 6 months without signs or symp- than 50% of cells. Therefore, methylation status of the
toms of tumor regrowth or recurrence of Cushing’s dis- MGMT gene promoter was not predictive of relative
ease, and there has been no MRI evidence of growth in MGMT expression by IHC in any of the cases. Marked
the residual tumor. Importantly, this patient had a sec- heterogeneity in MGMT expression was noted within
ond GKR treatment 5 months before starting temozo- many of the tumors, some with expression predominantly
lomide, so it is possible that the treatment response in in single regions of the tumor, some with predominant
this patient is in part the result of radiosurgery (thera- expression in the periphery of the tumor. Likewise, there
peutic response in Cushing’s patients has been shown to was significant heterogeneity of the intensity of MGMT
be delayed 3– 48 months after GKR) (30). expression as demonstrated in Table 2 and Fig. 3.
TABLE 2. Fifteen surgical specimens from seven patients with invasive or metastatic pituitary tumors
MGMT IHC
PCR: MGMT promoter Reviewer 1 Reviewer 2 Reviewer 3
Patient MGMT MGMT MGMT
no. Specimen Methylated Unmethylated positive Intensity Total positive Intensity Total positive Intensity Total
1 A X 3 1 4 3 1 4 3 2 5
2 A X 1 2 3 Negative 0 0 1 1 2
J Clin Endocrinol Metab, November 2010, 95(11):E280 –E290

B X 1 1 2 2 3 5 Negative 0 0
3 A X 2 2 4 2 2 4 3 2 5
Ba X 2 2 4 2 1 3 1 1 2
Ca X 2 1 3 1 3 4 2 1 3
4 A X 3 2 5 3 1 4 3 2 5
B X 2 1 3 2 3 5 3 2 5
5 A X 1 1 2 Negative 0 0 1 1 2
B X 1 1 2 2 2 4 1 1 2
6 Aa X 3 1 4 3 1 4 3 2 5
Ba N/A; necrotic N/A; necrotic N/A N/A Necrotic N/A N/A Necrotic N/A N/A Necrotic
specimen specimen
Ca X 3 2 5 3 1 4 3 2 5
7 A X 1 1 2 1 2 3 Negative 0 0
B X 1 1 2 1 2 3 1 1 2
Methylation status of the MGMT promoter by MSP and MGMT immunoreactivity are shown. MGMT positive: 1 ⫽ ⬍10% of cells positive in tumor; 2 ⫽ 10%–50%; 3 ⫽ ⬎ 50%. MGMT intensity: 1,
faint; 2, moderate; 3, equivalent to positive controls. N/A, Not available.
a
Postradiation surgical specimen.
jcem.endojournals.org
E285

Downloaded from https://academic.oup.com/jcem/article-abstract/95/11/E280/2835354 by guest on 16 June 2020


E286 Bush et al. Temozolomide for Aggressive Pituitary Tumors J Clin Endocrinol Metab, November 2010, 95(11):E280 –E290

this tumor and thus predict temozo-


lomide responsiveness; however, the
patient developed spinal metastasis
and increasing parasellar tumor bur-
den during 7 months of temozolomide
treatment, which was discontinued 2
months before her death to alleviate the
side effects of severe fatigue and head-
aches. The two tumors with the most
significant response to temozolomide

Downloaded from https://academic.oup.com/jcem/article-abstract/95/11/E280/2835354 by guest on 16 June 2020


therapy were cases 2 and 5 described
in Clinical response, both of which
were locally invasive with encroach-
ment on cranial nerves, optic appara-
tus, and midbrain structures. Like
case 7, these tumors were MGMT
positive in less than 10% of cells.
Thus, clinical response was not reli-
ably predicted in our cohort by either
MGMT promoter methylation or
MGMT IHC immunoreactivity.

