ArticlePDF Available

Design, Synthesis, and Structure–Activity Relationship Studies of 3-(Phenylethynyl)-1 H -pyrazolo[3,4- d ]pyrimidin-4-amine Derivatives as a New Class of Src Inhibitors with Potent Activities in Models of Triple Negative Breast Cancer

Authors:

Abstract and Figures

A series of 3-(phenylethynyl)-1H-pyrazolo[3,4-d]pyrimidin-4-amine derivatives were designed and synthesized. Structure-activity relationship (SAR) analysis of these compounds led to the discovery of compound 1j, which showed the highest inhibitory potency against the Src kinase and the most potent anti-viability activity against the typical TNBC cell line MDA-MB-231 among all the synthesized compounds. Further kinase inhibition assays showed that compound 1j was a multikinase inhibitor and potently inhibited Src (IC50: 0.0009 μM) and MAPK signaling protein kinases B-RAF and C-RAF. In an MDA-MB-231 xenograft mouse model, a once-daily dose of compound 1j at 30mg/kg for 18 days completely suppressed the tumor growth with a tumor inhibition rate large than 100% without obvious toxicity. It also displayed good pharmacokinetic properties in a preliminary pharmacokinetic assay. Western blot and immunohistochemical assays revealed that compound 1j significantly inhibited Src and MAPK signaling, and markedly induced apoptosis in tumor tissues.
Content may be subject to copyright.
Design, Synthesis, and StructureActivity Relationship Studies of
3(Phenylethynyl)1Hpyrazolo[3,4d]pyrimidin-4-amine Derivatives
as a New Class of Src Inhibitors with Potent Activities in Models
of Triple Negative Breast Cancer
Chun-Hui Zhang,
,§
Ming-Wu Zheng,
,§
Ya-Ping Li,
,§
Xing-Dong Lin,
Mei Huang,
Lei Zhong,
Guo-Bo Li,
Rong-Jie Zhang,
Wan-Ting Lin,
Yan Jiao,
Xiao-Ai Wu,
Jiao Yang,
Rong Xiang,
Li-Juan Chen,
Ying-Lan Zhao,
Wei Cheng,
Yu-Quan Wei,
and Sheng-Yong Yang*
,
State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy,
Sichuan University, Sichuan 610041, China
Department of Clinical Medicine, School of Medicine, Nankai University, Tianjin 300071, China
*
SSupporting Information
ABSTRACT: A series of 3-(phenylethynyl)-1H-pyrazolo[3,4-d]pyrimidin-4-
amine derivatives were designed and synthesized. Structureactivity relationship
(SAR) analysis of these compounds led to the discovery of compound 1j, which
showed the highest inhibitory potency against the Src kinase and the most potent
antiviability activity against the typical TNBC cell line MDA-MB-231 among all
the synthesized compounds. Further kinase inhibition assays showed that com-
pound 1j was a multikinase inhibitor and potently inhibited Src (IC50 =0.0009μM)
and MAPK signaling protein kinases B-RAF and C-RAF. In an MDA-MB-231
xenograft mouse model, a once-daily dose of compound 1j at 30 mg/kg for
18 days completely suppressed the tumor growth with a tumor inhibition rate
larger than 100% without obvious toxicity. It also displayed good pharmacokinetic properties in a preliminary pharmacokinetic
assay. Western blot and immunohistochemical assays revealed that compound 1j signicantly inhibited Src and MAPK signaling
and markedly induced apoptosis in tumor tissues.
1. INTRODUCTION
Breast cancer is one of the most common cancer types in women
worldwide. Within breast cancer, the triple-negativesubtype
(triple-negative breast cancer, TNBC), which lacks the expres-
sion of estrogen and progesterone receptor (ER/PR) and HER2,
is the most aggressive and also the most deadly.
1,2
TNBC has
been of great interest to oncologists because these cancers do not
benet from hormonal therapies or treatments targeted against
HER2.
3,4
Currently, chemotherapy is the only systemic therapy
and prognosis remains poor.
5,6
Compared with chemotherapy, targeted therapies have the
advantage of maximizing ecacy while often reducing toxicity.
However, unlike other molecular subtypes of breast cancer,
there is no validated specic biomarker for TNBC.
79
Preclinical
studies have identied several potential targets, and Src is one
of the targets that have recently received increasing interest.
10,11
Src is a nonreceptor tyrosine kinase and has been revealed to
be a critical regulator of a large number of intracellular signaling
pathways.
1214
Abnormal activation or amplication of Src
has been detected in TNBC and demonstrated to play a role in
proliferation, migration, and invasion of breast cancer cell
lines.
10,15,16
Furthermore, a number of recent studies have shown
that dysregulation of Src is strongly associated with tumor
metastasis and a poor prognosis of TNBC.
17,18
Src therefore
represents a rational molecular target for TNBC.
10,1922
Several
agents (see Figure 1) are currently available for inhibiting
Src,
2328
and dasatinib is the only one that is in clinical trials for
treating TNBC. Dasatinib is an orally active tyrosine kinase
inhibitor that targets Src and BCR-ABL as well as several other
kinases and has been approved for the treatment of imatinib-
resistant BCR-ABL-positive leukemia.
29
Though dasatinib has a
good potency against Src kinase (IC50 = 0.0003 μM), it just
showed moderate anti-TNBC activity. For example, the IC50
value of dasatinib against the typical TNBC cell line MDA-MB-231
is just 0.178 μM. The updated reports from clinical trial studies of
dasatinib in the treatment of TNBC indicated that single-agent
dasatinib just showed limited ecacy in TNBC.
30
Reasons
causing the limited ecacy of the Src inhibitor in treating TNBC
could be complicated but should be mainly due to the refractory
of TNBC and certain unknown imperfect properties of this
compound. Therefore, discovery of new Src inhibitors with
potent anti-TNBC activity is now strongly demanded for drug
research and development (R&D) against TNBC.
In an eort to discover Src inhibitors with high potency against
TNBC tumor, we recently performed a rational drug design in
Received: February 15, 2015
Article
pubs.acs.org/jmc
© XXXX American Chemical Society ADOI: 10.1021/acs.jmedchem.5b00270
J. Med. Chem. XXXX, XXX, XXXXXX
which scaold hopping was applied on ponatinib (see
Figure 2A). Ponatinib was selected, since we are interested in
the 1,2-diarylethyne structure motif; in addition to that it can
potently inhibit Src (IC50 = 0.003 μM).
31,32
Before the scaold
hopping, we collected a total of 209 potential hinge-binder
fragments derived from known kinase inhibitors; a hinge-binder
fragment indicates a moiety in a kinase inhibitor that can form
one to three hydrogen bonds with the hinge region of kinase
domain.
33
The collected hinge-binder fragments were used to
replace the imidazo[1,2-b]pyridazine moiety in ponatinib; the
imidazo[1,2-b]pyridazine moiety corresponds to the hinge-
binder of ponatinib. Molecular docking was then used to evaluate
the binding anities of constructed molecules with Src, and
GoldScore
34
was adopted to rank these compounds (for details,
see Supporting Information). Chemical structures of the top 100
compounds are given in Supporting Information (see Table S1).
Interestingly, the second ranked compound happened to be a
known potent Src inhibitor.
35
Encouraged by this result, we
decided to choose one from the top of these compounds to
synthesize; a compound was selected if it is a new compound and
synthetically accessible and can potentially form at least two
hydrogen bonds with the hinge region of kinase domain (for
details, see Supporting Information). The nally obtained com-
pound is 3-((4-amino-1-isopropyl-1H-pyrazolo[3,4-d]pyrimidin-
3-yl)ethynyl)-4-methyl-N-(4-((4-methylpiperazin-1-yl)methyl)-
3-(triuoromethyl)phenyl)benzamide (1a; Figure 2A), which
showed a slightly improved kinase inhibitory potency (IC50 =
0.002 μM) against Src compared with ponatinib. More importantly,
it exhibited an IC50 value of 0.069 μM against the TNBC MDA-
MB-231 cells in antiviability assays, which is approximately
2.5 times more potent than dasatinib and ponatinib (see Table 1).
This result indicated that improving the anti-TNBC potency of
Src inhibitors was feasible and encouraged us to carry out a
further structural optimization to compound 1a.
In this investigation, we shall perform structural modications
to compound 1a. A series of 3-(phenylethynyl)-1H-pyrazolo-
[3,4-d]pyrimidin-4-amine derivatives are synthesized, and
the structureactivity relationship (SAR) of this series of com-
pounds is discussed. To the most active compound in enzymatic
and cellular assays, an in depth anti-TNBC activities both in vitro
and in vivo are evaluated. The mechanism of action of this
compound is also investigated.
Figure 1. Chemical structures of representative Src inhibitors.
Figure 2. (A) Structures of ponatinib and compound 1a. (B) Schematic showing regions that are the focus of structural modications.
Journal of Medicinal Chemistry Article
DOI: 10.1021/acs.jmedchem.5b00270
J. Med. Chem. XXXX, XXX, XXXXXX
B
2. CHEMISTRY
All the target compounds in this investigation were readily
prepared using palladium-catalyzed Sonogashira coupling as a
key step (Schemes 13). Synthetic routes of compound 1ah
are schematically illustrated in Scheme 1. First, intermediate
3-iodo-1H-pyrazolo[3,4-d]pyrimidin-4-amine (2), which was
prepared through NIS iodination of commercially available
1H-pyrazolo[3,4-d]pyrimidin-4-amine, reacted with commer-
cially available haloalkanes or self-prepared methanesulfonates
to produce the key intermediates 3ai. Second, commercially
Table 1. Kinase Inhibitory Potency against Src and Antiviability Activities against MDA-MB-231 and HepG2 Cells of
Compounds 1ah
a
IC50 values were determined from KinaseProler of Eurons. The data represent the mean values of two independent experiments.
b
Each
compound was tested in triplicate; the data are presented as the mean ±SD.
Journal of Medicinal Chemistry Article
DOI: 10.1021/acs.jmedchem.5b00270
J. Med. Chem. XXXX, XXX, XXXXXX
C
Scheme 1. Synthetic Routes for Compounds 1ah
a
a
Reagents and conditions: (a) NIS, DMF, 80 °C, N2; (b) R3-X(Br, I) or R3-OMs, K2CO3, DMF, 80 °C, N2; (c) MeOH, conc H2SO4 cat,reux;
(d) ethynyltrimethylsilane, CuI, Pd(PPh3)4, triethylamine, THF, rt, N2; (e) (i) K2CO3, MeOH, rt, (ii) NaOH, H2O, rt; (f) diisobutylaluminum
hydride, anhydrous THF, N2, rt; (g) pyridinium p-toluenesulfonate, amine, sodium triacetoxyborohydride, EA, N2, 11050 °C; (h) (i) SOCl2,
80 °C, (ii) DIPEA, EA, 0 °C to rt ((iii) this step only for 10h, LiOH, THF/H2O, rt); (j) (PPh3)2PdCl2, CuI, DIPEA, DMF, 80 °C, N2.
Scheme 2. Synthetic Routes for Compounds 1ir
a
a
Reagents and conditions: (a) (PPh3)2PdCl2, CuI, DIPEA, DMF, 80 °C, N2; (b) TFA, DCM, 0 °C to rt; (c) acrylyl chloride, triethylamine,
DCM, 10 °C to rt.
Journal of Medicinal Chemistry Article
DOI: 10.1021/acs.jmedchem.5b00270
J. Med. Chem. XXXX, XXX, XXXXXX
D
available 3-iodo-4-methylbenzoic acid was estericated and
subsequently coupled with ethynyltrimethylsilane under
Sonogashira coupling conditions, followed by deprotection to
yield 3-ethynyl-4-methylbenzoic acid 6. Third, reduction of
benzonitrile 7acwith DIBAL cleanly aorded aldehyde 8ac
in excellent yield. 8acwere then coupled with amine through
a classical Borch reductive amination to produce 9a,c,d,fh.
Fourthly, 9a,c,d,fhtogether with 4-((4-methylpiperazin-1-
yl)methyl)aniline (9b) and 3-(triuoromethyl)aniline (9e),
which were purchased from market, reacted with 6under basic
conditions to give the key intermediates 10ah. Finally, target
compounds 1ahwere obtained through a palladium-catalyzed
Sonogashira coupling reaction between 10ahand 3a. Similarly,
target compounds 1ir, which contain various substituents at the
N-1 position of 1H-pyrazolo[3,4-d]pyrimidine (R3) (Scheme 2),
were prepared again through a Sonogashira coupling reaction
between 3biand 10a. Among them, 1q was produced by
deprotection of 1p, and amidation of 1q led to 1r.
The synthetic routes for compounds 14agare outlined in
Scheme 3. Terminal-alkyne-containing intermediates 11 was rst
prepared through a Sonogashira coupling reaction of 3b with
ethynyltrimethylsilane and a succeeding deprotection. Then com-
mercially available or self-prepared benzoic acids 12afwere
coupled with 4-((4-methylpiperazin-1-yl)methyl)-3-(triuoromethyl)-
aniline (9a) to produce iodobenzamides 13af, which under-
went Sonogashira coupling to yield nal compounds 14af, and
hydrolysis of 14f with BBr3led to compound 14g. Self-prepared
iodobenzoic acids 12c,dwere synthesized by esterication of
corresponding benzoic acids 15a,b, followed by iodination and
hydrolysis.
3. RESULTS AND DISCUSSIONS
3.1. SAR Analyses of 3-(Phenylethynyl)-1H-pyrazolo-
[3,4-d]pyrimidin-4-amine Derivatives. Since our goal here
is to discover Src inhibitors with high potency against TNBC,
both enzymatic and cellular assays were utilized to evaluate the
bioactivity of synthesized compounds in the SAR studies. For the
cellular assays, two tumor cell lines, MDA-MB-231 and HepG2,
were chosen. MDA-MB-231 is a typical human TNBC cell line
and expresses a high level of Src. HepG2 is a human liver cancer
cell line that expresses very low level of Src; HepG2 was used
here for ruling out the possible o-target eects and cellular toxic
eects.
The SAR analyses below will mainly focus on discussing the
inuences of various substituents at the following three regions
(see Figure 2B) on the bioactivities: the phenyl ring of the amino
terminal of amide (Ring A; R1,R
2), the N-1 position of 1H-
pyrazolo[3,4-d]pyrimidine (R3), and the phenyl ring of the
carboxyl terminal of amide (Ring B; R4).
3.1.1. Substitution Eects of the Phenyl Ring of the Amino
Terminal of Amide (Ring A; R1,R
2). In this section, we shall
explore the eects of dierent substituents at the meta-position
(R1) or para-position (R2) of ring A. In the rst step, we xed
R2as its original group, namely, (4-methylpiperazin-1-yl)methyl,
Scheme 3. Synthetic Routes for Compounds 14ag
a
a
Reagents and conditions: (a) Pd(PPh3)4, CuI, ethynyltrimethylsilane, triethylamine, DMF, 80 °C, N2; (b) K2CO3, MeOH, rt; (c) (i) SOCl2,80°C,
(ii) 4-((4-methylpiperazin-1-yl)methyl)-3-(triuoromethyl)aniline (7a), DIPEA, EA, 0 °C to rt; (d) CuI, (PPh3)2PdCl2, DIPEA, DMF, 80 °C, N2;
(e) BBr3, DCM, 78 °C to rt; (f) MeOH, conc H2SO4 cat,reux; (g) NaIO4, I2, conc H2SO4, acetic anhydride, acetic acid, 540 °C; (h) EtOH/H2O,
NaOH, rt.
Journal of Medicinal Chemistry Article
DOI: 10.1021/acs.jmedchem.5b00270
J. Med. Chem. XXXX, XXX, XXXXXX
E
and varied R1. Bioactivities of the resulting compounds (1ad)
are shown in Table 1. Obviously, replacement of triuoromethyl
group by hydrogen led to a slight drop in bioactivities in both
enzymatic and cellular assays (1b). Introduction of chloride or
bromide at R1did not improve the biochemical and cellular
potencies too much, rather likely increased the antiviability
activity against HepG2, implying a possible increase in toxicity. In
the second step, we thus xed R1as triuoromethyl group and
changed R2with dierent substituents. Compared with 1a,
substitution of R2by hydrogen (1e) or 4-methylmorpholine (1f)
decreased the enzymatic activity and the antiviability potency
against MDA-MB-231 cells. For replacement of R2with 1-ethyl-
4-methylpiperidine (1g), its enzymatic and cellular potencies
remained. However, introduction of 2-(4-methylpiperazin-1-
yl)ethanol (1h)atR
2slightly increased the antiviability against
HepG2, implying an increase in toxicity. Obviously, the results
indicated that modications of 1a on R1and R2had almost no
eect on the improvement of bioactivity. Therefore, in
subsequent structural modication processes, R1and R2will be
kept as their original triuoromethyl and (4-methylpiperazin-1-
yl)methyl, respectively.
3.1.2. Inuences of Various Substituents at the N-1 Position
of 1H-Pyrazolo[3,4-d]pyrimidine Moiety (R3). To examine the
possible inuences of substituents at the N-1 position of
1H-pyrazolo[3,4-d]pyrimidine moiety, various alkyl groups
with dierent size were rst used to replace the original
isopropyl. Bioactivities of the synthesized derivatives (1a,1ir)
are shown in Table 2. From Table 2, we can see that all the
derivatives have good bioactivities in both enzymatic and cellular
levels. The most active compound corresponds to 1j (Src, IC50 =
0.0009 μM; MDA-MB-231, IC50 = 0.011 μM), which contains an
ethyl group at R3. Compared with 1j, compounds with a smaller
size hydrophobic group (methyl, 1i), or a larger size hydrophobic
group (isopropyl (1a), cyclobutyl (1k), and cyclopentyl (1l)), at
the R3position aorded a slightly lower bioactivity, implying that
ethyl represents the most suitable size of alkyl substituent. Then,
various polar substituents including methoxymethyl (1m),
(S)-tetrahydrofuran-3-yl (1n), (R)-tetrahydrofuran-3-yl (1o),
1-(Boc)piperidin-4-yl) (1p), piperidin-4-yl (1q), and 1-acryl-
oylpiperidin-4-yl (1r) were used to replace the ethyl group.
However, bioactivities of these compounds (Table 2) still did not
exceed that of compound 1j.
3.1.3. Possible Impacts of Various Substituents at the C-4
Position of Phenyl Ring B (R4). To explore the possible impact of
dierent substituents at the C-4 position (R4) of phenyl ring B,
we synthesized compounds 14ag, which contain varied
substituents at R4and xed groups at R1,R
2, and R3as their
optimal forms. Bioactivities of compounds 14agare shown in
Table 3. From Table 3, we can see that replacement of methyl
group by hydrogen slightly decreased the biochemical and
cellular anti-TNBC activities. A bulky alkyl substituent at R4, such
as ethyl or i-Pr, remarkably decreased its bioactivities. Further, a
halide (Cl, F) or a polar group (methoxyl and hydroxyl) also
considerably decreased the bioactivity. These results indicated
that methyl is the best substituent group at R4.
