ArticlePDF AvailableLiterature Review

Recent Progress in the Development of Quinoline Derivatives for the Exploitation of Anti-Cancer Agents

Authors:

Abstract

Background Along with the progress in medicine and therapies, the exploitation of anti-cancer agents focused more on the vital signaling pathways and key biological macromolecules. With rational design and advanced synthesis, quinoline derivatives have been utilized frequently in medicinal chemistry, especially in developing anti-cancer drugs or candidates. Methods Using DOI searching, articles published before 2020 all over the world have been reviewed as comprehensively as possible. Results In this review, we selected the representative quinoline derivate drugs in market or clinical trials, classified them into five major categories with detailed targets according to their main mechanisms, discussed the relationship within the same mechanism, and generated a summative discussion with prospective expectations. For each mechanism, the introduction of the target was presented, with the typical examples of quinoline derivate drugs. Conclusion This review has highlighted the quinoline drugs or candidates, suited them into corresponding targets in their pathways, summarized and discussed. We hope that this review may help the researchers who are interested in discovering quinoline derivate anticancer agents obtain considerable understanding in this specific topic. Through the flourishing period and the vigorous strategies in clinical trials, quinoline drugs would be potential but facing new challenges in future.
Anti-Cancer Agents in Medicinal Chemistry
The journal for in-depth reviews and high quality research
papers on Anti-Cancer Agents
Impact
Factor:
2.049
ISSN: 1871-5206
eISSN: 1875-5992
Send Orders for Reprints to reprints@benthamscience.net
Anti-Cancer Agents in Medicinal Chemistry, 2021, 21, 825-838
825
REVIEW ARTICLE
Recent Progress in the Development of Quinoline Derivatives for the Exploitation of
Anti-Cancer Agents
Ruo-Jun Man1,‡, Nasreen Jeelani2,‡, Chongchen Zhou3,* and Yu-Shun Yang2,*
1College of Chemistry and Chemical Engineering, Guangxi University for Nationalities, Nanning, China; 2Institute of Chemistry and
BioMedical Sciences, School of Life Sciences, Nanjing University, Nanjing 210023, China; 3Henan Provincial Key Laboratory of
Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou University,
Zhengzhou, 450018, China
A R T I C L E H I S T O R Y
Received: December 31 , 2019
Revised: January 23, 2020
Accepted: February 10 , 2020
DOI:
10.2174/1871520620666200516150345
Abstract: Background: Along with the progress in medicine and therapies, the exploitation of anti-cancer agents
focused more on the vital signaling pathways and key biological macromolecules. With rational design and ad-
vanced synthesis, quinoline derivatives have been utilized frequently in medicinal chemistry, especially in develop-
ing anti-cancer drugs or candidates.
Methods: Using DOI searching, articles published before 2020 all over the world have been reviewed as compre-
hensively as possible.
Results: In this review, we selected the representative quinoline derivate drugs in market or clinical trials, classified
them into five major categories with detailed targets according to their main mechanisms, discussed the relationship
within the same mechanism, and generated a summative discussion with prospective expectations. For each mecha-
nism, the introduction of the target was presented, with the typical examples of quinoline derivate drugs.
Conclusion: This review has highlighted the quinoline drugs or candidates, suited them into corresponding targets
in their pathways, summarized and discussed. We hope that this review may help the researchers who are interested
in discovering quinoline derivate anti-cancer agents obtain considerable understanding of this specific topic.
Through the flourishing period and the vigorous strategies in clinical trials, quinoline drugs would be potential but
facing new challenges in the future.
Keywords: Anti-cancer agents, quinoline derivatives, marketed drugs, clinical trials, molecular targets, major mechanisms.
1. INTRODUCTION
Risking the health of people worldwide, cancer is the second
highest cause of death all over the world [1]. For quite a long pe-
riod, there has been a lack of available preventative measures, accu-
rate diagnose approaches and potent therapeutic schedules for most
of the cancers [2-4]. The major impediments are the complex
mechanisms and contributing factors in specific tumor types, and
what could be worse, the fast-paced life and environmental pollu-
tion nowadays might make the situations more sophisticated [5, 6].
Fortunately, along with the progress in medicine and therapies,
researchers have gradually comprehended the tumor pathogenesis
in the cellular and molecular scale, thus have revealed more and
more vital signaling pathways and key biological macromolecules
including membrane receptors and kinases [7-9]. As the consequent
assistance, the key nodes have been studied as specific targets,
which has enlarged the possibility of developing high efficiency,
low toxic and unique anti-cancer agents [10, 11]. Accordingly, in
the rational design procedures of anti-cancer agents, in considera-
tion of the relationship between targets and ligands, there are two
major strategies. One is analyzing the inner requirements of the
targets to polish a rough hit-compound, thus to obtain a well-crafted
*Address correspondence to these authors at the Institute of Chemistry and
BioMedical Sciences, School of Life Sciences, Nanjing University, Nanjing
210023, China; E-mail: ys_yang@nju.edu.cn and Henan Provincial Key
Laboratory of Children's Genetics and Metabolic Diseases, Children's
Hospital Affiliated to Zhengzhou University, Zhengzhou University,
Zhengzhou, 450018, China; E-mail: zhouchongchen@163.com
prodrug step-by-step [12]. The other is evaluating the actual po-
tency of the hit-compounds to deduce the modification demands
and rationally dispose the functional groups, thus to form a well-
organized candidate after screening [13]. Both strategies are actu-
ally based on structural factors, which infer the significance of dis-
cussing the exploitation of anti-cancer agents with certain backbone
structures [14, 15].
As one kind of widely used chemicals, quinoline derivatives
have been involved in fields such as medicinal chemistry [16], pes-
ticides [17], dyestuffs [18], analytical solvent [19], preservatives
[20] and rubber accelerators [21]. Although the flat conjugate struc-
ture with the capability of transferring electrons has brought them
applicable potential in solvability and photo-electric sensitivity, the
most attractive point is that it has appeared in the core position or
synthetic routes of quite a few drugs [22]. As the synthesis interme-
diates, the typical examples include the preparation of nicotinic acid
and hydroxyquinoline drugs [23]. As the structural component, we
would like to review this topic here with the classification based on
their molecular targets. In medicinal chemistry, although quinoline
derivatives have indicated broad activities including anti-bacterial
[24], anti-inflammatory [25], anti-malarial [26] and cardiotonic
effect [27], one of their most important roles is acting as anti-cancer
agents. Quinoline derivatives can exhibit fine results in treating
cancer via several different mechanisms, such as blocking the cell
cycle, inducing the cell apoptosis, inhibiting the vascular growth,
stopping the cell migration and activating the immune responses
[28-31]. Accordingly, along with the reported mechanisms, certain
target nodes through the involved pathways have been successfully
investigated to exploit hit-compounds, candidates, prodrugs and
1875-5992/21 $65.00+.00 © 2021 Bentham Science Publishers
826 Anti-Cancer Ag ents in Medicinal C hemistry, 2021, Vol. 21, No. 7 Man et al.
even listed drugs. In this review, we selected the representative
quinoline derivate drugs in market or clinical trials, classified them
into five major categories with detailed targets according to their
main mechanisms, discussed the relationship within the same
mechanism, and generated a summative discussion with prospective
expectations. We hope that this review may help the researchers
who are interested in discovering quinoline derivate anti-cancer
agents obtain considerable understanding of this specific topic.
2. QUINOLINE DERIVATE DRUGS IN VARIOUS
MECHANISMS
2.1. Through Receptor Tyrosine Kinases Mediated Pathways
The Receptor Tyrosine Kinases (RTKs) are a tremendous su-
perfamily of receptors [32]. They are not only receptors but also
enzymes. Their major biological function is acting as the receptors
for a large range of growth factors such as Epidermal Growth Fac-
tor (EGF), Platelet-Derived Growth Factor (PDGF), Vascular Endo-
thelial Growth Factor (VEGF), Fibroblast Growth Factor (FGF),
Nerve Growth Factor (NGF), insulin and the Insulin-lik e Growth
Factors (IGF), and the ephrins and angiopoietins [33-40]. Usually,
they can bind with the ligands, therefore, induce the phosphoryla-
tion of the tyrosine on the target protein. They commonly consist of
three sections, the ligand binding site outside the cell, the single
transmembrane hydrophobic helix and the tyrosine protein kinase
part in the cell [41]. RTKs have been reported to play vital roles in
the development and deterioration of cancers [42-44]. The mutation
of RTKs activates a series of signaling cascade and impacts the
expression or activation of downstream proteins. Focusing on the
quinoline derivate anti-cancer agents, we have illustrated a simpli-
fied pathway map with the involved targets in this review (Fig. 1).
The colored nodes are the targets of quinoline derivatives. The de-
tailed interactions with the ligands have been discussed for each
target in the following paragraphs, accompanied by the structures of
selected quinoline derivate drugs (Figs. 2-5).
2.1.1. Quinoline Derivatives as EGFR/HER2 Inhibitors
Epidermal Growth Factor Receptor (EGFR, ErbB1) and Human
Epidermal growth Factor Receptor-2 (HER2, ErbB2) both belong to
the ErbB family [45, 46]. The ErbB family includes four members,
EGFR, HER2, ErbB3 and ErbB4. They transport the signals to
major signal pathways such as Mitogen-Activated Protein Kinase
(MAPK) [47] or Phosphoinositide 3-Kinase/Protein KinaseB
(PI3K/Akt) [48] pathways, thus to regulate cell proliferation, migra-
tion, differentiation, apoptosis and movement [49-51]. The overex-
pression and amplification of the ErbB family and their ligands are
often observed in many forms of malignant tumors, which make the
ErbB family an important therapeutic target [52, 53]. Among them,
EGFR has been associated with Non-Small Cell Lung Cancer
(NSCLC) [54] and neuroglioma [55], while HER2 amplification is
found in breast, ovarian, bladder, NSCLC and several other tumor
types [56-58]. After being activated , the formation of EGFR
changes into dimer from monomer. The homologous dimer is
formed by two EGFR, while the heterologous dimer is usually
formed by EGFR and HER2 [59, 60]. Both preclinical and clinical
studies have convinced that bi-targeting ErbB inhibition is more
effective than single-targeting therapies [61].
The quinoline derivatives listed here are exactly the bi-targeting
inhibitors for EGFR and HER2. Though Trastuzumab (Herceptin®)
is still the most popular drug in curing HER2-positive breast can-
cers [62], the unavoidable drug resistance has pushed the research-
ers to find better solutions such as bi-targeting inhibition. As shown
in Fig. (2a), all three selected drugs have a similar core structure to
typical EGFR inhibitors (e.g., Erlotinib and Gefitinib) [63, 64]
whereas have replaced the quinazoline moiety by the quinoline.
Fig. (1). The simplified pathway map with the RTKs-mediated target proteins of quinoline derivate anti-cancer agents. The colored nodes are the targets of
quinoline derivatives. (A higher resolution / colour version of this figure is available in the electronic copy of the article).
Recent Progress in the Development of Quinoline Derivatives Anti-Cancer Agents in Medicinal Chemistry, 2021, Vol. 21, No. 7 827
NO
H
N
O
NCN
HN
Cl
O
N
Neratinib
Listed in 2017
HER2, ErbB4, EGFR
NO
H
N
O
CN
HN
O
N
Cl
N
Pyrotinib
Applying NDA
HER2, EGFR
NO
H
N
O
NCN
NH
F
Cl
Pelitinib
Phase II (Terminated)
EGFR, HER2
NO
O
NH2
O
H
N
F
Anlotinib
Listed in 2018
VEGFR3, PDGFR
β
, FGFRs,
VEGFR2, KIT
NO
H2N
OCl
H
N
O
H
N
O
Lenvatinib
Listed in 2015
PDGFR
α
, VEGFR3, FGFR2, VEGFR2,
KIT, FGFR3, RET, FGFR4, VEGFR1, FGFR1
NO
O
O
H
N
O O
H
N
F
Cabozantinib
Listed in 2012
VEGFR3, TYRO3, ROS, UFO, VEGFR2,
TIE2, c-Met/HGFR, KIT, NTRK2, RET, VEGFR1
NO
O
O
H
N
O O
H
N
F
F
N
O
Foretinib
Phase II
VEGFR2, c-Met/HGFR
NO
O
O
H2N
H
N
O
Lucitanib
Phase II
PDGFR
α
, VEGFR3, PDGFB,
VEGFR2, VEGFR1
NO
O
N
O
OH
N
O
PF-337210
Phase II
VEGFR2
NO
O
HO
FH
N
O
N
N
O
Ningetinib
Phase I
UFO, VEGFR2, c-Met/HGFR
NO
O
FH
N
S
N
H
O
H
N
O
TAS-115
Phase I
VEGFR2, c-Met/HGFR
N
NH
S
O
H
N
O
F3C
Thiophenib
Phase I
PDGFR
β
, VEGFR2, KIT
a) EGFR/HER2
Inhibitors
b) VEGFR/PDGFR/RET
Inhibitors
c) c-Met
Inhibitors
NO
O
NN
N
N
AMG-208
Phase II (Terminated)
c-Met/HGFR
N
NN
N
N
F
ONH
Capmatinib
Phase III
c-Met/HGFR
NN
N
N
N
F F
N
N
JNJ-38877605
Phase I
c-Met/HGFR
NN
N
N
N
F F
N
JNJ-38877618
Phase I
c-Met/HGFR
N
NN
N
N
N
N
NOH
PF-04217903
Phase I (Terminated)
c-Met/HGFR
S
NN
N
N
N
N
N
SGX-523
Phase I (Terminated)
c-Met/HGFR
Fig. (2). The structures of quinoline derivatives as anti-cancer agents for RTKs. (a) EGFR/HER2 inhibitors; (b) VEGFR/PDGFR/RET inhibitors; (c) c-Met
inhibitors.
828 Anti-Cancer Agents in Medicinal C hemistry, 2021, Vol. 21, No. 7 Man et al.
Neratinib Maleate was approved in July 2017 for the use as an ex-
tended adjuvant treatment in adult patients with early-stage HER2-
overexpressed/amplified breast cancer, to follow adjuvant trastu-
zumab-based therapy [65]. Its approval was granted to Puma Bio-
technology Inc. for the trade name Nerlynx. It exhibited antitumor
action against EGFR, HER2 and ErbB4 positive carcinomas [66].
With the modification in the pharmacokinetic group, Pyrotinib
Maleate was applying New Drug Application (NDA) by Jiangsu
Hengrui Medicine Co. [67]. Pelitinib (EKB-569; WAY-EKB 569),
who lost the pyridine moiety, indicated better potency for EGFR
(IC50=38.5nM) instead of HER2 (IC50=1255nM) [68]. It could also
exhibit inhibitory activity against Src and MEK/ERK. Exploited by
Pfizer, it was utilized in NSCLC and colorectal cancer but termi-
nated in phase II clinical trials. In general, for inhibiting the ErbB
family, the backbone was usually a 3-cyanoquinoline. Moreover,
quinoline derivatives could supply a good fit for the updated pursuit
of bi-targeting ErbB inhibition, thus might be potential for further
optimization of existing listed drugs.
2.1.2. Quinoline Derivatives as VEGFR/PDGFR/RET Inhibitors
Besides the ErbB family, other major RTKs, including Vascular
Endothelial Growth Factor Receptor (VEGFR), Platelet-Derived
Growth Factor Receptors (PDGFR) and RET, can also be inhibited
by quinoline derivatives. VEGF is a signal protein that stimulates
angiogenesis in cells with the function of promoting angiogenesis
and regeneration [69]. VEGF binds to VEGFR on the surface of the
cell membrane, and produces biological effects through a series of
signaling pathways, leading to angiogenesis [70]. In 1971, Folkman
first proposed the theory of tumor angiogenesis [71]. Accordingly,
tumor growth should depend on tumor angiogenesis. Moreover,
VEGFR can cause drug resistance by regulating the downstream
pathways to motivate endothelial and tumor cells to proliferate and
spread. Now VEGFR is one of the hottest targets for monoclonal
antibody in spite of PD-1 and TNF-α, while actually, there have
been several listed chemical drugs for VEGFR [72]. Among them,
there are also quinoline derivates.
