ArticlePDF AvailableLiterature Review

Novel targeted therapeutics: inhibitors of MDM2, ALK and PARP

Authors:

Abstract and Figures

We reviewed preclinical data and clinical development of MDM2 (murine double minute 2), ALK (anaplastic lymphoma kinase) and PARP (poly [ADP-ribose] polymerase) inhibitors. MDM2 binds to p53, and promotes degradation of p53 through ubiquitin-proteasome degradation. JNJ-26854165 and RO5045337 are 2 small-molecule inhibitors of MDM2 in clinical development. ALK is a transmembrane protein and a member of the insulin receptor tyrosine kinases. EML4-ALK fusion gene is identified in approximately 3-13% of non-small cell lung cancer (NSCLC). Early-phase clinical studies with Crizotinib, an ALK inhibitor, in NSCLC harboring EML4-ALK have demonstrated promising activity with high response rate and prolonged progression-free survival. PARPs are a family of nuclear enzymes that regulates the repair of DNA single-strand breaks through the base excision repair pathway. Randomized phase II study has shown adding PARP-1 inhibitor BSI-201 to cytotoxic chemotherapy improves clinical outcome in patients with triple-negative breast cancer. Olaparib, another oral small-molecule PARP inhibitor, demonstrated encouraging single-agent activity in patients with advanced breast or ovarian cancer. There are 5 other PARP inhibitors currently under active clinical investigation.
Content may be subject to copyright.
REVIEW Open Access
Novel targeted therapeutics: inhibitors of MDM2,
ALK and PARP
Yuan Yuan
1
, Yu-Min Liao
2
, Chung-Tsen Hsueh
1
and Hamid R Mirshahidi
1*
Abstract
We reviewed preclinical data and clinical development of MDM2 (murine double minute 2), ALK (anaplastic
lymphoma kinase) and PARP (poly [ADP-ribose] polymerase) inhibitors. MDM2 binds to p53, and promotes
degradation of p53 through ubiquitin-proteasome degradation. JNJ-26854165 and RO5045337 are 2 small-molecule
inhibitors of MDM2 in clinical development. ALK is a transmembrane protein and a member of the insulin receptor
tyrosine kinases. EML4-ALK fusion gene is identified in approximately 3-13% of non-small cell lung cancer (NSCLC).
Early-phase clinical studies with Crizotinib, an ALK inhibitor, in NSCLC harboring EML4-ALK have demonstrated
promising activity with high response rate and prolonged progression-free survival. PARPs are a family of nuclear
enzymes that regulates the repair of DNA single-strand breaks through the base excision repair pathway.
Randomized phase II study has shown adding PARP-1 inhibitor BSI-201 to cytotoxic chemotherapy improves
clinical outcome in patients with triple-negative breast cancer. Olaparib, another oral small-molecule PARP inhibitor,
demonstrated encouraging single-agent activity in patients with advanced breast or ovarian cancer. There are 5
other PARP inhibitors currently under active clinical investigation.
Introduction
Modern cancer therapeutics has evolved from non-spe-
cific cytotoxic agents that affect both normal and cancer
cells to targeted therapies and personalized medicine.
Targeted therapies are directed at unique molecular sig-
nature of cancer cells to produce greater efficacy with
less toxicity. The development and use of such thera-
peutics allow us to practice personalized medicine and
improve cancer care. In this review, we summarized pre-
clinical data and clinical development of three important
targeted therapeutics: murine double minute 2 (MDM2),
anaplastic lymphoma kinase (ALK) and poly [ADP-
ribose] polymerase (PARP) inhibitors.
Murine Double Minute 2
MDM2, also known as HDM2 in human, is a negative
regulator of tumor suppressor p53 [1]. MDM2 encodes
a90-kDaproteinwithap53bindingdomainattheN-
terminus, and a RING (really interesting gene) domain
at the C-terminus functioning as an E3 ligase responsi-
ble for p53 ubiquitylation [2]. When wild-type p53 is
activated by various stimuli such as DNA damage,
MDM2 binds to p53 at the N-terminus to inhibit the
transcriptional activation of p53, and promote the
degradation of p53 via ubiquitin-proteasome pathway
[3,4]. MDM2 is overexpressed in a variety of human
cancers, including melanoma, non-small cell lung can-
cer (NSCLC), breast cancer, esophageal cancer, leuke-
mia, non-Hodgkins lymphoma and sarcoma [5].
MDM2 can interfere with p53-mediated apoptosis and
growth arrest of tumor, which is the major oncogenic
activity of MDM2 [6,7]. Additionally, MDM2 can
cause carcinogenesis independent of p53 pathway [8].
In tumor with homozygous mutant p53, loss of
MDM2, which mimics the inhibition of the MDM2-
p53 interaction, can cause stabilization of mutant p53
and increased incidence of metastasis [9]. Overexpres-
sion of MDM2 has been shown to correlate positively
with poor prognosis in sarcoma, glioma and acute lym-
phocytic leukemia [10]. In NSCLC, there have been
conflicting results as to whether MDM2 overexpres-
sion is associated with worse or better prognosis, but
the subset analysis has demonstrated a poor prognostic
factor for early-stage NSCLC patients, particularly
those with squamous cell histology [11].
* Correspondence: hmirshah@llu.edu
1
Division of Medical Oncology and Hematology, Loma Linda University
Medical Center, Loma Linda, CA 92354, USA
Full list of author information is available at the end of the article
Yuan et al.Journal of Hematology & Oncology 2011, 4:16
http://www.jhoonline.org/content/4/1/16 JOURNAL OF HEMATOLOGY
& ONCOLOGY
© 2011 Yuan et al; licensee BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons
Attribution License (http://creative commons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in
any medium, pro vided the original work is properly cited.
Preclinical development of MDM2 inhibitors
Inhibition of MDM2 can restore p53 activity in cancers
containing wild-type p53, leading to anti-tumor effects
with apoptosis and growth inhibition [12-14]. Animal
studies have shown reactivation of p53 function can
lead to the suppression of lymphoma, soft tissue sar-
coma, and hepatocellular carcinoma [15-17]. Ventura et
al. have designed a reactivatable p53 knockout animal
model by a a Cre-loxP-based strategy, which a transcrip-
tion-translation stop cassette flanked by loxP sites (LSL)
is inserted in the first intron of the endogenous wild-
type p53 locus leading to silencing of p53 expression.
Cells from homozygous p53LSL/LSL mice are function-
ally equivalent to p53 null (p53-/-) cells, and p53LSL/
LSL mice are prone to develop lymphoma and sarcoma.
Due to the presence of flanking loxP sites, the stop cas-
sette can be excised by the Cre recombinase, which
causes reactivation of p53 expression and regression of
autochthonous lymphomas and sarcomas in mice [16].
These results have provided an encouraging direction
for p53-target therapeutic strategy utilizing inhibition of
MDM2. Since the interaction and functional relationship
between MDM2 and p53 have been well characterized,
small-molecule inhibitors of MDM2 have been devel-
oped by high-throughput screening of chemical libraries
[18-20]. As shown in table 1, there are three main cate-
gories of MDM2 inhibitors: inhibitors of MDM2-p53
interaction by targeting to MDM2, inhibitor of MDM2-
p53 interaction by targetingtop53,andinhibitorsof
MDM2 E3 ubiquitin ligase. The binding sites and
mechanism of action for these inhibitors are further illu-
strated in Figure 1.
Nutlins, consisting of nutlin 1, 2 and 3, analogs of cis-
imidazoline, fit in the binding pocket of p53 in MDM2
and inhibit the interaction between MDM2 and p53
[21,22]. Nutlin-3, an analog of the series, has the most
potent binding capacity and lowest inhibition concentra-
tion, induced p53 levels, and activated p53 transcrip-
tional activity [23]. Nutlin-3 has been shown to exhibit a
broad activity against various cancer models with wild-
type p53, such as breast, colon, neuroblastoma, mantle
cell lymphoma and osteosarcoma [24-27]. Nutlin-3 acti-
vates p53 and induces apoptosis and cellular senescence
in myeloid and lymphoid leukemic cells {Hasegawa,
2009 #149}. In the absence of functional p53, nutlin-3
interrupts the interaction between p73 and MDM2, and
increases p73 transcriptional activity, leading to
enhanced apoptosis and growth inhibition of leukemic
cell [30].
Table 1 MDM2 inhibitors in development
Chemical series Therapeutics Development stage
Inhibitors of MDM2-p53 interaction by targeting to MDM2
Cis-imidazoline RO5045337 (RG7112; Nutlin-3) Phase I:
Advanced solid tumors and hematological malignancy
Benzodiazepinedione TDP521252 & TDP665759 Preclinical
Spiro-oxindoles MI-219,
MI-319 & other MI compounds
Preclinical
Isoquinolinone PXN727 & PXN822 Preclinical
Inhibitor of MDM2-p53 interaction by targeting to p53
Thiophene RITA
(NSC 652287)
Preclinical
E3 Ligase Inhibitors
5-Deazaflavin HLI98 compounds Preclinical
Tryptamine JNJ-26854165 Phase I:
Advanced solid tumors
RITA, reactivation of p53 and induction of tumor cell apoptosis.
Reference; [23,36,37,130-132,134-139].
Figure 1 Schematic representation of the MDM2 and p53
proteins, and the binding areas for small-molecule inhibitors.
Nutlin, cis-imidazoline; TDP, benzodiazepinedione; MI, spiro-
oxindoles; PXN, isoquinolinone; HLI98, 5-deazaflavin; JNJ-26854165,
tryptamine; RITA, thiophene; RING, really interesting new gene
(signature domain of E3 ligase). Binding of either HLI98 or JNJ-
26854165 to RING domain of MDM2 can block the interaction of
ubiquitinated MDM2-p53 protein complex to the proteasome.
Yuan et al.Journal of Hematology & Oncology 2011, 4:16
http://www.jhoonline.org/content/4/1/16
Page 2 of 14
MDM4 (also known as MDMX), an MDM2 homolog,
binds p53 and inhibits p53 activity without causing
degradation of p53 degradation [31]. Furthermore,
despite the similarity between MDM2 and MDM4,
MDM2 inhibitors such as nutlin-3 are far less effective
against MDM4 [32]. Small-molecule inhibitor of MDM4
has been developed through a reporter-based drug
screening [33]. MDM4 inhibitor not only can activate
p53 and induce apoptosis in breast cancer MCF-7 cells,
but can also synergize with MDM2 inhibitor for p53
activation and induction of apoptosis.
Clinical development of MDM2 inhibitors
JNJ-26854165, a novel tryptamine derivative, is an oral
MDM2 inhibitor. Pre-clinical studies have shown bind-
ing of JNJ-26854165 to RING domain of MDM2 inhibits
the interaction of MDM2-p53 complex to the protea-
some, and increases p53 level [19]. Furthermore, induc-
tion of apoptosis and anti-proliferation independent of
p53 in various tumor models including breast cancer,
multiple myeloma and leukemia were shown [34-36].
The presence of p53-independent apoptotic activity in
addition to p53-mediated apoptosis is regarded as an
advantage to prevent the selection of p53 mutant sub-
clones in cancer during treatment of JNJ-26854165.
Results for phase I study (clinicaltrial.gov identifier:
NCT00676910) using continuously daily oral dosing in
patients with advanced solid tumors were presented in
2009 annual meeting of American Society of Clinical
Oncology (ASCO) [37]. Forty-seven patients were trea-
ted at 11 dose levels, ranging from 4 to 400 mg daily.
Treatment was well tolerated with frequent adverse
events in grade 1-2: nausea, vomiting, fatigue, anorexia,
insomnia, electrolyte imbalance, and mild renal/liver
function impairment. No hematological or cardiovascu-
lar toxicities were observed. One patient at 300 mg dose
level experienced dose-limiting toxicity (DLT) with
grade 3 asymptomatic QTc prolongation, which resolved
after discontinuation of treatment. Dose escalation was
stopped at 400 mg dose level due to 2 out of 3 patients
had DLT including one grade 3 skin rash and one grade
3 QTc prolongation. There was no objective response,
but 3 patients with prolonged SD including one breast
cancer overexpressing human epidermal growth factor
receptor 2. Pharmacokinetic study demonstrated linear
pharmacokinetics in 20 to 400 mg dose range, with pre-
clinical determined therapeutic concentration achieved
at dose level of 300 mg and above. Pharmacodynamic
study showed upregulation of p53 in skin, increase of
HDM2 levels in tumors, and increase of plasma macro-
phage inhibitory cytokine-1 (MIC-1) levels in dose-
dependent manner. MIC-1, a transforming growth fac-
tor-B superfamily cytokine, is induced by p53 activation,
and secreted MIC-1 levels can serve as a biomarker for
p53 activation [38]. Dose level of 350 mg was used on
expanded cohort of patients to confirm maximum toler-
ated dose, and trial with alternate dosing schedule to
minimize QTc prolongation was started with 150 mg
twice a day.
RO5045337 (RG7112), an oral formulation of nutlin-3,
is currently in phase I studies for patients with advanced
solid tumors (NCT00559533), and refractory acute leu-
kemias and chronic lymphocytic leukemia
(NCT00623870). Both studies are to determine the max-
imum tolerated dose and the optimal dosing schedule of
RO5045337, administered as monotherapy. Preliminary
data has shown acceptable safety profiles with responses
seen in patients with liposarcoma, acute myelogenous
leukemia and chronic lymphocytic leukemia.
