You are on page 1of 7

This article was downloaded by: [Universidad De Concepcion]

On: 29 November 2014, At: 12:21


Publisher: Taylor & Francis
Informa Ltd Registered in England and Wales Registered Number: 1072954 Registered office: Mortimer House,
37-41 Mortimer Street, London W1T 3JH, UK

Human Vaccines
Publication details, including instructions for authors and subscription information:
http://www.tandfonline.com/loi/khvi19

Therapeutic Vaccination with Epidermal Growth Factor


(EGF) in Advanced Lung Cancer: Analysis of Pooled
Data from Three Clinical Trials
Gisela Gonzlez, Tania Crombet, Elia Neninger, Carmen Viada & Agustn Lage
Published online: 01 Jan 2007.

To cite this article: Gisela Gonzlez, Tania Crombet, Elia Neninger, Carmen Viada & Agustn Lage (2007) Therapeutic
Vaccination with Epidermal Growth Factor (EGF) in Advanced Lung Cancer: Analysis of Pooled Data from Three Clinical Trials,
Human Vaccines, 3:1, 8-13, DOI: 10.4161/hv.3.1.3537
To link to this article: http://dx.doi.org/10.4161/hv.3.1.3537

PLEASE SCROLL DOWN FOR ARTICLE


Taylor & Francis makes every effort to ensure the accuracy of all the information (the Content) contained in
the publications on our platform. Taylor & Francis, our agents, and our licensors make no representations or
warranties whatsoever as to the accuracy, completeness, or suitability for any purpose of the Content. Versions
of published Taylor & Francis and Routledge Open articles and Taylor & Francis and Routledge Open Select
articles posted to institutional or subject repositories or any other third-party website are without warranty
from Taylor & Francis of any kind, either expressed or implied, including, but not limited to, warranties of
merchantability, fitness for a particular purpose, or non-infringement. Any opinions and views expressed in this
article are the opinions and views of the authors, and are not the views of or endorsed by Taylor & Francis. The
accuracy of the Content should not be relied upon and should be independently verified with primary sources
of information. Taylor & Francis shall not be liable for any losses, actions, claims, proceedings, demands,
costs, expenses, damages, and other liabilities whatsoever or howsoever caused arising directly or indirectly in
connection with, in relation to or arising out of the use of the Content.
This article may be used for research, teaching, and private study purposes. Terms & Conditions of access and
use can be found at http://www.tandfonline.com/page/terms-and-conditions
It is essential that you check the license status of any given Open and Open Select article to confirm
conditions of access and use.

[Human Vaccines 3:1, -13, January/February 2007]; 2007 Landes Bioscience

Research Paper

Therapeutic Vaccination with Epidermal Growth Factor (EGF)


in Advanced Lung Cancer
Analysis of Pooled Data from Three Clinical Trials
Abstract

2Hermanos Ameijeiras Hospital; Havana, Cuba

*Correspondence to: Gisela Gonzlez; Calle 216 esq 15, Atabey, Siboney, Playa;
Havana, Cuba; PO BOX 11600; Tel.: +53.7.2717645; Fax: +53.7.2720644;
Email: gisela@cim.sld.cu
Original manuscript submitted: 07/11/06
Manuscript accepted: 10/22/06

cancer vaccine, epidermal growth factor,


non-small cell lung cancer

Introduction

IEN

Abbreviations

CE

Key words

.D

ON

Previously published online as a Human Vaccines E-publication:


http://www.landesbioscience.com/journals/vaccines/abstract.php?id=3537

IST

RIB

UT
E

We have undertaken the analysis of pooled data from three pilot clinical trials of
vaccination with Epidermal Growth Factor (EGF) in patients with advanced non small
cell lung cancer (NSCLC), addressing particularly the issue of the relationship between
immunization and survival. Eightythree patients with advanced disease were included in
three pilot clinical trials and vaccinated with the EGF Vaccine. The trials were designed
to evaluate the immunogenicity and safety of the vaccine using different adjuvants,
cyclophosphamide pretreatment or not, and different dosage levels of the vaccine. The
vaccine elicited specific antiEGF antibody titers in 83% of subjects, and 49% developed
a good antiEGF antibody response. The adjuvant, the vaccine dose, and cyclophospha
mide pretreatment significantly influenced immunogenicity. Patients that seroconverted
survived significantly longer than patients who did not. Good antibody responders
survived significantly longer than poor responders. Pooled results from these trials confirm
that vaccination with EGF is safe and immunogenic in advanced NSCLC patients. The
association between good antibody responses and survival consistently appeared in
every single trial independently of the specific trial designs. Although these were small
pilot nonrandomized clinical trials not intended to confirm therapeutic effect, the survival
of the pooled patient population was statistically greater compared with 163 control
patients receiving standard treatment.

