82
Views
14
CrossRef citations to date
0
Altmetric
Review

Update on oncolytic viral therapy – targeting angiogenesis

, &
Pages 1031-1040 | Published online: 31 Jul 2013

Abstract

Oncolytic viruses (OVs) have the ability to selectively replicate in and lyse cancer cells. Angiogenesis is an essential requirement for tumor growth. Like OVs, the therapeutic effect of many angiogenesis inhibitors has been limited, leading to the development of more effective approaches to combine antiangiogenic therapy with OVs. Angiogenesis can be targeted either directly by OV infection of vascular endothelial cells, or by arming OVs with antiangiogenic transgenes, which are subsequently expressed locally in the tumor microenvironment. In this review, we describe the development and targeting of OVs, the role of angiogenesis in cancer, and the progress made in arming viruses with antiangiogenic transgenes. Future developments required to optimize this approach are addressed.

Introduction to oncolytic viral therapy

Cancer remains a leading cause of death despite incremental advances in surgery, chemotherapy, and radiotherapy. This has led to the development of new treatment strategies. With increasing knowledge of the genetic defects and molecular basis of cancer, gene therapy has become an attractive option, although clinical trials to date have shown only moderate efficacy. Since cancer usually results from a combination of genetic defects, strategies to eradicate cancer cells themselves are likely to have a greater efficacy than attempts to correct each genetic defect individually. The ideal vector has tumor-selective oncolytic properties in that, as well as delivering gene therapy, it can selectively enter and destroy cancer cells, without affecting surrounding normal cells. It should also be safe to administer, have minimal side effects, be easy to mass-produce and purify, and be genetically stable in storage and use.

While nonviral vectors, including naked DNA or DNA packaged in liposomes and dendrimers, have been used in cancer gene therapy,Citation1 it is viruses that have been recognized to be the most efficient means currently available for the delivery of therapeutic genes. The evolution of viruses over millions of years has resulted in their ability to evade our immune system, infect, and replicate efficiently in human cells before causing their death to facilitate viral spread.Citation2 While the viral vectors developed initially were nonreplicating, it is the ability of viruses to replicate, so amplifying a small input dose to maximize therapeutic effect, that really sets them apart from other vectors. Interest has therefore shifted to the development of oncolytic viruses (OVs). These are replication-selective in that they have the ability to replicate in and cause death of tumor cells, while sparing normal cells.Citation3 This property may be inherent or enhanced through genetic engineering and the viruses armed to deliver therapeutic transgenes, such as angiogenesis inhibitors.

The use of viruses for cancer treatment is not a new concept since, at the beginning of the 20th century, it was observed that a flu-like disease resulted in decreased tumor load in a patient with leukemia and that rabies vaccination was followed by regression of cervical cancer.Citation4 OVs can be engineered to express foreign genes and, therefore, deliver gene therapy on a continuous basis to augment their therapeutic effect. This combination of mechanisms means that resistance, which often limits standard chemotherapy, is less likely to occur.Citation5

Barriers to oncolytic viruses

Over the last two decades, thousands of OVs have been developed, resulting in many clinical trials but few examples of clinical efficacy.Citation6 Replication-selective oncolytic adenovirus is the most well-researched, and dl1520 (or H101 in the People’s Republic of China) was approved as the world’s first oncolytic virotherapy for the treatment of head and neck cancer.Citation7 This has also been administered by intratumoral injection into patients with locally advanced pancreatic tumors in Phase I/II trials.Citation8 Although treatments were well tolerated, efficacy was poor. The main barriers to oncolytic viral therapy have been recognized as viral attenuation caused by the genetic engineering to improve tumor selectivity, the effects of the tumor microenvironment, and the host immune response.Citation9,Citation10

Tumor selectivity

The first generation of OVs were inherently tumor selective in their replication and included Newcastle virusCitation11 and reovirus.Citation12,Citation13 Second generation OVs were engineered to limit replication to tumors by inserting tumor-specific promoters, such as the prostate-specific antigen (PSA), upstream of the genes required for viral replication.Citation14 The deletion of genes, such as thymidine kinase (TK), that are themselves required for viral replication in normal cells, but not in tumor cells also enhances tumor-selectivity. Many other strategies have been developed in order to increase the selectivity of OVs.Citation10 However, the efficiency of cell lysis and cell-to-cell transfer of OVs alone in vivo is often poor since these modifications can result in the attenuation of antitumor potency.

Tumor microenvironment

The importance of the interaction between cancer cells and their environment is well recognized.Citation15 Tumor development has many similarities with wound healing, as in tumor development, there is also a persistent inflammatory response, with many of the same cytokines released from the fibroblasts, which attract immune cells, including macrophages, which in turn, release further cytokines that promote angiogenesis.Citation16 Considering these complex interactions, it is not surprising that OVs targeting the tumors alone have had limited efficacy.

