You are on page 1of 9

Current Treatment Options in Oncology (2010) 11:3644

DOI 10.1007/s11864-010-0120-6

Lung Cancer

Exciting New Targets in Lung


Cancer Therapy: ALK, IGF-1R,
HDAC, and Hh
Joel W. Neal, MD, PhD*
Lecia V. Sequist, MD, MPH
Address
*Stanford Cancer Center, 875 Blake Wilbur Drive,
Stanford, CA 94305, USA.
E-mail: jwneal@stanford.edu

Springer Science+Business Media, LLC 2010

Opinion statement
The anaplastic lymphoma kinase (ALK) inhibitor crizotinib will become an integral
addition to the treatment of patients with non-small cell lung cancer (NSCLC) harboring genetic ALK translocations. The insulin-like growth factor receptor (IGF-1R)
monoclonal antibody figitumumab, while initially promising, appears to increase
toxicity and death in combination with chemotherapy in the treatment of patients
with NSCLC of squamous histology; therefore, clinical development of this class of
agents will need to proceed with caution. The histone deacetylation (HDAC) inhibitor
vorinostat did not demonstrate an improvement in overall survival (OS) compared with
placebo in a large randomized trial, but other agents in this class may have greater
selectivity and efficacy. Inhibitors of the hedgehog (Hh) signaling pathways have
some early clinical promise in both NSCLC and small cell lung cancer (SCLC), and larger
studies using these agents are eagerly anticipated.

Introduction
Lung cancer is still the leading cause of cancer-related
death in the United States despite extensive research
efforts and the development of new therapeutic
approaches [1]. However, ongoing research into the
molecular basis of lung cancer has yielded insight into
various molecular pathways that are deregulated during
the process of tumorigenesis. This article summarizes
four signaling pathways of emerging importance in

lung cancer: anaplastic lymphoma kinase (ALK), insulin-like growth factor receptor (IGF-1R), histone
deacetylation (HDAC), and hedgehog (Hh). Inhibitors
targeting these pathways have been used investigationally to treat both non-small cell lung cancer
(NSCLC) and small cell lung cancer (SCLC), and may
lead to novel therapeutic strategies to complement
traditional chemotherapy.

ALK
Anaplastic lymphoma kinase is an orphan member of the insulin
superfamily of receptor tyrosine kinases normally expressed only in
the central nervous system, small intestine, and testis [2, 3]. The
normal function of ALK is poorly understood, but many anaplastic