Discussion
To our knowledge, this is the largest
published cohort of patients treated
with temozolomide for aggressive pitu-
itary tumors (invasive adenomas and
carcinomas). The lack of clinical guide-
FIG. 3. IHC for MGMT. A and B, Diffuse pattern of stain (score 2); C, strong stain (score 3); lines for chemotherapy management of
D, example of heterogeneous stain in a single focus; E, internal positive control endothelial
cells; F, internal positive control sinus mucosa.
life-threatening pituitary tumors has
led to significant heterogeneity in the
clinical use of temozolomide for these
IHC statistical analysis rare tumors. The single-center aspect of this cohort has
The free-marginal ␬ statistic for the semiquantitative allowed for some uniformity in regard to the multimodal-
IHC analysis of MGMT positivity was 0.40, which sug- ity treatment algorithm and temozolomide dosing. None-
gests poor to moderate agreement by three reviewers on
theless, our data underscore the limitations of our current
the proportion of cells with MGMT immunoreactivity
understanding of aggressive pituitary tumors and the un-
within any given specimen (␬ ⬎ 0.70 indicates good in-
derlying molecular features that determine the heteroge-
teroperator reliability). With a dichotomous outcome
neity in tumor behavior and response to multimodality
measure of positive (scores of 1, 2, or 3) vs. negative, the
therapy. We believe that our data raise more questions
free-marginal ␬ score is 0.62, suggesting moderate inter-
than are answered, and we caution against any conclu-
operator agreement.
sions regarding what type of aggressive pituitary tumors
Correlation of clinical outcomes and molecular may or may not respond to temozolomide based on this
studies limited data set. Previously published case reports dem-
The MGMT promoter regions in the two postradiation onstrate at least a partial response to temozolomide in
surgical specimens from the most aggressive tumor (case 7, some aggressive tumors arising from all anterior pituitary
Table 1) in our cohort remained unmethylated, but IHC cell types including functioning and nonfunctioning pitu-
immunoreactivity for MGMT was less than 10% of cells itary adenomas and pituitary carcinomas.
in both specimens. The lack of MGMT expression would Most patients receiving treatment for aggressive pitu-
be expected to predict impaired DNA repair capacity in itary tumors in our center undergo at least two operations;
J Clin Endocrinol Metab, November 2010, 95(11):E280 –E290 jcem.endojournals.org E287

the second procedure is routinely undertaken to decom- contrast to other published findings (31, 32), our data
press compromised neural structures, reduce symptoms, suggest that MGMT promoter methylation and MGMT
or create adequate space from the optic chiasm to allow expression by IHC are not clinically useful in predicting
for focused radiation therapy. Radiation therapy with tumor response to temozolomide therapy. The intraop-
GammaKnife is routinely performed for treating residual erator heterogeneity in MGMT IHC scores in our series is
tumor seen either intraoperatively or by MRI after a sec- in line with recent large studies that have evaluated
ond surgical procedure. In those cases in which there is MGMT expression in gliomas; the intraoperator variabil-
tumor adjacent to optic nerves or other barriers to GKR ity seen across the various scoring methods used in these
treatment, conventional fractioned radiation therapy is studies reflects the significant intratumor variability of im-
recommended. In cases 1 and 4, the patients refused ra- munoreactivity patterns (multiple regions within a single

Downloaded from https://academic.oup.com/jcem/article-abstract/95/11/E280/2835354 by guest on 16 June 2020