3.2. Preliminary in Vivo Antitumor Assays for Screening
Compounds with the Most Potent Anti-TNBC Activity. The
SAR analyses above led to the discovery of a number of Src
inhibitors that exhibited higher potency than dasatinib and
ponatinib in in vitro antiviability assays against human TNBC
MDA-MB-231 cells. To screen compounds with potent anti-
TNBC activity in vivo, we chose three compounds, namely, 1n,
1i, and 1j, to carry out a preliminary in vivo anti-TNBC study in a
MDA-MB-231 xenograft mouse model. The three compounds
were chosen because they are potent Src inhibitors with an IC50
value less than 10 nM and ranked as the top three most active
ones among all the compounds in the in vitro antiviability assay
against the TNBC MDA-MB-231 cells. In the preliminary in vivo
assays, compound 1j was still the most active one (see Figure 3).
Therefore, further in-depth studies including in vitro and in vivo
anti-TNBC activities, and the mechanism of action, were sub-
sequently carried out with compound 1j.
3.3. Biochemical Activities of Compound 1j against
Various Recombinant Human Protein Kinases. The kinase
inhibition prole of compound 1j against a panel of selected
recombinant human protein kinases is presented in Table 4.
Compound 1j potently inhibited Src (IC50 = 0.0009 μM), Yes
(IC50 = 0.0008 μM), Fyn (IC50 = 0.005 μM), and Blk (IC50 =
0.019 μM), which all belong to the Src family kinases (SFKs).
Compound 1j also exhibited considerable potency against
several other kinases, including B-RAFV600E (IC50 = 0.015 μM),
B-RAF (IC50 = 0.092 μM), C-RAF (IC50 = 0.027 μM), BCR-ABL
(IC50 = 0.001 μM), EphA2 (IC50 = 0.016 μM), EphB2 (IC50 =
0.026 μM), TrkA (IC50 = 0.027 μM), DDR2 (IC50 = 0.128 μM),
TAK1 (IC50 = 0.061 μM), Btk (IC50 = 0.067 μM), IKKα(IC50 =
0.353 μM), IKKβ(IC50 = 0.164 μM), Axl (IC50 = 0.578 μM),
PDGFRα(IC50 = 0.890 μM), JAK2 (IC50 = 2.911 μM), and
EGFR(IC50 = 3.518 μM). Compound 1j displayed almost
no inhibitory activity against 18 other tested protein kinases
(IC50 >10μM). These data demonstrate that compound 1j is a
multikinase inhibitor with high potencies against Src family
kinases (SFKs) and several other kinases including MAPK
signaling protein kinases B-RAF and C-RAF.
3.4. Antiviability Activities of Compound 1j against
Various Cancer Cell Lines. The antiviability potencies of
compound 1j against various tumor cell lines including TNBC,
and other breast cancer subtypes, as well as other selected cancer
types, were measured using the MTT assay method. As shown in
Table 5, compound 1j displayed potent activities against TNBC
cell lines MDA-MB-231 and MDA-MB-435 with IC50 values of
0.011 and 0.005 μM, respectively. It also exhibited considerable
potencies against several other cell lines, including MDA-MB-
436 (TNBC, IC50 = 0.316 μM), MDA-MB-453 (TNBC, IC50 =
0.325 μM), MM.1S (myeloma, IC50 = 0.872 μM), U-87 MG
(glioblastoma, IC50 = 0.742 μM). For other 12 cell lines including
BT474 (breast cancer), MCF-7 (breast cancer), SKBR-3 (breast
cancer), HBL-1 (lymphadenoma), OCI-LY10 (lymphadenoma),
RAMOS (lymphadenoma), THP-1 (leukemia), HepG2 (hep-
atocarcinoma), Hela (cervical cancer), H358 (lung cancer),
A2058 (melanoma), PANC-1 (pancreatic cancer), compound 1j
just showed very weak activity or no activity (IC50 >10μM).
Colony-forming assays were then used to examine the anti-
proliferation activity of compound 1j. It was found that com-
pound 1j at concentrations larger than 0.01 μM signicantly
inhibited the colony formation of MDA-MB-231 cells (see
Figure 4A). Dasatinib also considerably decreased the formation
of colonies of MDA-MB-231 cells at concentrations of >0.01 μM.
Nevertheless, in terms of the potency in inhibiting the formation
of colonies, compound 1j is obviously more potent than dasatinib.
3.5. Eects of Compound 1j on Cell Apoptosis and Cell
Cycle Progression. Aow cytometry assay was performed to
examine cell apoptosis upon treatment with compound 1j.As
shown in Figure 4B, compound 1j induced apoptosis in MDA-
MB-231 cells in a concentration-dependent manner. At a con-
centration of 0.3 μM, an apoptosis rate of 34.4% was observed
after 48 h. We further examined the inuence of compound 1j
Journal of Medicinal Chemistry Article
DOI: 10.1021/acs.jmedchem.5b00270
J. Med. Chem. XXXX, XXX, XXXXXX
F
Table 2. Kinase Inhibitory PotencyagainstSrc and Antiviability Activities against MDA-MB-231and HepG2 Cells of Compounds 1irand1a
a
IC50 values were determined from KinaseProler of Eurons. The data represent the mean values of two independent experiments.
b
Each
compound was tested in triplicate; the data are presented as the mean ±SD.
c
nd means not determined.
Journal of Medicinal Chemistry Article
DOI: 10.1021/acs.jmedchem.5b00270
J. Med. Chem. XXXX, XXX, XXXXXX
G
treatment on the cell cycle of MDA-MB-231 cells using ow
cytometry. The results indicated that compound 1j could induce
cell cycle arrest in G0/G1phase (Figure 4C). As a positive
control, dasatinib also showed the same eects in both ow
cytometry assays but was relatively weaker than compound 1j in
terms of the potency. Taken together, these data demonstrated
that compound 1j could eectively lead to cell apoptosis and
G0/G1phase arrest in MDA-MB-231.
3.6. Eects of Compound 1j on Tumor Cell Migration
and Invasion. The eects of compound 1j on tumor cell
migration and invasion were assessed using wound healing assays
and transwell assays, respectively. As displayed in Figure 5A, after
treatment of compound 1j in concentrations of >0.03 μM for
20 h, the migration of MDA-MB-231 cells was signicantly
inhibited. Results from the transwell assays showed that 0.1 μM
compound 1j potently blocked the invasion ability of MDA-MB-
231 (Figure 5B). Again, dasatinib also exhibited the same eects
but was relatively weaker than compound 1j in terms of the
potency. These results demonstrated that 1j could eciently
inhibit the migration and invasion of tumor cells.
3.7. Inactivation of Key Signaling Proteins in Intact
Cells. The ability of compound 1j to inhibit the activation of key
signaling proteins in intact cells was assessed using Western blot.
After a 5 h treatment with increasing concentrations of com-
pound 1j, MDA-MB-231 cells were harvested and lysed for an
IP/wt assay. As shown in Figure 6, compound 1j inhibited Src
phosphorylation in a dose-dependent manner with an estimated
IC50 value of 0.03 μM. Consistent with the inhibition of Src
activation, the phosphorylation of its downstream signaling
Figure 3. Preliminary in vivo anti-TNBC assays of compounds 1n,1i,
and 1j. Daily oral administration of compounds 1n,1i, and 1j at
concentrations of 40 mg kg1d1, respectively, was initiated when the
MDA-MB-231 tumors reached approximately 200 mm3in volume
(three mice per group). Points indicate mean tumor volume (mm3);
bars indicate SD.
Table 4. Kinase Inhibition Prole of Compound 1j against
Human Src and a Panel of Other Selected Protein Kinases
a
kinase IC50
(μM) kinase IC50
(μM) kinase IC50
(μM)
Src 0.0009 TAK1 0.061 CHK1 >10
Src (T341M) 0.030 Btk 0.067 FAK >10
Yes 0.0008 IKKα0.353 GSK3β>10
Fyn 0.005 IKKβ0.164 JNK1α1 >10
Blk 0.019 Axl 0.578 MAPK1 >10
B-RAF (V600E) 0.015 PDGFRα0.890 MEK1 >10
B-RAF 0.092 JAK2 2.911 mTOR >10
C-RAF 0.027 EGFR 3.518 PAK1 >10
BCR-ABL 0.001 Aurora A >10 Pim-1 >10
EphA2 0.016 Ark5 >10 PKBα>10
EphB2 0.026 CDK2 >10 PKBβ>10
TrkA 0.027 CDK7 >10 PKCα>10
DDR2 0.128 TBK1 >10 PI3Kα>10
a
IC50 values were determined from KinaseProler of Eurons. The
data represent the mean values of two independent experiments.
Table 5. Antiviability Activities of Compound 1j against
Various Cancer Cell Lines
a
cell line tumor type IC50 (μM)
MDA-MB-231 TNBC 0.011
MDA-MB-435 TNBC 0.005
MDA-MB-436 TNBC 0.316
MDA-MB-453 TNBC 0.325
BT474 breast cancer >10
MCF-7 breast cancer 7.25
SKBR-3 breast cancer 4.523
HBL-1 lymphadenoma 10
OCI-LY10 lymphadenoma 2.168
RAMOS lymphadenoma 10
MM.1S myeloma 0.872
THP-1 leukemia 5.11
U-87 MG glioblastoma 0.742
HepG2 hepatocarcinoma 8.672
Hela cervical cancer 5.410
H358 lung cancer 1.665
A2058 melanoma 1.908
PANC-1 pancreatic cancer 6.556
a
Each compound was tested in triplicate; the data are presented as the
mean ±SD.
Table 3. Kinase Inhibitory Potency against Src and
Antiviability Activities against MDA-MB-231 and HepG2
Cells of Compounds 14ag and 1j
anti-cell viability (IC50,μM)
b
compd R4
kinase inhibition
(IC50,μM),
a
Src MDA-MB-231 HepG2
1j Me 0.0009 0.011 ±0.001 >5
14a H 0.005 0.024 ±0.002 >5
14b Cl 0.011 0.052 ±0.004 >5
14c Et 0.083 0.418 ±0.029 >5
14d i-Pr 1.409 1.225 ±0.090 >5
14e Fnd
c
1.743 ±0.222 >5
14f OMe 0.059 0.321 ±0.041 4.640 ±0.387
14g OH 3.191 >5 >5
a
IC50 values were determined from KinaseProler of Eurons. The
data represent the mean values of two independent experiments.
b
Each compound was tested in triplicate; the data are presented as the
mean ±SD.
c
nd means not determined.
Journal of Medicinal Chemistry Article
DOI: 10.1021/acs.jmedchem.5b00270
J. Med. Chem. XXXX, XXX, XXXXXX
H
protein FAK was signicantly inhibited at concentrations of
>0.03 μM. Additionally, phosphorylation of the MAPK signaling
proteins, MEK and ERK, was also strongly inhibited, which could
be due to the inactivation of B-RAF and C-RAF (see Table 4),
upstream signaling proteins of MAPK, by compound 1j. It is also
noteworthy that compound 1j has no impact on the phos-
phorylation of AKT, indicating no eect on the AKT signaling.
Similar to compound 1j, dasatinib also eciently inhibited Src
and FAK and showed no eect on AKT signaling. However,
dierent from compound 1j, dasatinib just displayed very weak
inhibitory potency against the ERK phosphorylation, suggesting
less inuence on the MAPK signal pathway. From here, it is
reasonable to conclude that one of the main causes that com-
pound 1j showed a higher antiproliferation activity against
TNBC cells than dasatinib could be due to it being able to inhibit
MAPK signaling more eciently than dasatinib, in addition to
the potent inhibition of Src signaling.
3.8. In Vivo Eects of Compound 1j. The in vivo anti-
TNBC activities of compound 1j were evaluated using an MDA-
MB-231 xenograft model. When the tumor grew to a volume of
150200 mm3, the mice were grouped and treated orally once
daily with 7.5, 15, or 30 mg kg1d11j for 18 days. The tumor
Figure 4. (A) 1j inhibited colony formation of MDA-MB-231 cells. MDA-MB-231 cells were seeded in six-well plates and treated with 1j or dasatinib for
12 days. Colonies were stained with crystal violet and pictures taken. (B, C) 1j induced apoptosis and G0/G1cell cycle arrest of MDA-MB-231 cells.
MDA-MB-231 cells were harvested after treatment with various concentrations of 1j and dasatinib for 24 and 48 h. Cells were stained with an annexin
V-FITC apoptosis detection kit or cell cycle and apoptosis analysis kit, respectively.
Journal of Medicinal Chemistry Article
DOI: 10.1021/acs.jmedchem.5b00270
J. Med. Chem. XXXX, XXX, XXXXXX
I
volumes were measured every 3 days. At all doses given,
compound 1j markedly inhibited the tumor growth. Notably,
a30mgkg
1d1dose of compound 1j completely stopped
the tumor growth with a tumor inhibitory rate of >100%
(Figure 7A) without obvious toxicity. On the contrary, dasatinib
(40 mg kg1d1) and paclitaxel (10 mg kg1week1), a clinically
used TNBC drug, just showed moderate anti-TNBC eects in
the same model (Figure 7A).
To better understand the mechanism of antitumor activities
in vivo, the abilities of compound 1j to inhibit Src and MAPK
signaling, and cell proliferation (Ki67), as well as its eect on
apoptosis in tumor tissues were also evaluated using tumor
tissues isolated from MDA-MB-231 tumor xenograft models at
the end of the treatment with histological and immunohis-
tochemical techniques. A signicant decrease in the phosphor-
ylation of Src and MEK/ERK was observed in the compound 1j
treated groups compared with the control groups (Figure 7B),
indicating inhibition of Src and MAPK signal pathways. These
tumors exhibited reduced staining of Ki67, implying a reduction
in the number of proliferating cells in the tumor tissues. Further,
more TUNEL-positive cells with AP-Red staining were observed,
indicating a signicant increased apoptosis in the treatment
group when compared with the control group.
3.9. Pharmacokinetic Characteristics of Compound 1j.
A preliminary pharmacokinetic property assessment of com-
pound 1j was carried out on rats. The plasma concentration
versus time prole is presented in Figure 7C, and the key
pharmacokinetic parameters of compound 1j are summarized in
Table 6. After per os administration of 10 mg/kg compound 1j,
the area under the concentrationtime curve (AUC0−∞) was
about 4772.5 μgL
1h and the half-life (t1/2) was about 12.76 h.
In addition, compound 1j achieved a maximum plasma
concentration (Cmax) of 214.63 μg/L within about 12 h and
showed a clearance rate (CL) of 2.1 L h1kg1and apparent
distribution volume (Vss) of 38.57 L/kg. All of these indicated
that compound 1j has good pharmacokinetic properties.
4. CONCLUDING REMARKS
In this study, a series of new Src inhibitors bearing a
3-(phenylethynyl)-1H-pyrazolo[3,4-d]pyrimidin-4-amine scaf-
fold were designed and synthesized. Structureactivity relation-
ships of these compounds were discussed based on enzymatic
and cellular activities. A number of compounds showed high
potencies in both biochemical and cell functional assays. Three
most active compounds were selected to conduct a preliminary in
vivo anti-TNBC assay, and 1j exhibited the most potent in vivo
anti-TNBC activity. Then, further in depth studies were carried
out on 1j for its in vitro and in vivo anti-TNBC activities.
Figure 5. (A) 1j inhibited MDA-MB-231 cell migration in wound healing assay. Cells were wounded by the pipet and then treated with various
concentrations of compounds for 20 h. Scale bar, 100 μm. (B) 1j inhibited MDA-MB-231 cells invasion in transwell invasion assay. The bottom
chambers of the transwells were lled with 600 μL of DMEM with 10% FBS, while the top chambers were seeded with 1 ×105MDA-MB-231 cells in
200 μL of DMEM and treated with dierent concentrations of compounds for 24 h. Scale bar, 50 μm.
Figure 6. 1j inhibited the phosphorylation of Src and its downstream
signaling proteins and the activation of MAPK signaling proteins in cell
cultures. MDA-MB-231 cells were treated with 1j or dasatinib for 20 h.
Cells were lysed, and the proteins were analyzed by Western blot
analysis.
Journal of Medicinal Chemistry Article
DOI: 10.1021/acs.jmedchem.5b00270
J. Med. Chem. XXXX, XXX, XXXXXX
J
The results showed that compound 1j was a multikinase inhib-
itor with high potencies against Src and several other kinases
including MAPK signaling protein kinases B-RAF and C-RAF.
In cellular assays, compound 1j signicantly inhibited the
proliferation of TNBC MDA-MB-231 and MDA-MB-435 cells
and displayed good selectivity for these TNBC cells against
other cancer cells. This compound could eectively block the
migration and invasion of MDA-MB-231 cells and lead to
apoptosis and G0/G1phase arrest. In an MDA-MB-231 xenograft
mouse model, a once-daily dose of compound 1j at 30 mg/kg
for 18 days led to complete tumor regression without obvious
toxicity. Western blot and immunohistochemical analyses
indicated that compound 1j potently inhibited Src and MAPK
signal pathways. Finally, it is also important to mention that
although dasatinib has a comparable potency with compound 1j
in Src inhibition, it has a relatively weaker activity than com-
pound 1j in inhibiting MAPK signaling; dysregulation of MAPK
signaling has been demonstrated to be associated with tumor
development and drug resistance of TNBC.
21,36,37
This could be
one of the most important reasons that compound 1j showed
more potent anti-TNBC activity than dasatinib.
5. EXPERIMENTAL SECTION
Kinase Inhibition Assays. Kinase inhibition proles were obtained
using KinaseProler services provided by Eurons, and ATP con-
centrations used are the ATP Kmof corresponding kinases.
Cell Lines and Cell Culture Conditions. All of the cell lines used
were obtained from the American Type Culture Collection (Manassas,
VA, USA). These cell lines were cultured in the designated medium
containing 10% fetal bovine serum (FBS) (v/v) at 37 °C in a humidied
5% CO2incubator according to ATCC guidelines.
