Strengthening Sweden’s resilience through pharmaceutical manufacturing

In an increasingly unstable world, Sweden is taking decisive steps to secure access to critical pharmaceuticals during crises and war.

As part of a broader government initiative, highlighted in a directive to the National Board of Health and Welfare and the Medical Products Agency, Sweden is investing in domestic manufacturing preparedness for life-saving medicines.

As illustrated in a recent article, NorthX Biologics demonstrates how local infrastructure can serve global needs. An example is the company’s expansion in Matfors, Sweden, where cutting-edge capabilities in vaccine and advanced therapeutics production are being built. Facilities like these are crucial, not only for public health resilience during pandemics but also as part of Sweden’s national security infrastructure.

In times when resilience and supply security are critical, we believe the key isn’t to start from scratch, but to build on what already exists. With diverse competencies, decades of experience, and established GMP facilities, we are a trusted partner in ensuring Sweden’s healthcare sovereignty.

Sweden’s strategy is clear: independence in producing critical healthcare products is no longer optional, it’s essential.

Read the article in Sundsvalls Tidning (in Swedish): Inifrån svenska storsatsningen i Matfors – Sundsvalls Tidning

Manufacturing stem cells for regenerative therapies

Stem cells have revolutionized the field of regenerative medicine, offering promising solutions for various medical conditions, including difficult-to-heal skin wounds. This review focuses on the background and manufacturing processes of skin cell-based therapies, particularly keratinocytes and adipose-derived mesenchymal stem cells (AD-MSCs), as highlighted in the thesis by Hady Shahin[1]. This article provides an overview of services offered by Contract Development and Manufacturing Organizations (CDMOs) involved in cell-based solution production.

Stem cell basics

Stem cells have unique abilities to self-renew and to recreate functional tissues. They can develop into many different cell types in the body during early life and growth[2]. Researchers study many different types of stem cells, including pluripotent stem cells (embryonic stem cells and induced pluripotent stem cells) and non-embryonic or somatic stem cells (commonly called adult stem cells)[2]. Pluripotent stem cells have the ability to differentiate into cells of the 3 main germ layers in the adult body[2].  Adult stem cells are found in specific anatomical locations and can differentiate to yield the specialized cell types of that tissue or organ[2]. They serve as an internal repair system that generates replacements for cells lost through normal wear and tear, injury, or disease.

Properties of stem cells

Stem cells have the remarkable potential to renew themselves and to differentiate into various specialized cell types[2].  When a stem cell divides, the resulting two daughter cells may be both stem cells, a stem cell and a more differentiated cell, or both more differentiated cells[2]. Discovering the mechanism behind self-renewal may make it possible to understand how cell fate is regulated during normal embryonic development and post-natally, or mis-regulated during aging or in the development of cancer[2].

The skin and regenerative therapies

The skin, the largest organ of the body, serves as a protective barrier against the external environment. It consists of three distinct layers: the epidermis, dermis, and subcutaneous adipose tissue[1]. The epidermis, primarily composed of keratinocytes, plays a crucial role in maintaining skin integrity and facilitating wound healing. Keratinocytes move from the basal layer to the surface, undergoing differentiation and forming a protective barrier[1].

Difficult-to-heal wounds, such as those caused by chronic diseases, trauma, or burns, pose significant challenges in clinical practice. These wounds often result in prolonged pain, infection, and impaired quality of life[1]. Regenerative advanced therapy medicinal products (ATMPs), including cell-based approaches, offer promising solutions for enhancing wound healing and improving patient outcomes[1].

Autologous vs allogenic skin ATMPs

Keratinocytes —the most abundant cell type in the epidermis— are instrumental in the re-epithelialization process during wound healing. They proliferate and migrate to cover the wound bed, forming new epidermal layers[1]. An autologous therapeutic approach involves harvesting skin biopsies from a patient’s healthy donor sites, isolating keratinocytes, expanding them, and reapplying them to the same patient after thorough characterization, quality control, and safety testing. The classical method for culturing keratinocytes includes enzymatic digestion of the epidermis, followed by expansion in culture media [1]. However, a major challenge in this process is the use of animal-derived products, which poses regulatory hurdles [1]. To address these challenges, Shahin’s thesis proposes a xeno-free workflow for keratinocyte isolation and expansion. The study validates the use of a xeno-free workflow to manufacture human keratinocytes as ATMP [1]. This approach ensures the production of keratinocytes that comply with regulatory standards, making them suitable for clinical applications [1].

