scispace - formally typeset
Search or ask a question
Journal ArticleDOI

Targeted Therapies in Breast Cancer: Implications for Advanced Oncology Practice.

01 Jul 2014-Journal of the advanced practitioner in oncology (J Adv Pract Oncol)-Vol. 5, Iss: 4, pp 246-260
TL;DR: The clinical development and utilization of targeted therapies currently in use or in clinical trials are described, with a focus on considerations for the oncology advanced practitioner.
Abstract: The systemic therapeutic management of breast cancer has undergone significant transformation in the past decade. Without targeted therapies, conventional treatment with cytotoxic agents has reached the limit of its potential in terms of patient survival for most types of cancer. Enhanced understanding of the pathogenesis of tumor cell growth and metastasis has led to the identification of signaling growth pathways as targets for these directed therapies. Novel therapies targeted to HER2/neu, epidermal growth factor receptor (EGFR), vascular endothelial growth factor (VEGF), poly(ADP ribose) polymerase (PARP), mammalian target of rapamycin (mTOR), histone deacetylase (HDAC), the heat shock protein, and cyclin-dependent kinase (CDK) inhibitors have been developed and have demonstrated some efficacy in breast cancer. Recognition and management of the toxicities associated with targeted therapies is imperative. This review will describe the clinical development and utilization of targeted therapies currently in use or in clinical trials, with a focus on considerations for the oncology advanced practitioner.

Content maybe subject to copyright    Report

246
REVIEW
Targeted Therapies in Breast
Cancer: Implications for
Advanced Oncology Practice
LAURA BOURDEANU, PhD, and THEHAN LUU, MD
From The Sage Colleges, Troy, New York, and
City of Hope National Medical Center, Duarte,
California
Authors' disclosures of potential conflicts of
interest are found at the end of this article.
Correspondence to: Laura Bourdeanu, PhD,
The Sage Colleges, Department of Nursing, 65
1st Street, Troy, NY 12180. E-mail: lbourdeanu@
yahoo.com
© 2014 Harborside Press®
Abstract
The systemic therapeutic management of breast cancer has undergone
significant transformation in the past decade. Without targeted thera-
pies, conventional treatment with cytotoxic agents has reached the limit
of its potential in terms of patient survival for most types of cancer. En-
hanced understanding of the pathogenesis of tumor cell growth and me-
tastasis has led to the identification of signaling growth pathways as tar-
gets for these directed therapies. Novel therapies targeted to HER2/neu,
epidermal growth factor receptor (EGFR), vascular endothelial growth
factor (VEGF), poly(ADP ribose) polymerase (PARP), mammalian target
of rapamycin (mTOR), histone deacetylase (HDAC), the heat shock pro-
tein, and cyclin-dependent kinase (CDK) inhibitors have been developed
and have demonstrated some ecacy in breast cancer. Recognition and
management of the toxicities associated with targeted therapies is im-
perative. This review will describe the clinical development and utiliza-
tion of targeted therapies currently in use or in clinical trials, with a focus
on considerations for the oncology advanced practitioner.
J Adv Pract Oncol 2014;5:246–260
D
uring the past decade,
the systemic thera-
peutic management of
breast cancer has un-
dergone a significant transforma-
tion. Without targeted therapies,
conventional treatment with cyto-
toxic agents has maximized its po-
tential in terms of patient survival
for most types of cancer. Enhanced
understanding of the pathogenesis
of tumor cell growth and metasta-
sis has led to the identification of
signaling growth pathways as tar-
gets for these directed therapies.
ANTI-HER2/neu THERAPY
Trastuzumab
The HER2/neu oncogene, a
transmembrane tyrosine kinase re-
ceptor belonging to the epidermal
growth factor receptor (EGFR) fam-
ily, has been shown to be amplified
in up to 30% of human breast can-

