ArticlePDF Available

Gefitinib response of erlotinib-refractory lung cancer involving meninges - Role of EGFR mutation

Authors:

Abstract

A 70-year-old Japanese-American woman who had never smoked was diagnosed with stage IV non-small-cell lung cancer with rib metastases. She had previously been well and she had no family history of malignancy. While receiving treatment with erlotinib, an epidermal growth factor receptor small-molecule inhibitor, she progressed and developed new brain metastases. She failed further chemotherapy treatments and subsequently developed extensive symptomatic leptomeningeal carcinomatosis associated with diplopia, hemiparesis, weight loss, and incontinence. Chest X-ray, head and chest CT scan, R2 lymph-node biopsy, histopathology, immunohistochemistry, MRI of head and spine, lumbar puncture, laser microdissection and EGFR genomic DNA sequencing of the R2 lymph node and cerebrospinal fluid tumor cells. Erlotinib-refractory stage IV lung adenocarcinoma and end-stage symptomatic leptomeningeal metastases with a novel double L858R + E884K somatic mutation of the EGFR. Carboplatin, paclitaxel and erlotinib, whole-brain radiotherapy, temozolomide with and without irinotecan, and gefitinib.
CASE STUDY
www.nature.com/clinicalpractice/onc
Gefitinib response of erlotinib-refractory lung
cancer involving meninges—role of EGFR mutation
Nicholas W Choong, Sascha Dietrich, Tanguy Y Seiwert, Maria S Tretiakova, Vidya Nallasura,
Gareth C Davies, Stanley Lipkowitz, Aliya N Husain, Ravi Salgia and Patrick C Ma*
This article offers the opportunity to earn one
Category 1 credit toward the AMA Physician’s
Recognition Award.
THE CASE
A 70-year-old Japanese-American woman,
who had never smoked, was diagnosed with
T2N2M1 stage IV non-small-cell lung cancer
(NSCLC) with rib metastases in April 2002.
She had initially presented to her primary-
care physician in December 2001 with a cough
of several months’ duration. She lost 3.6 kg
over 6 months and weighed 41 kg at presenta-
tion. She was otherwise healthy with no family
history of malignancy. A chest X-ray performed
in February 2002 showed a right hilar mass.
In March 2002, a chest CT confirmed a right
middle lobe mass, measuring 5 × 6 cm, and
enlarged hilar and mediastinal lymph nodes. A
bone scan 8 days later showed multiple rib meta-
static foci. The patient underwent bronchoscopy
and mediastinoscopy in April 2002, and biop-
sies of two involved R2 lymph nodes confirmed
adenocarcinoma (Figure 1A). Histopathologic
and immunohistochemical examination of
this biopsy specimen revealed strong positive
staining for cytokeratin 7 and thyroid tran-
scription factor-1 (data not shown). Epidermal
growth factor receptor (EGFR) expression
was strongly positive and predominantly
membranous (Figure 1A, inset).
The patient was immediately referred to the
chest oncology clinic of a large cancer center in
the US midwest. Her physical examination and
laboratory findings were unremarkable. She
was enrolled in a phase III clinical trial and
received first-line treatment with carboplatin
(area under the curve 6) and paclitaxel
(200 mg/ m2 every 3 weeks). She was also rand-
omized to receive concurrent erlotinib (150 mg
daily).1 She completed six cycles of this therapy
and attained a maximal tumor reduction of
42%. After 11 months of single-agent erlotinib
and disease stabilization, however, she
developed ataxia and leg weakness. An
Background A 70-year-old Japanese–American woman who had never smoked
was diagnosed with stage IV non-small-cell lung cancer with rib metastases. She
had previously been well and she had no family history of malignancy. While
receiving treatment with erlotinib, an epidermal growth factor receptor small-
molecule inhibitor, she progressed and developed new brain metastases. She
failed further chemotherapy treatments and subsequently developed extensive
symptomatic leptomeningeal carcinomatosis associated with diplopia,
hemiparesis, weight loss, and incontinence.
Investigations Chest X-ray, head and chest CT scan, R2 lymph-node
biopsy, histopathology, immunohistochemistry, MRI of head and
spine, lumbar puncture, laser microdissection and EGFR genomic DNA
sequencing of the R2 lymph node and cerebrospinal fluid tumor cells.
Diagnosis Erlotinib-refractory stage IV lung adenocarcinoma and
end-stage symptomatic leptomeningeal metastases with a novel double
L858R + E884K somatic mutation of the EGFR.
Management Carboplatin, paclitaxel and erlotinib, whole-brain
radiotherapy, temozolomide with and without irinotecan, and gefitinib.
KEYWORDS epidermal growth factor receptor, leptomeningeal
carcinomatosis, lung cancer, mutation, small-molecule inhibitor
CME
NW Choong and TY Seiwert are Fellows in the Section of Hematology–Oncology,
MS Tretiakova is a research pathologist in the Department of Pathology,
V Nallasura is a senior research assistant in the Aerodigestive Tract Translational
Research Laboratories, AN Husain is Professor of Pathology, and R Salgia is
Associate Professor of Medicine and Director of the Thoracic Oncology Research
Program at the Pritzker School of Medicine, all at the University of Chicago,
Chicago, IL, USA. S Dietrich is a final year medical student and PhD candidate
from the University of Jena, Germany, and an elective research fellow in the
Thoracic Oncology Research program at the University of Chicago, Chicago, IL,
USA. GC Davies is a visiting Fellow, and S Lipkowitz is an investigator in the
Laboratory of Cellular and Molecular Biology at the National Cancer Institute,
Bethesda, MD, USA. PC Ma is Assistant Professor of Medicine and clinician–
scientist in the Thoracic Oncology Program, Case Western Reserve University and
Case Comprehensive Cancer Center, Cleveland, OH, USA.