PDGFR system mainly affects the development of the embryo,
especially the angiogenesis and organ formation in the process of
development, while in the adult stage, under physiological state, it
is related to the repair of tissue and wound healing [73]. However,
the overexpression of PDGFR will lead to the occurrence and de-
velopment of tumors [74]. As a kind of transmembrane glycopro-
tein, PDGFR is composed of α and β protein tyrosine kinase
subunits. Furthermore, the α subunit can trigger the downstream
enzyme effectors (PI3K, MAPK, Src, SHP2, etc) and non-enzyme
effectors (Crk, Sch, Grbs, etc) [75], thus affect the cell growth,
survival, proliferation and differentiation.
RET protein, encoded by the corresponding proto oncogene, is
a transmembrane tyrosine kinase receptor that can activate a series
of downstream pathways after itself being activated [76]. Despite
PI3K, MAPK, Src, it can also activate the Focal Adhesion Kinase
(FAK) pathway [77].
Here we listed the VEGFR/PDGFR/RET multi-target tyrosinase
inhibitors with quinoline as the pharmacodynamic group (Fig. 2b).
Anlotinib Dihydrochloride, launched by CTTQ Pharmaceutical Co.,
Ltd. and listed in 2018, was a high-profile drug for NSCLC [78]. It
could inhibit a wide range of RTKs, including VEGFRs, PDGFRs,
FGFRs and KIT, whereas RET was not involved. Recently, based
on the clinical trial, Anlotinib was also approved for soft tissue
sarcoma [79]. Previously marketed drugs raised well-known exam-
ples, such as Lenvatinib Mesylate (listed in 2015) [80] and Cabo-
zantinib S-malate (listed in 2012) [81]. Both of them are different
from specific inhibitors for one or two unique targets, instead, they
are both available for almost ten targets, of course, with
VEGFR/PDGFR/RET. Especially for Cabozantinib, its targets can
even include TYRO3, ROS, UFO, TIE2, c-Met/HGFR and NTRK2.
Lenvatinib seemed particularly effective in the treatment of liver
cancer induced by hepatitis B, which provided new therapeutic
options for patients with liver cancer in a later period [82]. Mean-
while, Cabozantinib was approved by the FDA for refractory or
relapsed medullary thyroid carcinoma, and later renal cancer that
could not be treated by Sutent (Sunitinib) [83]. Other candidates in
Phase II clinical trials include Foretinib [84], Lucitanib [85] and
PF-337210 [86], while the ones in Phase II clinical trials contain
Ningetinib Tosylate [87], TAS-115 [88] and Thiophenib Tosylate
[89]. All of them indicated fewer targets than Lenvatinib or Cabo-
zantinib. The similar target of these candidates was VEGFR2, as we
demonstrated above, the hottest target for monoclonal antibody as
well. With a glance at all the mentioned drugs and candidates, we
can find that these compounds often have the quinoline core modi-
fied with cyclopropyl or urea-like groups.
2.1.3. Quinoline Derivatives as c-Met Inhibito rs
Although the multi-targeted drug Cabozantinib also inferred the
inhibitory effect against c-Met, quinoline moiety could be em-
ployed in specific c-Met inhibitors. In RTKs, c-Met is the only
receptor protein which can highly be affined with Hepatocyte
Growth Factor (HGF) [90]. The interactions between HGF and c-
Met can cause the phosphorylation and a series of biochemical
procedures to activate the signal pathway.
It is a pity that till now, there has been none listed drug for c-
Met inhibition. Crtzotinib might be one potential choice but should
not be emphasized here due to the lack of quinoline [91]. Within
quinoline derivatives, the most potential one is Capmatinib (Fig.
2c), which was developed by Incyte, obtained by Novartis, and is in
Phase III clinical trials now [92]. In ASCO 2019 conference, the
announced data of Capmatinib in treating NSCLC suggested the
curing potential in Metexon14-skipping locally later period or me-
tastatic situations. As shown, Capmatinib links the quinoline group
to the nitrogen heterocyclic ring with a methylene bridge. With a
similar structural feature, there are other candidates in Phase I clini-
cal trials, such as JNJ-38877605 [93], JNJ-38877618 [94], PF-
04217903 [95] and SGX-523 [96]. They were all developed for
treating solid tumors. As a unique case, AMG-208 [97], terminated
in Phase II clinical trials, has a methoxyl group on the quinoline and
a longer linker. On the whole, all the quinoline derivatives designed
for c-Met inhibition bear the highly conservative backbone, and are
all specifically targeted at the c-Met/HGFR pair.
2.1.4. Quinoline Derivatives as PI3K Inhibitors
Seeking the downstream of RTKs, MAPK and PI3K-Akt-
mTOR pathways are two major pathways. Some of the quinoline
derivatives can block them by inhibiting RTKs, while at the same
time, PI3K itself has been studied as the target of quinoline derivate
drugs (Fig. 3). PI3K-Akt-mTOR pathway plays an important role in
cell growth, differentiation, apoptosis and other aspects. Therefore,
many members of this signal transduction are key drug targets in
the process of cancer, immunity and thrombosis control [98, 99].
PI3K can act as both Ser/Thr kinase and phosphatidylinositol
kinase, and it can be divided into three categories with differed
structures and functions. The most widely investigated one is Class
I PI3K [100]. Among the four catalytic subunits (α, β, δ, γ), δ and γ
are limited within leukocytes, whereas the rest are widely distrib-
uted in various cells. The downstream signal pathway of PI3K is
complex due to some feedback loops. Each of the four catalytic
isomers of Class I PI3K preferentially regulates specific signal
transduction and tumor cell survival, which depends on the type of
malignant tumor and the genetic or epigenetic changes.
Most of the quinoline derivatives here are PI3K pan-blockers
(Fig. 3a). As a typical example, Dactolisib, launched by Novartis
and in Phase II clinical trials now [101], has been set into the trials
including bladder cancer, pancreatic cancer, breast cancer, renal cell
Recent Progress in the Development of Quinoline Derivatives Anti-Cancer Agents in Medicinal Chemistry, 2021, Vol. 21, No. 7 829
cancer, and prostate cancer [102]. Its structural backbone is an imi-
dazolone-combined quinoline, while it also has an extra quinoline
group linked with the backbone. Another candidate developed by
Novartis, BGT-226, was terminated in Phase II clinical trials [103].
It retained the backbone of imidazolone-combined quinoline but
lost the other quinoline moiety. Similar situations in structural fea-
tures happened in LY-3023414 (in Phase II clinical trials for
NSCLC by Eli Lilly) [104] and Panulisib (in Phase I clinical trials
for solid tumor by Piramal Phytocare) [105]. Besides, also for PI3K
pan-blocker, GlaxoSmithKline exploited a candidate with alterna-
tive backbone, and the so-named Omipalisib inferred good potency
in Phase I clinical trials for the treatment of idiopathic pulmonary
fibrosis, solid tumors and lymphoma [106]. Despite the quinoline
backbone, its sulfonamide group is somehow attractive. Meanwhile,
GlaxoSmithKline attempted to develop the specific inhibitor for
PI3Kα, however, as a result, GSK-615 seemed to suffer a failure in
Phase I clinical trials for solid tumors and lymphoma [107]. Gener-
ally, although most of the quinoline derivate candidates suggested
splendid efficiency, unfortunately, none of them achieved the status
of being marketed at present.
2.1.5. Quinoline Derivatives as Src/Abl Inhibitors
Src (Proto-oncogene Tyrosine-protein Kinase) is actually not
involved in the traditional sense of RTKs. Discovered by Peyton
Rous in 1910, the protooncogene c-src opened the researches on the
structure, regulation, localization and function of the corresponding
protein Src [108]. Src can act as the inserted step within the typical
RTKs mediated pathways. By responding to the growth factors
directly or transporting the signals from G protein coupled recep-
tors, Src then activates the typical pathways including MAPK,
PI3K, FAK and STAT3, thus regulates the subsequent survival,
proliferation and motility [109]. The RTK inhibitor Imatinib could
show potency for most of the chronic myeloid leukemia patients,
while the following Nilotinib and Dasatinib were used to overcome
the drug resistance [110-112]. However, the toxicity and further
drug resistance made the requirement for new agents urgent.
Bosutinib, launched by Pfizer, was marketed in 2012 to act as a
potent orally-taken Src/Abl dual inhibitor [113]. It was over 15-fold
more potent than Imatinib, and low-toxic. As shown in Fig. (3b), its
3-cyanoquinoline backbone is quite similar to the above mentioned
EGFR/HER2 inhibitors, but the other linked groups are obviously
different.
2.2. Through DNA Related Mechanisms
DNA related mechanisms have been utilized in treating cancer
for a long period [114]. Till now, the platinum containing drugs are
the most widely used chemotherapeutics [115]. After the first ex-
ample, Cisplatin, being marketed in 1978, over 40 years evolution of
these drugs has not only promoted the drugs into the therapies for a
variety of cancers, but also broadened the corresponding mechanisms
around DNA. Since binding to the DNA sequences is cell cycle
nonspecific, these agents usually indicated relatively strong nephro-
toxicity, hematotoxicity and neurotoxicity [116]. Quinoline deriva-
tives selected in this review were designed for other DNA related
mechanisms with specific protein targets (Fig. 4). They inhibit their
specific targets and subsequently cause the corresponding effect to
impact the morphology or sequences of DNA, therefore, finally
induce the apoptosis of cancer cells. Based on the concrete mecha-
nisms, the quinoline derivate drugs or candidates are detailed as
follows.
2.2.1. Quinoline Derivatives as DNA Topoisomerase Inhibitors
DNA Topoisomerase (TOP) is a general term of enzymes that
catalyze the mutual transformation of DNA topoisomers [117]. It is a
vital kind of biological enzymes in both eukaryotic and prokaryotic
N
N
N
N
O
NC
Dactolisib
Phase II
PI3K
β
, mTORC2, mTORC1,
PI3K
δ
, PI3K
α
, PI3K
γ
N
N
N
N
O
O
NCF
3
HN
BGT-226
Phase II (Terminated)
Class I PI3K, mTORC2, mTORC1
N
N
N
N
O
HO
O
LY-3023414
Phase II
Class I PI3K, mTOR
N
N
N
N
N
N
CN
NC
H
2
N
F
3
C
Panulisib
Phase I
Class I PI3K, mTORC2, mTORC1
N
N
HN
S
O
O
GSK-615
Phase I (Terminated)
PI3K
α
N
N
N
NO
HN
S OO
F
FOmipalis ib
Phase I
PI3K
β
, mTORC2, mTORC1,
PI3K
δ
, PI3K
α
, PI3K
γ
a) PI3K
Inhibitors
b) Src/Abl
Inhibitor
N
O
ON
N
CN
HN
Cl Cl
O
Bosutinib
Listed in 2012
LYN, TPK HCK, Src, Bcr-Abl
Fig. (3). The structures of quinoline derivatives as anti-cancer agents for other RTKs-mediated pathways: (a) PI3K inhibitors; (b) Src/Abl inhibitors.
830 Anti-Cancer Agents in Medicinal C hemistry, 2021, Vol. 21, No. 7 Man et al.
N
N
O
N
H
O
HO
O
Belotecan
Listed in 2003
TOP1
N
N
O
O
O
HO
O
O
N
N
Irinotecan
Listed in 2011
TOP1
N
HN
OH
NS
O
O
Amsacrine
Listed
TOP2
N
N
O
O
O
HO
Camptothecin
Listed
TOP1
N
N
O
O
O
HO
Rubitecan
Applying NDA
TOP1
NO
2
N
N
O
O
O
HO
NH
2
9-Aminocamptothecin
Phase III (Terminated)
TOP1
N
N
O
O
O
HO
HO
Si
N
N
O
O
O
HO
Si
Cositecan
Phase III
TOP1
AR-67
Phase II (Terminated)
TOP1
O
N
O
N
OH
N
O
O
O
O
N
O
2
N
O
2
NNO
2
O
2
N
TLC-388 HCl
Phase II
TOP1
N
N
O
O
HO O
F
F
Diflomotecan
Phase II (Terminated)
TOP1
N
N
O
O
OH
O
O
HO
DRF-1042
Phase II
TOP1
N
N
O
O
O
HO
O
O
N
N
Lurtotecan
Phase II (Terminated)
TOP1
N
HN
OH
NS
O
O
Asulacrine
Phase II (Terminated)
TOP2
ONH
N
HN O
N
Acridine
Phase II (Terminated)
TOP2, TOP1
N
N
O
O
O
HO
N
O
Gimatecan
Phase II (Terminated)
TOP1
N
N
O
O
O
HO
NN
TP-300
Phase II
TOP1
N
O
HO NH
N
BMS-247615
Phase II
TOP2, TOP1
N
H
O
NO
2
N
N
N
Pyrazoloacridine
Phase II (Terminated)
TOP2, TOP1
N
N
O
O
O
HO
N
ONH
2
Namitecan
Phase I
TOP1
N
N
O
O
O
HO
O
O
N
N
Simmitecan
Phase I
TOP1
N
N
O
O
O
O
O
CZ-48
Phase I
TOP1
N
N
O
O
O
HO
Cl
N
Elomotecan
Phase I
TOP2, TOP1
N
N
O
O
O
O
O
NO
2
CZ-112
Phase I (Terminated)
TOP1
N
N
O
O
O
O
O
H
N
Genz-644282
Phase I
TOP1
N
N
O
O
O
HO
O
OO
OO
Tenifatecan
Phase I (Terminated)
TOP1
N
N
O
O
O
O
O
H
N
O
N
H
N
N
H
N
H
S
OO
O
HO OH
Afeletecan
Phase I (Terminated)
TOP1
a) DNA TOP
Inhibitors
b) ATM
Inhibitors
N
N
N
NO
O
ON
AZD-0156
Phase I
ATM N
N
N
NO
ON
F
AZD-1390
Phase I
ATM
Fig. (4). The structures of quinoline derivatives as anti-cancer agents for DNA-related pathways: (a) DNA TOP inhibitors; (b) ATM inhibitors.
Recent Progress in the Development of Quinoline Derivatives Anti-Cancer Agents in Medicinal Chemistry, 2021, Vol. 21, No. 7 831
cells [118]. All through the processes of DNA transcription, replica-
tion, chromosome separation and gene expression, TOP participates
in the regulation. With the unaffected higher level of expression in
tumor cells than in normal ones, the TOP family has been regarded
as suitable targets in anti-cancer strategies [119].
