Oncolytic virus

Last updated

An oncolytic virus is a virus that preferentially infects and kills cancer cells. As the infected cancer cells are destroyed by oncolysis, they release new infectious virus particles or virions to help destroy the remaining tumour. [1] [2] Oncolytic viruses are thought not only to cause direct destruction of the tumour cells, but also to stimulate host anti-tumour immune system responses. [3] [4] Oncolytic viruses also have the ability to affect the tumor micro-environment in multiple ways. [5] [6]

Contents

The potential of viruses as anti-cancer agents was first realised in the early twentieth century, although coordinated research efforts did not begin until the 1960s. [7] A number of viruses including adenovirus, reovirus, measles, herpes simplex, Newcastle disease virus, and vaccinia have been clinically tested as oncolytic agents. [8] Most current oncolytic viruses are engineered for tumour selectivity, although there are naturally occurring examples such as reovirus and the senecavirus, [9] resulting in clinical trials. [10]

The first oncolytic virus to be approved by a national regulatory agency was genetically unmodified ECHO-7 strain enterovirus RIGVIR, which was approved in Latvia in 2004 for the treatment of skin melanoma; [11] the approval was withdrawn in 2019. An oncolytic adenovirus, a genetically modified adenovirus named H101, was approved in China in 2005 for the treatment of head and neck cancer. [12] In 2015, talimogene laherparepvec (OncoVex, T-VEC), an oncolytic herpes virus which is a modified herpes simplex virus, became the first oncolytic virus to be approved for use in the United States and the European Union, for the treatment of advanced inoperable melanoma. [13]

On December 16, 2022, the Food and Drug Administration approved nadofaragene firadenovec-vncg (Adstiladrin, Ferring Pharmaceuticals) for adult patients with high-risk Bacillus Calmette-Guérin (BCG) unresponsive non-muscle invasive bladder cancer (NMIBC) with carcinoma in situ (CIS) with or without papillary tumors. [14]

History

A connection between cancer regression and viruses has long been theorised, and case reports of regression noted in cervical cancer, Burkitt lymphoma, and Hodgkin lymphoma, after immunisation or infection with an unrelated virus appeared at the beginning of the 20th century. [15] Efforts to treat cancer through immunisation or virotherapy (deliberate infection with a virus), began in the mid-20th century. [15] [16] As the technology to create a custom virus did not exist, all early efforts focused on finding natural oncolytic viruses. During the 1960s, promising research involved using poliovirus, [17] adenovirus, [15] Coxsackie virus, [18] ECHO enterovirus RIGVIR, [19] and others. [16] The early complications were occasional cases of uncontrolled infection (resulting in significant morbidity and mortality); an immune response would also frequently develop. While not directly harmful to the patient, [15] the response destroyed the virus thus preventing it from destroying the cancer. [17] Early efforts also found that only certain cancers could be treated through virotherapy. [18] Even when a response was seen, these responses were neither complete nor durable. [15] The field of virotherapy was nearly abandoned for a time, as the technology required to modify viruses didn't exist whereas chemotherapy and radiotherapy technology enjoyed early success. However, now that these technologies have been thoroughly developed and cancer remains a major cause of mortality, there is still a need for novel cancer therapies, garnering this once-sidelined therapy renewed interest. [15] [20]

Herpes simplex virus

Herpes simplex virus (HSV) was one of the first viruses to be adapted to attack cancer cells selectively, because it was well understood, easy to manipulate and relatively harmless in its natural state (merely causing cold sores) so likely to pose fewer risks. The herpes simplex virus type 1 (HSV-1) mutant 1716 lacks both copies of the ICP34.5 gene, and as a result is no longer able to replicate in terminally differentiated and non-dividing cells but will infect and cause lysis very efficiently in cancer cells, and this has proved to be an effective tumour-targeting strategy. [21] [22] In a wide range of in vivo cancer models, the HSV1716 virus has induced tumour regression and increased survival times. [23] [24] [25]

In 1996, the first approval was given in Europe for a clinical trial using the oncolytic virus HSV1716. From 1997 to 2003, strain HSV1716 was injected into tumours of patients with glioblastoma multiforme, a highly malignant brain tumour, with no evidence of toxicity or side effects, and some long-term survivors. [26] [27] [28] Other safety trials have used HSV1716 to treat patients with melanoma and squamous-cell carcinoma of head and neck. [29] [30] Since then other studies have shown that the outer coating of HSV1716 variants can be targeted to specific types of cancer cells, [31] and can be used to deliver a variety of additional genes into cancer cells, such as genes to split a harmless prodrug inside cancer cells to release toxic chemotherapy, [32] or genes which command infected cancer cells to concentrate protein tagged with radioactive iodine, so that individual cancer cells are killed by micro-dose radiation as well as by virus-induced cell lysis. [33]

Other oncolytic viruses based on HSV have also been developed and are in clinical trials. [34] One that has been approved by the FDA for advanced melanoma is Amgen's talimogene laherparepvec. [35]

Oncorine (H101)

The first oncolytic virus to be approved by a regulatory agency was a genetically modified adenovirus named H101 by Shanghai Sunway Biotech. It gained regulatory approval in 2005 from China's State Food and Drug Administration (SFDA) for the treatment of head and neck cancer. [12] [36] Sunway's H101 and the very similar Onyx-15 (dl1520) have been engineered to remove a viral defense mechanism that interacts with a normal human gene p53 , which is very frequently dysregulated in cancer cells. [36] Despite the promises of early in vivo lab work, these viruses do not specifically infect cancer cells, but they still kill cancer cells preferentially. [36] While overall survival rates are not known, short-term response rates are approximately doubled for H101 plus chemotherapy when compared to chemotherapy alone. [36] It appears to work best when injected directly into a tumour, and when any resulting fever is not suppressed. [36] Systemic therapy (such as through infusion through an intravenous line) is desirable for treating metastatic disease. [37] It is now marketed under the brand name Oncorine. [38]

Mechanisms of action

Immunotherapy

With advances in cancer immunotherapy such as immune checkpoint inhibitors, increased attention has been given to using oncolytic viruses to increase antitumor immunity. [39] There are two main considerations of the interaction between oncolytic viruses and the immune system.[ citation needed ]

Immunity as an obstacle

A major obstacle to the success of oncolytic viruses is the patient immune system which naturally attempts to deactivate any virus. This can be a particular problem for intravenous injection, where the virus must first survive interactions with the blood complement and neutralising antibodies. [40] It has been shown that immunosuppression by chemotherapy and inhibition of the complement system can enhance oncolytic virus therapy. [41] [42] [43]

Pre-existing immunity can be partly avoided by using viruses that are not common human pathogens. However, this does not avoid subsequent antibody generation. Yet, some studies have shown that pre-immunity to oncolytic viruses doesn't cause a significant reduction in efficacy. [44]

Alternatively, the viral vector can be coated with a polymer such as polyethylene glycol, shielding it from antibodies, but this also prevents viral coat proteins adhering to host cells. [45]

Another way to help oncolytic viruses reach cancer growths after intravenous injection, is to hide them inside macrophages (a type of white blood cell). Macrophages automatically migrate to areas of tissue destruction, especially where oxygen levels are low, characteristic of cancer growths, and have been used successfully to deliver oncolytic viruses to prostate cancer in animals. [46]

Immunity as an ally

Although it poses a hurdle by inactivating viruses, the patient's immune system can also act as an ally against tumors; infection attracts the attention of the immune system to the tumour and may help to generate useful and long-lasting antitumor immunity. [47] [48] One important mechanism is the release of substances by tumor lysis, such as tumor-associated antigens and danger associated-molecular patterns (DAMPs), which can elicit an antitumor immune response. [49] This essentially produces a personalised cancer vaccine.