Correspondence
*Division of Hematology–Oncology, Case School of Medicine, Case Western Reserve University,
University Hospitals of Cleveland and Ireland Cancer Center, Case Comprehensive Cancer Center,
Wolstein Research Building WRB 2-123, 10900 Euclid Avenue, Cleveland, OH 44106, USA
patrick.ma@case.edu
Both authors contributed equally to this work
Received 23 October 2005 Accepted 16 November 2005
www.nature.com/clinicalpractice
doi:10.1038/ncponc0400
SUMMARY
50 NATURE CLINICAL PRACTICE ONCOLOGY JANUARY 2006 VOL 3 NO 1
ncponc_2005_288.indd 50ncponc_2005_288.indd 50 13/12/05 1:21:27 pm13/12/05 1:21:27 pm
Nature Publishing Group
©2006
CASE STUDY
JANUARY 2006 VOL 3 NO 1 CHOONG ET AL. NATURE CLINICAL PRACTICE ONCOLOGY 51
www.nature.com/clinicalpractice/onc
investigative head CT scan showed new
multiple brain metastases in her right cere-
bellum and left parietal lobe. Erlotinib was
stopped, and in November 2003 the patient
underwent standard whole-brain radiation
therapy. Thereafter, she was enrolled in a phase
II trial of combination oral temozolomide
(75 mg/m2 daily on days 1–15) and intravenous
irinotecan (100 mg/m2 on days 8 and 15 in a
21-day cycle); this therapy was not tolerated,
and was dis continued after one cycle because
of severe diarrhea and dehydration. The patient
then remained clinically stable, off chemo-
therapy, until August 2004 when she developed
new-onset hemiparesis associated with radic-
ular pain in her left leg, left-gaze diplopia, and
incontinence of the bowel and bladder. She
became wheelchair-bound as a result of
I II III
TT
F
Forward Forward
Reverse
Involved R2 lymph node
G L858R A K
TTGGGCTGGCCAAAC TGGCAT TGGAATC
A L E884K S I
AA T T
A
B
C
TT
F
Metastatic CSF tumor cells
G L858R A K
TTGGGCTGGCCAAAC TGGCAT TGGAATC
A L E884K S I
AA T T
TT
F
Wild-type EGFR (CSF leukocytes)
G L858 A K
TTGGGCTGGCCAAAC TGGCAT TGGAATC
A L E884 S I
AA T T
Forward Forward
Forward Forward
Reverse
Reverse Reverse
Reverse Reverse
Figure 1 Lung adenocarcinoma tumor cells and EGFR somatic mutations. (A) Hematoxylin and eosin staining of lung
adenocarcinoma in the involved mediastinal R2 lymph node at time of diagnosis, demonstrating the typical acinar pattern of
glandular differentiation (upper panel). Intense EGFR staining of the adenocarcinoma tumor cells (inset). Laser microdissection of the
tumor cells in the lymph node is shown (lower panels) with digital micrographs taken before (I, left), during (II, middle) and after (III,
right) the UV laser firing. Magnification ×200. (B) Cerebrospinal fluid (CSF) with non-small-cell lung cancer tumor cell (black arrow)
and normal leukocyte (arrow outline) identified. Magnification ×400. (C) Chromatogram of the EGFR DNA sequencing, showing
missense mutations L858R (CTGCGG, heterozygous) in exon 21 and E884K (GAAAAA, heterozygous) in exon 22 seen in the
involved R2 lymph node (top panel) and also in the metastatic CSF tumor cells (middle panel). Wild-type EGFR sequence seen in
normal leukocytes isolated from CSF (bottom panel) is shown as a control, and confirms the somatic nature of L858R and E884K
mutations. Presence of the two mutations was confirmed in both tumor samples by reamplification and sequencing in both sense
(Forward) and antisense (Reverse) directions.
ncponc_2005_288.indd 51ncponc_2005_288.indd 51 13/12/05 1:21:31 pm13/12/05 1:21:31 pm
Nature Publishing Group
©2006
CASE STUDY
52 NATURE CLINICAL PRACTICE ONCOLOGY CHOONG ET AL. JANUARY 2006 VOL 3 NO 1
www.nature.com/clinicalpractice/onc
weakness in her left leg (quadriceps strength
grade 1/5). CT scan showed that her brain
metastases and pulmonary disease (Figure 2A)
burdens were unchanged, with no mass effect
and little interval progression since her last
completed therapies. Brain MRI in August 2004
showed postradiation therapy changes in the
peri ventricular white matter. There were subtle
areas of leptomeningeal enhancement in the
posterior fossa suggestive of leptomeningeal
carcinomatosis. Spine MRI ruled out the possi-
bility of spinal-cord compression, but revealed
extensive spinal leptomeningeal metastases
with multiple well-demarcated intrathecal
isointense masses that demonstrated intense
enhancement with contrast administration,
consistent with drop metastases. The disease
was predominantly localized to the lumbar
region surrounding the cauda equina, and was
shown to have worsened on the MRI in October
2004 (Figure 3A,B), despite two cycles of
single-agent oral temozolomide (150 mg/m2 on
days 1–5 in a 28-day cycle). The patient had
declined lumbar puncture and intrathecal
chemotherapy.
Gefitinib (250 mg daily) was initiated in
October 2004, and the patient experienced
significant symptom improvement within
3 weeks. She started gaining weight, the pain in
her leg improved, she was able to move her left
leg and her diplopia also improved. The patient
then received follow-up consultations in the
clinic every 1–2 months. After 6 weeks on gefit-
inib, she was able to ambulate with walker assist-
ance, her diplopia and incontinence completely
resolved, and muscle strength in her left quad-
riceps improved to grade 4/5. Brain and spine
MRI showed a dramatic decrease in meningeal
ABC
Figure 2 Response to gefitinib in the primary tumor and thoracic involved lymph nodes. CT scan of the
chest (A) prior to gefitinib, (B) 2 months after starting gefitinib, and (C) 4.5 months after starting gefitinib,
demonstrating reduction in the size of the involved lymph node (white arrow) and the right middle lobe
tumor (arrow outline).
ABCD
Figure 3 Response to gefitinib in extensive leptomeningeal carcinomatosis. MRI scan of the lower
thoracic and lumbar spine (A,B) prior to starting gefitinib, and (C,D) 2 months after gefitinib monotherapy,
demonstrating a dramatic reduction in the number and enhancement of leptomeningeal metastases (solid
white arrows), correlating with the positive clinical response.
ncponc_2005_288.indd 52ncponc_2005_288.indd 52 13/12/05 1:21:33 pm13/12/05 1:21:33 pm
Nature Publishing Group
©2006
CASE STUDY
JANUARY 2006 VOL 3 NO 1 CHOONG ET AL. NATURE CLINICAL PRACTICE ONCOLOGY 53
www.nature.com/clinicalpractice/onc
enhancement and nodular tumor mass burden
(Figure 3C,D) after only 6 weeks of gefitinib
therapy. Analysis of cerebrospinal fluid (CSF)
confirmed the presence of residual metastatic
NSCLC cells (Table 1; Figure 1B). Her right
middle lobe mass had decreased from 2.2 cm to
1.5 cm in the short axis over the first 2 months,
and it continued to respond on subsequent
evaluations. Hilar and media stinal lymph nodes
showed a similar response (Figure 2A–C).
At a follow-up visit 4 months after starting
gefitinib, the patient’s neuro logical exami-
nation showed further improvement, and
she was ambulating independently with a
walker. Six months after starting gefitinib,
her ECOG (Eastern Cooperative Oncology
Group) performance status improved from 4
to 2. No significant adverse effects—specifically
diarrhea, rash, or interstitial pneumonitis—
were reported. Unfortunately, 8 months after
initiating gefitinib, the patient had a witnessed
food aspiration with resultant acute respiratory
distress. She died of aspiration pneumonia in
June 2005, 7 days after hospital admission.