Since the TOP family can be generally divided into TOP1 and
TOP2, quinoline derivate drugs for these two subtypes are quite
different in structures (Fig. 4a). Most of them are TOP1 inhibitors
with a typical backbone mimicking Camptothecin [120]. When we
look into the listed drugs, despite the original Camptothecin, Be-
lotecan Hydrochloride (listed in 2003) [121] and Irinotecan Hydro-
chloride (listed in 2011) [122] are all in the similar structural pat-
tern and targeted at TOP1. Amsacrine, also listed, is a TOP2 inhibi-
tor for treating leukemia [123]. Its backbone is acridine, being dis-
tinguished from the Camptothecin-mimicking core for TOP1. In
spite of the marketed drugs, there are a large number of candidates
in their clinical trials for inhibiting the TOP family. As typical
strategies of developing TOP1 inhibitory anti-cancer agents, the
modification on Camptothecin led to several practical choices such
as adding substitutes, enlarging the aliphatic ring and linking other
functional fragments. The examples which are close to achieving
marketed drugs include Rubitecan (applying NDA) [124], 9-
Aminocamptothecin (terminated in Phase III) [125] and Cositecan
(in Phase III) [126]. All of these three candidates followed the
choice of adding suitable substitutes. Focusing on TOP1, other
Camptothecin-like candidates can be organized as follows: AR-67
(terminated in Phase II) [127], DRF-1042 (in Phase II) [128], Lur-
totecan (in Phase II) [129], Gimatecan (terminated in Phase II)
[130], TP-300 (in Phase II) [131], Namitecan (in Phase I) [132],
Simmitecan (in Phase I) [133], CZ-48 (in Phase I) [134], CZ-112
(in Phase I) [135] and Genz-644282 (in Phase I) [136] for adding
substitutes; Diflomotecan (terminated in Phase II) [137] for enlarg-
ing the aliphatic ring; TLC-388 (in Phase II) [138], Tenifatecan
(terminated in Phase I) [139] and Afeletecan (terminated in Phase I)
[140] for linking other functional fragments. Especially, one unique
case is Elomotecan (in Phase I) [141], because it bears an aliphatic
ring-enlarged Camptothecin backbone but exhibited the activities
for inhibiting both TOP2 and TOP1. Besides, there are several
acridine derivate candidates, whose designed target should be TOP2.
Actually, however, only Asulacrine (terminated in Phase II) [142]
was reported to specifically inhibit TOP2. Both of the other two
examples, Acridine (terminated in Phase II) [143] and Pyrazoloac-
ridine (terminated in Phase II) [144], did not present enough selec-
tivity for TOP2 from TOP1. Additionally, as a unique case, BMS-
247615 (in Phase II) [145], now being developed by Bristol-Myers
Squibb, is a TOP2/TOP1 bi-targeting inhibitor with a backbone of
indenoquinoline, which is different from neither Camptothecin nor
acridine. As a whole, no matter applied for TOP1, TOP2 or both of
them, exploiting quinoline derivatives for inhibiting the TOP family
seems a practical orientation with a considerable fever.
2.2.2. Quinoline Derivatives as ATM Kinase Inhibitors
Ataxia Telangiectasia Mutant (ATM) kinase inhibitors have
potential chemical or radiation sensitization and antitumor activities
due to blocking the ATM-induced signal transduction [146]. This
kind of blocking can prevent the activation of DNA damage check-
points, therefore, impact the DNA damage repair and lead to cell
death of ATM overexpressed tumor cells [147]. Meanwhile, ATM
N
ON
NOH
H
H
F
F
Zosuquidar
Phase III (Terminated)
PGY1
ON
N
HO
O
N
Dofequidar
Phase III (Terminated)
PGY1
O
N
N
N
N
O
O
Laniquidar
Phase II
PGY1
N
NOH
H
Cinchonine
Phase I (Terminated)
PGY1
a) PGY1
Inhibitors
N
N
N
NH2
Imiquimod
Listed in 1997
TLR7
N
N
N
NH2
H
N
S
O
O
PF-4878691
Phase II (Terminated)
TLR7
N
N
N
NH2
O
N
H
O
16
MEDI-9197
Phase I
TLR7, TLR8
N
HN
O
Cl
H
F
BMS-986205
Phase III
IDO
N
N
NH2
N
N
O
O
Crenolanib
Phase III
PDGFRs, FLT3
N
H2N
O
NN N
Galunisertib
Phase III
TGFBR1
O
N
N
O
O
N
Cl
H
N
OOH
Meclinertant
Phase III (Terminated)
NTS
b) TLR
Agonists
c) Other
Targets
Fig. (5). The structures of quinoline derivatives as anti-cancer agents through glycoprotein transmembrane transport, immune enhancement and other mechanisms:
(a) PGY1 inhibitors; (b) TLR agonists; (c) agents for other targets.
832 Anti-Cancer Agents in Medicinal C hemistry, 2021, Vol. 21, No. 7 Man et al.
inhibitors can also make tumor cells sensitive to chemotherapy and
radiotherapy.
ATM is a relatively new anti-cancer target, so the exploitation
of its inhibitors is still preliminary at present. Of course, quinoline
derivatives for this target are also in the initial stage (Fig. 4b).
AZD-0156 [148] and AZD-1390 [149], the selected two examples,
were both developed by AstraZeneca, and are in their Phase I clini-
cal trials for treating solid tumors. Their structures are similar to the
above mentioned PI3K inhibitors, but the detailed parts have been
polished.
2.3. Through Glycoprotein Transmembrane Transport
Mechanism
The glycoprotein here is P-glycoprotein (P-gp), a member of
the ATP-Binding Cassette (ABC) transmembrane transporter super-
family [150]. It can pump the chemotherapeutic drugs outside the
cells to reduce the intracellular drug concentration. P-gp inhibitors,
conversely, can directly bind to P-gp, block the pumping-out and
cause accumulation of the drugs, therefore, reverse the drug resis-
tance. As shown in Fig. (5a), quinoline derivatives usually act as
the inhibitors of PGY1, one kind of P-gp. Almost all of them are
facing difficulty on the road to marketed drugs. Both Zosuquidar
Trihydrochloride (developed by Kanisa for acute myeloid leuke-
mia) [151] and Dofequidar Fumarate (developed by Bayer for
NSCLC and breast cancer) [152] were terminated in their Phase III
clinical trials. Laniquidar (developed by Janssen for breast cancer)
[153] is in Phase II clinical trials with no progress. Cinchonine
[154], an alkaloid with special backbone, was also terminated in
Phase I for treating solid tumors.
2.4. Through the Immune Enhancement Mechanism
As the target of the quinoline derivatives, Toll-Like Receptors
(TLRs) are widely expressed in the natural immune system [155].
Recent researches connected their expression with the cancer
physiology, and indicated that TLR7/TLR8 agonists could activate
several immune pathways at the same time, leading to effective
anti-tumor immunity [156]. In Fig. (5b), the selected TLR agonists
are all Imidazoquinoline derivatives with alkyl chain modification.
Among them, Imiquimod was listed in 1997 for inhibiting TLR7 by
Minnesota Mining and Manufacturing (3M) Company [157]. How-
ever, another attempt, PF-4878691, also targeting at TLR7, was
terminated in Phase II [158]. Now, MEDI-9197, designed for both
TLR7 and TLR8, has been promoted in Phase I for treating solid
tumors [159].
2.5. Through Other Mechanisms
In Fig. (5c), quinoline derivatives as anti-cancer agents via
other mechanisms have been displayed. BMS-986205 (developed
by Bristol-Myers Squibb), inhibiting Indoleamine 2,3-Dioxygenase
(IDO), is in Phase III clinical trials for solid tumors, bladder cancer
and melanoma [160]. Crenolanib Besylate (developed by Pfizer), as
a pan-FLT3 (FMS-like tyrosine kinase 3) inhibitor, is in Phase III
for AML [161]. Galunisertib (developed by Eli Lilly and Com-
pany), targeting at TGF-β Receptor type-1 (TGFBR1), is in Phase
III for Myelodysplastic Syndromes and solid tumors [162]. As a
negative example, only Meclinertant (developed by Sanofi), affect-
ing Neurotensin (NTS), was terminated in Phase III for prostate
cancer and colorectal cancer [163].
CONCLUSION
In summary, along with the deeper understanding of the vital
nodes in pathways and the principle for structural modification,
various quinoline derivatives have been developed into anti-cancer
agents. According to specific structural features, they can be ap-
plied in treating can cer via several mechanisms. Two major mecha-
nisms are RTKs inhibition and DNA obstruction. Acting as RTK
inhibitors, quinoline derivate drugs indicated the preference for
relatively upstream proteins, which were the receptors themselves
(EGFR/HER2, VEGFR, PDGFR, RET, c-Met) or the ones closely
next to the receptors (Src, PI3K). Impacting DNA, quinoline de-
rivate drugs inhibited their specific targets and subsequently caused
the corresponding effect to impact the morphology or sequences of
DNA, thus overcame the disadvantage of lacking selectivity from
normal cells. Among the selected compounds, ten are marketed
drugs. Half of them are involved in the RTKs inhibition, while four
are DNA topoisomerase inhibitors. The rest one is TLR agonist. We
can generally demonstrate that VEGFR2 and DNA TOP1 are the
most discovered targets for quinoline derivatives, and new research
target such as ATM kinase has been promoted to clinical trials. In
these drugs and candidates, quinoline moiety not only works as the
core, but serves as the linked functional group. These hints inferred
the significance of quinoline in both structural extension and phar-
macophore mimicking.
Furthermore, if we broaden our eyesight, we would find that the
market of small molecule drugs is gradually eroded by macromo-
lecular drugs. Thus in the future, the advanced parameters to de-
velop quinoline derivate drugs should be considered at least on two
orientations. One is continuously seeking for candidates with high
potency and low toxicity. The other is addressing the relationship
between quinoline-derived compounds and macromolecular drugs.
Still, there is a need to find the equilibrium in potency and mor-
phology.
Indeed, quinoline drugs experienced the flourishing period and
the vigorous strategies in clinical trials, and their future would be
potential but facing new challenges.
CONSENT FOR PUBLICATION
Not applicable.
FUNDING
This study was sponsored by Guangxi University for Nationali-
ties Research Funded Project, China (No. 2018KJQD13), Guangxi
Minority Preparatory Education Base Research Project, China (No.
2017B002), and Guangxi Biological Polysaccharide Separation,
Purification and Modification Research Platform, China (No.
GKZY18076005).
CONFLICT OF INTEREST
The author declares no conflict of interest, financial or other-
wise.
ACKNOWLEDGEMENTS
Declared none.
REFERENCES
[1] Smith, R.A.; Manassaram-Baptiste, D.; Brooks, D.; Doroshenk, M.;
Fedewa, S.; Saslow, D.; Brawley, O.W.; Wender, R. Cancer
screening in the United States, 2015: A review of current American
cancer society guidelines and current issues in cancer screening.
CA Cancer J. Clin., 2015, 65(1), 30-54.
http://dx.doi.org/10.3322/caac.21261 PMID: 25581023
[2] Marmot, M.G.; Altman, D.G.; Cameron, D.A.; Dewar, J.A.;
Thompson, S.G.; Wilcox, M. Independent UK Panel on Breast
Cancer Screening. The benefits and harms of breast cancer screen-
ing: an independent review. Lancet, 2012, 380(9855), 1778-1786.
http://dx.doi.org/10.1016/S0140-6736(12)61611-0 PMID:
23117178
[3] Chu, K.F.; Dupuy, D.E. Thermal ablation of tumours: Biological
mechanisms and advances in therapy. Nat. Rev. Cancer, 2014,
14(3), 199-208.
http://dx.doi.org/10.1038/nrc3672 PMID: 24561446
Recent Progress in the Development of Quinoline Derivatives Anti-Cancer Agents in Medicinal Chemistry, 2021, Vol. 21, No. 7 833
[4] Duan, Y.; Liu, W.; Tian, L.; Mao, Y.; Song, C. Targeting tubulin-
colchicine site for cancer therapy: Inhibitors, antibody-drug conju-
gates and degradation agents. Curr. Top. Med. Chem., 2019,
19(15), 1289-1304.
http://dx.doi.org/10.2174/1568026619666190618130008 PMID:
31210108
[5] Satarug, S. Long-term exposure to cadmium in food and cigarette
smoke, liver effects and hepatocellular carcinoma. Curr. Drug Me-
tab., 2012, 13(3), 257-271.
http://dx.doi.org/10.2174/138920012799320446 PMID: 22455552
[6] van den Dungen, M.W.; Rijk, J.C.W.; Kampman, E.; Steegenga,
W.T.; Murk, A.J. Steroid hormone related effects of marine persis-
tent organic pollutants in human H295R adrenocortical carcinoma
cells. Toxicol. In Vitro, 2015, 29(4), 769-778.
http://dx.doi.org/10.1016/j.tiv.2015.03.002 PMID: 25765474
[7] Roberts, L.R.; Gores, G.J. Hepatocellular carcinoma: Molecular
pathways and new therapeutic targets. Semin. Liver Dis., 2005,
25(2), 212-225.
http://dx.doi.org/10.1055/s-2005-871200 PMID: 15918149
[8] Liu, L.; Cao, Y.; Chen, C.; Zhang, X.; McNabola, A.; Wilkie, D.;
Wilhelm, S.; Lynch, M.; Carter, C. Sorafenib blocks the
RAF/MEK/ERK pathway, inhibits tumor angiogenesis, and induces
tumor cell apoptosis in hepatocellular carcinoma model
PLC/PRF/5. Cancer Res., 2006, 66(24), 11851-11858.
http://dx.doi.org/10.1158/0008-5472.CAN-06-1377 PMID:
17178882
[9] Duan, Y.T.; Sangani, C.B.; Liu, W.; Soni, K.V.; Yao, Y. New
promises to cure cancer and other genetic diseases/disorders: Epi-
drugs through epigenetics. Curr. Top. Med. Chem., 2019, 19(12),
972-994.
http://dx.doi.org/10.2174/1568026619666190603094439 PMID:
31161992
[10] Han, H.; Hurley, L.H. G-quadruplex DNA: A potential target for
anti-cancer drug design. Trends Pharmacol. Sci., 2000, 21(4), 136-
142.
http://dx.doi.org/10.1016/S0165-6147(00)01457-7 PMID:
10740289
[11] Hare, J.I.; Lammers, T.; Ashford, M.B.; Puri, S.; Storm, G.; Barry,
S.T. Challenges and strategies in anti-cancer nanomedicine devel-
opment: An industry perspective. Adv. Drug Deliv. Rev., 2017, 108,
25-38.
http://dx.doi.org/10.1016/j.addr.2016.04.025 PMID: 27137110
[12] Chinthala, Y.; Thakur, S.; Tirunagari, S.; Chinde, S.; Domatti,
A.K.; Arigari, N.K.; Srinivas, K.V.N.S.; Alam, S.; Jonnala, K.K.;
Khan, F.; Tiwari, A.; Grover, P. Synthesis, docking and ADMET
studies of novel chalcone triazoles for anti-cancer and anti-diabetic
activity. Eur. J. Med. Chem., 2015, 93, 564-573.
http://dx.doi.org/10.1016/j.ejmech.2015.02.027 PMID: 25743216
[13] Roy, K.K.; Singh, S.; Saxena, A.K. Integration-mediated prediction
enrichment of quantitative model for Hsp90 inhibitors as anti-
cancer agents: 3D-QSAR study. Mol. Divers., 2011, 15(2), 477-
489.
http://dx.doi.org/10.1007/s11030-010-9269-y PMID: 20740314
[14] Cragg, G.M.; Grothaus, P.G.; Newman, D.J. Impact of natural
products on developing new anti-cancer agents. Chem. Rev., 2009,
109(7), 3012-3043.
http://dx.doi.org/10.1021/cr900019j PMID: 19422222
[15] Liu, W.; Wang, X.; Zhu, H.; Duan, Y. Precision tumor medicine
and drug targets. Curr. Top. Med. Chem., 2019, 19(17), 1488-1489.
http://dx.doi.org/10.2174/156802661917190828111130 PMID:
31592750
[16] Afzal, O.; Kumar, S.; Haider, M.R.; Ali, M.R.; Kumar, R.; Jaggi,
M.; Bawa, S. A review on anticancer potential of bioactive hetero-
cycle quinoline. Eur. J. Med. Chem., 2015, 97, 871-910.
http://dx.doi.org/10.1016/j.ejmech.2014.07.044 PMID: 25073919
[17] Kaur, K.; Jain, M.; Reddy, R.P.; Jain, R. Quinolines and structur-
ally related heterocycles as antimalarials. Eur. J. Med. Chem.,
2010, 45(8), 3245-3264.
