Gene Therapy and Oncolytic Viruses


Introduction

Genetic alterations are the driving force behind cancer development and progression. It follows that cancer could potentially be treated by correcting these alterations using gene therapy or by agents that kill cells by mechanisms based on these genetic alterations. Approaches of these kinds have a number of potential advantages. Vectors and viruses can be engineered in countless ways to achieve specificity and potency ( Table 54-1 ) and can be designed and tested using tools that are routine in contemporary molecular biology laboratories. For these reasons, a dazzling array of creative concepts has been described over the past 20 years. However, few of these have been tested extensively in the clinic, and none has yet completed a successful Phase III clinical trial in the United States or Europe. Therefore, gene therapy and oncolytic viral therapy have not yet lived up to their promise and have not yet entered the mainstream of medical oncology, by any means. In this chapter, we discuss the potential and challenges of these novel technologies. We focus on the main strategies that have been evaluated clinically. These include the design of agents that kill cancer cells by gene replacement or by disruption of oncogenic signaling pathways. For example, therapeutic agents have been developed that reintroduce the wild-type p53 tumor suppressor gene or destroy RNA encoding oncogenic K-RAS. The delivery of toxic genes and genes that convert prodrugs into toxic metabolites has undergone extensive clinical testing and has produced exciting results. Furthermore, we highlight strategies that aim at modulating the immune response in order to achieve anticancer effects. Finally, we discuss the basic and clinical aspects of the use of replication-competent viruses, either in their natural configuration or genetically modified, to selectively kill cancer cells ( Table 54-2 ).

Table 54-1
Gene Therapy and Oncolytic Viruses—Basic Concepts
Class Example
Vectors Viral Naturally cancer-selective viruses Reovirus, Newcastle disease virus (NDV), vesicular stomatitis virus (VSV), measles, vaccinia virus
Engineered viruses Adenovirus: ONYX015; herpes simplex virus (HSV): OncoVEXGM-CSF; vaccinia virus: JX-594
Nonviral Bacteria Listeria monocytogenes : CRS-207
Plasmids Plasmid encoding IL-12
RNAi siRNA against RRM2: CALAA-01
Strategies to achieve tumor killing Cell lysis/killing via viral replication All replication-competent viruses
Direct targeting of tumor genetics: knockdown of oncogenes or essential pathway components, reexpression of tumor suppressor genes KRasG12D: siG12D LODER;
p53: AdCMV-p53
Delivery of cytotoxic or pro-apoptotic genes Bcl-2; Survivin
Delivery of immune-stimulatory genes GM-CSF: JX594, OncoVEXGM-CSF; TNF: TNFerade
Activation of innate and adaptive immunity, induction of memory OncoVEXGM-CSF, JX594
Delivery of anti-anti angiogenic genes and enzymes Anti-FLT1 ribozyme
Delivery of prodrug converting enzymes Thymidine kinase, carboxylesterase, cytosinedeaminase
Delivery of transporters Sodium-iodide symporter (NIS)
Chemosensitization by viral replication All replication-competent viruses
Strategies to achieve tumor selectivity Vector size: preferential exit from leaky tumor vasculature (fenestrations of ∼100-400 nm); enhanced permeability retention effect (EPR) CALAA-01 (70-nm diameter); vaccinia virus (200 nm)
Vector engineering to match specific tumor genetics Rb: adenovirus Delta24
Targeting cell surface receptors overexpressed by cancer cells for delivery Transferrin receptor: CALAA-01 siRNA delivering nanoparticles with transferrin as a targeting ligand
α v β-Integrin: Adenovirus RGD
Deletion of viral genes inducing S-phase, and/or other genes required for replication in quiescent cells Adenovirus: E1A and E1B deletions;
vaccinia: thymidine kinase deletion
Use of tumor- and/or tissue-specific promoters Adenovirus CG0070: E2F-1 driven expression of E1A
FLT1, Fms-like tyrosine kinase 1, VEGFR1; RGD, Arg-Gly-Asp; RRM2, ribonucleotide reductase subunit M2.