Exciting New Targets in Lung Cancer Therapy

Neal and Sequist

37

large cell lymphomas activate ALK oncogenic signaling via genetic


chromosomal translocation, by forming fusion proteins between the
ALK kinase domain and partner proteins such as nucleophosmin
(NPM) [4]. Aberrant ALK expression and signaling have also been
observed in solid tumors, including lung cancers. In a subset of
NSCLC, genetic translocation of two genes in the short arm of chromosome 2 leads to the expression of a fusion protein between the
N-terminal portion of echinoderm microtubule-associated protein-like
4 (EML4) and the C-terminal kinase domain of ALK [5]. This causes
aberrant activation of downstream oncogenic signaling pathways such
as MAP kinase, PI3-Kinase, and STAT, leading to cell proliferation,
invasion, and inhibition of apoptosis [6].
Anaplastic lymphoma kinase translocations in NSCLC were first
detected using a cDNA expression library from an NSCLC tumor to
transform 3T3 fibroblasts [5]. In the initial report, 5 out of 75 Japanese NSCLC tumor samples of unselected histologic subtype contained evidence of an EML4ALK gene fusion. The presence of the
gene fusion was mutually exclusive of mutations in epidermal growth
factor receptor (EGFR) and KRAS, which are other common oncogenic
driving mutations in NSCLC. In a subsequently reported series, tumor
samples that were being screened for EGFR mutations also underwent
ALK screening, resulting in a population weighted toward females,
Asian ethnicity, never/light smoking history, and adenocarcinoma
histology. This study identified 19/141 tumors containing an ALK
translocation (13%) [7], but in an unselected population of patients
with NSCLC, the frequency is likely closer to 35%, since ALK
translocations are associated with light smoking history and adenocarcinoma histologic subtype [8, 9]. However, ALK-mutated tumors
commonly have a unique signet ring cell histologic feature and are
generally less gender-associated than EGFR mutations [8]. Detection
of an ALK tyrosine kinase rearrangement is achieved through a variety
of techniques, including break-apart fluorescence in situ hybridization
(FISH) to detect the genetic translocation [7], multiplexed RT-PCR to
detect the fusion transcript [10], or immunohistochemistry (IHC)
using an antibody to detect aberrant ALK protein expression [11].
The development of tyrosine kinase inhibitors against the ALK kinase
allowed exploration of the hypothesis that tumors harboring the ALK
translocation are oncogene addicted to this signaling pathway. This
term describes the phenomenon in which tumors with an activated
oncogene depend on continued signaling for proliferation and survival. One example of this is in lung cancers harboring activating
mutations in the EGFR tyrosine kinase, in which disruption of EGFR
signaling results in widespread apoptosis of tumor cells [12]. By
analogy, it was believed that ALK-positive tumors may be sensitive to
ALK tyrosine kinase inhibitors, as they are insensitive to EGFR
tyrosine kinase inhibitors [7]. Patients with ALK-positive tumors
were enrolled on an expansion arm of a phase I clinical trial of an
oral ALK tyrosine kinase inhibitor, crizotinib (PF-02341066). The
maximum tolerated dose was determined to be 250 mg twice daily,
and major toxicities included nausea, vomiting, diarrhea, fatigue,
and transaminitis, as well as a characteristic visual disturbance when
transitioning from dark to light. In a preliminary report, among 82
patients with ALK-positive NSCLC treated in the expansion phase of
the phase 1 study, the response rate was 63% and the disease control
rate was 87% [13]. To date, the median progression-free survival
(PFS) and overall survival (OS) have not yet been reached, as the

38

Lung Cancer
majority of patients are still receiving treatment after a median
follow-up of 6.4 months.
Based on these promising results, a phase III registrational trial is
currently enrolling patients with metastatic NSCLC containing ALK
translocations. This trial randomizes 318 patients with ALK FISHpositive, advanced-stage NSCLC to second-line crizotinib vs pemetrexed or docetaxel, with the aim of demonstrating superior PFS in this
selected group of patients. In addition, a companion phase II singlearm study is enrolling patients who are ALK-positive but either (1)
ineligible for the phase III study or (2) developed progressive disease
on the chemotherapy arm of the phase III trial. In addition, a first-line
trial is under development for patients with previously untreated ALKpositive lung cancer. If this drug is established to be superior to
standard chemotherapy, it should become a new standard of care in
the fraction of patients with ALK-positive NSCLC. In addition, other
highly potent ALK tyrosine kinase inhibitors are in the early stages of
clinical development, and other targeted agents such as HSP-90
inhibitors may inhibit this pathway as well [14].

Insulin-like growth factor type 1 receptor


The IGF pathway is an ancient signaling system which is used for the
regulation of carbohydrate energy balance at the level of the organism,
as well as control of individual cell growth [15]. Signaling through the
IGF pathway occurs when the IGF ligands, IGF-1 or IGF-2, bind to the
extracellular domain of the IGF-1R receptor, which is modulated by
the presence of extracellular IGF binding proteins (IGFBPs) (for review
see Ref. [16]). Signals downstream of the IGF-1R tyrosine kinase
include the MAP kinase pathway, which activates cellular proliferation,
as well as the PI3K pathway, which inhibits apoptosis. Many normal
tissues and tumor cells express IGF receptors as well as the closely
related insulin receptor, which can form heterotetramers with IGF-1R,
leading to alterations in its signaling ability. Normally, circulating
IGF-1 and IGF-2 are produced by the liver in response to growth hormone (GH) stimulation, as well as in tissues in an autocrine and paracrine manner. There is mounting evidence from population-based
studies that higher circulating levels of IGF-1 can increase the risk of
breast and prostate cancer, and perhaps even lung cancer [1720].
Preclinical models have suggested that inhibition of this pathway may
potentiate chemotherapy, EGFR inhibitors, and even radiation effects in
NSCLC cell lines [2123]. Two classes of inhibitors are being developed
to target the IGF pathway in various tumor types: monoclonal antibodies to the IGF-1R extracellular domain and small molecule inhibitors of the IGF-1R tyrosine kinase domain.
A handful of IGF-1R antibodies are in various stages of clinical
development, including figitumumab (CP-751, 871), robatumumab
(R1507), cixutumumab (IMC-A12), dalotuzumab (MK0646), and
AMG479. A unique toxicity of this class of agents is hyperglycemia,
which results from inhibition of negative feedback to the pituitary
from IGF-1R signaling, with resultant hyperglycemia and insulin
resistance from growth hormone oversecretion, increased IGF-1 levels,
and increased IGFBP levels [18]. In lung cancer, figitumumab has
been tested most extensively. In a phase I dose-escalation trial in
combination with carboplatin and paclitaxel, 40% of patients with
NSCLC had a response, including one complete response [24]. In
a subsequent phase II trial, 156 patients were randomized to