247
TARGETED THERAPIES IN BREAST CANCER
REVIEW
cer cell lines (Slamon et al., 1987). Identification
of HER2/neu led to the development of trastu-
zumab (Herceptin), a humanized monoclonal an-
tibody of the IgG1 type directed against the extra-
cellular portion of human EGFR HER2/neu, and
revolutionized the management of both early and
advanced breast cancer. Pivotal phase II and III
clinical trials of trastuzumab given in combination
with chemotherapy to women with early-stage
and metastatic breast cancer (MBC) have dem-
onstrated that trastuzumab is associated with sig-
nificantly longer overall survival (OS), longer time
to tumor progression (TTP), and longer duration
of response (Slamon et al., 2004; Romond et al.,
2005; Piccart-Gebhart et al, 2005; Joensuu et al.,
2006; Robert et al., 2006; Pierga et al., 2010; Marty
et al., 2005; Inoue et al., 2010).
Ado-Trastuzumab Emtansine
Ado-trastuzumab emtansine (Kadcyla) is an
antibody-drug conjugate designed to combine the
biological activity of trastuzumab with targeted
delivery of a potent microtubule-disrupting agent,
DM1 (a maytansine derivative), to HER2/neu-
expressing cancer cells (Lewis Phillips et al.,
2008). In a phase I study, ado-trastuzumab emtan-
sine showed clinical activity in heavily pretreated
patients with HER2/neu-overexpressing meta-
static breast cancer (Krop et al., 2010).
The recommended dose for phase II trials was
determined to be 3.6 mg/kg every 3 weeks. The
phase II studies confirmed this strong activity in
patients with HER2/neu-positive MBC whose
disease progressed while receiving HER2/neu-
directed therapy or who were previously treated
with an anthracycline, a taxane, capecitabine,
lapatinib (Tykerb), and trastuzumab, with overall
response (OR) rates in the range of 23.9% to 39.5%
(Burris 3rd et al., 2011).
The open-label phase III trial (EMILIA)
comparing ado-trastuzumab emtansine vs.
capecitabine and lapatinib in HER2/neu-positive
locally advanced or metastatic breast cancer previ-
ously treated with trastuzumab and a taxane con-
firmed that ado-trastuzumab emtansine signifi-
cantly improved progression-free survival (PFS,
p < .001) and OS was not reached vs. 23.3 months
(p = .005) compared with capecitabine and lapa-
tinib (Blackwell et al., 2012).
The primary results from a phase III trial,
called the TH3RESA trial, showed that ado-
trastuzumab emtansine increased PFS in patients
whose cancer was inoperable or had recurred or
metastasized after several treatments including
trastuzumab and lapatinib and were treated with
ado-trastuzumab emtansine vs. physician’s choice
of treatment (6.2 vs. 3.3 months, respectively,
p < .0001). The interim analysis of OS showed a
trend in favor of ado-trastuzumab emtansine, but
it did not reach a level of statistically significant
benefit (Wildiers et al., 2013).
Pertuzumab
Pertuzumab (Perjeta) is a monoclonal anti-
body that binds to the dimerization domain of
HER2/neu and prevents receptor dimerization,
thus preventing HER2/neu-mediated intracel-
lular signaling (Franklin et al., 2004). Data from
a phase I trial demonstrated the dosage of pertu-
zumab to be > 5 mg/kg given every 3 weeks (Agus
et al., 2005).
A phase II trial in patients with HER2/neu-
negative disease suggested that pertuzumab had
some activity as a single agent; however, the ben-
efit was so limited that further investigation of
single-agent pertuzumab in unselected patients
with HER2/neu-negative disease was unwarrant-
ed (Gianni et al., 2010).
Another phase II trial assessed the ecacy and
safety profile of pertuzumab in combination with
trastuzumab in patients with HER2/neu-positive
breast cancer whose disease had progressed during
prior trastuzumab-based therapy. Patients received
trastuzumab weekly (4 mg/kg loading dose, then
2 mg/kg every week) or every 3 weeks (8 mg/kg
loading dose, then 6 mg/kg every 3 weeks) and per-
tuzumab every 3 weeks (840 mg loading dose, then
420 mg every 3 weeks). Treatment continued until
disease progression or excessive toxicity. Overall,
the combination of pertuzumab and trastuzumab
was well tolerated, and adverse events were mild to
moderate (Baselga et al., 2010).
A subsequent phase III trial (CLEOPATRA)
assessed the activity of pertuzumab in patients
with HER2/neu-positive adenocarcinoma of the
breast with locally recurrent or metastatic disease.
Patients were randomized (1:1) to receive docetax-
el, trastuzumab, and pertuzumab or docetaxel,