Table 54-2
Cancer-Selective Viruses in Clinical Trials
Virus Genus Virus Product Name/Strain Genome Basis of Selectivity (Other) Genetic Modification(s) Clinical Phase Route of Administration Ref.
Enterovirus: Coxsackievirus A21, CVA21 CAVATAK ssRNA + High levels of CAV21 receptors (ICAM1, DAF) expressed by cancer cells None Phase II malignant melanoma Intratumoral NCT00832559
Newcastle Disease Virus (NDV) NDV-HUJ ssRNA Defective IFN response in transformed cells None Phase I/II
glioblastoma, sarcoma, neuroblastoma
IV NCT01174537
Parvovirus H-1
(apathogenic rat virus)
ParvOryx01 ssDNA Defective IFN response in transformed cells; viral replication only in proliferating cells None Phase I/IIa
glioblastoma multiforme
Intratumoral/ intracerebral or IV/intracerebral NCT01301430
Reovirus dsRNA PKR suppression by Ras and oncogenic EGFR None
Respiratory enteric orphan virus (REOLYSIN) Phase II NSCLC;
Phase II prostate cancer;
Phase II colorectal cancer;
Phase II pancreatic adenocarcinoma;
Phase II melanoma;
Phase II lung squamous cell carcinoma;
Phase II head and neck carcinoma;
Phase III head and neck carcinoma
IV

NCT00861627;
NCT01619813;
NCT01622543;
NCT00998322;
NCT00984464;
NCT00998192;
NCT00753038;
NCT01166542

Wild-type reovirus Phase II fallopian tube cancer,
ovarian cancer
Primary peritoneal cavity cancer;
Phase II pancreatic cancer
IV NCT01199263;
NCT01280058
Seneca-valley virus/ senecavirus Senecavirus-001, NTX-010 ssRNA + Phase II NSCLC IV NCT01017601
Adenovirus CG0070 DNA E2F-1 promoter-driven expression of E1A GM-CSF + Phase II/III
Non–muscle-invasive bladder cancer
Bladder instillation NCT01438112
Herpes Simplex Virus OncoVEX GM-CSF DNA ICP34.5 -
(ICP34.5 related to eIF2 phosphatase regulatory subunit GADD34, involved in translational regulation)
ICP47 -
(immune stimulating, as ICP47 blocks antigen processing in HSV-infected cells),
GM-CSF +
(immune stimulating)
Phase III
Malignant melanoma
Intratumoral NCT01368276,NCT00769704
Vaccinia DNA Natural tumor selectivity: permeable tumor vasculature; selective replication in metabolically active tumor environment
Replication activated by epidermal growth factor receptor EGFR/Ras pathway signaling; cancer cell resistance to IFNs
GL-ONC1, (attenuated Lister strain ) Thymidine kinase - Luciferase-GFP fusion +
LacZ +
GusA +
Hemagglutinin -
Phase I/II
Peritoneal carcinomatosis
Intraperitoneal NCT01443260
JX-594,
(Wyeth strain)
Thymidine kinase - GM-CSF,
B-galactosidase
Ph2
Hepatocellular carcinoma;
Ph1/2 colorectal carcinoma
IV NCT00554372,
NCT01171651,
NCT01387555,
NCT01394939
Phase I/II, Phase II, or Phase III clinical trials. June 2012. Clinicaltrials.gov .
EGFR, epidermal growth factor receptor; GFP, green fluorescent protein; GusA, β-glucuronidase; ICAM1, intercellular adhesion molecule 1; IFN, interferon; IV, intravenous; DAF, decay accelerating factor; GM-CSF, granulocyte macrophage colony-stimulating factor; LacZ, β-galactosidase; NSCLC, non–small-cell lung cancer; PKR, protein kinase R.

Killing Cancer Cells by Gene Replacement and Gene Knockout

Gene Replacement

Cancers develop through loss of function of key regulatory genes known as tumor suppressors as well as through activation of proto-oncogenes. Loss of p14, p16, p53, or PTEN occurs at high frequencies in most tumor types. Loss of Adenomatous Polyposis Coli ( APC ) occurs in the majority of colorectal cancers, and loss of RB is frequent in small-cell lung cancers, among many other examples. Mutations or deletions inactivate products of these genes, or their expression is suppressed by hypermethylation. Clonal evolution of tumors depends on loss of these functions: replacement of functional versions of these genes should therefore reverse this process. Indeed, when tumor suppressors are reexpressed in tumor cells by gene delivery, tumor cells die or growth is arrested. This has been shown for APC, p16, p21, p27, RB, p14ARF, p53, PTEN, APC, and BRCA1, BRCA2, among others.