Collagen in the extracellular matrix (ECM) has been found to be a physical barrier to the spread of OVs within tumors. It has been shown that inducing collagen degradation with a bacterial collagenase improved the spread and efficacy of an oncolytic herpes simplex virus.Citation17 The construction of an oncolytic adenovirus expressing the collagenase matrix metalloproteinase (MMP)-9 also increased viral spread in human pancreatic and lung cancer models.Citation18 The inevitable hypoxic environment in most tumors has also been found to attenuate oncolytic adenoviral therapy.Citation19 OVs can also increase the vascular permeability in tumors, stimulating angiogenesis.Citation20 Antiangiogenic therapy may itself reduce the innate immune response by stabilizing the tumor vasculature and decreasing the immune cell infiltration of tumors.Citation21

Host immune response

The host immune response to virus-infected tumors may be the most significant limitation to oncolytic viral therapy. Soon after virus delivery to tumors, an innate immune response is observed, with recruitment of neutrophils, natural killer cells, and macrophages.Citation22 Inhibition of the innate immune response may enhance the efficacy of OVs.Citation9 However, the step-wise deletion of virus virulence genes that normally cause evasion of the host immune response has been used to improve OV tumor selectivity. The host immune response is a double-edged sword for OV-based therapeutics. On the one hand, a vigorous host immune response to the OV can result in rapid viral clearance before the virus is able to exert a therapeutic effect or even result in tumor progression due to immunosuppression. The efficacy of multiple injections of the same virus may be further limited by a neutralizing antibody response.Citation23 However, the host immune response may be critical to the efficacy of oncolytic viral therapy. This may be mediated via innate immune effectors, adaptive antiviral immune responses eliminating infected cells, or adaptive antitumor immune responses.Citation24

Choice of transgene expression by oncolytic viruses

The choice of genes possible for cancer gene therapy is vast. The main mechanisms of action of potential therapeutic genes are those considered to be corrective, immunomodulatory, and cytoreductive, all of which have been delivered by OVs.

Corrective genes

Unregulated cell growth in cancers is caused by mutations in oncogenes or tumor suppressor genes (TSGs). Therefore, inhibiting oncogenes or upregulating TSGs may restore normal cell growth and division. The most commonly targeted TSG is p53, as mutations of this gene are estimated to be found in over half of all malignancies.Citation25,Citation26 In 2003, a recombinant, nonreplicating adenovirus expressing p53 (rAd-53 or Gendicine™; Schenzhen SiBiono Gene Tech Co, Ltd, Schenzhen, People’s Republic of China) was approved by the State Food and Drug Administration of China for the treatment of head and neck squamous cell carcinoma and became the first licensed gene therapy product in the world. Another commonly mutated TSG is p16, which has been delivered by oncolytic adenovirus to treat gastric cancer xenografts.Citation27 However, since most cancers result from defects in several genes, this strategy has not been effective, and focus has moved to targeting signaling pathways.

Immunomodulatory genes

As discussed previously, the host’s innate immune response to viruses can lead to their rapid clearance thus limiting transgene expression and antitumor efficacy. The adaptive immune response may preclude repeated virus administration through the formation of circulating antibodies. Many malignant cells express tumor-associated antigens (TAAs). Recognition of TAAs by antigen-presenting cells leads to the activation of TAA-specific CD8+ cytotoxic T lymphocytes (CTLs). These in turn, cause tumor cell death through direct lysis. Tumors avoid CTL destruction by limiting TAA presentation and reducing the expression of major histocom-patibility complex class I (MHC I), which is also required for antigen recognition.Citation28 Cancer gene therapy can enhance tumor cell recognition through increased TAA presentation or upregulated MHC I expression.Citation29 Cytokine delivery by gene therapy, such as oncolytic adenovirus- or vaccinia virus (VV)-delivered interferon β, has shown potential in modulating the host immune response to improve tumor clearance.Citation30,Citation31 Granulocyte-macrophage colony-stimulating factor (GM-CSF)-armed OVs (herpes simplex virus [HSV] and VV) have shown encouraging results in clinical trials,Citation32,Citation33 although tumor-derived GM-CSF has also been recently demonstrated to drive the progression of cancer.Citation34,Citation35 The mechanism is thought to be similar in both situations, with GM-CSF suppressing T cell immunity, allowing OVs and tumor cells, respectively, to evade the host immune response.

Cytoreductive genes

Cytoreductive therapy targets cancer cells either directly or indirectly. Methods include gene-directed enzyme prodrug therapy (GDEPT) (also called suicide gene therapy) and antiangiogenic therapy. Acting in cancer cells only, GDEPT expresses a gene encoding an enzyme that converts a prodrug into a potential cytotoxin. The cytotoxin is produced exclusively within the target tumor to cause tumor cell death and regression.Citation36 One example is an oncolytic adenovirus expressing a fusion protein that converts 5-fluorocytosine into the commonly used chemotherapeutic agent 5-fluorouracil.Citation37 Virus-delivered GDEPT has entered Phase I clinical trials.Citation38,Citation39 The requirement of angiogenesis for the growth of all tumors has led to the discovery of a wide variety of angiogenesis inhibitors. This strategy will be now be explored in detail.