http://dx.doi.org/10.1016/j.ejmech.2010.04.011 PMID: 20466465
[18] Gupta, V.K.; Mittal, A.; Gajbe, V. Adsorption and desorption stud-
ies of a water soluble dye, Quinoline Yellow, using waste materi-
als. J. Colloid Interface Sci., 2005, 284(1), 89-98.
http://dx.doi.org/10.1016/j.jcis.2004.09.055 PMID: 15752789
[19] Zaoui, F.; Didi, M.A.; Villemin, D. Investigation of 7-
((dioctylamino)methyl)quinoline-8-ol for uptake and removal of
uranyl ions. J. Radioanal. Nucl. Chem., 2013, 295(1), 419-424.
http://dx.doi.org/10.1007/s10967-012-1789-8
[20] Dinç Zor, Ş.; Aşçı, B.; Aksu Dönmez, Ö.; Yıldırım Küçükkaraca,
D. Simultaneous determination of potassium sorbate, sodium ben-
zoate, quinoline yellow and sunset yellow in lemonades and lemon
sauces by HPLC using experimental design. J. Chromatogr. Sci.,
2016, 54(6), 952-957.
http://dx.doi.org/10.1093/chromsci/bmw027 PMID: 26951541
[21] Sributr, A.; Yamsaengsung, W.; Wimolmala, E.; Kositchaiyong,
A.; Isarangkura, K.; Sombatsompop, N. Effects of solution and
solid forms of 2-hydroxypropyl-3-piperazinyl-quinoline carboxylic
acid methacrylate on antibacterial, physical and mechanical proper-
ties of polypropylene sheeting. J. Plast. Film Sheeting, 2015, 31(3),
248-268.
http://dx.doi.org/10.1177/8756087914561137
[22] Kumar, S.; Bawa, S.; Gupta, H. Biological activities of quinoline
derivatives. Mini Rev. Med. Chem., 2009, 9(14), 1648-1654.
http://dx.doi.org/10.2174/138955709791012247 PMID: 20088783
[23] Woodward, C.F.; Badgett, C.O.; Kaufman, J.G. Chemical-catalytic
liquid-phase oxidation of nicotine, beta-picoline, and quinoline to
nicotinic acid. Ind. Eng. Chem., 1944, 36, 544-546.
http://dx.doi.org/10.1021/ie50414a012
[24] Eswaran, S.; Adhikari, A.V.; Shetty, N.S. Synthesis and antimicro-
bial activities of novel quinoline derivatives carrying 1,2,4-triazole
moiety. Eur. J. Med. Chem., 2009, 44(11), 4637-4647.
http://dx.doi.org/10.1016/j.ejmech.2009.06.031 PMID: 19647905
[25] Baba, A.; Kawamura, N.; Makino, H.; Ohta, Y.; Taketomi, S.;
Sohda, T. Studies on disease-modifying antirheumatic drugs: Syn-
thesis of novel quinoline and quinazoline derivatives and their anti-
inflammatory effect. J. Med. Chem., 1996, 39(26), 5176-5182.
http://dx.doi.org/10.1021/jm9509408 PMID: 8978845
[26] Hu, Y.Q.; Gao, C.; Zhang, S.; Xu, L.; Xu, Z.; Feng, L.S.; Wu, X.;
Zhao, F. Quinoline hybrids and their antiplasmodial and antimalar-
ial activities. Eur. J. Med. Chem., 2017, 139, 22-47.
http://dx.doi.org/10.1016/j.ejmech.2017.07.061 PMID: 28800458
[27] Wang, H.; Fang, G.Q.; Wang, K.; Wu, Z.Y.; Yao, Q.Q. Determina-
tion of dopamine using 2-(4-boronophenyl)quinoline-4-carboxylic
acids as fluorescent probes. Anal. Lett., 2019, 52(4), 713-727.
http://dx.doi.org/10.1080/00032719.2018.1488258
[28] Martirosyan, A.R.; Rahim-Bata, R.; Freeman, A.B.; Clarke, C.D.;
Howard, R.L.; Strobl, J.S. Differentiation-inducing quinolines as
experimental breast cancer agents in the MCF-7 human breast can-
cer cell model. Biochem. Pharmacol., 2004, 68(9), 1729-1738.
http://dx.doi.org/10.1016/j.bcp.2004.05.003 PMID: 15450938
[29] Yang, Y.; Shi, L.; Zhou, Y.; Li, H.Q.; Zhu, Z.W.; Zhu, H.L. De-
sign, synthesis and biological evaluation of quinoline amide deriva-
tives as novel VEGFR-2 inhibitors. Bioorg. Med. Chem. Lett.,
2010, 20(22), 6653-6656.
http://dx.doi.org/10.1016/j.bmcl.2010.09.014 PMID: 20943391
[30] Tsai, C.C.; Liu, H.F.; Hsu, K.C.; Yang, J.M.; Chen, C.; Liu, K.K.;
Hsu, T.S.; Chao, J.I. 7-Chloro-6-piperidin-1-yl-quinoline-5,8-dione
(PT-262), a novel ROCK inhibitor blocks cytoskeleton function
and cell migration. Biochem. Pharmacol., 2011, 81(7), 856-865.
http://dx.doi.org/10.1016/j.bcp.2011.01.009 PMID: 21276421
[31] El-Sonbati, A.Z.; Diab, M.A.; Mohamed, G.G.; Saad, M.A.;
Morgan, S.M.; El-Sawy, S.E.A. Polymer complexes. LXXVII.
Synthesis, characterization, spectroscopic studies and immune re-
sponse in cattle of quinoline polymer complexes. Appl. Organomet.
Chem., 2019, 33(8), e4973.
http://dx.doi.org/10.1002/aoc.4973
[32] Lemmon, M.A.; Schlessinger, J. Cell signaling by receptor tyrosine
kinases. Cell, 2010, 141(7), 1117-1134.
http://dx.doi.org/10.1016/j.cell.2010.06.011 PMID: 20602996
[33] Normanno, N.; De Luca, A.; Bianco, C.; Strizzi, L.; Mancino, M.;
Maiello, M.R.; Carotenuto, A.; De Feo, G.; Caponigro, F.; Salo-
mon, D.S. Epidermal Growth Factor Receptor (EGFR) signaling in
cancer. Gene, 2006, 366(1), 2-16.
http://dx.doi.org/10.1016/j.gene.2005.10.018 PMID: 16377102
[34] Andrae, J.; Gallini, R.; Betsholtz, C. Role of platelet-derived
growth factors in physiology and medicine. Genes Dev., 2008,
22(10), 1276-1312.
http://dx.doi.org/10.1101/gad.1653708 PMID: 18483217
[35] Ferrara, N. Vascular endothelial growth factor: Basic science and
clinical progress. Endocr. Rev., 2004, 25(4), 581-611.
http://dx.doi.org/10.1210/er.2003-0027 PMID: 15294883
[36] Turner, N.; Grose, R. Fibroblast growth factor signalling: From
development to cancer. Nat. Rev. Cancer, 2010, 10(2), 116-129.
834 Anti-Cancer Agents in Medicinal C hemistry, 2021, Vol. 21, No. 7 Man et al.
http://dx.doi.org/10.1038/nrc2780 PMID: 20094046
[37] Carter, B.D.; Kaltschmidt, C.; Kaltschmidt, B.; Offenhäuser, N.;
Böhm-Matthaei, R.; Baeuerle, P.A.; Barde, Y.A. Selective activa-
tion of NF-κB by nerve growth factor through the neurotrophin re-
ceptor p75. Science, 1996, 272(5261), 542-545.
http://dx.doi.org/10.1126/science.272.5261.542 PMID: 8614802
[38] Pollak, M. The insulin and insulin-like growth factor receptor fam-
ily in neoplasia: An update. Nat. Rev. Cancer, 2012, 12(3), 159-
169.
http://dx.doi.org/10.1038/nrc3215 PMID: 22337149
[39] Pasquale, E.B. Eph receptors and ephrins in cancer: Bidirectional
signalling and beyond. Nat. Rev. Cancer, 2010, 10(3), 165-180.
http://dx.doi.org/10.1038/nrc2806 PMID: 20179713
[40] Fagiani, E.; Christofori, G. Angiopoietins in angiogenesis. Cancer
Lett., 2013, 328(1), 18-26.
http://dx.doi.org/10.1016/j.canlet.2012.08.018 PMID: 22922303
[41] Li, E.; Hristova, K. Role of receptor tyrosine kinase transmembrane
domains in cell signaling and human pathologies. Biochemistry,
2006, 45(20), 6241-6251.
http://dx.doi.org/10.1021/bi060609y PMID: 16700535
[42] Gschwind, A.; Fischer, O.M.; Ullrich, A. The discovery of receptor
tyrosine kinases: Targets for cancer therapy. Nat. Rev. Cancer,
2004, 4(5), 361-370.
http://dx.doi.org/10.1038/nrc1360 PMID: 15122207
[43] Ostman, A.; Böhmer, F.D. Regulation of receptor tyrosine kinase
signaling by protein tyrosine phosphatases. Trends Cell Biol., 2001,
11(6), 258-266.
http://dx.doi.org/10.1016/S0962-8924(01)01990-0 PMID:
11356362
[44] Hubbard, S.R.; Miller, W.T. Receptor tyrosine kinases: Mecha-
nisms of activation and signaling. Curr. Opin. Cell Biol., 2007,
19(2), 117-123.
http://dx.doi.org/10.1016/j.ceb.2007.02.010 PMID: 17306972
[45] Reid, A.; Vidal, L.; Shaw, H.; de Bono, J. Dual inhibition of ErbB1
(EGFR/HER1) and ErbB2 (HER2/neu). Eur. J. Cancer, 2007,
43(3), 481-489.
http://dx.doi.org/10.1016/j.ejca.2006.11.007 PMID: 17208435
[46] Wang, S.E.; Narasanna, A.; Perez-Torres, M.; Xiang, B.; Wu, F.Y.;
Yang, S.; Carpenter, G.; Gazdar, A.F.; Muthuswamy, S.K.;
Arteaga, C.L. HER2 kinase domain mutation results in constitutive
phosphorylation and activation of HER2 and EGFR and resistance
to EGFR tyrosine kinase inhibitors. Cancer Cell, 2006, 10(1), 25-
38.
http://dx.doi.org/10.1016/j.ccr.2006.05.023 PMID: 16843263
[47] Johnson, G.L.; Lapadat, R. Mitogen-activated protein kinase path-
ways mediated by ERK, JNK, and p38 protein kinases. Science,
2002, 298(5600), 1911-1912.
http://dx.doi.org/10.1126/science.1072682 PMID: 12471242
[48] Liu, P.; Cheng, H.; Roberts, T.M.; Zhao, J.J. Targeting the phos-
phoinositide 3-kinase pathway in cancer. Nat. Rev. Drug Discov.,
2009, 8(8), 627-644.
http://dx.doi.org/10.1038/nrd2926 PMID: 19644473
[49] Sun, Y.; Liu, W.Z.; Liu, T.; Feng, X.; Yang, N.; Zhou, H.F. Signal-
ing pathway of MAPK/ERK in cell proliferation, differentiation,
migration, senescence and apoptosis. J. Recept. Signal Transduct.
Res., 2015, 35(6), 600-604.
http://dx.doi.org/10.3109/10799893.2015.1030412 PMID:
26096166
[50] English, J.M.; Cobb, M.H. Pharmacological inhibitors of MAPK
pathways. Trends Pharmacol. Sci., 2002, 23(1), 40-45.
http://dx.doi.org/10.1016/S0165-6147(00)01865-4 PMID:
11804650
[51] Vanhaesebroeck, B.; Leevers, S.J.; Panayotou, G.; Waterfield,
M.D. Phosphoinositide 3-kinases: A conserved family of signal
transducers. Trends Biochem. Sci., 1997, 22(7), 267-272.
http://dx.doi.org/10.1016/S0968-0004(97)01061-X PMID:
9255069
[52] Lacouture, M.E. Mechanisms of cutaneous toxicities to EGFR
inhibitors. Nat. Rev. Cancer, 2006, 6(10), 803-812.
http://dx.doi.org/10.1038/nrc1970 PMID: 16990857
[53] Chong, C.R.; Jänne, P.A. The quest to overcome resistance to
EGFR-targeted therapies in cancer. Nat. Med., 2013, 19(11), 1389-
1400.
http://dx.doi.org/10.1038/nm.3388 PMID: 24202392
[54] Jänne, P.A.; Yang, J.C.H.; Kim, D.W.; Planchard, D.; Ohe, Y.;
Ramalingam, S.S.; Ahn, M.J.; Kim, S.W.; Su, W.C.; Horn, L.;
Haggstrom, D.; Felip, E.; Kim, J.H.; Frewer, P.; Cantarini, M.;
Brown, K.H.; Dickinson, P.A.; Ghiorghiu, S.; Ranson, M.
AZD9291 in EGFR inhibitor-resistant non-small-cell lung cancer.
N. Engl. J. Med., 2015, 372(18), 1689-1699.
http://dx.doi.org/10.1056/NEJMoa1411817 PMID: 25923549
[55] Hovinga, K.E.; McCrea, H.J.; Brennan, C.; Huse, J.; Zheng, J.;
Esquenazi, Y.; Panageas, K.S.; Tabar, V. EGFR amplification and
classical subtype are associated with a poor response to bevacizu-
mab in recurrent glioblastoma. J. Neurooncol., 2019, 142(2), 337-
345.
http://dx.doi.org/10.1007/s11060-019-03102-5 PMID: 30680510
[56] Cho, H.S.; Mason, K.; Ramyar, K.X.; Stanley, A.M.; Gabelli, S.B.;
Denney, D.W., Jr; Leahy, D.J. Structure of the extracellular region
of HER2 alone and in complex with the Herceptin Fab. Nature,
2003, 421(6924), 756-760.
http://dx.doi.org/10.1038/nature01392 PMID: 12610629
[57] Verma, S.; Miles, D.; Gianni, L.; Krop, I.E.; Welslau, M.; Baselga,
J.; Pegram, M.; Oh, D.Y.; Diéras, V.; Guardino, E.; Fang, L.; Lu,
M.W.; Olsen, S.; Blackwell, K. EMILIA Study Group. Trastuzu-
mab emtansine for HER2-positive advanced breast cancer. N. Engl.
J. Med., 2012, 367(19), 1783-1791.
http://dx.doi.org/10.1056/NEJMoa1209124 PMID: 23020162
[58] Arteaga, C.L.; Sliwkowski, M.X.; Osborne, C.K.; Perez, E.A.;
Puglisi, F.; Gianni, L. Treatment of HER2-positive breast cancer:
Current status and future perspectives. Nat. Rev. Clin. Oncol.,
2011, 9(1), 16-32.
http://dx.doi.org/10.1038/nrclinonc.2011.177 PMID: 22124364
[59] Bublil, E.M.; Pines, G.; Patel, G.; Fruhwirth, G.; Ng, T.; Yarden,
Y. Kinase-mediated quasi-dimers of EGFR. FASEB J., 2010,
24(12), 4744-4755.
PMID: 20682838
[60] Arkhipov, A.; Shan, Y.; Kim, E.T.; Dror, R.O.; Shaw, D.E. Her2
activation mechanism reflects evolutionary preservation of asym-
metric ectodomain dimers in the human EGFR family. eLife, 2013,
2, e00708.
http://dx.doi.org/10.7554/eLife.00708 PMID: 23878723
[61] Xu, J.; Du, Y.; Liu, X.J.; Zhu, B.Y.; Zhang, S.H.; Li, L.; Li, Y.;
Wang, X.F.; Shan, C.K.; Wang, R.Q.; Zhen, Y.S. Recombinant
EGFR/MMP-2 bi-targeted fusion protein markedly binding to non-
small-cell lung carcinoma and exerting potent therapeutic efficacy.