In contrast, normal cells receiving additional copies of these genes appear to be unaffected, at least in the cases that have been tested so far. This has been documented most clearly for p53: delivery of this tumor suppressor to normal bronchial epithelial cells had no effect on cell growth, suggesting a therapeutic window of more than 2 orders of magnitude. This may be because tumor cells have additional defects that make them more sensitive to the effects of reexpressing p53. For example, almost all tumor cells have a defect in the RB checkpoint and therefore are less able to undergo growth arrest. Normal cells, in contrast, undergo G 1 arrest when p53 is active, and this can protect them from apoptosis. In addition, the negative regulator of p53, Mdm2, is itself a p53 effector. In tumor cells lacking p53, mdm2 levels are often low. In normal cells, Mdm2 constantly degrades p53 and maintains low p53 activity. Therefore, p53 expressed ectopically in normal cells is degraded, whereas in cancer cells it is relatively stable. In addition, p53 promotes a bystander effect on uninfected tumor cells, possibly through antiangiogenesis, secretion of soluble proapoptotic proteins, and immune upregulation. In agreement with these theoretical considerations, clinical safety and some evidence for anti-tumor activity of an adenoviral vector expresses wild-type p53 under the control of a Rous-sarcoma-virus promoter (Gendicine) have been demonstrated and led to approval of this agent in China for use in head and neck cancer. Fever and flu-like symptoms were the main adverse events; no severe side effects occurred. Long-lasting responses with this agent were observed in studies in patients with head and neck cancer. Very high response rates, including 64% complete responses, were observed following intratumoral injection of Gendicine in combination with radiation therapy. However, a similar virus that delivers wild-type p53 under the control of a constitutively active CMV promoter (AdCMV-p53; Advexin) failed to complete a Phase III clinical trial successfully in the United States, and development of this agent stopped in 2008. This example illustrates the fact that poor delivery systems and inefficient access to tumor cells have limited the potential of gene-replacement therapy for treating cancer, despite compelling theoretical arguments for its development.

Gene Knockout

Antisense

Activation of proto-oncogenes such as Ras, B-Raf, Myc , or EGFR through various mechanisms is a key factor in carcinogenesis. For example, mutations of the Ras oncogene occur in many types, including pancreatic cancer, where about 90% of cases carry a mutation of the K-ras gene. Knockout of oncogenes driving tumor development in transgenic mouse models of cancer can lead to complete regression, confirming their potential value as drug targets. Antisense technology has been studied as a strategy to knock down expression of oncogenes. This approach is based on the possibility of inhibiting transcription of a particular mRNA by transfecting short double-stranded DNA oligonucleotides into target cells that bind and inactivate the target mRNA in a sequence-specific manner. The development of oligonucleotides with modified DNA backbone that increases the stability of the molecule in vivo allowed for the development of clinical protocols involving intravenous application of antisense molecules targeting, for example, mutant Kras or the anti-apoptotic genes BCL-2 and Survivin. Clinical trials with oblimersen sodium (G3139), an antisense oligonucleotide directed against BCL-2, have demonstrated increased survival of patients with advanced melanoma in combination with chemotherapy compared to chemotherapy alone. An interesting variant of the antisense approach that is currently being tested clinically is GRN163L, a lipid-modified 13-mer DNA-oligonucleotide that acts as a telomerase RNA template antagonist.

RNAi

The discovery of the possibility of silencing gene expression in mammalian cells with high efficiency using RNA interference (RNAi) has spurred renewed interest in the gene knock-down as a therapeutic strategy. This approach, first described in the worm Caenorhabditis elegans , takes advantage of a cellular gene silencing machinery that involves the RNA-induced silencing complex (RISC) and degrades double-stranded RNAs with high efficacy. The introduction of short RNAs with complementary sequence to any cellular transcript activates this mechanism. RNAi can silence target genes with high efficacy. However, off-target effects that result in sequence-specific though unpredictable knock-down of additional genes represent a potential problem that has not been fully resolved at this point. Nevertheless, novel targeting and delivery mechanisms have been developed that hold the promise of therapeutic application of RNAi-based agents in cancer. For instance, a recently reported nanoparticle-based delivery system shows great promise for systemic delivery of siRNA. These synthetic nanoparticles consist of a cyclodextrin-based polymer, a human transferrin protein (TF) targeting ligand displayed on the exterior of the nanoparticle to engage TF receptors (TFR) on the surface of the cancer cells, a hydrophilic polymer (polyethylene glycol [PEG] used to promote nanoparticle stability in biological fluids), and siRNA designed to reduce the expression of the Ribonucleotide Reductase subunit M2 (RRM2). Tumor-specific targeting is achieved via the interaction of the TF targeting ligand with TFR, which is known to be upregulated in malignant cells. Moreover, the size of these nanoparticles, about 70 nm in diameter, favors their exit from the bloodstream in leaky tumor vasculature and accumulation in the tumor bed via the enhanced permeability and retention (EPR) effect. RRM2, an enzyme catalyzing a rate-limiting step in DNA synthesis, is an established anticancer target. These particles (CALAA-01), currently in Phase I clinical studies, have been administered systemically to humans and have shown specific gene inhibition—a reduction in both mRNA and protein.