DISCUSSION OF DIAGNOSIS
Multifocal seeding of the arachnoid membrane
and pia mater by metastatic tumor cells is known
as leptomeningeal carcinomatosis. Brain paren-
chymal metastases occur in 30% of NSCLC
patients, while leptomeningeal carcinomatosis
occurs in only 5%.2 Leptomeningeal carcinoma-
tosis carries a devastating prognosis and often
represents a terminal event,3 with a median
survival of 1.8 months in NSCLC, even with
active treatment.4 As improved therapies have
created a trend towards prolonged survival, the
incidence of this advanced complication is likely
to increase. Leptomeningeal carcinomatosis
commonly presents with cranial nerve palsies,
mental changes, stroke-like symptoms, cerebellar
signs, and, less commonly, autonomic distur-
bances. The condition can be readily diagnosed
by MRI and lumbar puncture with CSF analysis.
Radiographic studies may show subarachnoid
masses, diffuse meningeal contrast enhance-
ment, or hydrocephalus without a mass lesion.2
The presence of malignant cells in the CSF is
considered the gold standard in the diagnosis
of leptomeningeal carcinomatosis.3 Mutations
within the EGFR tyrosine kinase domain,
particularly L858R (exon 21) and exon 19 short
in-frame deletions, have been associated with the
response to EGFR-targeted therapy (gefitinib and
erlotinib).5–7 Therefore, in this case, genomic
analysis of EGFR was performed on both the
original pretreatment tumor biopsy and also on
tumor cells found in the CSF.
DIFFERENTIAL DIAGNOSIS
The differential diagnosis for leptomeningeal
carcinomatosis includes various chronic states
of meningitis and acute spinal-cord compres-
sion. Careful CSF examination and MRI should
be performed to distinguish these conditions.
TREATMENT AND MANAGEMENT
The standard treatment for leptomeningeal
carcinomatosis is intrathecal chemotherapy
(methotrexate, liposomal cytarabine, and thio-
peta) and neuroaxis radiation therapy. The effi-
cacy of systemic chemotherapy is usually limited
because tumor cells in the CSF are protected by
the blood–brain barrier. Radiotherapy is indi-
cated for bulky disease, as intrathecal chemo-
therapy can be limited by diffusion to 2–3 mm
penetration into tumor nodules. A recent study
favors liposomal cytarabine over methotrexate as
the drug of choice.3 Moreover, potential impair-
ment of CSF circulation in patients might result
in uneven distribution of intra thecal chemo-
therapeutics within the sub arachnoid space.
Aggressive approaches such as those described
carry significant toxicity,3 and result in reversal
or palliation of neurological symptoms in only
a minority of patients.4 The patient in this case
study was previously treated with erlotinib
in combination with carboplatin/paclitaxel
(TRIBUTE trial)1 with subsequent progressive
central nervous system (CNS) metastases when
receiving erlotinib. The TRIBUTE trial did not
show any statistically significant survival benefit
of adding erlotinib to standard chemotherapy.
Table 1 Laboratory analysis of the patient’s cerebrospinal fluid.
Test Result
Glucose (normal range: 2.2–4.4 mM 3.2 mM
Protein (normal range: 0.15–0.45 g/l) 0.57 g/l
Lactate dehydrogenase
(normal range: 0–26 U/l)
46 U/l
Red blood cell count (normally, there
are no red blood cells in the CSF)
12,017/μl
White cell count (normal range: 0–5/μl) 33/μl (neutrophils 58%, lymphocytes
26%, monocytes 9%, tumor cells 4%)
Cytology Positive (non-small-cell carcinoma)
ncponc_2005_288.indd 53ncponc_2005_288.indd 53 13/12/05 1:21:34 pm13/12/05 1:21:34 pm
Nature Publishing Group
©2006
CASE STUDY
54 NATURE CLINICAL PRACTICE ONCOLOGY CHOONG ET AL. JANUARY 2006 VOL 3 NO 1
www.nature.com/clinicalpractice/onc
The clinical response resulting from use of the
alternative anti-EGFR quinazoline inhibitors
in the setting of progressive CNS disease has
not been previously described. In this case, the
role of gefitinib at the time of the patient’s lepto-
meningeal disease progression was unclear, in
light of her previous erlotinib failure. As a result
of the minimal adverse effect profile of the EGFR
tyrosine kinase inhibitor (TKI), gefitinib was
initiated instead of hospice referral, after fully
informed discussion of the limited treatment
options. A prompt and striking response was
Erlotinib inhibition Gefitinib inhibition
IB: p-EGFR [Y1068] IB: p-EGFR [Y1068]
ErbB1
ErbB2
ErbB3
ErbB1: VP IKWMALES I LHR IYTHQSDVWSY
ErbB2: VP IKWMALES I LRRRFTHQSDVWSY
ErbB3: VP IKWMALES I HFGKYTHQSDVWSY
E884K (GAA>AAA)
Amino acid alignment
CLUSTAL W (1.82) mulitiple sequence alignment
GTGCCTATCAAGTGGATGGCATTGGAATCAATTTTACACAGAATCTATACCCACCAGAGT 60
GTGCCCATCAAGTGGATGGCGCTGGAGTCCATTCTCCGCCGGCGGTTCACCCACCAGAGT 60
ACTCCAATTAAGTGGATGGCCCTTGAGAGTATCCACTTTGGGAAATACACACACCAGAGT 60
** ** *********** * ** ** * * ** *********
ErbB1
ErbB2
ErbB3
GATGTCTGGAGCTACG 76
GATGTGTGGAGTTATG 76
GATGTCTGGAGCTATG 76
***** ***** ** *
IB: β-actin
U 0.01 0.1 1 5 U 0.01 0.1 1 5 U 0.01 0.1 1 5 U 0.01 0.1 1 5 μM U 0.01 0.1 1 5 U 0.01 0.1 1 5 U 0.01 0.1 1 5 U 0.01 0.1 1 5 μM
IB: β-actin
WT L858R E884K L858R + E884K WT L858R E884K L858R + E884K
A
CD
B
80
60
40
20
0
Erlotinib Gefitinib
Percent phospho-EGFR level of untreated control
L858R
L858R + E884K
Figure 4 EGFR mutations and differential effects on the sensitivity of the receptor towards inhibition by erlotinib and gefitinib. COS-
7 cells transiently transfected with EGFR plasmid constructs (wild-type, L858R, E884K, and L858R + E884K [created by site-directed
mutagenesis as previously described16]) were treated with increasing concentrations of either (A) erlotinib or (B) gefitinib, in the
presence of EGF (100 ng/ml, 30 min). Whole cell lysates were collected after EGF stimulation, separated on 7.5% SDS-PAGE and
electrotransferred onto nitrocellulose membrane for immunoblotting. The membrane was probed with an antibody against phospho-
EGFR [pY1068] (upper panel) and β-actin (lower panel) as a loading control. Equivalent receptor expression was also confirmed
with an anti-EGFR immunoblot (see Supplementary Figure 1 online). Two separate immunoblotting experiments were used for
quantitative analysis of the phospho-EGFR signal intensity. U, cells untreated with TKI; WT, wild-type. (C) Percentage (%) phospho-
EGFR signal level compared with the untreated control (100%) plotted for EGFRL858R-COS-7and EGFRL858R + E884K-COS-7 cell lines
treated with either of the two EGFR TKIs at 0.1 μM. The EGF-stimulated phospho-EGFR activation of L858R mutation is modulated
by the E884K mutation differentially with erlotinib (resistant) and gefitinib (sensitizing) as shown here at the in vitro concentration
of 0.1 μM. The percentage phospho-EGFR signal level (%) relative to untreated control (100%) was shown for both the L858R and
L858R + E884K mutations to illustrate how E884K differentially modulates the sensitizing effect of L858R on the receptor towards the
two different inhibitors. (D) Multiple sequence alignment performed by the CLUSTAL W alignment program for exon 22 of ErbB1, 2,
and 3 of the ErbB receptor family. The black arrow shows the conserved nucleotide G that was mutated in our patient’s tumor EGFR
gene and the arrow outline and bold text shows the conserved amino acid residue glutamic acid, E884. The methods are further
described in the Supplementary methods section; please go to the article online for details. *Conserved nucleotides among the
three sequences; IB, immunoblot.