Pharmacol. Res., 2017, 126, 66-76.
http://dx.doi.org/10.1016/j.phrs.2017.04.001 PMID: 28392461
[62] Izumi, Y.; Xu, L.; di Tomaso, E.; Fukumura, D.; Jain, R.K. Tumour
biology: Herceptin acts as an anti-angiogenic cocktail. Nature,
2002, 416(6878), 279-280.
http://dx.doi.org/10.1038/416279b PMID: 11907566
[63] Tsao, M.S.; Sakurada, A.; Cutz, J.C.; Zhu, C.Q.; Kamel-Reid, S.;
Squire, J.; Lorimer, I.; Zhang, T.; Liu, N.; Daneshmand, M.; Mar-
rano, P.; da Cunha Santos, G.; Lagarde, A.; Richardson, F.;
Seymour, L.; Whitehead, M.; Ding, K.; Pater, J.; Shepherd, F.A.
Erlotinib in lung cancer - molecular and clinical predictors of out-
come. N. Engl. J. Med., 2005, 353(2), 133-144.
http://dx.doi.org/10.1056/NEJMoa050736 PMID: 16014883
[64] Pao, W.; Miller, V.A.; Politi, K.A.; Riely, G.J.; Somwar, R.; Zak-
owski, M.F.; Kris, M.G.; Varmus, H. Acquired resistance of lung
adenocarcinomas to gefitinib or erlotinib is associated with a sec-
ond mutation in the EGFR kinase domain. PLoS Med., 2005, 2(3),
e73.
http://dx.doi.org/10.1371/journal.pmed.0020073 PMID: 15737014
[65] Kotecki, N.; Gombos, A.; Awada, A. Adjuvant therapeutic ap-
proaches of HER2-positive breast cancer with a focus on neratinib
maleate. Expert Rev. Anticancer Ther., 2019, 19(6), 447-454.
http://dx.doi.org/10.1080/14737140.2019.1613892 PMID:
31082272
[66] Burstein, H.J.; Sun, Y.; Dirix, L.Y.; Jiang, Z.; Paridaens, R.; Tan,
A.R.; Awada, A.; Ranade, A.; Jiao, S.; Schwartz, G.; Abbas, R.;
Powell, C.; Turnbull, K.; Vermette, J.; Zacharchuk, C.; Badwe, R.
Neratinib, an irreversible ErbB receptor tyrosine kinase inhibitor, in
patients with advanced ErbB2-positive breast cancer. J. Clin. On-
col., 2010, 28(8), 1301-1307.
http://dx.doi.org/10.1200/JCO.2009.25.8707 PMID: 20142587
[67] Blair, H.A. Pyrotinib: First global approval. Drugs, 2018, 78(16),
1751-1755.
http://dx.doi.org/10.1007/s40265-018-0997-0 PMID: 30341682
[68] Bryce, A.H.; Rao, R.; Sarkaria, J.; Reid, J.M.; Qi, Y.; Qin, R.;
James, C.D.; Jenkins, R.B.; Boni, J.; Erlichman, C.; Haluska, P.
Recent Progress in the Development of Quinoline Derivatives Anti-Cancer Agents in Medicinal Chemistry, 2021, Vol. 21, No. 7 835
Phase I study of temsirolimus in combination with EKB-569 in pa-
tients with advanced solid tumors. Invest. New Drugs, 2012, 30(5),
1934-1941.
http://dx.doi.org/10.1007/s10637-011-9742-1 PMID: 21881915
[69] Tammela, T.; Zarkada, G.; Wallgard, E.; Murtomäki, A.; Suchting,
S.; Wirzenius, M.; Waltari, M.; Hellström, M.; Schomber, T.; Pel-
tonen, R.; Freitas, C.; Duarte, A.; Isoniemi, H.; Laakkonen, P.;
Christofori, G.; Ylä-Herttuala, S.; Shibuya, M.; Pytowski, B.;
Eichmann, A.; Betsholtz, C.; Alitalo, K. Blocking VEGFR-3 sup-
presses angiogenic sprouting and vascular network formation. Na-
ture, 2008, 454(7204), 656-660.
http://dx.doi.org/10.1038/nature07083 PMID: 18594512
[70] Kiselyov, A.; Balakin, K.V.; Tkachenko, S.E. VEGF/VEGFR
signalling as a target for inhibiting angiogenesis. Expert Opin. In-
vestig. Drugs, 2007, 16(1), 83-107.
http://dx.doi.org/10.1517/13543784.16.1.83 PMID: 17155856
[71] Sitohy, B.; Nagy, J.A.; Dvorak, H.F. Anti-VEGF/VEGFR therapy
for cancer: Reassessing the target. Cancer Res., 2012, 72(8), 1909-
1914.
http://dx.doi.org/10.1158/0008-5472.CAN-11-3406 PMID:
22508695
[72] Bhargava, P.; Robinson, M.O. Development of second-generation
VEGFR tyrosine kinase inhibitors: Current status. Curr. Oncol.
Rep., 2011, 13(2), 103-111.
http://dx.doi.org/10.1007/s11912-011-0154-3 PMID: 21318618
[73] Dibb, N.J.; Dilworth, S.M.; Mol, C.D. Switching on kinases: On-
cogenic activation of BRAF and the PDGFR family. Nat. Rev.
Cancer, 2004, 4(9), 718-727.
http://dx.doi.org/10.1038/nrc1434 PMID: 15343278
[74] Board, R.; Jayson, G.C. Platelet-Derived Growth Factor Receptor
(PDGFR): A target for anticancer therapeutics. Drug Resist. Up-
dat., 2005, 8(1-2), 75-83.
http://dx.doi.org/10.1016/j.drup.2005.03.004 PMID: 15939344
[75] Jechlinger, M.; Sommer, A.; Moriggl, R.; Seither, P.; Kraut, N.;
Capodiecci, P.; Donovan, M.; Cordon-Cardo, C.; Beug, H.;
Grünert, S. Autocrine PDGFR signaling promotes mammary can-
cer metastasis. J. Clin. Invest., 2006, 116(6), 1561-1570.
http://dx.doi.org/10.1172/JCI24652 PMID: 16741576
[76] Worby, C.A.; Vega, Q.C.; Zhao, Y.; Chao, H.H.J.; Seasholtz, A.F.;
Dixon, J.E. Glial cell line-derived neurotrophic factor signals
through the RET receptor and activates mitogen-activated protein
kinase. J. Biol. Chem., 1996, 271(39), 23619-23622.
http://dx.doi.org/10.1074/jbc.271.39.23619 PMID: 8798576
[77] Webb, D.J.; Donais, K.; Whitmore, L.A.; Thomas, S.M.; Turner,
C.E.; Parsons, J.T.; Horwitz, A.F. FAK-Src signalling through pax-
illin, ERK and MLCK regulates adhesion disassembly. Nat. Cell
Biol., 2004, 6(2), 154-161.
http://dx.doi.org/10.1038/ncb1094 PMID: 14743221
[78] Syed, Y.Y. Anlotinib: First global approval. Drugs, 2018, 78(10),
1057-1062.
http://dx.doi.org/10.1007/s40265-018-0939-x PMID: 29943374
[79] Shen, G.; Zheng, F.; Ren, D.; Du, F.; Dong, Q.; Wang, Z.; Zhao,
F.; Ahmad, R.; Zhao, J. Anlotinib: A novel multi-targeting tyrosine
kinase inhibitor in clinical development. J. Hematol. Oncol., 2018,
11(1), 120.
http://dx.doi.org/10.1186/s13045-018-0664-7 PMID: 30231931
[80] Tahara, M.; Kiyota, N.; Yamazaki, T.; Chayahara, N.; Nakano, K.;
Inagaki, L.; Toda, K.; Enokida, T.; Minami, H.; Imamura, Y.; Sa-
saki, T.; Suzuki, T.; Fujino, K.; Dutcus, C.E.; Takahashi, S. Len-
vatinib for anaplastic thyroid cancer. Front. Oncol., 2017, 7, 25.
http://dx.doi.org/10.3389/fonc.2017.00025 PMID: 28299283
[81] Elisei, R.; Schlumberger, M.J.; Müller, S.P.; Schöffski, P.; Brose,
M.S.; Shah, M.H.; Licitra, L.; Jarzab, B.; Medvedev, V.; Kreissl,
M.C.; Niederle, B.; Cohen, E.E.W.; Wirth, L.J.; Ali, H.; Hessel, C.;
Yaron, Y.; Ball, D.; Nelkin, B.; Sherman, S.I. Cabozantinib in pro-
gressive medullary thyroid cancer. J. Clin. Oncol., 2013, 31(29),
3639-3646.
http://dx.doi.org/10.1200/JCO.2012.48.4659 PMID: 24002501
[82] Cabanillas, M.E.; Habra, M.A. Lenvatinib: Role in thyroid cancer
and other solid tumors. Cancer Treat. Rev., 2016, 42, 47-55.
http://dx.doi.org/10.1016/j.ctrv.2015.11.003 PMID: 26678514
[83] Yakes, F.M.; Chen, J.; Tan, J.; Yamaguchi, K.; Shi, Y.; Yu, P.;
Qian, F.; Chu, F.; Bentzien, F.; Cancilla, B.; Orf, J.; You, A.; Laird,
A.D.; Engst, S.; Lee, L.; Lesch, J.; Chou, Y.C.; Joly, A.H. Cabo-
zantinib (XL184), a novel MET and VEGFR2 inhibitor, simultane-
ously suppresses metastasis, angiogenesis, and tumor growth. Mol.
Cancer Ther., 2011, 10(12), 2298-2308.
http://dx.doi.org/10.1158/1535-7163.MCT-11-0264 PMID:
21926191
[84] Simiczyjew, A.; Dratkiewicz, E.; Van Troys, M.; Ampe, C.;
Styczeń, I.; Nowak, D. Combination of EGFR inhibitor Lapatinib
and MET inhibitor Foretinib inhibits migration of triple negative
breast cancer cell lines. Cancers (Basel), 2018, 10(9), 335.
http://dx.doi.org/10.3390/cancers10090335 PMID: 30227653
[85] Gras, J. Lucitanib hydrochloride dual FGFR/VEGFR inhibitor
treatment of NSCLC and breast cancer. Drugs Future, 2015, 40(8),
509-521.
[86] Tomillero, A.; Moral, M.A. Gateways to clinical trials. Method
Find. Exp. Clin., 2010, 32(7), 518-548.
http://dx.doi.org/10.1358/mf.2010.32.7.1549223
[87] Xi, N.; Zhang, Y.J.; Wang, Z.H.; Wu, Y.J.; Wang, T.J.
CT053PTSA, a novel c-MET and VEGFR2 inhibitor, potently sup-
presses angiogenesis and tumor growth. Cancer Res., 2014, 74(19),
Meeting Abstract 1755.
[88] Doi, T.; Matsubara, N.; Kawai, A.; Naka, N.; Takahashi, S.; Ue-
mura, H.; Yamamoto, N. Phase I study of TAS-115, a novel oral
multi-kinase inhibitor, in patients with advanced solid tumors. In-
vest. New Drug, 2020, 38(4), 1175-1185.
[89] Macpherson, I.R.; Poondru, S.; Simon, G.R.; Gedrich, R.; Brock,
K.; Hopkins, C.A.; Stewart, K.; Stephens, A.; Evans, T.R.J. A
phase 1 study of OSI-930 in combination with erlotinib in patients
with advanced solid tumours. Eur. J. Cancer, 2013, 49(4), 782-789.
http://dx.doi.org/10.1016/j.ejca.2012.09.036 PMID: 23099006
[90] Gherardi, E.; Birchmeier, W.; Birchmeier, C.; Vande Woude, G.
Targeting MET in cancer: Rationale and progress. Nat. Rev. Can-
cer, 2012, 12(2), 89-103.
http://dx.doi.org/10.1038/nrc3205 PMID: 22270953
[91] Cui, J.J.; Tran-Dubé, M.; Shen, H.; Nambu, M.; Kung, P.P.; Pair-
ish, M.; Jia, L.; Meng, J.; Funk, L.; Botrous, I.; McTigue, M.;
Grodsky, N.; Ryan, K.; Padrique, E.; Alton, G.; Timofeevski, S.;
Yamazaki, S.; Li, Q.; Zou, H.; Christensen, J.; Mroczkowski, B.;
Bender, S.; Kania, R.S.; Edwards, M.P. Structure based drug de-
sign of crizotinib (PF-02341066), a potent and selective dual in-
hibitor of Mesenchymal-Epithelial Transition factor (c-MET)
kinase and Anaplastic Lymphoma Kinase (ALK). J. Med. Chem.,
2011, 54(18), 6342-6363.
http://dx.doi.org/10.1021/jm2007613 PMID: 21812414
[92] Baltschukat, S.; Engstler, B.S.; Huang, A.; Hao, H.X.; Tam, A.;
Wang, H.Q.; Liang, J.; DiMare, M.T.; Bhang, H.C.; Wang, Y.;
Furet, P.; Sellers, W.R.; Hofmann, F.; Schoepfer, J.; Tiedt, R.
Capmatinib (INC280) is active against models of non-small cell
lung cancer and other cancer types with defined mechanisms of
MET activation. Clin. Cancer Res., 2019, 25(10), 3164-3175.
http://dx.doi.org/10.1158/1078-0432.CCR-18-2814 PMID:
30674502
[93] Lolkema, M.P.; Bohets, H.H.; Arkenau, H.T.; Lampo, A.; Barale,
E.; de Jonge, M.J.A.; van Doorn, L.; Hellemans, P.; de Bono, J.S.;
Eskens, F.A.L.M. The c-Met tyrosine kinase inhibitor JNJ-
38877605 causes renal toxicity through species-specific insoluble
metabolite formation. Clin. Cancer Res., 2015, 21(10), 2297-2304.
http://dx.doi.org/10.1158/1078-0432.CCR-14-3258 PMID:
25745036
[94] Pennacchietti, S.; Cazzanti, M.; Bertotti, A.; Rideout, W.M., III;
Han, M.; Gyuris, J.; Perera, T.; Comoglio, P.M.; Trusolino, L.; Mi-
chieli, P. Microenvironment-derived HGF overcomes genetically
determined sensitivity to anti-MET drugs. Cancer Res., 2014,
74(22), 6598-6609.
http://dx.doi.org/10.1158/0008-5472.CAN-14-0761 PMID:
25217525
[95] Cui, J.J.; McTigue, M.; Nambu, M.; Tran-Dubé, M.; Pairish, M.;
Shen, H.; Jia, L.; Cheng, H.; Hoffman, J.; Le, P.; Jalaie, M.; Goetz,
G.H.; Ryan, K.; Grodsky, N.; Deng, Y.L.; Parker, M.; Timofeevski,
S.; Murray, B.W.; Yamazaki, S.; Aguirre, S.; Li, Q.; Zou, H.;
Christensen, J. Discovery of a novel class of exquisitely selective
Mesenchymal-Epithelial Transition factor (c-MET) protein kinase
inhibitors and identification of the clinical candidate 2-(4-(1-
(quinolin-6-ylmethyl)-1H-[1,2,3]triazolo[4,5-b]pyrazin-6-yl)-1H-
pyrazol-1-yl)ethanol (PF-04217903) for the treatment of cancer. J.