Many cases of spontaneous remission of cancer have been recorded. Though the cause is not fully understood, they are thought likely to be a result of a sudden immune response or infection. [50] Efforts to induce this phenomenon have used cancer vaccines (derived from cancer cells or selected cancer antigens), or direct treatment with immune-stimulating factors on skin cancers. [51] Some oncolytic viruses are very immunogenic and may by infection of the tumour, elicit an anti-tumor immune response, especially viruses delivering cytokines or other immune stimulating factors. [52] [53]

Viruses selectively infect tumor cells because of their defective anti-viral response. [39] Imlygic, an attenuated herpes simplex virus, has been genetically engineered to replicate preferentially within tumor cells and to generate antigens that elicit an immune response. [39]

Oncolytic behaviour of wild-type viruses

Vaccinia virus

Vaccinia virus (VACV) is arguably the most successful live biotherapeutic agent because of its critical role in the eradication of smallpox, one of the most deadly diseases in human history. Long before the smallpox eradication campaign was launched, VACV was exploited as a therapeutic agent for the treatment of cancer. In 1922, Levaditi and Nicolau reported that VACV was able to inhibit the growth of various tumors in mice and rats. This was the first demonstration of viral oncolysis in the laboratory. This virus was subsequently shown to selectively infect and destroy tumor cells with great potency, while sparing normal cells, both in cell cultures and in animal models. Since vaccinia virus has long been recognized as an ideal backbone for vaccines due to its potent antigen presentation capability, this combines well with its natural oncolytic activities as an oncolytic virus for cancer immunotherapy. [54]

Vesicular stomatitis virus

Vesicular stomatitis virus (VSV) is a rhabdovirus, consisting of 5 genes encoded by a negative sense, single-stranded RNA genome. In nature, VSV infects insects as well as livestock, where it causes a relatively localized and non-fatal illness. The low pathogenicity of this virus is due in large part to its sensitivity to interferons, a class of proteins that are released into the tissues and bloodstream during infection. These molecules activate genetic anti-viral defence programs that protect cells from infection and prevent spread of the virus. However, in 2000, Stojdl, Lichty et al. [55] demonstrated that defects in these pathways render cancer cells unresponsive to the protective effects of interferons and therefore highly sensitive to infection with VSV. Since VSV undergoes a rapid cytolytic replication cycle, infection leads to death of the malignant cell and roughly a 1000-fold amplification of virus within 24h. VSV is therefore highly suitable for therapeutic application, and several groups have gone on to show that systemically administered VSV can be delivered to a tumour site, where it replicates and induces disease regression, often leading to durable cures. [56] [57] [58] [59] Attenuation of the virus by engineering a deletion of Met-51 of the matrix protein ablates virtually all infection of normal tissues, while replication in tumour cells is unaffected. [56]

Recent research has shown that this virus has the potential to cure brain tumours, thanks to its oncolytic properties. [60]

Poliovirus

Poliovirus is a natural invasive neurotropic virus, making it the obvious choice for selective replication in tumours derived from neuronal cells. Poliovirus has a plus-strand RNA genome, the translation of which depends on a tissue-specific internal ribosome entry site (IRES) within the 5' untranslated region of the viral genome, which is active in cells of neuronal origin and allows translation of the viral genome without a 5' cap. Gromeier et al. (2000) [61] replaced the normal poliovirus IRES with a rhinovirus IRES, altering tissue specificity. The resulting PV1(RIPO) virus was able to selectively destroy malignant glioma cells, while leaving normal neuronal cells untouched. [62]

Reovirus

Reoviruses generally infect mammalian respiratory and bowel systems (the name deriving from an acronym, respiratory enteric orphan virus). Most people have been exposed to reovirus by adulthood; however, the infection does not typically produce symptoms. The reovirus' oncolytic potential was established after they were discovered to reproduce well in various cancer cell lines, lysing these cells. [63]

Reolysin is a formulation of reovirus intended to treat various cancers currently undergoing clinical trials. [64]

Senecavirus

Senecavirus, also known as Seneca Valley Virus, is a naturally occurring wild-type oncolytic picornavirus discovered in 2001 as a tissue culture contaminate at Genetic Therapy, Inc. The initial isolate, SVV-001, is being developed as an anti-cancer therapeutic by Neotropix, Inc. under the name NTX-010 for cancers with neuroendocrine features including small cell lung cancer and a variety of pediatric solid tumours.[ citation needed ]

RIGVIR

RIGVIR is a drug that was approved by the State Agency of Medicines of the Republic of Latvia in 2004. [65] It was also approved in Georgia [66] and Armenia. [67] It is wild type ECHO-7, a member of echovirus group. [68] The potential use of echovirus as an oncolytic virus to treat cancer was discovered by Latvian scientist Aina Muceniece in the 1960s and 1970s. [68] The data used to register the drug in Latvia is not sufficient to obtain approval to use it in the US, Europe, or Japan. [68] [69] As of 2017 there was no good evidence that RIGVIR is an effective cancer treatment. [70] [71] On 19 March 2019, the manufacturer of ECHO-7, SIA LATIMA, announced the drug's removal from sale in Latvia, quoting financial and strategic reasons and insufficient profitability. [72] However, several days later an investigative TV show revealed that State Agency of Medicines had run laboratory tests on the vials, and found that the amount of ECHO-7 virus is of a much smaller amount than claimed by the manufacturer. According to agency's lab director, "It's like buying what you think is lemon juice, but finding that what you have is lemon-flavored water". In March 2019, the distribution of ECHO-7 in Latvia has been stopped. [73] Based on the request of some patients, medical institutions and physicians were allowed to continue use despite the suspension of the registration certificate. [74]

Semliki Forest virus

Semliki Forest virus (SFV) is a virus that naturally infects cells of the central nervous system and causes encephalitis. A genetically engineered form has been pre-clinically tested as an oncolytic virus against the severe brain tumour type glioblastoma. The SFV was genetically modified with microRNA target sequences so that it only replicated in brain tumour cells and not in normal brain cells. The modified virus reduced tumour growth and prolonged survival of mice with brain tumours. [75] The modified virus was also found to efficiently kill human glioblastoma tumour cell lines. [75]

Other

The maraba virus, first identified in Brazilian sandflies, is being tested clinically. [76]

Coxsackievirus A21 is being developed by Viralytics under trade name Cavatak. [77] Coxsackievirus A21 belongs to Enterovirus C species. [78]

Influenza A is one of the earliest viruses anecdotally reported to induce cancer regression. [79] This has prompted preclinical development of genetically engineered oncolytic influenza A viruses. [80] Murine Respirovirus, which is frequently called Sendai virus in scientific literature, has shown some oncolytic properties that are decibed in the section Murine respirovirus as an oncolytic agent.

Engineering oncolytic viruses

Directed evolution

An innovative approach of drug development termed "directed evolution" involves the creation of new viral variants or serotypes specifically directed against tumour cells via rounds of directed selection using large populations of randomly generated recombinant precursor viruses. The increased biodiversity produced by the initial homologous recombination step provides a large random pool of viral candidates which can then be passed through a series of selection steps designed to lead towards a pre-specified outcome (e.g. higher tumor specific activity) without requiring any previous knowledge of the resultant viral mechanisms that are responsible for that outcome. The pool of resultant oncolytic viruses can then be further screened in pre-clinical models to select an oncolytic virus with the desired therapeutic characteristics. [81]

Directed evolution was applied on human adenovirus, one of many viruses that are being developed as oncolytic agents, to create a highly selective and yet potent oncolytic vaccine. As a result of this process, ColoAd1 (a novel chimeric member of the group B adenoviruses) was generated. This hybrid of adenovirus serotypes Ad11p and Ad3 shows much higher potency and tumour selectivity than the control viruses (including Ad5, Ad11p and Ad3) and was confirmed to generate approximately two logs more viral progeny on freshly isolated human colon tumour tissue than on matching normal tissue. [81]

Attenuation

Attenuation involves deleting viral genes, or gene regions, to eliminate viral functions that are expendable in tumour cells, but not in normal cells, thus making the virus safer and more tumour-specific. Cancer cells and virus-infected cells have similar alterations in their cell signalling pathways, particularly those that govern progression through the cell cycle. [82] A viral gene whose function is to alter a pathway is dispensable in cells where the pathway is defective, but not in cells where the pathway is active.[ citation needed ]

The enzymes thymidine kinase and ribonucleotide reductase in cells are responsible for DNA synthesis and are only expressed in cells which are actively replicating. [83] These enzymes also exist in the genomes of certain viruses (E.g. HSV, vaccinia) and allow viral replication in quiescent(non-replicating) cells, [84] so if they are inactivated by mutation the virus will only be able to replicate in proliferating cells, such as cancer cells.