ncponc_2005_288.indd 54ncponc_2005_288.indd 54 13/12/05 1:21:35 pm13/12/05 1:21:35 pm
Nature Publishing Group
©2006
CASE STUDY
JANUARY 2006 VOL 3 NO 1 CHOONG ET AL. NATURE CLINICAL PRACTICE ONCOLOGY 55
www.nature.com/clinicalpractice/onc
seen with gefitinib, resulting in a significant
reduction in the burden of leptomeningeal
carcinomatosis with corresponding improve-
ment in quality of life and performance status.
Interestingly, this response was not associated
with any toxicity. Notably, the patient was not
receiving any medications that might change
the serum levels of the EGFR inhibitors, such as
CYP3A4 inducers or inhibitors. Although studies
have reported good and prolonged responses
of brain metastases to gefitinib,8–10 successful
treatment of leptomeningeal metastasis has not
been reported. Furthermore, in patients who
have a response to gefitinib or erlotinib in their
primary tumor, brain and leptomeninges are
often sites of treatment failure.11 This report is
the first, to the authors’ knowledge, to describe
the response of lepto meningeal carcinoma-
tosis and extracranial disease to gefitinib after
erlotinib failure.
In the era of molecular-targeted therapy,
treatment options are often dependent on the
molecular and genetic profile of the tumor cells.
In this case, approximately 300 tumor cells from
the R2 lymph node were laser micro dissected
for genomic DNA extraction (Figure 1A,
I–III) and sequenced for the entire EGFR gene
(exons 1–28). Two EGFR missense mutations
within the tyrosine kinase domain were identi-
fied. Both mutations were present in the initial
pretreatment R2 lymph-node biopsy and in the
malignant cells from the CSF (four tumor cells
isolated by laser microdissection) (Figure 1C).
The first somatic mutation was L858R (exon
21, heterozygous), where leucine was substi-
tuted by arginine. The second mutation was a
novel E884K mutation (exon 22, heterozygous),
where glutamic acid was substituted by lysine.
The wild-type EGFR sequence was identified
in the patient’s CSF leukocytes (Figure 1B,C),
thereby confirming the somatic nature of the
mutations. We also identified two single nucle-
otide polymorphisms, Q787Q and T903T (data
not shown).
WT L858R T790M E884K L858R+
E884K
S S++ R+++ R++ R+
S
Erlotinib
Gefitinib S++ R+++ S++
Class I Class IIIClass II
S+++
EGFR
WT
EGFR
L858R
EGFR
T790M
EGFR
E884K
EGFR
L858R+E884K
Figure 5 Summary of EGFR mutations and their effects on sensitivity and resistance towards inhibition
by EGFR small molecule inhibitors. The known somatic missense mutations of EGFR and their effects on
sensitivity and resistance towards erlotinib or gefitinib inhibition are shown schematically. L858R sensitizes
the receptor to both inhibitors to a similar extent, and more than the wild-type receptor. T790M is the
recently reported resistant mutation against both EGFR inhibitors. E884K gives the receptor a sensitizing
effect towards gefitinib, while rendering it more resistant to erlotinib. The double mutation L858R + E884K
also sensitizes the receptor to gefitinib inhibition, with a synergistic effect when compared with E884K
alone, and confers resistance to erlotinib. Of note, the most sensitizing mutation towards gefitinib inhibition
is EGFRL858R + E884K, identified in this patient. Hence, the effects of EGFR mutations on TKIs can be divided
into: Class I, sensitizing responses; Class II, resistant responses; and Class III, differential responses.
S, sensitive; R, resistant.
ncponc_2005_288.indd 55ncponc_2005_288.indd 55 13/12/05 1:21:35 pm13/12/05 1:21:35 pm
Nature Publishing Group
©2006
CASE STUDY
56 NATURE CLINICAL PRACTICE ONCOLOGY CHOONG ET AL. JANUARY 2006 VOL 3 NO 1
www.nature.com/clinicalpractice/onc
In order to further delineate the correla-
tion of EGFR mutational status and inhibitor
responsiveness, we performed in vitro trans-
fection studies in COS-7 cells using expres-
sion vectors containing wild-type (EGFRWT),
EGFRL858R, EGFRE884K, and EGFRL858R + E884K
constructs (Figure 4A,B). The L858R muta-
tion sensitized the receptor to both erlotinib
and gefitinib inhibition. Interestingly, we also
found that the EGFRE884K mutation alone
decreased the sensitivity of the receptor to
erlotinib inhibition compared with both wild-
type and L858R (Figure 4A). E884K abrogated
the sensitizing effect of the L858R mutation
towards erlotinib inhibition (Figure 4C). The
relative erlotinib resistance is dose-dependent;
this dose dependency appeared to be overcome
by increasing the dose to 1 μM. Conversely,
EGFRE884K and EGFRL858R + E884K were both
found to increase the gefitinib-sensitivity of
the mutated receptor to a greater extent than
EGFRWT (Figure 4B). Most interestingly, when
the two gefitinib-sensitizing mutations—L858R
and E884K—coexisted, they synergistically
sensitized the receptor to gefitinib inhibition,
with dramatic inhibition of phospho-EGFR
seen at 0.1 μM (Figure 4B,C). This double
mutant form is more sensitive to gefitinib inhi-
bition than either L858R or E884K alone. In
summary, while the E884K mutation increases
sensitivity of the EGFR to gefitinib inhibition,
it reduces sensitivity of the receptor to erlotinib
inhibition (Figure 4C).