Med. Chem., 2012, 55(18), 8091-8109.
http://dx.doi.org/10.1021/jm300967g PMID: 22924734
836 Anti-Cancer Agents in Medicinal C hemistry, 2021, Vol. 21, No. 7 Man et al.
[96] Chessari, G.; Woodhead, A.J. From fragment to clinical candidate--
a historical perspective. Drug Discov. Today, 2009, 14(13-14),
668-675.
http://dx.doi.org/10.1016/j.drudis.2009.04.007 PMID: 19427404
[97] Hong, D.S.; Rosen, P.; Lockhart, A.C.; Fu, S.; Janku, F.; Kurzrock,
R.; Khan, R.; Amore, B.; Caudillo, I.; Deng, H.; Hwang, Y.C.;
Loberg, R.; Ngarmchamnanrith, G.; Beaupre, D.M.; Lee, P. A first-
in-human study of AMG 208, an oral MET inhibitor, in adult pa-
tients with advanced solid tumors. Oncotarget, 2015, 6(21), 18693-
18706.
http://dx.doi.org/10.18632/oncotarget.4472 PMID: 26155941
[98] Hennessy, B.T.; Smith, D.L.; Ram, P.T.; Lu, Y.; Mills, G.B. Ex-
ploiting the PI3K/AKT pathway for cancer drug discovery. Nat.
Rev. Drug Discov., 2005, 4(12), 988-1004.
http://dx.doi.org/10.1038/nrd1902 PMID: 16341064
[99] DeBerardinis, R.J.; Lum, J.J.; Hatzivassiliou, G.; Thompson, C.B.
The biology of cancer: Metabolic reprogramming fuels cell growth
and proliferation. Cell Metab., 2008, 7(1), 11-20.
http://dx.doi.org/10.1016/j.cmet.2007.10.002 PMID: 18177721
[100] Vanhaesebroeck, B.; Guillermet-Guibert, J.; Graupera, M.; Bi-
langes, B. The emerging mechanisms of isoform-specific PI3K sig-
nalling. Nat. Rev. Mol . Cell Biol ., 2010, 11(5), 329-341.
http://dx.doi.org/10.1038/nrm2882 PMID: 20379207
[101] Serra, V.; Markman, B.; Scaltriti, M.; Eichhorn, P.J.; Valero, V.;
Guzman, M.; Botero, M.L.; Llonch, E.; Atzori, F.; Di Cosimo, S.;
Maira, M.; Garcia-Echeverria, C.; Parra, J.L.; Arribas, J.; Baselga,
J. NVP-BEZ235, a dual PI3K/mTOR inhibitor, prevents PI3K sig-
naling and inhibits the growth of cancer cells with activating PI3K
mutations. Cancer Res., 2008, 68(19), 8022-8030.
http://dx.doi.org/10.1158/0008-5472.CAN-08-1385 PMID:
18829560
[102] Maira, S.M.; Stauffer, F.; Brueggen, J.; Furet, P.; Schnell, C.;
Fritsch, C.; Brachmann, S.; Chène, P.; De Pover, A.; Schoemaker,
K.; Fabbro, D.; Gabriel, D.; Simonen, M.; Murphy, L.; Finan, P.;
Sellers, W.; García-Echeverría, C. Identification and characteriza-
tion of NVP-BEZ235, a new orally available dual phosphatidyli-
nositol 3-kinase/mammalian target of rapamycin inhibitor with po-
tent in vivo antitumor activity. Mol. Cancer Ther., 2008, 7(7),
1851-1863.
http://dx.doi.org/10.1158/1535-7163.MCT-08-0017 PMID:
18606717
[103] Williams, R. Discontinued drugs in 2012: Oncology drugs. Expert
Opin. Investig. Drugs, 2013, 22(12), 1627-1644.
http://dx.doi.org/10.1517/13543784.2013.847088 PMID: 24102323
[104] Ding, Y.; Liu, J.G.; Calley, J.N.; Qian, H.R.; Iversen, P.W.; Ebert,
P.J.; Beckmann, R.P.; Donoho, G.P.; Martinez, R.; Wu, W.J.; Lin,
A.B.; Bowden, E.; Aggarwal, A. PI3K/AKT signaling pathway is
transcriptionally elevated in prexasertib-resistant TNBC PDX mod-
els. Cancer Res., 2018, 78(13), Meeting Abstract 2586.
[105] Jalota-Badhwar, A.; Bhatia, D.R.; Boreddy, S.; Joshi, A.; Venka-
traman, M.; Desai, N.; Chaudhari, S.; Bose, J.; Kolla, L.S.; Deore,
V.; Yewalkar, N.; Kumar, S.; Sharma, R.; Damre, A.; More, A.;
Sharma, S.; Agarwal, V.R. P7170: A novel molecule with unique
profile of mTORC1/C2 and activin receptor-like kinase 1 inhibition
leading to antitumor and antiangiogenic activity. Mol. Cancer
Ther., 2015, 14(5), 1095-1106.
http://dx.doi.org/10.1158/1535-7163.MCT-14-0486 PMID:
25700704
[106] Basu, D.; Salgado, C.M.; Bauer, B.; Khakoo, Y.; Patel, J.R.; Hoehl,
R.M.; Bertolini, D.M.; Zabec, J.; Brzozowski, M.R.; Reyes-
Múgica, M. The dual PI3K/mToR inhibitor Omipal-
isib/GSK2126458 inhibits clonogenic growth in oncogenically-
transformed cells from neurocutaneous melanocytosis. Cancer Ge-
nom. Proteom., 2018, 15(4), 239-248.
http://dx.doi.org/10.21873/cgp.20082 PMID: 29976629
[107] Braz, J.C.; Gill, R.M.; Corbly, A.K.; Jones, B.D.; Jin, N.; Vlahos,
C.J.; Wu, Q.; Shen, W. Selective activation of
PI3Kalpha/Akt/GSK-3β signalling and cardiac compensatory hy-
pertrophy during recovery from heart failure. Eur. J. Heart Fail.,
2009, 11(8), 739-748.
http://dx.doi.org/10.1093/eurjhf/hfp094 PMID: 19633101
[108] Thomas, S.M.; Brugge, J.S. Cellular functions regulated by Src
family kinases. Annu. Rev. Cell Dev. Biol., 1997, 13, 513-609.
http://dx.doi.org/10.1146/annurev.cellbio.13.1.513 PMID: 9442882
[109] Fabian, M.A.; Biggs, W.H., III; Treiber, D.K.; Atteridge, C.E.;
Azimioara, M.D.; Benedetti, M.G.; Carter, T.A.; Ciceri, P.; Edeen,
P.T.; Floyd, M.; Ford, J.M.; Galvin, M.; Gerlach, J.L.; Grotzfeld,
R.M.; Herrgard, S.; Insko, D.E.; Insko, M.A.; Lai, A.G.; Lélias,
J.M.; Mehta, S.A.; Milanov, Z.V.; Velasco, A.M.; Wodicka, L.M.;
Patel, H.K.; Zarrinkar, P.P.; Lockhart, D.J. A small molecule-
kinase interaction map for clinical kinase inhibitors. Nat. Biotech-
nol., 2005, 23(3), 329-336.
http://dx.doi.org/10.1038/nbt1068 PMID: 15711537
[110] Demetri, G.D.; von Mehren, M.; Blanke, C.D.; Van den Abbeele,
A.D.; Eisenberg, B.; Roberts, P.J.; Heinrich, M.C.; Tuveson, D.A.;
Singer, S.; Janicek, M.; Fletcher, J.A.; Silverman, S.G.; Silberman,
S.L.; Capdeville, R.; Kiese, B.; Peng, B.; Dimitrijevic, S.; Druker,
B.J.; Corless, C.; Fletcher, C.D.M.; Joensuu, H. Efficacy and safety
of imatinib mesylate in advanced gastrointestinal stromal tumors.
N. Engl. J. Med., 2002, 347(7), 472-480.
http://dx.doi.org/10.1056/NEJMoa020461 PMID: 12181401
[111] Kantarjian, H.; Giles, F.; Wunderle, L.; Bhalla, K.; O’Brien, S.;
Wassmann, B.; Tanaka, C.; Manley, P.; Rae, P.; Mietlowski, W.;
Bochinski, K.; Hochhaus, A.; Griffin, J.D.; Hoelzer, D.; Albitar,
M.; Dugan, M.; Cortes, J.; Alland, L.; Ottmann, O.G. Nilotinib in
imatinib-resistant CML and Philadelphia chromosome-positive
ALL. N. Engl. J. Med., 2006, 354(24), 2542-2551.
http://dx.doi.org/10.1056/NEJMoa055104 PMID: 16775235
[112] Talpaz, M.; Shah, N.P.; Kantarjian, H.; Donato, N.; Nicoll, J.;
Paquette, R.; Cortes, J.; O’Brien, S.; Nicaise, C.; Bleickardt, E.;
Blackwood-Chirchir, M.A.; Iyer, V.; Chen, T.T.; Huang, F.; Decil-
lis, A.P.; Sawyers, C.L. Dasatinib in imatinib-resistant Philadelphia
chromosome-positive leukemias. N. Engl. J. Med., 2006, 354(24),
2531-2541.
http://dx.doi.org/10.1056/NEJMoa055229 PMID: 16775234
[113] Khoury, H.J.; Cortes, J.E.; Kantarjian, H.M.; Gambacorti-Passerini,
C.; Baccarani, M.; Kim, D.W.; Zaritskey, A.; Countouriotis, A.;
Besson, N.; Leip, E.; Kelly, V.; Brümmendorf, T.H. Bosutinib is
active in chronic phase chronic myeloid leukemia after imatinib
and dasatinib and/or nilotinib therapy failure. Blood, 2012, 119(15),
3403-3412.
http://dx.doi.org/10.1182/blood-2011-11-390120 PMID: 22371878
[114] Abad, E.; Graifer, D.; Lyakhovich, A. DNA damage response and
resistance of cancer stem cells. Cancer Lett., 2020, 474, 106-117.
http://dx.doi.org/10.1016/j.canlet.2020.01.008 PMID: 31968219
[115] Dasari, S.; Tchounwou, P.B. Cisplatin in cancer therapy: Molecular
mechanisms of action. Eur. J. Pharmacol., 2014, 740, 364-378.
http://dx.doi.org/10.1016/j.ejphar.2014.07.025 PMID: 25058905
[116] Rabik, C.A.; Dolan, M.E. Molecular mechanisms of resistance and
toxicity associated with platinating agents. Cancer Treat. Rev.,
2007, 33(1), 9-23.
http://dx.doi.org/10.1016/j.ctrv.2006.09.006 PMID: 17084534
[117] Champoux, J.J. DNA topoisomerases: Structure, function, and
mechanism. Annu. Rev. Biochem., 2001, 70, 369-413.
http://dx.doi.org/10.1146/annurev.biochem.70.1.369 PMID:
11395412
[118] Wang, J.C. Cellular roles of DNA topoisomerases: A molecular
perspective. Nat. Rev. Mol. Cell Biol., 2002, 3(6), 430-440.
http://dx.doi.org/10.1038/nrm831 PMID: 12042765
[119] Banerjee, S.; Veale, E.B.; Phelan, C.M.; Murphy, S.A.; Tocci,
G.M.; Gillespie, L.J.; Frimannsson, D.O.; Kelly, J.M.;
Gunnlaugsson, T. Recent advances in the development of 1,8-
naphthalimide based DNA targeting binders, anticancer and fluo-
rescent cellular imaging agents. Chem. Soc. Rev., 2013, 42(4),
1601-1618.
http://dx.doi.org/10.1039/c2cs35467e PMID: 23325367
[120] Pommier, Y. Topoisomerase I inhibitors: Camptothecins and be-
yond. Nat. Rev. Cancer, 2006, 6(10), 789-802.
http://dx.doi.org/10.1038/nrc1977 PMID: 16990856
[121] Lee, D.H.; Kim, S.W.; Suh, C.; Lee, J.S.; Lee, J.H.; Lee, S.J.;
Ryoo, B.Y.; Park, K.; Kim, J.S.; Heo, D.S.; Kim, N.K. Belotecan,
new camptothecin analogue, is active in patients with small-cell
lung cancer: results of a multicenter early phase II study. Ann. On-
col., 2008, 19(1), 123-127.
http://dx.doi.org/10.1093/annonc/mdm437 PMID: 17823384
[122] Vredenburgh, J.J.; Desjardins, A.; Herndon, J.E., II; Dowell, J.M.;
Reardon, D.A.; Quinn, J.A.; Rich, J.N.; Sathornsumetee, S.; Gu-
rurangan, S.; Wagner, M.; Bigner, D.D.; Friedman, A.H.; Fried-
man, H.S. Phase II trial of bevacizumab and irinotecan in recurrent
malignant glioma. Clin. Cancer Res., 2007, 13(4), 1253-1259.
http://dx.doi.org/10.1158/1078-0432.CCR-06-2309 PMID:
17317837
Recent Progress in the Development of Quinoline Derivatives Anti-Cancer Agents in Med icinal C hemistry, 2021, Vol. 21, No. 7 837
[123] Ketron, A.C.; Denny, W.A.; Graves, D.E.; Osheroff, N. Amsacrine
as a topoisomerase II poison: Importance of drug-DNA interac-
tions. Biochemistry, 2012, 51(8), 1730-1739.
http://dx.doi.org/10.1021/bi201159b PMID: 22304499
[124] Clark, J.W. Rubitecan. Expert Opin. Investig. Drugs, 2006, 15(1),
71-79.
http://dx.doi.org/10.1517/13543784.15.1.71 PMID: 16370935
[125] Pond, C.D.; Marshall, K.M.; Barrows, L.R. Identification of a small
topoisomerase I-binding peptide that has synergistic antitumor ac-
tivity with 9-aminocamptothecin. Mol. Cancer Ther., 2006, 5(3),
739-745.
http://dx.doi.org/10.1158/1535-7163.MCT-05-0377 PMID:
16546989
[126] Munster, P.N.; Daud, A.I. Preclinical and clinical activity of the
topoisomerase I inhibitor, karenitecin, in melanoma. Expert Opin.
Investig. Drugs, 2011, 20(11), 1565-1574.
http://dx.doi.org/10.1517/13543784.2011.617740 PMID: 21985236
[127] Tsakalozou, E.; Howard, D.; Leggas, M. In vitro and ex vivo anti-
leukemia activity of AR-67, a novel lipophilic camptothecin. Can-
cer Res., 2011, 71(8), Meeting Abstract 3547.
http://dx.doi.org/10.1158/1538-7445.AM2011-3547
[128] Chatterjee, A.; Digumarti, R.; Katneni, K.; Upreti, V.V.; Mamidi,
R.N.; Mullangi, R.; Surath, A.; Srinivas, M.L.; Uppalapati, S.; Ji-
watani, S.; Srinivas, N.R. Safety, tolerability, and pharmacokinetics
of a capsule formulation of DRF-1042, a novel camptothecin ana-
log, in refractory cancer patients in a bridging phase I study. J.
Clin. Pharmacol., 2005, 45(4), 453-460.
http://dx.doi.org/10.1177/0091270004270225 PMID: 15778426
[129] Giles, F.J.; Tallman, M.S.; Garcia-Manero, G.; Cortes, J.E.; Tho-
mas, D.A.; Wierda, W.G.; Verstovsek, S.; Hamilton, M.; Barrett,
E.; Albitar, M.; Kantarjian, H.M. Phase I and pharmacokinetic
study of a low-clearance, unilamellar liposomal formulation of lur-
totecan, a topoisomerase 1 inhibitor, in patients with advanced leu-
kemia. Cancer, 2004, 100(7), 1449-1458.
http://dx.doi.org/10.1002/cncr.20132 PMID: 15042679
[130] Zou, J.; Li, S.; Chen, Z.; Lu, Z.; Gao, J.; Zou, J.; Lin, X.; Li, Y.;
Zhang, C.; Shen, L. A novel oral camptothecin analog, gimatecan,
exhibits superior antitumor efficacy than irinotecan toward eso-
phageal squamous cell carcinoma in vitro and in vivo. Cell Death
Dis., 2018, 9(6), 661.
http://dx.doi.org/10.1038/s41419-018-0700-0 PMID: 29855512
[131] Propper, D.; Jones, K.; Anthoney, D.A.; Mansoor, W.; Ford, D.;
Eatock, M.; Agarwal, R.; Inatani, M.; Saito, T.; Abe, M.; Evans,
T.R.J. Phase II study of TP300 in patients with advanced gastric or
gastro-oesophageal junction adenocarcinoma. BMC Cancer, 2016,
16(1), 779.
http://dx.doi.org/10.1186/s12885-016-2828-6 PMID: 27724887
[132] Joerger, M.; Hess, D.; Delmonte, A.; Gallerani, E.; Fasolo, A.;
Gianni, L.; Cresta, S.; Barbieri, P.; Pace, S.; Sessa, C. Integrative
population pharmacokinetic and pharmacodynamic dose finding
approach of the new camptothecin compound namitecan (ST1968).