Anaplastic Lymphoma Kinase
ALK is a 1620 amino acid transmembrane protein, con-
sisting of extracellular domain with amino-terminal sig-
nal peptide, intracellular domain with a
juxtamembranous segment harboring a binding site for
insulin receptor substrate-1, and a carboxy-terminal
kinase domain [39]. ALK is a member of the insulin
receptor tyrosine kinases, and the physiological function
of ALK remains unclear [40]. Translocation of ALK
occurs in about 50% of anaplastic large-cell lymphoma
(ALCL), and 80% of them have the t (2; 5) chromosomal
translocation with NPM-ALK expression [41]. The t (2;
5) translocation generates a fusion protein with carboxy-
terminal kinase domain of ALK on chromosome 2, and
the amino-terminal portion of nucleophosmin (NPM)
on chromosome 5. NPM is the most common fusion
partner of ALK, but at least six other fusion partners
have been identified. In these fusion proteins, the
amino-terminal portion is responsible for protein oligo-
merization, which activates ALK kinase and downstream
signaling such as Akt, STAT3, and extracellular signal-
regulated kinase 1 and 2 [42] (Figure 2).
Mutations of ALK have been identified in 6-12% of
sporadic neuroblastoma, and preclinical studies have
demonstrated these mutations promote ALK kinase
activity leading to oncogenic events [43]. It has been
postulated that activation of ALK provides oncogenic
addiction to tumors harboring activating mutation or
translocation of ALK [44]. Knock-down of ALK by
small hairpin RNA targeting ALK in NPM-ALK-con-
taining tumor models gives raise to growth inhibition
and apoptosis [45]. This indicates inhibition of ALK
may be an effective therapeutic strategy for tumors har-
boring ALK activation.
Echinoderm microtubule-associated protein-like 4
(EML4) is a 120 KDa cytoplasmic protein, which
involves in the formation of microtubules and microtu-
bule binding protein [46]. EML4-ALK is a novel fusion
Yuan et al.Journal of Hematology & Oncology 2011, 4:16
http://www.jhoonline.org/content/4/1/16
Page 3 of 14
gene arising from an inversion on the short arm of
chromosome 2 [Inv (2)(p21p23)] that joined exons 1-13
of EML4 to exons 20-29 of ALK [47,48]. Soda et al.
identified this fusion gene as a transforming activity in
mouse 3T3 fibroblasts from DNA of lung cancer in a
Japanese man with a smoking history in 2007 [48].
EML4-ALK fusion protein consists of the complete tyro-
sine kinase domain of ALK at and the carboxy-terminal,
and promoter of the 5partner gene controls the tran-
scription of the resulting fusion gene. Multiple variants
of EML4-ALK have been identified, and all the variants
encode the same cytoplasmic portion of ALK but differ-
ent truncations of EML4 (at exons 2, 6, 13, 14, 15, 18,
and 20). In lung cancer the chimeric protein involves
ALK fused most commonly but not exclusively to
EML4. Other rare fusion partners are TRK-fused gene
11 (TFG 11) and KIF5B (Kinesin heavy chain) [47-51].
ALK gene rearrangements and the resulting fusion pro-
teins in tumor specimen can be identified by fluorescent
in situ hybridization (FISH), immunohistochemistry
(IHC), and reverse transcription-polymerase chain reac-
tion (RT-PCR).
The presence of EML4-ALK fusion is identified in
approximately 3-13% of NSCLC, and mutually exclusive
with the presence of epidermal growth factor receptor
(EGFR) mutation [48,52-55]. EML4-ALK fusion
Figure 2 Schematic representation of the EML-4 and ALK translocation. A) Fusion of the N-terminal portion of EML4 (comprising the basic
region, the HELP domain and part of the WD-repeat region) to the intracellular region of ALK (including the tyrosine kinase domain). TM,
transmembrane domain. B) Both the ALK gene and the EML4 gene plot to chromosome 2p, but have opposite orientations. In the NSCLC EML4
is interrupted at a position 3.6 kb downstream of exon 13 and is attached to a position 297 bp upstream of exon 21 of ALK, creating the EML4-
ALK (variant 1) fusion gene.
Yuan et al.Journal of Hematology & Oncology 2011, 4:16
http://www.jhoonline.org/content/4/1/16
Page 4 of 14
transcript is not identified in other cancer types such as
gastrointestinal and breast cancers [56]. Shaw et al.
investigated the clinical features of NSCLC patients har-
boring EML4-ALK fusion rearrangement [55]. Among
141 patients, they found 19 (13%) patients carried the
EML4-ALK rearrangement, 31 (22%) harbored an acti-
vating EGFR mutation, and 91 (65%) were wild type for
both ALK and EGFR (designated WT/WT). EML4-
ALK-positive patients were significantly younger than
patients with either EGFR mutation or WT/WT (P <
0.001 and P = 0.005, respectively). EML4-ALK-positive
patients were more likely to be men than patients with
either EGFR mutation or WT/WT (P = 0.036 and P =
0.039, respectively). EML4-ALK-positive patients were
significantly never or light smokers compared with the
WT/WT patients (P < 0.001), and did not benefit from
treatment with EGFR tyrosine kinase inhibitors (TKIs).
Eighteen EML4-ALK-positive patients had adenocarci-
noma and one patient had mixed adenosquamous his-
tology.However,patientswithEML4-ALK-positive
NSCLC did not have exclusively adenocarcinoma histol-
ogy in two other studies [51,53].
Focusing on the clinical outcome, Shaw et al. exam-
ined 477 NSCLC patients, and identified 43 patients
(9%) with EML4-ALK rearrangements, 99 patients (21%)
with EGFR mutations, and 335 patients (70%) with WT/
WT [57]. EML4-ALK-positive patients were significantly
younger (median age 54 vs 64 years old, p < 0.001) and
more likely to be never or light smokers (90% vs 37%, p
< 0.001), compared with WT/WT patients. There was
no difference in overall survival (OS) between patients
with EML4-ALK fusion and EGFR mutation (1-year OS:
82% vs 81%, p = 0.79); however, both groups demon-
strated a longer OS than WT/WT patients (1-year OS
66%, p < 0.001). This data suggests the better outcome
in patients with EML4-ALK rearrangement vs. patients
with WT/WT may be related to differences in biology,
demographic features, and availability of targeted
therapies.
Preclinical development of ALK inhibitors
The development of ALK small-molecule inhibitors has
been hampered due to lack of ALK protein structure.
Initial testing and development of ALK inhibitors were
done with naturally occurring sources such as stauros-
porine and HSP90 inhibitors, which are not potent and
specific inhibitors of ALK [58]. Subsequently, using
homology modeling to assist the screening and synth-
esis, more potent and specific ALK inhibitors have been
developed [59]. Although there are multiple partners for
the ALK translocation, all the fusion proteins contain
the ALK kinase domain and should be susceptible to
ALK kinase inhibition. As shown in table 2, there are at
least 9 different chemical classes of small-molecule inhi-
bitors of ALK being developed.
PF-2341066 (Crizotinib), derivative of aminopyridine,
was initially developed as a potent, orally bioavailable,
ATP-competitive small-molecule inhibitor of c-MET
and hepatocyte growth factor receptor [60]. Further
investigation has indicated Crizotinib is a potent inhibi-
tor of ALK as well, and half maximal inhibitory concen-
tration (IC
50
) for either c-MET or ALK overexpressing
cell line is ~20 nM. Crizotinib suppresses the prolifera-
tion of ALCL cell line with ALK activation, but not in
ALCL cell lines without ALK activation. Crizotinib inhi-
bits phosphorylation of ALK, and causes complete
regression of ALCL harboring NPM-ALK fusion in
xenograft model [61]. Crizotinib also inhibits the prolif-
eration in NSCLC (such as H3122) and neuroblastoma
cell lines harboring ALK activation [62]. Experiments
Table 2 ALK inhibitors in development
Chemical series Therapeutics Development stage
Aminopyridine PF-2341066 (crizotinib) Phase II/III: NSCLC; phase I/II: advanced solid tumors, neuroblastoma, and ALCL
Diaminopyrimidine CEP-28122 Preclinical
IND application expected
Structure undisclosed AP-26113 Preclinical
IND application expected in 2011
Structure undisclosed X-276 Preclinical
Pyridoisoquinoline F91873 and F91874 Preclinical
Pyrrolopyrazole PHA-E429 Preclinical
Indolocarbazole CEP-14083 and CEP-14513 Preclinical
No further development
Pyrrolopyrimidine GSK1838705A Preclinical
No further development
Dianilinopyrimidine NVP-TAE684 Preclinical
No further development
NSCLC, non-small cell lung cancer; ALCL, anaplastic large cell lymphoma
Reference [60,61,64,65,140-150]
Yuan et al.Journal of Hematology & Oncology 2011, 4:16
http://www.jhoonline.org/content/4/1/16
Page 5 of 14
using NCI-H441 NSCLC xenografts showed a 43%
decrease in mean tumor volume, with 3 of 11 mice
exhibiting a >30% decrease in tumor mass and 3 animals
with no evidence of tumor at the end of the 38-day cri-
zotinib treatment [60]. Crizotinib is currently under-
going active clinical investigation in NSCLC.
Additionally, phase I/II study is conducted in patients
with advanced malignancy such as ALCL or neuroblas-
toma (NCT00939770).
Second-generation ALK inhibitors such as AP-26113
and X-276 are considered more potent and selective
inhibitors of ALK than crizotinib. AP-26113, an orally
bioavailable inhibitor of ALK with undisclosed structure,
is developed by Ariad [63]. During preclinical investiga-
tion, AP-26113 has been shown to inhibit not only the
wild-type ALK but also mutant forms of ALK, which
are resistant to the first-generation ALK inhibitor such
as crizotinib. Further studies have demonstrated AP-
26113 is at least 10-fold more potent and selective in
ALK inhibition than crizotinib [64,65].
Clinical development of ALK inhibitors
In 2009 annual meeting of ASCO, Kwat et al. reported
the results of phase I dose escalation study and
expanded phase II study of crizotinib [66]. Thirty-seven
patients with advanced solid tumors including 3 NSCLC
patients were enrolled in phase I study. The maximum
tolerated dose of crizotinib was 250 mg orally twice a
day,and2fatigueDLTwerenotedinthenextdose
level at 300 mg twice a day. The major side effects were
fatigue, nausea, vomiting and diarrhea; but were man-
ageable and reversible. There was 1 partial response
(PR) in a sarcoma patient with ALK rearrangement.
Additionally, a dramatic clinical response was observed
in a NSCLC patient harboring EML4-ALK rearrange-
ment. Therefore, an expanded phase II study using 250
mg of crizotinib twice a day was conducted in 27
NSCLC patients harboring EML4-ALK tumor deter-
mined by FISH. In the first 19 evaluable patients, there
were 17 patients with adenocarcinoma (90%) and 14
non-smokers (74%). Overall response rate (RR) was 53%,
and disease control rate (DCR; complete response [CR]/
PR/stable disease [SD]) was 79% at 8 weeks. Only 4
patients (21%) progressed after 8 weeks of treatment,
despite more than 60% of patients received 2 or more
lines of treatment prior to entering this study.
Bang et al. presented the follow up results on the
expanded phase II study of crizotinib in NSCLC patients
with EML4-ALK rearrangement in 2010 annual meeting
of ASCO [67]. Eighty-two patients were evaluable, with
96% adenocarcinoma, 76% never-smokers and ~95%
having prior treatment. Overall RR was 57%, with esti-
mated 6-month progression-free survival (PFS) rate of
72%, and DCR of 87% at 8 weeks. The median
progression-free survival was not yet mature, and the
median duration of treatment was 25.5 weeks. Radiolo-
gical responses typically were observed at the first or
second restaging CT scan. Main side effects were nau-
sea, diarrhea and visual disturbance on light/dark
accommodation without abnormality on eye examina-
tion. The results of this phase II study have been
recently published [68].
Based on these encouraging results, a randomized
phase III trial comparing crizotinib to standard second-
line cytotoxic chemotherapy docetaxel and pemetrexed
in patients with ALK-positive NSCLC has now com-
menced (NCT00932893). The combination of erlotinib
and crizotinib is also being tested in patients who failed
prior chemotherapies regardless of EML4/ALK translo-
cation status (NCT00965731). A phase III study to eval-
uate crizotinib as first line therapy in EML4-ALK
translocation patients compare to standard platinum
based chemotherapy is underway (NCT01154140).
Poly ADP-Ribose Polymerases
PARPs are a family of nuclear enzymes that regulates
the repair of DNA single-strand breaks (SSBs) through
the base-excision repair (BER) pathway [69]. Upon DNA
damage, PARP cleaves nicotinamide adenine dinucleo-
tide (NAD
+
) to generate poly (ADP-ribose) (PAR) poly-
mers, which are added onto DNA, histones, DNA repair
proteins and PARP [70,71]. These hetero- and auto-
modification processes mediated by PARP lead to
recruitment of repair machinery to facilitate BER pro-
cess. Among the 17 members of PARP, PARP-1 and
PARP-2 are the only members known to be activated by
DNA damage and may compensate for each other [72].
PARP-1 is best characterized and responsible for most if
not all the DNA-damage-dependent PAR synthesis;
exhibits with N-terminal DNA-binding domain, central
auto-modification domain, and C-terminal catalytic
domain, which is the signaturefor PARP family.
Although lacks of central auto-modification domain,
PARP-2 shares ~70% homology of catalytic domain as
PARP-1, and provides residual PARP activity (~15%) in
the absence of PARP-1 [73]. The physiological functions
of PARP-1 and PARP-2 have been further explored in
knockout models [74]. Double PARP-1 and PARP-2
knockout mice are lethal at the embryonic stage. Knock-
out of either PARP-1 or PARP-2 results in increased
genomic instability by accumulation of DNA SSBs, and
causes hypersensitivity to ionizing radiation and alkylat-
ing agents. Additionally, PARP-1 also plays important
roles in cellular responses to ischemia, inflammation
and necrosis.