Another well-established anticancer target is the KRAS oncogene. Most pancreatic adenocarcinomas are caused by a somatic mutation in KRAS , most commonly KRAS G12D . In vitro and animal studies suggest that selective inhibition of the mutant, but not the WT KRAS expression, leads to apoptosis of targeted cancer cells. An siRNA specifically targeting mRNA encoding KRAS G12D, coated by a biodegradable polymeric matrix, siG12D LODER (Local Drug EluteR), is currently in Phase 0/I to evaluate its safety and tolerability. Apart from these two particular RNAi therapeutics, at least another six are currently undergoing clinical testing, thanks to recent advancements in the understanding of RNAi biology and in the areas of RNAi specificity, stability, and delivery. Most are delivered employing synthetic carriers, such as cationic liposomes, anionic liposomes, and polymeric particles. As more attention is focused on safe and effective methods for delivering siRNA to tumors, the clinical value of this approach will likely increase dramatically. siRNA-based therapy has the potential of addressing undruggable oncogene targets, combinatorial approaches to cancer cell killing, and drug resistance, but this potential will not be realized until the challenges of delivery and uptake have been fully addressed.

Killing Cancer Cells by Delivering Prodrug Converting Enzymes and Other Enzymes

For more than 60 years chemotherapeutic agents have been used for the treatment of cancer. However, their use is often limited by damage to normal cells, drug resistance, and low chemical stability. One strategy to overcome limitations of classical chemotherapeutic agents is the use of prodrugs. A prodrug is a fairly nontoxic compound that needs to be transformed before acting as a pharmacon. Such a transformation can be catalyzed by endogenous enzymes, in which case tissue distribution of such endogenous enzymes dictates where the active pharmacon is produced. Alternatively, gene-directed enzyme prodrug therapy (GDEPT) can specifically deliver such enzymes to diseased cells where they can activate nontoxic prodrugs into toxic agents. Progress in this field has been reviewed recently by Duarte and colleagues.

Thymidine Kinase

The first GDEPT system described was the thymidine kinase gene of the herpes simplex virus (HSV-TK) in combination with the prodrug nucleoside analog ganciclovir. The enzyme thymidine kinase (TK) is naturally present in bacteria, viruses, and mammals, where it is involved in the salvage pathway of nucleotide biosynthesis. Thus, high TK activity is found in proliferating cells such as cancer cells. TK also converts ganciclovir to ganciclovir monophosphate, which is subsequently converted by cellular kinases into the toxic ganciclovir triphosphate nucleotide. The HSV-TK is three orders of magnitude more efficient than any human kinase catalyzing this first activation step. Hence, several gene therapy approaches combining HSV-TK and ganciclovir have been developed.

For instance, retroviral vectors were used to deliver HSV-TK in a brain tumor model. Because retroviruses integrate only in proliferating cells, gene delivery and expression would be tumor selective in the context of normal, nonproliferating brain cells. A further advantage of this approach was thought to result from the “bystander effect,” the killing of uninfected neighboring cells that occurs when HSV-TK–expressing cells are exposed to ganciclovir, which can be observed in vitro and in vivo. To increase transduction of target cells with retroviruses, virus-producing cells (VPCs) were used to inoculate target tumors instead of virus suspension. Preclinical studies in a rat glioma model demonstrated that this delivery of a retrovirus expressing HSV-TK resulted in high transduction levels and frequent tumor regressions following ganciclovir administration. Initial clinical studies were promising and demonstrated in responses of small glioblastomas following VPC injection. However, when standard therapy (surgical resection and radiotherapy) was compared to standard therapy plus injection of retrovirus-producing cells in a Phase III trial, no differences in progression-free and overall survival were observed between the two groups. Smaller, preoperative studies suggested that lack of transduction of tumor cells is the dominant reason for the failure of this approach. Despite the disappointing results of this trial, it represents an early and innovative effort to use gene therapy to kill cancer cells selectively.

Cytosine Deaminase

In contrast to thymidine kinase, the enzyme cytosine deaminase (CD) is not present in mammalian cells, but in several bacteria and fungi. It catalyzes the amidine hydrolysis of cytosine to uracil and ammonia, and several cytosine analogs such as halogenated cytosines are substrates as well. One such substrate is the prodrug 5′-fluorocytosine (5′-FC) which is activated by CD to 5′-fluorouracil (5′-FU). 5′-FU kills cells mainly by inhibition of thymidilate synthetase and by incorporation into DNA. The safety and efficacy of a CD and 5′-FC prodrug therapy are currently being assessed in Phase I/II trials in which 5′-FC and a recombinant Bifidobacterium longum (a live bacterium normally found in the digestive tract) that has been modified to produce CD are given orally to patients with various solid tumors.

You're Reading a Preview

Become a Clinical Tree membership for Full access and enjoy Unlimited articles

Become membership

If you are a member. Log in here