Exciting New Targets in Lung Cancer Therapy

Neal and Sequist

39

chemotherapy alone or chemotherapy plus figitumumab. While the


addition of figitumumab appeared to increase the response rate in
both adenocarcinoma and squamous cell histology, the response rate
among patients with squamous cell histolology was an astonishing
78%, which was subsequently duplicated in an expansion cohort [25].
Based on these results, a large study randomized 681 patients with
non-adenocarcinoma histology to carboplatin and paclitaxel with and
without figitumumab. However, in December 2009, this phase III
study was prematurely terminated, due to a trend toward worse OS in
the group receiving figitumumab and chemotherapy (10.3 months vs.
8.5 months, hazard ratio (HR) 1.23, P = 0.051), perhaps due to an
increase in infection and cardiovascular events [26]. In a subgroup
analysis, patients with high levels of IGF-1 (>1 ng/ml) had a trend
toward improved survival and less toxicity from the combined treatment; however, IGF-1 levels did not appear to be predictive of
response in the phase II study [27].
Clinical development may continue for other indications such as refractory Ewings sarcoma, in which figitumumab appears to have single agent
activity [28], but the phase III trial with figitumumab in combination
with erlotinib in the second-line treatment of non-adenocarcinoma
NSCLC was closed in March 2010 due to futility. Clinical trials testing
other monoclonal IGF-1R antibodies both along and in combination are
ongoing in both NSCLC and SCLC.
The second class of IGF-1R inhibitors, the small molecule TKIs, are
less clinically developed. Unlike the monoclonal antibodies, which
specifically inhibit IGF-1R, these drugs generally inhibit both IGF-1R
and insulin receptor signals by binding to the ATP binding site in the
kinase domain. In theory, this may result in more severe hyperglycemic disturbances. In a preliminary report of toxicity from a phase I
trial with the small molecule IGF-1R inhibitor OSI-906, adverse events
included hyperglycemia, nausea, vomiting, fatigue, lethargy, and
diarrhea, but these were generally less than grade 3 [29]. Future
development of this drug and other molecules with a similar spectrum
of activity, such as BMS-754807, will hopefully include diseasespecific trials in lung cancer.

Histone deacetylases
Acetylation of histones is one of the many post-translational modifications, which occurs on these DNA-packaging proteins, which generally leads to increased accessibility of promoter regions and
increased transcription of genes in localized areas of chromosomes
[30]. This process is opposed by the histone deacetylase classes of
enzymes, which promote condensation of chromatin and repression
of gene expression. Small molecule HDAC inhibitors bind to the zinccontaining catalytic domain and have varying specificity against the
distinct classes of HDAC enzymes. Many cancers express high levels of
HDAC and appear to have an increased sensitivity to HDAC inhibitors
relative to normal cells [31]. Interestingly, the activity of many other
proteins involved in cancer is also modulated through acetylation,
such as p53 [32] c-MYC [33], and the molecular chaperone HSP90
[34]. Therefore, the pleiotropic mechanisms by which HDAC inhibitors exhibit antitumor effects likely include altering gene expression
and cell differentiation via histone acetylation, as well as modifying
the cell cycle and lowering the apoptotic threshold by changing the
acetylation state of other cellular proteins [31].