Targeting the PARP-mediated DNA repair pathway is
a promising therapeutic approach for potentiating the
effects of chemotherapy and radiation therapy and
Yuan et al.Journal of Hematology & Oncology 2011, 4:16
http://www.jhoonline.org/content/4/1/16
Page 6 of 14
overcoming drug resistance [75]. However, the most
exciting use of PARP inhibitors may be utilizing a phe-
nomenon called synthetic lethality [76]. Synthetic lethal-
ity is a cellular condition in which simultaneous loss of
two nonessential mutations results in cell death, which
dose not occur if either gene products is present and
functional [77]. Tumors with DNA repair defects, such
as those arising from patients with BRCA mutations
were found to be more sensitive to PARP inhibition due
to synthetic lethality. The BRCA1 and BRCA2 gene
encodes large proteins that coordinate the homologous
recombination repair double strand breaks (DSBs) path-
way [78]. Since BRCA1/2-mutated tumors cannot utilize
homologous recombination to repair DSBs, exposing
these cells to PARP inhibitor, which shuts down BER
rescue pathway, will lead to accumulation of DNA
damage, genomic instability and cell death (Figure 3).
Preclinical development of PARP inhibitors
Inhibition of PARP has been developed in the laboratory
for more than 30 years, with analogues mimicking nicoti-
namide component of NAD
+
for binding to catalytic site
of PARP [70,79]. Preclinical data reporting efficacy of
PARP inhibitors in a BRCA mutated population was initi-
ally reported in 2005 [80,81]. Bryant et al. revealed that
low concentrations of PARP inhibitors produced cyto-
toxicity on BRCA2-deficient cell lines with defects in
homologous recombination, but not in cell lines with
intact homologous recombination. When BRCA2 func-
tion was restored in these cell lines, the cells were no
longer subject to inhibition of PARP. In other breast can-
cer cell lines such as MCF-7 and MDA-MB-231, similar
sensitivity to PARP inhibition was observed when
BRCA2 was depleted. Similarly, Farmer et al. demon-
strated that PARP inhibitors NU1025 and AG14361 were
highly cytotoxic in BRCA2-deficient VC-8 cells [80].
Additionally, cell death increased when BRCA1/2 defi-
cient cells were transfected with small interfering RNA
targeting PARP-1. Enhanced sensitivity to PARP inhibi-
tion in BRCA-deficient cells was observed when DNA-
damaging agents were added in vitro. These preclinical
data serve as proof-of-concept for synthetic lethality in
BRCA-deficient cell lines and provide important rationale
for studying PARP inhibitors in patients with BRCA1/2-
asssociated breast and ovarian cancer.
Further investigations have identified triple-negative
breast cancer (TNBC, breast cancer without over-expres-
sion of estrogen, progesterone and HER2-neu receptors,
accounts for about 15 percent of all breast cancers) and
sporadic serous ovarian cancer without mutations of
BRCA1/2 but exhibit properties of BRCA1- or BRCA2-
deficient cells, known as BRCAness[82]. BRCAness
cancers have defects in homologous recombination due
to dysfunctional BRCA1/2 from epigenetic modification,
and/or deficiency in proteins involved in homologous
recombination repair pathways, such as RAD51, RAD54,
DSS1, RPA1, ATM, CHK2 and PTEN [83-85]. Preclinical
studies have shown BRCAness cancer cells are more sen-
sitive to PARP inhibition especially in the presence of
DNA-damaging agents such as cisplatin, vs. non-BRCA-
ness [86]. These important findings have further
expanded the therapeutic application of PARP inhibitors
in cancers with acquired defect in homologous recombi-
nation other than germline BRCA mutations.
As shown in table 3, there are currently 9 different
PARP inhibitors at different stages of clinical development,
and at least 3 highly selective PARP inhibitors in preclini-
cal development. Because both PARP-1 and PARP-2 share
high degree of homology in catalytic domain, most of the
PARP inhibitors under clinical development do not have
significant differential activity against either PARP-1 or
PARP-2 [69]. Using x-ray crystal structure and homology
Figure 3 Schematic representation of PARP and BRCA
mediated DNA repair in cells without exposure to PARP
inhibitor and BRCA mutation (A), and synthetic lethality in cells
with BRCA mutation exposing to PARP inhibitor (B).
Yuan et al.Journal of Hematology & Oncology 2011, 4:16
http://www.jhoonline.org/content/4/1/16
Page 7 of 14
modeling, highly selective inhibitors against either PARP-1
or PARP-2 have been successfully developed [87-89].
Over-activation of PARP-1 due to DNA damage from
ischemic event is responsible for post-ischemic cell death
in neurons and myocardial cells, and PARP-1 knockout
mice are more resistant to the damage from ischemic
insults [90,91]. PARP inhibitors such as INO-1001 and
MP-124 have been studied in animal models and clinical
settings as neuroprotectant and cardiac protectant during
ischemic insults [92-94].
PARP-5a and PARP-5b, also known as tankyrase 1
and tankyrase 2, are involved in telomere metabolism
and Wnt/b-catenin signaling [69]. Moreover, tankyrase
inhibition imposes selective lethality on BRCA deficient
cell lines [95]. XAV939, a small molecule which sup-
presses b-catenin-mediated transcription by stabiling
axin and degrading b-catenin, is found to inhibit tan-
kyrases [96]. Molecule like XAV939 can be used to tar-
get cancers harboring BRCA (such as breast cancer)
and/or dysregulated Wnt-b-catenin signaling (such as
colorectal cancer) without affecting PARP-1.
Clinical development of PARP inhibitors
Seven PARP inhibitors are currently in clinical devel-
opment in oncology. Most of phase I studies have used
pharmacodynamic analysis of PARP-1 activity in per-
ipheral blood mononuclear cells (PBMCs) to determine
the optimal PARP inhibitory dose. There are 2 main
investigational approaches: single-agent study in
BRCA-associated and BRCAness cancers; combination
study with DNA-damage agent and/or radiation. BSI-
201 (Sanofi-Aventis) is currently in a phase III trial for
TNBC in combination with gemcitabine and carbopla-
tin. AZD2281 (Astra-Zeneca), AG-014966 (Pfizer) and
ABT-888 (Abott), are in phase II clinical trials as sin-
gle agent or in combination with chemotherapy. MK-
4827 (Merck), CEP-9722 (Cephalon) and E7016 (Eisai)
are in phase I clinical trials. INO-1001 (Inotek) is no
longer in clinical development after completion of a
phase IB study in combination with temozolomide in
patients with advanced melanoma [97], and there is no
updated information available on this compound
[98,99].
Table 3 PARP inhibitors in development
Chemical
series
Therapeutics Development stage
Benzamide BSI-201
(iniparib)
Phase III
Gemcitabine and carboplatin ± BSI-201 in breast and lung cancers; phase I/II: single agent or combination with
chemotherapy in various cancer types including glioma and ovarian cancer
Phthalazinone AZD2281
(olaparib)
Phase I/II
Single agent or combination with chemotherapy in various cancer types including breast, ovarian and colorectal
cancers
Tricyclic indole AG-014699
(PF-01367338)
Phase II
Single agent in BRCA-associated breast or ovarian cancer; Phase I: combination with chemotherapy in advanced
solid tumors
Benzimidazole ABT-888
(Veliparib)
Phase II
Combination with chemotherapy in various cancer types including breast cancer, colorectal cancer, glioblastoma
multiforme and melanoma; phase I: combination with radiation
Indazole MK-4827 Phase I
Single agent; combination with carboplatin-containing regimens
Pyrrolocarbazole CEP-9722 Phase I
Combination with temozolomide in advanced solid tumors
Phthalazinone E7016
(GPI-21016)
Phase I
Combination with temozolomide in advanced solid tumors
Isoindolinone INO-1001 Phase I
Combination with temozolomide in melanoma (completed) without further investigation in oncology; phase II in
cardiovascular disease
Structure
undisclosed
MP-124 Phase I in acute ischemic stroke
Structure
undisclosed
LT-00673 Preclinical
Structure
undisclosed
NMS-P118 Preclinical
Structure
undisclosed
XAV939 Preclinical, highly selective against PARP-5 (tankyrase)
Reference; [94,96,97,100,101,103,106,108,115-117,119,122-125,151-156]
Yuan et al.Journal of Hematology & Oncology 2011, 4:16
http://www.jhoonline.org/content/4/1/16
Page 8 of 14
BSI-201 (Iniparib)
BSI-201 is different from other PARP inhibitors, due to
drug discovery from interacting with DNA binding
domain of PARP-1 instead of catalytic site of PARP
[100]. By disrupting the binding between PARP-1 and
DNA, BSI-201, a noncompetitive PARP-1 inhibitor,
attenuates PARP-1 activation. Phase I study of BSI-201 in
advanced solid tumors has demonstrated good tolerabil-
ity without an identified MTD with dose levels ranging
from 0.5 mg/kg to 8.0 mg/kg IV twice weekly. The most
common adverse event was gastrointestinal toxicity
(39%). At dose level of 2.8 mg/kg, PARP was inhibited in
PBMCsbygreaterthan50%afterasingledose,with
greater inhibition observed (80% or more) after multiple
dosing [101]. A phase IB study combining BSI-201 with
various chemotherapeutic agents such topotecan, gemci-
tabine, temozolomide, and carboplatin/paclitaxel in
patients with advanced solid tumors has shown accepta-
ble safety profiles at doses levels ranging from 1.1 to 8.0
mg/kg iv twice a week [102]. Significant PARP inhibition
was again noted at dose levels of 2.8 mg/kg or higher. Of
55 patients in this study, there were one CR (ovarian can-
cer), 5 PR (2 breast cancer, and 3 other cancer types) and
19 SD. In 2009, OShaughnessy et al. presented the
results of a randomized phase II study comparing gemci-
tabine plus carboplatin with or without BSI-201 (5.6 mg/
kg; iv; biweekly n days 1, 4, 8, and 11 every 21 days) in
patients with TNBC [103]. The addition of BSI-201
improved RR from 16% to 48% (p = 0.002), and DCR
from 21% to 62%. Median PFS was improved from 3.3 to
6.9 months (hazard ratio [HR] 0.34, p < 0.001). Final
result of this phase II study was reported at 2009 San
Antonio Breast Cancer Symposium with overall survival
was improved from 7.7 to 12.2 month (HR 0.5, p = 0.005)
[104]. Its noted that no significant difference in myelo-
toxicity was seen between the two treatment arms. An
updated analysis reported at 2010 European Society for
Medical Oncology meeting showed PFS was improved
from 3.6 months to 5.9 months (HR 0.59) and DCR was
improved from 33.9% to 55.7% (p = 0.015), median over-
all survival benefit remain similar (7.7 months vs. 12.3
months, HR 0.57). A randomized phase III study compar-
ing gemcitabine plus carboplatin with or without BSI-201
in patients with TNBC is currently underway
(NCT00938652). Similar treatment design is used for an
ongoing phase III study in patients with stage IV squa-
mous cell lung cancer (NCT01082549). BSI-201 is also
currently being evaluated as single agent or combination
with chemotherapy in phase I/II studies in various cancer
types including glioma and ovarian cancer.
AZD2281 (Olaparib)
Fong et al. reported the results of phase I study of ola-
parib, which is an oral small-molecule PARP inhibitor
[105,106]. The frequently occurred toxicities were nau-
sea, vomiting, diarrhea, and fatigue. Maximum tolerated
dose (MTD) was identified at 400 mg twice daily, with
grade 3 fatigue and mood alteration DLT noted in one
of eight patients at this dose level. Grade 4 thrombocy-
topenia and grade 3 somnolence occurred in two of five
patients receiving 600 mg twice daily. In a group of 19
patients with breast, ovarian or prostate caners with
known BRCA mutation, RR of 47% and DCR of 63%
were observed without profound difference in toxicity
profiles in comparison with non-BRCA mutated patients
[106]. The subsequent phase II study in 27 breast cancer
patients with BRCA mutation (18 BRCA1 deficient and
9 BRCA2 deficient) showed RR of 41% and median PFS
of 5.7 months [107]. The pooled analysis of 50 ovarian
cancer patients with BRCA1/2-mutation treated on
phase I and II studies (11 on phase I, and 39 on phase
phase II receiving olaparib 200 mg twice daily) showed
RR of 40% and DCR of 46%, predominately in platinum-
sensitive group [108].
Two subsequent Phase II studies evaluating olaparib
in previously treated BRCA1/2-mutated breast cancer
and ovarian cancer patients were recently reported
[104,109,110]. In both studies, patients were treated
with either 100 mg or 400 mg of olaparib twice daily.
Fifty-seven ovarian cancer patients and 54 breast cancer
patients were studies respectively. Overall RR in the
ovarian cancer study was 33% in the high-dose group
and 13% in the low-dose group. Overall RR in the breast
cancer study was 41% in the high-dose group and 22%
in the low-dose group.
Interestingly, reported in 2010 annual meeting of
ASCO, a provocative phase II study of olaparib showed
promising results for women with high-grade serous
ovarian cancer regardless of BRCA mutation status
[111]. Patients with advanced breast or ovarian cancer
were treated with single agent olaparib 400 mg twice
daily continuously for 28-day cycle. Of 64 women with
ovarian cancer in the study, the overall RR was 41.2%
and 23.9%, respectively, for patients with and without
BRCA mutations. However, no response was seen in the
24 patients with TNBC treated with olaparib. This is the
first single-agent trial demonstrating promising activity
of olaparib in high-grade non-BRCA mutated sporadic
serous ovarian caner. The mechanism could be attribu-
ted by underlying DNA repair abnormalities, which may
lead to BRCAness[82,112].