diation. Temozolomide is rarely used to treat pituitary tumor can vastly differ in regard to the frequency and the
tumors at our center and is only used as last-line treatment relative intensity of the immunoreactivity) (33–35). Fur-
for life-threatening pituitary tumors that have been refrac- thermore, our MSP data suggest that MGMT promoter
tory to standard treatment modalities. We use the 21 d on, methylation is rare among invasive pituitary tumors and
7 d off temozolomide regimen—a “dose-dense” sched- does not explain the wide variability of IHC expression
ule—that delivers a higher dose of temozolomide per unit seen in our cohort of tumors.
time (75 mg delivered 21/28 d vs. 150 or 200 mg delivered Clonality studies employing X-chromosome inactiva-
5/28 d). There is a theoretical advantage to the dose-dense tion analyses have demonstrated that the majority of spo-
schedule in that it may deplete tumor cells of their MGMT radic human pituitary adenomas are monoclonal (36).
and thereby circumvent an important mechanism of re- This implies that these tumors arise from de novo somatic
sistance in tumors that lack promoter methylation. This genetic changes in a single pituitary cell. Our DNA meth-
regimen is associated with a greater risk of lymphopenia ylation data suggest that the heterogeneity of MGMT ex-
than standard 5/28-d regimens. However, the association pression within these monoclonal tumor cell populations
with higher risk of other side effects or secondary malig- may result not from epigenetic modification of the MGMT
nancies is unclear. promoter but instead from a unique transcriptional mi-
Our clinical results suggest that temozolomide induces croenvironment dictated by nuclear receptors and their
clinically significant tumor volume regression in a minor- corepressors and coactivators or at the level of mRNA
ity of invasive pituitary tumors [two of seven patients translation.
(29%)], but stabilization of tumor growth occurred in The alkylating action of temozolomide can cause epi-
four additional cases [four of seven patients (57%)]. In genetic modification of DNA by methylation of gene pro-

FIG. 4. Pre- and posttemozolomide treatment specimens from case 2. A and B, Surgical specimen before GammaKnife radiation and
temozolomide treatment showing extensive cellular atypia (A) and high Ki-67 labeling index (18%) (B). C–F, Surgical specimen after treatments
and collected during CSF leak correction showed focal islands of tumor cells entrapped on fibrous connective tissue with intense inflammatory
reaction (C). Extensive cellular pleomorphism was present (D), but adenoma cells were still immunoreactive for ACTH (E). Ki-67 labeling was
practically absent on tumor cells, but present in inflammatory cells seen on the specimen (F).
E288 Bush et al. Temozolomide for Aggressive Pituitary Tumors J Clin Endocrinol Metab, November 2010, 95(11):E280 –E290

moter sites, which disrupts protein synthesis. In vitro stud- 5 in our series, a prolactinoma, was one of the two tumors
ies suggest that temozolomide arrests tumor growth with significant tumor regression. There is a preponder-
through cell cycle arrest rather than direct cytotoxic mech- ance of prolactinomas among those tumors reported in the
anisms. As such, complete remission is not an expected literature to be temozolomide-responsive (14 –17, 31, 42).
outcome of temozolomide treatment; instead, growth ar- However, we believe this is indicative of the overrepre-
rest, diminished tumor volume, and biochemical control sentation of prolactinomas among functional tumors that
are the goals of temozolomide treatment. Tumor cells un- progress to a more malignant phenotype (13) rather
dergoing cell replication are most vulnerable to alkylating than a true molecular predisposition to temozolomide
agents. This was demonstrated in the recent publication responsiveness.
from Kovacs et al. (17) in which Ki-67 staining of post- Complete remission is not an expected outcome of te-

Downloaded from https://academic.oup.com/jcem/article-abstract/95/11/E280/2835354 by guest on 16 June 2020