Residue E884 is conserved in members of the
EGF receptor family (Figure 4D), and it is unclear
how common the mutation E884K is among
the NSCLC patient population. Whether or not
there are racial differences in the frequency of
this mutation has yet to be determined. Recent
sequencing studies, in which most known muta-
tions were identified, only sequenced the EGFR
gene from exons 18–21.12,13 Since the E884K
mutation is present in exon 22, it would not
have been identified. Among Asians, a high
frequency of EGFR tyrosine kinase mutations
in adeno carcinomas have been identified—55%
in Taiwan13 and 49% in Japan.12 It has been
reported that double missense mutations of the
EGFR often involve L858R.5–7
This case report demonstrates that, according
to molecular profiling, it is possible to achieve
a meaningful response from an EGFR TKI,
even in the setting of a terminal condition such
as extensive leptomeningeal metastases. To the
authors’ knowledge, this differential response
between erlotinib and gefitinib, mediated
by the EGFR mutation E884K, has not been
reported before. The T790M mutation of
the EGFR has recently been shown to confer
resistance to both gefitinib and erlotinib.14,15
This case provides new evidence to expand the
spectrum of the effects of EGFR mutations on
small-molecule inhibitors (Figure 5).
CONCLUSION
To the authors’ knowledge, this is the first case
to describe a striking response to gefitinib in a
patient with leptomeningeal metastases and
erlotinib-refractory lung adenocarcinoma. The
case shows that some EGFR mutations can
modulate differential responses to erlotinib and
gefitinib. Eventually, molecular and mutational
profiling of lung tumors might help to predict
differential response to TKIs and could guide
the selection of optimal therapeutics. Patients
with lepto meningeal metastases represent a
unique subgroup of patients with advanced
disease, who might still benefit from individual-
ized targeted therapy with small molecule TKIs
selected based on the patient’s mutational profile.
Further biochemical and functional assays are
needed to better understand the alteration of cell
signaling that is mediated by these mutations.
Furthermore, structural studies such as X-ray
crystallography of the novel mutant receptor
EGFRL858R + E884K are warranted, to elucidate
the structural mechanism behind the differen-
tial responses towards the inhibitors conferred
by these mutations.
Supplementary information
is available on the
Nature Clinical Practice Oncology website.
References
1 Herbst RS et al. (2005) TRIBUTE: a phase III trial
of erlotinib hydrochloride (OSI-774) combined
with carboplatin and paclitaxel chemotherapy in
advanced non-small-cell lung cancer. J Clin Oncol
23: 5892–5899
2 Grossman SA and Krabak MJ (1999) Leptomeningeal
carcinomatosis. Cancer Treat Rev 25: 103–119
3 Chamberlain MC (2005) Neoplastic meningitis. J Clin
Oncol 23: 3605–3613
4 Herrlinger U et al. (2004) Leptomeningeal metastasis:
survival and prognostic factors in 155 patients.
J Neurol Sci 223: 167–178
5 Paez JG et al. (2004) EGFR mutations in lung cancer:
correlation with clinical response to gefitinib therapy.
Science 304: 1497–1500
6 Pao W et al. (2004) EGF receptor gene mutations are
common in lung cancers from “never smokers” and are
associated with sensitivity of tumors to gefitinib and
erlotinib. Proc Natl Acad Sci USA 101: 13306–13311
Acknowledgments
S Dietrich is supported by
a Boehringer Ingelheim
Foundation Research
Fellowship, Germany.
TY Seiwert is supported by
a CALGB research grant
award. GC Davies
and S Lipkowitz are
supported by the Intramural
Research Program of
the NIH, National Cancer
Institute, Center for Cancer
Research. R Salgia is
supported by NIH/NCI-R01
award, American Cancer
Society Award (National), and
Institutional Cancer Research
Awards from the University of
Chicago Cancer Center with
the American Cancer Society
and the V-Foundation.
PC Ma is supported by
NIH/NCI-K08 award, the
American Association
for Cancer Research–
AstraZeneca–Cancer
Prevention and Treatment
Translational Lung Cancer
Research Fellowship, and
American Cancer Society
(Illinois Division)-LUNGevity
Foundation Lung Cancer
Treatment Research Award.
ncponc_2005_288.indd 56ncponc_2005_288.indd 56 13/12/05 1:21:36 pm13/12/05 1:21:36 pm
Nature Publishing Group
©2006
CASE STUDY
JANUARY 2006 VOL 3 NO 1 CHOONG ET AL. NATURE CLINICAL PRACTICE ONCOLOGY 57
www.nature.com/clinicalpractice/onc
7 Lynch TJ et al. (2004) Activating mutations in
the epidermal growth factor receptor underlying
responsiveness of non-small-cell lung cancer to
gefitinib. N Engl J Med 350: 2129–2139
8 Namba Y et al. (2004) Gefitinib in patients with
brain metastases from non-small-cell lung cancer:
review of 15 clinical cases. Clin Lung Cancer 6:
123–128
9 Ceresoli GL et al. (2004) Gefitinib in patients with
brain metastases from non-small-cell lung cancer: a
prospective trial. Ann Oncol 5: 1042–1047
10 Hotta K et al. (2004) Effect of gefitinib (‘Iressa’,
ZD1839) on brain metastases in patients with
advanced non-small-cell lung cancer. Lung Cancer 46:
255–261
11 Omuro AM et al. (2005) High incidence of disease
recurrence in the brain and leptomeninges in patients
with nonsmall cell lung carcinoma after response to
gefitinib. Cancer 103: 2344–2348
12 Kosaka T et al. (2004) Mutations of the epidermal
growth factor receptor gene in lung cancer. Cancer
Res 64: 8919–8923
13 Huang SF et al. (2004) High frequency of epidermal
growth factor receptor mutations with complex
patterns in non-small cell lung cancers related to
gefitinib responsiveness in Taiwan. Clin Cancer Res
10: 8195–8203
14 Kobayashi S et al. (2005) EGFR mutation and
resistance of non-small-cell lung cancer to gefitinib.
N Engl J Med 352: 786–792
15 Pao W et al. (2005) Acquired resistance of lung
adenocarcinomas to gefitinib or erlotinib is associated
with a second mutation in the EGFR kinase domain.
PLoS Med 2:e73
16 Ma PC et al. (2005) Functional expression
and mutations of c-Met and its therapeutic inhibition
with SU11274 and small interfering RNA in non-small
cell lung cancer. Cancer Res 65: 1479–1488
Competing interests
The authors declared
they have no competing
interests.
ncponc_2005_288.indd 57ncponc_2005_288.indd 57 13/12/05 1:21:37 pm13/12/05 1:21:37 pm
Nature Publishing Group
©2006
... Most tyrosine kinase domain mutations (85-90%) had a deletion in exon 19 and substitution of L858R in exon 21. Recent studies have also reported a rare exon 22 mutation (E884K) that may reduce sensitivity to different EGFR inhibitors [72]. Several mutations at the cytoplasmic region cause destabilization of the conformation, upregulation of kinase activity, and irregularly promote downstream signaling pathways by avoiding cell apoptosis [73][74][75]. ...