Introduction to angiogenesis

Angiogenesis is the growth of new capillary blood vessels from existing vessels. This is necessary for the growth of all tumors beyond 2 or 3 mm in diameter.Citation40 Folkman proposed that the acquisition of an “angiogenic switch” was necessary for tumor growth and metastasis.Citation41 He first recognized this as an important factor in the development of other chronic diseases, such as atherosclerosis, chronic liver disease, and rheumatoid arthritis. This has led to the development of a huge range of antiangiogenic therapies, some of which are now in widespread clinical use, including small molecules and monoclonal antibody inhibitors.Citation42,Citation43 Viruses have been recognized as offering the possibility of targeted delivery of angiogenesis inhibitors, providing local expression of these proteins on a continual basis in order to maximize efficacy and limit side effects. This review will focus on the angiogenesis inhibitors delivered by replicating OVs for cancer therapy, forming an update from our previous review on this topic.Citation44

Specific importance of angiogenesis in cancer

Angiogenesis is regulated by the balance of angiogenic growth factors and inhibitors, which are released from endothelial cells (ECs), monocytes, platelets, and smooth muscle and tumor cells.Citation45 The normal physiological situation in solid organ vasculature is an excess of inhibitors. When an excess of growth factors is present, as is frequently the case in tumors, the balance is tipped in favor of angiogenesis.

Vascular endothelial growth factor (VEGF) and other growth factors released from tumors bind to receptors on both the endothelial cells of nearby blood vessels and circulating bone marrow-derived epithelial progenitor cells, resulting in their activation, proliferation, and production of enzymes.Citation46 The resulting enzymes dissolve holes in the basement membrane of the surrounding blood vessels, allowing proliferating ECs to migrate out toward the tumor. MMPs produced by ECs dissolve the surrounding stroma, allowing the ECs to advance toward the tumor, using integrins including αvβ3 and αvβ6 to direct them.Citation47 The ECs then remodel and form tubes, which connect into loops through the tumor mass, so forming complete blood vessels. Structural support cells, such as smooth muscle cells, then follow, although tumor blood vessels remain leaky and have a poorly formed basement membrane, two factors that have been proposed to aid the inherent tumor specificity of some viral vectors.

The development of angiogenesis inhibitors has become a broad and active area of cancer research.Citation45,Citation48Citation51 A wide range of angiogenesis inhibitors have been discovered and the most widely studied include protease inhibitors, tyrosine kinase inhibitors, chemokines, interleukins (ILs) (eg, IL-8, IL-12, IL-18), and proteolytic fragments of diverse molecules (eg, endostatin, angiostatin, vaculostatin, canstatin). These antiangiogenic molecules function in multiple ways, including inhibiting endothelial cell proliferation, migration, protease activity and tubule formation, as well as inducing apoptosis.

Antiangiogenic agents offer lower toxicity than most conventional chemotherapy, allowing long-term use.Citation48,Citation52 VEGF is the most commonly targeted angiogenic growth factor, and the first angiogenesis inhibitor to be licensed in Europe was bevacizumab. This class I inhibitor is an antibody that specifically blocks VEGF and was licensed in January 2005 for the treatment of metastatic colorectal carcinoma, following its success in clinical trials.Citation53 However, resistance has been seen in patients with other tumor types, where multiple angiogenic factors may be produced by primary tumors.Citation54

Despite great promise, the results obtained with the use of these peptide inhibitors alone in clinical trials have been disappointing, and regimes combining angiogenesis inhibitors with standard chemotherapeutic regimes are often required.Citation42,Citation43 Many angiogenesis inhibitors are not directly cytotoxic to tumor cells, but need to be expressed on a continuous basis to inhibit ECs effectively. Initial efforts focused on the targeting of VEGF- and tumor-derived VEGF-signaling. However, resistance was observed, and a greater understanding of the mechanisms of drug resistanceCitation50 and the concept that sustained local delivery of angiogenesis inhibitors to tumors may be more effective has led to the development of antiangiogenic cancer gene therapy.Citation45

Targeting endothelial cells with oncolytic viruses

Some OVs display an innate ability to infect ECs in tumors, while sparing those in normal vessels.Citation55 Intravenous delivery of a vesicular stomatitis virus in a murine colorectal carcinoma xenograft model showed direct infection of ECs. This induced neutrophil infiltration, leading to microclot formation with tumor-associated vasculature, resulting in a large bystander effect of cell death within the tumor.Citation56 An HSV delivered intravenously in a murine ovarian carcinoma model has also been found to specifically infect tumor-associated ECs, causing cell death while sparing the ECs in normal organs.Citation57 In a Phase I clinical trial, intravenous delivery of oncolytic VV engineered to target cells with activation of the Ras/mitogen-activated protein kinase (MAPK) pathway was found to selectively infect tumor-associated ECs, sparing normal ECs.Citation58 This provides a useful platform for the further development of OVs armed with therapeutic transgenes in the future.

Arming oncolytic viruses with angiogenesis inhibitors

While nanoparticles, liposomes, and naked plasmid DNA electroporation have all been used to deliver antiangiogenesis gene therapy, it is viruses that are the most promising vectors for the delivery of angiogenesis inhibitors.Citation59 The ability of OVs to infect and selectively amplify the input dose of virus in the target tumor has been exploited to address some of the limitations of nonreplicating viruses.Citation23,Citation60 OVs can selectively target and kill tumor ECs as well as tumor cells, although there is evidence that they can themselves increase vascular permeability in tumors, stimulating angiogenesis.Citation20 The combination of antiangiogenic therapy delivered by an OV may prevent this through reduction of the host immune response, by stabilizing tumor vasculature and decreasing immune cell infiltration.Citation21 The selective expression of angiogenesis inhibitors in the tumor microenvironment prevents further tumor growth, allowing the viral progeny produced through replication to spread through tumors, infecting and inducing lysis of cancer cells in order to achieve tumor clearance.