248
BOURDEANU and LUU
REVIEW
trastuzumab, and placebo. The median PFS in-
creased significantly by 6.1 months in the pertu-
zumab group (hazard ratio [HR] for disease pro-
gression or death, 0.62; 95% confidence interval
[CI] = 0.51–0.75; p < .001). The interim analysis of
OS data showed a strong trend toward a survival
benefit with pertuzumab/trastuzumab/docetax-
el therapy, although it did not reach significance
(Baselga & Swain, 2010).
Side Eects
Although there are similarities in the side-
eect profiles of all three of these drugs, there
are some adverse events that are unique to each
agent. The most common adverse reactions as-
sociated with trastuzumab include headache, di-
arrhea, nausea, chills, infection, congestive heart
failure, insomnia, cough, and rash (Robert et al.,
2006; Pierga et al., 2010; Marty et al., 2005; Inoue
et al., 2010). The most common side eects associ-
ated with pertuzumab are diarrhea, alopecia, neu-
tropenia, nausea, rash, and peripheral neuropathy.
Finally, the most common side eects associated
with ado-trastuzumab emtansine are thrombo-
cytopenia, epistaxis, eye-tearing/conjunctivitis
disorder, and elevated liver enzymes (Baselga et
al., 2010; Baselga & Swain, 2010; Agus et al., 2005;
Blackwell et al., 2012; Burris 3rd et al., 2011; Gianni
et al., 2010; Krop et al., 2010).
One of the most concerning side eects of
HER2/neu therapy is cardiac dysfunction or
failure. Cardiac toxicity occurs in 7% to 28%
of patients treated with trastuzumab alone or
in combination with anthracycline-based che-
motherapy, and in 1.2% of patients treated with
pertuzumab in combination with chemotherapy
(Agus et al., 2005; Baselga et al., 2010; Baselga &
Swain 2010; Gianni et al., 2010; Inoue et al., 2010;
Marty et al., 2005; Pierga et al., 2010; Robert
et al., 2006; Slamon et al., 2001; Wardley et al.,
2010). Anti-HER2/neu therapy–induced cardiac
failure may be severe, and in some cases associ-
ated with death.
Other concerning grade 3 side eects of anti-
HER2/neu therapy include neutropenia, leuko-
penia, thrombocytopenia, diarrhea, elevated liver
enzymes, and palmar-plantar erythrodysesthesia
(Agus et al., 2005; Baselga et al., 2010; Baselga &
Swain, 2010; Gianni et al., 2010; Inoue et al., 2010;
Marty et al., 2005; Pierga et al., 2010; Robert et al.,
2006; Slamon et al., 2001; Wardley et al., 2010).
These side eects have generally been observed
when the therapy is used in combination with oth-
er antineoplastic agents. Other less common and
grade < 3 side eects are listed in Table 1.
HER2 AND EGFR PATHWAY
INHIBITORS
Lapatinib
Lapatinib is a reversible dual EGFR/HER1 and
HER2 tyrosine kinase inhibitor (TKI) that acts in-
tracellularly, directly targeting the TK domains
of HER1 and HER2 and inhibiting the receptor
phosphorylation, leading to inhibition of down-
stream pathways that control cell proliferation
and survival (Tevaarwerk & Kolesar, 2009). The
combination of lapatinib and capecitabine showed
clinical activity in a phase I study of patients with
advanced solid tumors at a dose of 1,500 mg/day
(Chu et al., 2007).
Several phase II trials examined the ecacy
of lapatinib in HER2/neu-positive MBC patients
who failed to respond to trastuzumab therapy. The
OR rate was 4% to 8%, whereas 15% to 46% of pa-
tients had stable disease and 13% to 22% remained
progression-free at 16 weeks after treatment with
lapatinib (Burstein et al., 2008; Gajria et al., 2012;
Johnston et al., 2009; Jagiello-Gruszfeld et al.,
2010; Rugo et al., 2012).
The ecacy of lapatinib was evaluated in
phase III trials, which led to its US Food and Drug
Administration (FDA) approval in combination
with capecitabine and in combination with letro-
zole for HER2/neu-positive MBC (Blackwell et al.,
2010; Cameron et al., 2008; Di Leo et al., 2008).
Lapatinib, as a single agent or in combination
with capecitabine, was also assessed for the treat-
ment of brain metastases in patients with HER2/
neu-positive MBC. Lapatinib alone resulted in
objective CNS responses of 3% to 6%, while the
addition of capecitabine resulted in an objective
CNS response of 20% in patients who received
prior whole-brain radiation (Lin et al., 2008). Sev-
eral other studies of lapatinib plus capecitabine
reported response rates of 31.8% to 38.5% (Lin et
al., 2011; Sutherland et al., 2010). The combina-
tion of lapatinib plus capecitabine in patients with
HER2/neu-positive MBC who have not received

