Knockout of p21-Activated Kinase 4 Stimulates MHC I Expression of Pancreatic Cancer Cells via an Autophagy-Independent Pathway
Cancers,
Год журнала:
2025,
Номер
17(3), С. 511 - 511
Опубликована: Фев. 3, 2025
Background/Objectives:
Pancreatic
ductal
adenocarcinoma
(PDA)
is
one
of
the
most
malignant
solid
cancers.
KRAS
mutation
accounts
for
over
90%
cases.
p21-activated
kinases
(PAKs)
act
downstream
and
are
involved
in
tumorigenesis.
The
inhibition
PAK4
suppresses
PDA
by
stimulating
tumor
infiltration
cytotoxic
T
cells.
major
histocompatibility
complex
class
I
(MHC
I)
a
key
presenting
antigens
to
MHC
degradation
via
autophagy
promotes
immune
evasion
pancreatic
cancer.
We
investigated
effect
on
expression
autophagy.
Methods:
In
this
study,
using
proteomic
analysis,
fluorescence-activated
cell
sorting
(FACS),
immunoblotting,
we
examined
knockout
(KO)
human
cells
identify
mechanism
stimulation
inhibition.
Results:
found
that
KO
increased
two
lines:
MiaPaCa-2
PANC-1.
also
cancer
However,
chloroquine
(CQ)
did
not
affect
apoptosis
death.
More
importantly,
CQ
alter
stimulated
KO,
indicating
an
autophagy-independent
pathway.
Conclusions:
identified
role
cells,
which
independent
Язык: Английский
Phosphoproteomics identifies determinants of PAK inhibitor sensitivity in leukaemia cells
Cell Communication and Signaling,
Год журнала:
2025,
Номер
23(1)
Опубликована: Март 13, 2025
Abstract
Background
The
P21
activated
kinases
(PAK)
are
frequently
dysregulated
in
cancer
and
have
central
roles
oncogenic
signalling,
prompting
the
development
of
PAK
inhibitors
(PAKi)
as
anticancer
agents.
However,
such
compounds
not
reached
clinical
use
because,
at
least
partially,
there
is
a
limited
mechanistic
understanding
their
mode
action.
Here,
we
aimed
to
characterize
functional
molecular
responses
PAKi
(PF-3758309,
FRAX-486
IPA-3)
multiple
acute
myeloid
leukaemia
(AML)
models
gain
insights
on
biochemical
pathways
affected
by
these
this
disease
identify
determinants
response
patient
samples.
Methods
We
mined
phosphoproteomic
datasets
primary
AML,
used
proteomics
phosphoproteomics
profile
impact
immortalized
(P31/Fuj
MV4-11),
AML
cells
from
8
patients.
These
omics
were
integrated
with
gene
dependency
data
which
proteins
targeted
necessary
for
proliferation
AML.
studied
effect
cell
cycle
progression,
proliferation,
differentiation
apoptosis.
Finally,
input
machine
learning
that
predicted
ex
vivo
two
independent
(with
36
50
cases,
respectively)
PF-3758309
markers
response.
Results
found
PAK1
activation–
measured
data–
was
predictive
poor
prognosis
cases.
most
effective
reducing
inducing
apoptosis
lines.
In
lines
cells,
inhibited
PAK,
AMPK
PKCA
activities,
reduced
c-MYC
transcriptional
activity
expression
ribosomal
proteins,
FLT3
pathway
FLT3-ITD
mutated
cells.
STAT5
phosphorylation
Tyr699.
Functionally,
cell-growth,
induced
apoptosis,
blocked
progression
promoted
model-dependent
manner.
ML
modelling
accurately
classified
samples
sensitive
or
resistant
treatment,
highlighted
PHF2
Ser705
robust
biomarker.
Conclusions
summary,
our
define
proteomic,
immortalised
suggest
route
personalise
treatments
based
inhibitors.
Язык: Английский
The Peptide PROTAC Modality: A New Strategy for Drug Discovery
MedComm,
Год журнала:
2025,
Номер
6(4)
Опубликована: Март 24, 2025
ABSTRACT
In
recent
years,
proteolysis
targeting
chimera
(PROTAC)
technology
has
made
significant
progress
in
the
field
of
drug
development.
Traditional
drugs
mainly
focus
on
inhibiting
or
activating
specific
proteins,
while
PROTAC
provides
new
ideas
for
treating
various
diseases
by
inducing
degradation
target
proteins.