Combinations of olaparib and chemotherapy agents
have been explored. Myelosuppresion decreases toler-
ability when combine olaparib with chemotherapy
agents [113]. Dent et al. reported a phase I/II study of
olaparib in combination with weekly paclitaxel as first
or second-line treatment in patients with metastatic
TNBC [114]. Olaparib 200 mg twice daily was given
Yuan et al.Journal of Hematology & Oncology 2011, 4:16
http://www.jhoonline.org/content/4/1/16
Page 9 of 14
continuously with paclitaxel 90 mg/m
2
weekly for 3 of 4
weeks. Toxicity included 58% neutropenia, 63% diarrhea,
58% nausea, and 53% fatigue, and most were grade 1-2
except neutropenia. Of 19 patients treated in two
cohorts, RR of 33 to 40% and median PFS of 5.2 to 6.3
months were observed.
AG-014699 (PF-01367338)
AG-014699, an intravenous PARP inhibitor, was stu-
died in combination with temozolomide in advanced
solid tumors [115]. PARP inhibitory dose was decided
at 12 mg/m
2
IV daily for 5 days every 4 weeks based
on 74% to 97% inhibition of peripheral blood lympho-
cyte PARP activity. Mean tumor PARP inhibition at 5
h was 92% (range, 46-97%). No significant toxicity was
seen from AG-014699 alone, and AG-014699 showed
linear pharmacokinetics without interaction with
temozolomide. A phase II study with this combination
as 1
st
line treatment of 40 patients with metastatic
melanoma showed RR of 10% and SD of 10%, with
significant bone marrow suppression being the major
toxicity [116]. Currently, this compound is in phase II
study as single agent in patients with advanced
BRCA1/2 mutated breast or ovarian cancer
(NCT00664781), and in phase I study in combination
with cytotoxic agents in patients with advanced solid
tumor (NCT01009190).
ABT-888 (Veliparib)
ABT-888 is an oral PARP inhibitor. Preclinical studies
in breast cancer, melanoma and glioma models demon-
strated that ABT-888 potentates the chemotherapy
effect of a number of agents including temozolomide,
platinum, and irinotecan, as well as radiation [117]. Tan
et al. reported the preliminary result of a phase I trial of
ABT-888 in combination with cyclophosphamide in
patients with advanced solid tumors [118]. ABT-888 50
mg twice daily can be safely combined with cyclopho-
sphamide 750 mg/m
2
. ABT-888 does not alter the phar-
macokinetics of cyclophosphamide. This study is still
ongoing to determine the MTD of ABT-888 and cyclo-
phosphamide combination.
A phase I study of ABT-888 in combination with
metronomic cyclophosphamide revealed activity in
BRCA mutated ovarian cancer and TNBC [119]. A
phase II trial of ABT-888 40 mg twice daily on days 1
to 7 in combination with temozolomide 150 mg/m
2
,
days 1-5 on a 28 days cycles for metastatic breast cancer
was well tolerated [120]. However, activity was limited
to BRCA mutation carriers. Of 8 patients with BRCA1/2
mutation, 37.5% RR and 62.5% DCR were observed.
Medial PFS was 5.5 months in BRCA mutation carriers
vs. 1.8 months in non-carriers. This study calls into
question of BRCAnessfor at least this PARP inhibitor.
ABT-888 is currently being evaluated in many phase I/II
studies in combination with chemotherapy or radiation
in patients with advanced solid tumors.
MK-4827
MK-4827 is an orally bioavailable PARP inhibitor. This
compound displays potent PARP-1 and PARP-2 inhibi-
tion, and inhibits proliferation of breast cancer cells
with mutant BRCA-1 and BRCA-2 with IC
50
in the
range of 10-100 nM [121]. Sandhu et al reported phase
I result of MK-4827 in 59 patients with advanced solid
tumors in 2010 annual meeting of ASCO [122]. MTD
was identified at 300 mg daily with common toxicities
in nausea/vomiting, fatigue and thrombocytopenia. Two
out of six patients on 400 mg daily experienced DLT
with grade 4 thrombocytopenia was seen in 2 out of 6
patients received 400 mg daily. Antitumor activity was
observed in patients with BRCA-deficient cancers (32%
PR in 19 patients with ovarian cancer, and 50% PR in 4
patients with breast cancer). Additionally, PR was seen
in 1 patient with sporadic platinum-sensitive ovarian
cancer. These findings have shown good tolerability and
promising antitumor activity of MK-4827 in both
BRCA-deficient and sporadic cancers. Phase I study in
expanded cohorts with sporadic ovarian and prostate
cancers is currently underway (NCT00749502). Phase IB
dose escalation study of MK-4827 in combination with
carboplatin, carboplatin/paclitaxel or carboplatin/doxil
in patients with advanced solid tumors has also been
activated (NCT01110603).
CEP-9722
Preclinical studies have shown CEP-9722 enhances cel-
lular sensitivity toward temozolomide, irinotecan and
radiation in various cancer types such as glioblastoma,
colon cancer, neuroblastoma, and rhabdomyosarcoma
[123]. CEP-9722 is currently undergoing phase I trial as
single agent and in combination with temozolomide in
advanced solid tumor (NCT00920595).
E7016
E7016 (previously known as GPI-21016) is an orally
bioavailable PARP inhibitor. In murine leukemia
model, E7016 enhances cisplatin-induced cytotoxicity
and ameliorated cisplatin-induced neuropathy at the
same time, suggesting a role to improve the therapeu-
tic margin of certain cytotoxic agent [124]. Further
study in human glioblastoma cell line and xenograft,
E7016 enhances tumor radiosensitivity, and synergizes
with combination treatment of temozolomide and
radiation [125]. There is an ongoing phase I study with
dose escalation of E7016 in combination with temozo-
lomide in patients with advanced solid tumors and
gliomas (NCT01127178).
Yuan et al.Journal of Hematology & Oncology 2011, 4:16
http://www.jhoonline.org/content/4/1/16
Page 10 of 14
Summary
We reviewed preclinical data and clinical development
of MDM2, ALK and PARP inhibitors. Cancer treatment
is entering an exciting chapter in targeted therapies and
personalized medicine due to the advance of molecular
biology and medicinal chemistry. Most likely several
compounds from this review will be approved for clini-
cal use in the years to come. Many questions remain to
be answered: (1) what are the long-term safety and toxi-
cities of these inhibitors (2) how to use biomarkers to
selectpatientswhowillbenefitmostfromtheseinhibi-
tors (3) how to combine these targeted therapies with
cytotoxic agents or other treatment modality such as
radiation modality in selected patient population?
More than 50 percent of human tumors contain a
mutation or deletion of the p53 gene. Mutation of p53
can confer dominant-negative or gain-of-function effects
[126]. Dominant-negative effects lead to suppression of
wild-type p53 protein in heterozygous mutant cells and
a p53 null phenotype; gain-of-function effects result in
promotion of tumor development. There have been con-
cerns on the exposure of MDM2 inhibitors to tumors
with mutant p53, which potentially can have deleterious
effects due to stabilization of mutant p53 [127].
Cautions need to be taken with long-term use of
PARP inhibitors. PARP-1 serves important roles in
other cellular function such as transcription regulation,
initiation of a unique cell death pathway, restarting
stalled replication forks, and modulation of cellular
responses to ischemia, inflammation and necrosis [128].
Previous studies indicated that genetic ablation of
PARP-1 in combination with p53 knockout increased
cancer incidence in mice [129]. This raises concern that
long-term PAPR-1 inhibition could potentially increase
the risk of secondary malignancies.
Author details
1
Division of Medical Oncology and Hematology, Loma Linda University
Medical Center, Loma Linda, CA 92354, USA.
2
Department of Internal
Medicine, China Medical University Hospital, Taichung, Taiwan, China.
Authorscontributions
HRM and CTH designed the paper. YY, YML, CTH and HRM wrote the paper.
All authors read and approved the final manuscript.
Competing interests
The authors declare that they have no competing interests.
Received: 27 January 2011 Accepted: 20 April 2011
Published: 20 April 2011
References
1. Fakharzadeh SS, Trusko SP, George DL: Tumorigenic potential associated
with enhanced expression of a gene that is amplified in a mouse tumor
cell line. EMBO J 1991, 10(6):1565-9.
2. Haupt Y, et al:Mdm2 promotes the rapid degradation of p53. Nature
1997, 387(6630):296-9.
3. Bond GL, Hu W, Levine AJ: MDM2 is a central node in the p53 pathway:
12 years and counting. Curr Cancer Drug Targets 2005, 5(1):3-8.
4. Piette J, Neel H, Marechal V: Mdm2: keeping p53 under control. Oncogene
1997, 15(9):1001-10.
5. Momand J, et al:The MDM2 gene amplification database. Nucleic Acids
Res 1998, 26(15):3453-9.
6. Montes de Oca Luna R, Wagner DS, Lozano G: Rescue of early embryonic
lethality in mdm2-deficient mice by deletion of p53. Nature 1995,
378(6553):203-6.
7. de Rozieres S, et al:The loss of mdm2 induces p53-mediated apoptosis.
Oncogene 2000, 19(13):1691-7.
8. Steinman HA, et al:An alternative splice form of Mdm2 induces p53-
independent cell growth and tumorigenesis. J Biol Chem 2004,
279(6):4877-86.
9. Terzian T, et al:The inherent instability of mutant p53 is alleviated by
Mdm2 or p16INK4a loss. Genes Dev 2008, 22(10):1337-44.
10. Onel K, Cordon-Cardo C: MDM2 and prognosis. Mol Cancer Res 2004,
2(1):1-8.
11. Heist RS, et al:MDM2 polymorphism, survival, and histology in early-
stage non-small-cell lung cancer. J Clin Oncol 2007, 25(16):2243-7.
12. Wasylyk C, et al:p53 mediated death of cells overexpressing MDM2 by
an inhibitor of MDM2 interaction with p53. Oncogene 1999,
18(11):1921-34.
13. Chene P, et al:A small synthetic peptide, which inhibits the p53-hdm2
interaction, stimulates the p53 pathway in tumour cell lines. J Mol Biol
2000, 299(1):245-53.
14. Wang H, et al:Anti-tumor efficacy of a novel antisense anti-MDM2
mixed-backbone oligonucleotide in human colon cancer models: p53-
dependent and p53-independent mechanisms. Mol Med 2002,
8(4):185-99.
15. Xue W, et al:Senescence and tumour clearance is triggered by p53
restoration in murine liver carcinomas. Nature 2007, 445(7128):656-60.
16. Ventura A, et al:Restoration of p53 function leads to tumour regression
in vivo. Nature 2007, 445(7128):661-5.
17. Martins CP, Brown-Swigart L, Evan GI: Modeling the therapeutic efficacy of
p53 restoration in tumors. Cell 2006, 127(7):1323-34.
18. Kussie PH, et al:Structure of the MDM2 oncoprotein bound to the p53
tumor suppressor transactivation domain. Science 1996, 274(5289):948-53.
19. Patel S, Player MR: Small-molecule inhibitors of the p53-HDM2 interaction
for the treatment of cancer. Expert Opin Investig Drugs 2008,
17(12):1865-82.
20. Cheok CF, et al:Translating p53 into the clinic. Nat Rev Clin Oncol advance
online publication; 2010.
21. Klein C, Vassilev LT: Targeting the p53-MDM2 interaction to treat cancer.
Br J Cancer 2004, 91(8):1415-9.
22. Vassilev LT: Small-molecule antagonists of p53-MDM2 binding: research
tools and potential therapeutics. Cell Cycle 2004, 3(4):419-21.
23. Vassilev LT, et al:In vivo activation of the p53 pathway by small-
molecule antagonists of MDM2. Science 2004, 303(5659):844-8.
24. Tovar C, et al:Small-molecule MDM2 antagonists reveal aberrant p53
signaling in cancer: implications for therapy. Proc Natl Acad Sci USA 2006,
103(6):1888-93.
25. Shangary S, S Wang: Small-molecule inhibitors of the MDM2-p53 protein-
protein interaction to reactivate p53 function: a novel approach for
cancer therapy. Annu Rev Pharmacol Toxicol 2009, 49:223-41.
26. Van Maerken T, et al:Antitumor activity of the selective MDM2
antagonist nutlin-3 against chemoresistant neuroblastoma with wild-
type p53. J Natl Cancer Inst 2009, 101(22):1562-74.
27. Tabe Y, et al:MDM2 antagonist nutlin-3 displays antiproliferative and
proapoptotic activity in mantle cell lymphoma. Clin Cancer Res 2009,
15(3):933-42.
28. Hasegawa H, et al:Activation of p53 by Nutlin-3a, an antagonist of
MDM2, induces apoptosis and cellular senescence in adult T-cell
leukemia cells. Leukemia 2009, 23(11):2090-2101.
29. Kojima K, et al:MDM2 antagonists induce p53-dependent apoptosis in
AML: implications for leukemia therapy. Blood 2005, 106(9):3150-9.
30. Lau LM, et al:HDM2 antagonist Nutlin-3 disrupts p73-HDM2 binding and
enhances p73 function. Oncogene 2008, 27(7):997-1003.
31. Jackson MW, Berberich SJ: MdmX protects p53 from Mdm2-mediated
degradation. Mol Cell Biol 2000, 20(3):1001-7.
Yuan et al.Journal of Hematology & Oncology 2011, 4:16
http://www.jhoonline.org/content/4/1/16
Page 11 of 14
32. Wade M, et al:Hdmx modulates the outcome of p53 activation in
human tumor cells. J Biol Chem 2006, 281(44):33036-44.
33. Wang H, et al:A Small-Molecule Inhibitor of MDMX Activates p53 and
Induces Apoptosis. Molecular Cancer Therapeutics 2010, 10(1):69-79.
34. Arts J, et al:JNJ-26854165 - a novel HDM2 antagonist in clinical
development showing broad-spectrum preclinical antitumour activity
against solid malignancies. Proc Am Assoc Cancer Res 2008, 49, Abstract
1592.
35. Stuhmer T, et al:A first-in-class HDM2-inhibitor (JNJ-26854165) in phase I
development shows potent activity against multiple myeloma (MM)
cells in vitro and ex vivo. J Clin Oncol (Meeting Abstracts) 2008,
26(15_suppl):14694.
