Science - USA (2020-08-21)

(Antfer) #1

RESEARCH ARTICLE SUMMARY



DRUG DEVELOPMENT


An orally available non-nucleotide STING agonist


with antitumor activity


Bo-Sheng Pan, Samanthi A. Perera†, Jennifer A. Piesvaux, Jeremy P. Presland,
Gottfried K. Schroeder†, Jared N. Cumming, B. Wesley Trotter*†, Michael D. Altman,
Alexei V. Buevich, Brandon Cash, Saso Cemerski, Wonsuk Chang, Yiping Chen, Peter J. Dandliker,
Guo Feng, Andrew Haidle, Timothy Henderson, James Jewell, Ilona Kariv, Ian Knemeyer,
Johnny Kopinja, Brian M. Lacey, Jason Laskey, Charles A. Lesburg, Rui Liang, Brian J. Long, Min Lu,
Yanhong Ma, Ellen C. Minnihan, Greg O’Donnell, Ryan Otte, Laura Price, Larissa Rakhilina,
Berengere Sauvagnat, Sharad Sharma, Sriram Tyagarajan, Hyun Woo, Daniel F. Wyss, Serena Xu,
David Jonathan Bennett†, George H. Addona†


INTRODUCTION:Activation of the STING (stimu-
lator of interferon genes) protein by its natural
ligand, cyclic guanosine monophosphate–
adenosine monophosphate (cGAMP), triggers
signaling responses, inducing the release of
type I interferons and other proinflammatory
cytokines. STING-controlled interferon pro-
duction is involved in antiviral defense as well
as antitumor immunity. Pharmacological ac-
tivation of STING is considered a promising
therapeutic strategy for cancer.


RATIONALE:First-generation STING agonists
are cyclic dinucleotide (CDN) analogs of cGAMP.
When administered systemically in animal
models, they induce inflammatory cytokines
equipotently in tumor and normal tissues,
owing to ubiquitous STING expression. Thus,


CDN-based STING agonists currently under-
going clinical trials are dosed by direct intra-
tumor injection, which limits their application
to a narrow set of tumors. To address a broad
spectrum of cancers, STING agonists that are
suitable for systemic administration and pref-
erentially target tumors are needed. We iden-
tified a previously unknown compound (MSA-2)
that exhibits such behavior through its dis-
tinctive mechanism of action. Moreover, MSA-2
is amenable to oral administration, a desir-
able delivery route because of convenience and
low cost.

RESULTS:MSA-2 was identified in a phenotypic
screen for chemical inducers of interferon-b
secretion (see the figure, top). In cell-free as-
says, MSA-2 binds human and mouse STING.

MSA-2 is orally available, manifesting similar
oral and subcutaneous exposure in mice. In
tumor-bearing mice, MSA-2 induced elevations
of interferon-bin plasma and tumors by both
routes of administration. Well-tolerated regi-
mens of MSA-2 induced tumor regressions in
mice bearing MC38 syngeneic tumors. Most
mice that exhibited complete regression were
resistant to reinoculation of MC38 cells, sug-
gesting establishment of durable antitumor
immunity. In tumor models that were moder-
ately or poorly responsive to PD-1 blockade,
combinations of MSA-2 and anti–PD-1 anti-
body were superior in inhibiting tumor growth
and prolonging survival over monotherapy
(see the figure, right).
Structural studies showed that MSA-2 was
bound as a noncovalent dimer to STING in a
“closed-lid”conformation (see the figure, left).
Each bound MSA-2 interacted with both mono-
mers of the STING homodimer (depicted in blue
and orange). The simplest model that can ac-
count for all observed equilibrium and kinetic
behaviors of MSA-2 is as follows: MSA-2 in
solution exists as monomers and noncovalent
dimers in an equilibrium that strongly favors
monomers; MSA-2 monomers cannot bind
STING, whereas the noncovalent MSA-2 dimers
bind STING with nanomolar affinity (see the
figure, center). The model was further sup-
ported by findings that covalently tethered
dimers of MSA-2 analogs exhibited nanomolar
affinity for STING.
Simulations and experimental analyses pre-
dicted that MSA-2, a weak acid, would exhibit
substantially higher cellular potency in an
acidified tumor microenvironment than nor-
mal tissue, owing to increased cellular entry
and retention combined with the inherently
steep MSA-2 concentration dependence of
STING occupancy (see the figure, bottom). It
is likely that preferential activation of STING
by MSA-2 in tumors substantially contrib-
utes to the observed favorable in vivo anti-
tumor activity and tolerability profile of this
compound.

CONCLUSION:In this work, we describe the
identification, in vivo antitumor properties,
and mechanism of action of MSA-2, an orally
available human STING agonist. MSA-2 could
prove valuable for the discovery and design of
human STING agonists suitable for systemic
administration in the clinic.▪

RESEARCH


Panet al.,Science 369 , 935 (2020) 21 August 2020 1of1


The list of author affiliations is available in the full article online.
*These authors contributed equally to this work.
†Corresponding author. Email: samanthi_perera@merck.
com (S.A.P.); [email protected] (G.K.S.);
[email protected] (B.W.T.); jonathan.bennett@
merck.com (D.J.B.); [email protected] (G.H.A.)
Cite this article as B.-S. Panet al.,Science 369 , eaba6098
(2020). DOI: 10.1126/science.aba6098

READ THE FULL ARTICLE AT
https://doi.org/10.1126/science.aba6098

STING agonist MSA-2.Identified in a cell-based screen, MSA-2 is bound to STING as a noncovalent dimer.
Extensive experimental analysis indicates that MSA-2 predimerization is required for binding. Acidic tumor
microenvironments favor permeable, uncharged MSA-2. Intracellular MSA-2 is“trapped”(deprotonated) and
accumulation drives MSA-2 dimerization, preferentially activating STING intratumorally. Orally dosed MSA-2
is well tolerated in mice, exhibiting STING-dependent antitumor activity, as monotherapy and combined with
antibodies against PD1 (anti-PD1). Me, methyl group; IFNb, interferon-b.

Free download pdf