40

Lung Cancer
Multiple HDAC inhibitors are in clinical development to target a wide
variety of solid and hematologic malignancies, including vorinostat
(MK-0683/SAHA), romidepsin (FK-228/depsipeptide), entinostat
(SNDX-275/MS-275), and panobinostat (LBH589). Of these, the
development of vorinostat is most matured in lung cancer. Vorinostat
is FDA approved as oral monotherapy for the third-line treatment of
cutaneous T-cell lymphoma based on a response rate of 30% in
otherwise refractory patients [35]. The most common side effects were
diarrhea, fatigue, nausea, dysguesia, and thrombocytopenia, but were
generally mild. In lung cancer, a phase II trial of single agent vorinostat in 14 refractory patients with NSCLC demonstrated no objective responses, but disease was transiently stabilized in 8 patients [36].
Based on this, carboplatin and paclitaxel were combined with escalating doses of vorinostat was tested in patients with solid tumors, and
partial responses were observed in 10 of 19 patients with advancedstage NSCLC, which is higher than expected with chemotherapy alone
[37]. Similarly encouraging results were observed in a randomized
phase II study of 94 patients, in which 34% of patients receiving
chemotherapy plus vorinostat responded to treatment, as compared
with 12% of patients receiving chemotherapy and placebo, with a
trend toward improved survival in the vorinostat arm [38]. However,
the subsequent phase III randomized trial of 253 patients was terminated prematurely due to failure to demonstrate an improvement
in response rate, PFS, or OS in the vorinostat arm [39]. While this
represents a setback for this combination, other clinical trials are
ongoing in small cell lung cancer, and in NSCLC with vorinostat in
combination with other targeted agents. One particularly intriguing
combination is vorinostat plus erlotinib, an epidermal growth factor
receptor tyrosine kinase inhibitor (EGFR-TKI). The rationale for
combining HDAC inhibitors with EGFR-TKIs is based on preclinical
data that HDAC inhibition restores sensitivity to EGFR-TKIs after the
development of resistance, and also dramatically reduces the concentration of erlotinib necessary for cell death in a variety of NSCLC
cell lines [40, 41]. Entinostat and romidepsin are similarly being
tested in combination with erlotinib, and results of these trials may
yield different results as all of these agents have different activity
against individual HDAC proteins.

Hedgehog
The Hh signaling pathway was named for its role in development in
fruit flies, but it also remains highly evolutionarily conserved in
mammals. Hh signaling, represented in Fig. 1, is initiated by binding
of ligand to the transmembrane patched (Ptch) proteins, which normally repress the transmembrane smoothened (Smo) protein. Without the negative repression by Ptch, Smo can activate the Gli
transcription factor, resulting in activation of target genes important in
development and cancer, including IGFBP3, BCL2, N-MYC, and likely
many others [4244]. The connection between the Hh pathway and
cancer was first identified in patients with nevoid basal cell carcinoma
syndrome (Gorlin syndrome). These patients have a strong genetic
predisposition for basal cell cancer (BCC) and medulloblastoma,
caused by a loss-of-function mutation in the PTCH1 gene, resulting in
constitutive activation of Hh signaling after a second genetic hit to the
remaining functional allele [45]. Interestingly, most sporadic BCCs
also contain mutations which activate the Hh signaling [4648], and

Exciting New Targets in Lung Cancer Therapy

Hh

Ptch

Smo

Neal and Sequist

41

Cyc

Gli activation
Gene transcription

Figure 1. Schematic of Hedgehog signaling pathway. Activation of Hedgehog signaling is initiated by binding of