temozolomide treatment specimen demonstrated a greater mozolomide therapy, but tumor stabilization or reduction
proportion of cells in G0 (fewer Ki-67-positive cells) than of tumor volume can improve clinical outcomes. Any ben-
the pretreatment specimen without any evidence of apo- efit of long-term therapy with a tumorstatic agent such as
ptosis or necrotic cell death. Our own pre- and posttemo- temozolomide is typically limited by the increased likeli-
zolomide surgical specimens from case 2 confirm these hood of adverse effects with cumulative doses. Cytopenias
findings, with Ki-67 labeling index in the pretreatment are the most likely to occur, but myelodysplasia syndrome
specimen being very high at 18% and subsequently unde- and secondary malignancies including myeloid leukemia
tectable in the posttreatment specimen (Fig. 4). A popu- have been observed with temozolomide (43). Thus, in our
lation of apparently viable corticotroph tumor cells is ev- opinion, temozolomide remains a last-line of defense for
ident in both pre- and posttemozolomide specimens. life-threatening pituitary tumors, and the duration of ther-
Radiation therapy is known to exert some antitumor apy must be determined on a patient-by-patient basis.
effects via DNA methylation (37, 38); however, the The lack of prospective clinical trial data for these rare
MGMT promoter in our five postradiation specimens re- aggressive tumors necessitates multidisciplinary patient
mained unmethylated (prior conventional radiation in care teams to provide patients with the most informed
two specimens from case 3, and prior conventional and treatment options and demands multicenter cooperation to
GKR treatment in three specimens from case 6, Table 2), facilitate the future clinical studies that are needed to estab-
and MGMT IHC does not suggest significant impact of lish treatment guidelines for aggressive pituitary tumors.
radiation on MGMT expression. This suggests that pre-
temozolomide radiation treatment may not have the same
therapeutic synergy with temozolomide that has been ob- Acknowledgments
served in glioblastomas (22, 39, 40). A recent publication
by Su et al. (41) that evaluated MGMT promoter meth- Address all correspondence and requests for reprints to: M. Beatriz
ylation in sputum samples of uranium miners showed in- S. Lopes, M.D., Ph.D., Division of Neuropathology, P.O. Box
creasing promoter methylation with cumulative low-dose 800214, Charlottesville, Virginia 22908-0214. E-mail: msl2e@
virginia.edu.
radiation exposure. Multicenter investigation will be nec-
Disclosure Summary: All authors have nothing to declare.
essary to establish the treatment effect of the combined
effects of radiation and temozolomide. Evaluation of dif-
ferential tumor response depending on interval between
radiation and temozolomide along with the study of the References
molecular effects of single-dose radiation treatment vs.
1. Meij BP, Lopes MB, Ellegala DB, Alden TD, Laws Jr ER 2002 The
chronic or intermittent low-dose radiation will be neces- long-term significance of microscopic dural invasion in 354 patients
sary to establish definitive treatment guidelines. with pituitary adenomas treated by transsphenoidal surgery. J Neu-
We conclude that adjuvant medical therapy with temo- rosurg 96:195–208
2. Pernicone PJ, Scheithauer BW, Sebo TJ, Kovacs KT, Horvath E,
zolomide may be considered for the management of life-
Young Jr WF, Lloyd RV, Davis DH, Guthrie BL, Schoene WC 1997
threatening invasive pituitary tumors that have failed to Pituitary carcinoma: a clinicopathologic study of 15 cases. Cancer
respond to conventional treatments. Based on our cumu- 79:804 – 812
3. Melmed S, Sternberg R, Cook D, Klibanski A, Chanson P, Bonert V,
lative experience and previous reported data, we do not
Vance ML, Rhew D, Kleinberg D, Barkan A 2005 A critical analysis
believe that there are any molecular or clinical features of of tumor shrinkage during primary medical therapy in acromegaly.
these tumors that adequately predict tumor response; J Clin Endocrinol Metab 90:4405– 4410
therefore, we do not recommend that tumor cell lineage or 4. Buchfelder M, Weigel D, Droste M, Mann K, Saller B, Brübach K,
Stalla GK, Bidlingmaier M, Strasburger CJ 2009 Investigators of
MGMT molecular analysis influence the clinical decision German Pegvisomant Observational Study. Pituitary tumor size in
for treatment with temozolomide in individual cases. Case acromegaly during pegvisomant treatment: experience from MR
J Clin Endocrinol Metab, November 2010, 95(11):E280 –E290 jcem.endojournals.org E289