Cell Viability Assays. The viability of cells was determined using the
MTT assay method. The cell lines were seeded (150030 000 cells per
well, depending on the cell type) in 96-well plates. After incubation for
24 h in serum-containing media, the cells were treated with inhibitors
(010 μg/mL) diluted with culture medium for 72 h at 37 °C under a
5% CO2atmosphere. Then 20 μL of the MTT reagent (5 mg/mL) was
added to each well, and the plates were incubated for 24 h at 37 °C. For
the adherent cells, the media and MTT were carefully aspirated from
each well, and the formazan crystals were dissolved in 150 μL of 100%
DMSO. For the suspended cells, 50 μL of 20% acidied SDS (w/v) was
used to dissolve the oxidative product, and the cells were incubated
overnight. Finally, the absorbance at 570 nm was read using a Multiskan
Figure 7. (A) In vivo antitumor ecacy of 1j against MDA-MB-231 tumor xenograft models. Daily oral administration of 1j at concentrations of 7.5, 15,
30 mg kg1d1and paclitaxel at a dose of 10 mg kg1week1though tail vein injection were initiated when the MDA-MB-231 tumors reached
appoximately 200 mm3in volume (6 mice per group). Points indicate mean tumor volume (mm3); bars indicate SD; iv, intravenous. (B) Mechanism of
action of 1j in human tumor xenograft models. Mice bearing MDA-MB-231 tumor xenograft which was treated with 1j at 30 mg/kg were humanly
euthanized at the end of the experiment, and the tumor tissues were removed for further immunohistochemistry analysis and TUNEL detection. Scale
bar, 50 μm. (C) Plasma concentrationtime curve of 1j in SD rats after a single oral dose of 10 mg/kg. Blood was collected at the indicated times, and the
plasma concentrations were determined by LC/MS. Points, mean; bars, SD; n=5.
Table 6. Pharmacokinetic Parameters of Compound 1j after
Oral Administration to SD Rats (n=5)
parameter po (10 mg/kg)
AUC0−∞
a
(μgL
1h) 4772.50
t1/2
b
(h) 12.76
Cmax
c
(μg/L) 214.63
Tmax
d
(h) 12.00
CL
e
(L h1kg1) 2.10
Vss
f
(L/kg) 38.57
a
Mean area under the plasma concentrationtime curve.
b
Mean half-
life associated with the terminal slope.
c
Mean peak plasma con-
centration.
d
Mean time to reach maximum plasma concentration.
e
Mean body clearance.
f
Mean volume of distribution at steady state.
Journal of Medicinal Chemistry Article
DOI: 10.1021/acs.jmedchem.5b00270
J. Med. Chem. XXXX, XXX, XXXXXX
K
MK3 ELISA photometer (Thermo Scientic). All experiments were
performed in triplicate. The IC50 values were calculated using GraphPad
Prism software.
Colony Formation Assay. Cells were seeded in six-well plates at a
density of 5000 per well and treated with vehicle, dasatinib, or 1j the next
day. The medium containing vehicle or 1j was replaced every 4 days.
Cells were xed with methanol and stained with crystal violet after
treatment for 12 days.
Wound Healing Assay. MDA-MB-231 cells were cultured to
conuence in 24-well plates and wounded using a sterilized pipet tip to
make a straight scratch. Cells were rinsed with physiological saline
gently, and then PBS was replaced with DMEM medium containing
vehicle, dasatinib, or 1j. Pictures were taken by an OLYMPUS digital
camera and analyzed by AxioVision Rel 4.8 (Carl Zeiss) after 20 h.
Transwell Invasion Assay. The cell invasion assay was performed
as described previously.
38
The cells were photographed with Leica
DM2500.
Cell Cycle Progression and Apoptosis Assays. A total of 2 ×105
MDA-MB-231 cells were plated in a six-well plate and treated with 1j
and dasatinib for 24 and 48 h at 37 °C. After incubation, the cells were
harvested and washed with ice-cold PBS. The cell cycle progression was
analyzed using a cell cycle and apoptosis analysis kit (Beyotime), and the
apoptosis ratio was performed with an annexin V-FITC Apoptosis
Detection Kit (keygentec).
Western Blot Analysis. After treatment with a series of con-
centrations of 1j for 20 h at 37 °C, MDA-MB-231 cells were harvested,
washed with ice-cold physiological saline, and lysed with RIPA lysis
buer (Beyotime) including 1% cocktail (Sigma-Aldrich). Whole-cell
protein lysates were prepared and centrifuged for 10 min at 12 000 rpm
and 4 °C to remove any insoluble material. The total proteins were
determined using the Bradford method, and an equivalent quantity of
protein was combined with an SDSPAGE loading buer (Beyotime)
in boiled water for 5 min. Cell lysates were separated by SDSPAGE
and electrotransferred onto PVDF membranes (Millipore). The PVDF
membranes were incubated with each antibody and detected according
to the immunoblot analysis principle. The antibodies were purchased
from Cell Signaling Technology, and the dilutions of the antibodies were
according to the instruction from Cell Signaling Technology.
In Vivo Models. The animal studies were conducted under the
approval of the Experimental Animal Management Committee of
Sichuan University. MDA-MB-231 cells were harvested during the
exponential-growth phase, washed 3 times with serum-free medium,
followed by resuspension at a concentration of 5 ×107per mL. A total of
100 μL of cell suspension was injected into SCID mice (56 weeks)
subcutaneously. After the tumors had grown to 150200 mm3, all the
mice were randomized into 6 groups (6 mice for each group) and dosed
with 1j (7.5, 15, or 30 mg kg1d1), dasatinib, paclitaxel (10 mg/kg/
week), or vehicle. The compounds were dissolved in sterilization
water with 25% (v/v) PEG400 plus 5% DMSO and administered orally.
Mice were monitored for side eects every day, and tumor growth was
measured every 3 days. The volume was calculated as follows: tumor
size = ab2/2 (a, long diameter; b, short diameter).
Histopathology and IHC. SCID mice bearing tumors were treated
with control or 1j as described before. At the indicated time after dosing,
individual mice were humanely euthanized. The tumors were xed with
formalin and embedded in paran. Sections measuring 48μmin
thickness were prepared for histological and immunostaining with the
Ki67 antibody (Thermo Fisher Scientic, Fremont, CA). Apoptosis was
determined using transferase-mediated dUTP nick-end labeling
(TUNEL) and AP-Red staining (Roche Applied Science). The tissues
of heart, liver, spleen, lung, and kidney were stained with hematoxylin
and eosin (H&E). Finally, images were acquired on an Olympus digital
camera attached to a light microscope.
Pharmacokinetic Assessments. The pharmacokinetics analysis of
1j was conducted in male SpragueDawley rats (Chinese Academy of
Medical Science, Beijing, China). Briey, catheters were surgically
placed into the jugular veins of the rats to collect serial blood samples.
The animals were administered a single dose of 10 mg/kg 1j by oral
gavage after fasting overnight. Blood was collected at the indicated
time and centrifuged immediately to isolate plasma. The plasma
concentrations were determined using high performance liquid
chromatography with tandem mass spectrometric detection (3200
QTRAP system, Applied Biosystems). Noncompartmental pharmaco-
kinetic parameters were tted using DAS software (Enterprise, version
2.0, Mathematical Pharmacology Professional Committee of China).
Chemistry Methods. All reagents and solvents were obtained from
commercial suppliers and used without further purication unless
otherwise indicated. Anhydrous solvents were dried and puried by
conventional methods prior use. Column chromatography was carried
out on silica gel (300400 mesh). All reactions were monitored by thin-
layer chromatography (TLC), and silica gel plates with uorescence
F-254 were used and visualized with UV light. All of the nal compounds
were puried to >95% purity, as determined by high-performance liquid
chromatography (HPLC). HPLC analysis was performed on a Waters
2695 HPLC system with the use of a Kromasil C18 reversed-column
(4.6 mm ×250 mm, 5 μm). The binary solvent system (A/B) was as
follows: 10 mmol of ammonium acetate in water (pH 9) (A) and
acetonitrile (B), A/B = 32/68. The absorbance was detected at 254 nm,
and the ow rate was 1 mL/min. 1H NMR and 13C NMR spectra were
recorded on a Bruker AV-400 spectrometer at 400 and 100 MHz,
respectively. Coupling constants (J) are expressed in hertz (Hz). Spin
multiplicities are described as s (singlet), br s (broad singlet), t (triplet),
q (quartet), and m (multiplet). Chemical shifts (δ) are listed in parts per
million (ppm) relative to tetramethylsilane (TMS) as an internal
standard. Mass spectral (MS) data were acquired on a Waters Q-TOF
Premier mass spectrometer (Micromass, Manchester, U.K.).
3-Iodo-1H-pyrazolo[3,4-d]pyrimidin-4-amine (2). A solution
of 1H-pyrazolo[3,4-d]pyrimidin-4-amine (20 g, 148 mmol) and
N-iodosuccinimide (50 g, 222 mmol) in DMF (150 mL) was stirred
at 80 °C for 1012 h under an N2atmosphere. A second batch of
N-iodosuccinimide (3.92 g, 2 mmol) was added and the solution stirred
for additional 12 h. Upon standing at room temperature, a precipitate
was formed which was separated by ltration and washed with
dimethylformamide and ethanol to aord 15.1 g of the title compound.
The ltrate was concentrated in vacuo to about one-half of the original
volume, and 500 mL of water was added. The precipitated product was
separated by ltration and washed with ethanol to aord a second batch
of the product (18.8 g, combined yield 33.9 g, 87.8% yield). 1H NMR
(400 MHz, DMSO-d6)δ11.06 (br s, 1H), 8.17 (s, 1H), 7.91 (br s, 1H),
6.76 (br s, 1H). MS m/z(ESI): 262.1 [M + H]+.
3-Iodo-1-isopropyl-1H-pyrazolo[3,4-d]pyrimidin-4-amine
(3a). To a suspension of 2(5.0 g, 19.2 mmol) in anhydrous N,N-
dimethylformamide (40 mL) under an N2atmosphere, potassium car-
bonate (5.3 g, 38.4 mmol) and 2-bromopropane (1.9 mL, 20.1 mmol)
were added sequentially. The resulting mixture was stirred at 80 °C for
5 h and then was allowed to cool to room temperature. The mixture was
ltered. The ltrate was concentrated in vacuo and then partitioned
between water and DCM. The organic layer was dried with sodium
sulfate and concentrated in vacuo. The crude product was puried using
silica gel chromatography with a methanol/dichloromethane gradient to
aord the desired product as a yellow solid (5.4 g, 92.8% yield). 1H
NMR (400 MHz, DMSO-d6)δ8.19 (s, 1H), 7.72 (br s, 1H), 6.72 (br s,
1H), 4.994.93 (m, 1H), 1.42 (d, J= 6.7 Hz, 6H). MS m/z(ESI): 304.0
[M + H]+. Compounds 3biwere synthesized by using a similar pro-
cedure; yield 51.192.0%.
1-Ethyl-3-iodo-1H-pyrazolo[3,4-d]pyrimidin-4-amine (3b).
1H NMR (400 MHz, DMSO-d6)δ8.20 (s, 1H), 7.82 (br s, 1H), 6.67
(br s, 1H), 4.30 (q, J= 7.2 Hz, 2H), 1.35 (t, J= 7.2 Hz, 3H). MS m/z
(ESI): 290.0 [M + H]+.
3-Iodo-1-methyl-1H-pyrazolo[3,4-d]pyrimidin-4-amine (3c).
1H NMR (400 MHz, DMSO-d6)δ8.21 (s, 1H), 7.87 (br s, 1H), 6.67
(br s, 1H), 3.88 (s, 3H). MS m/z(ESI): 275.9 [M + H]+.
1-Cyclobutyl-3-iodo-1H-pyrazolo[3,4-d]pyrimidin-4-amine
(3d). 1H NMR (400 MHz, DMSO-d6)δ8.19 (s, 1H), 7.69 (br s, 1H),
6.64 (br s, 1H), 5.325.13 (m, 1H), 2.712.54 (m, 2H), 2.36 (d, J= 4.7
Hz, 2H), 1.931.76 (m, 2H). MS m/z(ESI): 316.1 [M + H]+.
1-Cyclopentyl-3-iodo-1H-pyrazolo[3,4-d]pyrimidin-4-amine
(3e). 1H NMR (400 MHz, DMSO-d6)δ8.19 (s, 1H), 7.78 (br s, 1H),
6.70 (br s, 1H), 5.284.97 (m, 1H), 2.05 (dt, J= 12.1, 7.9 Hz, 2H),
Journal of Medicinal Chemistry Article
DOI: 10.1021/acs.jmedchem.5b00270
J. Med. Chem. XXXX, XXX, XXXXXX
L
1.991.88 (m, 2H), 1.881.78 (m, 2H), 1.721.59 (m, 2H). MS m/z
(ESI): 330.1 [M + H]+.
3-Iodo-1-(methoxymethyl)-1H-pyrazolo[3,4-d]pyrimidin-4-
amine (3f). 1H NMR (400 MHz, DMSO-d6)δ8.26 (s, 1H), 7.87 (br s,
1H), 6.70 (br s, 1H), 5.54 (s, 2H), 3.26 (s, 3H). MS m/z(ESI): 306.0
[M + H]+.
(S)-3-Iodo-1-(tetrahydrofuran-3-yl)-1H-pyrazolo[3,4-d]-
pyrimidin-4-amine (3g). 1H NMR (400 MHz, DMSO-d6)δ8.21 (s,
1H), 7.89 (br s, 1H), 6.62 (br s, 1H), 5.505.28 (m, 1H), 4.103.96
(m, 2H), 3.923.80 (m, 2H), 2.432.23 (m, 2H). MS m/z(ESI): 332.1
[M + H]+.
(R)-3-Iodo-1-(tetrahydrofuran-3-yl)-1H-pyrazolo[3,4-d]-
pyrimidin-4-amine (3h). 1H NMR (400 MHz, DMSO-d6)δ8.21 (s,
1H), 7.89 (br s, 1H), 6.63 (br s, 1H), 5.605.28 (m, 1H), 4.153.96
(m, 2H), 3.943.76 (m, 2H), 2.442.22 (m, 2H). MS m/z(ESI): 332.1
[M + H]+.
tert-Butyl 4-(4-Amino-3-iodo-1H-pyrazolo[3,4-d]pyrimidin-
1-yl)piperidine-1-carboxylate (3i). 1H NMR (400 MHz, DMSO-
d6)δ8.20(s, 1H), 7.79 (br s, 1H), 6.72 (br s, 1H), 4.874.75 (m, 1H),
4.06 (d, J= 10.8 Hz, 2H), 2.95 (br s, 2H), 1.991.81 (m, 4H), 1.43
(s, 9H). MS m/z(ESI): 445.3 [M + H]+.
3-Ethynyl-4-methylbenzoic Acid (6). 3-Iodo-4-methylbenzoate
(4). A solution of 3-iodo-4-methylbenzoic acid (15 g, 57.14 mmol)
and conc H2SO4(cat., 3 mL) in MeOH (100 mL) was heated at
65 °C for 24 h. The reaction mixture was cooled, and the solvent was
evaporated. The residue was dissolved in Et2O, washed with saturated
aqueous NaHCO3(3×), H2O, brine, dried over MgSO4, and the solvent
was evaporated to give the methyl 3-iodo-4-methylbenzoate 4(14.8 g,
94.0%) as an orange liquid.
Methyl 4-Methyl-3-((trimethylsilyl)ethynyl)benzoate (5). 4(14.8 g,
53.6 mmol), Pd(PPh3)4(3.3 g, 2.86 mmol), and CuI (1.1 g, 5.74 mmol)
were in suspended absolute THF (100 mL). The mixture underwent
three cycles of vacuum/lling with N2.Triethylamine(5mL,35.6mmol)
and trimethylsilylacetylene (2.65 mL, 18.7 mmol) were then added.
The mixture was stirred at room temperature overnight. Then the
mixture was ltered, ltrate concentrated in vacuo, and the residue
was diluted with EA (3×) and 0.5 M ammonia solution for extraction.
The combined organic phases were dried over sodium sulfate, ltered,
and concentrated in vacuo. The crude product was puried using silica
gel chromatography with a petroleum ether/ethyl acetate gradient to
aord the methyl 4-methyl-3-((trimethylsilyl)ethynyl)benzoate (5)asa
yellow liquid.
5was dissolved in methanol (80 mL). K2CO3(7.4 g, 53.6 mmol) was
added and stirred for 10 min at rt. Then to the mixture were added
NaOH (2.1 g, 53.6 mmol) and water (20 mL), and the mixture was
stirred for another 3 h. The mixture was acidied with hydrochloric acid
(pH 4) and extracted with DCM (3×). The combined organic phases
were washed with saturated sodium chloride solution and concentrated
under reduced pressure. Chromatographic purication through a short
silica gel frit with a petroleum ether/ethyl acetate gradient yields 6(6.8 g,
74.1% yield). 1H NMR (400 MHz, DMSO-d6)δ13.04 (br s, 1H), 7.93
(s, 1H), 7.85 (d, J= 8.0 Hz, 1H), 7.43 (d, J= 8.0 Hz, 1H), 4.48 (s, 1H),
2.45 (s, 3H). MS m/z(ESI): 159.0 [M H].
4-((4-Methylpiperazin-1-yl)methyl)-3-(triuoromethyl)-
aniline (9a). 4-Amino-2-trifluoromethylbenzaldehyde (8a). 4-Amino-2-
(triuoromethyl)benzonitrile (7a) (11.2 g, 60 mmol) was charged into a
500 mL three-necked, round-bottomed ask and underwent three cycles
of vacuum/lling with N2. Dry THF (100 mL) was then added and a
solution of diisobutylaluminum hydride (100 mL, 1.5 M in toluene) was
added at 2330 °C over 20 min. Upon complete addition, the resulting
solution was stirred for an additional 30 min. After the reaction was
complete, the mixture was cooled to 10 °C and methanol (18 mL) was
carefully added. Then the mixture was stirred for an additional 2 h at rt
and an aqueous saturated Rochelle salt solution was added dropwise.
After the quench was complete, the mixture was stirred at 4550 °C for
10 min. tert-Butyl methyl ether (150 mL) was added and stirred for
10 min. Organic layers were separated, and more tert-butyl methyl ether
was added for extraction. The combined organic phases were dried over
MgSO4and concentrated under vacuum. The residue was used directly
in the next step. MS m/z(ESI): 188.0 [M H].