Article
Full-text available
Lung cancer has a high prevalence, with a growing number of new cases and mortality every year. Furthermore, the survival rate of patients with non-small-cell lung carcinoma (NSCLC) is still quite low in the majority of cases. Despite the use of conventional therapy such as tyrosine kinase inhibitor for Epidermal Growth Factor Receptor (EGFR), which is highly expressed in most NSCLC cases, there was still no substantial improvement in patient survival. This is due to the drug’s ineffectiveness and high rate of resistance among individuals with mutant EGFR. Therefore, the development of new inhibitors is urgently needed. Understanding the EGFR structure, including its kinase domain and other parts of the protein, and its activation mechanism can accelerate the discovery of novel compounds targeting this protein. This study described the structure of the extracellular, transmembrane, and intracellular domains of EGFR. This was carried out along with identifying the binding pose of commercially available inhibitors in the ATP-binding and allosteric sites, thereby clarifying the research gaps that can be filled. The binding mechanism of inhibitors that have been used clinically was also explained, thereby aiding the structure-based development of new drugs.
... The previous studies demonstrated that erlotinib after failure of gefitinib may produce clinical benefit against patients who had a PFS of more than 6 months during gefitinib treatment or a long SD on prior gefitinib therapy [12,16,17]. Although it is unknown why the duration of the previous EGFR-TKI treatment is linked to the prediction of duration of re-treatment, the reasons, such as different sensitivity of 1st TKI, and presence of sensitive or resistant mutations are presumed [18][19][20]. Considering these findings, it can be suggested that the duration of maintenance with the previous 1st TKI could affect the efficacy of 2nd TKI, as a re-treatment after 1st TKI failure. To our knowledge, this is the first study to examine the effectiveness of afatinib as a re-challenge based on the treatment duration of prior 1st TKI; however, the exact mechanism remains unknown. ...
Article
Full-text available
Background Re-challenge of erlotinib after gefitinib failure is reported to yield some benefit in patients with non-small cell lung cancer (NSCLC) harboring epidermal growth factor receptor (EGFR) mutation. However, little is known about the re-challenge of afatinib after 1st generate on EGFR tyrosine kinase inhibitor (TKI) failure. Methods From May 2015 to August 2018, 62 patients with advanced NSCLC harboring sensitive EGFR mutation received afatinib after gefitinib and/or erlotinib failure at our institution was included in our retrospective study. Results The overall response rate (ORR) and disease control rate (DCR) of afatinib as re-challenge were 17.0% and 79.2%, respectively. The median time on treatment of 1st generation EGFR-TKI (1st TKI) was 14 months. By multivariate analysis, smoking, performance status (PS), and time on treatment of 1st TKI with more than 10 months were confirmed to be independent prognostic factors predicting a worse progression-free survival (PFS), and significant prognostic markers for overall survival (OS) were PS and time on treatment of 1st TKI with more than 10 months, especially in patients with exon 19 deletion. Conclusions Re-challenge of afatinib was identified as one of the therapeutic options after 1st TKI failure in the patients with advanced NSCLC harboring EGFR mutation when the time of treatment by prior 1st TKI is more than 10 months.
Article
Full-text available
Purpose Leptomeningeal metastasis (LM) is a severe complication of non-small cell lung cancer (NSCLC). In patients with NSCLC LM harboring epidermal growth factor receptor (EGFR) mutations, osimertinib is favored over alternative EGFR tyrosine kinase inhibitors (TKIs). However, the efficacy of osimertinib relative to other EGFR-TKIs is not well established for patients with LM. We aimed to compare the efficacy of EGFR-TKIs in EGFR-mutated NSCLC LM. Methods This systematic review and meta-analysis performed according to PRISMA guidelines included studies of adult patients with EGFR-mutated NSCLC and a diagnosis of LM who received an EGFR-TKI for the treatment of LM. We searched Medline ALL, Embase, Cochrane Central Register of Controlled Trials, Scopus, and Web of Science Core Collection. The evaluation of biases was done by using the Ottawa-Newscastle scale. The hazard ratio was used as the parameter of interest for overall survival (OS) and central nervous system-specific progression-free survival (PFS). Results 128 publications were included with 243 patients and 282 lines of EGFR-TKI for NSCLC LM that met inclusion criteria. The median PFS in patients receiving any EGFR-TKI was 9.1 months, and the median OS was 14.5 months. In univariate analyses of the entire cohort, osimertinib treatment demonstrated significantly prolonged PFS, but not OS, compared to other EGFR-TKIs. Osimertinib demonstrated significantly prolonged PFS and OS in the subset of patients who were previously treated with EGFR-TKIs, but not in EGFR-TKI naïve patients. Conclusion Osimertinib is associated with improved outcomes compared to other EGFR-TKIs, particularly in patients previously treated with EGFR-TKIs. An important limitation is that most patients were derived from retrospective reports. These results highlight the need for prospective studies for this difficult-to-treat patient population.
Article
Herein, we report the synthesis of a series of new quinazoline sulfonamide conjugates 2-16 and their evaluation as potential anticancer agents via dual targeting of EGFRT790M and VEGFR-2. The newly synthesized compounds were designed based on the structure requirements of the target receptors and were confirmed using spectral data. The compounds were evaluated for their cytotoxicity against four cancer cell lines (HepG2, MCF-7, HCT116 and A549) using MTT assay. The most active compounds were further evaluated for their inhibitory activity against EGFRT790M and VEGFR-2. Compound 15 showed the most significant cytotoxic activity with IC50 = 0.0977 µM against MCF-7 and the most potent inhibitory activity against both EGFR and VEGFR with IC50 = 0.0728 and 0.0523 µM, respectively. Compound 15 was able to induce apoptosis in MCF-7 cells and cell cycle arrest at the G2/M phase. The relative safety profile of 15 was assessed using HEK-293 normal cell line and an ADMET profile was carried out. Radiosensitizing evaluation of 15 proved its significant ability to sensitize the cancer cell to the effect of radiation after being subjected to a single dose of 8 Gy gamma irradiation. Molecular docking studies revealed that 15 could bind to the ATP-binding site of EGF and VEGF receptors, inhibiting their activity.
