Science - USA (2021-12-10)

(Antfer) #1
SCIENCE science.org

By Alan Melcher^1 , Kevin Harrington^1 ,
Richard Vile^2

O

ncolytic virus (OV) therapy is now
something of a misnomer. The initial
paradigm was that naturally occur-
ring or genetically engineered viruses
would preferentially infect, replicate
in, and lyse cancer cells relative to
normal cells, leading to selective direct cy-
totoxicity. The immune system used to be
considered detrimental, given its potential to
recognize and respond to the virus, prevent
its replication, and limit viral spread and tu-
mor cell killing ( 1 ). This view has now been
largely replaced by a model in which infec-
tion of the tumor by the virus makes it more
visible to the immune system for recognition
and attack. Tumor cell killing by the virus
and/or immune effector cells effectively gen-
erates an in situ vaccine, with tumor antigens
released into a microenvironment modified
by viral infection to reverse tumor-induced
immune suppression.
By 2020, there were ~100 reported clini-
cal trials using OVs, treating more than 3000
cancer patients. Approvals were granted in
different countries for picorna-, adeno-, and
herpes viruses, although only one agent
was approved by the US Food and Drug
Administration (FDA). The growing rec-
ognition of the immune basis of oncolytic
virotherapy coincided with the success of
immunotherapy in the clinic, particularly
immune checkpoint inhibitors (ICIs), which
block negative regulatory elements that
control immune tolerance, specifically the
programmed cell death protein 1 (PD-1) axis
and cytotoxic T lymphocyte–associated an-
tigen 4 ( CTLA-4). These drugs revealed that
the immune system can and does respond
to cancer, but tumors evolve to escape im-
mune detection through immunoediting ( 2 ).
However, if the immune system can be reen-
ergized and reactivated, immune control of
the tumor can be reestablished.
The number of OVs being tested in pre-
clinical models, with a particular focus on
their immunogenicity, has since increased.
However, in some ways the field has been
struggling with an embarrassment of (po-
tential) riches. It has been unclear how to

prioritize the most promising preclinical
agents to take forward to clinical testing.
Additionally, how to accurately measure
the immunogenicity of tumor cell death
after OV infection—particularly for viruses
that do not readily infect mouse cells (e.g.,
coxsackie virus, adenovirus, and measles vi-
rus) and so cannot be tested reliably in pre-
clinical models—has been a challenge. The
variety of OVs covers DNA, single-stranded
RNA (ssRNA), and double-stranded RNA
(dsRNA) viruses, and the immunobiologi-
cal consequences after infection of tumor
and/or other cells varies, including the sig-
naling pathways activated, such as retinoic
acid–inducible gene I (RIG-I)–like recep-
tors for viral RNA and the cyclic GMP–AMP
synthase (cGAS)–stimulator of interferon
genes (STING) pathway for cytosolic DNA.
The degree to which these pathways remain
intact within cancer cells, and the relative
contribution of the response to infection of
nonmalignant cells in the tumor microen-
vironment (such as cancer-associated fibro-
blasts, endothelial cells, macrophages, and
other immune cell subtypes), adds further
complexity to understanding how tumor
immunogenicity is generated by OVs.
Much current work is focused on the abil-
ity to genetically modify OVs to encode trans-
genes, such as cytokines, ICIs, tumor-associ-
ated antigens, bispecific T cell engagers, and
microRNAs, as payloads to boost the antitu-
mor immune response. However, the choice
of these encoded transgenes has often been
based on limited mechanistic understand-
ing. For example, granulocyte-macrophage
colony-stimulating factor (GM-CSF) is ex-
pressed in many OVs being clinically tested,
even though the evidence supporting the
benefit of virally encoded GM-CSF is limited.
A further challenge is the inevitable antiviral
(alongside antitumor) immune response acti-
vated by treatment. Even if viruses to which
patients have not been previously exposed
are used, neutralizing antiviral antibodies
(NAbs) will increase with treatment, poten-
tially compromising repeat administrations,
particularly if the route of administration is
systemic. Even though intravenously deliv-
ered OVs (e.g., adenovirus and vaccinia virus)
can infect tumors in patients ( 3 , 4 ), despite
the presence of NAbs in the case of reovirus
( 5 ), concerns about sufficient delivery to tu-
mors have led to the administration of many
OVs—including talimogene laherparepvec

(T-Vec), a clinically approved OV for the
treatment of advanced melanoma—being re-
stricted to direct intratumoral injection.
The rapid expansion of ICIs into the clinic
has affected the development of OVs and
other immunotherapeutics in unexpected
ways. Usually, advances in the clinic lag years
behind the laboratory discoveries that under-
pin them, but this has reversed. To improve
on the benefit from ICIs, which remains lim-
ited to an overall response rate of only 10 to
15% of all cancer patients (although this is
higher in certain tumor types and some pa-
tient groups), combination strategies have
been empirically tested without sufficient
understanding of the immunobiology of why
they might (or might not) work. Trials of ICI
combinations with chemotherapy or radio-
therapy are relatively easy to design because
the ICI is added to current standard-of-care
regimens, and such trials have met with con-
siderable success ( 6 , 7 ). However, immuno-
therapy-OV combinations have so far been
disappointing. In almost all such studies, the
collection and analysis of patient samples
has been inadequate and/or unreported, so
it remains generally unknown why combina-
tions, which showed promise in preclinical
models, have failed to translate into patient
benefit. This lack of correlative, translational
data particularly applies to negative trials
(which are at least as important, in terms of
mechanistic understanding, as positive stud-
ies), leaving the immune changes that occur
in patients after treatment unknown.
There have been few large, randomized con-
trolled trials including OVs so far, and those
that have been reported have been discour-
aging. A study of vocimagene amiretrorepvec
(TOCA 511), a replicating retrovirus encoding
cytosine deaminase, which converts the pro-
drug 5-fluorocytosine into 5-fluorouracil,
failed in a phase 2/3 study of direct injection
into the surgical cavity on first or second re-
section of high-grade glioma, randomized
against standard of care ( 8 ). A second phase
2b study involving pexastimogene devacire-
pvec (Pexa-Vec), a vaccinia virus encoding
GM-CSF, was also unsuccessful when tested
in sorafenib-refractory hepatocellular carci-
noma (HCC) ( 9 ). Both studies have plausible
explanations for their lack of success. In the
TOCA 511 study, patients had fewer treat-
ment cycles than in earlier phase trials, and
for Pexa-Vec, the poor responses of advanced
HCC are notoriously difficult to reverse.

CANCER

Oncolytic virotherapy as immunotherapy


Recognizing immune responses to oncolytic virotherapy opens the way for new combinations


(^1) The Institute of Cancer Research, London, UK. (^2) Mayo
Clinic, Rochester, MN, USA. Email: [email protected];
[email protected]; [email protected]
10 DECEMBER 2021 • VOL 374 ISSUE 6573 1325

Free download pdf