OT
D

1Center of Molecular Immunology; Havana, Cuba

An increasing body of scientific literature is showing that the therapeutic vaccine


approach in cancer is feasible and promising.1,2 More than 50 cancer vaccines are now
reported to be in clinical trials, and more than 10 have reached the Phase 3 trial stage.3
Many tumorassociated antigens have been identified, providing a basis for the design
of therapeutic vaccines; moreover it is now clear that, even when the tumorassociated
antigen is completely self, both cellular and humoral immune responses can be safely
elicited with an appropriate immunization procedure.49
However, early disappointment is coming from the fact that these successful vaccinations have not yet translated into a clear clinical advantage for a target cancer. There also is
often an undue generalization of data obtained in a particular situation. A substantial part
of the accumulated clinical experience has been obtained in melanoma. Less data exist in
patients with most common tumors, and few data exist in advanced lung cancer, the most
prevalent and fatal malignancy worldwide.
During the last ten years we have been studying the effect of vaccination against EGF,
first in the preclinical setting and subsequently in cancer patients. In laboratory animals we
have demonstrated that antibody responses can be elicited against EGF and that such an
antibody responses correlate with survival when animals are challenged with EGF receptor
(EGFR) expressing tumors.10,11
The clinical experience with the EGF vaccine began with a small pilot clinical trial
where 10 patients with epidermoidorigin tumors in advanced stages were vaccinated.
Results from this trial demonstrated that, in such a patient population, vaccination was
safe and immunogenic.7
Afterwards, new clinical trials were focused on patients with advanced (stages IIIb
and IV) NSCLC. More than 80 NSCLC patients have been vaccinated, in a series of
three consecutive pilot clinical trials, exploring diverse adjuvants, dosages and vaccination
schedules, before launching a randomized study. Some of the results of each of these
early pilot trials have been previously published.8,9 In the present paper we undertake the

20

06

LA

ND

ES

BIO

SC

EGF epidermal growth factor


NSCLC non-small cell lung cancer
EGFR epidermal growth factor receptor
WHO World Health Organization
PS
performance status
ECOG Eastern Cooperation Oncology

Group
ELISA enzyme-linked immunosorbent

assay
GAR good antibody responders
PAR
poor antibody responders

Downloaded by [Universidad De Concepcion] at 12:21 29 November 2014

Gisela Gonzlez1,*
Tania Crombet1
Elia Neninger2
Carmen Viada1
Agustn Lage1

Human Vaccines

2007; Vol. 3 Issue 1

Vaccination with EGF for Lung Cancer Therapy

Table 1

Main features of the three reported trial designs

Downloaded by [Universidad De Concepcion] at 12:21 29 November 2014

Trial
Group
N Dose

(EGF Equivalent)

Cyclophosphamide
Pretreatment

Adjuvant Induction Schedule Reimmunizations

1st trial

1a
1b

10
50 mg
No
10

Alum
Montanide

Days 0, 7, 14, 21, 51

At decrease in antibody titers

2nd trial

2a
2b

10
50 mg
Yes
10

Alum
Montanide

Days 0, 7, 14, 21, 51

At decrease in antibody titers

3rd trial

3a
3b

21
22

Alum

Days 0, 7, 14, 21, 51

Monthly

71 mg
142 mg

No

analysis of the pooled data from a series of 83 patients immunized


with the EGF Vaccine in three clinical trials, addressing in particular
the relationship between immunization and survival, and looking for
the consistency of the early findings about safety, immunogenicity
and association between immune response and survival.