Tumour targeting

There are two main approaches for generating tumour selectivity: transductional and non-transductional targeting. [85]

Double targeting with both transductional and non-transductional targeting methods is more effective than any one form of targeting alone. [86]

Reporter genes

Viral luciferase expression in a mouse tumour Viral luciferase expression in a mouse tumour.jpg
Viral luciferase expression in a mouse tumour

Both in the laboratory and in the clinic it is useful to have a simple means of identifying cells infected by the experimental virus. This can be done by equipping the virus with "reporter genes" not normally present in viral genomes, which encode easily identifiable protein markers. One example of such proteins is GFP (green fluorescent protein) which, when present in infected cells, will cause a fluorescent green light to be emitted when stimulated by blue light. [87] [88] An advantage of this method is that it can be used on live cells and in patients with superficial infected lesions, it enables rapid non-invasive confirmation of viral infection. [89] Another example of a visual marker useful in living cells is luciferase, an enzyme from the firefly which in the presence of luciferin, emits light detectable by specialized cameras. [87]

Vaccinia virus infected cells expressing beta-glucuronidase (blue colour) Viral LACZ gene expression.JPG
Vaccinia virus infected cells expressing beta-glucuronidase (blue colour)

The E. coli enzymes beta-glucuronidase and beta-galactosidase can also be encoded by some viruses. These enzymes, in the presence of certain substrates, can produce intense colored compounds useful for visualizing infected cells and also for quantifying gene expression.[ citation needed ]

Modifications to improve oncolytic activity

Oncolytic viruses can be used against cancers in ways that are additional to lysis of infected cells.

Suicide genes

Viruses can be used as vectors for delivery of suicide genes, encoding enzymes that can metabolise a separately administered non-toxic pro-drug into a potent cytotoxin, which can diffuse to and kill neighbouring cells. One herpes simplex virus, encoding a thymidine kinase suicide gene, has progressed to phase III clinical trials. The herpes simplex virus thymidine kinase phosphorylates the pro-drug, ganciclovir, which is then incorporated into DNA, blocking DNA synthesis. [90] The tumour selectivity of oncolytic viruses ensures that the suicide genes are only expressed in cancer cells, however a "bystander effect" on surrounding tumour cells has been described with several suicide gene systems. [91]

Suppression of angiogenesis

Angiogenesis (blood vessel formation) is an essential part of the formation of large tumour masses. Angiogenesis can be inhibited by the expression of several genes, which can be delivered to cancer cells in viral vectors, resulting in suppression of angiogenesis, and oxygen starvation in the tumour. The infection of cells with viruses containing the genes for angiostatin and endostatin synthesis inhibited tumour growth in mice. Enhanced antitumour activities have been demonstrated in a recombinant vaccinia virus encoding anti-angiogenic therapeutic antibody and with an HSV1716 variant expressing an inhibitor of angiogenesis. [92] [93]

Radioiodine

Adenoviral NIS gene expression in a mouse tumour (Located at the crosshairs) following intravenous delivery of virus (Left) compared to an uninfected control mouse (Right) Adenoviral NIS gene expression in a mouse tumour.jpg
Adenoviral NIS gene expression in a mouse tumour (Located at the crosshairs) following intravenous delivery of virus (Left) compared to an uninfected control mouse (Right)

Addition of the sodium-iodide symporter (NIS) gene to the viral genome causes infected tumour cells to express NIS and accumulate iodine. When combined with radioiodine therapy it allows local radiotherapy of the tumour, as used to treat thyroid cancer. The radioiodine can also be used to visualise viral replication within the body by the use of a gamma camera. [87] This approach has been used successfully preclinically with adenovirus, measles virus and vaccinia virus. [94] [95] [96]

Approved therapeutic agents

Oncolytic viruses in conjunction with existing cancer therapies

It is in conjunction with conventional cancer therapies that oncolytic viruses have often showed the most promise, since combined therapies operate synergistically with no apparent negative effects. [103]

Clinical trials

Onyx-015 (dl1520) underwent trials in conjunction with chemotherapy before it was abandoned in the early 2000s. The combined treatment gave a greater response than either treatment alone, but the results were not entirely conclusive. [104] Vaccinia virus GL-ONC1 was studied in a trial combined with chemo- and radiotherapy as Standard of Care for patients newly diagnosed with head & neck cancer. [105] Herpes simplex virus, adenovirus, reovirus and murine leukemia virus are also undergoing clinical trials as a part of combination therapies. [106]

Pre-clinical research

Chen et al. (2001) [107] used CV706, a prostate-specific adenovirus, in conjunction with radiotherapy on prostate cancer in mice. The combined treatment resulted in a synergistic increase in cell death, as well as a significant increase in viral burst size (the number of virus particles released from each cell lysis). No alteration in viral specificity was observed.[ citation needed ]

SEPREHVIR (HSV-1716) has also shown synergy in pre-clinical research when used in combination with several cancer chemotherapies. [108] [109]

The anti-angiogenesis drug bevacizumab (anti-VEGF antibody) has been shown to reduce the inflammatory response to oncolytic HSV and improve virotherapy in mice. [110] A modified oncolytic vaccinia virus encoding a single-chain anti-VEGF antibody (mimicking bevacizumab) was shown to have significantly enhanced antitumor activities than parental virus in animal models. [111]

In fiction

In science fiction, the concept of an oncolytic virus was first introduced to the public in Jack Williamson's novel Dragon's Island, published in 1951, although Williamson's imaginary virus was based on a bacteriophage rather than a mammalian virus. [112] Dragon's Island is also known for being the source of the term "genetic engineering". [113]

The plot of the Hollywood film I Am Legend is based on the premise that a worldwide epidemic was caused by a viral cure for cancer. [114]

See also

Related Research Articles

<span class="mw-page-title-main">Sedoreoviridae</span> Family of viruses

Sedoreoviridae is a family of double-stranded RNA viruses. Member viruses have a wide host range, including vertebrates, invertebrates, plants, protists and fungi. They lack lipid envelopes and package their segmented genome within multi-layered capsids. Lack of a lipid envelope has allowed three-dimensional structures of these large complex viruses to be obtained, revealing a structural and likely evolutionary relationship to the cystovirus family of bacteriophage. There are currently 97 species in this family, divided among 15 genera in two subfamilies. Reoviruses can affect the gastrointestinal system and respiratory tract. The name "reo-" is an acronym for "respiratory enteric orphan" viruses. The term "orphan virus" refers to the fact that some of these viruses have been observed not associated with any known disease. Even though viruses in the family Reoviridae have more recently been identified with various diseases, the original name is still used.

<span class="mw-page-title-main">Cancer immunotherapy</span> Artificial stimulation of the immune system to treat cancer

Cancer immunotherapy is the stimulation of the immune system to treat cancer, improving on the immune system's natural ability to fight the disease. It is an application of the fundamental research of cancer immunology and a growing subspecialty of oncology.

Virotherapy is a treatment using biotechnology to convert viruses into therapeutic agents by reprogramming viruses to treat diseases. There are three main branches of virotherapy: anti-cancer oncolytic viruses, viral vectors for gene therapy and viral immunotherapy. These branches use three different types of treatment methods: gene overexpression, gene knockout, and suicide gene delivery. Gene overexpression adds genetic sequences that compensate for low to zero levels of needed gene expression. Gene knockout uses RNA methods to silence or reduce expression of disease-causing genes. Suicide gene delivery introduces genetic sequences that induce an apoptotic response in cells, usually to kill cancerous growths. In a slightly different context, virotherapy can also refer more broadly to the use of viruses to treat certain medical conditions by killing pathogens.

<span class="mw-page-title-main">Herpes simplex virus</span> Species of virus

Herpes simplex virus1 and 2, also known by their taxonomic names Human alphaherpesvirus 1 and Human alphaherpesvirus 2, are two members of the human Herpesviridae family, a set of viruses that produce viral infections in the majority of humans. Both HSV-1 and HSV-2 are very common and contagious. They can be spread when an infected person begins shedding the virus.

Viral vectors are tools commonly used by molecular biologists to deliver genetic material into cells. This process can be performed inside a living organism or in cell culture. Viruses have evolved specialized molecular mechanisms to efficiently transport their genomes inside the cells they infect. Delivery of genes or other genetic material by a vector is termed transduction and the infected cells are described as transduced. Molecular biologists first harnessed this machinery in the 1970s. Paul Berg used a modified SV40 virus containing DNA from the bacteriophage λ to infect monkey kidney cells maintained in culture.