4-Amino-2-triuoromethylbenzaldehyde (8a) (1.13 g, 6 mmol) was
dissolved in ethyl acetate (35 mL), and aqueous 1 M HCl solution
(20 mL) was added. The mixture was stirred rapidly at 2035 °C for
5 min. A solution of aqueous 1 M NaOH (15 mL) was charged to the
mixture and stirred for an additional 5 min. The organic layer was
separated, washed with 10% (w/w) aqueous NaCl solution (20 mL),
and charged to a 100 mL reactor. 1-Methylpiperazine (3.0 g, 30 mmol),
pyridinium p-toluenesulfonate (150.8 mg, 0.6 mmol), and toluene
(15 mL) were charged. The mixture underwent azeotropic distillation
under DeanStark conditions with a nal vessel temperature of 102
110 °C. After each 15 mL portion of distillate had been collected, fresh
ethyl acetate (15 mL) was charged to the reaction mixture. This
distillationaddition operation was repeated until a total of 45 mL of
ethyl acetate was collected. The mixture was cooled to 50 °C, and
sodium triacetoxyborohydride (2.54 g, 12 mmol) was added in portions
at 5060 °C. The reaction mixture was cooled to 1015 °C and
quenched with water (25 mL) over 10 min while maintaining the batch
temperature below 20 °C. The organic layer was separated, washed with
water (2 ×25 mL), dried over MgSO4, and concentrated in vacuo. The
residue was washed with petroleum ether/ethyl acetate (v/v = 5), and
the title compound 9a was obtained as a white solid (1.26 g, 76.1%
yield). 1H NMR (400 MHz, CDCl3): δ7.47 (d, J= 8.4 Hz, 1H), 6.91 (d,
J= 2.4 Hz, 1H), 6.79 (dd, J= 8.4, 2.4 Hz, 1H), 3.76 (br s, 2H), 3.52 (s,
2H), 2.702.35 (m, 8H), 2.28 (s, 3H); MS (ESI) m/z274.2 [M + H]+.
Compounds 9cdand 9fhwere synthesized by using a similar
procedure; yield 51.185.0%.
4-(Morpholinomethyl)-3-(triuoromethyl)aniline (9f). 1H
NMR (400 MHz, DMSO-d6)δ7.30 (d, J= 8.4 Hz, 1H), 6.85 (d, J=
2.1 Hz, 1H), 6.726.75 (m, 1H), 5.45 (s, 2H), 3.603.50 (m, 4H), 3.39
(s, 2H), 2.402.30 (m, 4H). MS (ESI) m/z261.1 [M + H]+.
4-((4-Ethylpiperazin-1-yl)methyl)-3-(triuoromethyl)aniline
(9g). 1H NMR (400 MHz, DMSO-d6)δ7.29 (d, J= 8.2 Hz, 1H), 6.87
(d, J= 2.2 Hz, 1H), 6.76 (dd, J= 8.2, 2.2 Hz, 1H,), 5.42 (s, 2H), 3.39 (s,
2H), 2.342.26 (m, 10H), 0.97 (t, J= 5 Hz, 3H). MS (ESI) m/z288.2
[M + H]+.
2-(4-(4-Amino-2-(triuoromethyl)benzyl)piperazin-1-yl)-
ethan-1-ol (9h). 1H NMR (400 MHz, CDCl3)δ7.47 (d, J= 8.3 Hz,
1H), 6.92 (d, J= 2.0 Hz, 1H), 6.79 (dd, J= 8.2, 2.0 Hz, 1H), 3.77 (br s,
2H), 3.60 (t, J= 5.4 Hz, 2H), 3.53 (s, 2H), 2.76 (br s, 1H), 2.562.49
(m, 10H). MS (ESI) m/z304.2 [M + H]+.
3-Ethynyl-4-methyl-N-(4-((4-methylpiperazin-1-yl)methyl)-
3-(triuoromethyl)phenyl)benzamide (10a). 3-Ethynyl-4-methyl-
benzoic acid 6(571 mg, 3.56 mmol) was reuxed in SOCl2for 2 h,
concentrated in vacuo, and redissolved in ethyl acetate (8 mL) to aord
3-ethynyl-4-methylbenzoyl chloride solution.
3-((4-Methylpiperazin-1-yl)methyl)-5-(triuoromethyl)aniline 9a
(812 mg, 2.97 mmol) and DIPEA (806 mg, 6.24 mmol) were dissolved
in ethyl acetate (10 mL) and cooled to 0 °C. Then 3-ethynyl-4-
methylbenzoyl chloride ethyl acetate solution was dropwise added,
resulting in an o-white suspension. The suspension was allowed to stir
for 1 h at rt. To the reaction mixture was added 30 mL of water, and
the aqueous phase was extracted with ethyl acetate (3 ×20 mL). The
combined organic phase was separated, dried over anhydrous sodium
sulfate, ltered, and the ltrate was evaporated to dryness under reduced
pressure to give a solid residue. The solid was puried by ash column
chromatography using MeOH in dichloromethane to give 10a as an o-
white solid (1.1 g, 88% yield). 1H NMR (400 MHz, CDCl3)δ8.33 (s,
1H), 7.91 (s, 1H), 7.86 (d, J= 13.7 Hz, 2H), 7.73 (t, J= 7.1 Hz, 2H), 7.28
(s, 1H), 3.60 (s, 2H), 3.32 (s, 1H), 2.48 (br s, 11H), 2.30 (s, 3H). MS
m/z(ESI): 416.3 [M + H]+. Compounds 10bgwere synthesized by
using a similar procedure; yield 79.188.2%.
3-Ethynyl-4-methyl-N-(4-((4-methylpiperazin-1-yl)methyl)-
phenyl)benzamide (10b). 1H NMR (400 MHz, CDCl3)δ7.91 (d, J=
10.3 Hz, 2H), 7.76 (d, J= 7.8 Hz, 1H), 7.57 (d, J= 7.7 Hz, 2H), 7.31 (d,
J= 7.7 Hz, 3H), 3.49 (s, 2H), 3.35 (s, 1H), 2.51 (s, 3H), 2.48 (br s, 8H),
2.32 (s, 3H). MS m/z(ESI): 348.2 [M + H]+.
N-(3-Chloro-4-((4-methylpiperazin-1-yl)methyl)phenyl)-3-
ethynyl-4-methylbenzamide (10c). 1H NMR (400 MHz, CDCl3)δ
8.86 (s, 1H), 7.99 (s, 1H), 7.89 (s, 1H), 7.81 (d, J= 7.9 Hz, 1H), 7.49 (d,
Journal of Medicinal Chemistry Article
DOI: 10.1021/acs.jmedchem.5b00270
J. Med. Chem. XXXX, XXX, XXXXXX
M
J= 8.3 Hz, 1H), 7.357.29 (m, 2H), 3.64 (s, 2H), 3.33 (s, 1H), 3.03
2.78 (m, 11H), 2.47 (s, 3H). MS m/z(ESI): 382.2 [M + H]+.
N-(3-Bromo-4-((4-methylpiperazin-1-yl)methyl)phenyl)-3-
ethynyl-4-methylbenzamide (10d). 1H NMR (400 MHz, CDCl3)δ
9.30 (s, 1H), 8.10 (s, 1H), 8.03 (s, 1H), 7.86 (d, J= 7.9 Hz, 1H), 7.63 (d,
J= 8.4 Hz, 1H), 7.32 (d, J= 8.4 Hz, 1H), 7.27 (d, J= 7.9 Hz, 1H), 3.63 (s,
2H), 3.32 (s, 1H), 3.042.94 (m, 8H), 2.82 (s, 3H), 2.48 (s, 3H). MS
m/z(ESI): 426.1 [M + H]+.
3-Ethynyl-4-methyl-N-(3-(triuoromethyl)phenyl)-
benzamide (10e). 1H NMR (400 MHz, DMSO-d6)δ10.55 (s, 1H),
8.25 (s, 1H), 8.11 (s, 1H), 8.07 (d, J= 8.4 Hz, 1H), 7.92 (d, J= 8.0 Hz,
1H), 7.60 (d, J= 8.0 Hz, 1H), 7.47 (t, J= 8.8 Hz, 2H), 4.54 (s, 1H), 2.47
(s, 3H). MS m/z(ESI): 304.1 [M + H]+.
3-Ethynyl-4-methyl-N-(4-(morpholinomethyl)-3-
(triuoromethyl)phenyl)benzamide (10f). 1H NMR (400 MHz,
CDCl3)δ7.98 (s, 1H), 7.95 (s, 1H), 7.88 (d, J= 8.0 Hz, 1H), 7.87 (s,
1H), 7.827.75 (m, 2H), 7.34 (d, J= 8.0 Hz, 1H), 3.813.66 (m, 4H),
3.63 (s, 2H), 3.36 (s, 1H), 2.52 (s, 3H), 2.512.41 (m, 4H). MS m/z
(ESI): 403.2 [M + H]+.
N-(4-((4-Ethylpiperazin-1-yl)methyl)-3-(triuoromethyl)-
phenyl)-3-ethynyl-4-methylbenzamide Hydrochloride Salt
(10g). 1H NMR (400 MHz, DMSO-d6)δ10.57 (s, 1H), 10.24 (br s,
1H), 8.22 (s, 1H), 8.11 (d, J= 8.9 Hz, 2H), 7.92 (d, J= 8.2 Hz, 1H), 7.72
(d, J= 8.5 Hz, 1H), 7.48 (d, J= 8.2 Hz, 1H), 4.55 (s, 1H), 3.67 (s, 2H),
3.43 (d, J= 12.1 Hz, 2H), 3.183.04 (m, 2H), 3.032.81 (m, 6H), 2.47
(s, 3H), 1.24 (t, J= 7.2 Hz, 3H). MS m/z(ESI): 430.2 [M + H]+.
3-Ethynyl-N-(4-((4-(2-hydroxyethyl)piperazin-1-yl)methyl)-
3-(triuoromethyl)phenyl)-4-methylbenzamide (10h). 3-Ethynyl-
4-methylbenzoic acid 6(1.04 g, 6.5 mmol) was reuxed in SOCl2for 2 h,
concentrated in vacuo, and redissolved in ethyl acetate (15 mL) to aord
3-ethynyl-4-methylbenzoyl chloride solution.
2-(4-(4-Amino-2-(triuoromethyl)benzyl)piperazin-1-yl)ethan-1-ol
9h (910 mg, 3 mmol) and DIPEA (840 mg, 6.5 mmol) were dissolved in
ethyl acetate (20 mL) and cooled to 0 °C. Then 3-ethynyl-4-
methylbenzoyl chlorideethyl acetate solution was dropwise added,
resultinginano-white suspension. The suspension was allowed to stir for
1 h at rt. To the reaction mixture was added 40 mL of water, and the
aqueous phase was extracted with ethyl acetate (3×30 mL). The combined
organic phase was separated, dried over anhydrous sodium sulfate, ltered,
and the ltrate was evaporated to dryness under reduced pressure to give a
solid residue. The solid combined with LiOH (500 mg) was dissolved
in THF/H2O (v/v = 3, 16 mL) and was stirred at 25 °C overnight. The
mixture was diluted with EA (3×30 mL) and brine (30 mL) for extraction.
Combined organic phases were dried over MgSO4, concentrated in vacuo.
The residue was puried by ash column chromatography using MeOH in
dichloromethane to give 10h as a solid (1.65 g, 61.7% yield).
3-((4-Amino-1-isopropyl-1H-pyrazolo[3,4-d]pyrimidin-3-yl)-
ethynyl)-4-methyl-N-(4-((4-methylpiperazin-1-yl)methyl)-3-
(triuoromethyl)phenyl)benzamide (1a). 3a (151.6 mg, 0.5 mmol),
10a (207.7 mg, 0.5 mmol), CuI (9.5 mg, 0.05 mmol), and (PPh3)2PdCl2
(17.5 mg, 0.025 mmol) were suspended in anhydrous DMF (3 mL). The
mixture underwent three cycles of vacuum/lling with N2. Then DIPEA
(165 μL, 1 mmol) was added with syringe. The mixture was stirred at
80 °C for 8 h and then quenched with water. DCM and 5% ammonia were
added for extraction. The combined organic layer was concentrated in
vacuo and the crude product was puried using silica gel chromatography
with a methanol/dichloromethane gradient to aord the title compound
1a as a white solid (136 mg, 46.0% yield). 1H NMR (400 MHz, DMSO-
d6)δ10.58 (s, 1H), 8.29 (s, 1H), 8.18 (s, 2H), 8.05 (d, J= 8.3 Hz, 1H),
7.91 (d, J= 8.0 Hz, 1H), 7.64 (d, J= 8.3 Hz, 1H), 7.46 (d, J= 8.0 Hz, 1H),
5.014.95 (m, 1H), 3.58 (s, 2H), 3.04 (br s, 4H), 2.62 (s, 3H), 2.56 (br s,
4H), 2.43 (s, 3H), 1.40 (d, J= 6.6 Hz, 6H). 13C NMR (100 MHz, DMSO-
d6)δ164.72, 157.80, 156.29, 152.32, 144.14, 138.53, 132.03, 131.46,
131.37, 131.09, 129.97, 128.75, 127.55 (d, J= 29.5 Hz), 124.89, 123.57,
122.93, 121.57, 117.42, 100.77, 91.09, 85.55, 56.65, 52.79, 49.54, 48.89,
42.71, 42.36, 21.76, 20.54. HRMS m/z(ESI) calcd for C31H34F3N8O
[M + H]+591.2808; found, 591.2806. Compounds 1bpwere synthesized
by using a similar procedure; yield 39.058.1%.
3-((4-Amino-1-isopropyl-1H-pyrazolo[3,4-d]pyrimidin-3-yl)-
ethynyl)-4-methyl-N-(4-((4-methylpiperazin-1-yl)methyl)-
phenyl)benzamide (1b). 1H NMR (400 MHz, DMSO-d6)δ10.54 (s,
1H), 8.288.25 (m, 3H), 8.09 (s, 1H), 7.94 (d, J= 7.9 Hz, 1H), 7.87 (br
s, 1H), 7.75 (d, J= 8.2 Hz, 1H), 7.36 (d, J= 8.2 Hz, 1H), 5.095.03 (m,
1H), 3.70 (s, 2H), 2.49 (s, 3H), 2.46 (br s, 8H), 2.24 (s, 3H), 1.49 (d, J=
6.4 Hz, 6H). 13C NMR (100 MHz, DMSO-d6)δ164.00, 157.87, 156.26,
152.28, 141.15, 136.75, 135.52, 133.62, 131.61, 130.74, 130.01, 127.29
(d, J= 30.1 Hz), 125.10, 125.02, 123.80, 121.76, 121.32, 100.80, 91.76,
84.76, 57.40, 54.47, 52.40, 48.87, 45.33, 21.74, 19.89. HRMS m/z(ESI)
calcd for C30H35N8O[M+H]
+523.2934; found, 523.2928.
3-((4-Amino-1-isopropyl-1H-pyrazolo[3,4-d]pyrimidin-3-yl)-
ethynyl)-N-(3-chloro-4-((4-methylpiperazin-1-yl)methyl)-
phenyl)-4-methylbenzamide (1c). 1H NMR (400 MHz, DMSO-d6)
δ10.43 (s, 1H), 8.32 (s, 1H), 8.26 (s, 1H), 7.98 (s, 1H), 7.94 (d, J= 8.0
Hz, 1H), 7.72 (d, J= 8.4 Hz, 1H), 7.54 (d, J= 8.0 Hz, 1H), 7.44 (d, J=
8.4 Hz, 1H), 5.134.99 (m, 1H), 3.53 (s, 2H), 2.58 (s, 3H), 2.43 (br s,
8H), 2.20 (s, 3H), 1.49 (d, J= 6.6 Hz, 6H). HRMS m/z(ESI) calcd for
C30H34ClN8O[M+H]
+557.2544; found, 557.2538.
3-((4-Amino-1-isopropyl-1H-pyrazolo[3,4-d]pyrimidin-3-yl)-
ethynyl)-N-(3-bromo-4-((4-methylpiperazin-1-yl)methyl)-
phenyl)-4-methylbenzamide (1d). 1H NMR (400 MHz, DMSO-d6)
δ10.44 (s, 1H), 8.33 (s, 1H), 8.27 (s, 1H), 8.16 (s, 1H), 7.95 (d, J= 8.0
Hz, 1H), 7.80 (d, J= 8.2 Hz, 1H), 7.54 (d, J= 8.2 Hz, 1H), 7.44 (d, J=
8.0 Hz, 1H), 5.194.94 (m, 1H), 3.56 (s, 2H), 2.73 (br s, 11H), 2.58 (s,
3H), 1.49 (d, J= 6.0 Hz, 6H). HRMS m/z(ESI) calcd for C30H34BrN8O
[M + H]+601.2039; found, 601.2040.
3-((4-Amino-1-isopropyl-1H-pyrazolo[3,4-d]pyrimidin-3-yl)-
ethynyl)-4-methyl-N-(3-(triuoromethyl)phenyl)benzamide
(1e). 1H NMR (400 MHz, DMSO-d6)δ10.63 (s, 1H), 8.36 (s, 1H),
8.26 (s, 2H), 8.09 (d, J= 7.9 Hz, 1H), 7.97 (d, J= 7.7 Hz, 1H), 7.88 (br s,
1H), 7.61 (t, J= 7.9 Hz, 1H), 7.54 (d, J= 7.9 Hz, 1H), 7.46 (d, J= 7.5 Hz,
1H), 6.74 (br s, 1H), 5.204.92 (m, 1H), 2.58 (s, 3H), 1.48 (d, J= 6.4
Hz, 6H). HRMS m/z(ESI) calcd for C25H22F3N6O[M+H]
+479.1807;
found, 479.1813.
3-((4-Amino-1-isopropyl-1H-pyrazolo[3,4-d]pyrimidin-3-yl)-
ethynyl)-4-methyl-N-(4-(morpholinomethyl)-3-(triuoromethyl)-
phenyl)benzamide (1f). 1H NMR (400 MHz, DMSO-d6)δ10.56 (s,
1H), 8.34 (s, 1H), 8.27 (s, 1H), 8.23 (s, 1H), 8.09 (d, J= 8.3 Hz, 1H),
7.97 (d, J= 7.9 Hz, 1H), 7.75 (d, J= 8.3 Hz, 1H), 7.55 (d, J= 7.9 Hz,
1H), 5.125.03 (m, 1H), 3.59 (s, 6H), 2.59 (s, 3H), 2.39 (s, 4H), 1.49
(d, J= 6.8 Hz, 6H). HRMS m/z(ESI) calcd for C30H31F3N7O2[M + H]+
578.2491; found, 578.2488.
3-((4-Amino-1-isopropyl-1H-pyrazolo[3,4-d]pyrimidin-3-yl)-
ethynyl)-N-(4-((4-ethylpiperazin-1-yl)methyl)-3-
(triuoromethyl)phenyl)-4-methylbenzamide (1g). 1HNMR
(400 MHz, DMSO-d6)δ10.59 (s, 1H), 8.35 (s, 1H), 8.27 (s, 1H),
8.24 (s, 1H), 8.10 (d, J= 8.2 Hz, 1H), 7.98 (dd, J= 7.9, 1.1 Hz, 1H), 7.73
(d, J= 8.6 Hz, 1H), 7.55 (d, J= 8.2 Hz, 1H), 5.105.03 (m, 1H), 3.62 (s,
2H), 2.59 (s, 3H), 2.55 (br s, 10H), 1.49 (d, J= 6.7 Hz, 6H), 1.10 (t, J=
6.9 Hz, 3H). HRMS m/z(ESI) calcd for C32H36F3N8O[M+H]
+
605.2964; found, 605.2962.