Article
Full-text available
Background: Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) are an effective treatment for non-small-cell lung cancer (NSCLC) harbouring EGFR mutations. The development of isolated central nervous system (CNS) metastases is a relevant clinical problem in patients who respond well to EGFR-TKIs. Methods: We present a patient with isolated progression of brain metastases during treatment of EGFRmutated NSCLC with an EGFR-TKI and review the treatment options in this setting, including the evidence for and toxicity of treatment with high-dose TKIs. Results: Oligometastatic CNS progression during TKI therapy may be treated locally. Both whole brain radiotherapy (WBRT) and stereotactic brain irradiation are well tolerated and effective in this setting. The use of high-dose pulsed TKIs is intended to increase the concentration of TKI in the brain and has been reported to be effective and without significant toxicity in case reports and small case series. These therapeutic options are illustrated in the case of a 44-year-old NSCLC patient who developed CNS progression after WBRT during second-line erlotinib and was treated locally with stereotactic radiosurgery (SRS) and, upon further CNS progression, with high-dose pulsed erlotinib. This resulted in intracerebral response; however, significant haemorrhage also occurred. Severe haemorrhage has not previously been described as a complication of high-dose pulsed erlotinib. Conclusion: Possible explanations for isolated CNS progression during TKI treatment include inadequate dosing across the blood—brain barrier and longer survival on TKIs. The efficacy and tolerability of high- dose pulsed TKIs for CNS metastases has been previously reported. None of the cases reported showed the severe haemorrhage and cerebral oedema that developed in our patient. Simultaneous anticoagulation as well as previous SRS may have predisposed our patient to haemorrhage and may prove to be relative contraindications to high-dose pulsed erlotinib. Most centres only see a few patients in this clinical situation, and co-operative efforts are needed to collect and analyse similar cases and to develop appropriate treatment strategies.
Article
The Epidermal Growth Factor Receptor (EGFR) has been targeted through the development of selective tyrosine kinase inhibitors (TKIs) and monoclonal antibodies (mAb). These molecules have shown effectiveness in a subset of patients with specific genetic alterations (i.e. gain-of-function EGFR mutations or EGFR gene amplification) and have been approved for their use in non-small-cell lung cancer (NSCLC), colorectal cancer (CRC), pancreatic cancer and head and neck cancer. In addition, extensive research is being performed in many other tumour types hoping for a future approval. However, the majority of the patients show no benefit from these molecules due to primary mechanisms of resistance, already present before treatment or show disease progression upon the acquisition of drug resistance mechanisms during the treatment. At present, the majority of patients display resistance due to alterations in genes related to the EGFR signalling pathway that eventually circumvent EGFR inhibition and allow cancer progression. Thus, in this review article we focus on the molecular mechanisms underlying drug resistance via genetic alterations leading to resistance to all anti-EGFR drugs approved by the FDA and/or EMA. We also discuss novel approaches to surmount these chemoresistance modalities.
Article
Mic Abstract : Patients with advanced non-small-cell lung cancer (NSCLC) harboring epidermal growth factor receptor (EGFR) mutation need novel treatments, especially for those with brain metastasis. This phase 1b study aimed to evaluate the safety and preliminary efficacy of epitinib (HMPL-813) for EGFR-mutated NSCLC with brain metastasis. Results suggested that epitinib 160 mg once daily had a manageable safety profile and showed a promising efficacy. Background : Non-small-cell lung cancer (NSCLC) had poor prognosis in patients with brain metastasis. The trial evaluated the safety and efficacy of epitinib (HMPL-813), an epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI), for EGFR-mutant NSCLC with brain metastasis. Patients and Methods : This open-label, dose-expansion phase Ib study (ClinicalTrials.gov: NCT02590952) was conducted at seven Chinese centers and enrolled patients with EGFR-mutant advanced NSCLC with brain metastasis. Epitinib was administered at 120 mg or 160 mg, orally QD. The primary endpoint was safety and tolerability. Results : Between April 2015 and April 2019, 72 patients were enrolled and received epitinib at 120 mg (n=30) or 160 mg (n=42). Treatment-related adverse events (TRAEs) of grade ≥3 occurred in 13 (43.3%) patients in 120 mg group and 21 (50.0%) in 160 mg group. The objective response rate (ORR) was 53.6% (95% CI 33.9%–72.5%) in 120 mg group and 40.5% (25.6%–56.7%) in 160 mg group. The median duration of response in the 120 mg and 160 mg groups were 7.40 months (95% CI 3.70–7.40) and 9.10 months (6.50–12.00), respectively. The median progression-free survival were 7.40 months (95% CI 5.40–9.20) and 7.40 months (5.50–10.00), respectively. Conclusion : In patients with EGFR-mutant NSCLC with brain metastasis, epitinib was well tolerable with a promising efficacy. According to the comprehensive assessment on safety and efficacy, 160 mg QD could be the recommended phase 2 dose.
Article
Full-text available
Background: EGFR tyrosine kinase inhibitors (TKIs) have revolutionized the therapeutic approach for EGFR mutated patients. However, acquired resistance to EGFR-TKI therapy is unavoidable. Repeat biopsy cannot be used, and peripheral blood detection shows a low positive rate in cases of brain-only disease progression. Methods: Droplet digital polymerase chain reaction (PCR) (ddPCR) was performed on the plasma and cerebrospinal fluid (CSF) samples of 79 lung adenocarcinoma (LUAD) patients with EGFR mutations and central nervous system (CNS) metastasis. The differences in the EGFR mutation status between the paired plasma and CSF samples were assessed, and the role of CSF testing as a predictor of overall survival was evaluated. Results: The CSF of patients with neurological symptoms, EGFR-TKI treatment, or leptomeningeal metastasis (LM) had a significantly higher positive rate of EGFR mutation compared to the plasma samples (P=0.001, P=0.035, P=0.019, respectively). Moreover, EGFR mutation status in CSF was consistent with neurological symptoms and LM (kappa =0.455, P<0.001; kappa =0.508, P<0.001; respectively). For the patients with brain metastasis, EGFR mutation-positive rate in CSF samples was lower than that in plasma samples (28.3% vs. 64.2%, P<0.001), while the patients with LM had the opposite result (84.6% vs. 38.5%, P=0.004). Moreover, patients with EGFR mutation in their CSF experienced worse survival [hazard ratio (HR) =2.93, 95% confidence interval (CI): 1.45-5.92; P=0.003, P adjust <0.0001]. Conclusions: The EGFR mutation status of CSF was different from that of plasma and is correlated with patient prognosis. CSF could be helpful in detecting the EGFR mutation status of patients, particularly in cases of LM.