hedgehog ligand (Hh) to the transmembrane patched (Ptch) proteins. Ptch normally represses the transmembrane
smoothened (Smo) protein. Without the negative repression by Ptch, Smo can activate the Gli transcription
factor, resulting in activation of the transcription factor target genes important in development and cancer.
Cyclopamine (Cyc) and its derivatives bind to Smo and inhibit Hedgehog signaling
multiple solid and hematologic malignancies have dysregulation of
this pathway. In lung cancer, Ptch mutations have not been observed,
but ligand-dependent activation of the Hh pathway appears to be
important for the growth of many SCLC cell lines [49]. Transduction
of Hh signaling, as measured by Gli1 expression, is even more frequent in ex vivo NSCLC and SCLC tumor samples than in lung cancer
cell lines [50, 51], suggesting a potential role for a stromal paracrine
effect in these tumors [52]. There is also growing evidence that Hh
signaling is critical for survival of cancer stem cells, such that inhibitors of this pathway might kill the relatively small population of
chemotherapy-resistant self-renewing cells within a tumor [53].
There are a number of druggable targets within the Hh pathway.
Cyclopamine, a natural steroidal alkaloid product of the corn lily
plant, binds to Smo and inhibits downstream signaling [54]. A
number of cyclopamine derivatives with improved pharmacokinetic
properties have entered clinical development in lung cancer, including
GDC-0449, IPI-926, and BMS-833923/XL-139. Molecules which target multiple other steps in the Hh pathway have also been recently
identified [55, 56]. Of the cyclopamine derivatives, the most extensively studied is GDC-0449. This agent has elicited remarkable
responses in metastatic BCC and medulloblastoma [57, 58]. In lung
cancer, the Eastern Cooperative Oncology Group (ECOG) 1508 phase
II trial is enrolling patients with SCLC for treatment with cisplatin and
etoposide, in combination with either GDC0449 or the IGF-1R antibody cixutumumab. BMS-833923 is also being evaluated for safety in
a phase Ib trial in combination with carboplatin and etoposide in
SCLC. If the Hh inhibitors are indeed active against putative SCLC
stem cells, these agents might confer durable responses in a disease
that often rapidly becomes chemoresistant.

Conclusions
Research into the molecular mechanisms of tumorigenesis has yielded
the development of novel therapeutic agents which hopefully will
augment the standard regimens of chemotherapy in lung cancer. The
ALK inhibitors appear to be effective in a subset of NSCLC driven by
characteristic EML4-ALK translocations, which underscores the
importance of early testing of new drugs in molecularly selected

42

Lung Cancer
patient populations. Recent large trials of both the IGF-1R inhibitor
figitumumab and the HDAC inhibitor vorinostat failed to demonstrate a benefit in unselected patients with NSCLC, but other ongoing
trials with these and newer agents may help to identify a particular
subgroup of patients for whom this therapy is most appropriate.
Finally, the Hh inhibitors have demonstrated preclinical efficacy in the
treatment of SCLC, and trials are ongoing to determine whether these
agents might augment the efficacy of chemotherapy in this disease.
Hopefully over the next few years, novel pathway inhibitors will
become an integral part of our therapeutic arsenal in the battle against
lung cancer.

References and Recommended Reading


Papers of particular interest, published recently, have been highlighted as:

Of importance
Of major importance
1.

American Cancer Society: Cancer Facts & Figures 2009.


Atlanta: American Cancer Society; 2009.
2. Morris SW, Naeve C, Mathew P, et al.: ALK, the
chromosome 2 gene locus altered by the t(2;5) in
non-Hodgkins lymphoma, encodes a novel neural
receptor tyrosine kinase that is highly related to
leukocyte tyrosine kinase (LTK). Oncogene 1997,
14(18):21752188.
3. Iwahara T, Fujimoto J, Wen D, et al.: Molecular
characterization of ALK, a receptor tyrosine kinase
expressed specifically in the nervous system. Oncogene 1997, 14(4):439449.
4. Morris SW, Kirstein MN, Valentine MB, et al.: Fusion
of a kinase gene, ALK, to a nucleolar protein gene,
NPM, in non-Hodgkins lymphoma. Science 1994,
263(5151):12811284.
5. Soda M, Choi YL, Enomoto M, et al.: Identification of
the transforming EML4-ALK fusion gene in nonsmall-cell lung cancer. Nature 2007, 448(7153):561
566.
6. Amin HM, Lai R: Pathobiology of ALK+ anaplastic
large-cell lymphoma. Blood 2007, 110(7):22592267.
7. Shaw AT, Yeap BY, Mino-Kenudson M, et al.: Clinical
features and outcome of patients with non-small-cell
lung cancer who harbor EML4-ALK. J Clin Oncol
2009, 27(26):42474253.
8. Rodig SJ, Mino-Kenudson M, Dacic S, et al.: Unique
clinicopathologic features characterize ALK-rearranged lung adenocarcinoma in the western population. Clin Cancer Res 2009, 15(16):52165223.
9. Koivunen JP, Mermel C, Zejnullahu K, et al.: EML4ALK fusion gene and efficacy of an ALK kinase
inhibitor in lung cancer. Clin Cancer Res 2008,
14(13):42754283.
10. Takeuchi K, Choi YL, Soda M, et al.: Multiplex reverse
transcription-PCR screening for EML4-ALK fusion
transcripts. Clin Cancer Res 2008, 14(20):66186624.
11. Mino-Kenudson M, Chirieac LR, Law K, et al.: A novel,
highly sensitive antibody allows for the routine
detection of ALK-rearranged lung adenocarcinomas
by standard immunohistochemistry. Clin Cancer Res
2010, 16(5):15611571.