36. Kojima K, et al:The novel tryptamine derivative JNJ-26854165 induces
wild-type p53- and E2F1-mediated apoptosis in acute myeloid and
lymphoid leukemias. Mol Cancer Ther 2010, 9(9):2545-57.
37. Tabernero J, et al:Phase I pharmacokinetic (PK) and pharmacodynamic
(PD) study of HDM-2 antagonist JNJ-26854165 in patients with
advanced refractory solid tumors. J Clin Oncol (Meeting Abstracts) 2009,
27(15S):3514.
38. Yang H, et al:Macrophage inhibitory cytokine-1: a novel biomarker for
p53 pathway activation. Mol Cancer Ther 2003, 2(10):1023-9.
39. Wasik MA, et al:Anaplastic lymphoma kinase (ALK)-induced
malignancies: novel mechanisms of cell transformation and potential
therapeutic approaches. Semin Oncol 2009, 36(2 Suppl 1):S27-35.
40. Chiarle R, et al:The anaplastic lymphoma kinase in the pathogenesis of
cancer. Nat Rev Cancer 2008, 8(1):11-23.
41. Morris SW, et al:Fusion of a kinase gene, ALK, to a nucleolar protein
gene, NPM, in non-Hodgkins lymphoma. Science 1994, 263(5151):1281-4.
42. Amin HM, Lai R: Pathobiology of ALK+ anaplastic large-cell lymphoma.
Blood 2007, 110(7):2259-67.
43. Chen Y, et al:Oncogenic mutations of ALK kinase in neuroblastoma.
Nature 2008, 455(7215):971-974.
44. Sharma SV, Settleman J: Oncogene addiction: setting the stage for
molecularly targeted cancer therapy. Genes Dev 2007, 21(24):3214-31.
45. Piva R, et al:Ablation of oncogenic ALK is a viable therapeutic approach
for anaplastic large-cell lymphomas. Blood 2006, 107(2):689-97.
46. Houtman SH, et al:Echinoderm microtubule-associated protein like
protein 4, a member of the echinoderm microtubule-associated protein
family, stabilizes microtubules. Neuroscience 2007, 144(4):1373-82.
47. Choi YL, et al:Identification of novel isoforms of the EML4-ALK
transforming gene in non-small cell lung cancer. Cancer Res 2008,
68(13):4971-6.
48. Soda M, et al:Identification of the transforming EML4-ALK fusion gene in
non-small-cell lung cancer. Nature 2007, 448(7153):561-6.
49. Takeuchi K, et al:Multiplex reverse transcription-PCR screening for EML4-
ALK fusion transcripts. Clin Cancer Res 2008, 14(20):6618-24.
50. Takeuchi K, et al:KIF5B-ALK, a novel fusion oncokinase identified by an
immunohistochemistry-based diagnostic system for ALK-positive lung
cancer. Clin Cancer Res 2009, 15(9):3143-9.
51. Wong DW, et al:The EML4-ALK fusion gene is involved in various
histologic types of lung cancers from nonsmokers with wild-type EGFR
and KRAS. Cancer 2009, 115(8):1723-33.
52. Perner S, et al:EML4-ALK fusion lung cancer: a rare acquired event.
Neoplasia 2008, 10(3):298-302.
53. Inamura K, et al:EML4-ALK fusion is linked to histological characteristics
in a subset of lung cancers. J Thorac Oncol 2008, 3(1):13-7.
54. Shinmura K, et al:EML4-ALK fusion transcripts, but no NPM-, TPM3-,
CLTC-, ATIC-, or TFG-ALK fusion transcripts, in non-small cell lung
carcinomas. Lung Cancer 2008, 61(2):163-9.
55. Shaw AT, et al:Clinical features and outcome of patients with non-small-
cell lung cancer who harbor EML4-ALK. J Clin Oncol 2009, 27(26):4247-53.
56. Fukuyoshi Y, et al:EML4-ALK fusion transcript is not found in
gastrointestinal and breast cancers. Br J Cancer 2008, 98(9):1536-9.
57. Shaw AT, et al:Prognostic versus predictive value of EML4-ALK
translocation in metastatic non-small cell lung cancer. J Clin Oncol
(Meeting Abstracts) 2010, 28(15_suppl):7606.
58. Li R, Morris SW: Development of anaplastic lymphoma kinase (ALK)
small-molecule inhibitors for cancer therapy. Med Res Rev 2008,
28(3):372-412.
59. Gunby RH, et al:Structural insights into the ATP binding pocket of the
anaplastic lymphoma kinase by site-directed mutagenesis, inhibitor
binding analysis, and homology modeling. J Med Chem 2006,
49(19):5759-68.
60. Zou HY, et al:An Orally Available Small-Molecule Inhibitor of c-Met, PF-
2341066, Exhibits Cytoreductive Antitumor Efficacy through
Antiproliferative and Antiangiogenic Mechanisms. Cancer Research 2007,
67(9):4408-4417.
61. Christensen JG, et al:Cytoreductive antitumor activity of PF-2341066, a
novel inhibitor of anaplastic lymphoma kinase and c-Met, in
experimental models of anaplastic large-cell lymphoma. Mol Cancer Ther
2007, 6(12 Pt 1):3314-22.
62. McDermott U, et al:Genomic alterations of anaplastic lymphoma kinase
may sensitize tumors to anaplastic lymphoma kinase inhibitors. Cancer
Res 2008, 68(9):3389-95.
63. Shakespeare WC, et al:Discovery of potent and selective orally active
inhibitors of anaplastic lymphoma kinase (ALK). Proc Am Assoc Cancer Res
2009, 50, Abstract 3738.
64. Zhang S, et al:AP26113, a potent ALK inhibitor, overcomes mutations in
EML4-ALK that confer resistance to PF-02341066 (PF1066). Proc Am Assoc
Cancer Res 2010, 51, Abstract LB-298.
65. Rivera VM, et al:Efficacy and pharmacodynamic analysis of AP26113, a
potent and selective orally active inhibitor of Anaplastic Lymphoma
Kinase (ALK). Proc Am Assoc Cancer Res 2010, 51, Abstract 3623.
66. Kwak EL, et al:Clinical activity observed in a phase I dose escalation trial
of an oral c-met and ALK inhibitor, PF-02341066. J Clin Oncol (Meeting
Abstracts) 2009, 27(15S):3509.
67. Bang Y, 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 (Meeting Abstracts) 2010, 28(18_suppl):3.
68. Kwak EL, et al:Anaplastic lymphoma kinase inhibition in non-small-cell
lung cancer. N Engl J Med 2010, 363(18):1693-703.
69. Rouleau M, et al:PARP inhibition: PARP1 and beyond. Nat Rev Cancer
2010, 10(4):293-301.
70. Durkacz BW, et al:(ADP-ribose)n participates in DNA excision repair.
Nature 1980, 283(5747):593-596.
71. Burkle A: Poly(ADP-ribose). The most elaborate metabolite of NAD+. FEBS
J2005, 272(18):4576-89.
72. Huber A, et al:PARP-1, PARP-2 and ATM in the DNA damage response:
functional synergy in mouse development. DNA Repair (Amst) 2004, 3(8-
9):1103-8.
73. Shieh WM, et al:Poly(ADP-ribose) polymerase null mouse cells synthesize
ADP-ribose polymers. J Biol Chem 1998, 273(46):30069-72.
74. Yelamos J, Schreiber V, Dantzer F: Toward specific functions of poly(ADP-
ribose) polymerase-2. Trends Mol Med 2008, 14(4):169-78.
75. Ratnam K, Low JA: Current development of clinical inhibitors of poly
(ADP-ribose) polymerase in oncology. Clin Cancer Res 2007, 13(5):1383-8.
76. Puhalla S: PARP inhibitors: what we know and what we have yet to
know. Oncology (Williston Park) 2010, 24(1):62, 65-6.
77. Comen EA, Robson M: Inhibition of poly(ADP)-ribose polymerase as a
therapeutic strategy for breast cancer. Oncology (Williston Park) 2010,
24(1):55-62.
78. Carden CP, Yap TA, Kaye SB: PARP inhibition: targeting the Achillesheel
of DNA repair to treat germline and sporadic ovarian cancers. Curr Opin
Oncol 2010, 22(5):473-80.
79. Plummer ER, Calvert H: Targeting poly(ADP-ribose) polymerase: a two-
armed strategy for cancer therapy. Clin Cancer Res 2007, 13(21):6252-6.
80. Farmer H, et al:Targeting the DNA repair defect in BRCA mutant cells as
a therapeutic strategy. Nature 2005, 434(7035):917-21.
81. Bryant HE, et al:Specific killing of BRCA2-deficient tumours with
inhibitors of poly(ADP-ribose) polymerase. Nature 2005,
434(7035):913-7.
82. Turner N, Tutt A, Ashworth A: Hallmarks of BRCAnessin sporadic
cancers. Nat Rev Cancer 2004, 4(10):814-9.
83. McCabe N, et al:Deficiency in the repair of DNA damage by
homologous recombination and sensitivity to poly(ADP-ribose)
polymerase inhibition. Cancer Res 2006, 66(16):8109-15.
84. Mendes-Pereira AM, et al:Synthetic lethal targeting of PTEN mutant cells
with PARP inhibitors. EMBO Mol Med 2009, 1(6-7):315-22.
Yuan et al.Journal of Hematology & Oncology 2011, 4:16
http://www.jhoonline.org/content/4/1/16
Page 12 of 14
85. Konstantinopoulos PA, et al:Gene expression profile of BRCAness that
correlates with responsiveness to chemotherapy and with outcome in
patients with epithelial ovarian cancer. J Clin Oncol 2010, 28(22):3555-61.
86. Hastak K, Ford E, Alli JM: Synergistic chemosensitivity of triple-negative
breast cancer cell lines to poly(ADP-Ribose) polymerase inhibition,
gemcitabine, and cisplatin. Cancer Res 2010, 70(20):7970-80.
87. Ishida J, et al:Discovery of potent and selective PARP-1 and PARP-2
inhibitors: SBDD analysis via a combination of X-ray structural study and
homology modeling. Bioorg Med Chem 2006, 14(5):1378-90.
88. Yoo AR, et al:Effects of a newly developed tricyclic PARP-1 inhibitor, on
ischemic stroke. Drug Dev Res 2010, 71(4):253-260.
89. Pellicciari R, et al:On the way to selective PARP-2 inhibitors. Design,
synthesis, and preliminary evaluation of a series of isoquinolinone
derivatives. ChemMedChem 2008, 3(6):914-23.
90. Eliasson MJ, et al:Poly(ADP-ribose) polymerase gene disruption renders
mice resistant to cerebral ischemia. Nat Med 1997, 3(10):1089-95.
91. Graziani G, Battaini F, Zhang J: PARP-1 inhibition to treat cancer, ischemia,
inflammation. Pharmacol Res 2005, 52(1):1-4.
92. Szabo C: Cardioprotective effects of poly(ADP-ribose) polymerase
inhibition. Pharmacol Res 2005, 52(1):34-43.
93. Radovits T, et al:Single dose treatment with PARP-inhibitor INO-1001
improves aging-associated cardiac and vascular dysfunction. Exp Gerontol
2007, 42(7):676-85.
94. Matsuura S, et al:MP-124, a novel poly(adp-ribose)polymerase (PARP)
inhibitor, ameliorates the ischemic damage in non-human primate
model (2009 International Stroke Conference). Stroke 2009, 40(4), Abstract
P251.
95. McCabe N, et al:Targeting Tankyrase 1 as a therapeutic strategy for
BRCA-associated cancer. Oncogene 2009, 28(11):1465-1470.
96. Huang S-MA, et al:Tankyrase inhibition stabilizes axin and antagonizes
Wnt signalling. Nature 2009, 461(7264):614-620.
97. Bedikian AY, et al:A phase IB trial of intravenous INO-1001 plus oral
temozolomide in subjects with unresectable stage-III or IV melanoma.
Cancer Invest 2009, 27(7):756-63.
98. Inotek Pharmaceuticals Pipeline. [http://www.inotekcorp.com/content/
pipeline_overview.asp], [cited 2011 March 18].
99. Annunziata CM, OShaughnessy J: Poly (ADP-ribose) polymerase as a
novel therapeutic target in cancer. Clin Cancer Res 2010, 16(18):4517-26.
100. Ferraris DV: Evolution of poly(ADP-ribose) polymerase-1 (PARP-1)
inhibitors. From concept to clinic. J Med Chem 2010, 53(12):4561-84.
101. Kopetz S, et al:First in human phase I study of BSI-201, a small molecule
inhibitor of poly ADP-ribose polymerase (PARP) in subjects with
advanced solid tumors. J Clin Oncol (Meeting Abstracts) 2008,
26(15_suppl):3577.
102. Mahany JJ, et al:A phase IB study evaluating BSI-201 in combination
with chemotherapy in subjects with advanced solid tumors. J Clin Oncol
(Meeting Abstracts) 2008, 26(15_suppl):3579.
103. OShaughnessy JOC, Pippen J, et al:Efficacy of BSI-201, a poly (ADP-ribose)
ploymerase-1(PARP1) inhibitor, in combination with gemcitabine/
carboplatin (G/C) in patients with metastatic triple-negative breast
cancer (TNBC): Results of a randomized phase II trial. J Clin Oncol
(Meeting Abstracts) 2009, 27(15S):3.
104. Audeh MW, et al:Phase II trial of the oral PARP inhibitor olaparib
(AZD2281) in BRCA-deficient advanced ovarian cancer. J Clin Oncol
(Meeting Abstracts) 2009, 27(15S):5500.
105. Fong PC, et al:AZD2281 (KU-0059436), a PARP (poly ADP-ribose
polymerase) inhibitor with single agent anticancer activity in patients
with BRCA deficient ovarian cancer: Results from a phase I study. J Clin
Oncol (Meeting Abstracts) 2008, 26(15_suppl):5510.