249
TARGETED THERAPIES IN BREAST CANCER
REVIEW
Table 1. Anti-HER2/neu Therapy
Agent Status Most common adverse events
Box warning or rare but
serious adverse events
Trastuzumab
a
FDA approved for the
treatment of HER2/neu-
overexpressing breast cancer
Fever, nausea, vomiting, infusion
reactions, diarrhea, infections,
increased cough, headache,
fatigue, dyspnea, rash, neutropenia,
anemia, and myalgia
CHF, significant decline
in left ventricular cardiac
function, severe infusion
reactions, pulmonary
toxicity
Ado-trastuzumab
emtansine
b
FDA approved for the
treatment of patients with
HER2/neu-positive MBC
who previously received
trastuzumab and a taxane,
separately or in combination
Fatigue, nausea, musculoskeletal
pain, thrombocytopenia, headache,
increased transaminases, and
constipation
Hepatotoxicity, left
ventricular dysfunction,
pulmonary toxicity,
infusion-related reactions,
hypersensitivity
reactions,
thrombocytopenia,
neurotoxicity
Pertuzumab
c
FDA approved in combination
with trastuzumab and
docetaxel for the treatment
of patients with HER2/neu-
positive MBC who have not
received prior anti-HER2/neu
therapy or chemotherapy for
metastatic disease
Diarrhea, alopecia, neutropenia,
nausea, fatigue, rash, PN
Left ventricular
dysfunction, infusion-
associated reactions,
hypersensitivity
reactions/anaphylaxis
Note. FDA = US Food and Drug Administration; CHF = congestive heart failure; MBC = metastatic breast cancer;
PN = peripheral neuropathy.
a
Information from Inoue et al. (2010), Marty et al. (2005), Pierga et al. (2010), Robert et al. (2006), Slamon et al. (2001),
Wardley et al. (2010).
b
Information from Blackwell et al. (2012), Burris 3rd et al. (2011), Krop et al. (2010).
c
Information from Agus et al. (2005), Baselga & Swain (2010), Baselga et al. (2010), Gianni et al. (2010).
whole-brain radiation resulted in an objective re-
sponse of 57% (Bachelot et al., 2013).
Neratinib
Neratinib, a highly selective irreversible inhib-
itor of the kinase activity of HER2/neu and EGFR,
showed antitumor activity as a single agent in pa-
tients with trastuzumab-pretreated MBC (Burst-
ein et al., 2010; Tsou et al., 2005). Phase I/II trials
evaluating the safety and ecacy of neratinib plus
vinorelbine or paclitaxel in HER2/neu-positive
MBC patients previously treated with anti-HER2/
neu therapy reported the maximum tolerated dose
(MTD) of neratinib to be 240 mg with promising
antitumor activity, with an OR rate of 57% and
71%, respectively, and no unexpected toxicities
(Chow et al., 2010; Awada et al., 2013).
Currently, neratinib is being studied in combina-
tion with temsirolimus (Torisel) in HER2/neu-posi-
tive or triple-negative MBC, as monotherapy vs. lapa-
tinib plus capecitabine in trastuzumab pretreated
HER2/neu-positive MBC, and in combination with
paclitaxel vs. paclitaxel plus trastuzumab in the first-
line treatment of HER2/neu-positive MBC (Clinical-
Trials.gov identifiers NCT01111825, NCT00777101,
and NCT00915018, respectively). Neratinib is also
being investigated in the adjuvant setting upon com-
pletion of trastuzumab-based therapy, as well as for
neoadjuvant treatment in locally advanced HER2/
neu-positive breast cancer (NCT01008150).
Afatinib
Afatinib is an irreversible dual inhibitor of
EGFR/HER1 and HER2 TKI (Minkovsky & Ber-
ezov, 2008). This first phase I study evaluated the
feasibility of oral dosing of afatinib in patients with
solid tumors for 14 days on followed by 14 days o
treatment and determined the MTD to be 70 mg
once daily (Eskens et al., 2008). Another phase I
trial assessing continuous administration of afatinib
in patients with solid tumors recommended a phase
II study dose of 50 mg once daily (Yap et al., 2010).
A phase II trial assessing the ecacy and
safety of afatinib in extensively pretreated pa-

250
BOURDEANU and LUU
REVIEW
tients with HER2/neu-negative MBC found that
afatinib had limited activity in HER2/neu-nega-
tive breast cancer (Schuler et al., 2012). Another
phase II trial evaluated afatinib monotherapy in
patients with HER2/neu-positive MBC after fail-
ure of trastuzumab treatment. Data demonstrated
11% of evaluable patients had a partial response,
37% had stable disease as best response, and 46%
achieved clinical benefit. Median PFS was 15.1
weeks and median OS was 61.0 weeks. The data
revealed that afatinib monotherapy has promising
clinical activity in extensively pretreated HER2/
neu-positive breast cancer patients whose disease
had progressed following trastuzumab treatment
(Lin et al., 2012).
Other ongoing phase II studies are currently
assessing the activity of afatinib in HER2/neu-pos-
itive MBC alone or in combination with a variety
of agents, such as vinorelbine, trastuzumab, lapa-
tinib, and letrozole (ClinicalTrials.gov Identifiers
NCT01325428 and NCT01531764, NCT01325428,
NCT00826267, NCT00708214, respectively),
in dierent settings, including brain metastases
and as neoadjuvant therapy (NCT01441596 and
NCT01594177, respectively). An ongoing phase
III study is comparing the addition of afatinib or
trastuzumab to vinorelbine in HER2/neu-pos-
itive MBC patients who progressed on or after
one prior trastuzumab-based treatment regimen
(NCT01125566).
Side Eects
The most commonly observed side eects
with all HER2/neu and EGFR inhibitors are acne-
like rash or folliculitis, diarrhea, and fatigue. Pal-
mar-plantar erythrodysesthesia, nausea, vomiting,
and fatigue were frequently seen with lapatinib.
Although not common, decreased left ventricular
ejection fraction, prolonged QT interval, and hep-
atotoxicity have been reported. Patients receiving
Table 2. HER2 and EGFR Pathway Inhibitors
Agent Status Adverse events
Box warning or
rare but serious
adverse events
Lapatinib
a
FDA-approved combination with (1)
capecitabine, for the treatment of patients
with advanced cancer or MBC whose
tumors overexpress HER2/neu and who
have received prior therapy including an
anthracycline, a taxane, and trastuzumab,
(2) letrozole for the treatment of
postmenopausal women with hormone
receptor–positive MBC that overexpresses
the HER2/neu receptor for whom hormonal
therapy is indicated
Diarrhea, PPE, nausea, rash,
vomiting, fatigue
Hepatotoxicity
Neratinib
b
Investigational Diarrhea, nausea, vomiting,
fatigue
Cardiotoxicity
Afatinib
c
Investigational Diarrhea, rash, fatigue Dehydration,
hyponatremia,
leukocytoclastic
vasculitis,
reduction in
cardiac LVEF,
acute renal
failure, reduced
general state
Note. FDA = US Food and Drug Administration; MBC = metastatic breast cancer; PPE = palmar-plantar
erythrodysesthesia; LVEF = left ventricular ejection fraction.
a
Information from Blackwell et al. (2010), Burstein et al. (2008), Cameron et al. (2008), Chu et al. (2007), Di Leo et al.
(2008), Gajria et al. (2012), Jagiello-Gruszfeld et al. (2010), Johnston et al. (2009), Rugo et al. (2012).
b
Information from Awada et al. (2013), Chow et al. (2010).
c
Information from Lin et al. (2012), Schuler et al. (2012), Yap et al. (2010).