12.

Sharma SV, Fischbach MA, Haber DA, Settleman J:


Oncogenic shock: explaining oncogene addiction
through differential signal attenuation. Clin Cancer
Res 2006, 12(14 Pt 2):4392s4395s.
13. Bang Y, Kwak EL, Shaw AT, et al.: Clinical activity of
the oral ALK inhibitor PF-02341066 in ALK-positive
patients with non-small cell lung cancer (NSCLC). J
Clin Oncol 2010, 28(7s): Abstract 3.
This remarkable phase I clinical trial demonstrates that
patients with NSCLC and genetic translocation of the ALK
gene respond to crizotinib, an oral ALK tyrosine kinase
inhibitor.
14. Li R, Morris SW: Development of anaplastic lymphoma kinase (ALK) small-molecule inhibitors for
cancer therapy. Med Res Rev 2008, 28(3):372412.
15. De Meyts P: Insulin and its receptor: structure,
function and evolution. Bioessays 2004, 26(12):1351
1362.
16. Pollak M: Insulin and insulin-like growth factor signalling in neoplasia. Nat Rev Cancer 2008, 8(12):915
928.
This is a comprehensive review of the discovery of IGF and
the emerging role of this signaling pathway in cancer.
17. Schernhammer ES, Holly JM, Hunter DJ, Pollak MN,
Hankinson SE: Insulin-like growth factor-I, its binding proteins (IGFBP-1 and IGFBP-3), and growth
hormone and breast cancer risk in The Nurses
Health Study II. Endocr Relat Cancer 2006, 13(2):583
592.
18. Pollak MN, Schernhammer ES, Hankinson SE: Insulinlike growth factors and neoplasia. Nat Rev Cancer
2004, 4(7):505518.
19. Chan JM, Stampfer MJ, Ma J, et al.: Insulin-like
growth factor-I (IGF-I) and IGF binding protein-3 as
predictors of advanced-stage prostate cancer. J Natl
Cancer Inst 2002, 94(14):10991106.
20. Spitz MR, Barnett MJ, Goodman GE, Thornquist MD,
Wu X, Pollak M: Serum insulin-like growth factor
(IGF) and IGF-binding protein levels and risk of
lung cancer: a case-control study nested in the betaCarotene and Retinol Efficacy Trial Cohort. Cancer
Epidemiol Biomarkers Prev 2002, 11(11):14131418.

Exciting New Targets in Lung Cancer Therapy


21.

22.

23.

24.

25.

26.

27.

28.

29.

30.

31.

32.

33.

34.