<span class="mw-page-title-main">Genetically modified virus</span> Species of virus

A genetically modified virus is a virus that has been altered or generated using biotechnology methods, and remains capable of infection. Genetic modification involves the directed insertion, deletion, artificial synthesis or change of nucleotide bases in viral genomes. Genetically modified viruses are mostly generated by the insertion of foreign genes intro viral genomes for the purposes of biomedical, agricultural, bio-control, or technological objectives. The terms genetically modified virus and genetically engineered virus are used synonymously.

Oncolytics Biotech Inc. is a Canadian company headquartered in Calgary, Alberta, that is developing an intravenously delivered immuno-oncolytic virus called pelareorep for the treatment of solid tumors and hematological malignancies. Pelareorep is a non-pathogenic, proprietary isolate of the unmodified reovirus that: induces selective tumor lysis and promotes an inflamed tumor phenotype through innate and adaptive immune responses.

Pelareorep is a proprietary isolate of the unmodified human reovirus being developed as a systemically administered immuno-oncological viral agent for the treatment of solid tumors and hematological malignancies. Pelareorep is an oncolytic virus, which means that it preferentially lyses cancer cells. Pelareorep also promotes an inflamed tumor phenotype through innate and adaptive immune responses. Preliminary clinical trials indicate that it may have anti-cancer effects across a variety of cancer types when administered alone and in combination with other cancer therapies.

JX-594 is an oncolytic virus is designed to target and destroy cancer cells. It is also known as Pexa-Vec, INN pexastimogene devacirepvec) and was constructed in Dr. Edmund Lattime's lab at Thomas Jefferson University, tested in clinical trials on melanoma patients, and licensed and further developed by SillaJen.

Herpes simplex research includes all medical research that attempts to prevent, treat, or cure herpes, as well as fundamental research about the nature of herpes. Examples of particular herpes research include drug development, vaccines and genome editing. HSV-1 and HSV-2 are commonly thought of as oral and genital herpes respectively, but other members in the herpes family include chickenpox (varicella/zoster), cytomegalovirus, and Epstein-Barr virus. There are many more virus members that infect animals other than humans, some of which cause disease in companion animals or have economic impacts in the agriculture industry.

<span class="mw-page-title-main">Jennerex</span>

Jennerex Biotherapeutics, Inc. was an American private biopharmaceutical company that developed the oncolytic viruses JX-594 and JX-929 among others. By creating oncolytic viruses that can (1) kill tumor cells directly through lysis, (2) activate the immune system by delivering genes that encode immunostimulants and by overcoming tumor cell-induced immunological tolerance, and (3) reduce tumor nutrient supply through the destruction of blood vessels, Jennerex aimed to create a novel approach to treating and possibly curing cancer.

<span class="mw-page-title-main">Talimogene laherparepvec</span> Gene therapy medication

Talimogene laherparepvec, sold under the brand name Imlygic, is a biopharmaceutical medication used to treat melanoma that cannot be operated on; it is injected directly into a subset of lesions which generates a systemic immune response against the recipient's cancer. The final four year analysis from the pivotal phase 3 study upon which TVEC was approved by the FDA showed a 31.5% response rate with a 16.9% complete response (CR) rate. There was also a substantial and statistically significant survival benefit in patients with earlier metastatic disease and in patients who hadn't received prior systemic treatment for melanoma. The earlier stage group had a reduction in the risk of death of approximately 50% with one in four patients appearing to have met, or be close to be reaching, the medical definition of cure. Real world use of talimogene laherparepvec have shown response rates of up to 88.5% with CR rates of up to 61.5%.

<span class="mw-page-title-main">Oncolytic herpes virus</span>

Many variants of herpes simplex virus have been considered for viral therapy of cancer; the early development of these was thoroughly reviewed in the journal Cancer Gene Therapy in 2002. This page describes the most notable variants—those tested in clinical trials: G207, HSV1716, NV1020 and Talimogene laherparepvec. These attenuated versions are constructed by deleting viral genes required for infecting or replicating inside normal cells but not cancer cells, such as ICP34.5, ICP6/UL39, and ICP47.

Adenovirus varieties have been explored extensively as a viral vector for gene therapy and also as an oncolytic virus.

GL-ONC1 is an investigational therapeutic product consisting of the clinical grade formulation of the laboratory strain GLV-1h68, an oncolytic virus developed by Genelux Corporation. GL-ONC1 is currently under evaluation in Phase I/II human clinical trials in the United States and Europe.

Akseli Hemminki July 27, 1973 (Helsinki) is a Finnish specialist in Oncology and Radiotherapy, Professor of Oncology and founder of two biotechnology companies.

Infected cell protein 34.5 is a protein expressed by the γ34.5 gene in viruses such as herpes simplex virus; it blocks a cellular stress response to viral infection. It shares the C-terminal regulatory domain with protein phosphatase 1 subunit 15A/B.

Adeno-associated virus (AAV) has been researched as a viral vector in gene therapy for cancer treatment as an Oncolytic Virus. Currently there are not any FDA approved AAV cancer treatments, as the first FDA approved AAV treatment was approved December 2017. However, there are many Oncolytic AAV applications that are in development and have been researched.

<span class="mw-page-title-main">Viral vector vaccine</span> Type of vaccine

A viral vector vaccine is a vaccine that uses a viral vector to deliver genetic material (DNA) that can be transcribed by the recipient's host cells as mRNA coding for a desired protein, or antigen, to elicit an immune response. As of April 2021, six viral vector vaccines, four COVID-19 vaccines and two Ebola vaccines, have been authorized for use in humans.

Transgene S.A. is a French biotechnology company founded in 1979. It is based in Illkirch-Graffenstaden, near Strasbourg, and develops and manufactures immunotherapies for the treatment of cancer.