Citations
More filters
Journal ArticleDOI
TL;DR: Identification of selected cancer-specific genomic alterations from cfDNA may be a noninvasive way to monitor disease progression, predict PFS, and offer targeted therapy.
Abstract: Due to the spatial and temporal genomic heterogeneity of breast cancer, genomic sequencing obtained from a single biopsy may not capture the complete genomic profile of tumors. Thus, we propose that cell-free DNA (cfDNA) in plasma may be an alternate source of genomic information to provide comprehensive data throughout a patient’s clinical course. We performed a retrospective chart review of 100 patients with stage 4 or high-risk stage 3 breast cancer. The degree of agreement between genomic alterations found in tumor DNA (tDNA) and cfDNA was determined by Cohen’s Kappa. Clinical disease progression was compared to mutant allele frequency using a two-sided Fisher’s exact test. The presence of mutations and mutant allele frequency was correlated with progression-free survival (PFS) using a Cox proportional hazards model and a log-rank test. The most commonly found genomic alterations were mutations in TP53 and PIK3CA, and amplification of EGFR and ERBB2. PIK3CA mutation and ERBB2 amplification demonstrated robust agreement between tDNA and cfDNA (Cohen’s kappa = 0.64 and 0.77, respectively). TP53 mutation and EGFR amplification demonstrated poor agreement between tDNA and cfDNA (Cohen’s kappa = 0.18 and 0.33, respectively). The directional changes of TP53 and PIK3CA mutant allele frequency were closely associated with response to therapy (p = 0.002). The presence of TP53 mutation (p = 0.0004) and PIK3CA mutant allele frequency [p = 0.01, HR 1.074 (95 % CI 1.018–1.134)] was excellent predictors of PFS. Identification of selected cancer-specific genomic alterations from cfDNA may be a noninvasive way to monitor disease progression, predict PFS, and offer targeted therapy.

50 citations


Cites background from "Targeted Therapies in Breast Cancer..."

  • ...Genome sequencing technology advancements have led to a deeper understanding of cancer biology, and they have enabled identification of cancer-specific genomic alterations for potential targeted therapies [1, 3]....

    [...]

Journal ArticleDOI
TL;DR: Anti‐EGFR family mAbs are shown to be one of the most promising approaches in cancer therapy for both haematologic malignancies and solid tumours.
Abstract: OBJECTIVES Monoclonal antibody-based of cancer therapy has been considered as one of the most successful therapeutic strategies for both haematologic malignancies and solid tumours in the last two decades. Epidermal growth factor receptor (EGFR) family signalling pathways play a key role in the regulation of cell proliferation, survival and differentiation. Hence, anti-EGFR family mAbs is one of the most promising approaches in cancer therapy. KEY FINDINGS Here, recent advances in anti-EGFR mAb including approved or successfully tested in preclinical and clinical studies have been reviewed. Although we focus on monoclonal antibodies against the EGF receptor, but the mechanisms underlying the effects of EGFR-specific mAb in cancer therapy, to some extend the resistance to existing anti-EGFR therapies and some therapeutic strategies to overcome resistance such as combination of mAbs on different pathways are briefly discussed as well. SUMMARY The EGFR family receptors, is considered as an attractive target for mAb development to inhibit their consecutive activities in tumour growth and resistance. However, due to resistance mechanisms, the combination therapies may become a good candidate for targeting EGFR family receptors.