Patients and Methods


Immunogens and immunization schedules. The vaccine was
composed of human recombinant EGF, obtained from recombinant
E. coli, conjugated to the P64K recombinant protein from Neisseria
meningitidis12,13 as carrier. Glutaraldehyde (0.05%) was added to the
protein mixture for conjugation, the reaction proceeded for 1 hour
and then the conjugate was purified by dialysis and filtered in sterile
conditions. The final conjugate contained 2 EGF molecules for each
P64k molecule and was administered with an adjuvant.
For preparations in which alum was used as adjuvant, conjugates
were mixed after filtration with 2 mg alum/dose; adsorption was
achieved by constant stirring at room temperature for 1 hour under
sterile conditions. All procedures were performed according to good
manufacturing practices. When Montanide ISA51 was used as the
adjuvant, it was mixed until emulsification with equal volume of the
conjugate immediately before injection.
The clinical trials. Pooled data from three pilot clinical trials were
used for this analysis. In the first trial, 20 patients were randomized
to vaccination with EGFP64K adsorbed to alum (ten patients) or
emulsified in Montanide ISA 51(ten patients). In the second trial,
additional 20 patients were similarly randomized but all they received
a single dose of cyclophosphamide (200 mg/m2) three days prior
to the first vaccination. In the third trial, another 43 patients were
randomized to receive either a single dose of the vaccine (equivalent
to 71 mg EGF) or a double dose (equivalent to 142 mg EGF). The
main features of dose and immunization schedules in the three clinical
trials are described in Table 1. Protocols were approved by the Ethics
Committee of the participating hospitals and also received clearance
from the Cuban National Regulatory Authority (CECMED).
Toxicity was graded according to the World Health Organization
(WHO) standard toxicity scale. Performance status (PS) was evaluated
according to the Eastern Cooperation Oncology Group (ECOG)
criteria.
Eligibility criteria. Patients with histologically proven NSCLC
at advanced stages (IIIb or IV) progressing after initial treatment or
considered not amenable to any other modality of oncospecific treatment were eligible, and were included in the trials four weeks after
finishing their last oncospecific treatment.
Other eligibility criteria were age between 18 and 80 years, PS
between 0 and 2, normal liver, kidney and bone marrow functions,
no pregnancy or lactation, no severe uncontrolled comorbidity, no
www.landesbioscience.com

Table 2

Characteristics of patients included in the three


reported trials and in the concurrent control




n

Age

Patients Included Concurrent


in the Trials
Controls
83
163
58.8 9.5
57.4 9.4

Gender

Male
Female

81 %
19 %

65 %
35 %

Performance
status (WHO)

0
1
2

13 %
69 %
18 %

2%
53%
45%

Clinical stage

III
IV

48 %
52 %

67%
33%

31.3 %
55.4 %

100%

8.4 %
2.4 %
2.4 %

Previous oncospecific no treatment


treatment
polychemotherapy
(before inclusion)
alone or combined

with RT and/or

surgery

RT alone

surgery alone

surgery + RT

secondary malignancies, and no previous history of hypersensitivity


to foreign proteins. All patients signed informed consent before being
included in the trials.
Measurement of antibody titers. Antibody titers against human
EGF were measured through an enzymelinked immunosorbent
assay (ELISA) as previously described.711 Antibody titer was defined
as the higher serum dilution producing an absorbance measurement
higher than the blank plus three standard deviations.
The antibody response was considered positive (seroconversion)
when antibody titers were at least twice their preimmunization
values. However a more stringent response criteria was introduced,
classifying patients as good antibody responders (GAR) when the
antibody response reached titers 1:4000 and at least 4X the preimmunization values, and as poor responders (PAR) if not.
Concurrent non-randomized control. Due to regulatory reasons,
there was a time lag between consecutive clinical trials with the EGF
Vaccine. During this intermediate time, patients newly diagnosed
and complying with inclusion criteria were treated by standard procedures, and then included in the control group and closely monitored
similarly to patients in the Clinical Trials. This group of 163 NSCLC
patients was provisionally taken as a reference point for survival analysis. Table 2 summarizes the main characteristics of patients included
in the trials and in the concurrent nonrandomized control.
Statistical analysis.
Statistical significance of differences in
percentages of qualitative variables was assessed through ChiSquare

Human Vaccines

Vaccination with EGF for Lung Cancer Therapy

Table 3

Trial
Group
%
%

Seroconversion
GAR

Downloaded by [Universidad De Concepcion] at 12:21 29 November 2014

1st trial

1a (Alum)

1b (Montanide)

2nd trial

2a (Cyclophosphamide/Alum)

78 %

22 %

100 %

73 %

90 %

30 %

2b (Cyclophosphamide/Montanide)