3-((4-Amino-1-isopropyl-1H-pyrazolo[3,4-d]pyrimidin-3-yl)-
ethynyl)-N-(4-((4-(2-hydroxyethyl)piperazin-1-yl)methyl)-3-
(triuoromethyl)phenyl)-4-methylbenzamide (1h). 1HNMR
(400 MHz, DMSO-d6)δ10.56 (s, 1H), 8.34 (s, 1H), 8.26 (s, 1H),
8.22 (s, 1H), 8.07 (d, J= 8.2 Hz, 1H), 7.96 (d, J= 8.0 Hz, 1H), 7.72 (d,
J= 8.2 Hz, 1H), 7.55 (d, J= 8.0 Hz, 1H), 5.194.94 (m, 1H), 4.37 (s,
1H), 3.56 (s, 2H), 3.48 (d, J= 5.4 Hz, 2H), 2.58 (s, 3H), 2.38 (br s, J=
6.1 Hz, 10H), 1.49 (d, J= 6.3 Hz, 6H). HRMS m/z(ESI) calcd for
C32H36F3N8O2[M + H]+621.2913; found, 621.2917.
3-((4-Amino-1-methyl-1H-pyrazolo[3,4-d]pyrimidin-3-yl)-
ethynyl)-4-methyl-N-(4-((4-methylpiperazin-1-yl)methyl)-3-
(triuoromethyl)phenyl)benzamide (1i). 1H NMR (400 MHz,
DMSO-d6)δ10.60 (s, 1H), 8.33 (s, 1H), 8.26 (s, 1H), 8.22 (s, 1H), 8.09
(d, J= 8.5 Hz, 1H), 7.95 (d, J= 8.1 Hz, 1H), 7.69 (d, J= 8.5 Hz, 1H),
7.52 (d, J= 8.1 Hz, 1H), 3.93 (s, 3H), 3.62 (s, 2H), 2.92 (br s, 4H), 2.58
(br s, 7H), 2.48 (s, 3H). 13C NMR (100 MHz, DMSO-d6)δ164.79,
157.79, 156.57, 153.26, 144.19, 138.48, 132.12, 131.46, 131.38, 131.30,
129.96, 128.69, 127.53 (d, J= 31.0 Hz), 125.04, 123.52, 121.61, 117.36,
117.30, 99.55, 91.12, 85.41, 56.82, 53.27, 50.27, 33.97, 20.50, 17.33.
HRMS m/z(ESI) calcd for C29H30F3N8O[M+H]
+563.2495; found,
563.2497.
Journal of Medicinal Chemistry Article
DOI: 10.1021/acs.jmedchem.5b00270
J. Med. Chem. XXXX, XXX, XXXXXX
N
3-((4-Amino-1-ethyl-1H-pyrazolo[3,4-d]pyrimidin-3-yl)-
ethynyl)-4-methyl-N-(4-((4-methylpiperazin-1-yl)methyl)-3-
(triuoromethyl)phenyl)benzamide (1j). 1H NMR (400 MHz,
DMSO-d6)δ10.56 (s, 1H), 8.34 (s, 1H), 8.27 (s, 1H), 8.22 (d, J= 1.7
Hz, 1H), 8.07 (d, J= 8.5 Hz, 1H), 7.97 (d, J= 8.1 Hz, 1H), 7.72 (d, J=
8.5 Hz, 1H), 7.55 (d, J= 8.1 Hz, 1H), 4.38 (q, J= 7.2 Hz, 2H), 3.57 (s,
2H), 2.58 (s, 3H), 2.39 (br s, 8H), 2.16 (s, 3H), 1.42 (t, J= 7.2 Hz, 3H).
13C NMR (100 MHz, DMSO-d6)δ164.77, 157.86, 156.52, 152.75,
144.17, 138.19, 132.23, 132.20, 131.38, 131.32, 129.99, 128.72, 127.75
(d, J= 30.0 Hz), 125.11, 123.56, 123.02, 121.62, 117.31, 100.19, 91.18,
85.47, 57.50, 54.78, 52.75, 45.78, 41.99, 20.55, 14.68. HRMS m/z(ESI)
calcd for C30H32F3N8O[M+H]
+577.2651; found, 577.2652.
3-((4-Amino-1-cyclobutyl-1H-pyrazolo[3,4-d]pyrimidin-3-yl)-
ethynyl)-4-methyl-N-(4-((4-methylpiperazin-1-yl)methyl)-3-
(triuoromethyl)phenyl)benzamide (1k). 1H NMR (400 MHz,
DMSO-d6)δ10.56 (s, 1H), 8.35 (s, 1H), 8.26 (s, 1H), 8.22 (s, 1H), 8.08
(d, J= 8.5 Hz, 1H), 7.97 (d, J= 8.0 Hz, 1H), 7.72 (d, J= 8.5 Hz, 1H),
7.55 (d, J= 8.0 Hz, 1H), 5.385.29 (m, 1H), 3.57 (s, 2H), 2.762.62
(m, 2H), 2.59 (s, 3H), 2.41 (br s, 10H), 2.18 (s, 3H), 1.951.80 (m,
2H). 13C NMR (100 MHz, DMSO-d6)δ164.66, 157.82, 156.43, 152.71,
144.14, 138.18, 132.13, 132.10, 131.33, 131.26, 129.95, 128.74, 127.42
(d, J= 30.0 Hz), 125.30, 123.51, 122.98, 121.54, 117.27, 100.77, 91.25,
85.45, 57.43, 54.68, 52.61, 50.39, 45.62, 29.36, 20.53, 14.41. HRMS m/z
(ESI) calcd for C32H34F3N8O[M+H]
+603.2808; found, 603.2808.
3-((4-Amino-1-cyclopentyl-1H-pyrazolo[3,4-d]pyrimidin-3-
yl)ethynyl)-4-methyl-N-(4-((4-methylpiperazin-1-yl)methyl)-3-
(triuoromethyl)phenyl)benzamide (1l). 1H NMR (400 MHz,
DMSO-d6)δ10.59 (s, 1H), 8.33 (s, 1H), 8.25 (s, 1H), 8.22 (s, 1H), 8.11
(d, J= 8.5 Hz, 1H), 7.95 (d, J= 7.9 Hz, 1H), 7.70 (d, J= 8.5 Hz, 1H),
7.54 (d, J= 7.9 Hz, 1H), 5.295.14 (m, 1H), 3.65 (s, 2H), 3.032.87
(m, 4H), 2.75 (s, 3H), 2.57 (s, 3H), 2.452.26 (m, 4H), 2.142.07 (m,
2H), 2.051.93 (m, 2H), 1.931.78 (m, 2H), 1.741.61 (m, 2H).
HRMS m/z(ESI) calcd for C33H36F3N8O[M+H]
+617.2964; found,
617.2968.
3-((4-Amino-1-(methoxymethyl)-1H-pyrazolo[3,4-d]-
pyrimidin-3-yl)ethynyl)-4-methyl-N-(4-((4-methylpiperazin-1-
yl)methyl)-3-(triuoromethyl)phenyl)benzamide (1m). 1H NMR
(400 MHz, DMSO-d6)δ10.57 (s, 1H), 8.36 (s, 1H), 8.32 (s, 1H), 8.22
(s, 1H), 8.07 (d, J= 8.3 Hz, 1H), 7.98 (d, J= 7.9 Hz, 1H), 7.72 (d, J= 8.3
Hz, 1H), 7.55 (d, J= 7.9 Hz, 1H), 5.63 (s, 2H), 3.58 (s, 2H), 3.31 (s,
3H), 2.59 (s, 3H), 2.40 (br s, 8H), 2.19 (s, 3H). HRMS m/z(ESI) calcd
for C30H32F3N8O2[M + H]+593.2600; found, 593.2597.
(S)-3-((4-Amino-1-(tetrahydrofuran-3-yl)-1H-pyrazolo[3,4-d]-
pyrimidin-3-yl)ethynyl)-4-methyl-N-(4-((4-methylpiperazin-1-
yl)methyl)-3-(triuoromethyl)phenyl)benzamide (1n). 1H NMR
(400 MHz, DMSO-d6)δ10.59 (s, 1H), 8.36 (s, 1H), 8.28 (s, 1H), 8.23
(s, 1H), 8.10 (d, J= 8.0 Hz, 1H), 7.97 (d, J= 7.6 Hz, 1H), 7.72 (d, J= 8.0
Hz, 1H), 7.55 (d, J= 7.6 Hz, 1H), 5.585.41 (m, 1H), 4.183.98 (m,
2H), 3.91 (dd, J= 13.4, 9.1 Hz, 2H), 3.62 (s, 2H), 2.59 (s, 3H), 2.54
(s, 3H), 2.44 (br s, 8H), 2.382.29 (m, 2H). 13C NMR (100 MHz,
DMSO-d6)δ164.63, 157.85, 156.51, 153.17, 144.19, 138.18, 132.12,
131.34, 131.25, 129.98, 129.94, 128.80, 127.42 (d, J= 29.3 Hz), 125.45,
123.52, 123.01, 121.52, 117.27, 100.83, 91.37, 85.36, 71.57, 67.39, 57.48,
54.76, 52.73, 45.75, 31.82, 20.56, 14.66. HRMS m/z(ESI) calcd for
C32H34F3N8O2[M + H]+619.2757; found, 619.2756.
(R)-3-((4-Amino-1-(tetrahydrofuran-3-yl)-1H-pyrazolo[3,4-
d]pyrimidin-3-yl)ethynyl)-4-methyl-N-(4-((4-methylpiperazin-
1-yl)methyl)-3-(triuoromethyl)phenyl)benzamide (1o). 1H
NMR (400 MHz, DMSO-d6)δ10.60 (s, 1H), 9.98 (br s, 1H), 9.44
(br s, 1H), 8.35 (s, 1H), 8.28 (s, 1H), 8.24 (s, 1H), 8.12 (d, J= 8.2 Hz,
1H), 7.98 (d, J= 8.0 Hz, 1H), 7.72 (d, J= 8.2 Hz, 1H), 7.55 (d, J= 8.0
Hz, 1H), 5.615.36 (m, 1H), 4.193.98 (m, 2H), 3.91 (dd, J= 12.3, 7.6
Hz, 2H), 3.67 (s, 2H), 3.08 (s, 6H), 2.91 (br s, 2H), 2.74 (s, 3H), 2.59 (s,
3H), 2.432.33 (m, 2H). HRMS m/z(ESI) calcd for C32H34F3N8O2
[M + H]+619.2757; found, 619.2761.
tert-Butyl 4-(4-Amino-3-((2-methyl-5-((4-((4-methylpipera-
zin-1-yl)methyl)-3-(triuoromethyl)phenyl)carbamoyl)-
phenyl)ethynyl)-1H-pyrazolo[3,4-d]pyrimidin-1-yl)piperidine-
1-carboxylate (1p). 1H NMR (400 MHz, DMSO-d6)δ10.60 (s, 1H),
8.33 (s, 1H), 8.24 (s, 1H), 8.22 (s, 1H), 8.09 (d, J= 8.0 Hz, 1H), 7.95 (d,
J= 7.7 Hz, 1H), 7.69 (d, J= 8.0 Hz, 1H), 7.52 (d, J= 7.7 Hz, 1H),
4.984.78 (m, 1H), 4.104.01 (m, 2H), 3.61 (s, 2H), 3.032.81 (m,
6H), 2.55 (br s, 7H), 2.48 (s, 3H), 1.991.90 (m, 4H), 1.41 (s, 9H).
HRMS m/z(ESI) calcd for C38H45F3N8O[M+H]
+732.3597; found,
732.3598.
3-((4-Amino-1-(piperidin-4-yl)-1H-pyrazolo[3,4-d]pyrimidin-
3-yl)ethynyl)-4-methyl-N-(4-((4-methylpiperazin-1-yl)methyl)-
3-(triuoromethyl)phenyl)benzamide (1q). 1p (1.46 g, 2 mmol)
was dissolved in DCM (20 mL) and cooled to 0 °C. Then 10 mL of
CF3COOH was slowly added and stirred at rt for 2 h. The mixture was
concentrated and suspended in water. Aqueous sodium hydrogen
carbonate was added and adjusted to pH 8. Then the mixture was
ltered and washed with water. The solid was dried in vacuo and
recrystallized in acetone. Then the mixture was ltered to aord 1q as a
light yellow solid (1.07 g, 84.9% yield). 1H NMR (400 MHz, DMSO-d6)
δ10.56 (s, 1H), 8.35 (s, 1H), 8.28 (s, 1H), 8.22 (s, 1H), 8.08 (d, J= 7.9
Hz, 1H), 7.98 (d, J= 7.9 Hz, 1H), 7.72 (d, J= 8.2 Hz, 1H), 7.55 (d, J=
8.2 Hz, 1H), 5.094.98 (m, 1H), 4.364.32 (m, 1H), 3.58 (s, 2H),
3.213.08 (m, 2H), 2.99 (d, J= 7.0 Hz, 2H), 2.58 (s, 3H), 2.42 (br s,
8H), 2.31 (d, J= 11.7 Hz, 2H), 2.22 (s, 3H), 2.12 (d, J= 11.6 Hz, 2H).
HRMS m/z(ESI) calcd for C33H37F3N9O[M+H]
+632.3073; found,
632.3071.
3-((1-(1-Acryloylpiperidin-4-yl)-4-amino-1H-pyrazolo[3,4-d]-
pyrimidin-3-yl)ethynyl)-4-methyl-N-(4-((4-methylpiperazin-1-
yl)methyl)-3-(triuoromethyl)phenyl)benzamide (1r). 1q (510 mg,
0.807 mmol) and triethylamine (223 μL, 1.6 mmol) were dissolved in
10 mL of DCM. Then the mixture was cooled to 10 °C, and to it was
dropped a solution of acrylyl chloride (72 μL, 0.888 mmol) in
dichloromethane (2 mL) within 0.5 h. Then the mixture was stirred at rt
for an additional 0.5 h and quenched with water. DCM and more water
were added for extraction. The combined organic layer was dried with
sodium sulfate, ltered, concentrated, and the crude product was puried
using silica gel chromatography with a methanol/dichloromethane gradient
to aord the 1r as a yellow solid (440 mg, 72.3%). 1H NMR (400 MHz,
DMSO-d6)δ10.54 (s, 1H), 8.33 (s, 1H), 8.27 (s, 1H), 8.21 (s, 1H), 8.06 (d,
J=7.7Hz,1H),7.96(d,J= 7.7 Hz, 1H), 7.71 (d, J= 7.8 Hz, 1H), 7.54 (d,
J= 7.8 Hz, 1H), 6.88 (dd, J= 17.7, 10.2 Hz, 1H), 6.14 (d, J= 17.7 Hz, 1H),
5.71 (d, J= 10.2 Hz, 1H), 5.084.94 (m, 1H), 4.56 (d, J= 10.5 Hz, 1H),
4.21 (d, J= 10.9 Hz, 1H), 3.57 (s, 2H), 2.972.84(m,1H),2.732.62 (m,
1H),2.57(s,3H),2.39(brs,8H),2.19(s,3H),2.01(brs,4H).HRMSm/z
(ESI) calcd for C36H39F3N9O2[M + H]+686.3179; found, 686.3181.
1-Ethyl-3-ethynyl-1H-pyrazolo[3,4-d]pyrimidin-4-amine
(11). 1-Ethyl-3-((trimethylsilyl)ethynyl)-1H-pyrazolo[3,4-d]pyrimidin-4-
amine. 3b (5.15 g, 17.8 mmol), CuI (339 mg, 1.78 mmol), and Pd(PPh3)4
(1 g, 0.89 mmol) were suspended in anhydrous DMF (25 mL). The
mixture underwent three cycles of vacuum/lling with N2.Triethylamine
(5 mL, 35.6 mmol) and trimethylsilylacetylene (2.65 mL, 18.7 mmol)
were then added. The mixture was stirred at 80 °C for 5 h and then
quenched with water. EA and more water were added for extraction. The
combined organic layer was dried with sodium sulfate, ltered, con-
centrated, and the crude product was puried using silica gel chroma-
tography with a methanol/dichloromethane gradient to aord the
1-ethyl-3-((trimethylsilyl)ethynyl)-1H-pyrazolo[3,4-d]pyrimidin-4-
amine as a yellow solid.
1-Ethyl-3-((trimethylsilyl)ethynyl)-1H-pyrazolo[3,4-d]pyrimidin-4-
amine was dissolved in methanol. Then K2CO3(4.9 g, 35.6 mmol) was
added, and the mixture was stirred for 10 min at room temperature. The
mixture was ltered, and the ltrate was concentrated in vacuo and
then partitioned between water and DCM. The organic layer was dried
with sodium sulfate and concentrated in vacuo. The crude product
was puried using silica gel chromatography with a methanol/
dichloromethane gradient to aord the title compound 11 as a white
solid (1.49 g, 44.7% yield). 1H NMR (400 MHz, DMSO-d6)δ8.20 (s,
1H), 7.82 (br s, 1H), 6.67 (br s, 1H), 4.30 (q, J= 7.2 Hz, 2H), 1.35 (t, J=
7.2 Hz, 3H). MS m/z(ESI): 188.2 [M + H]+.
3-Iodo-4-isopropylbenzoic Acid (12d). Methyl 4-isopropylben-
zoate (16b). A solution of 4-isopropylbenzoic acid 15b (1 g, 6.1 mmol),
and conc H2SO4(cat., 1 mL) in MeOH (10 mL) was heated at 65 °C for
20 h. The reaction mixture was cooled, and the solvent was evaporated.
The residue was dissolved in Et2O, washed with saturated aqueous
NaHCO3(3×), brine, dried over MgSO4, and the solvent was evaporated
Journal of Medicinal Chemistry Article
DOI: 10.1021/acs.jmedchem.5b00270
J. Med. Chem. XXXX, XXX, XXXXXX
O
to give the 16b (998 mg, 92.0%) as an orange liquid. 1H NMR (400
MHz, DMSO-d6)δ7.89 (d, J= 8.3 Hz, 2H), 7.40 (d, J= 8.3 Hz, 2H),
3.84 (s, 3H), 3.072.86 (m, 1H), 1.22 (d, J= 6.9 Hz, 6H). MS m/z
(ESI): 179.1 [M + H]+.