Br. J. Clin. Pharmacol., 2015, 80(1), 128-138.
http://dx.doi.org/10.1111/bcp.12583 PMID: 25580946
[133] Hu, Z.; Sun, Y.; Du, F.; Niu, W.; Xu, F.; Huang, Y.; Li, C. Accu-
rate determination of the anticancer prodrug simmitecan and its ac-
tive metabolite chimmitecan in various plasma samples based on
immediate deactivation of blood carboxylesterases. J. Chromatogr.
A, 2011, 1218(38), 6646-6653.
http://dx.doi.org/10.1016/j.chroma.2011.07.042 PMID: 21839460
[134] Zhao, Z.Y.; Xie, X.J.; Li, W.H.; Liu, J.; Chen, Z.; Zhang, B.; Li, T.;
Li, S.L.; Lu, J.G.; Zhang, L.R.; Zhang, L.H.; Xu, Z.S.; Lee, H.C.;
Zhao, Y.J. A cell-permeant mimetic of NMN activates SARM1 to
produce cyclic ADP-Ribose and induce non-apoptotic cell death.
Science, 2019, 15, 452-466. http://dx.doi.org/10.1016/j.isci.2019.05.001
[135] Liehr, J.G.; Harris, N.J.; Mendoza, J.; Ahmed, A.E.; Giovanella,
B.C. Pharmacology of camptothecin esters. Ann. N.Y. Acad. Sci.,
2000, 922, 216-223.
http://dx.doi.org/10.1111/j.1749-6632.2000.tb07040.x PMID:
11193897
[136] Kurtzberg, L.S.; Roth, S.; Krumbholz, R.; Crawford, J.; Bormann,
C.; Dunham, S.; Yao, M.; Rouleau, C.; Bagley, R.G.; Yu, X.J.;
Wang, F.; Schmid, S.M.; Lavoie, E.J.; Teicher, B.A. Genz-644282,
a novel non-camptothecin topoisomerase I inhibitor for cancer
treatment. Clin. Cancer Res., 2011, 17(9), 2777-2787.
http://dx.doi.org/10.1158/1078-0432.CCR-10-0542 PMID:
21415217
[137] Kroep, J.R.; Gelderblom, H. Diflomotecan, a promising homo-
camptothecin for cancer therapy. Expert Opin. Investig. Drugs,
2009, 18(1), 69-75.
http://dx.doi.org/10.1517/13543780802571674 PMID: 19053883
[138] Ghamande, S.; Lin, C.C.; Cho, D.C.; Shapiro, G.I.; Kwak, E.L.;
Silverman, M.H.; Tseng, Y.; Kuo, M.W.; Mach, W.B.; Hsu, S.C.;
Coleman, T.; Yang, J.C.H.; Cheng, A.L.; Ghalib, M.H.; Chuad-
hary, I.; Goel, S. A phase 1 open-label, sequential dose-escalation
study investigating the safety, tolerability, and pharmacokinetics of
intravenous TLC388 administered to patients with advanced solid
tumors. Invest. New Drugs, 2014, 32(3), 445-451.
http://dx.doi.org/10.1007/s10637-013-0044-7 PMID: 24271274
[139] Williams, R. Discontinued drugs in 2011: Oncology drugs. Expert
Opin. Investig. Drugs, 2013, 22(1), 9-34.
http://dx.doi.org/10.1517/13543784.2013.739605 PMID: 23127145
[140] Slingerland, M.; Gelderblom, H. The fate of camptothecin glyco-
conjugate: report of a clinical hold during a phase II study of BAY
56-3722 (formerly BAY 38-3441), in patients with recurrent or me-
tastatic colorectal cancer resistant/refractory to irinotecan. Invest.
New Drugs, 2012, 30(3), 1208-1210.
http://dx.doi.org/10.1007/s10637-011-9679-4 PMID: 21547368
[141] Trocóniz, I.F.; Cendrós, J.M.; Soto, E.; Pruñonosa, J.; Perez-
Mayoral, A.; Peraire, C.; Principe, P.; Delavault, P.; Cvitkovic, F.;
Lesimple, T.; Obach, R. Population pharmacoki-
netic/pharmacodynamic modeling of drug-induced adverse effects
of a novel homocamptothecin analog, elomotecan (BN80927), in a
Phase I dose finding study in patients with advanced solid tumors.
Cancer Chemother. Pharmacol., 2012, 70(2), 239-250.
http://dx.doi.org/10.1007/s00280-012-1906-y PMID: 22699813
[142] See, E.; Zhang, W.; Liu, J.; Svirskis, D.; Baguley, B.C.; Shaw, J.P.;
Wang, G.; Wu, Z. Physicochemical characterization of asulacrine
towards the development of an anticancer liposomal formulation
via active drug loading: Stability, solubility, lipophilicity and ioni-
zation. Int. J. Pharm., 2014, 473(1-2), 528-535.
http://dx.doi.org/10.1016/j.ijpharm.2014.07.033 PMID: 25079434
[143] Korth, C.; May, B.C.H.; Cohen, F.E.; Prusiner, S.B. Acridine and
phenothiazine derivatives as pharmacotherapeutics for prion dis-
ease. Proc. Natl. Acad. Sci. USA, 2001, 98(17), 9836-9841.
http://dx.doi.org/10.1073/pnas.161274798 PMID: 11504948
[144] Adjei, A.A. Current status of pyrazoloacridine as an anticancer
agent. Invest. New Drugs, 1999, 17(1), 43-48.
http://dx.doi.org/10.1023/A:1006242321596 PMID: 10555121
[145] Fortune, J.M.; Velea, L.; Graves, D.E.; Utsugi, T.; Yamada, Y.;
Osheroff, N. DNA topoisomerases as targets for the anticancer
drug TAS-103: DNA interactions and topoisomerase catalytic inhi-
bition. Biochemistry, 1999, 38(47), 15580-15586.
http://dx.doi.org/10.1021/bi991792g PMID: 10569942
[146] Stankovic, T.; Kidd, A.M.J.; Sutcliffe, A.; McGuire, G.M.; Robin-
son, P.; Weber, P.; Bedenham, T.; Bradwell, A.R.; Easton, D.F.;
Lennox, G.G.; Haites, N.; Byrd, P.J.; Taylor, A.M.R. ATM muta-
tions and phenotypes in ataxia-telangiectasia families in the British
Isles: expression of mutant ATM and the risk of leukemia, lym-
phoma, and breast cancer. Am. J. Hum. Genet., 1998, 62(2), 334-
345.
http://dx.doi.org/10.1086/301706 PMID: 9463314
[147] Jeggo, P.A.; Carr, A.M.; Lehmann, A.R. Splitting the ATM: Distinct
repair and checkpoint defects in ataxia-te langiectasia. Trends Genet.,
1998, 14(8), 312-316.
http://dx.doi.org/10.1016/S0168-9525(98)01511-X PMID:
9724963
[148] Hann, M.M.; Alderton, W.; Davenport, R.; Williams, P. Recent
disclosures of clinical candidates: Highlights from the Society of
Medicines Research Symposium. Drug Future, 2017, 42(2), 125-
129.
http://dx.doi.org/10.1358/dof.2017.042.02.2592800
[149] Simoneaux, R.; Font, H. AACR-NCI-EORTC International Con-
ference on molecular targets and cancer therapeutics: Discovery,
biology, and clinical applications Drug Future, Philadelphia, Penn-
sylvania, USA October 27-30, 2017.
[150] Aller, S.G.; Yu, J.; Ward, A.; Weng, Y.; Chittaboina, S.; Zhuo,
R.P.; Harrell, P.M.; Trinh, Y.T.; Zhang, Q.H.; Urbatsch, I.L.;
Chang, G. Structure of P-glycoprotein reveals a molecular basis for
poly-specific drug binding. Science, 2009, 323(5922), 1718-1722.
http://dx.doi.org/10.1126/science.1168750
838 Anti-Cancer Agents in Medicinal Chemistry, 2021, Vol. 21, No. 7 Man et al.
[151] Dantzig, A.H.; Law, K.L.; Cao, J.; Starling, J.J. Reversal of
multidrug resistance by the P-glycoprotein modulator, LY335979,
from the bench to the clinic. Curr. Med. Chem., 2001, 8(1), 39-50.
http://dx.doi.org/10.2174/0929867013373903 PMID: 11172691
[152] Katayama, R.; Koike, S.; Sato, S.; Sugimoto, Y.; Tsuruo, T.; Fujita,
N. Dofequidar fumarate sensitizes cancer stem-like side population
cells to chemotherapeutic drugs by inhibiting ABCG2/BCRP-
mediated drug export. Cancer Sci., 2009, 100(11), 2060-2068.
http://dx.doi.org/10.1111/j.1349-7006.2009.01288.x PMID:
19673889
[153] Luurtsema, G.; Schuit, R.C.; Klok, R.P.; Verbeek, J.; Leysen, J.E.;
Lammertsma, A.A.; Windhorst, A.D. Evaluation of [11C]laniquidar
as a tracer of P-glycoprotein: Radiosynthesis and biodistribution in
rats. Nucl. Med. Biol., 2009, 36(6), 643-649.
http://dx.doi.org/10.1016/j.nucmedbio.2009.03.004 PMID: 19647170
[154] Yu, Y.; Xin, Y.; Yang, H.F.; Liu, Z.M.; Liu, Y.L.; Shen, G.L.; Yu,
R.Q. Electrochemical sensor for cinchonine based on a competitive
host-guest complexation. Anal. Chim. Acta, 2005, 528(2), 135-142.
http://dx.doi.org/10.1016/j.aca.2004.10.041
[155] Akira, S.; Takeda, K. Toll-like receptor signalling. Nat. Rev. Im-
munol., 2004, 4(7), 499-511.
http://dx.doi.org/10.1038/nri1391 PMID: 15229469
[156] Grimmig, T.; Matthes, N.; Hoeland, K.; Tripathi, S.; Chandraker,
A.; Grimm, M.; Moench, R.; Moll, E.M.; Friess, H.; Tsaur, I.; Bla-
heta, R.A.; Germer, C.T.; Waaga-Gasser, A.M.; Gasser, M. TLR7
and TLR8 expression increases tumor cell proliferation and pro-
motes chemoresistance in human pancreatic cancer. Int. J. Oncol.,
2015, 47(3), 857-866.
http://dx.doi.org/10.3892/ijo.2015.3069 PMID: 26134824
[157] Stanley, M.A. Imiquimod and the imidazoquinolones: Mechanism
of action and therapeutic potential. Clin. Exp. Dermatol., 2002,
27(7), 571-577.
http://dx.doi.org/10.1046/j.1365-2230.2002.01151.x PMID:
12464152
[158] Fidock, M.D.; Souberbielle, B.E.; Laxton, C.; Rawal, J.; Delpuech-
Adams, O.; Corey, T.P.; Colman, P.; Kumar, V.; Cheng, J.B.;
Wright, K.; Srinivasan, S.; Rana, K.; Craig, C.; Horscroft, N.; Per-
ros, M.; Westby, M.; Webster, R.; van der Ryst, E. The innate im-
mune response, clinical outcomes, and ex vivo HCV antiviral effi-
cacy of a TLR7 agonist (PF-4878691). Clin. Pharmacol. Ther.,
2011, 89(6), 821-829.
http://dx.doi.org/10.1038/clpt.2011.60 PMID: 21451504
[159] Fakhari, A.; Nugent, S.; Elvecrog, J.; Vasilakos, J.; Corcoran, M.;
Tilahun, A.; Siebenaler, K.; Sun, J.; Subramony, J.A.; Schwarz, A.
Thermosensitive gel-based formulation for intratumoral delivery of
Toll-Like Receptor 7/8 dual agonist, MEDI9197. J. Pharm. Sci.,
2017, 106(8), 2037-2045.
http://dx.doi.org/10.1016/j.xphs.2017.04.041 PMID: 28456734
[160] Cheong, J.E.; Ekkati, A.; Sun, L. A patent review of IDO1 inhibi-
tors for cancer. Expert Opin. Ther. Pat., 2018, 28(4), 317-330.
http://dx.doi.org/10.1080/13543776.2018.1441290 PMID:
29473428
[161] Galanis, A.; Ma, H.; Rajkhowa, T.; Ramachandran, A.; Small, D.;
Cortes, J.; Levis, M. Crenolanib is a potent inhibitor of FLT3 with
activity against resistance-conferring point mutants. Blood, 2014,
123(1), 94-100.
http://dx.doi.org/10.1182/blood-2013-10-529313 PMID: 24227820
[162] Gueorguieva, I.; Tabernero, J.; Melisi, D.; Macarulla, T.; Merz, V.;
Waterhouse, T.H.; Miles, C.; Lahn, M.M.; Cleverly, A.; Benhadji,
K.A. Population pharmacokinetics and exposure-overall survival
analysis of the transforming growth factor-β inhibitor galunisertib
in patients with pancreatic cancer. Cancer Chemother. Pharmacol.,
2019, 84(5), 1003-1015.
http://dx.doi.org/10.1007/s00280-019-03931-1 PMID: 31482224
[163] Baxendale, I.R.; Cheung, S.; Kitching, M.O.; Ley, S.V.; Shearman,
J.W. The synthesis of neurotensin antagonist SR 48692 for prostate
cancer research. Bioorg. Med. Chem., 2013, 21(14), 4378-4387.
http://dx.doi.org/10.1016/j.bmc.2013.04.075 PMID: 23721919
... [1][2][3][4][5] Since chemotherapy is the only semi-synthetic derivative drug that the FDA has approved, finding and developing new anticancer agents that are actively effective is necessary to save millions of lives worldwide. 6,7 After our extensive research, we have found that quinoline is currently one of the most potent pharmaceutically active scaffolds and forms the basis for many natural compounds with remarkable pharmacological properties, like antifungal, 8-10 anti-inflammatory, 11,12 antibacterial, [13][14][15] anti-malarial, 16,17 anticancer, [18][19][20] and anti-tuberculosis effects. 21,22 Among the most important quinoline derivatives [23][24][25] that are used as a basic substance in some medicines is 4,7-dichloroquinoline. ...
... Due to the well-established pharmacokinetics and pharmacodynamics of these medications, a change to the established medication is an additional option for innovative drug discovery. According to reports, quinoline derivatives exhibit a variety of biological functions, including those of Anti-Cancer [21] , antioxidant and antibacterial [22] , antifungal [23] , antiinflammatory, anti-Alzheimer, anti-convulsant, anti-oxidant [24] , Antitubercular [25] , Anti-SARS-Cov-2 [26] , Nsp16-Nsp10) inhibitors [27] . Well-known antibiotics that fall within the category of antibacterial chemicals contain the quinoline nucleus [28] , called quinolones (Figure 1). ...
Article
The purpose of this article is to synthesize some novel quinoline-3-Carboxylic acid derivatives, analyze them, and assess their antibacterial potential. With the help of elemental analysis, IR, NMR, and mass spectral data, the synthesized derivatives were identified. Derivatives' antibacterial activity was determined using the cup and plate diffusion method. At doses of 50 μg/ml and 100 μg/ml, the substance demonstrated substantial antibacterial potential against the tested strains. To predict the pharmacokinetic properties (ADME) of these derivatives, in-silico investigations were also carried out. For the current study, the in silico Swiss ADME-assisted results were shown to be suitable for the derivation and synthesis of efficient antibacterial drugs.