References

  1. Ferguson MS, Lemoine NR, Wang Y (2012). "Systemic delivery of oncolytic viruses: hopes and hurdles". Advances in Virology. 2012: 1–14. doi: 10.1155/2012/805629 . PMC   3287020 . PMID   22400027.
  2. Casjens S (2010). "Oncolytic virus". In Mahy BW, Van Regenmortel MH (eds.). Desk Encyclopedia of General Virology. Boston: Academic Press. p. 167. ISBN   978-0-12-375146-1.
  3. Melcher A, Parato K, Rooney CM, Bell JC (June 2011). "Thunder and lightning: immunotherapy and oncolytic viruses collide". Molecular Therapy. 19 (6): 1008–16. doi:10.1038/mt.2011.65. PMC   3129809 . PMID   21505424.
  4. Lichty BD, Breitbach CJ, Stojdl DF, Bell JC (August 2014). "Going viral with cancer immunotherapy". Nature Reviews. Cancer. 14 (8): 559–67. doi:10.1038/nrc3770. PMID   24990523. S2CID   15182671.
  5. De Silva, Naomi; Atkins, Harold; Kirn, David H.; Bell, John C.; Breitbach, Caroline J. (1 April 2010). "Double trouble for tumours: Exploiting the tumour microenvironment to enhance anticancer effect of oncolytic viruses". Cytokine & Growth Factor Reviews. Recent Advances in the Development of Oncolytic Viruses as Cancer Therapeutics. 21 (2): 135–141. doi:10.1016/j.cytogfr.2010.02.007. ISSN   1359-6101. PMID   20338801.
  6. "Using Viruses to Treat Cancer | Science-Based Medicine". sciencebasedmedicine.org. 28 September 2022. Retrieved 4 November 2022.
  7. Alemany R (March 2013). "Viruses in cancer treatment". Clinical & Translational Oncology. 15 (3): 182–8. doi: 10.1007/s12094-012-0951-7 . PMID   23143950. S2CID   6123610.
  8. Donnelly OG, Errington-Mais F, Prestwich R, Harrington K, Pandha H, Vile R, Melcher AA (July 2012). "Recent clinical experience with oncolytic viruses". Current Pharmaceutical Biotechnology. 13 (9): 1834–41. doi:10.2174/138920112800958904. PMID   21740364.
  9. Roberts MS, Lorence RM, Groene WS, Bamat MK (August 2006). "Naturally oncolytic viruses". Current Opinion in Molecular Therapeutics. 8 (4): 314–21. PMID   16955694.
  10. Rudin CM, Poirier JT, Senzer NN, Stephenson J, Loesch D, Burroughs KD, Reddy PS, Hann CL, Hallenbeck PL (February 2011). "Phase I clinical study of Seneca Valley Virus (SVV-001), a replication-competent picornavirus, in advanced solid tumors with neuroendocrine features". Clinical Cancer Research. 17 (4): 888–95. doi:10.1158/1078-0432.CCR-10-1706. PMC   5317273 . PMID   21304001.
  11. "Rigvir šķīdums injekcijām". Medicinal product register of the Republic of Latvia. 29 April 2004. Retrieved 8 December 2016.
  12. 1 2 Frew SE, Sammut SM, Shore AF, Ramjist JK, Al-Bader S, Rezaie R, Daar AS, Singer PA (January 2008). "Chinese health biotech and the billion-patient market". Nature Biotechnology. 26 (1): 37–53. doi:10.1038/nbt0108-37. PMC   7096943 . PMID   18183014.
  13. Broderick J (29 April 2015). "FDA Panels Support Approval of T-VEC in Melanoma". OncLive. Retrieved 24 August 2015.
  14. Research, Center for Drug Evaluation and (29 December 2022). "FDA approves first adenoviral vector-based gene therapy for high-risk Bacillus Calmette-Guérin unresponsive non-muscle invasive bladder cancer". FDA.
  15. 1 2 3 4 5 6 Kuruppu D, Tanabe KK (May 2005). "Viral oncolysis by herpes simplex virus and other viruses". Cancer Biology & Therapy. 4 (5): 524–31. doi: 10.4161/cbt.4.5.1820 . PMID   15917655.
  16. 1 2 Voroshilova MK (1989). "Potential use of nonpathogenic enteroviruses for control of human disease". Progress in Medical Virology. Fortschritte der Medizinischen Virusforschung. Progrès en Virologie Médicale. 36: 191–202. PMID   2555836.
  17. 1 2 Pond AR, Manuelidis EE (August 1964). "Oncolytic Effect of Poliomyelitis Virus on Human Epidermoid Carcinoma (Hela Tumor) Heterologously Transplanted to Guinea Pigs". The American Journal of Pathology. 45 (2): 233–49. PMC   1907181 . PMID   14202523.
  18. 1 2 Kunin CM (December 1964). "Cellular Susceptibility to Enteroviruses". Bacteriological Reviews. 28 (4): 382–90. doi:10.1128/MMBR.28.4.382-390.1964. PMC   441234 . PMID   14244713.
  19. Chumakov PM, Morozova VV, Babkin IV, Baĭkov IK, Netesov SV, Tikunova NV (2012). "[Oncolytic enteroviruses]". Molekuliarnaia Biologiia (in Russian). 46 (5): 712–25. doi:10.1134/s0026893312050032. PMID   23156670. S2CID   3716727.
  20. Kelly E, Russell SJ (April 2007). "History of oncolytic viruses: genesis to genetic engineering". Molecular Therapy. 15 (4): 651–9. doi: 10.1038/sj.mt.6300108 . PMID   17299401.
  21. MacLean AR, ul-Fareed M, Robertson L, Harland J, Brown SM (March 1991). "Herpes simplex virus type 1 deletion variants 1714 and 1716 pinpoint neurovirulence-related sequences in Glasgow strain 17+ between immediate early gene 1 and the 'a' sequence". The Journal of General Virology. 72 ( Pt 3) (3): 631–9. doi: 10.1099/0022-1317-72-3-631 . PMID   1848598.
  22. Brown SM, Harland J, MacLean AR, Podlech J, Clements JB (September 1994). "Cell type and cell state determine differential in vitro growth of non-neurovirulent ICP34.5-negative herpes simplex virus types 1 and 2". The Journal of General Virology. 75 ( Pt 9) (9): 2367–77. doi: 10.1099/0022-1317-75-9-2367 . PMID   8077935.
  23. Kesari S, Randazzo BP, Valyi-Nagy T, Huang QS, Brown SM, MacLean AR, Lee VM, Trojanowski JQ, Fraser NW (November 1995). "Therapy of experimental human brain tumors using a neuroattenuated herpes simplex virus mutant". Laboratory Investigation; A Journal of Technical Methods and Pathology. 73 (5): 636–48. PMID   7474937.
  24. McKie EA, MacLean AR, Lewis AD, Cruickshank G, Rampling R, Barnett SC, Kennedy PG, Brown SM (September 1996). "Selective in vitro replication of herpes simplex virus type 1 (HSV-1) ICP34.5 null mutants in primary human CNS tumours--evaluation of a potentially effective clinical therapy". British Journal of Cancer. 74 (5): 745–52. doi:10.1038/bjc.1996.431. PMC   2074706 . PMID   8795577.
  25. Randazzo BP, Bhat MG, Kesari S, Fraser NW, Brown SM (June 1997). "Treatment of experimental subcutaneous human melanoma with a replication-restricted herpes simplex virus mutant". The Journal of Investigative Dermatology. 108 (6): 933–7. doi: 10.1111/1523-1747.ep12295238 . PMID   9182825.
  26. Rampling R, Cruickshank G, Papanastassiou V, Nicoll J, Hadley D, Brennan D, Petty R, MacLean A, Harland J, McKie E, Mabbs R, Brown M (May 2000). "Toxicity evaluation of replication-competent herpes simplex virus (ICP 34.5 null mutant 1716) in patients with recurrent malignant glioma". Gene Therapy. 7 (10): 859–66. doi: 10.1038/sj.gt.3301184 . PMID   10845724.
  27. Papanastassiou V, Rampling R, Fraser M, Petty R, Hadley D, Nicoll J, Harland J, Mabbs R, Brown M (March 2002). "The potential for efficacy of the modified (ICP 34.5(-)) herpes simplex virus HSV1716 following intratumoural injection into human malignant glioma: a proof of principle study". Gene Therapy. 9 (6): 398–406. doi: 10.1038/sj.gt.3301664 . PMID   11960316.
  28. Harrow S, Papanastassiou V, Harland J, Mabbs R, Petty R, Fraser M, Hadley D, Patterson J, Brown SM, Rampling R (November 2004). "HSV1716 injection into the brain adjacent to tumour following surgical resection of high-grade glioma: safety data and long-term survival". Gene Therapy. 11 (22): 1648–58. doi: 10.1038/sj.gt.3302289 . PMID   15334111.
  29. MacKie RM, Stewart B, Brown SM (February 2001). "Intralesional injection of herpes simplex virus 1716 in metastatic melanoma". Lancet. 357 (9255): 525–6. doi:10.1016/S0140-6736(00)04048-4. PMID   11229673. S2CID   34442464.