Since our last review on this subject was published,Citation44 there have been 17 new OVs armed with antiangiogenic genes reported in animal models, making a total of 32 studies in the literature (). There are still no clinical trials reported nor registered to date.

Table 1 Oncolytic viruses expressing inhibitors of the VEGF pathway

Table 2 Oncolytic viruses expressing cytokines and chemokines

Table 3 Oncolytic viruses expressing other endogenous inhibitors

Targeting VEGF

VEGF has a key role in the signaling pathways that mediate angiogenesis, tumor growth, and metastasis. Monoclonal antibodies against VEGF are now in widespread clinical use in oncology. Since VEGF is highly expressed in many cancers, this pathway has been targeted by many OVs expressing angiogenesis inhibitors. The first reported OV expressing an angiogenesis inhibitor was a first-generation oncolytic adenovirus (ONYX-015, E1B55k, and E3B-deleted adenovirus) armed with a soluble VEGF-receptor inhibitor, sFlt-1, which was effective in an animal model of human colorectal cancer.Citation62 The combination of a second-generation oncolytic adenovirus (dl922/47, with E1ACR2 mutation and E3B deletion) and a nonreplicating adenovirus expressing Flk1-Fc, a soluble ectodomain of the VEGF receptor, was more effective than either virus alone.Citation63 Another E1B55 kDa-deleted oncolytic adenovirus was effective in a human colorectal model by expressing vascular endothelial growth inhibitor (VEGI).Citation64 Ad-Δ7B-KOX (E1B19 kDa and E1B55 kDa gene-deleted), an oncolytic adenovirus expressing a VEGF promoter targeted artificial zinc-finger protein inserted into the E3 gene, reduced VEGF expression, and increased the survival of animals bearing human glioblastoma xenografts.Citation65

More recently, a group has published results of both oncolytic adenovirusCitation66 (Ad5/3 serotype with hypoxia inducible factor [HIF]-promoter) and VVCitation67 (TK- and vaccinia growth factor-deleted) expressing VEGF-1-immunoglobulin (Ig), a soluble inhibitor that binds VEGF without inducing vascular EC mitogenesis.Citation68 VV has also been used to deliver a single chain antibody to VEGF, which was effective in a canine xenograft model.Citation69 Targeting the VEGF pathway has been effective in animal models and shows promise for translation to clinical studies in the future.

Targeting interleukins and chemokines

IL-24 is a good candidate for expression by OVs, as it is an effective antiangiogenic cytokine and as well, induces apoptosis and reduced growth in many tumors.Citation70,Citation71 Three different oncolytic adenoviruses have been engineered to express IL-24,Citation72Citation74 with coexpression of arrestin, and these were effective in a melanoma model.Citation74 IL-8 has been targeted, as it promotes angiogenesis, tumor growth, and metastasis.Citation75 An oncolytic adenovirus delivering small interfering ribonucleic acid (siRNA) against IL-8 was effective in a range of human xenograft models, including metastatic breast cancer.Citation76 IL-18 has also been identified as an angiogenic inhibitor and tumor suppressor.Citation77 An oncolytic E1b55 kDa-deleted adenovirus expressing IL-18 was effective in a human renal carcinoma xenograft model. IL-12Citation78 and chemokine platelet factor 4 (PF4)Citation79 are also potent antiangiogenic agents, and oncolytic HSVs armed with IL-12 or PF4 have shown promise in animal models.Citation80Citation82

Targeting matrix metalloproteinases

MMPs play a pivotal role in angiogenesis by degrading the stroma of the ECM that surrounds blood vessels, leading to EC proliferation, migration, and new capillary formation.Citation83 Tissue inhibitors of metalloproteinases (TIMP) have been developed for antiangiogenic therapy. The only reported virus expressing a TIMP is AdΔ24TIMP-3, an oncolytic adenovirus expressing TIMP-3. However, despite reducing levels of MMP-2, this did not lead to decreased tumor growth or improved survival in a human glioma model.Citation84

Other endogenous inhibitors

Endostatin has displayed the broadest anticancer spectrum of all endogenous inhibitors currently identified and inhibits migration of tumor cells and ECs as well as invasion of tumor cells.Citation85 The endostatin gene has been engineered to be expressed by oncolytic adenovirus, adeno-associated virus and HSV,Citation86Citation93 showing better efficacy than control or nonreplicating viruses expressing endostatin.

Angiostatin, a 38 kDa fragment of plasminogen, inhibits EC proliferation and migration as well as inducing apoptosis. The strategy of combining angiogenesis inhibitors that work through different pathways has been investigated since they tend to exhibit a low side-effect profile when compared with conventional chemotherapy. An oncolytic adenovirus expressing angiostatin has shown promise when delivered locally following systemic bevacizumab.Citation94 Endostatin and angiostatin have been found to act synergistically when used in combination, which led to the development of an endostatin-angiostatin fusion gene.Citation95 This fusion gene has been incorporated into oncolytic HSV, adenovirus, and VV.Citation96Citation98 We found that oncolytic VV expressing the fusion protein displayed superior efficacy over ONYX-015 in a head and neck cancer model.Citation99