The allogeneic approach, on the other hand, involves Adipose-Derived Mesenchymal Stem Cells (AD-MSCs) as a promising alternative to address the scalability challenges associated with keratinocytes, which are mature cells. AD-MSCs are multipotent stem cells isolated from adipose (fat) tissue. They possess the ability to differentiate into various cell types, including osteoblasts, chondrocytes, and adipocytes[1]. AD-MSCs are particularly attractive for regenerative therapies due to their ease of isolation, high yield, and immunomodulatory properties[1].

In the context of wound healing, AD-MSCs contribute to tissue repair by promoting angiogenesis, reducing inflammation, and enhancing collagen synthesis[1]. Shahin’s thesis explores the potential of AD-MSCs as an alternative to keratinocytes for treating difficult-to-heal wounds[1]. The study highlights the differentiation of AD-MSCs into keratinocyte-like cells through direct co-culture with keratinocytes[1]. This approach leverages the paracrine signaling between the two cell types to enhance the differentiation process[1].

Manufacturing cell therapies for clinical use

The manufacturing of stem cells for clinical applications involves several critical steps, including cell isolation, expansion, and quality control. Ensuring compliance with Good Manufacturing Practice (GMP) guidelines is essential to produce safe and effective cell-based therapies[1].

Every cell therapy product is special, NorthX Biologics adopts a flexible manufacturing operation where we together with the client tailor a manufacturing and testing process meeting the specific product’s exact requirements.

Proposed workflow for manufacturing a cell-based ATMP for wound healing

1. Cell isolation

Keratinocytes are typically isolated from skin biopsies using enzymatic digestion. Shahin’s thesis validates the use of a completely xeno-free keratinocytes extraction method, ensuring the production of GMP-compliant keratinocytes in a timely manner[1]. AD-MSCs are isolated from adipose tissue through enzymatic digestion and centrifugation[1]. The high yield of AD-MSCs from adipose tissue makes them a viable option for large-scale production[1].

2. Cell expansion

The expansion of keratinocytes and AD-MSCs requires optimized culture conditions to maintain cell viability and functionality. Shahin’s study demonstrates the use of xeno-free culture media for keratinocyte expansion, eliminating the need for animal-derived products[1]. For AD-MSCs, the co-culture with keratinocytes enhances their differentiation into keratinocyte-like cells, providing a scalable approach for producing epidermal cells [1].. Rigorous quality control is needed to ensure such in-vitro cell manipulation is safe and does not compromise the properties of the cells. Therefore, thorough characterization and stability testing are needed for cell-therapies to be considered safe for clinical use and to fulfil stringent regulatory requirements for ATMPs.

At NorthX Biologics, we provide an extensive range of testing services tailored to the advanced products we manufacture, to support designing a comprehensive analytics panel for your ATMP (consulting on release criteria with the regulatory bodies).

3. Cell transportation

As cell therapy manufacturing for clinical use must be conducted under strict control in a GMP facility, the final cell solution often needs to be transported from the production site to the treatment site, which may be several hours away. Shahin’s study demonstrated that the cell solution can be transported for up to 24 hours under controlled conditions while maintaining cell functionality and characteristics.

This finding allowed the research team to establish human serum albumin as the preferred carrier solution for keratinocytes in clinical treatments, ensuring their viability and functionality during transport. Additionally, it was validated as the final formulation solution for administration.

At NorthX Biologics, we offer tailored fill & finish solutions with integrated analytical support.

4.  Ensuring quality and safety in cell-based ATMP manufacturing

Ensuring the quality and safety of cell-based ATMPs is paramount. Shahin’s thesis emphasizes the importance of thorough characterization of keratinocytes and AD-MSCs, including the assessment of cell viability, differentiation potential, and functionality[1]. The use of cell and molecular characterization methods, including but not limited to immunophenotyping, gene and protein expression analyses are instrumental tools for monitoring and ensuring the quality of the produced cells [1].