  30. Mace AT, Ganly I, Soutar DS, Brown SM (August 2008). "Potential for efficacy of the oncolytic Herpes simplex virus 1716 in patients with oral squamous cell carcinoma". Head & Neck. 30 (8): 1045–51. doi:10.1002/hed.20840. PMID   18615711. S2CID   43914133.
  31. Conner J, Braidwood L, Brown SM (December 2008). "A strategy for systemic delivery of the oncolytic herpes virus HSV1716: redirected tropism by antibody-binding sites incorporated on the virion surface as a glycoprotein D fusion protein". Gene Therapy. 15 (24): 1579–92. doi: 10.1038/gt.2008.121 . PMID   18701918.
  32. Braidwood L, Dunn PD, Hardy S, Evans TR, Brown SM (June 2009). "Antitumor activity of a selectively replication competent herpes simplex virus (HSV) with enzyme prodrug therapy". Anticancer Research. 29 (6): 2159–66. PMID   19528476.
  33. Sorensen A, Mairs RJ, Braidwood L, Joyce C, Conner J, Pimlott S, Brown M, Boyd M (April 2012). "In vivo evaluation of a cancer therapy strategy combining HSV1716-mediated oncolysis with gene transfer and targeted radiotherapy". Journal of Nuclear Medicine. 53 (4): 647–54. doi: 10.2967/jnumed.111.090886 . PMID   22414636.
  34. Turnbull S, West EJ, Scott KJ, Appleton E, Melcher A, Ralph C (December 2015). "Evidence for Oncolytic Virotherapy: Where Have We Got to and Where Are We Going?". Viruses. 7 (12): 6291–312. doi: 10.3390/v7122938 . PMC   4690862 . PMID   26633468.
  35. Conry RM, Westbrook B, McKee S, Norwood TG (February 2018). "Talimogene laherparepvec: First in class oncolytic virotherapy". Human Vaccines & Immunotherapeutics. 14 (4): 839–846. doi:10.1080/21645515.2017.1412896. PMC   5893211 . PMID   29420123.
  36. 1 2 3 4 5 Garber K (March 2006). "China approves world's first oncolytic virus therapy for cancer treatment". Journal of the National Cancer Institute. 98 (5): 298–300. doi: 10.1093/jnci/djj111 . PMID   16507823.
  37. Ayllón Barbellido S, Campo Trapero J, Cano Sánchez J, Perea García MA, Escudero Castaño N, Bascones Martínez A (January 2008). "Gene therapy in the management of oral cancer: review of the literature" (PDF). Medicina Oral, Patologia Oral y Cirugia Bucal. 13 (1): E15–21. PMID   18167474.
  38. Guo J, Xin H (November 2006). "Chinese gene therapy. Splicing out the West?". Science. 314 (5803): 1232–5. doi:10.1126/science.314.5803.1232. PMID   17124300. S2CID   142897522.
  39. 1 2 3 Marin-Acevedo JA, Soyano AE, Dholaria B, Knutson KL, Lou Y (January 2018). "Cancer immunotherapy beyond immune checkpoint inhibitors". Journal of Hematology & Oncology. 11 (1): 8. doi: 10.1186/s13045-017-0552-6 . PMC   5767051 . PMID   29329556.
  40. Schmidt C (May 2013). "Awaiting a moment of truth for oncolytic viruses". Journal of the National Cancer Institute. 105 (10): 675–6. doi: 10.1093/jnci/djt111 . PMID   23650626.
  41. Kottke T, Thompson J, Diaz RM, Pulido J, Willmon C, Coffey M, Selby P, Melcher A, Harrington K, Vile RG (January 2009). "Improved systemic delivery of oncolytic reovirus to established tumors using preconditioning with cyclophosphamide-mediated Treg modulation and interleukin-2". Clinical Cancer Research. 15 (2): 561–9. doi:10.1158/1078-0432.CCR-08-1688. PMC   3046733 . PMID   19147761.
  42. Lolkema MP, Arkenau HT, Harrington K, Roxburgh P, Morrison R, Roulstone V, Twigger K, Coffey M, Mettinger K, Gill G, Evans TR, de Bono JS (February 2011). "A phase I study of the combination of intravenous reovirus type 3 Dearing and gemcitabine in patients with advanced cancer". Clinical Cancer Research. 17 (3): 581–8. doi: 10.1158/1078-0432.CCR-10-2159 . PMID   21106728.
  43. Magge D, Guo ZS, O'Malley ME, Francis L, Ravindranathan R, Bartlett DL (June 2013). "Inhibitors of C5 complement enhance vaccinia virus oncolysis". Cancer Gene Therapy. 20 (6): 342–50. doi:10.1038/cgt.2013.26. PMC   4060830 . PMID   23661042.
  44. Heo J, Reid T, Ruo L, Breitbach CJ, Rose S, Bloomston M, Cho M, Lim HY, Chung HC, Kim CW, Burke J, Lencioni R, Hickman T, Moon A, Lee YS, Kim MK, Daneshmand M, Dubois K, Longpre L, Ngo M, Rooney C, Bell JC, Rhee BG, Patt R, Hwang TH, Kirn DH (March 2013). "Randomized dose-finding clinical trial of oncolytic immunotherapeutic vaccinia JX-594 in liver cancer". Nature Medicine. 19 (3): 329–36. doi:10.1038/nm.3089. PMC   4268543 . PMID   23396206.
  45. Wonganan P, Croyle MA (February 2010). "PEGylated Adenoviruses: From Mice to Monkeys". Viruses. 2 (2): 468–502. doi: 10.3390/v2020468 . PMC   3185605 . PMID   21994645.
  46. Muthana, Munitta; Rodrigues, Samuel; Chen, Yung-Yi; Welford, Abigail; Hughes, Russell; Tazzyman, Simon; Essand, Magnus; Morrow, Fiona; Lewis, Claire E. (15 January 2013). "Macrophage delivery of an oncolytic virus abolishes tumor regrowth and metastasis after chemotherapy or irradiation". Cancer Research. 73 (2): 490–495. doi: 10.1158/0008-5472.CAN-12-3056 . ISSN   1538-7445. PMID   23172310.
  47. Tong AW, Senzer N, Cerullo V, Templeton NS, Hemminki A, Nemunaitis J (July 2012). "Oncolytic viruses for induction of anti-tumor immunity". Current Pharmaceutical Biotechnology. 13 (9): 1750–60. doi:10.2174/138920112800958913. PMID   21740355.
  48. Naik JD, Twelves CJ, Selby PJ, Vile RG, Chester JD (July 2011). "Immune recruitment and therapeutic synergy: keys to optimizing oncolytic viral therapy?". Clinical Cancer Research. 17 (13): 4214–24. doi:10.1158/1078-0432.CCR-10-2848. PMC   3131422 . PMID   21576084.
  49. Keller, Brian A.; Bell, John C. (1 September 2016). "Oncolytic viruses—immunotherapeutics on the rise". Journal of Molecular Medicine. 94 (9): 979–991. doi:10.1007/s00109-016-1453-9. ISSN   1432-1440. PMID   27492706. S2CID   253638074.
  50. O'Regan B, Hirshberg C (1993). Spontaneous Remission: An Annotated Bibliography. Sausalito, California: Institute of Noetic Sciences. ISBN   978-0-943951-17-1. Archived from the original on 21 March 2015. Retrieved 31 March 2013.[ page needed ]
  51. Lattime E (2013). Gene Therapy of Cancer: Translational Approaches from Preclinical Studies to Clinical Implementation. Academic Press. ISBN   978-0-12-394295-1.[ page needed ]
  52. Mastrangelo MJ, Lattime EC (December 2002). "Virotherapy clinical trials for regional disease: in situ immune modulation using recombinant poxvirus vectors". Cancer Gene Therapy. 9 (12): 1013–21. doi: 10.1038/sj.cgt.7700538 . PMID   12522440.
  53. Lundstrom K (2018). "New frontiers in oncolytic viruses: optimizing and selecting for virus strains with improved efficacy". Biologics: Targets and Therapy. 12: 43–60. doi: 10.2147/BTT.S140114 . PMC   5810530 . PMID   29445265.
  54. "Oncolytic Viruses in cancer therapy" . Retrieved 11 April 2023.
  55. Stojdl DF, Lichty B, Knowles S, Marius R, Atkins H, Sonenberg N, Bell JC (July 2000). "Exploiting tumor-specific defects in the interferon pathway with a previously unknown oncolytic virus". Nature Medicine. 6 (7): 821–5. doi:10.1038/77558. PMID   10888934. S2CID   8492631.
  56. 1 2 Stojdl DF, Lichty BD, tenOever BR, Paterson JM, Power AT, Knowles S, Marius R, Reynard J, Poliquin L, Atkins H, Brown EG, Durbin RK, Durbin JE, Hiscott J, Bell JC (October 2003). "VSV strains with defects in their ability to shutdown innate immunity are potent systemic anti-cancer agents". Cancer Cell. 4 (4): 263–75. doi: 10.1016/S1535-6108(03)00241-1 . PMID   14585354.