32 citations


Cites background from "Targeted Therapies in Breast Cancer..."

  • ...[70] Anit-HER2 mAbs and derivatives Approved monoclonal antibodies Trastuzumab (HERCEPTIN) Trastuzumab is a humanized monoclonal antibody which binds to domain IV of HER2....

    [...]

  • ...[7,33,104] It is more effective when HER3 activates HER2....

    [...]

  • ...It is worth noting that Pertuzumab and trastuzumab bind to different domains of HER2.([71,84,86]) Pertuzumab inhibits HER2 dimerization with other activated HER-family members such as HER3....

    [...]

  • ...[82,83] Pertuzumab (PERJETA) Pertuzumab is a humanized monoclonal antibody which binds to domain II of HER2....

    [...]

  • ...Szymanska M et al. A combination of two antibodies recognizing nonoverlapping epitopes of HER2 induces kinase activity-dependent internalization of HER2....

    [...]

Journal ArticleDOI
Hong Chen1, Jingjing Wang1, Hong Yang1, Dan Chen1, Panpan Li1 
TL;DR: The finding that FOXM1 may be a downstream target gene of the Hh signaling pathway in cervical cancer provides a potential novel diagnostic and therapeutic target for cervical cancer.
Abstract: Forkhead box M1 (FOXM1) and hedgehog (Hh) signaling pathway are implicated in the formation and development of human tumors, including cervical cancer. Previous studies have indicated that FOXM1 may be a downstream target gene of the Hh signaling pathway, but their association in cervical cancer is largely unknown. In the present study, the expression of FOXM1 and Hh signaling molecules was evaluated by immunohistochemical analysis in a tissue microarray that contained 70 cervical cancer tissues and 10 normal cervical tissues. In addition, the association of these molecules with clinicopathological parameters, and the association between FOXM1 and various molecules involved in the Hh signaling pathway was investigated. The results indicated that FOXM1 and Hh signaling molecules were overexpressed in cervical cancer tissues. The protein expression levels of FOXM1, glioma-associated oncogene 1 (GLI1) and smoothened (SMO) correlated with the clinical stage of the tumors, while the protein expression levels of Sonic Hh (SHh), patched 1 (PTCH1) and GLI1 correlated with the pathological grade of the tumors. The expression levels of GLI1 were lower in tissues without lymph node metastasis than in tissues with lymph node metastasis. In addition, FOXM1 expression correlated with GLI1, SHh and PTCH1 expression in cancer tissues. These findings confirmed the participation of FOXM1 and the Hh signaling pathway in cervical cancer. Furthermore, the finding that FOXM1 may be a downstream target gene of the Hh signaling pathway in cervical cancer provides a potential novel diagnostic and therapeutic target for cervical cancer.

25 citations

Journal ArticleDOI
TL;DR: The results showed that LCL161 can induce multiple forms of programmed cell death in breast cancer cells, and may thus offer promise as an anticancer agent in diverse genotypic backgrounds.
Abstract: There is an urgent requirement for the development of novel targeted therapies to treat breast cancer, which is the most comment type of malignancy among women. The evasion of apoptosis is a hallmark of cancer, and is often due to the upregulation of inhibitor of apoptosis proteins (IAPs) in tumor cells. Second mitochondrial‑derived activator of caspase/direct IAP‑binding protein with low PI is a natural IAP antagonist, which is found in the mitochondrion; this protein has a motif, which binds to a surface groove on the baculovirus IAP repeat domains of the IAPs. In the present study, the effects of the LCL161 Smac mimetic, a small molecule IAP antagonist, on breast cell lines was examined. The results from MTT and colony formation assays demonstrated that LCL161 markedly inhibited the proliferation and induced the apoptosis of MDA‑MB‑231 and MCF‑7 cell lines. As determined by western blotting, cIAP1 was degraded in the breast cancer cells, which occurred in an LCL161‑dependent manner. Upon caspase activation, LCL161 treatment induced necroptosis, another form of programmed cell death. The downregulation of receptor‑interacting protein kinase‑1 via small interfering RNA protected the cells from LCL161‑induced necroptosis. Taken together, the results of the present study showed that LCL161 can induce multiple forms of programmed cell death in breast cancer cells, and may thus offer promise as an anticancer agent in diverse genotypic backgrounds.

15 citations


Cites background from "Targeted Therapies in Breast Cancer..."

  • ...As knowledge of the underlying molecular mechanisms responsible for tumor development and chemotherapeutic resistance has increased, contemporary treatment of breast cancer has entered an era of targeted therapies based on molecular typing, and examples of these molecular targeted therapeutics for breast cancer include tyrosine kinase inhibitors (TKIs), angiogenesis inhibitors and agents that perturb DNA repair (8)....

    [...]