Lee YJ, Imsumran A, Park MY, et al.: Adenovirus


expressing shRNA to IGF-1R enhances the chemosensitivity of lung cancer cell lines by blocking IGF-1
pathway. Lung Cancer 2007, 55(3):279286.
Guix M, Faber AC, Wang SE, et al.: Acquired resistance to EGFR tyrosine kinase inhibitors in cancer
cells is mediated by loss of IGF-binding proteins.
J Clin Invest 2008, 118(7):26092619.
Iwasa T, Okamoto I, Suzuki M, et al.: Inhibition of
insulin-like growth factor 1 receptor by CP-751, 871
radiosensitizes non-small cell lung cancer cells. Clin
Cancer Res 2009, 15(16):51175125.
Karp DD, Pollak MN, Cohen RB, et al.: Safety, pharmacokinetics, and pharmacodynamics of the insulin-like growth factor type 1 receptor inhibitor
figitumumab (CP-751, 871) in combination with
paclitaxel and carboplatin. J Thorac Oncol 2009,
4(11):13971403.
Karp DD, Paz-Ares LG, Novello S, et al.: Phase II study
of the anti-insulin-like growth factor type 1 receptor
antibody CP-751, 871 in combination with paclitaxel
and carboplatin in previously untreated, locally
advanced, or metastatic non-small-cell lung cancer.
J Clin Oncol 2009, 27(15):25162522.
Jassem J, Langer CJ, Karp DD, et al.: Randomized,
open label, phase III trial of figitumumab in combination with paclitaxel and carboplatin versus
paclitaxel and carboplatin in patients with nonsmall cell lung cancer (NSCLC). J Clin Oncol 2010,
28(7s):Abstract 7500.
Gualberto A, Dolled-Filhart MP, Hixon ML, et al.:
Molecular bases for sensitivity to figitumumab
(CP-751, 871) in NSCLC. J Clin Oncol 2009, 27(15s):
Abstract 8091.
Olmos D, Postel-Vinay S, Molife LR, et al.: Safety,
pharmacokinetics, and preliminary activity of the
anti-IGF-1R antibody figitumumab (CP-751, 871) in
patients with sarcoma and Ewings sarcoma: a phase
1 expansion cohort study. Lancet Oncol 2010,
11(2):129135.
Lindsay CR, Chan E, Evans TR, et al.: Phase I dose
escalation study of continuous oral dosing of OSI906, an insulin like growth factor-1 receptor (IGF-1R)
tyrosine kinase inhibitor, in patients with advanced
solid tumors. J Clin Oncol 2009, 27(18s): Abstract
2559.
Gregory PD, Wagner K, Horz W: Histone acetylation
and chromatin remodeling. Exp Cell Res 2001,
265(2):195202.
Schrump DS: Cytotoxicity mediated by histone
deacetylase inhibitors in cancer cells: mechanisms
and potential clinical implications. Clin Cancer Res
2009, 15(12):39473957.
Gu W, Roeder RG: Activation of p53 sequence-specific DNA binding by acetylation of the p53 C-terminal domain. Cell 1997, 90(4):595606.
Patel JH, Du Y, Ard PG, et al.: The c-MYC oncoprotein
is a substrate of the acetyltransferases hGCN5/PCAF
and TIP60. Mol Cell Biol 2004, 24(24):1082610834.
Kovacs JJ, Murphy PJ, Gaillard S, et al.: HDAC6 regulates Hsp90 acetylation and chaperone-dependent
activation of glucocorticoid receptor. Mol Cell 2005,
18(5):601607.

35.

36.

37.

38.

39.

40.

41.

42.

43.

44.

45.
46.

47.

48.

49.