  57. Ahmed M, Cramer SD, Lyles DS (December 2004). "Sensitivity of prostate tumors to wild type and M protein mutant vesicular stomatitis viruses". Virology. 330 (1): 34–49. doi: 10.1016/j.virol.2004.08.039 . PMID   15527832.
  58. Ebert O, Harbaran S, Shinozaki K, Woo SL (April 2005). "Systemic therapy of experimental breast cancer metastases by mutant vesicular stomatitis virus in immune-competent mice". Cancer Gene Therapy. 12 (4): 350–8. doi: 10.1038/sj.cgt.7700794 . PMID   15565179.
  59. Porosnicu M, Mian A, Barber GN (December 2003). "The oncolytic effect of recombinant vesicular stomatitis virus is enhanced by expression of the fusion cytosine deaminase/uracil phosphoribosyltransferase suicide gene". Cancer Research. 63 (23): 8366–76. PMID   14678998.
  60. Bridle BW, Stephenson KB, Boudreau JE, Koshy S, Kazdhan N, Pullenayegum E, Brunellière J, Bramson JL, Lichty BD, Wan Y (August 2010). "Potentiating cancer immunotherapy using an oncolytic virus". Molecular Therapy. 18 (8): 1430–9. doi:10.1038/mt.2010.98. PMC   2927075 . PMID   20551919.
  61. Gromeier M, Lachmann S, Rosenfeld MR, Gutin PH, Wimmer E (June 2000). "Intergeneric poliovirus recombinants for the treatment of malignant glioma". Proceedings of the National Academy of Sciences of the United States of America. 97 (12): 6803–8. Bibcode:2000PNAS...97.6803G. doi: 10.1073/pnas.97.12.6803 . JSTOR   122718. PMC   18745 . PMID   10841575.
  62. Goetz C, Gromeier M (2010). "Preparing an oncolytic poliovirus recombinant for clinical application against glioblastoma multiforme". Cytokine & Growth Factor Reviews. 21 (2–3): 197–203. doi:10.1016/j.cytogfr.2010.02.005. PMC   2881183 . PMID   20299272.
  63. Lal R, Harris D, Postel-Vinay S, de Bono J (October 2009). "Reovirus: Rationale and clinical trial update". Current Opinion in Molecular Therapeutics. 11 (5): 532–9. PMID   19806501.
  64. Thirukkumaran C, Morris DG (2009). "Oncolytic Viral Therapy Using Reovirus". Gene Therapy of Cancer. Methods in Molecular Biology. Vol. 542. pp. 607–34. doi:10.1007/978-1-59745-561-9_31. ISBN   978-1-934115-85-5. PMID   19565924.
  65. "Latvijas Zāļu reģistrs". www.zva.gov.lv. Retrieved 17 December 2017.
  66. "Georgia Today".
  67. "Latvian Rigvir anti-cancer medicine registered in Armenia". The Baltic Course. 11 May 2016. Retrieved 3 January 2018.
  68. 1 2 3 Babiker, HM; Riaz, IB; Husnain, M; Borad, MJ (2017). "Oncolytic virotherapy including Rigvir and standard therapies in malignant melanoma". Oncolytic Virotherapy. 6: 11–18. doi: 10.2147/OV.S100072 . PMC   5308590 . PMID   28224120.
  69. "Feasibility study for registration of medicine RIGVIR with the European Medicine Agency". European Commission. 8 January 2016. Archived from the original on 2 November 2016. Retrieved 2 November 2016. However, further use and commercialisation in the EU is prevented as EU regulations require cancer medicines to be registered centrally through the European Medicine Agency (EMA). National registrations are not considered.
  70. Gorski D (18 September 2017). "Rigvir: Another unproven and dubious cancer therapy to be avoided". Science-Based Medicine.
  71. Gorski, David (25 September 2017). "Ty Bollinger's "The Truth About Cancer" and the unethical marketing of the unproven cancer virotherapy Rigvir". Science-Based Medicine.
  72. "Rigvir medication distribution in Latvia halted temporarily". 19 March 2019.
  73. "Rigvir cancer treatment at center of fresh controversy". eng.lsm.lv.
  74. "Apturēta Rigvir reģistrācija; informācija esošajiem pacientiem | Zāļu valsts aģentūra".
  75. 1 2 Ramachandran M, Yu D, Dyczynski M, Baskaran S, Zhang L, Lulla A, Lulla V, Saul S, Nelander S, Dimberg A, Merits A, Leja-Jarblad J, Essand M (March 2017). "Safe and Effective Treatment of Experimental Neuroblastoma and Glioblastoma Using Systemically Delivered Triple MicroRNA-Detargeted Oncolytic Semliki Forest Virus". Clinical Cancer Research. 23 (6): 1519–1530. doi: 10.1158/1078-0432.CCR-16-0925 . PMID   27637889.
  76. Clinical trial number NCT02285816 for "MG1 Maraba/MAGE-A3, With and Without Adenovirus Vaccine, With Transgenic MAGE-A3 Insertion in Patients With Incurable MAGE-A3-Expressing Solid Tumours (I214)" at ClinicalTrials.gov
  77. Annels, Nicola E; Mansfield, David; Arif, Mehreen; Ballesteros-Merino, Carmen; Simpson, Guy R; Denyer, Mick; Sandhu, Sarbjinder S; Melcher, Alan; Harrington, Kevin J; Davies, BronwYn; Au, Gough; Grose, Mark; Bagwan, Izhar N; Fox, Bernard A.; Vile, Richard G; Mostafid, Hugh; Shafren, Darren; Pandha, Hardev (2019). "Viral targeting of non-muscle invasive bladder cancer and priming of anti-tumour immunity following intravesical Coxsackievirus A21" (PDF). Clinical Cancer Research. 25 (19): 5818–5831. doi: 10.1158/1078-0432.CCR-18-4022 . ISSN   1078-0432. PMID   31273010.
  78. Van Leer-Buter, Coretta C.; Poelman, Randy; Borger, Renze; Niesters, Hubert G. M.; Tang, Y.-W. (2016). "Newly Identified Enterovirus C Genotypes, Identified in the Netherlands through Routine Sequencing of All Enteroviruses Detected in Clinical Materials from 2008 to 2015". Journal of Clinical Microbiology. 54 (9): 2306–2314. doi:10.1128/JCM.00207-16. ISSN   0095-1137. PMC   5005491 . PMID   27358467.
  79. Dock, George. "The influence of complicating diseases upon leukaemia". The American Journal of the Medical Sciences (1827-1924); Philadelphia. 127 (4).
  80. Kabiljo, Julijan; Laengle, Johannes; Bergmann, Michael (2020). "From threat to cure: understanding of virus-induced cell death leads to highly immunogenic oncolytic influenza viruses". Cell Death Discovery. 6: 48. doi:10.1038/s41420-020-0284-1. ISSN   2058-7716. PMC   7288254 . PMID   32542113.
  81. 1 2 Kuhn I, Harden P, Bauzon M, Chartier C, Nye J, Thorne S, Reid T, Ni S, Lieber A, Fisher K, Seymour L, Rubanyi GM, Harkins RN, Hermiston TW (June 2008). "Directed evolution generates a novel oncolytic virus for the treatment of colon cancer". PLOS ONE. 3 (6): e2409. Bibcode:2008PLoSO...3.2409K. doi: 10.1371/journal.pone.0002409 . PMC   2423470 . PMID   18560559.
  82. Chow AY. "Cell Cycle Control by Oncogenes and Tumor Suppressors: Driving the Transformation of Normal Cells into Cancerous Cells". Nature Education. 3 (9): 7. Retrieved 5 April 2013.
  83. "Thymidine kinase". Medical Dictionary. Merriam-Webster. Retrieved 5 April 2013.
  84. Gentry GA (1992). "Viral thymidine kinases and their relatives". Pharmacology & Therapeutics. 54 (3): 319–55. doi:10.1016/0163-7258(92)90006-L. PMID   1334563.
  85. 1 2 3 4 Singh PK, Doley J, Kumar GR, Sahoo AP, Tiwari AK (October 2012). "Oncolytic viruses & their specific targeting to tumour cells". The Indian Journal of Medical Research. 136 (4): 571–84. PMC   3516024 . PMID   23168697.
  86. Davydova J, Le LP, Gavrikova T, Wang M, Krasnykh V, Yamamoto M (June 2004). "Infectivity-enhanced cyclooxygenase-2-based conditionally replicative adenoviruses for esophageal adenocarcinoma treatment". Cancer Research. 64 (12): 4319–27. doi: 10.1158/0008-5472.CAN-04-0064 . PMID   15205347.
  87. 1 2 3 Haddad D, Chen CH, Carlin S, Silberhumer G, Chen NG, Zhang Q, Longo V, Carpenter SG, Mittra A, Carson J, Au J, Gonen M, Zanzonico PB, Szalay AA, Fong Y (2012). Gelovani JG (ed.). "Imaging characteristics, tissue distribution, and spread of a novel oncolytic vaccinia virus carrying the human sodium iodide symporter". PLOS ONE. 7 (8): e41647. Bibcode:2012PLoSO...741647H. doi: 10.1371/journal.pone.0041647 . PMC   3422353 . PMID   22912675.