100 %

100 %

3rd trial

3a (Alum/single dose)

52.9 %

41.2 %

3b (Alum/double dose)

95.2 %

38.1 %

Figure 1. (A) Two patients with characteristic kinetics of antibody response;


arrows indicates (1) the end of the induction period and (2) the first reimmu
nization. Abscissa is time after the first vaccination and ordinate is antibody
titers; (B) Kinetics of antibody response in the group of patients that received
a low dose of cyclophosphamide before vaccination and Montanide ISA 51
as adjuvant; this treatment group showed the strongest antibody response.
The abscissa is time after the first vaccination, and the ordinate is geometric
mean of maximal antibody titer.

tests. Comparisons between antibody titers were done using the


MannWhitney UTest when two groups were compared or the
KruskalWallis test when groups were more than two. Estimations
of survival times were made with KaplanMeier curves and were
compared using the Logrank test.14,15

Results
The EGF vaccine was safe and immunogenic. There were some
natural antibody titers in most patients before vaccination. The
geometric mean of patients baseline antiEGF antibodies was of
1:224. As we have previously published,16 these natural antibodies
are EGFspecific in the sense that they can be blocked in the assay
with added EGF. After EGF vaccination, 83% of vaccinated patients
10

at least doubled their baseline antiEGF antibody


titers (considered as seroconversion), and 49%
increased their baseline levels up to 4 fold, with
titers >1:4000 being considered GAR.
In patients showing seroconversion, the median
Geometric
value
of maximal antibody titers was 1:4000 and
Means Maximal
geometric mean 1:3954 (range of 1:100 1:400
Ab titers
000). The behavior of the immunogenicity data
1023
in each trial is described on Table 3.
6456
The adjuvant and the Cyclophosphamide
2291
pretreatment significantly increased immunoge10471
nicity. Patients vaccinated using Montanide ISA
1246
51 as adjuvant had higher % seroconversion and
2519
% GAR as well as increased geometric mean
maximal antiEGF titers (p < 0.05). Patients
pretreated with Cyclophosphamide before vaccination had increased geometric mean maximal antiEGF titers
(p < 0.05). These effects appeared as a nonsignificant trend in the
previously published separated trials but achieved statistical significance in the pooled data. The trend to an increased antibody
response in patients vaccinated with higher doses did not reach
statistical significance in this series.
GAR was related to performance status, being 70% for patients
with PS0, 52% for patients with PS1, and 17% for patients with
PS2 (p = 0.03). Other analyzed variables (sex, age, clinical stage, and
previous treatment) had no influence on immunogenicity.
The immune responses were shortlasting. After the last vaccine
dose of the induction period, antibody titers started to decrease at
2.64 1.89 months. After the decrease, reimmunization elicited a
recovery of antibody titers to similar values, without producing the
booster effect characteristic of many antiinfectious vaccines. Typical
kinetics of antibody titers for two patients is shown in Figure 1A,
and the mean behavior of antibody titers for the ten patients treated
with the vaccine in Montanide adjuvant and with cyclophosphamide
pretreatment is shown in Figure 1B.
Vaccination was well tolerated, producing only mild (Grade I or
II) toxicity limited to pain at the site of injection (11%), chills (23%),
tremors (17%), nausea (16%), headaches (21%), hot flashes (11%),
and fever (16%), which disappeared after standard medications.
Antibody response was associated with better survival. Within the
group of vaccinated patients (Fig. 2), survival was better in patients
that seroconverted (mean 11.06 months, median 8.4 months) when
compared with patients that did not (mean 5.67 months; median
3.5 months) .There was an additional advantage in survival for GAR
(mean 12.2 months, median 8.37 months) as compared with PAR
(mean 8.07 months, median 8.07 months).
When the association between good antibody response and survival
was analyzed within the subgroup of patients with Performance
Status 0 and 1, the association remained statistically significant.
Vaccinated patients had better survival that concurrent control.
Although these clinical trials were not designed to evaluate the effect
of vaccination on survival, a preliminary assessment of this effect
could be obtained by comparing survival of vaccinated patients with
that of a concurrent (although nonrandomized) control group. As
shown in Figure 3A, vaccinated patients survived significantly longer
(mean 9.13 months; median 8.0 months) than concurrent controls
(mean 4.85 month; median survival 4.53 months). This survival
advantage continued to be evident inside the subgroup of patients
with similar performance status (Fig. 3B).