Especially
peptide
PROTACs,
due
to
their
unique
structural
and
functional
characteristics,
they
have
become
a
hot
research
topic.
This
review
detailed
description
key
components,
mechanisms,
design
principles
elaborates
applications
skin‐related
diseases,
oncology,
other
potential
therapeutic
fields,
analyzes
advantages
challenges,
looks
forward
future
development
prospects.
The
not
only
opens
up
paths
development,
but
also
solving
resistance
safety
issues
faced
traditional
small‐molecule
drugs.
Compared
with
PROTACs
such
as
multitargeting,
biodegradability,
low
toxicity,
flexibility
design.
With
deepening
continuous
maturity
technology,
are
expected
one
important
strategies
discovery,
providing
hope
treatment
more
intractable
diseases.
Peptide
ushering
era
precision
medicine.
Язык: Английский
The pathogenesis and therapeutic implications of metabolic reprogramming in renal cell carcinoma
Cell Death Discovery,
Год журнала:
2025,
Номер
11(1)
Опубликована: Апрель 19, 2025
Abstract
Renal
cell
carcinoma
(RCC),
a
therapeutically
recalcitrant
genitourinary
malignancy,
exemplifies
the
profound
interplay
between
oncogenic
signaling
and
metabolic
adaptation.
Emerging
evidence
positions
reprogramming
as
central
axis
of
RCC
pathogenesis,
characterized
by
dynamic
shifts
in
nutrient
utilization
that
transcend
canonical
Warburg
physiology
to
encompass
lipid
anabolism,
glutamine
auxotrophy,
microenvironment-driven
plasticity.
This
orchestrated
rewiring
cellular
energetics
sustains
tumor
proliferation
under
hypoxia
while
fostering
immunosuppression
through
metabolite-mediated
T
exhaustion
myeloid-derived
suppressor
activation.
Crucially,
exhibits
heterogeneity
across
histological
subtypes
intratumoral
regions—a
feature
increasingly
recognized
determinant
therapeutic
resistance.
Our
review
systematically
deciphers
molecular
architecture
metabolism,
elucidating
how
VHL/HIF
mutations,
mTOR
pathway
dysregulation,
epigenetic
modifiers
converge
reshape
glucose
flux,
droplet
biogenesis,
amino
acid
catabolism.
We
present
novel
insights
into
spatial
zonation
within
tumors,
where
pseudohypoxic
niches
engage
lactate
shuttling
cholesterol
efflux
adjacent
vasculature,
creating
pro-angiogenic
immunosuppressive
microdomains.
Therapeutically,
we
evaluate
first-in-class
inhibitors
targeting
rate-limiting
enzymes
de
novo
lipogenesis
proposing
biomarker-driven
strategies
overcome
compensatory
highlight
synergy
glutaminase
PD-1
blockade
reinvigorating
CD8
+
function,
role
lipid-loaded
cancer-associated
fibroblasts
shielding
tumors
from
ferroptosis.
Finally,
outline
translational
roadmap
integrating
multi-omics
profiling,
functional
metabolomics,
biology
match
vulnerabilities
with
precision
therapies.
Язык: Английский
Advancements in delivery Systems for Proteolysis-Targeting Chimeras (PROTACs): Overcoming challenges and expanding biomedical applications
Yawei Yu,
Weitong Hu,
Yihua Xu
и другие.
Journal of Controlled Release,
Год журнала:
2025,
Номер
unknown, С. 113719 - 113719
Опубликована: Апрель 1, 2025
Язык: Английский
Design and optimization strategies of PROTACs and its Application, Comparisons to other targeted protein degradation for multiple oncology therapies
M. Malarvannan,
S. Unnikrishnan,
S. Monohar
и другие.
Bioorganic Chemistry,
Год журнала:
2024,
Номер
154, С. 107984 - 107984
Опубликована: Ноя. 22, 2024
Язык: Английский
Peptides as Targeting Agents and Therapeutics: A Brief Overview
Biomacromolecules,
Год журнала:
2024,
Номер
25(11), С. 6923 - 6935
Опубликована: Окт. 24, 2024
The
controllability
and
specificity
of
peptides
make
them
ideal
for
targeting
therapeutic
delivery
systems
as
agents
that
interfere
with
the
essential
functions
pathogens
tumors.