106. Fong PC, et al:Inhibition of poly(ADP-ribose) polymerase in tumors from
BRCA mutation carriers. N Engl J Med 2009, 361(2):123-34.
107. Tutt A, et al:Phase II trial of the oral PARP inhibitor olaparib in BRCA-
deficient advanced breast cancer. J Clin Oncol (Meeting Abstracts) 2009,
27(18S):CRA501.
108. Fong PC, et al:Poly(ADP)-Ribose Polymerase Inhibition: Frequent Durable
Responses in BRCA Carrier Ovarian Cancer Correlating With Platinum-
Free Interval. J Clin Oncol 2010, 28(15):2512-2519.
109. Audeh MW, et al:Oral poly(ADP-ribose) polymerase inhibitor olaparib in
patients with BRCA1 or BRCA2 mutations and recurrent ovarian cancer:
a proof-of-concept trial. The Lancet 2010, 376(9737):245-251.
110. Tutt A, et al:Oral poly(ADP-ribose) polymerase inhibitor olaparib in
patients with BRCA1 or BRCA2 mutations and advanced breast cancer: a
proof-of-concept trial. The Lancet 2010, 376(9737):235-244.
111. Gelmon KA, et al:Can we define tumors that will respond to PARP
inhibitors? A phase II correlative study of olaparib in advanced serous
ovarian cancer and triple-negative breast cancer. J Clin Oncol (Meeting
Abstracts) 2010, 28(15_suppl):3002.
112. Tan DS, et al:BRCAnesssyndrome in ovarian cancer: a case-control
study describing the clinical features and outcome of patients with
epithelial ovarian cancer associated with BRCA1 and BRCA2 mutations. J
Clin Oncol 2008, 26(34):5530-6.
113. Giaccone G, et al:A phase I combination study of olaparib (AZD2281;
KU-0059436) and cisplatin (C) plus gemcitabine (G) in adults with solid
tumors. J Clin Oncol (Meeting Abstracts) 2010, 28(15_suppl):3027.
114. Dent RA, et al:Safety and efficacy of the oral PARP inhibitor olaparib
(AZD2281) in combination with paclitaxel for the first- or second-line
treatment of patients with metastatic triple-negative breast cancer:
Results from the safety cohort of a phase I/II multicenter trial. J Clin
Oncol (Meeting Abstracts) 2010, 28(15_suppl):1018.
115. Plummer R, et al:Phase I study of the poly(ADP-ribose) polymerase
inhibitor, AG014699, in combination with temozolomide in patients with
advanced solid tumors. Clin Cancer Res 2008, 14(23):7917-23.
116. Plummer R, et al:First and final report of a phase II study of the poly
(ADP-ribose) polymerase (PARP) inhibitor, AG014699, in combination
with temozolomide (TMZ) in patients with metastatic malignant
melanoma (MM). J Clin Oncol (Meeting Abstracts) 2006, 24(18_suppl):8013.
117. Kummar S, et al:Phase 0 clinical trial of the poly (ADP-ribose)
polymerase inhibitor ABT-888 in patients with advanced malignancies. J
Clin Oncol 2009, 27(16):2705-11.
118. Tan AR, et al:Preliminary results of a phase I trial of ABT-888, a poly
(ADP-ribose) polymerase (PARP) inhibitor, in combination with
cyclophosphamide. J Clin Oncol (Meeting Abstracts) 2010,
28(15_suppl):3000.
119. Kummar S, et al:A phase I study of ABT-888 (A) in combination with
metronomic cyclophosphamide (C) in adults with refractory solid
tumors and lymphomas. J Clin Oncol (Meeting Abstracts) 2010,
28(15_suppl):2605.
120. Isakoff SJ, et al:A phase II trial of the PARP inhibitor veliparib (ABT888)
and temozolomide for metastatic breast cancer. J Clin Oncol (Meeting
Abstracts) 2010, 28(15_suppl):1019.
121. Jones P, et al:Discovery of 2-{4-[(3S)-Piperidin-3-yl]phenyl}-2H-indazole-7-
carboxamide (MK-4827): A Novel Oral Poly(ADP-ribose)polymerase
(PARP) Inhibitor Efficacious in BRCA-1 and -2 Mutant Tumors. Journal of
Medicinal Chemistry 2009, 52(22):7170-7185.
122. Sandhu SK, et al:First-in-human trial of a poly(ADP-ribose) polymerase
(PARP) inhibitor MK-4827 in advanced cancer patients (pts) with
antitumor activity in BRCA-deficient and sporadic ovarian cancers. J Clin
Oncol (Meeting Abstracts) 2010, 28(15_suppl):3001.
123. Miknyoczki S, et al:The selective poly(ADP-ribose) polymerase-1(2)
inhibitor, CEP-8983, increases the sensitivity of chemoresistant tumor
cells to temozolomide and irinotecan but does not potentiate
myelotoxicity. Mol Cancer Ther 2007, 6(8):2290-2302.
124. Lapidus RG, et al:PARP inhibitors enhance the effect of cisplatin against
tumors and ameliorate cisplatin-induced neuropathy. Proc Am Assoc
Cancer Res 2006, 47, Abstract 506.
125. Russo AL, et al:In vitro and in vivo radiosensitization of glioblastoma
cells by the poly (ADP-ribose) polymerase inhibitor E7016. Clin Cancer
Res 2009, 15(2):607-12.
126. Sigal A, Rotter V: Oncogenic mutations of the p53 tumor suppressor: the
demons of the guardian of the genome. Cancer Res 2000, 60(24):6788-93.
127. Marine JC, Lozano G: Mdm2-mediated ubiquitylation: p53 and beyond.
Cell Death Differ 2010, 17(1):93-102.
128. Patel A, Kaufmann SH: Development of PARP inhibitors: an unfinished
story. Oncology (Williston Park) 2010, 24(1):66-68.
129. Tong WM, et al:Null mutation of DNA strand break-binding molecule
poly(ADP-ribose) polymerase causes medulloblastomas in p53(-/-) mice.
Am J Pathol 2003, 162(1):343-52.
130. Grasberger BL, et al:Discovery and cocrystal structure of
benzodiazepinedione HDM2 antagonists that activate p53 in cells. J Med
Chem 2005, 48(4):909-12.
Yuan et al.Journal of Hematology & Oncology 2011, 4:16
http://www.jhoonline.org/content/4/1/16
Page 13 of 14
131. Koblish HK, et al:Benzodiazepinedione inhibitors of the Hdm2:p53
complex suppress human tumor cell proliferation in vitro and sensitize
tumors to doxorubicin in vivo. Mol Cancer Ther 2006, 5(1):160-9.
132. Ding K, et al:Structure-Based Design of Spiro-oxindoles as Potent,
Specific Small-Molecule Inhibitors of the MDM2âˆp53 Interaction. Journal
of Medicinal Chemistry 2006, 49(12):3432-3435.
133. Shangary S, et al:Temporal activation of p53 by a specific MDM2
inhibitor is selectively toxic to tumors and leads to complete tumor
growth inhibition. Proc Natl Acad Sci USA 2008, 105(10):3933-3938.
134. Mohammad R, et al:An MDM2 antagonist (MI-319) restores p53
functions and increases the life span of orally treated follicular
lymphoma bearing animals. Mol Cancer 2009, 8(1):115.
135. Rothweiler U, et al:Isoquinolin-1-one Inhibitors of the MDM2-p53
Interaction. ChemMedChem 2008, 3(7):1118-1128.
136. Nieves-Neira W, et al:DNA protein cross-links produced by NSC 652287, a
novel thiophene derivative active against human renal cancer cells. Mol
Pharmacol 1999, 56(3):478-84.
137. Issaeva N, et al:Small molecule RITA binds to p53, blocks p53-HDM-2
interaction and activates p53 function in tumors. Nat Med 2004,
10(12):1321-8.
138. Yang Y, et al:Small molecule inhibitors of HDM2 ubiquitin ligase activity
stabilize and activate p53 in cells. Cancer Cell 2005, 7(6):547-559.
139. Wilson JM, et al:Synthesis of 5-deazaflavin derivatives and their
activation of p53 in cells. Bioorg Med Chem 2007, 15(1):77-86.
140. CEP-28122. 2010 [http://www.cephalon.com].
141. Ott GR, et al:Discovery of a potent Inhibitor of anaplastic lymphoma
kinase with in vivo antitumor activity. ACS Med Chem Lett 2010,
1(9):493-498.
142. Lovly CM, et al:Preclinical development of a selective, potent small
molecule ALK inhibitor. Proc Am Assoc Cancer Res 2010, 51, Abstract 1788.
143. Kruczynski A, et al:Antitumor activity of pyridoisoquinoline derivatives
F91873 and F91874, novel multikinase inhibitors with activity against
the anaplastic lymphoma kinase. Anti-Cancer Drugs 2009, 20(5):364-372.
144. Fancelli D, et al:1,4,5,6-Tetrahydropyrrolo[3,4-c]pyrazoles:??Identification
of a Potent Aurora Kinase Inhibitor with a Favorable Antitumor Kinase
Inhibition Profile. Journal of Medicinal Chemistry 2006, 49(24):7247-7251.
145. Bossi RT, et al:Crystal Structures of Anaplastic Lymphoma Kinase in
Complex with ATP Competitive Inhibitors. Biochemistry 2010,
49(32):6813-6825.
146. Wan W, et al:Anaplastic lymphoma kinase activity is essential for the
proliferation and survival of anaplastic large-cell lymphoma cells. Blood
2006, 107(4):1617-1623.
147. Ardini E, et al:Anaplastic Lymphoma Kinase: Role in specific tumours,
and development of small molecule inhibitors for cancer therapy. Cancer
Letters 2010, 299(2):81-94.
148. Sabbatini P, et al:GSK1838705A inhibits the insulin-like growth factor-1
receptor and anaplastic lymphoma kinase and shows antitumor activity
in experimental models of human cancers. Mol Cancer Ther 2009,
8(10):2811-2820.
149. Galkin AV, et al:Identification of NVP-TAE684, a potent, selective, and
efficacious inhibitor of NPM-ALK. Proc Natl Acad Sci USA 2007,
104(1):270-275.
150. Chen Z, et al:Inhibition of ALK, PI3K/MEK, and HSP90 in Murine Lung
Adenocarcinoma Induced by EML4-ALK Fusion Oncogene. Cancer
Research 2010.
151. Brock WA, et al:Radiosensitization of human and rodent cell lines by
INO-1001, a novel inhibitor of poly(ADP-ribose) polymerase. Cancer Lett
2004, 205(2):155-60.
152. Cheng CL, et al:Poly(ADP-ribose) polymerase-1 inhibition reverses
temozolomide resistance in a DNA mismatch repair-deficient malignant
glioma xenograft. Mol Cancer Ther 2005, 4(9):1364-8.
153. MP-124. [http://www.mt-pharma-america.com/development.php], [cited
2010 December 16].
154. Wang B, et al:Novel PARP inhibitors with potent antitumor activity as
single-agent and combination therapies. Mol Cancer Ther 2009,
8(Supplement 1), Abstract 121.
155. Plummer R: Perspective on the pipeline of drugs being developed with
modulation of DNA damage as a target. Clin Cancer Res 2010,
16(18):4527-31.
156. Montagnoli A, et al:NMS-P118, a Parp-1 selective inhibitor with efficacy
in DNA repair deficient tumor models in 22nd EORTC-NCI-AACR
Symposium on Molecular Targets and Cancer Therapeutics. Berlin,
Germany 2010.
doi:10.1186/1756-8722-4-16
Cite this article as: Yuan et al.: Novel targeted therapeutics: inhibitors of
MDM2, ALK and PARP. Journal of Hematology & Oncology 2011 4:16.
Submit your next manuscript to BioMed Central
and take full advantage of:
Convenient online submission
Thorough peer review
No space constraints or color figure charges
Immediate publication on acceptance
Inclusion in PubMed, CAS, Scopus and Google Scholar
Research which is freely available for redistribution
Submit your manuscript at
www.biomedcentral.com/submit
Yuan et al.Journal of Hematology & Oncology 2011, 4:16
http://www.jhoonline.org/content/4/1/16
Page 14 of 14
... Aberrant MDM2 protein expression is documented in a wide variety of human tumors and is thought to be due to gene amplification, transcriptional as well as posttranslational regulation [3]. Evidences have shown the involvement of proteasome system (Ubiquitinproteasome system, UPS) in MDM2 degradation [3][4][5]. Indeed, several Deubiquitinating Enzymes (DUBs), such as USP15 and USP7, have been reported to regulate MDM2 expression or transcriptional activity [6][7][8][9][10]. ...
... Recent studies have identified E3 ubiquitin ligase MDM2 as a novel therapeutic target in cervical cancer, unveiling a great treatment opportunity for cervical cancer patients [4,[19][20][21][22][23]. MDM2, known as Murine double minute 2, is known to be a negative regulator of p53 tumor suppressor gene [22]. ...
Article
Full-text available
Previous studies have demonstrated that the antitumor potential of IU1 (a pharmacological compound), which was mediated by selective inhibition of proteasome-associated deubiquitinase ubiquitin-specific protease 14 (USP14). However, the underlying molecular mechanisms remain elusive. It has been well established that mdm2 (Murine double minute 2) gene was amplified and/or overexpressed in a variety of human neoplasms, including cervical cancer. Furthermore, MDM2 is critical to cervical cancer development and progression. Relatively studies have reported that USP15 and USP7 stabilized MDM2 protein levels by removing its ubiquitin chain. In the current study, we studied the cell proliferation status after IU1 treatment and the USP14-MDM2 protein interaction in cervical cancer cells. This study experimentally revealed that IU1 treatment reduced MDM2 protein expression in HeLa cervical cancer cells, along with the activation of autophagy-lysosomal protein degradation and promotion of ubiquitin-proteasome system (UPS) function, thereby blocked G0/G1 to S phase transition, decreased cell growth and triggered cell apoptosis. Thus, these results indicate that IU1 treatment simultaneously targets two major intracellular protein degradation systems, ubiquitin-proteasome and autophagy-lysosome systems, which leads to MDM2 degradation and contributes to the antitumor effect of IU1.