Another fragment of plasminogen, kringle 5, inhibits EC proliferation more effectively than angiostatin.Citation100 Kringle 5 and a mutant kringle 5 (mK5, with leucine71 changed to arginine) have been delivered by a first-generation onco-lytic adenovirus (ONYX-015, E1B55 kDa-deleted).Citation101 The ZD55-mK5 virus significantly suppressed tumor growth and improved survival in a human colorectal xenograft model. ONYX-015 has also been used to express canstatin, a 24 kDa fragment of type IV collagen.Citation102 An oncolytic HSV expressing vasculostatin, a fragment of brain-specific angiogenesis inhibitor-1, has shown promise in a human pancreatic model.Citation103

Fibroblast growth factor (FGF) signaling is another important mediator of both EC and tumor cell migration essential in angiogenesis. A novel oncolytic HSV armed with a dominant-negative FGF receptor has been developed and has been shown to be more effective than its unarmed counterpart at inhibiting tumor growth and angiogenesis, in both human and mouse tumor models in vivo.Citation104

Future developments

Many different angiogenesis inhibitors have been used to arm OVs. Most have shown promise in animal models, but none have reached clinical trials. The major barriers limiting the efficacy of OVs are still tumor selectivity, the effect on the tumor microenvironment, and the host immune response to virus-infected tumor cells.Citation9 Many strategies are being developed to overcome these obstacles and to optimize OV delivery. Lessons should be learnt from standard chemotherapeutic regimes, which often combine multiple agents with different mechanisms of action. The same approach should be used with angiogenesis inhibitors, where less toxicity is usually observed when compared with most standard chemotherapy drugs.

Many antiangiogenic drugs have the ability to constrict abnormal leaky tumor vessels.Citation105 This ability to temporarily normalize tumor vasculature provides a therapeutic window during which systemic delivery of OVs may be improved. Clinical studies should concentrate on combining OVs with standard treatment regimes in order to enhance their efficacy.

Disclosure

The authors report no conflicts of interest in this work.

