Oncolytic viruses

Viral vectors and vaccines

Mounting research indicates that therapies centered around oncolytic viruses are making exciting strides in healthcare: these viruses have not only demonstrated a knack for effectively targeting and lysing tumor cells, operating as therapeutic agents that can spur a broad immune response, but they’ve also shown the remarkable ability to harness the power of a patient’s immune system, kickstarting it to recognize and destroy cancer cells. The task now on hand is to translate this promising concept into practical clinical applications beneficial to patients.

viral vector and vaccine manufacturing CDMO

The emerging landscape of oncolytic viruses

While only a handful of oncolytic virus therapies have received regulatory approval, the pipeline promises many more in the offing. Clinical trials have revealed the efficacy of oncolytic viruses in eliminating cancer cells that have proven resistant to standard therapies. When used in tandem with other cancer treatments, oncolytic viruses have shown significantly enhanced effectiveness. Over 200 clinical trials are in various stages, using a variety of viral backbones, such as adenovirus, herpes simplex virus (HSV) or reovirus. Oncolytic viruses are generally well tolerated; the most common side effects being brief bouts of fever, aches, and fatigue lasting for a couple of days.  Above all, they offer an innovative method to destroy tumors directly and activate the immune system to eradicate cancer cells throughout the body.

However, the path to the development and manufacturing of oncolytic viruses –like any emerging field– isn’t without its challenges. The actual production of oncolytic viruses, for instance, involves intricate processes and requires specialized expertise as well as advanced facilities. With the constantly evolving and complex regulatory landscape, navigating through it presents another hurdle. Furthermore, given that most oncolytic virus therapies spring from academic labs, the methods used for virus propagation and purification may not translate directly to scalable GMP conditions. These challenges underscore a strategic role for a CDMO partner with robust experience and proven track-record in oncolytic virus therapies.

Oncolytic virus clinical trial_CDMO_types of oncolytic viruses
Overview of  oncolytic virus clinical trials (N=225, as of November 2021, data analyzed from clinicaltrials.gov and EudraCT) per type of oncolytic virus (adenovirus, herpes simplex virus, vaccinia virus, reovirus, coxsackievirus, measles virus, vesicular stomatitis virus, and  other). Adapted from Oncolytic viruses: challenges and considerations in an evolving clinical landscape, Ulrich M Lauer & Julia Beil, Future Oncology, VOL 18, NO. 24, 2022

Development and manufacture of oncolytic viruses: the path to scale

To meet the growing demand for oncolytic viruses, there is a need to explore scalable manufacturing processes. While steps such as ultrafiltration/diafiltration and column chromatography are already scalable using existing technologies, scaling up the infection process for licensed product manufacturing typically requires the investigation of bioreactor technology. Potential methods might involve using adherent cells attached to microbeads or suspended cell cultures which can facilitate the production of oncolytic viruses at higher titers. It would be ideal if cell proliferation could occur in a serum-free medium to exclude animal-derived components in line with Good Manufacturing Practice (GMP). High-volume, disposable stirred tank bioreactors designed for single use might be most suitable for virus production. The downstream purification process, particularly the separation from subcellular structures, might necessitate improvements depending on the size and density of the virus particle.

Proper quality control, a critical part of GMP, assures relevant and necessary tests are performed and that all essential materials fulfill acceptable quality standards – this includes sampling, specifications, testing, documentation, and release procedures. By validating and qualifying these processes, we guarantee that our manufactured oncolytic viruses meet the highest standards of purity, potency, and safety for clinical use.

Overview of production, purification, and characterization of oncolytic viruses_clinical grade_CDMO
Overview of production, purification, and characterization of clinical grade oncolytic viruses (Adenovirus, Herpesvirus, Reovirus, Measles virus). Adapted from Moving oncolytic viruses into the clinic: clinical-grade production, purification, and characterization of diverse oncolytic viruses, Ungerechts et al., Mol Ther Methods Clin Dev. 2016; 3: 16018.

The initial step in the development and manufacture of oncolytic viruses begins with a certified, GMP-compliant master cell bank and a characterized master seed virus. The raw virus supernatant is primarily produced by infecting adherent producer cells at a low rate of infection, and then harvesting the oncolytic virus when extensive cytopathic effect is observed. The purification procedure generally consists of five standard steps that apply to all oncolytic virus platforms, including clarification to eliminate cellular debris, nuclease treatment to break down host cell nucleic acids, ion-exchange/size exclusion chromatography for virus purification, ultracentrifugation or tangential flow ultrafiltration/diafiltration for concentration and buffer interchange, and a terminal sterile filtration step.

In conclusion, the development and manufacture of oncolytic viruses for clinical trials and beyond require careful consideration of scalable production methods and adherence to rigorous quality control standards. By navigating these challenges and partnering with experienced CDMOs, the full potential of oncolytic virus therapies can be realized, offering new hope in the fight against cancer.

Development and manufacture of oncolytic viruses: the path to scale

To meet the growing demand for oncolytic viruses, there is a need to explore scalable manufacturing processes. While steps such as ultrafiltration/diafiltration and column chromatography are already scalable using existing technologies, scaling up the infection process for licensed product manufacturing typically requires the investigation of bioreactor technology. Potential methods might involve using adherent cells attached to microbeads or suspended cell cultures which can facilitate the production of oncolytic viruses at higher titers. It would be ideal if cell proliferation could occur in a serum-free medium to exclude animal-derived components in line with Good Manufacturing Practice (GMP). High-volume, disposable stirred tank bioreactors designed for single use might be most suitable for virus production. The downstream purification process, particularly the separation from subcellular structures, might necessitate improvements depending on the size and density of the virus particle.