Immunogenicity descriptors in the randomization groups


from all trials (% of seroconversion, % of GAR and geometric
means of maximal antibody titers)

Human Vaccines

2006; Vol. 3 Issue 1

Downloaded by [Universidad De Concepcion] at 12:21 29 November 2014

Vaccination with EGF for Lung Cancer Therapy

Figure 2.
Survival functions for (A) patients with seroconversion (-) (mean 11.6
months; median 8.4 months) compared with patients without seroconversion
(-) (mean 5.67 months; median 3.5 months) (log rank test: p = 0.0049) and
for (B) Good Antibody Responders [GAR] (-) (mean 12.2 months; median
8.37 months) compared with Poor Antibody Responders [PAR] (-) (mean SV
8.07 months; median 8.07 months) (log rank test: p = 0.036).

There was no significant effect of age, as a standalone variable,


on survival. However, the effect of vaccination on survival was more
evident in younger patients (Table 4), although there was no difference in the immune response to the vaccine (% seroconversion, %
GAR, geometric mean maximal antiEGF titers) between older and
younger patients. The survival advantage of GAR remained evident
for either younger or older patients.
Increased survival times for vaccinated patients continued to be
statistically significant after stratification by sex and clinical stage

Discussion
The EGF and its cell membrane receptor have recently become
appealing targets for new anticancer treatments17 mainly because
EGFR is overexpressed in cancer cells from many of the most
prevalent epithelial tumors, such as lung, colon, head and neck,
cervix and breast neoplasms,1822 and moreover there is evidence of
its participation in the malignant cell physiology.17
www.landesbioscience.com

Figure 3. Survival functions for (A) vaccine group (-) (mean 9.83 months;
median 8.0 months) and concurrent controls () (mean 6.2 months; median
4.1 months) (log rank test: p=0.0000) and (B) vaccine group with PS 0 and
1 (-) (mean 10.47 months; median 8.23 months) and concurrent controls
with PS 0 and 1 (-) (mean 6.04 months; median 4.2 months); the survival
advantage continued to be statistically significant inside the subgroup of
patients with PS 0 and 1 (log rank test:
p
= 0,0000). Six months survival
was achieved by 60.8% of vaccinated and 37.0% of control patients (Chi
square: p
= 0.001). Twelve month survival was achieved by 26.6% of vac
cinated and 8.6% of control patients (Chi square:
p = 0.000).