References

  • KanedaYTabataYNon-viral vectors for cancer therapyCancer Sci200697534835416630130
  • YoungLSSearlePFOnionDMautnerVViral gene therapy strategies: from basic science to clinical applicationJ Pathol2006208229931816362990
  • HawkinsLKLemoineNRKirnDOncolytic biotherapy: a novel therapeutic plafformLancet Oncol200231172611905600
  • KellyERussellSJHistory of oncolytic viruses: genesis to genetic engineeringMol Ther200715465165917299401
  • KirnDMartuzaRLZwiebelJReplication-selective virotherapy for cancer: biological principles, risk management and future directionsNat Med20017778178711433341
  • LiuTCGalanisEKirnDClinical trial results with oncolytic virotherapy: a century of promise, a decade of progressNat Clin Pract Oncol20074210111717259931
  • GarberKChina approves world’s first oncolytic virus therapy for cancer treatmentJ Natl Cancer Inst200698529830016507823
  • HechtJRBedfordRAbbruzzeseJLA phase I/II trial of intratumoral endoscopic ultrasound injection of ONYX-015 with intravenous gemcitabine in unresectable pancreatic carcinomaClin Cancer Res20039255556112576418
  • LiuTCKirnDGene therapy progress and prospects cancer: oncolytic virusesGene Ther2008151287788418418413
  • WongHHLemoineNRWangYOncolytic viruses for cancer therapy: overcoming the obstaclesViruses2010217810620543907
  • SinkovicsJGHorvathJCNewcastle disease virus (NDV): brief history of its oncolytic strainsJ Clin Virol200016111510680736
  • CominsCHeinemannLHarringtonKMelcherADe BonoJPandhaHReovirus: viral therapy for cancer ‘as nature intended’Clin Oncol (R Coll Radiol)200820754855418583112
  • ThorneSHHwangTHO’GormanWERational strain selection and engineering creates a broad-spectrum, systemically effective oncolytic poxvirus, JX-963J Clin Invest2007117113350335817965776
  • LiXZhangYPKimHSGene therapy for prostate cancer by controlling adenovirus E1a and E4 gene expression with PSES enhancerCancer Res20056551941195115753394
  • BissellMJRadiskyDPutting tumours in contextNat Rev Cancer200111465411900251
  • DvorakHFTumors: wounds that do not heal. Similarities between tumor stroma generation and wound healingN Engl J Med198631526165016593537791
  • McKeeTDGrandiPMokWDegradation of fibrillar collagen in a human melanoma xenograft improves the efficacy of an oncolytic herpes simplex virus vectorCancer Res20066652509251316510565
  • ChengJSauthoffHHuangYHuman matrix metalloproteinase-8 gene delivery increases the oncolytic activity of a replicating adenovirusMol Ther200715111982199017653103
  • PipiyaTSauthoffHHuangYQHypoxia reduces adenoviral replication in cancer cells by downregulation of viral protein expressionGene Ther2005121191191715690061
  • AghiMRabkinSDMartuzaRLAngiogenic response caused by oncolytic herpes simplex virus-induced reduced thrombospondin expression can be prevented by specific viral mutations or by administering a thrombospondin-derived peptideCancer Res200767244044417234749
  • KurozumiKHardcastleJThakurREffect of tumor microenvironment modulation on the efficacy of oncolytic virus therapyJ Natl Cancer Inst200799231768178118042934
  • ChioccaEAThe host response to cancer virotherapyCurr Opin Mol Ther2008101384518228180
  • ParatoKASengerDForsythPABellJCRecent progress in the battle between oncolytic viruses and tumoursNat Rev Cancer200551296597616294217
  • PrestwichRJHarringtonKJPandhaHSVileRGMelcherAAErringtonFOncolytic viruses: a novel form of immunotherapyExpert Rev Anticancer Ther1020088101581158818925850
  • ShiraishiKKatoSHanSYIsolation of temperature-sensitive p53 mutations from a comprehensive missense mutation libraryJ Biol ChemJan 22004279134835514559903
  • PengZCurrent status of gendicine in China: recombinant human Ad-p53 agent for treatment of cancers. Hum Gene TherSep200516910161027
  • MaJHeXWangWE2F promoter-regulated oncolytic adenovirus with p16 gene induces cell apoptosis and exerts antitumor effect on gastric cancerDig Dis Sci20095471425143119034663
  • OchsenbeinAFPrinciples of tumor immunosurveillance and implications for immunotherapyCancer Gene Ther20029121043105512522443
  • VileRGRussellSJLemoineNRCancer gene therapy: hard lessons and new coursesGene Ther2000712810680008
  • StermanDHRecioACarrollRGA phase I clinical trial of single-dose intrapleural IFN-beta gene transfer for malignant pleural mesothelioma and metastatic pleural effusions: high rate of antitumor immune responsesClin Cancer Res20071315 Pt 14456446617671130
  • KirnDHWangYLe BoeufFBellJThorneSHTargeting of interferonbeta to produce a specific, multi-mechanistic oncolytic vaccinia virusPLoS Med2007412e35318162040
  • HeoJReidTRuoLRandomized dose-finding clinical trial of oncolytic immunotherapeutic vaccinia JX-594 in liver cancerNat Med201319332933623396206
  • BreitbachCJBurkeJJonkerDIntravenous delivery of a multi-mechanistic cancer-targeted oncolytic poxvirus in humansNature201147773629910221886163
  • Pylayeva-GuptaYLeeKEHajduCHMillerGBar-SagiDOncogenic Kras-induced GM-CSF production promotes the development of pancreatic neoplasiaCancer Cell201221683684722698407
  • BayneLJBeattyGLJhalaNTumor-derived granulocyte-macrophage colony-stimulating factor regulates myeloid inflammation and T cell immunity in pancreatic cancerCancer Cell201221682283522698406
  • GrecoODachsGUGene directed enzyme/prodrug therapy of cancer: historical appraisal and future prospectivesJ Cell Physiol20011871223611241346
  • DiasJDLiikanenIGuseKTargeted chemotherapy for head and neck cancer with a chimeric oncolytic adenovirus coding for bifunctional suicide protein FCU1Clin Cancer Res20101692540254920388844
  • BraybrookeJPSladeADeplanqueGPhase I study of MetXia-P450 gene therapy and oral cyclophosphamide for patients with advanced breast cancer or melanomaClin Cancer Res20051141512152015746054