Neal and Sequist

43

Olsen EA, Kim YH, Kuzel TM, et al.: Phase IIb multicenter trial of vorinostat in patients with persistent,
progressive, or treatment refractory cutaneous T-cell
lymphoma. J Clin Oncol 2007, 25(21):31093115.
Traynor AM, Dubey S, Eickhoff JC, et al.: Vorinostat
(NSC# 701852) in patients with relapsed non-small
cell lung cancer: a Wisconsin Oncology Network
phase II study. J Thorac Oncol 2009, 4(4):522526.
Ramalingam SS, Parise RA, Ramanathan RK, et al.:
Phase I and pharmacokinetic study of vorinostat, a
histone deacetylase inhibitor, in combination with
carboplatin and paclitaxel for advanced solid malignancies. Clin Cancer Res 2007, 13(12):36053610.
Ramalingam SS, Maitland ML, Frankel P, et al.: Carboplatin and Paclitaxel in combination with either
vorinostat or placebo for first-line therapy of
advanced non-small-cell lung cancer. J Clin Oncol
2010, 28(1):5662.
Belani C, Ramalingam S, Kalemkerian G, et al.: Randomized, double-blind phase II/III study of first-line
paclitaxel (P) plus carboplatin (C) in combination
with vorinostat or placebo in patients with advanced
non-small-cell lung cancer (NSCLC). Eur J Cancer
Suppl 2009, 7(2):507, Abstract O-9007.
Witta SE, Gemmill RM, Hirsch FR, et al.: Restoring
E-cadherin expression increases sensitivity to epidermal growth factor receptor inhibitors in lung
cancer cell lines. Cancer Res 2006, 66(2):944950.
Zhang W, Peyton M, Xie Y, et al.: Histone deacetylase
inhibitor romidepsin enhances anti-tumor effect of
erlotinib in non-small cell lung cancer (NSCLC) cell
lines. J Thorac Oncol 2009, 4(2):161166.
Ho L, Stojanovski A, Whetstone H, et al.: Gli2 and p53
cooperate to regulate IGFBP-3-mediated chondrocyte apoptosis in the progression from benign to
malignant cartilage tumors. Cancer Cell 2009,
16(2):126136.
Bigelow RL, Chari NS, Unden AB, et al.: Transcriptional regulation of bcl-2 mediated by the sonic
hedgehog signaling pathway through gli-1. J Biol
Chem 2004, 279(2):11971205.
Oliver TG, Grasfeder LL, Carroll AL, et al.: Transcriptional profiling of the Sonic hedgehog response: a
critical role for N-myc in proliferation of neuronal
precursors. Proc Natl Acad Sci USA 2003,
100(12):73317336.
Gorlin RJ: Nevoid basal cell carcinoma (Gorlin)
syndrome. Genet Med 2004, 6(6):530539.
Gailani MR, Stahle-Backdahl M, Leffell DJ, et al.: The
role of the human homologue of Drosophila patched in sporadic basal cell carcinomas. Nat Genet
1996, 14(1):7881.
Xie J, Murone M, Luoh SM, et al.: Activating
smoothened mutations in sporadic basal-cell carcinoma. Nature 1998, 391(6662):9092.
Reifenberger J, Wolter M, Knobbe CB, et al.: Somatic
mutations in the PTCH, SMOH, SUFUH and TP53
genes in sporadic basal cell carcinomas. Br J Dermatol
2005, 152(1):4351.
Watkins DN, Berman DM, Burkholder SG, Wang B,
Beachy PA, Baylin SB: Hedgehog signalling within
airway epithelial progenitors and in small-cell lung
cancer. Nature 2003, 422(6929):313317.

44
50.

51.

52.

53.

54.

Lung Cancer
Vestergaard J, Pedersen MW, Pedersen N, et al.:
Hedgehog signaling in small-cell lung cancer: frequent in vivo but a rare event in vitro. Lung Cancer
2006, 52(3):281290.
Yuan Z, Goetz JA, Singh S, et al.: Frequent requirement of hedgehog signaling in non-small cell lung
carcinoma. Oncogene 2007, 26(7):10461055.
Yauch RL, Gould SE, Scales SJ, et al.: A paracrine
requirement for hedgehog signalling in cancer.
Nature 2008, 455(7211):406410.
Zhao C, Chen A, Jamieson CH, et al.: Hedgehog signalling is essential for maintenance of cancer stem
cells in myeloid leukaemia. Nature 2009,
458(7239):776779.
Chen JK, Taipale J, Cooper MK, Beachy PA: Inhibition
of Hedgehog signaling by direct binding of cyclopamine to Smoothened. Genes Dev 2002,
16(21):27432748.

55.

Stanton BZ, Peng LF, Maloof N, et al.: A small molecule that binds Hedgehog and blocks its signaling
in human cells. Nat Chem Biol 2009, 5(3):154156.
56. Hyman JM, Firestone AJ, Heine VM, et al.: Smallmolecule inhibitors reveal multiple strategies for
Hedgehog pathway blockade. Proc Natl Acad Sci USA
2009, 106(33):1413214137.
57. Rudin CM, Hann CL, Laterra J, et al.: Treatment of
medulloblastoma with hedgehog pathway inhibitor
GDC-0449. N Engl J Med 2009, 361(12):11731178.
58. Von Hoff DD, LoRusso PM, Rudin CM, et al.: Inhibition of the hedgehog pathway in advanced basalcell carcinoma. N Engl J Med 2009, 361(12):1164
1172
Report of a phase I clinical trial demonstrating that basal
cell carcinoma respond dramatically to Hedgehog pathway
inhibitors.

You might also like