... Echinoderm microtubule-associated protein-like 4 (EML4)-ALK is the most common ALK gene arrangement, and it is found in 3%-13% of NSCLC. 4 Different ALK fusion genes and EML4-ALK variants have been reported in NSCLC, and they respond to ALK inhibitors differently. 5 The incidence of uncommon ALK gene rearrangements in ALKpositive Chinese NSCLC is 18%-19%, and the clinical efficacy of ALK inhibitors needs further study. ...
Article
Full-text available
A 55‐year‐old Chinese man with a right lung mass and lymph node metastasis (T4N3M0 IIIB) was diagnosed with lung adenocarcinoma after a CT‐guided biopsy. With the wide application of next‐generation sequencing (NGS) in tumour detection, we found a rare CCDC85A‐ALK fusion. The patient received alectinib, which had marked efficacy. This is the first report of a lung adenocarcinoma patient harbouring a new uncommon anaplastic lymphocyte kinase fusion that showed a remarkable response to alectinib. NGS aids in selecting treatment in non‐small cell lung cancer patients.
... Desde entonces, se han publicado más de 60 artículos que referencian la importancia de las alteraciones en este gen, asociadas con el desarrollo de patologías como el CPCNP (≈5%), el tumor miofibrolástico inflamatorio (50%), los linfomas cutáneos (2%-30%), el neuroblastoma (6%-8%), el cáncer de tiroides (6%), el cáncer de mama (2%-4%) y el carcinoma de esófago (≈1%) [8][9][10][11] . En paralelo, se han diseñado y evaluado inhibidores del ALK de origen natural como las estauroporinas o los inhibidores de la HSP90 (hit shock protein 90), que no son específicos ni potentes; posteriormente, se incluyeron nueve subtipos diferentes de moléculas sintéticas pequeñas, entre ellas las aminopirimidinas (crizotinib), diaminopirimidinas (CEP-28122), piridoisoquinolinas (F91873 y F91874), pirrolopirasoles (PHA-E429), indolocarbasoles (CEP-14083 y CEP-14513), una pirrolopirimidina (GSK1838705A), una dianilinopirimidina (NVP-TAE684) y dos moléculas sin estructura claramente definida (AP26113 y X276) [12][13][14][15] . ...
Article
Full-text available
Los rearreglos del gen con actividad quinasa del linfoma anaplásico (ALK) ocurren infrecuentemente en el cáncer de pulmón de célula no pequeña (CPCNP) y constituyen un paradigma para la terapia dirigida contra oncogenes conductores en esta enfermedad. El crizotinib, un inhibidor del ALK disponible por vía oral, ofrece ventajas significativas para los pacientes positivos con toxicidad leve y beneficio clínico, motivos por los que fue aprobado para uso clínico en un subgrupo del cáncer de pulmón definido molecularmente. En la actualidad, se están desarrollando múltiples inhibidores del ALK dirigidos a maximizar la respuesta en presencia de menos efectos adversos, al igual que a controlar los mecanismos de resistencia subyacentes. Esta información permitirá optimizar en el futuro el tratamiento de los pacientes con cáncer de pulmón ALK positivo.
... Genomic analyses have revealed that HR deficiency (HRD) contributes to~50% of HG-SOC cases [6,8]. Exposing these cells to PARPi will lead to DNA damage accumulation, cytotoxic genomic instability, and eventually synthetic lethality [9]. PARPi have been successfully implemented for recurrent HG-SOCs by leveraging inherent HRD [2]. ...
Article
Full-text available
PARP inhibitors (PARPi) are efficacious in treating high-grade serous ovarian cancer (HG-SOC) with homologous recombination (HR) deficiency. However, they exhibit suboptimal efficiency in HR-proficient cancers. Here, we found that the expression of CCAAT/enhancer-binding protein β (C/EBPβ), a transcription factor, was inversely correlated with PARPi sensitivity in vitro and in vivo, both in HR-proficient condition. High C/EBPβ expression enhanced PARPi tolerance; PARPi treatment in turn induced C/EBPβ expression. C/EBPβ directly targeted and upregulated multiple HR genes (BRCA1, BRIP1, BRIT1, and RAD51), thereby inducing restoration of HR capacity and mediating acquired PARPi resistance. C/EBPβ is a key regulator of the HR pathway and an indicator of PARPi responsiveness. Targeting C/EBPβ could induce HR deficiency and rescue PARPi sensitivity accordingly. Our findings indicate that HR-proficient patients may benefit from PARPi via targeting C/EBPβ, and C/EBPβ expression levels enable predicting and tracking PARPi responsiveness during treatment.
... 12,89 The first study was on DNA repair and used NAD + -like compounds that were found to bind to the catalytic site of PARP. 89,90 Due to the role of PARP-1 in the detection and repair of DNA damage, the search for pharmacological inhibitors remains the subject of intense research. Preclinical data showing the efficacy of PARP inhibitors in the suppression of growth of BRCA mutant cells was first published in 2005. ...
Article
Full-text available
Sepsis is not only a threat to the health of individual patients but also presents a serious epidemiological problem. Despite intensive research, modern sepsis therapy remains based primarily on antimicrobial treatment and supporting the functions of failing organs. Finding a cure for sepsis represents a great and as yet unfulfilled need in modern medicine. Research results indicate that the activity of poly (adenosine diphosphate (ADP)-ribose) polymerase (PARP) may play an important role in the inflammatory response and the cellular metabolic disorders found in sepsis. Mechanisms by which PARP-1 may contribute to inflammation and metabolic disorders include effects on the regulation of gene expression, impaired metabolism, cell death, and the release of alarmins. These findings suggest that inhibition of this enzyme may be a promising solution for the treatment of sepsis. In studies using experimental sepsis models, inhibition of PARP-1 has been shown to ameliorate the inflammatory response and increase survival. This action was described, among others, for olaparib, a PARP-1 inhibitor approved for use in oncology. While the results of current research are promising, the use of PARP inhibitors in non-oncological diseases raises some concerns, mainly related to the enzyme’s role in deoxyribonucleic acid (DNA) repair. However, the results of studies on experimental models indicate the effectiveness of even short-term PARP-1 inhibition and do not confirm concerns regarding its impact on the integrity of nuclear DNA. Current research presents PARP inhibition as a potential solution for the treatment of sepsis and indicates the need for further research.
... Pharmaceutical inhibition of PARP-1 [50] may arrest the progression of cancer in part by boosting oxidative metabolism and opposing the Warburg effect (i.e., the reliance of cancer cells on anaerobic glycolysis even when oxygen is plentiful, an effect crucial for malignant transformation). Because they inhibit PARP-1, such drugs would increase sirtuin 1 activity because of the increase in NAD 1 that would ensue. ...
Chapter
This chapter summarizes the actions of the seven mammalian sirtuins, their protein targets, intracellular localization, and the pathways they modulate, with an emphasis on their important roles in aging and longevity. Sirtuins are downregulated with aging. Their wide-ranging activities help ameliorate aging-related mitochondrial dysfunction, genomic instability, inflammation, autophagy, and others. This review emphasizes the intricate molecular mechanisms by which each sirtuin protects against decline in cell function with aging. Genetic variants in sirtuins may contribute to aging and life span. Nutraceuticals and pharmacological agents able to boost sirtuin activity have clinical potential in helping to treat aging-related conditions such as cardiovascular disease, type 2 diabetes, dementia, osteoporosis, and arthritis. In some cancers, however, sirtuins appear to represent a “double-edged sword” by exacerbating or even contributing to cancer etiology, leading to development of sirtuin inhibitors for therapy. In general, activation of sirtuins has the potential to extend the period of healthy life span and could even enhance life span itself.
... The first small molecule Nutlin-3a that can selectively inhibit p53-MDM2 interaction through inducing p53 expression, enhancing its transcriptional activity and effectively inhibiting the growth of tumors with wild-type p53 [5]. One Nutlin derivative RG7112 exhibited good dose tolerance and positive effects in the treatment of acute or chronic leukemia in phase I clinical trial [6,7]. Several other agents, such as RG7388, MI-73301, Kevertrin, MK8242 and DS-3032 have been investigated in clinical trials [3,8e11]. ...
Article
Blockage of p53-MDM2 protein-protein interaction has long been a promising strategy of drug development for cancers with wild type p53. In this study, we report a new p53-MDM2 interaction inhibitor, CYZ2017, which could induce p53 nuclear translocation and possess p53-dependent anti-proliferation activity in a range of cancer cells. CYZ2017 treatment led to increase of p53 levels and induced the transactivation of its target genes p21. In addition, CYZ2017 induced G0/G1 cell cycle arrest and apoptosis in HCT116 cells. Besides, CYZ2017 suppressed tumor growth in a HCT116 xenograft model without visible toxicity. These results support that CYZ2017 might be a promising p53-MDM2 interaction inhibitor with good anti-tumor activity. Our finding provides some cues for further investigation of developing anti-tumor drugs based on the blockage of p53-MDM2 interaction.
Article
Full-text available
Targeting protein-protein interactions (PPIs) by small molecule modulators (iPPIs) is an attractive strategy for drug therapy, and some iPPIs have already been introduced into the clinic. Blocking PPIs is however considered to be a more difficult task than inhibiting enzymes or antagonizing receptor activity. In this paper, we examine whether it is possible to predict the likelihood of molecules to act as iPPIs. Using our in-house iterative stochastic elimination (ISE) algorithm, we constructed two classification models that successfully distinguish between iPPIs from the iPPI-DB database and decoy molecules from either the Enamine HTS collection (ISE 1) or the ZINC database (ISE 2). External test sets of iPPIs taken from the TIMBAL database and decoys from Enamine HTS or ZINC were screened by the models: the area under the curve for the receiver operating characteristic curve was 0.85-0.89, and the Enrichment Factor increased from an initial 1 to as much as 66 for ISE 1 and 57 for ISE 2. Screening of the Enamine HTS and ZINC data sets through both models results in a library of ∼1.3 million molecules that pass either one of the models. This library is enriched with iPPI candidates that are structurally different from known iPPIs, and thus, it is useful for target-specific screenings and should accelerate the discovery of iPPI drug candidates. The entire library is available in Table S6.
Chapter
Full-text available
TNBC is the most difficult BC subtype to treat since it is itself divided into various subtypes As TNBC is associated with worse outcomes and thus doesn’t get benefit from hormonal therapy or therapies targeted to HER2. Thus, this specific type of BC must achieve some specific treatment options. The cornerstone of TNBC treatment is adjuvant/neoadjuvant CT, which includes taxanes, anthracyclines, and/or platinum drugs, as well as dose-dense TC (docetaxel/cyclophosphamide) and AC (doxorubicin/cyclophosphamide). Despite the fact that TNBC is susceptible to CT, current treatment choices are insufficient. TNBC treatment possibilities are at the front edge of the BC research field. TNBC individuals presently get a combination of treatments that include radiation, CT, surgery, recently developed immunotherapy, and targeted therapy. The average survival time for advanced TNBC is 1 year, which is much less than the average survival time for other advanced types of BC. As a result, identifying particular targets and developing more efficient, strong, and hopeful therapy for TNBC patients continues to be a significant clinical challenge. A great focus has been grown in the recently developed targets for TNBC, for instance, the signaling pathways like Hedgehog (Hh) pathway, Notch signaling pathway, Wnt/-catenin pathway; the target molecules like (mTOR) inhibitors, EGFR inhibitors, PARP1 inhibitors, angiogenesis inhibitors, chondroitin sulfate proteoglycan 4 (CSPG4) protein targeted monoclonal antibody and TGF-inhibitors. Furthermore, various target agents are in experimental trials for assessing their therapeutic role in treating TNBC. The need of an hour is that treatment strategy involving targeted therapy must involve such options that should result in the maximum survival rate and less adverse effects, so as to win the battle against this dreadful subtype of BC.
Article
Full-text available
Despite all the other cells that have the potential to prevent cancer development and metastasis through tumour suppressor proteins, cancer cells can upregulate the ubiquitin-proteasome system (UPS) by which they can degrade tumour suppressor proteins and avoid apoptosis. This system plays an extensive role in cell regulation organized in two steps. Each step has an important role in controlling cancer. This demonstrates the importance of understanding UPS inhibitors and improving these inhibitors to foster a new hope in cancer therapy. UPS inhibitors, as less invasive chemotherapy drugs, are increasingly used to alleviate symptoms of various cancers in malignant states. Despite their success in reducing the development of cancer with the lowest side effects, thus far, an appropriate inhibitor that can effectively inactivate this system with the least drug resistance has not yet been fully investigated. A fundamental understanding of the system is necessary to fully elucidate its role in causing/controlling cancer. In this review, we first comprehensively investigate this system, and then each step containing ubiquitination and protein degradation as well as their inhibitors are discussed. Ultimately, its advantages and disadvantages and some perspectives for improving the efficiency of these inhibitors are discussed.