  • PalmerDHMautnerVMirzaDVirus-directed enzyme prodrug therapy: intratumoral administration of a replication-deficient adenovirus encoding nitroreductase to patients with resectable liver cancerJ Clin Oncol20042291546155215051757
  • FolkmanJTumor angiogenesis: therapeutic implicationsN Engl J Med197128521118211864938153
  • FolkmanJRole of angiogenesis in tumor growth and metastasisSemin Oncol2002296 Suppl 16151812516034
  • LaskinJCrinòLFelipESafety and efficacy of first-line bevacizumab plus chemotherapy in elderly patients with advanced or recurrent nonsquamous non-small cell lung cancer: safety of avastin in lung trial (MO19390)J Thorac Oncol20127120321122173662
  • SobreroAAcklandSClarkeSPhase IV study of bevacizumab in combination with infusional fluorouracil, leucovorin and irinotecan (FOLFIRI) in first-line metastatic colorectal cancerOncology200977211311919628950
  • TysomeJRLemoineNRWangYCombination of anti-angiogenic therapy and virotherapy: arming oncolytic viruses with anti-angiogenic genesCurr Opin Mol Ther200911666466920072943
  • TandleABlazerDGIIILibuttiSKAntiangiogenic gene therapy of cancer: recent developmentsJ Transl Med2004212215219236
  • DomeBDobosJTovariJCirculating bone marrow-derived endothelial progenitor cells: characterization, mobilization, and therapeutic considerations in malignant diseaseCytometry A200873318619318000872
  • PandyaNMDhallaNSSantaniDDAngiogenesis – a new target for future therapyVascul Pharmacol200644526527416545987
  • MaJWaxmanDJCombination of antiangiogenesis with chemotherapy for more effective cancer treatmentMol Cancer Ther20087123670368419074844
  • SessaCGuibalADel ConteGRüeggCBiomarkers of angiogenesis for the development of antiangiogenic therapies in oncology: tools or decorations?Nat Clin Pract Oncol20085737839118560389
  • Bottsford-MillerJNColemanRLSoodAKResistance and escape from antiangiogenesis therapy: clinical implications and future strategiesJ Clin Oncol201230324026403423008289
  • AlbiniATosettiFLiVWNoonanDMLiWWCancer prevention by targeting angiogenesisNat Rev Clin Oncol20129949850922850752
  • BoehmTFolkmanJBrowderTO’ReillyMSAntiangiogenic therapy of experimental cancer does not induce acquired drug resistanceNature199739066584044079389480
  • HurwitzHFehrenbacherLNovotnyWBevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancerN Engl J Med2004350232335234215175435
  • RelfMLeJeuneSScottPAExpression of the angiogenic factors vascular endothelial cell growth factor, acidic and basic fibroblast growth factor, tumor growth factor beta-1, platelet-derived endothelial cell growth factor, placenta growth factor, and pleiotrophin in human primary breast cancer and its relation to angiogenesisCancer Res19975759639699041202
  • AngaritaFAAcunaSAOttolino-PerryKZerhouniSMcCartJAMounting a strategic offense: fighting tumor vasculature with oncolytic virusesTrends Mol Med201319637839223540715
  • BreitbachCJDe SilvaNSFallsTJTargeting tumor vasculature with an oncolytic virusMol Ther201119588689421364541
  • BenenciaFCourregesMCConejo-GarciaJROncolytic HSV exerts direct antiangiogenic activity in ovarian carcinomaHum Gene Ther200516676577815960607
  • BreitbachCJArulanandamRDe SilvaNOncolytic vaccinia virus disrupts tumor-associated vasculature in humansCancer Res20137341265127523393196
  • LauKBicknellRAntiangiogenic gene therapyGene Ther19996111793179510602373
  • KirnDHThorneSHTargeted and armed oncolytic poxviruses: a novel multi-mechanistic therapeutic class for cancerNat Rev Cancer200991647119104515
  • ClinicalTrials.govUnited States National Institute of Health2013http://www.clinicaltrials.gov/Accessed March 3, 2013
  • ZhangZZouWWangJSuppression of tumor growth by oncolytic adenovirus-mediated delivery of an antiangiogenic gene, soluble Flt-1Mol Ther200511455356215771958
  • ThorneSHTamBYKirnDHContagCHKuoCJSelective intratumoral amplification of an antiangiogenic vector by an oncolytic virus produces enhanced antivascular and anti-tumor efficacyMol Ther200613593894616469543
  • XiaoTFanJKHuangHLGuJFLiLYLiuXYVEGI-armed oncolytic adenovirus inhibits tumor neovascularization and directly induces mitochondria-mediated cancer cell apoptosisCell Res201020336737819918267
  • KangYAShinHCYooJYKimJHKimJSYunCONovel cancer antiangiotherapy using the VEGF promoter-targeted artificial zinc-finger protein and oncolytic adenovirusMol Ther20081661033104018398429
  • GuseKDiaconuIRajeckiMAd5/3–9HIF-Delta24-VEGFR-1-Ig, an infectivity enhanced, dual-targeted and antiangiogenic oncolytic adenovirus for kidney cancer treatmentGene Ther20091681009102019440223
  • GuseKSlonieckaMDiaconuIAntiangiogenic arming of an oncolytic vaccinia virus enhances antitumor efficacy in renal cell cancer modelsJ Virol201084285686619906926
  • OlofssonBKorpelainenEPepperMSVascular endothelial growth factor B (VEGF-B) binds to VEGF receptor-1 and regulates plasminogen activator activity in endothelial cellsProc Natl Acad Sci USA1998952011709117149751730
  • PatilSSGentschevIAdelfingerMVirotherapy of canine tumors with oncolytic vaccinia virus GLV-1h109 expressing an anti-VEGF single-chain antibodyPLoS One2012710e4747223091626
  • GuptaPSuZZLebedevaIVmda-7/IL-24: multifunctional cancer-specific apoptosis-inducing cytokinePharmacol Ther2006111359662816464504
  • LebedevaIVEmdadLSuZZmda-7/IL-24, novel anticancer cytokine: focus on bystander antitumor, radiosensitization and anti-angiogenic properties and overview of the phase I clinical experience (Review)Int J Oncol2007315985100717912425
  • FanJKWeiNDingMTargeting Gene-ViroTherapy for prostate cancer by DD3-driven oncolytic virus-harboring interleukin-24 geneInt J Cancer2010127370771719950222