... [78][79][80][81][82][83][84][85][86][87][88][89]. ...
Preprint
Nitrogen-based heterocyclic compounds are extensively used as corrosion inhibitors because of their ability to form strong coordination bonding with metallic atoms. A literature study showed that various reports are available on the anticorrosive behavior of N-heterocycles corrosion inhibitors. The present study is an effort to collect the information's covering the articles published from 2019 to the beginning of 2022, regarding the recent development of N-heterocyclic compounds, including pyridazine, quinoxaline, quinoline, indazole, pyridine, benzimidazole, and pyrimidine, as metallic corrosion inhibitors.
Article
Full-text available
In this work, the reactions of nonacarbonyldiiron with 8‐alkynyloxyquinoline ligands (L1‐L3) and their resultant iron carbonyl complexes (1a, 1b, 2, and 3) are described. The complexes including the ligands were fully characterized by using a variety of spectroscopic techniques. The four complexes were also crystallographically determined. In the reactions, the quinoline N atom underwent cyclization to form a quinolinium skeleton while the C‐H bond of the quinoline at position 2 or the substituted methyl group was cleaved to form Fe‐C bond(s). Meanwhile, the alkynyl triple bond was reduced to a single bond (1a) or double bonds (1b, 2, and 3) to further coordinate the iron in σ‐bonds or η‐bonds. Particularly, pathways of hydrogenation (1a and 2) and/or dehydrogenation (1b and 3) were accompanied by the reactions. Moreover, the oxidation states of the iron centers in these complexes were identified through analyses of their bond formation, FTIR and NMR spectroscopic signals (¹H NMR and DEPT‐135) in combination with their diamagnetic nature, that is, iron (0) for 2 and iron (I) for the rest of the diiron complexes.
Article
Full-text available
The syntheses of novel 2,4-bis[(substituted-aminomethyl)phenyl]phenylquinazolines 12 and 2,4-bis[(substituted-aminomethyl)phenyl]phenylquinolines 13 are reported here in six steps starting from various halogeno-quinazoline-2,4-(1H,3H)-diones or substituted anilines. The antiproliferative activities of the products were determined in vitro against a panel of breast (MCF-7 and MDA-MB-231), human adherent cervical (HeLa and SiHa), and ovarian (A2780) cell lines. Disubstituted 6- and 7-phenyl-bis(3-dimethylaminopropyl)aminomethylphenyl-quinazolines 12b, 12f, and 12i displayed the most interesting antiproliferative activities against six human cancer cell lines. In the series of quinoline derivatives, 6-phenyl-bis(3-dimethylaminopropyl)aminomethylphenylquinoline 13a proved to be the most active. G-quadruplexes (G4) stacked non-canonical nucleic acid structures found in specific G-rich DNA, or RNA sequences in the human genome are considered as potential targets for the development of anticancer agents. Then, as small aza-organic heterocyclic derivatives are well known to target and stabilize G4 structures, their ability to bind G4 structures have been determined through FRET melting, circular dichroism, and native mass spectrometry assays. Finally, telomerase inhibition ability has been also assessed using the MCF-7 cell line.
Article
Full-text available
A major global public health problem is the emergence of antimicrobial resistance to common commercial medications. Therefore, there is an urgent need for new antimicrobials with enhanced biological activity. In this regard , in this study 5 novel Quinolone derivative A, B, C, D, and E were synthesized and their structure was analyzed using UV light, FTIR, 1H NMR, and 13C NMR techniques. The structure of synthesized compounds was investigated. The well diffusion assay method was used to test the synthetic compounds' antibacterial properties in vitro against two Gram-positive (Staphylococcus aureus and Streptococcus pyogenes) and two Gram-negative (Escherichia coli and Klebsiella pneumoniae) bacteria. Ciprofloxacin drug was used as an antibiotic standard. Best activity was shown by compound C against E. coli with an inhibition zone of 30mm and 33 mm at 250and 500 μg/mL respectively. While compound B in Staphylococcus aureus has high inhibition zone of 42mm and 47mm at 250and 500μg/mL respectively. However compound D in Klebsiella pneumoniae has high inhibition zone of 32mm and 35mm at 250 and 500μg/mL respectively, while compound A for Streptococcus pyogenes has inhibition zone of 45mm and 47mm at 250 and 500μg/mL respectively.
Article
Full-text available
N-heterocyclic compounds are a natural and rich source of pharmacologically active molecules displaying anti-cancer properties through various antiproliferative mechanisms. Some of these N-heterocyclic compounds are already being utilized or evaluated in clinical settings for cancer treatment, highlighting their potential significance in discovering new anti-cancer agents. This study aims to gather information from articles published between 2019 and 2021 on the recent advancements in N-heterocyclic derivatives such as indazole, triazolopyrimidine, pyrazolopyrimidine, quinoxaline, benzimidazole, benzodiazepine, indole, and quinoline as promising anticancer agents, including their structure-activity relationships and mechanisms of action.
Article
Full-text available
The cancer stem cell (CSC) model defines tumors as hierarchically organized entities, containing a small population of tumorigenic CSC, or tumour-initiating cells, placed at the apex of this hierarchy. These cells may share common qualities with chemo- and radio-resistant cancer cells and contribute to self-renewal activities resulting in tumour formation, maintenance, growth and metastasis. Yet, it remains obscure what self-defense mechanisms are utilized by these cells against the chemotherapeutic drugs or radiotherapy. Recently, attention has been focused on the pivotal role of the DNA damage response (DDR) in tumorigenesis. In line with this note, an increased DDR that prevents CSC and chemoresistant cells from genotoxic pressure of chemotherapeutic drugs or radiation may be responsible for cancer metastasis. In this review, we focus on the current knowledge concerning the role of DDR in CSC and resistant cancer cells and describe the existing opportunities of re-sensitizing such cells to modulate therapeutic treatment effects.
Article
Full-text available
TAS-115 is a novel MET, VEGFR, FMS and PDGFR inhibitor, developed to improve the continuity of drug administration with a relatively short half-life. We assessed its tolerability, safety, pharmacokinetics, efficacy, and pharmacodynamics in patients with solid tumors. This open-label, dose-escalation phase I study of TAS-115 consisted of three parts: part 1 (TAS-115 was administered orally once daily [SID]); part 2 and an expansion part (SID in a 5 days on/2 days off [5-on/2-off] schedule for 21 days per cycle). In part 1 (200–800 mg SID administered to 21 patients), systemic exposure after single administration increased almost dose-proportionally. Three dose-limiting toxicities (DLTs) were observed in three patients: grade 3 rash (650 mg), thrombocytopenia with bleeding, and rash (800 mg). The maximum tolerated dose (MTD) was determined as 650 mg SID. In part 2, the 5-on/2-off schedule was evaluated at the MTD to improve treatment exposure. No DLTs were observed and no patients required treatment interruption in cycle 1. During part 2 and the expansion part (N = 61), grade ≥3 treatment-related adverse events were reported in 47 patients, with neutropenia (24.6%), hypophosphatemia (21.3%), anemia, and thrombocytopenia (14.8% each), and leukocytopenia (11.5%) occurring in ≥10% of patients. The best overall response was stable disease in 31 of 82 patients (37.8%). An apparent reduction in fluorodesoxyglucose-uptake and bone scan index was observed in some patients. TAS-115 was generally well tolerated, with manageable toxicities and recommended phase II dose was estimated as 650 mg SID, 5-on/2-off. Furthermore, promising antitumor activity was observed.
Article
Full-text available
Purpose To evaluate the exposure–overall survival (OS) relationship in patients with advanced pancreatic cancer treated with galunisertib plus gemcitabine (GG) or gemcitabine plus placebo (GP). Methods Galunisertib 300 mg/day was given orally as intermittent dosing and gemcitabine as per label. Galunisertib exposure metrics for each patient in the GG arm (n = 99) of a phase 2 study of pancreatic cancer were calculated. Parametric survival models were used to identify influential baseline and response covariates on OS. Results The population pharmacokinetics dataset included data from 297 patients/healthy subjects (age: 22–84 years, weight: 39–126 kg) across multiple studies, including this pancreatic cancer study. Galunisertib was rapidly absorbed with peak concentrations attained within 0.5–2 h and had an elimination half-life of 8 h. Between-subject variance on apparent clearance was estimated to be 47%. Age was the only characteristic to have a statistically significant effect on apparent clearance. A parametric Weibull survival model with treatment effect (dose) estimated a hazard ratio of 0.796, after adjusting for patient baseline factors that were significantly associated with OS. There was also a flat daily exposure–OS relationship within the observed exposure range, once all significant baseline covariates were included. Response covariates, such as reduction in CA19-9, time on treatment, and cumulative exposure over treatment cycles were also identified as significant factors for OS for patients with pancreatic cancer. Conclusions This analysis suggests that 300 mg/day galunisertib administered as 150 mg twice daily for 14 days on/14 days off treatment is an appropriate dosing regimen for patients with pancreatic cancer.
Article
Full-text available
A series of novel polymeric metal complexes of Co(II), Ni(II), Cu(II), Zn(II) and Cr(III) with 5,5′‐{(1E,1E′)‐1,4‐phenylenebis(diazene‐2,1‐diyl)}bis(quinolin‐8‐ol) (H2L) ligand were synthesized and characterized using elemental analyses, 1H NMR, mass, infrared, UV–visible and electron spin resonance (ESR) spectroscopies, magnetic moment and conductivity measurements as well as thermal analyses. The spectral and analytical data revealed the ligand adopted a neutral bidentate fashion when binding to metal ions via the nitrogen of azomethine of pyridine ring and the deprotonated hydroxyl group. Electronic and magnetic susceptibility measurements of the polymer complexes indicated octahedral geometry for all polymer complexes. The ESR spectral data provided information about the structures on the basis of Hamiltonian parameters and the degree of covalency. Molecular docking was used to predict the binding between the H2L and the receptor of breast cancer (3hb5). The immune response of the synthesized polymer complexes with bovine respiratory syncytial (BRS) vaccine in cattle was studied using serum neutralizations test (SNT). It was found that the isolated polymer complexes with BRS vaccine caused a significant increase in the antibody titre against BRS virus in SNT compared to BRS vaccine alone. The humeral immune response to bovine respiratory syncytial (BRS) vaccine and Cu(II), Co(II), Ni(II), Cr(III) and Zn(II) polymer complexes (1–5) was evaluated in vaccinated calves using serum neutralization test (SNT) which is characterized to be a rapid, practical and efficient method for evaluation. The data for 1–5 showed protective neutralizing serum antibody titre of 1.15, 1.12, 1.03, 0.90 and 1.13, respectively. The highest level of antibody of 1–5 was 1.98, 1.95, 1.95, 1.80 and 1.84, respectively.
Article
Full-text available
SARM1, an NAD-utilizing enzyme, regulates axonal degeneration. We show that CZ-48, a cell-permeant mimetic of NMN, activated SARM1 in vitro and in cellulo to cyclize NAD and produce a Ca²⁺ messenger, cADPR, with similar efficiency as NMN. Knockout of NMN-adenylyltransferase elevated cellular NMN and activated SARM1 to produce cADPR, confirming NMN was its endogenous activator. Determinants for the activating effects and cell permeability of CZ-48 were identified. CZ-48 activated SARM1 via a conformational change of the auto-inhibitory domain and dimerization of its catalytic domain. SARM1 catalysis was similar to CD38, despite having no sequence similarity. Both catalyzed similar set of reactions, but SARM1 had much higher NAD-cyclizing activity, making it more efficient in elevating cADPR. CZ-48 acted selectively, activating SARM1 but inhibiting CD38. In SARM1-overexpressing cells, CZ-48 elevated cADPR, depleted NAD and ATP, and induced non-apoptotic death. CZ-48 is a specific modulator of SARM1 functions in cells.
Article
Full-text available
Purpose The highly vascular malignant brain tumor glioblastoma (GBM) appears to be an ideal target for anti-angiogenic therapy; however, clinical trials to date suggest the VEGF antibody bevacizumab affects only progression-free survival. Here we analyze a group of patients with GBM who received bevacizumab treatment at recurrence and are stratified according to tumor molecular and genomic profile (TCGA classification), with the goal of identifying molecular predictors of the response to bevacizumab. Methods We performed a retrospective review of patients with a diagnosis of glioblastoma who were treated with bevacizumab in the recurrent setting at our hospital, from 2006 to 2014. Treatment was discontinued by the treating neuro-oncologists, based on clinical and radiographic criteria. Pre- and post-treatment imaging and genomic subtype were available on 80 patients. We analyzed time on bevacizumab and time to progression. EGFR gene amplification was determined by FISH. Results Patients with classical tumors had a significantly shorter time on bevacizumab than mesenchymal, and proneural patients (2.7 vs. 5.1 vs. 6.4 and 6.0 months respectively, p = 0.011). Classical subtype and EGFR gene amplification were significantly associated with a shorter time to progression both in univariate (p < 0.001 and p = 0.007, respectively) and multivariate analysis (both p = 0.010). Conclusion EGFR gene amplification and classical subtype by TCGA analysis are associated with significantly shorter time to progression for patients with recurrent GBM when treated with bevacizumab. These findings can have a significant impact on decision-making and should be further validated prospectively.
Article
Microtubules are essential for the mitotic division of cells and have been an attractive target for antitumour drugs due to the increased incidence of cancer and significant mitosis rate of tumour cells. In the past few years, tubulin-colchicine binding site, as one of the three binding pockets including taxol-, vinblastine- and colchicine-binding sites, has been focused on to design tubulin-destabilizing agents including inhibitors, antibody-drug conjugates and degradation agents. The present review is the first to cover a systemic and recent synopsis of tubulin-colchicine binding site agents. We believe that it would provide an increase in our understanding of receptor-ligand interaction pattern and consciousness of a series of challenges about tubulin target druggability.
Article
All the heritable alterations in gene expression and chromatin structure due to chemical modifications that do not involve changes in the primary gene nucleotide sequence is referred to as epigenetics. DNA methylation, histone modifications, and non-coding RNAs are distinct types of epigenetic inheritance. Epigenetic patterns have been linked to developmental stages, environmental exposure, and diet. Therapeutic strategies are now being developed to target human diseases such as cancer with mutations in epigenetic regulatory genes using specific inhibitors. Within the past two decades, seven epigenetic drugs have received regulatory approval and many others show their candidature in clinical trials. The current article represents a review of epigenetic heritance, diseases connected with epigenetic alterations and regulatory approved epigenetic drugs as future medicines.
Article
Introduction: Breast cancer (BC) is the most common cancer in women. Human epidermal growth factor receptor 2-positive (HER2-positive) BC represents up to 15% of all BC cases and is associated with a poor prognosis. Despite the substantial improvement obtained with the addition to the treatment of trastuzumab in this subtype of BC, disease recurrence can still occur. Areas covered: Anti-HER2 targeting drugs such as trastuzumab, pertuzumab, and T-DM1 have shown significant results in (neo)adjuvant setting. In this article, we will focus on available data for neratinib to reduce BC recurrence based mainly on the results of the ExteNET trial. This trial aimed to investigate whether neratinib can further reduce the risk of recurrence of patients diagnosed with HER2-positive BC. This trial demonstrated a significant reduction in the risk of invasive disease-free survival, particularly in hormone receptor-positive population. In addition, this review provides an expert opinion and analysis of the current situation in the adjuvant HER2-positive BC setting and in particular the escalation strategy of HER2 targeting. Expert opinion: The treatment landscape of HER2 positive BC in this setting will evolve in the coming years with a need for clinical and molecular perspective tools to guide therapy.