Table 4

Mean and median survival times for vaccinated


and control patients divided into age cohorts

Age Group

Treatment
Group

Mean

Median

Younger than 60

vaccinated
controls

11.04 1.52
4.67 0.65

8,37
3,33

<0.0001

Older than 60

vaccinated
controls

10.19 1.40
6.98 1.27

8,07
4,53

<0.02

Human Vaccines

11

Downloaded by [Universidad De Concepcion] at 12:21 29 November 2014

Vaccination with EGF for Lung Cancer Therapy

During the last ten years several small molecule inhibitors of


EGFRassociated Tyrosine Kinase and several monoclonal antibodies
against EGFR have been tested in clinical trials,2327 but the active
immunization approach (cancer vaccines) has been much less
explored.
We have undertaken precisely this approach and, after showing
that laboratory animals can be safely immunized against EGF10,11 and
that such immunization has antitumor activity, an EGFVaccine
was allowed to enter into clinical testing. Three pilot clinical trials
were performed, accruing more than 80 NSCLC, which explored
diverse dosage, schedule and adjuvanticity conditions. This has been
the only known published experience of human vaccination with
EGF.
The first striking observation was related to immunogenicity,
starting from the finding that natural autoantibodies (specific
IgG) against EGF do exist in humans before vaccination, both in
normal subjects and in cancer patients.16 This baseline autoantibody
response can be increased after immunization.
The immunogenicity of vaccines prepared with human recombinant EGF linked to a carrier protein has been consistently evident in
our clinical trials in diverse adjuvanticity and vaccination schedule
settings. More than 80% of patients elicited some antiEGF antibody
response, with approximately 50% of good responders and a median
titer of 1:4000.
Such immunization could be achieved at a price of mild toxicity,
limited to nausea, vomiting, tremors, chills, cephalea, fever, cramps,
or hot flashes (NCICTC grade I or II) that disappeared after medication and without any
evidence of autoimmunity symptoms. The
absence of secondary events attributable to autoimmunity is remarkable, since autoimmunity is a potential risk of cancer vaccines that
include selfepitopes. In fact, severe and fatal diabetes, arteritis and
myocarditis have been described in transgenic mice immunized with
dendritic cells.28 The induction of vitiligo is a frequent observation
of melanoma vaccine trials,29,30 and severe autoimmune events have
been described recently in a cancer vaccine trial when vaccination was
combined with antiCTLA4 antibodies.31
The absence of autoimmune events in our EGFvaccine trials
could be explained by the existence of a comfortable dosewindow
between antitumor therapeutic dose and an autoimmunityinducing
dose, or by the more trivial fact that optimal immunization schedules
have not yet been found.
As in any other therapeutic intervention, optimal biological effects
should be evaluated, and we therefore explored new adjuvants and
therapeutic schedules. A
lthough the switch from alum to Montanide
as adjuvant and the introduction of lowdose cyclophosphamide
pretreatment improved the immunogenicity of the vaccine, the level
of antibody titers (1: 4000) and the fact that these titers lasted only
23 months without additional vaccination suggest that we are still
far from the optimal immunization schedule. In fact, antibody titers
100 times above the median value have been occasionally found in
some immunized patients (maximal observed titers 1:400,000).
Another consistent finding in all our trials was the relationship
between the immune response and survival, in that GARs consistently had significantly longer survival times.
Can this association be interpreted in terms of a therapeutic effect
of the vaccine? One cannot be certain until the ongoing randomized Phase 2b clinical trial has been completed. However, vaccinated
patients, either with or without antibody responses, had significantly
longer survival times than the concurrent control group. Although
not the product of randomization, this group of patients had features
12

that made it provisionally adequate for this comparison: compliance


with the same inclusion criteria, simultaneity in time and treatment
in the same hospital by the same physician staff. To make survival
data comparable between vaccinated patients and historical controls,