  88. Poirier JT, Reddy PS, Idamakanti N, Li SS, Stump KL, Burroughs KD, Hallenbeck PL, Rudin CM (December 2012). "Characterization of a full-length infectious cDNA clone and a GFP reporter derivative of the oncolytic picornavirus SVV-001". The Journal of General Virology. 93 (Pt 12): 2606–13. doi: 10.1099/vir.0.046011-0 . PMID   22971818.
  89. Yu YA, Shabahang S, Timiryasova TM, Zhang Q, Beltz R, Gentschev I, Goebel W, Szalay AA (March 2004). "Visualization of tumors and metastases in live animals with bacteria and vaccinia virus encoding light-emitting proteins". Nature Biotechnology. 22 (3): 313–20. doi:10.1038/nbt937. PMID   14990953. S2CID   1063835.
  90. Freeman SM, Whartenby KA, Freeman JL, Abboud CN, Marrogi AJ (February 1996). "In situ use of suicide genes for cancer therapy". Seminars in Oncology. 23 (1): 31–45. PMID   8607030.
  91. Duarte S, Carle G, Faneca H, de Lima MC, Pierrefite-Carle V (November 2012). "Suicide gene therapy in cancer: where do we stand now?". Cancer Letters. 324 (2): 160–70. doi:10.1016/j.canlet.2012.05.023. hdl: 10316/24816 . PMID   22634584.
  92. Frentzen A, Yu YA, Chen N, Zhang Q, Weibel S, Raab V, Szalay AA (August 2009). "Anti-VEGF single-chain antibody GLAF-1 encoded by oncolytic vaccinia virus significantly enhances antitumor therapy". Proceedings of the National Academy of Sciences of the United States of America. 106 (31): 12915–20. Bibcode:2009PNAS..10612915F. doi: 10.1073/pnas.0900660106 . JSTOR   40484625. PMC   2722284 . PMID   19617539.
  93. Conner J, Braidwood L (July 2012). "Expression of inhibitor of growth 4 by HSV1716 improves oncolytic potency and enhances efficacy". Cancer Gene Therapy. 19 (7): 499–507. doi: 10.1038/cgt.2012.24 . PMID   22595793.
  94. Grünwald GK, Klutz K, Willhauck MJ, Schwenk N, Senekowitsch-Schmidtke R, Schwaiger M, Zach C, Göke B, Holm PS, Spitzweg C (June 2013). "Sodium iodide symporter (NIS)-mediated radiovirotherapy of hepatocellular cancer using a conditionally replicating adenovirus". Gene Therapy. 20 (6): 625–33. doi: 10.1038/gt.2012.79 . PMID   23038026.
  95. Penheiter AR, Wegman TR, Classic KL, Dingli D, Bender CE, Russell SJ, Carlson SK (August 2010). "Sodium iodide symporter (NIS)-mediated radiovirotherapy for pancreatic cancer". AJR. American Journal of Roentgenology. 195 (2): 341–9. doi:10.2214/AJR.09.3672. PMC   3117397 . PMID   20651188.
  96. Li H, Peng KW, Dingli D, Kratzke RA, Russell SJ (August 2010). "Oncolytic measles viruses encoding interferon beta and the thyroidal sodium iodide symporter gene for mesothelioma virotherapy". Cancer Gene Therapy. 17 (8): 550–8. doi:10.1038/cgt.2010.10. PMC   2907639 . PMID   20379224.
  97. Clinical trial number NCT00769704 for "Efficacy and Safety Study of OncoVEXGM-CSF Compared to GM-CSF in Melanoma" at ClinicalTrials.gov
  98. "FDA approves Amgen's Injected Immunotherapy for Melanoma". Reuters. 27 October 2015.
  99. Sheridan C (June 2015). "First oncolytic virus edges towards approval in surprise vote". Nature Biotechnology. 33 (6): 569–70. doi:10.1038/nbt0615-569. PMID   26057953. S2CID   205268968.
  100. "Amgen, Form 8-K, Current Report, Filing Date Jan 26, 2012" (PDF). secdatabase.com. Retrieved 8 January 2013.
  101. Clinical trial number NCT01161498 for "Study of Safety and Efficacy of OncoVEXGM-CSF With Cisplatin for Treatment of Locally Advanced Head and Neck Cancer" at ClinicalTrials.gov
  102. "Delytact scores a first with Japanese approval for malignant glioma". www.thepharmaletter.com. Retrieved 21 December 2022.
  103. Ottolino-Perry K, Diallo JS, Lichty BD, Bell JC, McCart JA (February 2010). "Intelligent design: combination therapy with oncolytic viruses". Molecular Therapy. 18 (2): 251–63. doi:10.1038/mt.2009.283. PMC   2839289 . PMID   20029399.
  104. Khuri FR, Nemunaitis J, Ganly I, Arseneau J, Tannock IF, Romel L, Gore M, Ironside J, MacDougall RH, Heise C, Randlev B, Gillenwater AM, Bruso P, Kaye SB, Hong WK, Kirn DH (August 2000). "a controlled trial of intratumoral ONYX-015, a selectively-replicating adenovirus, in combination with cisplatin and 5-fluorouracil in patients with recurrent head and neck cancer". Nature Medicine. 6 (8): 879–85. doi:10.1038/78638. PMID   10932224. S2CID   3199209.
  105. Mell LK, Brumund KT, Daniels GA, Advani SJ, Zakeri K, Wright ME, Onyeama SJ, Weisman RA, Sanghvi PR, Martin PJ, Szalay AA (October 2017). "Phase I Trial of Intravenous Oncolytic Vaccinia Virus (GL-ONC1) with Cisplatin and Radiotherapy in Patients with Locoregionally Advanced Head and Neck Carcinoma" (PDF). Clinical Cancer Research. 23 (19): 5696–5702. doi: 10.1158/1078-0432.CCR-16-3232 . PMID   28679776. S2CID   30604400.
  106. Suryawanshi YR, Zhang T, Essani K (March 2017). "Oncolytic viruses: emerging options for the treatment of breast cancer". Medical Oncology. 34 (3): 43. doi:10.1007/s12032-017-0899-0. PMID   28185165. S2CID   44562857.
  107. Chen Y, DeWeese T, Dilley J, Zhang Y, Li Y, Ramesh N, Lee J, Pennathur-Das R, Radzyminski J, Wypych J, Brignetti D, Scott S, Stephens J, Karpf DB, Henderson DR, Yu DC (July 2001). "CV706, a prostate cancer-specific adenovirus variant, in combination with radiotherapy produces synergistic antitumor efficacy without increasing toxicity". Cancer Research. 61 (14): 5453–60. PMID   11454691.
  108. Mace AT, Harrow SJ, Ganly I, Brown SM (August 2007). "Cytotoxic effects of the oncolytic herpes simplex virus HSV1716 alone and in combination with cisplatin in head and neck squamous cell carcinoma". Acta Oto-Laryngologica. 127 (8): 880–7. doi:10.1080/00016480601075381. PMID   17763002. S2CID   44252457.
  109. Toyoizumi T, Mick R, Abbas AE, Kang EH, Kaiser LR, Molnar-Kimber KL (December 1999). "Combined therapy with chemotherapeutic agents and herpes simplex virus type 1 ICP34.5 mutant (HSV-1716) in human non-small cell lung cancer". Human Gene Therapy. 10 (18): 3013–29. doi:10.1089/10430349950016410. PMID   10609661. S2CID   20072243.
  110. Currier MA, Eshun FK, Sholl A, Chernoguz A, Crawford K, Divanovic S, Boon L, Goins WF, Frischer JS, Collins MH, Leddon JL, Baird WH, Haseley A, Streby KA, Wang PY, Hendrickson BW, Brekken RA, Kaur B, Hildeman D, Cripe TP (May 2013). "VEGF blockade enables oncolytic cancer virotherapy in part by modulating intratumoral myeloid cells". Molecular Therapy. 21 (5): 1014–23. doi:10.1038/mt.2013.39. PMC   3666636 . PMID   23481323.
  111. Frentzen A, Yu YA, Chen N, Zhang Q, Weibel S, Raab V, Szalay AA (August 2009). "Anti-VEGF single-chain antibody GLAF-1 encoded by oncolytic vaccinia virus significantly enhances antitumor therapy". Proceedings of the National Academy of Sciences of the United States of America. 106 (31): 12915–20. Bibcode:2009PNAS..10612915F. doi: 10.1073/pnas.0900660106 . PMC   2722284 . PMID   19617539.
  112. Williamson J (2002). Dragon's Island and other stories . Waterville, Me.: Five Star. ISBN   978-0-7862-4314-3.[ page needed ]
  113. Stableford BM (2004). Historical dictionary of science fiction literature. Scarecrow Press. p. 133. ISBN   978-0-8108-4938-9.
  114. Dalhousie University (9 May 2008). "A Real-life 'I Am Legend?' Researcher Champions Development Of 'Reovirus' As Potential Treatment For Cancer". Science Daily.

Further reading