References
More filters
Journal ArticleDOI
09 Jan 1987-Science
TL;DR: Amplification of the HER-2/neu gene was a significant predictor of both overall survival and time to relapse in patients with breast cancer, and had greater prognostic value than most currently used prognostic factors in lymph node-positive disease.
Abstract: The HER-2/neu oncogene is a member of the erbB-like oncogene family, and is related to, but distinct from, the epidermal growth factor receptor. This gene has been shown to be amplified in human breast cancer cell lines. In the current study, alterations of the gene in 189 primary human breast cancers were investigated. HER-2/neu was found to be amplified from 2- to greater than 20-fold in 30% of the tumors. Correlation of gene amplification with several disease parameters was evaluated. Amplification of the HER-2/neu gene was a significant predictor of both overall survival and time to relapse in patients with breast cancer. It retained its significance even when adjustments were made for other known prognostic factors. Moreover, HER-2/neu amplification had greater prognostic value than most currently used prognostic factors, including hormonal-receptor status, in lymph node-positive disease. These data indicate that this gene may play a role in the biologic behavior and/or pathogenesis of human breast cancer.

11,597 citations


"Targeted Therapies in Breast Cancer..." refers background in this paper

  • ...ANTI-HER2/neu THERAPY Trastuzumab The HER2/neu oncogene, a transmembrane tyrosine kinase receptor belonging to the epidermal growth factor receptor (EGFR) family, has been shown to be amplified in up to 30% of human breast can- cer cell lines (Slamon et al., 1987)....

    [...]

Journal ArticleDOI
TL;DR: The addition of trastuzumab to chemotherapy was associated with a longer time to disease progression, a higher rate of objective response, a longer duration of response, and a lower rate of death at 1 year.
Abstract: Background The HER2 gene, which encodes the growth factor receptor HER2, is amplified and HER2 is overexpressed in 25 to 30 percent of breast cancers, increasing the aggressiveness of the tumor. Methods We evaluated the efficacy and safety of trastuzumab, a recombinant monoclonal antibody against HER2, in women with metastatic breast cancer that overexpressed HER2. We randomly assigned 234 patients to receive standard chemotherapy alone and 235 patients to receive standard chemotherapy plus trastuzumab. Patients who had not previously received adjuvant (postoperative) therapy with an anthracycline were treated with doxorubicin (or epirubicin in the case of 36 women) and cyclophosphamide with (143 women) or without trastuzumab (138 women). Patients who had previously received adjuvant anthracycline were treated with paclitaxel alone (96 women) or paclitaxel with trastuzumab (92 women). Results The addition of trastuzumab to chemotherapy was associated with a longer time to disease progression (median, 7.4 ...

10,532 citations


"Targeted Therapies in Breast Cancer..." refers background in this paper

  • ...…diarrhea, elevated liver enzymes, and palmar-plantar erythrodysesthesia (Agus et al., 2005; Baselga et al., 2010; Baselga & Swain, 2010; Gianni et al., 2010; Inoue et al., 2010; Marty et al., 2005; Pierga et al., 2010; Robert et al., 2006; Slamon et al., 2001; Wardley et al., 2010)....

    [...]

  • ...2% of patients treated with pertuzumab in combination with chemotherapy (Agus et al., 2005; Baselga et al., 2010; Baselga & Swain 2010; Gianni et al., 2010; Inoue et al., 2010; Marty et al., 2005; Pierga et al., 2010; Robert et al., 2006; Slamon et al., 2001; Wardley et al., 2010)....

    [...]

  • ...…and in 1.2% of patients treated with pertuzumab in combination with chemotherapy (Agus et al., 2005; Baselga et al., 2010; Baselga & Swain 2010; Gianni et al., 2010; Inoue et al., 2010; Marty et al., 2005; Pierga et al., 2010; Robert et al., 2006; Slamon et al., 2001; Wardley et al., 2010)....

    [...]

  • ...Other concerning grade 3 side effects of antiHER2/neu therapy include neutropenia, leukopenia, thrombocytopenia, diarrhea, elevated liver enzymes, and palmar-plantar erythrodysesthesia (Agus et al., 2005; Baselga et al., 2010; Baselga & Swain, 2010; Gianni et al., 2010; Inoue et al., 2010; Marty et al., 2005; Pierga et al., 2010; Robert et al., 2006; Slamon et al., 2001; Wardley et al., 2010)....

    [...]