timezero for survival analysis was taken one month after the end
of any other oncospecific treatment, and control patients surviving
less than 51 days (the equivalent to the vaccination period) were
excluded. This exclusion of control patients with very short survival
times eliminates bias in favor to the vaccinated group. In fact, such
a procedure makes survival analysis between control and vaccinated
groups more homogeneous: neither patients not completing the first
5 vaccination doses nor controls not surviving the equivalent 51 days
period were included. The survival advantage of vaccinated patients
was still statistically significant inside the group of patients with PS
0 and 1.
Therefore, with the conservative interpretation imposed by the
limitations of a nonrandomized study, these data are highly suggestive of a therapeutic effect of vaccination with EGF.
The effect of vaccination was more evident in the age group below
60 years old, but the survival advantage of GARs was evident in all
age groups.
The confirmation of this effect in a randomized trial, and the
deeper exploration of dosedependence and scheduledependence in
diverse combinations with other treatments, are paths of development currently being pursued.
References
1. Finn OJ. Cancer vaccines: Between the idea and the reality. Nat Rev Immun 2003;
3:63041.
2. Rosenberg SA. Progress in human tumor immunology and immunotherapy. Nature 2001;
411:38084.
3. New Medicines in Development: Biotechnology. Survey 2002. Copyright by the
Pharmaceutical Research and Manufacturers of America, NW. Washington, DC: 2005.
4. Renkvist N, Castelli C, Robbins PF, Parmiani G.
A listing of human tumor antigens recognized by T cells. Cancer Immunol Immunother 2001; 50:315.
5. Disis ML, Gooley TA, Rinn K, Davis D, Piepkorn M, Cheever MA, Knutson KL, Schiffman
K. Generation of Tcell immunity to the Her2/neu protein after active immunization with
Her2/neu peptidebased vacines. J Clin Oncol 2002; 20:262432.
6. Van der Bruggen P, Zhang Y, Chaux P, Stroobant V, Panichelli C, Schultz ES, Chapiro J, Van
Den Eynde BJ, Brasseur F, Boon T. Tumorspecific shared antigenic peptides recognized by
human T cells. Immunol Rev 2002; 166:5164.
7. Gonzlez G, Crombet T, Catal M, Mirabal V, Hernandez JC, Gonzlez Y, Marinello P,
Guillen G, Lage A. A novel cancer vaccine composed of humanrecombinant epidermal
growth factor linked to a carrier protein: Report of a pilot clinical trial. Ann Oncol 1998;
9:43135.
8. Gonzlez G, Crombet T, Torres F, Catal M, Alfonso L, Osorio M, Neninger E, Garca B,
Mulet A, Perez R, Lage A. Epidermal growth factorbased cancer vaccine for nonsmall cell
lung cancer therapy. Ann Oncol 2003; 14:46166.
9. Crombet T, Neninger E, Catal M, Garca B, Leonard I, Martnez L, Gonzlez G, Prez R,
Lage A. Treatment of NSCLC patients with an EGFbased vaccine report of a phase I trial.
Cancer Biol Ther 2006; 5:1459.
10. Gonzlez G, Snchez B, Surez E, Beausoleil I, Prez O, Lastre M, Lage A. Induction of
immunerecognition of self epidermal growth factor (EGF): Effect on EGFbiodistribution
and tumor growth. Vaccine Res 1996; 5:23344.
11. Gonzlez G, Pardo OL, Snchez B, Garca JL, Beausoleil I, Marinello P, Gonzlez Y,
Domarco A, Guilln G, Prez R, Lage A. Induction of immune recognition of self epidermal
growth factor II: Characterization of the antibody response and the use of a fusion protein.
Vaccine Res 1997; 6:91100.
12. Guillen G, Silva R. Alvarez A, Coizeau E, Novoa L, Selman M, Morales J, Gonzlez J,
Musacchio A, del Valle J, Delgado M, Tamayo B, Caballero E, Fernndez JR, Herrera L.
Cloning and expression of a high molecular weigh protein (PM6) from the Neisseria meningitidis strain B:4PL15. Evaluation of the immunogenicity and bacterial activity of antibodies
raised against the recombinant protein. In CondeGonzalez CJ, Morse S, Rice P, et al, eds.
Pathology and Immunobiology of Neisseriaceae. Cuernavaca, Mexico: Instituto Nacional de
Salud Publica, 1994:83440.
13. Silva R, Selman M, Guillen G, Herrera L, Fernndez JR, Novoa LI, Morales J, Moreira V,
Gonzlez S, Tamargo B, del Valle JA, Caballero E, Alvarez A, Coizeau E, Cruz S, Musacchio
A. Nucleotide sequence coding for an outer membrane protein from Neisseria meningitides
and use of said sequence in vaccine preparations USA patent No.5286484, 1994.
14. In: Neter J, Wasserman W, Kutner MH, eds. Applied Linear Statistical Models. 4th ed.
Homewood, IL: Irwin, 1996.