Peptides
can
also
mimic
natural
protein
structures
or
parts
thereof,
agonize
receptors,
be
conjugated
to
other
molecules
will
self-assemble.
In
this
short
Review,
we
discuss
research
from
last
ten
years
into
peptide
use
in
three
arenas:
treatment
cancer,
pathogens,
specific
organs
organelles.
These
studies
demonstrate
successful
application
Язык: Английский
PAK4-targeted PROTACs in clear cell renal cell carcinoma: a two-for-one targeted and immune therapy?
EBioMedicine,
Год журнала:
2024,
Номер
105, С. 105218 - 105218
Опубликована: Июнь 25, 2024
Renal
cell
carcinoma
(RCC)
affects
approximately
82,000
patients
and
causes
over
15,000
deaths
in
the
United
States
per
year.1Siegel
R.L.
Giaquinto
A.N.
Ahmedin
J.
Cancer
statistics,
2024.CA
J
Clin.
2024;
74:
12-49https://doi.org/10.3322/CAAC.21820Crossref
PubMed
Scopus
(0)
Google
Scholar
Clear
RCC
(ccRCC)
accounts
for
around
75%
of
cases
is
characterized
by
near-ubiquitous
biallelic
inactivation
VHL,
which
ultimately
activates
a
pseudohypoxia
program
driven
hypoxia
inducible
factor
(HIF)
family.
The
management
with
advanced
or
metastatic
ccRCC
has
been
revolutionized
therapies
targeting
HIF-VEGF
axis
immune
checkpoint
inhibitors
PD-1
CTLA-4
axes.
Combinations
these
pathways
have
thus
far
proven
to
be
most
effective
strategies
treatment
ccRCC.
Despite
improvements
ccRCC,
still
eventually
progress.
Further
improving
outcomes
will
likely
require
combinations
that
incorporate
novel
targets,
would
ideally
synergistic
additive
activity
as
well
minimally
overlapping
toxicities.2Saliby
R.M.
Saad
E.
Labaki
C.
et
al.Novel
targeted
renal
carcinoma:
building
on
successes
vascular
endothelial
growth
mTOR
inhibition.Hematol
Oncol
Clin
North
Am.
2023;
37:
1015-1026https://doi.org/10.1016/J.HOC.2023.05.022Summary
Full
Text
PDF
Scholar,3Braun
D.A.
Bakouny
Z.
Hirsch
L.
al.Beyond
conventional
immune-checkpoint
inhibition
—
immunotherapies
carcinoma.Nat
Rev
Oncol.
2021;
18:
199-214https://doi.org/10.1038/s41571-020-00455-zCrossref
(189)
In
this
study,
Xu
al.
developed
tested
efficacy
toxicity
proteolysis
chimera
(PROTAC)
degrades
P21-activated
kinase-4
(PAK4)
ccRCC.4Xu
S.
Ma
B.
Jian
Y.
Yao
Wang
Fan
Development
PAK4-targeting
PROTAC
therapy:
concurrent
cancer
proliferation
enhancement
response.eBioMedicine.
104:
105162https://doi.org/10.1016/j.ebiom.2024.105162Summary
(1)
PAK4
serine/threonine
kinase,
one
six
PAKs
all
share
structural
functional
homology.
overexpressed
multiple
cancers
associates
worse
various
tumor
types.
roles
cancer,
including
promotion
invasion,
metastasis,
proliferation,
epithelial-to-mesenchymal
transition
through
activation
Wnt/Beta-catenin,
MAPK,
PI3K/AKT
pathways.5Won
S.Y.
Park
J.J.
Shin
E.Y.
Kim
E.G.
signaling
health
disease:
defining
PAK4–CREB
axis.Exp
Mol
Med.
2019;
51:
1-9https://doi.org/10.1038/s12276-018-0204-0Crossref
(54)
also
shown
inhibit
ccRCC6Aboud
O.A.
Chen
C.H.
Senapedis
W.
Baloglu
Argueta
Weiss
R.H.
Dual
specific
NAMPT
KPT-9274
decreases
kidney
growth.Mol
Ther.
2016;
15:
2119https://doi.org/10.1158/1535-7163.MCT-16-0197Crossref
(116)
potentiate
immunotherapy
responses
other
types.7Abril-Rodriguez
G.
Torrejon
D.Y.
Liu
al.PAK4
improves
blockade
immunotherapy.Nat
Cancer.