Methyl 3-Iodo-4-isopropylbenzoate (17b). To 8 mL of acetic
acid and 4 mL of acetic anhydride in a 50 mL round-bottom ask were
added NaIO4(599 mg, 2.8 mmol) and powdered I2(507.62 mg,
2 mmol) at 510 °C with stirring. Then conc H2SO4(1 mL) was slowly
added dropwise while keeping the temperature at 510 °C followed
with methyl 4-isopropylbenzoate (998 mg, 5.6 mmol) at the same
temperature. A brown suspension was stirred for 1 h at room tem-
perature and 4 h at 40 °C (during this time a brown-violet solution
almost decolorized). After cooling, water (30 mL) was added at 0 °C and
product was extracted with CH2Cl2(3×20 mL). Extracts were washed
with 20% NaOH (approximately 3 mL; water layer was strongly alkaline
after extraction), dried over MgSO4, and evaporated to aord a yellow
oil. Crude product was puried using silica gel chromatography with a
petroleum ether gradient to aord the desired product as a light yellow
solid (1.56 g, 91.5% yield). 1H NMR (400 MHz, DMSO-d6)δ8.34 (d,
J= 1.7 Hz, 1H), 7.94 (dd, J= 8.1, 1.7 Hz, 1H), 7.49 (d, J= 8.1 Hz, 1H),
3.85 (s, 3H), 3.243.06 (m, 1H), 1.21 (d, J= 6.8 Hz, 6H). MS m/z
(ESI): 305.0 [M + H]+.
3-Iodo-4-isopropylbenzoic Acid (12d). To a solution of 17b
(1.56 g, 5.1 mmol) in ethanol (20 mL) was added 1 M NaOH (6 mL).
After stirring for 2 h at room temperature a clear pale yellow solution was
obtained. The mixture was concentrated in vacuo and suspended in
water. Then the solution was adjusted to pH 2 by addition of 1 M HCl
and the solvent was stripped o. The residue was washed with water and
taken up three times in toluene (30 mL) and evaporated to aord 12d as
a beige powder (1.33 g, 89.9% yield). 1H NMR (400 MHz, DMSO-d6)δ
13.10 (s, 1H), 8.32 (d, J= 1.6 Hz, 1H), 7.92 (dd, J= 8.1, 1.6 Hz, 1H),
7.46 (d, J= 8.1 Hz, 1H), 3.223.07 (m, 1H), 1.21 (d, J= 6.8 Hz, 6H).
MS m/z(ESI): 289.0 [M H]. Compound 12c was prepared by using
a similar procedure; yield 78.1%.
4-Ethyl-3-iodobenzoic Acid (12c). 1H NMR (400 MHz, DMSO-
d6)δ8.31 (s, 1H), 7.89 (d, J= 7.8 Hz, 1H), 7.44 (d, J= 7.8 Hz, 1H), 2.73
(q, J= 7.2 Hz, 2H), 1.16 (t, J=7.2Hz,3H).MSm/z(ESI): 275.0 [M H].
Compounds 13afwere prepared in a similar manner to that
described for 10a from 4-((4-methylpiperazin-1-yl)methyl)-3-
(triuoromethyl)aniline and dierent benzoic acid; yield 67.985.0%.
3-Iodo-N-(4-((4-methylpiperazin-1-yl)methyl)-3-
(triuoromethyl)phenyl)benzamide (13a). 1H NMR (400 MHz,
DMSO-d6)δ10.57 (s, 1H), 8.32 (s, 1H), 8.18 (s, 1H), 8.04 (d, J= 8.5 Hz,
1H), 7.98 (d, J= 7.8 Hz, 2H), 7.71 (d, J= 8.5 Hz, 1H), 7.36 (t, J= 7.9 Hz,
1H), 3.57 (s, 2H), 2.40 (br s, 8H), 2.17 (s, 3H). MS m/z(ESI): 504.1
[M + H]+.
4-Chloro-3-iodo-N-(4-((4-methylpiperazin-1-yl)methyl)-3-
(triuoromethyl)phenyl)benzamide Hydrochloride Salt (13b).
1H NMR (400 MHz, DMSO-d6)δ10.76 (s, 1H), 8.54 (s, 1H), 8.24 (s,
1H), 8.13 (d, J= 8.3 Hz, 1H), 8.01 (d, J= 8.3 Hz, 1H), 7.89 (br s, 1H),
7.77 (d, J= 8.4 Hz, 1H), 3.90 (br s, 2H), 3.47 (br s, 2H), 3.16 (br s, 4H),
2.78 (s, 3H), 2.71 (br s, 2H). MS m/z(ESI): 538.1 [M + H]+.
4-Ethyl-3-iodo-N-(4-((4-methylpiperazin-1-yl)methyl)-3-
(triuoromethyl)phenyl)benzamide (13c). 1H NMR (400 MHz,
CDCl3), δ8.29(s, 1H), 8.16 (s, 1H), 7.887.85 (m, 2H), 7.78 (d, J= 8.0
Hz, 1H), 7.74 (d, J= 8.4 Hz, 1H), 7.29 (d, J= 8.0 Hz, 1H), 3.62 (s, 2H),
2.77 (q, J= 7.6 Hz, 2H), 2.51 (br s, 8H), 2.41 (s, 3H), 1.22 (t, J= 7.6 Hz,
3H). MS m/z(ESI): 532.1 [M + H]+.
3-Iodo-4-isopropyl-N-(4-((4-methylpiperazin-1-yl)methyl)-3-
(triuoromethyl)phenyl)benzamide (13d). 1H NMR (400 MHz,
CDCl3)δ8.37 (s, 1H), 8.31 (s, 1H), 7.95 (d, J = 10.1 Hz, 2H), 7.90 (d,
J= 7.6 Hz, 1H), 7.57 (d, J= 6.4 Hz, 1H), 7.36 (d, J= 8.1 Hz, 1H), 3.73 (s,
2H), 3.273.23 (m, 1H), 3.04 (br s, 4H), 2.87 (s, 4H), 2.74 (s, 3H), 1.26
(d, J= 6.8 Hz, 6H). MS m/z(ESI): 546.1 [M + H]+.
4-Fluoro-3-iodo-N-(4-((4-methylpiperazin-1-yl)methyl)-3-
(triuoromethyl)phenyl)benzamide (13e). 1H NMR (400 MHz,
DMSO-d6)δ10.56 (s, 1H), 8.46 (d, J= 5.7 Hz, 1H), 8.17 (s, 1H),
8.03 (d, J= 8.3 Hz, 2H), 7.71 (d, J= 8.6 Hz, 1H), 7.45 (t, J= 8.2 Hz, 1H),
3.58 (s, 2H), 2.43 (br s, 8H), 2.24 (s, 3H). MS m/z(ESI): 522.1
[M + H]+.
3-Iodo-4-methoxy-N-(4-((4-methylpiperazin-1-yl)methyl)-3-
(triuoromethyl)phenyl)benzamide Hydrochloride Salt (13f).
1H NMR (400 MHz, DMSO-d6)δ10.43 (s, 1H), 9.62 (br s, 1H), 8.43
(d, J= 1.9 Hz, 1H), 8.19 (s, 1H), 8.147.97 (m, 2H), 7.69 (d, J= 8.5 Hz,
1H), 7.16 (d, J= 8.7 Hz, 1H), 3.92 (s, 3H), 3.65 (s, 2H), 3.15 (br s, 4H),
2.75 (s, 3H), 2.67 (br s, 4H). MS m/z(ESI): 534.1 [M + H]+.
Compounds 14afwere prepared in a similar manner to that
described for 1a from 1-ethyl-3-ethynyl-1H-pyrazolo[3,4-d]pyrimidin-
4-amine 11 and dierent iodobenzamide (13af); yield 37.951.6%.
3-((4-Amino-1-ethyl-1H-pyrazolo[3,4-d]pyrimidin-3-yl)-
ethynyl)-N-(4-((4-methylpiperazin-1-yl)methyl)-3-
(triuoromethyl)phenyl)benzamide (14a). 1H NMR (400 MHz,
DMSO-d6)δ10.65 (s, 1H), 8.36 (s, 1H), 8.27 (s, 1H), 8.23 (s, 1H),
8.138.01 (m, 2H), 7.97 (s, 1H), 7.73 (d, J= 7.6 Hz, 1H),7.65 (s, 1H),
4.37 (q, J= 7.2 Hz, 2H), 3.58 (s, 2H), 2.43 (br s, 8H), 2.23 (s, 3H), 1.42
(t, J= 7.2 Hz, 3H). HRMS m/z(ESI) calcd for C29H30F3N8O[M+H]
+
563.2495; found, 563.2499.
3-((4-Amino-1-ethyl-1H-pyrazolo[3,4-d]pyrimidin-3-yl)-
ethynyl)-4-chloro-N-(4-((4-methylpiperazin-1-yl)methyl)-3-
(triuoromethyl)phenyl)benzamide (14b). 1H NMR (400 MHz,
DMSO-d6)δ10.67 (s, 1H), 8.48 (s, 1H), 8.29 (s, 1H), 8.20 (s, 1H), 8.05
(s, 2H), 7.83 (d, J= 8.2 Hz, 1H), 7.73 (d, J= 8.2 Hz, 1H), 4.39 (d, J= 6.9
Hz, 2H), 3.57 (s, 2H), 2.40 (br s, 8H), 2.19 (s, 3H), 1.42 (t, J= 6.9 Hz,
3H). HRMS m/z(ESI) calcd for C29H29ClF3N8O[M+H]
+597.2105;
found, 597.2107.
3-((4-Amino-1-ethyl-1H-pyrazolo[3,4-d]pyrimidin-3-yl)-
ethynyl)-4-ethyl-N-(4-((4-methylpiperazin-1-yl)methyl)-3-
(triuoromethyl)phenyl)benzamide (14c). 1H NMR (400 MHz,
DMSO-d6)δ10.62 (s, 1H), 8.35 (s, 1H), 8.28 (s, 1H), 8.24 (s, 1H), 8.09
(d, J= 8.3 Hz, 1H), 8.00 (d, J= 8.0 Hz, 1H), 7.72 (d, J= 8.3 Hz, 1H),
7.56 (d, J= 8.0 Hz, 1H), 4.38 (q, J= 7.6 Hz, 2H), 3.60 (s, 2H), 2.95 (q,
J= 7.2 Hz, 2H), 2.51 (br s, 8H), 2.34 (s, 3H), 1.42 (t, J= 7.6 Hz, 3H),
1.29 (t, J= 7.2 Hz, 3H). HRMS m/z(ESI) calcd for C31H34F3N8O
[M + H]+591.2808; found, 591.2805.
3-((4-Amino-1-ethyl-1H-pyrazolo[3,4-d]pyrimidin-3-yl)-
ethynyl)-4-isopropyl-N-(4-((4-methylpiperazin-1-yl)methyl)-3-
(triuoromethyl)phenyl)benzamide (14d). 1H NMR (400 MHz,
DMSO-d6)δ10.58 (s, 1H), 8.33 (s, 1H), 8.28 (s, 1H), 8.23 (s, 1H), 8.08
(d, J= 8.1 Hz, 1H), 8.02 (d, J= 8.1 Hz, 1H), 7.72 (d, J= 8.4 Hz, 1H),
7.62 (d, J= 8.4 Hz, 1H), 4.39 (q, J= 7.2 Hz, 2H), 3.61 (s, 2H), 3.593.50
(m, 1H), 2.39 (s, 8H), 2.33 (s, 3H), 1.42 (t, J= 7.2 Hz, 3H), 1.32 (d, J=
6.9 Hz, 6H). HRMS m/z(ESI) calcd for C32H36F3N8O[M+H]
+
605.2964; found, 605.2970.
3-((4-Amino-1-ethyl-1H-pyrazolo[3,4-d]pyrimidin-3-yl)-
ethynyl)-4-uoro-N-(4-((4-methylpiperazin-1-yl)methyl)-3-
(triuoromethyl)phenyl)benzamide (14e). 1H NMR (400 MHz,
DMSO-d6)δ10.64 (s, 1H), 8.47 (d, J= 8.1 Hz, 1H), 8.28 (s, 1H), 8.20
(s, 1H), 8.158.08 (m, 1H), 8.05 (d, J= 8.1 Hz, 1H), 7.73 (d, J= 8.6 Hz,
1H), 7.59 (t, J= 9.0 Hz, 1H), 4.38 (q, J= 7.2 Hz, 2H), 3.57 (s, 2H), 2.39
(br s, 8H), 2.16 (s, 3H), 1.42 (t, J= 7.2 Hz, 3H). HRMS m/z(ESI) calcd
for C29H29F4N8O[M+H]
+581.2400; found, 581.2405.
3-((4-Amino-1-ethyl-1H-pyrazolo[3,4-d]pyrimidin-3-yl)-
ethynyl)-4-methoxy-N-(4-((4-methylpiperazin-1-yl)methyl)-3-
(triuoromethyl)phenyl)benzamide (14f). 1H NMR (400 MHz,
DMSO-d6)δ10.50 (s, 1H), 8.40 (s, 1H), 8.29 (s, 2H), 8.22 (s, 1H),
8.168.02 (m, 2H), 7.71 (d, J= 7.9 Hz, 1H), 7.34 (d, J= 8.8 Hz, 1H),
6.73 (br s, 1H), 4.36 (d, J= 6.7 Hz, 2H), 4.04 (s, 3H), 3.60 (s, 2H), 2.39
(br s, 8H), 2.34 (s, 3H), 1.42 (t, J= 6.7 Hz, 3H). HRMS m/z(ESI) calcd
for C30H32F3N8O2[M + H]+593.2600; found, 593.2598.
3-((4-Amino-1-ethyl-1H-pyrazolo[3,4-d]pyrimidin-3-yl)-
ethynyl)-4-hydroxy-N-(4-((4-methylpiperazin-1-yl)methyl)-3-
(triuoromethyl)phenyl)benzamide (14g). To a stirred suspension
of 3-((4-amino-1-ethyl-1H-pyrazolo[3,4-d]pyrimidin-3-yl)ethynyl)-4-
methoxy-N-(4-((4-methylpiperazin-1-yl)methyl)-3-(triuoromethyl)-
phenyl)benzamide 14f (296 mg, 0.5 mmol) in dry DCM (10 mL)
at 78 °C was added dropwise BBr3(1.0 M solution in DCM, 6 mL,
6 mmol) under an N2atmosphere. Then the reaction mixture was stirred
at this temperature for 20 min and moved to rt for another 8 h. The
reaction was then quenched by the dropwise addition of water. The
reaction mixture was poured into ammonia solution (30 mL, pH 8), and
more DCM was added for extraction. The combined organic extracts
Journal of Medicinal Chemistry Article
DOI: 10.1021/acs.jmedchem.5b00270
J. Med. Chem. XXXX, XXX, XXXXXX
P
were dried (Na2SO4) and the solvent was removed under reduced
pressure to give a solid which was puried by ash chromatography to
aord the title compound as a yellow solid (173.5 mg, 60.1% yield). 1H
NMR (400 MHz, DMSO-d6)δ10.68 (s, 1H), 8.41 (s, 1H), 8.31 (s, 1H),
8.28 (s, 1H), 8.14 (d, J= 8.6 Hz, 1H), 8.04 (d, J= 8.7 Hz, 1H), 7.98 (d,
J= 8.7 Hz, 1H), 7.73 (d, J= 8.6 Hz, 1H), 7.58 (s, 1H), 4.44 (q, J= 7.2 Hz,
2H), 3.67 (s, 2H), 3.11 (br s, 4H), 2.70 (s, 3H), 2.65 (br s, 4H), 1.46 (t,
J= 7.2 Hz, 3H). HRMS m/z(ESI) calcd for C29H30F3N8O2[M + H]+
579.2444; found, 579.2446.
ASSOCIATED CONTENT
*
SSupporting Information
Chemical structures of top 100 compounds, a full description of
the scaold hopping and molecular docking, and a csv le of
molecular formula strings. This material is available free of charge
via the Internet at http://pubs.acs.org.
AUTHOR INFORMATION
Corresponding Author
*Phone: +86-28-85164063. Fax: +86-28-85164060. E-mail:
yangsy@scu.edu.cn.
Author Contributions
§
C.-H.Z., M.-W.Z., and Y.-P.L. contributed equally to this work.
Notes
The authors declare no competing nancial interest.
ACKNOWLEDGMENTS
This work was supported by the 973 Program (Grant
2013CB967204), the National Natural Science Foundation
(Grant 81325021, 81172987, and 81473140), and the Program
for Changjiang Scholars and Innovative Research Team in
University (Grant IRT13031).
ABBREVIATIONS USED
TNBC, triple-negative breast cancer; ER, estrogen receptor; PR,
progesterone receptor; HER2, human epidermal growth factor
receptor 2; Src, sarcoma; B-RAF, serine/threonine protein kinase
B-RAF; C-RAF, RAF proto-oncogene serine/threonine protein
kinase; FAK, focal adhesion kinase; ERK, extracellular regulated
protein kinases; AKT, protein kinase B; MAPK, mitogen-
activated protein kinase; SFK, Src family kinase; SAR, structure
activity relationship; R&D, research and development; i-Pr,
isopropyl; NIS, 1-iodopyrrolidine-2,5-dione; DIBAL, diisobuty-
laluminum hydride; EA, ethyl acetate
REFERENCES
(1) Foulkes, W. D.; Smith, I. E.; Reis-Filho, J. S. Triple-negative breast
cancer. N. Engl. J. Med. 2010,363 (20), 19381948.
(2) Carey, L.; Winer, E.; Viale, G.; Cameron, D.; Gianni, L. Triple-
negative breast cancer: disease entity or title of convenience? Nat. Rev.
Clin. Oncol. 2010,7(12), 683692.
(3) Cleator, S.; Heller, W.; Coombes, R. C. Triple-negative breast
cancer: therapeutic options. Lancet Oncol. 2007,8(3), 235244.
(4) Higgins, M. J.; Baselga, J. Targeted therapies for breast cancer. J.
Clin. Invest. 2011,121 (10), 37973803.
(5) von Minckwitz, G.; Martin, M. Neoadjuvant treatments for triple-
negative breast cancer (TNBC). Ann. Oncol. 2012,23 (Suppl. 6), vi35
vi39.
(6) Mayer, I. A.; Abramson, V. G.; Lehmann, B. D.; Pietenpol, J. A.
New strategies for triple-negative breast cancerdeciphering the
heterogeneity. Clin. Cancer Res. 2014,20 (4), 782790.
(7) Craig, D. W.; OShaughnessy, J. A.; Kiefer, J. A.; Aldrich, J.; Sinari,
S.; Moses, T. M.; Wong, S.; Dinh, J.; Christoforides, A.; Blum, J. L.;
Aitelli, C. L.; Osborne, C. R.; Izatt, T.; Kurdoglu, A.; Baker, A.; Koeman,
J.; Barbacioru, C.; Sakarya, O.; De La Vega, F. M.; Siddiqui, A.; Hoang,
L.; Billings, P. R.; Salhia, B.; Tolcher, A. W.; Trent, J. M.; Mousses, S.;
Von Hoff, D.; Carpten, J. D. Genome and transcriptome sequencing in
prospective metastatic triple-negative breast cancer uncovers therapeu-
tic vulnerabilities. Mol. Cancer Ther. 2013,12 (1), 104116.