re-evaluations of the German Pegvisomant Observational Study. 22. Hegi ME, Diserens AC, Gorlia T, Hamou MF, de Tribolet N, Weller
Eur J Endocrinol 161:27–35 M, Kros JM, Hainfellner JA, Mason W, Mariani L, Bromberg JE,
5. Batista DL, Zhang X, Gejman R, Ansell PJ, Zhou Y, Johnson SA, Hau P, Mirimanoff RO, Cairncross JG, Janzer RC, Stupp R 2005
Swearingen B, Hedley-Whyte ET, Stratakis CA, Klibanski A 2006 MGMT gene silencing and benefit from temozolomide in glioblas-
The effects of SOM230 on cell proliferation and adrenocortico- toma. N Engl J Med 352:997–1003
tropin secretion in human corticotroph pituitary adenomas. J Clin 23. Widhalm G, Wolfsberger S, Preusser M, Woehrer A, Kotter MR,
Endocrinol Metab 91:4482– 4488 Czech T, Marosi C, Knosp E 2009 O(6)-methylguanine DNA meth-
6. Boscaro M, Ludlam WH, Atkinson B, Glusman JE, Petersenn S, yltransferase immunoexpression in nonfunctioning pituitary ade-
Reincke M, Snyder P, Tabarin A, Biller BM, Findling J, Melmed S, nomas: are progressive tumors potential candidates for temozolo-
Darby CH, Hu K, Wang Y, Freda PU, Grossman AB, Frohman LA, mide treatment? Cancer 115:1070 –1080
Bertherat J 2009 Treatment of pituitary-dependent Cushing’s dis- 24. Hsu SM, Raine L, Fanger H 1981 The use of antiavidin antibody and
ease with the multireceptor ligand somatostatin analog pasireotide avidin-biotin-peroxidase complex in immunoperoxidase tech-
(SOM230): a multicenter, phase II trial. J Clin Endocrinol Metab niques. Am J Clin Pathol 75:816 – 821

Downloaded from https://academic.oup.com/jcem/article-abstract/95/11/E280/2835354 by guest on 16 June 2020