Proper quality control, a critical part of GMP, assures relevant and necessary tests are performed and that all essential materials fulfill acceptable quality standards – this includes sampling, specifications, testing, documentation, and release procedures.

Adenovirus
  • Producer cells: HEK293, Per.C6, A549-derived pTG6559, N52.E6, All cells contain contiguous adenovirus E1A and E1B sequences
  • Culture vessels: Spinner, roller bottle, bioreactor, single-use bags
  • Virus purification method: a) CsCl-density gradient ultracentrifugation b) HPLC
  • Molecular characterization of virus identity: Endpoint PCR, restriction analysis, sequencing
  • Assays for analysis of efficiency of virus preparation: a) Potency assay at low MOI in different tumor cells versus normal cells, b) In vivo xenograft model
  • Other: In vivo: a) Biodistribution studies by PCR b) Virus replication in lungs of Syrian hamster
Herpesvirus
  • Producer cells: Vero, BHK
  • Culture vessels: Flasks, roller bottles, cell factories or bioreactors
  • Virus purification method: Centrifugation, size exclusion and ion exchange chromatography, membrane chromatography and filtration
  • Molecular characterization of virus identity: Antibody-based assays for virus species identity & presence of inserted genes, Southern blot and/or PCR
  • Assays for analysis of efficiency of virus preparation: Virus titration and potency of any inserted genes
  • Other: N/A
Reovirus
  • Producer cells: HEK293, suspension
  • Culture vessels: 100 L stir tank reactor
  • Virus purification method: Clarification of feed stock followed by UF/DF; anion exchange chromatography and size exclusion chromatography; terminal filtration
  • Molecular characterization of virus identity: Western blotting; Q-PCR and Sanger sequencing
  • Assays for analysis of efficiency of virus preparation: Virus titration
  • Other: N/A
Measles virus
  • Producer cells: Vero cells, adherent
  • Culture vessels: Cell factory
  • Virus purification method: Physical virus release from cells; clarification; DNase treatment; tangential flow filtration and diafiltration; polishing
  • Molecular characterization of virus identity:RNA by RT-PCR, sequencing; protein by western blot; transgene expression
  • Assays for analysis of efficiency of virus preparation: Virus titration; potency assay on test cells
  • Other: Stability (>6 years at ≤ -65°C)
Overview of production, purification, and characterization of clinical grade oncolytic viruses (Adenovirus, Herpesvirus, Reovirus, Measles virus). Adapted from Moving oncolytic viruses into the clinic: clinical-grade production, purification, and characterization of diverse oncolytic viruses, Ungerechts et al., Mol Ther Methods Clin Dev. 2016; 3: 16018.

The initial step in the development and manufacture of oncolytic viruses begins with a certified, GMP-compliant master cell bank and a characterized master seed virus. The raw virus supernatant is primarily produced by infecting adherent producer cells at a low rate of infection, and then harvesting the oncolytic virus when extensive cytopathic effect is observed. The purification procedure generally consists of five standard steps that apply to all oncolytic virus platforms, including clarification to eliminate cellular debris, nuclease treatment to break down host cell nucleic acids, ion-exchange/size exclusion chromatography for virus purification, ultracentrifugation or tangential flow ultrafiltration/diafiltration for concentration and buffer interchange, and a terminal sterile filtration step.

In conclusion, the development and manufacture of oncolytic viruses for clinical trials and beyond require careful consideration of scalable production methods and adherence to rigorous quality control standards. By navigating these challenges and partnering with experienced CDMOs, the full potential of oncolytic virus therapies can be realized, offering new hope in the fight against cancer.

Your CDMO of choice for oncolytic viruses

We are an end-to-end viral vector CDMO with a track record in developing and manufacturing a wide variety of oncolytic viruses. We have built processes from both adherent and suspension cell lines and offer upstream processing from small scale up to 1,000 L SUB scale. Our capabilities extend to viral vectors such as adenovirus, lentivirus, adeno-associated viruses (AAV), HSV and other viruses.

GMP recombinant protein manufacturing

We support you in developing and commercializing recombinant proteins and antibodies, enabling the treatment and prevention of life-threatening conditions.

GMP viral product manufacturing

We support your project with the development & commercialization of viral products enabling the treatment and prevention of life threatening conditions.

Aseptic fill and finish and lyophilization

We provide GMP certified drug product production capabilities with a small and large scale filling line, processing over a million vials per year.

Development of small to large scale production processes

We bring your product into the clinical phase with our small scale production, analytical development, lyophilization cycle development and upscaling services.

GMP viral product manufacturing

We support your project with the development & commercialization of viral products enabling the treatment and prevention of life threatening conditions.

GMP recombinant protein manufacturing

We support you in developing and commercializing recombinant proteins and antibodies, enabling the treatment and prevention of life-threatening conditions.

Aseptic fill and finish and lyophilization

We provide GMP certified drug product production capabilities with a small and large scale filling line, processing over a million vials per year.

Development of small to large scale production processes

We bring your product into the clinical phase with our small scale production, analytical development, lyophilization cycle development and upscaling services.