  • XiaoLLWuYMQianJThe antitumor efficacy of IL-24 mediated by E1A and E1B triple regulated oncolytic adenovirusCancer Biol Ther201010324225020574150
  • ChaiLLiuSMaoQA novel conditionally replicating adenoviral vector with dual expression of IL-24 and arresten inserted in E1 and the region between E4 and fiber for improved melanoma therapyCancer Gene Ther201219424725422193628
  • XieKInterleukin-8 and human cancer biologyCytokine Growth Factor Rev200112437539111544106
  • YooJYKimJHKimJShort hairpin RNA-expressing oncolytic adenovirus-mediated inhibition of IL-8: effects on antiangiogenesis and tumor growth inhibitionGene Ther200815963565118273054
  • CaoRFarneboJKurimotoMCaoYInterleukin-18 acts as an angiogenesis and tumor suppressorFASEB J199913152195220210593867
  • BrundaMJLuistroLWarrierRRAntitumor and antimetastatic activity of interleukin 12 against murine tumorsJ Exp Med19931784122312308104230
  • MaioneTEGrayGSPetroJInhibition of angiogenesis by recombinant human platelet factor-4 and related peptidesScience1990247493877791688470
  • LiuTCZhangTFukuharaHOncolytic HSV armed with platelet factor 4, an antiangiogenic agent, shows enhanced efficacyMol Ther200614678979717045531
  • VargheseSRabkinSDLiuRNielsenPGIpeTMartuzaRLEnhanced therapeutic efficacy of IL-12, but not GM-CSF, expressing oncolytic herpes simplex virus for transgenic mouse derived prostate cancersCancer Gene Ther200613325326516179929
  • WongRJChanMKYuZAngiogenesis inhibition by an oncolytic herpes virus expressing interleukin 12Clin Cancer Res200410134509451615240543
  • MeadowsKNBryantPVincentPAPumigliaKMActivated Ras induces a proangiogenic phenotype in primary endothelial cellsOncogene200423119220014712224
  • LamfersMLGianniDTungCHTissue inhibitor of metalloproteinase-3 expression from an oncolytic adenovirus inhibits matrix metalloproteinase activity in vivo without affecting antitumor efficacy in malignant gliomaCancer Res200565209398940516230403
  • FolkmanJEndogenous angiogenesis inhibitorsAPMIS20041127–849650715563312
  • LiGShamJYangJPotent antitumor efficacy of an E1B 55 kDa-deficient adenovirus carrying murine endostatin in hepatocellular carcinomaInt J Cancer2005113464064815389517
  • LiGCYangJMNieMMPotent antitumoral effects of a novel gene-viral therapeutic system CNHK300-mEndostatin in hepatocellular carcinomaChin Med J (Engl)2005118317918515740644
  • MullenJTDonahueJMChandrasekharSOncolysis by viral replication and inhibition of angiogenesis by a replication-conditional herpes simplex virus that expresses mouse endostatinCancer2004101486987715305421
  • SuCNaMChenJGene-viral cancer therapy using dual-regulated oncolytic adenovirus with antiangiogenesis gene for increased efficacyMol Cancer Res20086456857518344493
  • ZhangQNieMShamJEffective gene-viral therapy for telomerase-positive cancers by selective replicative-competent adenovirus combining with endostatin geneCancer Res200464155390539715289347
  • PanJGZhouXZengGWHanRFPotent antitumour activity of the combination of HSV-TK and endostatin armed oncolytic adeno-associated virus for bladder cancer in vitro and in vivoJ Surg Oncol2012105324925721953122
  • GoodwinJMSchmittADMcGinnCMAngiogenesis inhibition using an oncolytic herpes simplex virus expressing endostatin in a murine lung cancer modelCancer Invest201230324325022360364
  • YangLWangLSuXQSuppression of ovarian cancer growth via systemic administration with liposome-encapsulated adenovirus-encoding endostatinCancer Gene Ther2010171495719609295
  • ZhangWFulciGBuhrmanJSBevacizumab with angiostatin-armed oHSV increases antiangiogenesis and decreases bevacizumab-induced invasion in U87 gliomaMol Ther2012201374521915104
  • ScappaticciFAContrerasASmithRStatin-AE: a novel angiostatin-endostatin fusion protein with enhanced antiangiogenic and antitumor activityAngiogenesis20014426326812197471
  • LiXLiuYHLeeSJGardnerTAJengMHKaoCProstate-restricted replicative adenovirus expressing human endostatin-angiostatin fusion gene exhibiting dramatic antitumor efficacyClin Cancer Res200814129129918172281
  • YangCTLinYCLinCLOncolytic herpesvirus with secretable angiostatic proteins in the treatment of human lung cancer cellsAnticancer Res2005253B2049205416158944
  • TysomeJRBriatAAlusiGLister strain of vaccinia virus armed with endostatin-angiostatin fusion gene as a novel therapeutic agent for human pancreatic cancerGene Ther200916101223123319587709
  • TysomeJRWangPAlusiGLister vaccine strain of vaccinia virus armed with the endostatin-angiostatin fusion gene: an oncolytic virus superior to dl1520 (ONYX-015) for human head and neck cancerHum Gene Ther20112291101110821361787
  • CaoYChenAAnSSJiRWDavidsonDLlinásMKringle 5 of plasminogen is a novel inhibitor of endothelial cell growthJ Biol Chem19972723622924229289278456
  • FanJKXiaoTGuJFIncreased suppression of oncolytic adenovirus carrying mutant k5 on colorectal tumorBiochem Biophys Res Commun2008374219820318621025
  • HeXPSuCQWangXHE1B-55kD-deleted oncolytic adenovirus armed with canstatin gene yields an enhanced anti-tumor efficacy on pancreatic cancerCancer Lett20092851899819481338
  • HardcastleJKurozumiKDmitrievaNEnhanced antitumor efficacy of vasculostatin (Vstat120) expressing oncolytic HSV-1Mol Ther201018228529419844198
  • LiuTCZhangTFukuharaHDominant-negative fibroblast growth factor receptor expression enhances antitumoral potency of oncolytic herpes simplex virus in neural tumorsClin Cancer Res200612226791679917121900
  • JainRKFinnAVKolodgieFDGoldHKVirmaniRAntiangiogenic therapy for normalization of atherosclerotic plaque vasculature: a potential strategy for plaque stabilizationNat Clin Pract Cardiovasc Med20074949150217712362