94:115–122 25. Norton AJ, Jordan S, Yeomans P 1994 Brief, high-temperature heat
7. Yildiz F, Zorlu F, Erba° T, Atahan L 1999 Radiotherapy in the man- denaturation (pressure cooking): a simple and effective method of an-
agement of giant pituitary adenomas. Radiother Oncol 52:233–237 tigen retrieval for routinely processed tissues. J Pathol 173:371–379
8. Knosp E, Steiner E, Kitz K, Matula C 1993 Pituitary Adenomas with 26. Brennan RL, Prediger DJ 1981 Coefficient kappa: Some uses, mis-
invasion of the cavernous sinus space: a magnetic resonance imaging uses, and alternatives. Educ Psychol Meas 41:687– 699
classification compared with surgical findings. Neurosurgery 33: 27. Fleiss JL 1971 Measuring nominal scale agreement among many
610 – 618 raters. Psychol Bull 76:378 –382
9. Petterson T, MacFarlane IA, MacKenzie JM, Shaw MD 1992 Pro- 28. Randolph JJ Free-marginal multirater ␬: an alternative to Fleiss’
lactin secreting pituitary carcinoma. J Neurol Neurosurg Psychiatry fixed-marginal multirater ␬. Proc of Joensuu University Learning
55:1205–1206 and Instruction Symposium, Joensuu, Finland, 2005
10. Vaughan NJ, Laroche CM, Goodman I, Davies MJ, Jenkins JS 1985 29. Jackson TJ, Michel JL, Roberts R, Shepheard J, Cheng D, Rust J,
Pituitary Cushing’s disease arising from a previously nonfunctional Perry C 2009 Development of a validation algorithm for ‘present on
corticotrophic chromophobe adenoma. Clin Endocrinol (Oxf) 22: admission’ flagging. BMC Med Inform Decis Mak 9:48
147–153 30. Sheehan JM, Vance ML, Sheehan JP, Ellegala DB, Laws Jr ER 2000
11. Mixson AJ, Friedman TC, Katz DA, Feuerstein IM, Taubenberger Radiosurgery for Cushing’s disease after failed transsphenoidal sur-
JK, Colandrea JM, Doppman JL, Oldfield EH, Weintraub BD 1993 gery. J Neurosurg 93:738 –742
Thyrotropin-secreting pituitary carcinoma. J Clin Endocrinol 31. Hagen C, Schroeder HD, Hansen S, Hagen C, Andersen M 2009
Metab 76:529 –533 Temozolomide treatment of a pituitary carcinoma and two pituitary
12. Kasperlik-Zaluska AA, Wislawski J, Kaniewska J, Zborzil J, macroadenomas resistant to conventional therapy. Eur J Endocrinol
Frankiewicz E, Zgliczyñski S 1987 Cytostatics for acromegaly. 161:631– 637
Marked improvement in a patient with an invasive pituitary tu- 32. Mohammed S, Cusimano MD, Scheithauer BW, Rotondo F,
mour. Acta Endocrinol 116:347–349 Horvath E, Kovacs K 2010 O-methylguanine-DNA methyltrans-
13. Lopes MB, Scheithauer BW, Schiff D 2005 Pituitary carcinoma: ferase immunoexpression in a double pituitary adenoma: case re-
diagnosis and treatment. Endocrine 28:115–121 port. Neurosurgery 66:E421–E422; discussion E422
14. Neff LM, Weil M, Cole A, Hedges TR, Shucart W, Lawrence D, Zhu 33. Christmann M, Nagel G, Horn S, Krahn U, Wiewrodt D, Sommer
JJ, Tischler AS, Lechan RM 2007 Temozolomide in the treatment of C, Kaina B 3 February 2010 MGMT activity, promoter methylation
an invasive prolactinoma resistant to dopamine agonists. Pituitary and immunohistochemistry of pre-treatment and recurrent malig-
10:81– 86 nant gliomas: a comparative study on astrocytoma and glioblas-
15. Fadul CE, Kominsky AL, Meyer LP, Kingman LS, Kinlaw WB, toma. Int J Cancer doi: 10.1002/ijc.25229
Rhodes CH, Eskey CJ, Simmons NE 2006 Long-term response of 34. Preusser M, Charles Janzer R, Felsberg J, Reifenberger G, Hamou
pituitary carcinoma to temozolomide. J Neurosurg 105:621– 626 MF, Diserens AC, Stupp R, Gorlia T, Marosi C, Heinzl H,
16. Lim S, Shahinian H, Maya MM, Yong W, Heaney AP 2006 Temo- Hainfellner JA, Hegi M 2008 Anti-O6-methylguanine-methyltrans-
zolomide: a novel treatment for pituitary carcinoma. Lancet Oncol ferase (MGMT) immunohistochemistry in glioblastoma multi-
7:518 –520 forme: observer variability and lack of association with patient
17. Kovacs K, Horvath E, Syro LV, Uribe H, Penagos LC, Ortiz LD, survival impede its use as clinical biomarker. Brain Pathol 18:
Fadul CE 2007 Temozolomide therapy in a man with an aggressive 520 –532
prolactin-secreting pituitary neoplasm: morphological findings. 35. Mellai M, Caldera V, Annovazzi L, Chiò A, Lanotte M, Cassoni P,
Hum Pathol 38:185–189 Finocchiaro G, Schiffer D 2009 MGMT promoter hypermethyl-
18. Thearle MS, Freda PU, Bruce JN, Isaacson SR, Lee Y, Fine RL 4 ation in a series of 104 glioblastomas. Cancer Genomics Proteomics
December 2009 Temozolomide and capecitabine treatment of an 6:219 –227
aggressive corticotroph pituitary tumor. Pituitary doi:10.1007/ 36. Clayton RN, Farrell WE 2004 Pituitary tumour clonality revisited.
s11102-009-0211-1 Front Horm Res 32:186 –204
19. Roos WP, Batista LF, Naumann SC, Wick W, Weller M, Menck CF, 37. Koturbash I, Pogribny I, Kovalchuk O 2005 Stable loss of global
Kaina B 2007 Apoptosis in malignant glioma cells triggered by the DNA methylation in the radiation-target tissue: a possible mecha-
temozolomide-induced DNA lesion O6-methylguanine. Oncogene nism contributing to radiation carcinogenesis. Biochem Biophys Res
26:186 –197 Commun 337:526 –533
20. Esteller M, Hamilton SR, Burger PC, Baylin SB, Herman JG 1999 38. Kaup S, Grandjean V, Mukherjee R, Kapoor A, Keyes E, Seymour
Inactivation of the DNA repair gene O6-methylguanine-DNA meth- CB, Mothersill CE, Schofield PN 2006 Radiation-induced genomic
yltransferase by promoter hypermethylation is a common event in instability is associated with DNA methylation changes in cultured
primary human neoplasia. Cancer Res 59:793–797 human keratinocytes. Mutat Res 597:87–97
21. Hegi ME, Diserens AC, Godard S, Dietrich PY, Regli L, Ostermann 39. Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B,
S, Otten P, Van Melle G, de Tribolet N, Stupp R 2004 Clinical trial Taphoorn MJ, Belanger K, Brandes AA, Marosi C, Bogdahn U,
substantiates the predictive value of O-6-methylguanine-DNA Curschmann J, Janzer RC, Ludwin SK, Gorlia T, Allgeier A,
methyltransferase promoter methylation in glioblastoma patients Lacombe D, Cairncross JG, Eisenhauer E, Mirimanoff RO 2005
treated with temozolomide. Clin Cancer Res 10:1871–1874 European Organisation for Research and Treatment of Cancer
E290 Bush et al. Temozolomide for Aggressive Pituitary Tumors J Clin Endocrinol Metab, November 2010, 95(11):E280 –E290