1:
46-58https://doi.org/10.1038/s43018-019-0003-0Crossref
(91)
evaluated
its
line
mouse
models
patient-derived
organoids.
They
studied
profile
pharmacokinetics
vivo
goal
translating
PAK4-targeted
PROTAC's
use
Multiple
small
molecule
are
development,
though
insufficient
inadequate
selectivity
limited
their
potential
date.8Li
Lu
Q.
Xie
Yu
Zhang
A.
Recent
advances
development
p21-activated
kinase
4
anti-tumor
agents.Front
Pharmacol.
2022;
13https://doi.org/10.3389/FPHAR.2022.956220Crossref
PROTACs
promising
approach
selectively
effectively
degrade
oncogenic
targets
could
circumvent
limitations
classical
inhibitors.
build
approaches
protein
degradation,
namely
immunomodulatory
imide-containing
drugs
(IMiDs)
selective
estrogen
receptor
degraders
(SERDs)
Food
Drug
Administration
(FDA)-approved
myeloma
breast
respectively.9Fang
Han
al.Targeted
degrader
cancer:
advances,
challenges,
opportunities.Trends
Pharmacol
Sci.
44:
303-317https://doi.org/10.1016/j.tips.2023.03.003Summary
(14)
currently
phase
II
III
clinical
trials,
degradation
androgen
prostate
(Luxdegalutamide)
(Vepdegestrant).10Wang
X.
Qin
Z.L.
Li
N.
al.Annual
review
anticancer
agents
2022.Eur
Med
Chem.
267116166https://doi.org/10.1016/J.EJMECH.2024.116166Crossref
To
ccRCC-specific
loss
VHL
(another
E3
ubiquitin
ligase
commonly
used
PROTACs),
leveraged
MDM2
ligase.
authors
demonstrate
vitro
(in
organoids)
increases
CD8+
T
infiltration
into
tumors
(Fig.
1).
show
effect
mediated
unaffected
PROTAC.
propose
induces
an
response
can
combining
inhibitor,
given
highly
interesting
add
way
armamentarium
available
agents.
direct
effects
appear
seemingly
tolerable
may
allow
it
combined
VEGF-targeted
tyrosine
and/or
While
claim
potentiating
inhibitors,
not
fully
substantiated
data
despite
being
previously
observed
Further,
whether
humans
sufficient
single
agent
warrant
further
remains
seen,
particularly
since
existing
either
failed
trials
date
early
stages
development.
Finally,
present
evidence
but
presented
effectiveness
forms
(i.e.
non-clear
RCC)
sparse
warrants
investigation.
conclusion,
study
develops
potentially
represent
pathway
inhibition.
both
target
cell-autonomous
driver
activate
appealing,
investigation
determine
safety
Conceptualization:
Ziad
Bakouny.
Supervision:
David
Braun,
Ed
Reznik,
Ari
Hakimi.
Writing:
Bakouny,
All
read
approved
final
manuscript.
following
report
competing
interests
(all
outside
submitted
work):
Z.B.:
Honoraria
from
UpToDate;
Associate
Editor
at
Journal
Clinical
Oncology
Informatics
(JCO
CCI);
co-chair
American
Society
Oncology's
International
Medical
Graduate
Community
Practice
(ASCO
IMG
CoP);
co-founder
Oncologists
nonprofit
non-governmental
organization.
D.A.B.:
advisory
board
fees
Exelixis,
AVEO,
Eisai,
Elephas,
equity
Fortress
Biotech
(subsidiary)
CurIOS
Therapeutics,
consulting/personal
Expert
Now,
Adnovate
Strategies,
MDedge,
CancerNetwork,
Catenion,
OncLive,
Cello
Health
BioConsulting,
PWW
Consulting,
Haymarket
Network,
Aptitude
Health,
ASCO
Post/Harborside,
Targeted
Oncology,
Merck,
Pfizer,
MedScape,
Accolade
2nd
MD,
DLA
Piper,
AbbVie,
Compugen,
Link
Cell
Therapies,
Rock,
research
support
Exelixis
AstraZeneca,
work.
E.R.:
Consulting/personal
Xontogeny,
llc,
A.A.H.:
funding
This
work
was
supported
National
Institute
Center
Core
(grant
P30-CA008748)
supporting
Memorial
Sloan
Kettering
Center.
responseThis
peptide
drug
only
curtails
microenvironment
enhances
response.
Our
paves
innovative
cancer.
Full-Text
Open
Access
Язык: Английский