跳转到主要内容
搜索

2025 年 5 月 30 日

Gene therapies delivered in viral vectors offer an exciting and challenging approach to targeted cancer therapy. Systems modeling for oncolytic viruses can help drug developers characterize safe and effective single-agent and combination treatments involving oncolytic viruses.

Understanding viruses

The ability of viruses to insert their genome into a host cell is leveraged in modern molecular biology. For example, recombinant viral vectors can manipulate the genes of a cell or organism.

oncolytic virus

资料来源:https://www.sciencedirect.com/topics/immunology-and-microbiology/oncolytic-virus

Oncolytic viruses in cancer treatment

Since the mid-1800s, there have been multiple cases of cancer patients experiencing tumor regressions that coincided with viral infections. The viral infections activated the patient’s immune system, which then attacked the cancer. This was an early type of immuno-oncology treatment.

Early clinical trials tested the viability of purposefully infecting cancer patients with a virus [3]. Indeed, many patients experienced tumor regressions, however, many also experienced undesirable side effects of said viral infection (ex: hepatitis). Patients needed a safer alternative infection with dangerous viruses. Today, oncolytic viruses are an emerging class of cancer therapeutics.

Historical observations of viral infections associated with tumor regression

1896 – A woman with “myelogenous leukemia” went into remission after influenza infection, 37 years before influenza was determined to be a viral infection [1]

1953 – Chicken pox led to regression of lymphatic leukemia in a 4-year-old boy [2]

How do oncolytic viruses work as cancer therapeutics?

Non-cancerous cells and a competent immune system can arrest/eliminate a viral infection. By contrast, cancer cells often develop mutations that enable them to evade immune surveillance. However, such mutations also make them vulnerable to viral infections [4]. Modern research and development of oncolytic viruses is focused on

  1. creating “attenuated” recombinant viruses that can shrink tumors
  2. but have limited ability to infect healthy tissue
  3. and don’t cause undesirable side effects or toxicity.

There are two key mechanisms of action oncolytic viruses take. Most modern virotherapies utilize recombinant DNA technologies to attenuate a wild-type virus. This engineered virus preferentially infects cancer cells while having a limited ability to infect healthy cells. Additionally, recombinant oncolytic viruses can “reprogram” cancer cells to change their microenvironment to drive cancer cell killing.

As a vector for gene manipulation, an oncolytic virus could use several possible strategies to reprogram the tumor microenvironment. For example, an oncolytic virus could prevent cancerous cells from expressing proteins that enable immune system evasion. Alternatively, it could make infected cells express proteins that recruit immune cells to the tumor.

Challenges with the development and modeling of oncolytic viruses

Certain challenges are unique to the development of oncolytic viruses. One challenge with different oncolytic viruses is that they can have species-specific permissivity. A virus can have a different ability to infect and replicate in mouse versus human cells, for example. Therefore, it becomes difficult to translate preclinical doses to human applications.

Certara experts use modeling to bridge species differences and make human predictions. This approach can answer questions around toxicity and/or efficacy.

Another consideration is that oncolytic viruses are live, and upon clinical administration, will proliferate within the body, causing variable effective doses. Currently, there is not enough publicly available data to fully understand the relationship between viral replication and clinical outcomes. Understanding this relationship will be essential in establishing safe and effective doses. [5]

For small molecule drugs or monoclonal antibodies, we can correlate plasma drug measurements (pharmacokinetics; PK) to the efficacy/safety effects without understanding their mechanism of action. However, it’s difficult to relate the PK of the virus or other gene therapy vectors to efficacy/safety.

Understanding a viral therapy’s mechanism of action requires characterizing how cells take up genetic material and translate it into a protein. Mechanistic modeling can capture these subtleties because it models each step explicitly and uses species-specific information.

Unlike most drugs, the body doesn’t metabolize oncolytic viruses. Potential host’s antiviral immune responses include disabling the virus via circulating neutralizing antibodies or haemaggluttinin binding. In some cases, viruses evade immune detection.

Further research on how cancer patients respond to oncolytic viruses will be required to understand exposure and dose-response. [5]

Lastly, working with immunocompromised populations requires designing effective clinical trials and determining appropriate dosing regimens.

Market outlook

The market for oncolytic viruses currently has few approved therapies. However, the market is expected to grow rapidly over the next decade, with a projected CAGR of 27% from 2023 to 2033 [6].

Currently approved oncolytic viruses for clinical treatment

Oncorine (Shanghai Sunway Biotech) – approved by NMPA (China) in 2005 for nasopharyngeal carcinoma

Imlygic (Amgen) – approved by the FDA (USA) in 2015 for melanomas

Delytact (Daiichi Sankyo) – approved by Ministry of Health (Japan) in 2016 for glioblastomas

Antibody-based and cell therapies are still the dominant biotherapeutics for treating cancer. However, there is growing excitement around oncolytic viruses as mono- and combination therapies. A recent publication highlighted that the combination of intratumoral oncolytic virus (DNX-240) and a checkpoint PD1 inhibitor, pembrolizumab, “was safe with notable survival benefit in select patients.”[7] In June 2023, Oncolys announced preliminary data from a Phase II trial (NCT03921021) of its oncolytic virus treatment telomelysin combined with Merck’s Keytruda (pembrolizumab) in refractory gastroesophageal adenocarcinoma.

Certara’s experience with oncolytic viruses

To date, Certara scientists have worked on several oncolytic virus collaborations with such partners as OncoMyx. These collaborations helped predict whether the oncolytic virus will cause cytokine-mediated toxicity and the therapeutic and toxic effects of potential dosing schedules.

Using first principles, mechanistic models built at Certara can predict how uncertainties in virotherapy design lead to uncertainties in clinical efficacy/toxicity. Certara’s QSP software offers pre-built, validated model packs for gene therapy modalities allowing modelers and non-modelers to jumpstart their workflows.

Explore model packRequest a consultation on your oncolytic virus program.

参考文献

[1] Dock G. The influence of complicating diseases upon leukaemia. The American Journal of the Medical Sciences (1827-1924). 1904 Apr 1;127(4):563.

[2] Bierman HR, Crile DM, Dod KS, Kelly KH, Petrakis NI, White LP, Shimkin MB. Remissions in leukemia of childhood following acute infectious disease. Staphylococcus and streptococcus, varicella, and feline panleukopenias. Cancer. 1953 May;6(3):591-605.

[3] Kelly E, Russell SJ. History of oncolytic viruses: genesis to genetic engineering. Molecular therapy. 2007 Apr 1;15(4):651-9.

[4] Mondal M, Guo J, He P, Zhou D. Recent advances of oncolytic virus in cancer therapy. Human vaccines & immunotherapeutics. 2020 Oct 2;16(10):2389-402.

[5] Kaufman, H., Kohlhapp, F. & Zloza, A. Oncolytic viruses: a new class of immunotherapy drugs. Nat Rev Drug Discov 14, 642–662 (2015). https://doi.org/10.1038/nrd4663

[7] Nassiri, F., Patil, V., Yefet, L.S. et al. Oncolytic DNX-2401 virotherapy plus pembrolizumab in recurrent glioblastoma: a phase 1/2 trial. Nat Med 29, 1370–1378 (2023). https://doi.org/10.1038/s41591-023-02347-y

与 QSP 专家建立联系


沪ICP备2022021526号

Powered by Translations.com GlobalLink Web Software