Cytokine Conditioning Enhances Systemic Delivery ...

5 downloads 70 Views 2MB Size Report
Enhances Systemic Delivery and Therapy of an Oncolytic Virus, Molecular Therapy ... Moreover, pre-existing NAb to an oncolytic virus may, therefore, even be.
ACCEPTED ARTICLE PREVIEW

Accepted Article Preview: Published ahead of advance online publication Cytokine Conditioning Enhances Systemic Delivery and Therapy of an Oncolytic Virus

Elizabeth Ilett , Timothy Kottke, Oliver Donnelly, Jill Thompson, Candice Willmon, Rosa Diaz, Shane Zaidi, Matt Coffey, Peter Selby, Kevin Harrington, Hardev Pandha; Alan Melcher, Richard Vile

us

cr

ip t

Cite this article as: Elizabeth Ilett , Timothy Kottke, Oliver Donnelly, Jill Thompson, Candice Willmon, Rosa Diaz, Shane Zaidi, Matt Coffey, Peter Selby, Kevin Harrington, Hardev Pandha; Alan Melcher, Richard Vile Cytokine Conditioning Enhances Systemic Delivery and Therapy of an Oncolytic Virus, Molecular Therapy accepted article preview online 24 June 2014; doi:10.1038/mt.2014.118

A

cc

ep

te

d

m

an

This is a PDF file of an unedited peer-reviewed manuscript that has been accepted for publication. NPG is providing this early version of the manuscript as a service to our customers. The manuscript will undergo copyediting, typesetting and a proof review before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers apply.

Received 06 February 2014; accepted 18 June 2014; Accepted article preview online 24 June 2014

© 2014 The American Society of Gene & Cell Therapy. All rights reserved

ACCEPTED ARTICLE PREVIEW

Cytokine Conditioning Enhances Systemic Delivery and Therapy of an Oncolytic Virus

Elizabeth Ilett

1,2,

*, Timothy Kottke2,*, Oliver Donnelly1, Jill Thompson2, Candice Willmon2, Rosa

Diaz2, Shane Zaidi2, Matt Coffey3, Peter Selby1, Kevin Harrington4, Hardev Pandha5; Alan Melcher 1,#, Richard Vile1,2,6#

1

2

Leeds Institute of Cancer and Pathology, St. James’ University Hospital, Leeds, UK;

Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905; 3Oncolytics Biotech

rip

t

Incorporated, Calgary, Canada; 4The Institute of Cancer Research, 237 Fulham Road, London,

us c

SW3; 5University of Surrey, Guildford, UK; 6Department of Immunology, Mayo Clinic, Rochester,

an

MN 55905

m

Correspondence should be addressed to R.V. ([email protected]) / Mayo Clinic / Gugg 18

d

/ 200 First Street SW / Rochester, MN 55905 / Phone: 507-284-3178 / FAX: 507-266-2122.

ep te

* These authors contributed equally to this work;

A cc

# These authors contributed equally to this work

Running Title: Systemic delivery of oncolytic reovirus

There is no conflict of interest to disclose.

Key Words: Cancer immunotherapy, Oncolytic virus, Viral delivery, VSV, GM-CSF conditioning B16 melanoma

© 2014 The American Society of Gene & Cell Therapy. All rights reserved

ACCEPTED ARTICLE PREVIEW

ABSTRACT Optimum clinical protocols require systemic delivery of oncolytic viruses in the presence of an intact immune system.

We have shown that pre-conditioning with immune modulators, or

loading virus onto carrier cells ex vivo, enhances virus-mediated anti-tumor activity. Our early trials of systemic reovirus delivery, showed that, following infusion, reovirus could be recovered from blood cells – but not from plasma – suggesting that rapid association with blood cells may protect virus from NAb. We therefore postulated that stimulation of potential carrier cells directly

t

in vivo prior to i.v. viral delivery would enhance delivery of cell-associated virus to tumor. We

rip

show that mobilization of the CD11b+ cell compartment by GM-CSF immediately prior to i.v.

us c

reovirus, eliminated detectable tumor in mice with small B16 melanomas, and achieved highly significant therapy in mice bearing well-established tumors. Unexpectedly, cytokine conditioning

an

therapy was most effective in the presence of pre-existing NAb. Consistent with this, reovirus

m

bound by NAb effectively accessed monocytes/macrophages and was handed off to tumor

d

cells. Thus, pre-conditioning with cytokine, stimulated recipient cells in vivo for enhanced viral

ep te

delivery to tumors. Moreover, pre-existing NAb to an oncolytic virus may, therefore, even be

A cc

exploited for systemic delivery to tumors, in the clinic.