Human Vaccines

2006; Vol. 3 Issue 1

Downloaded by [Universidad De Concepcion] at 12:21 29 November 2014

Vaccination with EGF for Lung Cancer Therapy


15. In: Fleming T, Harrington D, eds. Counting Processes and Survival Analysis. New York, NY:
Wiley, 1991.
16. Gonzlez G, Montero E, Leon K, Cohen IR, Lage A, Autoimmunization to Epidermal
Growth Factor, a component of the immunological homunculus. Autoimm Rev 202;
1:8995
17. Lage A, Crombet T, Gonzlez G. Targeting epidermal growth factor receptor signaling: Early
results and future trends in oncology. Ann Med 2003; 35:32736.
18. Rusch V, Baselga J, CordonCardo C, Orazem J, Zaman M, Hoda S, McIntosh J, Kurie
J, Dmitrovsky E. Differential expression of the epidermal growth factor receptor and its
ligands in primary nonsmall cell ling cancers and adjacent benign lung. Cancer Res 1993;
53:237985.
19. Mayer A, Takimoto M, Fritz E, Schekkander G, Kofler K, Ludwing H. The prognostic
significance of proliferating cell nuclear antigen, epidermal growth factor receptor and mdr
gene expression in colorectal cancer. Cancer 1993; 71:245460.
20. Grandis JR, Melhem MF, Gooding WE. Levels of TGFa and EGFR in head and neck
squamous cell carcinoma and patient survival. J Natl Cancer Inst 1998; 90:82432.
21. Kohler M, Janz I, Wintzer HO, Wagner E, Bauknecht T. The expression of EGF receptors,
EGFlike factors and cmyc in ovarian and cervical carcinomas and their potential clinical
significance. Anticancer Res 1989; 9:153747.
22. Prez R, Pascual MR, Macias A, Lage A. Epidermal growth factor receptors in human breast
cancer. Breast Cancer Res Treat 1984; 4:18993.
23. Fukoka M, Yano S, Giaccone G, Tamura T, Nakagawa K, Douillard JY, Nishiwaki Y,
Vansteenkiste J, Kudoh S, Rischin D, Eek R, Horai T, Noda K, Takata I, Smith E, Averbuch
S, Macleod A, Feyereislova A, Dong RP, Baselga J. Multiinstitutional randomized phase II
trial of gefitinib for previously treated patients with advanced nonsmall cell lung cancer (The
IDEAL 1 Trial) [corrected]. J Clin Oncol 2004; 22:481120.
24. Kris M, Natale RB, Herbst RS, Lynch Jr TJ, Prager D, Belani CP, Schiller JH, Kelly K,
Spiridonis H, Sandler A, Albain KS, Cella D, Wolf MK, Averbuch SD, Ochs JJ, Kay AC.
Efficacy of gefitinib, an inhibitor of the epidermal growth factor receptor tyrosine kinase,
in symptomatic patients with nonsmall cell lung cancer: A randomized trial. JAMA 2003;
290:214958.
25. PrezSoler R, Chachoua A, Hammond LA, Rowinsky EK, Huberman M, Karp D, Rigas
J, Clark GM, Santabarbara P, Bonomi P. Determinants of tumor response and survival with
erlotinib in patients with nonsmall cell lung cancer. J Clin Oncol 2004; 22:323847.
26. Robert F, Ezekiel MP, Spencer SA, Meredith RF, Bonner JA, Khazaeli MB, Saleh MN, Carey
D, LoBuglio AF, Wheeler RH, Cooper MR, Waksal HW. Phase I study of antiepidermal
growth factor receptor antibody cetuximab in combination with radiation therapy in
patients with advanced head and neck cancer. J Clin Oncol 2001; 19:323443.
27. Crombet T, Osorio M, Cruz T, Roca C, del Castillo R, Mon R, IznagaEscobar N,
Figueredo R, Koropatnick J, Rengifo E, Fernndez E, Alvarez D, Torres D, Ramos M,
Leonard I, Perez R, Lage A. Use of the humanized antiepidermal growth factor receptor
monoclonal antibody hR3 in combination with radiotherapy in the treatment of advanced
head and neck cancer. J Clin Oncol 2004; 22:164654.
28. Ludewig B, Ochsenbein AF, Odermatt B, Paulin D, Hengartner H, Zinkernagel RM.
Immunotherapy with dendritic cells directed against tumor antigens shared with normal
host cells results in severe autoimmune disease. J Exp Med 2000; 191:795804.
29. Slingluff Jr CL, Petroni GR, Yamschikov GV, Barnd DL, Eastham S, Galavotti H, patterson JW, Deacon DH, Hibbits S, Teates D, Neese PY, Grosh WW, ChianeseBullock
KA, Woodson EM, Wiernasz CJ, Merrill P, Gibson J, Ross M, Engelhard VH. Clinical and
immunologic results of a randomized phase II trial of vaccination using four melanoma peptides either administered in granulocytemacrophage colonystimulating factor in adjuvant
or pulsed on dendritic cells. Clin Oncol 2003; 21:401626.
30. Dudley ME, Wunderlich JR, Robbins PF, Yang JC, Hwu P, Schwartzebtruber DJ, Topalian
SL, Sherry R, Restifo NP, Hubicki AM, Robinson MR, Raffeld M, Duray P, Seipp CA,
RogersFreezer L, Morton KE, Mavroukakis SA, White DE, Rosenberg SA. Cancer regression and autoimmunity in patients after clonal repopulation with antitumor lymphocytes.
Science 2002; 298:85054.
31. Phan GQ, Yang JC, Dherry RM, Hwu P, Topalian SL, Schwaetzentruber DJ, Restifo NP,
Haworth LR, Seipp CA, Freezer LJ, Morton KE, Mavroukakis SA, Duray PH, Steinberg
SM, Allison JP, Davis TA, Rosenberg SA. Cancer regression and autoimmunity induced by
cytotoxic T lymphocytesassociated antigen 4 blockade in patients with metastatic melanoma. Proc Natl Acad Sci USA 2003; 100:837277.

www.landesbioscience.com

Human Vaccines

13

You might also like