(8) Shah, S. P.; Roth, A.; Goya, R.; Oloumi, A.; Ha, G.; Zhao, Y.;
Turashvili, G.; Ding, J.; Tse, K.; Haffari, G.; Bashashati, A.; Prentice, L.
M.; Khattra, J.; Burleigh, A.; Yap, D.; Bernard, V.; McPherson, A.;
Shumansky, K.; Crisan, A.; Giuliany, R.; Heravi-Moussavi, A.; Rosner, J.;
Lai, D.; Birol, I.; Varhol, R.; Tam, A.; Dhalla, N.; Zeng, T.; Ma, K.; Chan,
S. K.; Griffith, M.; Moradian, A.; Cheng, S. W.; Morin, G. B.; Watson, P.;
Gelmon, K.; Chia, S.; Chin, S. F.; Curtis, C.; Rueda, O. M.; Pharoah, P.
D.; Damaraju, S.; Mackey, J.; Hoon, K.; Harkins, T.; Tadigotla, V.;
Sigaroudinia, M.; Gascard, P.; Tlsty, T.; Costello, J. F.; Meyer, I. M.;
Eaves, C. J.; Wasserman, W. W.; Jones, S.; Huntsman, D.; Hirst, M.;
Caldas, C.; Marra, M. A.; Aparicio, S. The clonal and mutational
evolution spectrum of primary triple-negative breast cancers. Nature
2012,486 (7403), 395399.
(9) Turner, N. C.; Reis-Filho, J. S. Tackling the diversity of triple-
negative breast cancer. Clin. Cancer. Res. 2013,19 (23), 63806388.
(10) Tryfonopoulos, D.; Walsh, S.; Collins, D. M.; Flanagan, L.; Quinn,
C.; Corkery, B.; McDermott, E. W.; Evoy, D.; Pierce, A.; ODonovan,
N.; Crown, J.; Duffy, M. J. Src: a potential target for the treatment of
triple-negative breast cancer. Ann. Oncol. 2011,22 (10), 22342240.
(11) Zhang, S.; Huang, W. C.; Li, P.; Guo, H.; Poh, S. B.; Brady, S. W.;
Xiong, Y.; Tseng, L. M.; Li, S. H.; Ding, Z.; Sahin, A. A.; Esteva, F. J.;
Hortobagyi, G. N.; Yu, D. Combating trastuzumab resistance by
targeting SRC, a common node downstream of multiple resistance
pathways. Nat. Med. 2011,17 (4), 461469.
(12) Kim, L. C.; Song, L.; Haura, E. B. Src kinases as therapeutic targets
for cancer. Nat. Rev. Clin. Oncol. 2009,6(10), 587595.
(13) Wheeler, D. L.; Iida, M.; Dunn, E. F. The role of Src in solid
tumors. Oncologist 2009,14 (7), 667678.
(14) Finn, R. S. Targeting Src in breast cancer. Ann. Oncol. 2008,19
(8), 13791386.
(15) Shields, B. J.; Wiede, F.; Gurzov, E. N.; Wee, K.; Hauser, C.; Zhu,
H. J.; Molloy, T. J.; OToole, S. A.; Daly, R. J.; Sutherland, R. L.; Mitchell,
C. A.; McLean, C. A.; Tiganis, T. TCPTP regulates SFK and STAT3
signaling and is lost in triple-negative breast cancers. Mol. Cell. Biol.
2013,33 (3), 557570.
(16) Hochgrafe, F.; Zhang, L.; OToole, S. A.; Browne, B. C.; Pinese,
M.; Porta Cubas, A.; Lehrbach, G. M.; Croucher, D. R.; Rickwood, D.;
Boulghourjian, A.; Shearer, R.; Nair, R.; Swarbrick, A.; Faratian, D.;
Mullen, P.; Harrison, D. J.; Biankin, A. V.; Sutherland, R. L.; Raftery, M.
J.; Daly, R. J. Tyrosine phosphorylation profiling reveals the signaling
network characteristics of Basal breast cancer cells. Cancer Res. 2010,70
(22), 93919401.
(17) Zhang, S.; Huang, W. C.; Zhang, L.; Zhang, C.; Lowery, F. J.;
Ding, Z.; Guo, H.; Wang, H.; Huang, S.; Sahin, A. A.; Aldape, K. D.;
Steeg, P. S.; Yu, D. SRC family kinases as novel therapeutic targets to
treat breast cancer brain metastases. Cancer Res. 2013,73 (18), 5764
5774.
(18) Park, B. J.; Whichard, Z. L.; Corey, S. J. Dasatinib synergizes with
both cytotoxic and signal transduction inhibitors in heterogeneous
breast cancer cell linesLessons for design of combination targeted
therapy. Cancer Lett. 2012,320 (1), 104110.
(19) Duffy, M. J.; McGowan, P. M.; Crown, J. Targeted therapy for
triple-negative breast cancer: Where are we? Int. J. Cancer 2012,131
(11), 24712477.
(20) Finn, R. S.; Dering, J.; Ginther, C.; Wilson, C. A.; Glaspy, P.;
Tchekmedyian, N.; Slamon, D. J. Dasatinib, an orally active small
molecule inhibitor of both the src and abl kinases, selectively inhibits
growth of basal-type/triple-negativebreast cancer cell lines growing in
vitro. Breast Cancer Res. Treat. 2007,105 (3), 319326.
(21) El Touny, L. H.; Vieira, A.; Mendoza, A.; Khanna, C.; Hoenerhoff,
M. J.; Green, J. E. Combined SFK/MEK inhibition prevents metastatic
outgrowth of dormant tumor cells. J. Clin. Invest. 2014,124 (1), 156
168.
Journal of Medicinal Chemistry Article
DOI: 10.1021/acs.jmedchem.5b00270
J. Med. Chem. XXXX, XXX, XXXXXX
Q
(22) Bianchi-Smiraglia, A.; Paesante, S.; Bakin, A. V. Integrin beta5
contributes to the tumorigenic potential of breast cancer cells through
the Src-FAK and MEK-ERK signaling pathways. Oncogene 2013,32
(25), 30493058.
(23) Boschelli, D. H.; Ye, F.; Wang, Y. D.; Dutia, M.; Johnson, S. L.;
Wu, B.; Miller, K.; Powell, D. W.; Yaczko, D.; Young, M.; Tischler, M.;
Arndt, K.; Discafani, C.; Etienne, C.; Gibbons, J.; Grod, J.; Lucas, J.;
Weber, J. M.; Boschelli, F. Optimization of 4-phenylamino-3-
quinolinecarbonitriles as potent inhibitors of Src kinase activity. J.
Med. Chem. 2001,44 (23), 39653977.
(24) Hennequin, L. F.; Allen, J.; Breed, J.; Curwen, J.; Fennell, M.;
Green, T. P.; Lambert-van der Brempt, C.; Morgentin, R.; Norman, R.
A.; Olivier, A.; Otterbein, L.; Plé
, P. A.; Warin, N.; Costello, G. N-(5-
Chloro-1,3-benzodioxol-4-yl)-7-[2-(4-methylpiperazin-1-yl)ethoxy]-5-
(tetrahydro-2H-pyran-4-yloxy)quinazolin-4-amine, a novel, highly
selective, orally available, dual-specific c-Src/Abl kinase inhibitor. J.
Med. Chem. 2006,49 (22), 64656488.
(25) Ma, J. G.; Huang, H.; Chen, S. M.; Chen, Y.; Xin, X. L.; Lin, L. P.;
Ding, J.; Liu, H.; Meng, L. H. PH006, a novel and selective Src kinase
inhibitor, suppresses human breast cancer growth and metastasis in vitro
and in vivo. Breast Cancer Res. Treat. 2011,130 (1), 8596.
(26) Kwarcinski, F. E.; Fox, C. C.; Steffey, M. E.; Soellner, M. B.
Irreversible Inhibitors of c-Src kinase that target a nonconserved
cysteine. ACS Chem. Biol. 2012,7(11), 19101917.
(27) Gushwa, N. N.; Kang, S.; Chen, J.; Taunton, J. Selective targeting
of distinct active site nucleophiles by irreversible Src-family kinase
inhibitors. J. Am. Chem. Soc. 2012,134 (50), 2021420217.
(28) Anbalagan, M.; Carrier, L.; Glodowski, S.; Hangauer, D.; Shan, B.;
Rowan, B. KX-01, a novel Src kinase inhibitor directed toward the
peptide substrate site, synergizes with tamoxifen in estrogen receptor α
positive breast cancer. Breast Cancer Res. Treat. 2012,132 (2), 391409.
(29) Das, J.; Chen, P.; Norris, D.; Padmanabha, R.; Lin, J.; Moquin, R.
V.; Shen, Z.; Cook, L. S.; Doweyko, A. M.; Pitt, S.; Pang, S.; Shen, D. R.;
Fang, Q.; de Fex, H. F.; McIntyre, K. W.; Shuster, D. J.; Gillooly, K. M.;
Behnia, K.; Schieven, G. L.; Wityak, J.; Barrish, J. C. 2-Aminothiazole as a
novel kinase inhibitor template. Structureactivity relationship studies
toward the discovery of N-(2-chloro-6-methylphenyl)-2-[[6-[4-(2-
hydroxyethyl)-1-piperazinyl)]-2-methyl-4-pyrimidinyl]amino)]-1,3-
thiazole-5-carboxamide (dasatinib, BMS-354825) as a potent pan-Src
kinase inhibitor. J. Med. Chem. 2006,49 (23), 68196832.
(30) Finn, R. S.; Bengala, C.; Ibrahim, N.; Roche, H.; Sparano, J.;
Strauss, L. C.; Fairchild, J.; Sy, O.; Goldstein, L. J. Dasatinib as a single
agent in triple-negative breast cancer: results of an open-label phase 2
study. Clin. Cancer. Res. 2011,17 (21), 69056913.
(31) Huang, W. S.; Metcalf, C. A.; Sundaramoorthi, R.; Wang, Y.; Zou,
D.; Thomas, R. M.; Zhu, X.; Cai, L.; Wen, D.; Liu, S.; Romero, J.; Qi, J.;
Chen, I.; Banda, G.; Lentini, S. P.; Das, S.; Xu, Q.; Keats, J.; Wang, F.;
Wardwell, S.; Ning, Y.; Snodgrass, J. T.; Broudy, M. I.; Russian, K.; Zhou,
T.; Commodore, L.; Narasimhan, N. I.; Mohemmad, Q. K.; Iuliucci, J.;
Rivera, V. M.; Dalgarno, D. C.; Sawyer, T. K.; Clackson, T.; Shakespeare,
W. C. Discovery of 3-[2-(imidazo[1,2-b]pyridazin-3-yl)ethynyl]-4-
methyl-N-{4-[(4-methylpiperazin-1-yl)methyl]-3-(trifluoromethyl)-
phenyl}benzamide (AP24534), a potent, orally active pan-inhibitor of
breakpoint cluster region-Abelson (BCR-ABL) kinase including the
T315I gatekeeper mutant. J. Med. Chem. 2010,53 (12), 47014719.
(32) OHare, T.; Shakespeare, W. C.; Zhu, X.; Eide, C. A.; Rivera, V.
M.; Wang, F.; Adrian, L. T.; Zhou, T.; Huang, W. S.; Xu, Q.; Metcalf, C.
A., 3rd; Tyner, J. W.; Loriaux, M. M.; Corbin, A. S.; Wardwell, S.; Ning,
Y.; Keats, J. A.; Wang, Y.; Sundaramoorthi, R.; Thomas, M.; Zhou, D.;
Snodgrass, J.; Commodore, L.; Sawyer, T. K.; Dalgarno, D. C.;
Deininger, M. W.; Druker, B. J.; Clackson, T. AP24534, a pan-BCR-ABL
inhibitor for chronic myeloid leukemia, potently inhibits the T315I
mutant and overcomes mutation-based resistance. Cancer Cell 2009,16
(5), 401412.
(33) Liu, Y.; Gray, N. S. Rational design of inhibitors that bind to
inactive kinase conformations. Nat. Chem. Biol. 2006,2(7), 358364.
(34) GOLD, version 5.0; Cambridge Crystallographic Data Centre:
Cambridge, U.K.
(35) Ren, X.; Pan, X.; Zhang, Z.; Wang, D.; Lu, X.; Li, Y.; Wen, D.;
Long, H.; Luo, J.; Feng, Y.; Zhuang, X.; Zhang, F.; Liu, J.; Leng, F.; Lang,
X.; Bai, Y.; She, M.; Tu, Z.; Pan, J.; Ding, K. Identification of GZD824 as
an orally bioavailable inhibitor that targets phosphorylated and
nonphosphorylated breakpoint cluster region-Abelson (Bcr-Abl) kinase
and overcomes clinically acquired mutation-induced resistance against
imatinib. J. Med. Chem. 2013,56 (3), 879894.
(36) Balko, J. M.; Cook, R. S.; Vaught, D. B.; Kuba, M. G.; Miller, T.
W.; Bhola, N. E.; Sanders, M. E.; Granja-Ingram, N. M.; Smith, J. J.;
Meszoely, I. M.; Salter, J.; Dowsett, M.; Stemke-Hale, K.; Gonzalez-
Angulo, A. M.; Mills, G. B.; Pinto, J. A.; Gomez, H. L.; Arteaga, C. L.
Profiling of residual breast cancers after neoadjuvant chemotherapy
identifies DUSP4 deficiency as a mechanism of drug resistance. Nat.
Med. 2012,18 (7), 10521059.
(37) Giltnane, J. M.; Balko, J. M. Rationale for targeting the Ras/
MAPK pathway in triple-negative breast cancer. Discovery Med. 2014,17
(95), 275283.
(38) Pan, Y.; Xu, Y.; Feng, S.; Luo, S.; Zheng, R.; Yang, J.; Wang, L.;
Zhong, L.; Yang, H. Y.; Wang, B. L.; Yu, Y.; Liu, J.; Cao, Z.; Wang, X.; Ji,
P.; Wang, Z.; Chen, X.; Zhang, S.; Wei, Y. Q.; Yang, S. Y. SKLB1206, a
novel orally available multikinase inhibitor targeting EGFR activating
and T790M mutants, ErbB2, ErbB4, and VEGFR2, displays potent
antitumor activity both in vitro and in vivo. Mol. Cancer Ther. 2012,11
(4), 952962.
Journal of Medicinal Chemistry Article
DOI: 10.1021/acs.jmedchem.5b00270
J. Med. Chem. XXXX, XXX, XXXXXX
R
... In an MDA-MB-231 xenograft mouse model, the administration of 1j at a daily dose of 30 mg/kg for 18 days, completely decreased tumor size (IC50 = 0.0009 μM). The tumor inhibition rate was over 100%, with little or no toxicity noticed [110]. ...
Article
Full-text available
Triple-negative breast cancer (TNBC) cells are devoid of estrogen receptors (ERs), progesterone receptor (PRs), and human epidermal growth factor receptor 2 (HER2), and it (TNBC) counts for about 10–15% of all breast cancers. TNBC is highly invasive, having a faster growth rate and a higher risk of metastasis and recurrence. Still, chemotherapy is one of the widely used options for treating TNBC. This study reviewed the histological and molecular characterization of TNBC subtypes, signaling pathways that are aberrantly expressed, and small molecules targeting these pathways, as either single agents or in combination with other therapeutic agents like chemotherapeutics, immunotherapeutics, and antibody–drug conjugates; their mechanisms of action, challenges, and future perspectives were also reviewed. A detailed analytical review was carried out using the literature collected from the SciFinder, PubMed, ScienceDirect, Google Scholar, ACS, Springer, and Wiley databases. Several small molecule inhibitors were found to be therapeutics for treating TNBC. The mechanism of action and the different signaling pathways through which the small molecules exert their effects were studied, including clinical trials, if reported. These small molecule inhibitors include buparlisib, everolimus, vandetanib, apatinib, olaparib, salidroside, etc. Some of the signaling pathways involved in TNBC, including the VEGF, PARP, STAT3, MAPK, EGFR, P13K, and SRC pathways, were discussed. Due to the absence of these biomarkers, drug development for treating TNBC is challenging, with chemotherapy being the main therapeutic agent. However, chemotherapy is associated with chemoresistance and a high toxicity to healthy cells as side effects. Hence, there is a continuous demand for small-molecule inhibitors that specifically target several signaling pathways that are abnormally expressed in TNBC. We attempted to include all the recent developments in this field. Any omission is truly unintentional.
... Compound IV was reported by Cheng et al. [32] as a dual B-RAF V600E /EGFR inhibitor with IC50 values of 51 and 8 nM, respectively. At the same time, the pyrazolopyrimidine V elicited significant inhibitory potency against Src, B-RAF WT/V600E and C-RAF with IC50 values of 0.9, 15, 92, and 27 nM, respectively [33]. The diarylamides quinoline ether VI was reported to possess moderate selective potency toward C-RAF (77% inhibition at 10 µM) [34]. ...
Article
Full-text available
The emergence of cancer resistance to targeted therapy represents a significant challenge in cancer treatment. Therefore, identifying new anticancer candidates, particularly those addressing oncogenic mutants, is an urgent medical demand. A campaign of structural modifications has been conducted to further optimize our previously reported 2-anilinoquinoline-diarylamides conjugate VII as a B-RAFV600E/C-RAF inhibitor. Considering the incorporation of a methylene bridge between the terminal phenyl and cyclic diamine, focused quinoline-based arylamides have been tailored, synthesized, and biologically evaluated. Among them, the 5/6-hydroxyquinolines 17b and 18a stood out as the most potent members, with IC50 values of 0.128 µM, 0.114 µM against B-RAFV600E, and 0.0653 µM, 0.0676 µM against C-RAF. Most importantly, 17b elicited remarkable inhibitory potency against the clinically resistant B-RAFV600K mutant with an IC50 value of 0.0616 µM. The putative binding mode of 17b and 18a were studied by molecular docking and molecular dynamics (MD). Moreover, the antiproliferative activity of all target compounds has been examined over a panel of NCI-60 human cancer cell lines. In agreement with cell-free assays, the designed compounds exerted superior anticancer impact over the lead quinoline VII against all cell lines at a 10 µM dose. Notably, both 17b and 18b showed highly potent antiproliferative activity against melanoma cell lines with growth percent under −90% (SK-MEL-29, SK-MEL-5, and UACC-62) at a single dose, while 17b maintained potency with GI50 values of 1.60–1.89 µM against melanoma cell lines. Taken together, 17b, a promising B-RAFV600E/V600K and C-RAF kinase inhibitor, may serve as a valuable candidate in the arsenal of anticancer chemotherapeutics.