Article
3514 Background: JNJ-26854165 is a novel first-in-clinic oral Human Double Minute-2 (HDM-2) ubiquitin ligase antagonist. It increases the level of HDM-2 client proteins (e.g. p53) by inhibiting the association of HDM-2-client protein complex with the proteosome. Preclinical studies have demonstrated potent anti-proliferative and apoptosis-inducing activity of JNJ-26854165 in a broad range of p53 wild type and mutant tumor models. Methods: A 3+3 dose escalation study with the primary objectives to determine the adverse event (AE) profile, dose limiting toxicities (DLT) and maximum tolerated dose of JNJ-26854165 is ongoing. Secondary objectives include evaluation of PK and PD activity. Eligible patients have ECOG PS≤2 and adequate hematological, renal and hepatic function. JNJ-26854165 is administered orally once daily on a continuous schedule (21-day cycles). Sequential skin and tumor biopsies are taken, and evaluations for HDM-2, p53 and other pathway related markers are performed, including further molecular analyses for HDM-2 activity. Results: 37 pts have been treated at 9 dose levels (DL), 4–300 mg qd. AEs included nausea, vomiting, fatigue, anorexia, insomnia, electrolyte disturbances, liver function tests and creatinine elevations, and asymptomatic QTc prolongation, mostly grade 1–2. A DLT, grade 3 QTcF prolongation, was observed in one patient at DL 9. The patient was asymptomatic and the QTc prolongation recovered after treatment discontinuation. PK data showed dose-proportionality. After dosing at 300 mg, steady state Cmax and AUC0–24h were 2–3 μg/mL and ≈50 μg.h/mL, respectively, well exceeding the expected effective exposure modeled by preclinical studies. PD data showed engagement of the targeted pathway such as dose-dependent p53 upregulation in skin, upregulation of HDM-2 levels in tumors after treatment, and an increase in plasma MIC-1 levels. No objective responses have been observed so far. Conclusions: JNJ-26854165 was well tolerated at doses beyond the expected effective exposure modeled by preclinical studies. PD activity was observed at the 150–300 mg dose levels. The trial continues with cohort expansion at 300 mg. Updated results will be presented. [Table: see text]
Article
5500 Background: Olaparib (AZD2281; KU-0059436) is a novel, orally active PARP inhibitor that induces synthetic lethality in homozygous BRCA-deficient cells. A phase I trial identified 400 mg bd as the maximum tolerated dose (MTD) with an initial signal of efficacy in BRCA-deficient cancers (ASCO 2008; abst 5510). The primary aim of this study was to test the efficacy of olaparib in confirmed BRCA1/BRCA2 carriers with advanced chemotherapy-refractory ovarian cancer. The secondary aim was to assess the safety and tolerability profile in ovarian cancer patients with BRCA1/2 deficiency. Methods: In an international, multicenter, proof-of-concept, single-arm, phase II study, two patient (pt) sequential cohorts received continuous oral olaparib in 28-day cycles, initially at the MTD, 400 mg bd (33 pts), and subsequently at 100 mg bd (24 pts), a previously shown clinically active and PARP inhibitory dose. Eligibility criteria included confirmed genetic BRCA1/2 mutation and recurrent, measurable, incurable disease (previous chemotherapy, median 3 lines). The primary efficacy endpoint was best objective response rate (ORR; RECIST) post baseline. Change in CA125 was a secondary efficacy endpoint. All adverse events were reported using CTCAE v3. Results: At this interim analysis dated October 31, 2008, of 57 enrolled pts (39 BRCA1 deficient and 18 BRCA2 deficient), 33 were evaluable at 400 mg bd and 24 at 100 mg bd. The confirmed RECIST ORR was 33% at 400 mg bd and 12.5% at 100 mg bd. Clinical benefit rate (ORR and/or confirmed >50% decline in CA125) was 57.6% at 400 mg bd and 16.7% at 100 mg bd. Toxicity was mainly mild in severity, reflecting grade 1/2 nausea (44%); fatigue (35%); and anemia (14%). Grade 3 toxicity occurred infrequently, and comprised primarily nausea (7%) and leukopenia (5%). Conclusions: Oral olaparib is well tolerated and highly active in advanced, chemotherapy-refractory BRCA-deficient ovarian cancer, with greater activity seen at the higher dose. Toxicity in BRCA1/2 carriers was similar to that seen in non-carriers. This study provides positive proof of the concept of the activity and tolerability of genetically defined targeted therapy with olaparib in BRCA-deficient ovarian cancers. [Table: see text]
Article
3509 Background: PF-02341066 (PF) is a selective, ATP-competitive, small molecule oral inhibitor of the c-Met/HGFR and ALK receptor tyrosine kinases that has not previously been tested in humans. A Phase 1 dose-escalation trial evaluating PF as an oral single agent was conducted to investigate safety, PK and PD in patients (pts) with advanced cancer (excluding leukemias). Methods: PF was administered under fasting conditions QD or BID on a continuous schedule to pts in successive dose-escalating cohorts at doses ranging from 50 mg QD to 300 mg BID. Pts with advanced cancer were enrolled in the study. Results: Thirty-seven pts were enrolled into the dose escalation part of the study. Tumor types included colorectal, pancreatic, sarcoma, ALCL and NSCLC. The MTD was 250 mg BID. Three DLTs were observed: grade 3 increase in ALT (1 pt at 200 mg QD) and grade 3 fatigue (2 pts at 300 mg BID). The most common AEs were nausea, emesis, fatigue and diarrhea. Nausea and emesis were independent of dose or duration of treatment. Mean AUC (30–57% CV) and Cmax (36–69% CV) increased proportionally with dose from 100 mg QD to 300 mg BID. The median terminal half-life was 46 hours. A 2- to 4-fold increase in the oral midazolam (MDZ) AUC was observed following 28-days of PF dosing at 100 mg QD (n = 3) and 300 mg BID (n = 2), respectively, suggesting PF to be an inhibitor of CYP3A. Ten pts have entered an enriched RP2D cohort of pts with tumors harboring c-Met amplification/gene mutation or ALK fusion genes. There has been 1 confirmed PR in a sarcoma pt with ALK rearrangement (inflammatory myofibroblastic tumor). Among 10 NSCLC pts whose tumors harbor EML4-ALK rearrangement, 1 pt has had a PR, 2 pts have achieved unconfirmed PR and 4 pts have had SD (3 have experienced reduction in tumor burden by ∼20% in measurable lesions and 1 has been treated for 28 weeks). Conclusions: The MTD of PF is 250 mg BID. The major AEs were fatigue or GI-related, and all AEs were manageable and reversible. There was no evidence of non-linear PK at PF doses >100 QD. Treatment with PF-02341066 resulted in promising clinical activity against tumors carrying activating ALK gene rearrangements. Further study of PF in pts with ALK-dependent tumors is warranted. [Table: see text]
Article
3 The full, final text of this abstract will be available in Part II of the 2009 ASCO Annual Meeting Proceedings, distributed onsite at the Meeting on May 30, 2009, and as a supplement to the June 20, 2009, issue of the Journal of Clinical Oncology. [Table: see text]
Article
3027 Background: Poly (ADP-ribose) polymerase (PARP) group of NAD+-dependent enzymes participate in multiple DNA- related functions. Increased PARP activity can lead to drug resistance. Olaparib (O), a potent oral inhibitor of PARP-1 and PARP-2 may partially overcome resistance to C and G when used in combination. Methods: Eligibility: histologically confirmed metastatic or unresectable solid tumors, PS 0–2, normal organ functions. Treatment at dose level 1 (DL1): O 100 mg orally bid on days (D) −2 to 2, G 500 mg/m2/h on D1 and D8, C 60 mg/m2 on D1 after G; q21 days ×6. Doses (same schedule) at DL −1: O 100 mg once daily, G 400 mg/m2/h, C 50 mg/m2. Peripheral blood mononuclear cells (PBMCs) were collected and processed within 2 hours and poly (ADP-ribose) (PAR) concentration determined by immunoassay. Results: From 5/2008 to 11/2009, 23 patients (pts) enrolled; 14 males, median age 52 yrs (25- 88). Tumor types: 8 NSCLC; 3 ovarian; 3 pancreatic; 2 esophageal; 2 thymic ca; 1 mesothelioma; 1 adrenocortical c...
Article
3 Background: PF-02341066 (PF-1066) is a selective, ATP-competitive, small molecule, orally bioavailable inhibitor of the ALK and MET/HGF receptor tyrosine kinases. EML4-ALK fusion oncogenes have been reported in approximately 4% of NSCLC. Patients with NSCLC harboring an ALK fusion were recruited into an expanded cohort at the recommended phase II dose within the first-in-patient monotherapy trial of PF-1066. Methods: Patients with ALK fusions, as determined by FISH using a break-apart probe to ALK, were enrolled into the expanded cohort irrespective of prior therapy. Treated brain metastases were allowed. PF-1066 was given orally at a dose of 250 mg BID. Responses were determined using RECIST with radiographic studies repeated every 8 weeks. The disease control rate (DCR) was determined based on the frequency of patients with RECIST CR, PR and stable disease at 8 weeks. Results: To date, 76 ALK+ NSCLC patients have been treated. The median number of prior treatments was 3 (range, 0-7). Most patients had adenocarcinoma histology and were never or former smokers. Mean plasma C trough was 292 ng/mL, which was above the predicted efficacious concentration from preclinical models (120 ng/mL). The median t 1/2 was ∼53 hours. To date, 50 patients are evaluable for response; ORR is 64% and DCR 90%. The median progression-free survival is not yet mature. The median duration of treatment is 25.5+ weeks. Radiological responses typically were observed at the first or second restaging CT scan. Gastrointestinal toxicities, including nausea (55%) and vomiting (39%), were the most frequent adverse events. Conclusions: The oral ALK inhibitor, PF-1066, demonstrated a high response rate in patients selected for ALK fusions and was associated with a good safety profile. A phase III study has been initiated. This study supports the concept of molecular selection of NSCLC patients for appropriately designed treatment. [Table: see text]
Article
2605 Background: Inhibition of poly (ADP-ribose) (PAR) polymerase (PARP) is expected to enhance the DNA-damaging effects of chemotherapy. PARP has also been implicated in endothelial cell migration. A is an orally available, potent inhibitor of PARP-1 and PARP-2. C is an alkylator that has effects on thrombospondin 1, endothelial cells and T reg cells. Methods: Objectives were to establish the safety, tolerability, and MTD of the combination of A with metronomic C; determine the PK of A; and evaluate PARP inhibition in PBMCs and tumor samples. Patients (pts) had histologically documented solid tumors and lymphoid malignancies that had progressed following standard therapy; ≥ 18 years old; KPS > 70%; adequate bone marrow, hepatic and renal function. Cycle length was 21 d. DLT was defined as drug-related gr ≥ 3 nonhematologic or gr 4 hematologic toxicities during cycle 1. PBMCs and optional tumor biopsies were collected, and PAR levels were determined using a validated ELISA. Results: Eighteen pts have been...
Article
CRA501 Background: Olaparib (AZD2281; KU-0059436) is a novel, orally active PARP inhibitor that induces synthetic lethality in homozygous BRCA-deficient cells. A phase I trial identified 400 mg bd as the maximum tolerated dose (MTD) with an initial signal of efficacy in BRCA-deficient ovarian cancers (ASCO 2008; abst 5510). The primary aim of this study was to test the efficacy of olaparib in confirmed BRCA1/BRCA2 carriers with advanced refractory breast cancer. The secondary aim was to assess safety and tolerability in this population. Methods: In an international, multicenter, proof-of-concept, single-arm, phase II study, two sequential patient (pt) cohorts received continuous oral olaparib in 28-day cycles initially at the MTD, 400 mg bd (27 pts), and subsequently at 100 mg bd, a previously identified PARP inhibitory dose (27 pts). Eligibility criteria included confirmed BRCA1/BRCA2 mutation and recurrent, measurable chemotherapy-refractory breast cancer. The primary efficacy endpoint was best objective response rate (ORR; RECIST) post baseline. Progression-free survival (PFS) and clinical benefit rate were secondary endpoints. All adverse events were reported using CTCAE v3. Results: On November 20, 2008, 54 pts exposed to a median of three prior lines of chemotherapy had been enrolled. 27 pts were dosed at 400 mg bd (18 BRCA1 deficient and 9 BRCA2 deficient), and 24 of these had databased RECIST assessments. The ORR (currently based on unconfirmed responses) was 38% (9/24) (400 mg bd). Causally-related toxicity was mainly mild (grade 1–2) in severity; 9/27 pts (33%) had fatigue; 7/27 (26%) had nausea; 4/27 (15%) had vomiting; and 1/27 (4%) had anemia. Causally-related grade 3 or higher toxicities were seen in 5 pts (19%) with fatigue (3 pts), nausea (2 pts), and anemia (1 pt). 27 pts were treated in the subsequent 100 mg bd cohort where no data are currently available. Conclusions: Olaparib at 400 mg bd is well tolerated and highly active in advanced chemotherapy-refractory BRCA-deficient breast cancer. Toxicity in BRCA1/BRCA2 carriers was similar to that reported previously in non-carriers. This first study with olaparib in BRCA-deficient breast cancers provides positive proof of concept for high activity and tolerability of a genetically defined targeted therapy. [Table: see text]
Article
1018 Background: Triple negative breast cancer (TNBC) shares molecular characteristics with BRCA1-/- breast cancer (BC). Preclinical and clinical data show that BRCA1-/- BC and TNBC are sensitive to inhibition of PARP1, an enzyme necessary for DNA repair and other critical cell functions. This phase 1 study evaluated the tolerability of olaparib, a potent PARP1 inhibitor, combined with weekly paclitaxel in patients with metastatic TNBC. This trial served as a safety evaluation to be followed by a phase II trial after tolerability was determined. Methods: Eligible patients (pts) had ≤ 1 prior cytotoxic regimens for ER-, PR-, and HER2- metastatic BC. Pts were treated with olaparib 200mg capsules PO BID continuously combined with paclitaxel 90mg/m2 IV weekly for 3 of 4 weeks. At this dose, a significant number of pts had delays due to neutropenia, thus a second cohort of 10 pts was enrolled with an amendment to the protocol to allow for GCSF (300 μ g SC days 3-5) as secondary prophylaxis following weekly pac...