Article
Full-text available
Objectives Urinary incontinence is estimated to affect up to 34% of women with gynecologic cancers. Chemotherapeutic effects on urinary symptoms have been scarcely studied. The aim of our study was to examine the impact of carboplatin/paclitaxel chemotherapy on urinary symptoms. Methods This was a prospective cohort study of all women undergoing adjuvant chemotherapy with carboplatin/paclitaxel after their primary debulking surgery for ovarian, fallopian tube, peritoneal, or endometrial cancers performed at a tertiary medical center by board certified gynecologic oncologists. We used validated questionnaires (incontinence impact questionnaire (IIQ-7), medical, epidemiologic, and social aspects of aging (MESA), urogenital distress inventory (UDI-6), the Sandvik severity index, and functional assessment of cancer therapy/GynecologicOncology Group-neurotoxicity (FACT/GOG-Ntx)) to assess the effects of carboplatin/paclitaxel therapy on the incidence and severity of urinary incontinence at three time points during the participants’ chemotherapy regimen: start of chemotherapy (pretreatment), during the fifth chemotherapy cycle (mid-cycle), and during the 6–12 week post-chemotherapy visit (post-treatment). Results We identified 62 women with ovarian, fallopian, peritoneal, and endometrial cancer who received carboplatin/paclitaxel therapy between May 2009 and December 2012 who met all of the inclusion criteria. Analysis of median IIQ-7 scores, across all time points, showed a statistically significant difference (0.0 (0.0 to 4.8), 0.0 (0.0 to 7.1), 0.0 (0.0 to 0.0), p=0.002, respectively). Examining pairwise differences between two treatment points, IIQ-7 pretreatment versus mid-treatment and pretreatment versus post-treatment, did not achieve significance (0.0 (0.0 to 2.4), p=0.13 and 0.0 (0.0 to 0.0), p>0.999, respectively), but the decrease in IIQ-7 mid-treatment versus post-treatment was statistically significant (0.0 (−2.4 to 0.0), p=0.003). Generalized estimating equation model analysis also showed significant changes in both median MESA for urge urinary incontinence (MESA-UUI) and median MESA for stress urinary incontinence (MESA-SUI) across all time points (p=0.003 and p=0.009, respectively). MESA-UUI and MESA-SUI pretreatment versus mid-treatment analysis achieved significance (2.0 (0.0 to 4.0), p=0.003 and 0.0 (0.0 to 2.0), p=0.01, respectively), demonstrating an increase in the incidence of stress urinary incontinence and urge urinary incontinence. There was a statistically significant association between the changes in FACT and UDI-6 scores from pretreatment versus mid-treatment, with a correlation coefficient of 0.37 (95% CI 0.08 to 0.61, p=0.005). Conclusion The study achieved its primary aim in demonstrating an impact of carboplatin/paclitaxel therapy on urinary incontinence severity and suggests if may be a factor leading to new onset or worsening urinary incontinence. As quality of life can be significantly impacted by these chemotherapeutic changes, further investigation is warranted to determine if the effects on urinary incontinence and neuropathy are transient or permanent.
Article
Full-text available
Receptor tyrosine kinase genes were sequenced in non–small cell lung cancer (NSCLC) and matched normal tissue. Somatic mutations of the epidermal growth factor receptor gene EGFR were found in 15of 58 unselected tumors from Japan and 1 of 61 from the United States. Treatment with the EGFR kinase inhibitor gefitinib (Iressa) causes tumor regression in some patients with NSCLC, more frequently in Japan. EGFR mutations were found in additional lung cancer samples from U.S. patients who responded to gefitinib therapy and in a lung adenocarcinoma cell line that was hypersensitive to growth inhibition by gefitinib, but not in gefitinib-insensitive tumors or cell lines. These results suggest that EGFR mutations may predict sensitivity to gefitinib.
Article
Neoplastic meningitis (NM) is a common problem in neuro-oncology occurring in approximately 5% of all patients with cancer, and is the third most common site of CNS metastases. NM is a disease affecting the entire neuraxis, and therefore clinical manifestations are pleomorphic affecting the spine, cranial nerves, and cerebral hemispheres. Because of craniospinal disease involvement, staging and treatment need to encompass all CSF compartments. Treatment of NM utilizes involved-field radiotherapy of bulky or symptomatic disease sites and intra-CSF drug therapy. The inclusion of concomitant systemic therapy may benefit patients with NM and may obviate the need for intra-CSF chemotherapy. At present, intra-CSF drug therapy is confined to three chemotherapeutic agents (ie, methotrexate, cytarabine, and thiotepa) administered by a variety of schedules either by intralumbar or intraventricular drug delivery. Although treatment of NM is palliative with an expected median patient survival of 2 to 6 months, it often affords stabilization and protection from further neurologic deterioration in patients with NM.
Article
Leptomeningeal carcinomatosis occurs in approximately 5% of patients with cancer. This disorder is being diagnosed with increasing frequency as patients live longer and as neuro-imaging studies improve. The most common cancers to involve the leptomeninges are breast cancer, lung cancer, and melanomas. Tumour cells reach the leptominges by hematogenous spread or by direct extension from pre-existing lesions and are then disseminated throughout the neuroaxis by the flow of the cerebrospinal fluid. Patients present with signs and symptoms from injury to nerves that traverse the subarachnoid space, direct tumour invasion into the brain or spinal cord, alterations in blood supply to the nervous system, obstruction of normal cerebrospinal fluid (CSF) flow pathways, or general interference with brain function. The diagnosis is most commonly made by lumbar puncture although the CSF cytology is persistently negative in about 10% of patients with leptomeningeal carcinomatosis. Radiologic studies may reveal subarachnoid masses, diffuse contrast enhancement of the meninges, or hydrocephalus without a mass lesion.
Article
Most patients with non-small-cell lung cancer have no response to the tyrosine kinase inhibitor gefitinib, which targets the epidermal growth factor receptor (EGFR). However, about 10 percent of patients have a rapid and often dramatic clinical response. The molecular mechanisms underlying sensitivity to gefitinib are unknown. We searched for mutations in the EGFR gene in primary tumors from patients with non-small-cell lung cancer who had a response to gefitinib, those who did not have a response, and those who had not been exposed to gefitinib. The functional consequences of identified mutations were evaluated after the mutant proteins were expressed in cultured cells. Somatic mutations were identified in the tyrosine kinase domain of the EGFR gene in eight of nine patients with gefitinib-responsive lung cancer, as compared with none of the seven patients with no response (P<0.001). Mutations were either small, in-frame deletions or amino acid substitutions clustered around the ATP-binding pocket of the tyrosine kinase domain. Similar mutations were detected in tumors from 2 of 25 patients with primary non-small-cell lung cancer who had not been exposed to gefitinib (8 percent). All mutations were heterozygous, and identical mutations were observed in multiple patients, suggesting an additive specific gain of function. In vitro, EGFR mutants demonstrated enhanced tyrosine kinase activity in response to epidermal growth factor and increased sensitivity to inhibition by gefitinib. A subgroup of patients with non-small-cell lung cancer have specific mutations in the EGFR gene, which correlate with clinical responsiveness to the tyrosine kinase inhibitor gefitinib. These mutations lead to increased growth factor signaling and confer susceptibility to the inhibitor. Screening for such mutations in lung cancers may identify patients who will have a response to gefitinib.