Brain Tumor and Radiotherapy Groups; National Cancer Institute promoter methylation of P16-INK4a and O6-methylguanine-DNA
of Canada Clinical Trials Group. Radiotherapy plus concomitant methyltransferase in workers at a Chinese uranium mine. J Occup
and adjuvant temozolomide for glioblastoma. N Engl J Med 352: Health 48:261–266
987–996 42. McCormack AI, McDonald KL, Gill AJ, Clark SJ, Burt MG, Campbell
40. Stupp R, Dietrich PY, Ostermann Kraljevic S, Pica A, Maillard I, KA, Braund WJ, Little NS, Cook RJ, Grossman AB, Robinson BG,
Maeder P, Meuli R, Janzer R, Pizzolato G, Miralbell R, Porchet F, Clifton-Bligh RJ 2009 Low O6-methylguanine-DNA methyltrans-
Regli L, de Tribolet N, Mirimanoff RO, Leyvraz S 2002 Promising ferase (MGMT) expression and response to temozolomide in aggres-
survival for patients with newly diagnosed glioblastoma multiforme sive pituitary tumours. Clin Endocrinol (Oxf) 71:226 –233
treated with concomitant radiation plus temozolomide followed by 43. Su YW, Chang MC, Chiang MF, Hsieh RK 2005 Treatment-related
adjuvant temozolomide. J Clin Oncol 20:1375–1382 myelodysplastic syndrome after temozolomide for recurrent high-
41. Su S, Jin Y, Zhang W, Yang L, Shen Y, Cao Y, Tong J 2006 Aberrant grade glioma. J Neurooncol 71:315–318

Downloaded from https://academic.oup.com/jcem/article-abstract/95/11/E280/2835354 by guest on 16 June 2020

Sign up for eTOC alerts today


to get the latest articles as soon as they are online.
http://endo.endojournals.org/subscriptions/etoc.shtml

You might also like