Journal ArticleDOI
14 Apr 2005-Nature
TL;DR: BRCA1 or BRCA2 dysfunction unexpectedly and profoundly sensitizes cells to the inhibition of PARP enzymatic activity, resulting in chromosomal instability, cell cycle arrest and subsequent apoptosis, illustrating how different pathways cooperate to repair damage.
Abstract: BRCA1 and BRCA2 are important for DNA double-strand break repair by homologous recombination, and mutations in these genes predispose to breast and other cancers. Poly(ADP-ribose) polymerase (PARP) is an enzyme involved in base excision repair, a key pathway in the repair of DNA single-strand breaks. We show here that BRCA1 or BRCA2 dysfunction unexpectedly and profoundly sensitizes cells to the inhibition of PARP enzymatic activity, resulting in chromosomal instability, cell cycle arrest and subsequent apoptosis. This seems to be because the inhibition of PARP leads to the persistence of DNA lesions normally repaired by homologous recombination. These results illustrate how different pathways cooperate to repair damage, and suggest that the targeted inhibition of particular DNA repair pathways may allow the design of specific and less toxic therapies for cancer.

5,650 citations


"Targeted Therapies in Breast Cancer..." refers background in this paper

  • ...PARP inhibitors have selective anticancer activity in BRCA1- and BRCA2-deficient cancers (Farmer et al., 2005; Bryant et al., 2005)....

    [...]

Journal ArticleDOI
TL;DR: Tastuzumab combined with paclitaxel after doxorubicin and cyclophosphamide improves outcomes among women with surgically removed HER2-positive breast cancer.
Abstract: Background We present the combined results of two trials that compared adjuvant chemotherapy with or without concurrent trastuzumab in women with surgically removed HER2-positive breast cancer. Methods The National Surgical Adjuvant Breast and Bowel Project trial B-31 compared doxorubicin and cyclophosphamide followed by paclitaxel every 3 weeks (group 1) with the same regimen plus 52 weeks of trastuzumab beginning with the first dose of paclitaxel (group 2). The North Central Cancer Treatment Group trial N9831 compared three regimens: doxorubicin and cyclophosphamide followed by weekly paclitaxel (group A), the same regimen followed by 52 weeks of trastuzumab after paclitaxel (group B), and the same regimen plus 52 weeks of trastuzumab initiated concomitantly with paclitaxel (group C). The studies were amended to include a joint analysis comparing groups 1 and A (the control group) with groups 2 and C (the trastuzumab group). Group B was excluded because trastuzumab was not given concurrently with paclit...

5,200 citations


"Targeted Therapies in Breast Cancer..." refers background in this paper

  • ...…with significantly longer overall survival (OS), longer time to tumor progression (TTP), and longer duration of response (Slamon et al., 2004; Romond et al., 2005; Piccart-Gebhart et al, 2005; Joensuu et al., 2006; Robert et al., 2006; Pierga et al., 2010; Marty et al., 2005; Inoue et al.,…...

    [...]

  • ...Pivotal phase II and III clinical trials of trastuzumab given in combination with chemotherapy to women with early-stage and metastatic breast cancer (MBC) have demonstrated that trastuzumab is associated with significantly longer overall survival (OS), longer time to tumor progression (TTP), and longer duration of response (Slamon et al., 2004; Romond et al., 2005; Piccart-Gebhart et al, 2005; Joensuu et al., 2006; Robert et al., 2006; Pierga et al., 2010; Marty et al., 2005; Inoue et al., 2010)....

    [...]

Journal ArticleDOI
TL;DR: One year of treatment with trastuzumab after adjuvant chemotherapy significantly improves disease-free survival among women with HER2-positive breast cancer.
Abstract: background Trastuzumab, a recombinant monoclonal antibody against HER2, has clinical activity in advanced breast cancer that overexpresses HER2. We investigated its efficacy and safety after excision of early-stage breast cancer and completion of chemotherapy. methods This international, multicenter, randomized trial compared one or two years of trastuzumab given every three weeks with observation in patients with HER2-positive and either node-negative or node-positive breast cancer who had completed locoregional therapy and at least four cycles of neoadjuvant or adjuvant chemotherapy. results Data were available for 1694 women randomly assigned to two years of treatment with trastuzumab, 1694 women assigned to one year of trastuzumab, and 1693 women assigned to observation. We report here the results only of treatment with trastuzumab for one year or observation. At the first planned interim analysis (median follow-up of one year), 347 events (recurrence of breast cancer, contralateral breast cancer, second nonbreast malignant disease, or death) were observed: 127 events in the trastuzumab group and 220 in the observation group. The unadjusted hazard ratio for an event in the trastuzumab group, as compared with the observation group, was 0.54 (95 percent confidence interval, 0.43 to 0.67; P<0.0001 by the log-rank test, crossing the interim analysis boundary), representing an absolute benefit in terms of disease-free survival at two years of 8.4 percentage points. Overall survival in the two groups was not significantly different (29 deaths with trastuzumab vs. 37 with observation). Severe cardiotoxicity developed in 0.5 percent of the women who were treated with trastuzumab. conclusions One year of treatment with trastuzumab after adjuvant chemotherapy significantly improves disease-free survival among women with HER2-positive breast cancer. (clinicaltrials.gov number, NCT 00045032.)

4,815 citations