© 2014 The American Society of Gene & Cell Therapy. All rights reserved

ACCEPTED ARTICLE PREVIEW

INTRODUCTION Oncolytic virotherapy is based on the concept that a replicating virus introduced into a tumor will rapidly spread through and lyse that tumor, with targeted replication being possible through natural, or engineered, selectivity1. Encouragingly, several viruses are currently entering later stage clinical trials and a randomized phase III study (OPTiM) using herpes simples virus (HSV) therapy for melanoma has achieved its primary endpoint, with a durable response rate of 16% seen in patients receiving HSV, compared with 3% in the control arm 2. Trials of this sort have

t

also highlighted the multi-component role of the immune system on the efficiency of virotherapy.

rip

Thus, anti-viral immune responses clearly impair virus delivery to tumors following systemic

against tumor8,

9

and/or virus4,

7

and tumor clearance often requires immune effectors

6-11

However, a major clinical challenge remains the

.

an

not always correlate with therapy6,

us c

administration and can restrict replication/oncolysis3-5. On the other hand, virus replication does

12-14

. In this respect, many barriers to efficient systemic delivery exist,

d

metastatic tumors1,

m

development of protocols for systemic delivery, in the presence of an intact immune system, to

ep te

including the tumor vasculature 15-17, virus inactivation (including by neutralizing antibody (NAb)), mislocalization, sequestration, and inadequate extravasation 13, 18-20.

A cc

In our own studies, we have developed the use of reovirus as a systemically delivered oncolytic agent in both pre-clinical models9, 13, 21-26 and in early Phase clinical trials14, 27-30. Reovirus has direct oncolytic activity against many human/murine tumor cells 29, 31, partly because of disruption of the PKR-mediated anti-viral response in malignant cells32, 33. In addition, we have shown that anti-tumor therapy is directly associated with immune activation by virus replication in tumors24, 25

. To mimic the clinical challenges of systemic delivery of oncolytic viruses, we developed a

murine model in which injection of reovirus into subcutaneous (s.c.) B16 melanomas generates therapy, but intravenous (i.v.) reovirus does not 13. However, we demonstrated that i.v. virus could achieve significant activity by conditioning the host with immune modulators (IL-2/Treg depletion or cyclophosphamide [CPA])13,

21, 22

, or by conditioning the tumor vasculature for

© 2014 The American Society of Gene & Cell Therapy. All rights reserved

ACCEPTED ARTICLE PREVIEW

increased reovirus localization/replication following i.v. delivery9, 23. In addition, we

12, 26, 34, 35

,

and others36, 37, have successfully used carrier cells of different types, loaded ex vivo, to protect viruses from neutralization and chaperone them into tumors 19.

However, translation of

strategies which require in vitro expansion of carrier cells, which are subsequently loaded with a replicating oncolytic virus, prior to intravenous (i.v.) delivery, are currently expensive and complex from a regulatory perspective. From our ongoing clinical program, we have shown in a Phase Ib, biological endpoint clinical

t

study (REO13) that, following i.v. injection of reovirus prior to planned resection of colorectal

rip

cancer liver metastases, reovirus could be specifically detected in patient tumors at the time of

us c

surgery, in spite of the presence of NAb in the circulation at baseline in all patients 38. Moreover, The REO13 study also demonstrated that, after systemic reovirus administration, replication

an

competent virus could be retrieved from mononuclear cells, granulocytes and platelets within

m

patient blood, but not from the plasma. These data suggested that, although free reovirus is

d

rapidly neutralized by NAb following i.v. injection, it may be successfully transported to tumors Therefore, based on these clinical observations, we

ep te

via protective carriage by blood cells.

hypothesized that i.v. injection of reovirus results in rapid adhesion to, or infection of, blood cells

A cc

which can protect the virus from neutralization, including by NAb; moreover, it may be possible to stimulate specific cell compartments prior to i.v. virus injection such that virus adhesion occurs selectively to a population of cell carriers which can traffic, and deliver virus, to tumors. Consistent with this hypothesis, we show here that, following i.v. administration into mice, reovirus associated predominantly with CD11b+ cells, and that stimulation of this compartment with GM-CSF prior to reovirus delivery significantly enhanced anti-tumor therapy by an immunemediated mechanism dependent on both NK cells and monocytes/macrophages. Interestingly, however, GM-CSF conditioning therapy was most effective in the presence of pre-existing NAb. Our data are significant in that they show that pre-existing NAb to an oncolytic virus may actually be exploited for systemic delivery to tumors and extend the previous use of ex vivo

© 2014 The American Society of Gene & Cell Therapy. All rights reserved

ACCEPTED ARTICLE PREVIEW

loaded cell carriers to a new concept of directed in vivo cell loading, thereby representing a readily testable, and translatable, method to enhance systemic delivery of oncolytic viruses in patients.

RESULTS Systemic delivery of reovirus in the presence of neutralizing antibodies Similar to the findings of our biological endpoint clinical study 38, 2 minutes after i.v. injection of reovirus, infectious virus was recovered from both plasma and cells of non reovirus-immune

rip

t

mice, but could only be recovered from the cellular fraction in virus-immune mice (Fig. 1A). By

us c

30 minutes after infusion, although very low levels (non-immune), or no (virus-immune), virus was recovered from plasma, infectious virus was still associated with the cell fraction, with

an

significantly increased levels recoverable from blood cells from virus-immune mice compared to

m

mice with no NAb (p