... SRC kinase inhibitor Dasatinib alone, particularly combining with cisplatin has been shown great potential in treating metastatic TNBC [23,25,26]. Moreover, emerging preclinical evidence has demonstrated that TNBC cells showed higher sensitivity to the c-SRC inhibitor than do other cancer subgroups by targeting BCSCs [27][28][29][30], further highlighting the potential significance of targeting BCSCs through inhibiting SRC activity in TNBC treatment. However, the role of SRC kinase in cancer stemness regulation and the underlying regulatory mechanisms are still obscured. ...
Article
Full-text available
SRC is the first identified oncogene, and its aberrant activation has been implicated as a driving event in tumor initiation and progression. However, its role in cancer stemness regulation and the underlying regulatory mechanism are still elusive. Here, we identified a YAP1 tyrosine phosphorylation-dependent YAP1-KLF5 oncogenic module, as the key downstream mediator of SRC kinase regulating cancer stemness and metastasis in triple-negative breast cancer (TNBC). SRC was overexpressed in TNBC patient tissues and its expression level was highly correlated with the tumor malignancy. SRC activation induced, while inhibition of SRC kinase reduced the cancer stemness, tumor cell growth and metastasis in vitro and in vivo. Transcriptomic and proteomic analysis revealed that SRC-mediated YAP1 tyrosine phosphorylation induced its interaction with Kruppel-like factor 5 (KLF5) to form a YAP1/TEAD-KLF5 complex in TNBC cells. YAP1-KLF5 association further promoted TEAD-mediated transcriptional program independently of canonical Hippo kinases, which eventually gave rise to the enhanced cancer stemness and metastasis. Disruption of YAP1-KLF5 module in TNBC cells dramatically attenuated the SRC-induced cancer stemness and metastasis in vitro and in vivo. Accordingly, co-upregulations of SRC and YAP1-KLF5 module in TNBC tissues were significantly positively correlated with the tumor malignance. Altogether, our work presents a novel tyrosine phosphorylation-dependent YAP1-KLF5 oncogenic module governing SRC-induced cancer stemness and metastasis in TNBC. Therefore, targeting YAP1/KLF5-mediated transcription may provide a promising strategy for TNBC treatment with SRC aberrantly activation.
... SRC kinase inhibitor Dasatinib alone, particularly combining with cisplatin has been shown great potential in treating metastatic TNBC 23,25,26 . Moreover, emerging preclinical evidences have demonstrated that TNBC cells showed higher sensitivity to the c-SRC inhibitor than do other cancer subgroups by targeting BCSCs [27][28][29][30] , further highlighting the potential significance of targeting BCSCs through inhibiting SRC activity in TNBC treatment. However, the role of SRC kinase in cancer stemness regulation and the underlying regulatory mechanisms are still obscured. ...
Preprint
Full-text available
SRC was the first identified oncogene, and its aberrant activation has been implicated as a driving event in tumor initiation and progression. However, its role in cancer stemness regulation and the underlying regulatory mechanism are still elusive. Here, we identified a YAP1 tyrosine phosphorylation-dependent YAP1-KLF5 oncogenic module, as the key downstream mediator of SRC kinase regulating cancer stemness and metastasis in triple negative breast cancer (TNBC). SRC was overexpressed in TNBC patient tissues and its expression level was highly correlated with the tumor malignancy. SRC activation induced, while inhibition of SRC kinase reduced the cancer stemness, tumor cell growth and metastasis in vitro and in vivo. Transcriptomic and proteomic analysis revealed that SRC-mediated YAP1 tyrosine phosphorylation induced its interaction with Kruppel-like factor 5 (KLF5) to form a YAP1/TEAD-KLF5 complex in TNBC cells. YAP1-KLF5 association further promoted TEAD-mediated transcriptional program independently of canonical Hippo kinases (MST-LATS), which eventually gave rise to the enhanced cancer stemness and metastasis. Disruption of YAP1-KLF5 module in TNBC cells dramatically attenuated the SRC-induced cancer stemness and metastasis in vitro and in vivo. Accordingly, co-upregulations of SRC and YAP1-KLF5 module in TNBC tissues were significantly positively correlated with the tumor malignance. Altogether, our work presents a novel tyrosine phosphorylation-dependent YAP1-KLF5 oncogenic module governing SRC-induced cancer stemness and metastasis in TNBC. Therefore, targeting of YAP1/KLF5-mediated transcription represents a promising strategy for TNBC treatment with SRC aberrantly activation.
Article
The physiological Src proto-oncogene is a protein tyrosine kinase receptor that served as the essential signalling pathway in different types of cancer. The etiology of cancer involved various signalling pathways and Src signalling pathways are also involved in those clusters. Although the dysregulation of Src kinase resulted in cancer being discovered in the late 19th century it is still considered a cult pathway because it is not much explored by different medicinal chemists and oncologists. In this review, we have elaborated about the structure and activation of Src kinase, as well as the development of Src kinase inhibitors. Our group also provided a comprehensive overview of Src inhibitors throughout the last two decades, including their biological activity, structure-activity relationship, and Src kinase selectivity. The Src binding pocket has been investigated in detail to better comprehend the interaction of Src inhibitors with amino acid residues. We have strengthened the literature with our contribution in terms of molecular docking and ADMET studies of top compounds. We hope that the current analysis will be a useful resource for researchers and provide glimpse of direction toward the design and development of more specific, selective, and potent Src kinase inhibitors.
Article
The pyrimidine and fused pyrimidine ring systems play vital roles to inhibit the c-Src kinase. The Src kinase is made of different domains but the kinase domain is responsible for inhibition of Src kinase. In which the kinase domain is the main domain that is made of several amino acids. The Src kinase is inhibited by its inhibitors when it is activated by phosphorylation. Although dysregulation of Src kinase caused cancer in the late nineteenth century, medicinal chemists have not explored it extensively; therefore it is still regarded as a cult pathway. There are numerous FDA-approved drugs on the market, yet novel anticancer drugs are still in demand. Existing medications have adverse effects and drug resistance owing to rapid protein mutation. In this review, we discussed the activation process of Src kinase, chemistry of pyrimidine ring and its different synthetic routes, as well as the recent development in c-Src kinase inhibitors containing pyrimidine and their biological activity, SAR, and selectivity. The c-Src binding pocket has been predicted in detail to discover the vital amino acids which will interact with inhibitors. The potent derivatives were docked to discover the binding pattern. The derivative 2 established three hydrogen bonds with the amino acid residues Thr341 and Gln278 and had the greatest binding energy of -13.0 kcal/mol. The top docked molecules were further studied for ADMET studies. The derivative 1, 2, and 43 did not show any violation of Lipinski's rule. All derivatives used for the prediction of toxicity showed toxicity.
Article
The 4-Aminopyrazolopyrimidine scaffold has been an interesting pharmacophore since the disclosure of the intimate connection between small-molecule inhibitors and the treatment of diseases. Modification of the 4-aminopyrazolopyrimidine scaffold according to different targets, especially tyrosine kinase and serine/threonine kinase, has resulted in a variety of small-molecule inhibitors. Kinase inhibitors with 4-aminopyrazolopyrimidine derivatives as scaffolds have been widely applied in the treatment of diseases. In this article, we summarized the reports on 4-aminopyrazolopyrimidine as well as its deformation and the application of its derivatives in designing small-molecule inhibitors and the treatment of diseases.
Article
Sorafenib is a clinically useful multiple kinase inhibitor for the treatment of kidney cancer, liver cancer and acute myelocytic leukemia, while it has shown weak efficacy in suppressing breast cancer. Since sirtuin2 (SIRT2) is an important epigenetic regulator and associated with several cancer types including breast cancer, development and evaluation of new SIRT2 inhibitors to probe their therapeutic potentials is currently desirable. A highly selective SIRT2 inhibitor named I was previously developed by us, which showed activity to inhibit non-small cell lung cancer cell lines in vitro. We herein report expanded screening of I and its structurally similar inactive compound II against other cancer cell lines, and found that I had a wide spectrum of anticancer activity while II had no such effects. The I-sorafenib combination treatment exerted obvious synergistic reduction on cell viability of MCF-7 cells. We observed that the combination treatment could suppress cell proliferation, survival and migration, arrest cell cycle at G0/G1 phase, and induce apoptosis in MCF-7 cells, when compared with the single treatment. In vivo studies revealed that the combination treatment showed stronger tumor growth inhibition (87%), comparing with I-(42.8%) or sorafenib-solely-treated groups (61.1%) in MCF-7 xenograft model. In conclusion, this work clearly revealed a potential synthetic lethality effect for I combined with sorafenib, and will probably offer a new strategy at least for breast cancer treatment.
Article
Full-text available
Breast cancer (BC) can recur as metastatic disease many years after primary tumor removal, suggesting that disseminated tumor cells survive for extended periods in a dormant state that is refractory to conventional therapies. We have previously shown that altering the tumor microenvironment through fibrosis with collagen and fibronectin deposition can trigger tumor cells to switch from a dormant to a proliferative state. Here, we used an in vivo preclinical model and a 3D in vitro model of dormancy to evaluate the role of Src family kinase (SFK) in regulating this dormant-to-proliferative switch. We found that pharmacological inhibition of SFK signaling or Src knockdown results in the nuclear localization of cyclin-dependent kinase inhibitor p27 and prevents the proliferative outbreak of dormant BC cells and metastatic lesion formation; however, SFK inhibition did not kill dormant cells. Dormant cell proliferation also required ERK1/2 activation. Combination treatment of cells undergoing the dormant-to-proliferative switch with the Src inhibitor (AZD0530) and MEK1/2 inhibitor (AZD6244) induced apoptosis in a large fraction of the dormant cells and delayed metastatic outgrowth, neither of which was observed with either inhibitor alone. Thus, targeting Src prevents the proliferative response of dormant cells to external stimuli, but requires MEK1/2 inhibition to suppress their survival. These data indicate that treatments targeting Src in combination with MEK1/2 may prevent BC recurrence.
Article
Full-text available
Despite better control of early stage disease and improved overall survival of patients with breast cancer, the incidence of life-threatening brain metastases continues to increase in some of these patients. Unfortunately, other than palliative treatments there is no effective therapy for this condition. In this study, we reveal a critical role for Src activation in promoting brain metastasis in a preclinical model of breast cancer, and we show how a Src-targeting combinatorial regimen can treat HER2+ brain metastases in this model. We found that Src was hyperactivated in brain-seeking breast cancer cells derived from human cell lines or from patients' brain metastases. Mechanistically, Src activation promoted tumor cell extravasation into the brain parenchyma via permeabilization of the blood-brain barrier. When combined with the EGFR/HER2 dual-targeting drug lapatinib, a Src-targeting combinatorial regimen prevented outgrowth of disseminated breast cancer cells through the induction of cell cycle arrest. More importantly, this combinatorial regimen inhibited the outgrowth of established experimental brain metastases, prolonging the survival of metastases-bearing mice. Our results provide a rationale for clinical evaluation of Src-targeting regimens to treat breast cancer patients suffering from brain metastasis.
Article
Full-text available
Triple negative breast cancer (TNBC) is characterized by the absence of expression of estrogen receptor, progesterone receptor, and HER-2. Thirty percent of patients recur after first line treatment, and metastatic TNBC (mTNBC) has a poor prognosis with median survival of one year. Here, we present initial analyses of whole genome and transcriptome sequencing data from 14 prospective metastatic mTNBC. We have cataloged the collection of somatic genomic alterations in these advanced tumors; particularly those that might inform targeted therapies. Genes mutated in multiple tumors included TP53, LRP1B, HERC1, CDH5, RB1, and NF1. Notable genes involved in focal structural events were CTNNA1, PTEN, FBXW7, BRCA2, WT1, FGFR1, KRAS, HRAS, ARAF, BRAF, and PGCP. Homozygous deletion of CTNNA1 was detected in 2 of 6 African Americans. RNA sequencing revealed consistent overexpression of the FOXM1 gene, when tumor gene expression was compared to nonmalignant breast samples. Using an outlier analysis of gene expression comparing one cancer to all the others, we detected expression patterns unique to each patient's tumor. Integrative DNA/RNA analysis provided evidence for deregulation of mutated genes, including the monoallelic expression of TP53 mutations. Finally, molecular alterations in several cancers supported targeted therapeutic intervention on clinical trials with known inhibitors, particularly for alterations in the RAS/RAF/MEK/ERK and PI3K/AKT/MTOR pathways. In conclusion, whole genome and transcriptome profiling of mTNBC have provided insights into somatic events occurring in this difficult to treat cancer. These genomic data have guided patients to investigational treatment trials and provide hypotheses for future trials in this irremediable cancer.
Article
Full-text available
Tyrosine phosphorylation-dependent signaling, as mediated by members of the epidermal growth factor receptor (EGFR) family (ErbB1 to -4) of protein tyrosine kinases (PTKs), Src family PTKs (SFKs), and cytokines such as interleukin-6 (IL-6) that signal via signal transducer and activator of transcription 3 (STAT3), is critical to the development and progression of many human breast cancers. EGFR, SFKs, and STAT3 can serve as substrates for the protein tyrosine phosphatase TCPTP (PTPN2). Here we report that TCPTP protein levels are decreased in a subset of breast cancer cell lines in vitro and that TCPTP protein is absent in a large proportion of “triple-negative” primary human breast cancers. Homozygous TCPTP deficiency in murine mammary fat pads in vivo is associated with elevated SFK and STAT3 signaling, whereas TCPTP deficiency in human breast cancer cell lines enhances SFK and STAT3 signaling. On the other hand, TCPTP reconstitution in human breast cancer cell lines severely impaired cell proliferation and suppressed anchorage-independent growth in vitro and xenograft growth in vivo. These studies establish TCPTP's potential to serve as a tumor suppressor in human breast cancer.
Article
"Triple negative" breast cancer (TNBC) is the most aggressive and least common clinical subtype of breast cancer. As its nomenclature implies, TNBC lacks specific biomarker expression marking response to an effective targeted therapy. The incidence of TNBC is higher in young minority women who suffer from high rates of early recurrence and death from their disease. Mounting preclinical evidence supports targeting the Ras/MAPK cell signaling pathway in the TNBC subtype, despite large genomic surveys such as The Cancer Genome Atlas demonstrating infrequent canonical mutations in this pathway. Due to the early spread of TNBC, targeted treatment in the neoadjuvant setting may offer the effective therapeutic punch needed to eliminate micro-metastatic disease and reduce mortality. Herein, we will review the evidence supporting clinical trials of targeted inhibitors of the Ras/MAPK pathway in TNBC, and discuss the obstacles and opportunities of this approach.
Article
Triple-negative breast cancer (TNBC) is a heterogeneous disease; gene expression analyses recently identified six distinct TNBC subtypes, each displaying a unique biology. Exploring novel approaches to treatment of these subtypes is critical because less than 30% of women with metastatic breast cancer survive five years and virtually all women with metastatic TNBC will ultimately die of their disease despite systemic therapy. To date, not a single targeted therapy has been approved for the treatment of TNBC and cytotoxic chemotherapy remains the standard treatment. We discuss the current and upcoming therapeutic strategies being explored in an attempt to "target" TNBC. Clin Cancer Res; 20(4); 782-90. ©2014 AACR.
Article
Triple-negative breast cancer (TNBC) comprises a highly diverse collection of cancers. Here, we review this diversity both in terms of gene expression subtypes and the repertoire of genetic events. Transcriptomic analyses of TNBC have revealed at least six subtypes, with the luminal androgen receptor (luminal AR) or molecular apocrine cancers forming a distinct group within triple-negative disease. Distinct from the gene expression subtypes, a diverse set of genetic events have been described in TNBC, with a number of potentially targetable genetic events found although all at relatively low frequency. Clinical trials to define the clinical utility of therapies targeting these low-frequency events will require substantial screening efforts to identify sufficient patients. Set against the diversity of TNBC, clinical studies of patients with triple-negative disease will need to be either focused on molecularly defined subsets with upfront molecular stratification, or powered for a secondary endpoint analysis of a molecularly defined subset. Such approaches will be crucial to realize the potential of precision medicine for patients with TNBCs. Clin Cancer Res; 19(23); 6380-8. ©2013 AACR.
Article
Various imatinib resistance-related mutations have been identified in different functional domains of Bcr-Abl kinase, among which the gatekeeper T315I point mutation poses a major challenge in the clinical management of chronic myelogenous leukemia (CML). Novel inhibitors are urgently needed to overcome drug resistance related to Bcr-Abl T315I and other mutations. Herein, we report GZD824 (10a) as a novel orally bioavailable inhibitor that targets a broad spectrum of Bcr-Abl mutants, including the gatekeeper T315I and p-loop mutations, which are associated with poor prognosis in CML. Compound 10a tightly bound phosphorylated and non-phosphorylated Bcr-Abl and inhibited their kinase activity with IC50 values in the low nM range. Compound 10a also strongly suppressed the proliferation of Bcr-Abl-positive CML cells and murine Ba/F3 cells ectopically expressing Bcr-Abl WT or resistance-related mutants with low-nM IC50 values. Compound 10a displayed good oral bioavailability (48.7%), a reasonable half-life (10.6 hrs) and promising in vivo anti-tumor efficacy. This compound induced the regression of tumors driven by wild-type Bcr-Abl or resistance-related mutants, such as T315I, G250E, Q252H, E255K and F317L, in mouse xenograft tumor models. The survival of mice bearing an allograft leukemia model that utilizes Ba/F3 cells harboring Bcr-Abl T315I was significantly improved by administration of 10a. Thus, compound 10a represents a promising lead candidate for the further development of Bcr-Abl inhibitors to overcome clinically acquired resistance against imatinib.
Article
Src-family tyrosine kinases play pivotal roles in human physiology and disease, and several drugs that target members of this family are in clinical use. None of these drugs appear to discriminate among closely related kinases. However, assessing their selectivity toward endogenous kinases in living cells remains a significant challenge. Here, we report the design of two Src-directed chemical probes, each consisting of a nucleoside scaffold with a 5'-electrophile. A 5'-fluorosulfonylbenzoate (1) reacts with the conserved catalytic lysine (Lys295) and shows little discrimination among related kinases. By contrast, a 5'-vinylsulfonate (2) reacts with a poorly conserved, proximal cysteine (Cys277) found in three Src-family and six unrelated kinases. Both 1 and 2 bear an alkyne tag and efficiently label their respective endogenous kinase targets in intact cells. Using 1 as a competitive probe, we determined the extent to which ponatinib, a clinical Bcr-Abl inhibitor, targets Src-family kinases. Remarkably, while ponatinib had little effect on en-dogenous Fyn or Src, it potently blocked the critical T-cell kinase, Lck. Probes 1 and 2 thus enable competitive profiling vs. distinct kinase subsets in living cells.