Conclusion

The thesis by Hady Shahin offers valuable insights into manufacturing cell-based regenerative solutions for skin healing. The proposed xeno-free workflows and co-culture techniques present promising methods for producing GMP-compliant cell therapies. These advancements pave the way for effective treatments for difficult-to-heal wounds, ultimately improving patient outcomes.

Hady Shahin, PhD
Production Scientist
NorthX Biologics

Hady is a Production Scientist at NorthX Biologics, providing bioprocess and CMC support for GMP manufacturing of drug substances (DS), contributing to the production of advanced biologics for clinical trials. Biotechnologist by training and SME in scaling up cell therapy solutions, with over 15 years of experience in regenerative medicine and stem cell research. Hady holds a PhD in Cell and Molecular Biology and an MSc in Biomedical Science, with a strong background in ATMP development and GMP manufacturing.

LinkedIn

References
  1. Shahin, H. (2023). Keratinocytes and Adipose-derived mesenchymal stem cells: The heir and the spare to regenerative cellular therapies for difficult-to-heal skin wounds. Linköping University Medical Dissertation No. 1880.
    https://liu.diva-portal.org/smash/get/diva2:1810734/FULLTEXT01.pdf
  2. National Institutes of Health. (2021). Stem Cell Basics. Retrieved from https://stemcells.nih.gov/info/basics/stc-basics.

Driving therapeutic innovation with OMVs and EVs

Outer membrane vesicles (OMVs) and extracellular vesicles (EVs) are revolutionizing biopharmaceuticals, offering cutting-edge vaccines and therapeutic delivery solutions. These nanoscale vesicles hold immense potential to enhance immune responses and serve as immune-silent drug carriers, opening new frontiers in medicine.

In this article, Isa Lindgren, Ph.D., Head of Analytics, and Ola Tuvesson, Chief Technology Officer, explore:

● The expanding applications of OMVs and EVs in vaccines and therapeutics

● Advances in vesicle-based drug delivery compared to viral vectors

● Key challenges in scaling up OMV and EV manufacturing

● NorthX Biologics’ expertise in bringing these innovative therapies to market

Discover how NorthX Biologics is driving innovation and overcoming manufacturing and analytical challenges to accelerate the clinical potential of OMV and EV-based therapies.

Fill out the form to read more.

Read about our Privacy Policy

Ola Tuvesson
Chief Technology Officer
NorthX Biologics

As CTO, Ola is leading NorthX Biologic’s development and project organization, focusing on delivering technologies and strategies to ensure high-end services within bioprocessing and analytics. He has more than 20 years’ experience from the pharma and biotech industry, ranging from early development to commercial GMP manufacturing. Ola has worked in several fields, including ATMP products, vaccines, and other biologicals. He has the essential knowledge to take a product from early pre-clinical development into clinical trials and to the market.

Isa Lindgren, Ph.D.
Head of Analytics
NorthX Biologics

Isa Lindgren, Ph.D., is Head of Analytics at NorthX Biologics, leading the QC and Analytical Development teams across the Matfors and Stockholm sites. With a background of 15+ years in life sciences research and experience from preclinical work at Chiesi Pharma in biologics and ATMPs, Isa has extensive expertise in analytics. Six years in the US have equipped her with valuable international experience for global communication and high-level customer care. Known for her technological acumen, she ensures NorthX Biologics remains a front-runner in analytics to deliver biologics at the highest quality. 

Robotic gloveless isolator technology enhances regional production of advanced therapeutics

To meet growing demands of advanced therapy manufacturing, production architectures must shift towards agile, efficient manufacturing systems. By integrating robotic gloveless isolator (RGI) technology into bioproduction, manufacturers can meet the needs of plasmid DNA, viral vector, and mRNA-based medicines while maintaining compliance with stringent regulatory standards.

Read the full article by Peter Boman (NorthX Biologics), and  Zach Hartman (Cytiva), in Cell & Gene Therapy Insights here.

From lab to clinic: Large-scale recombinant protein production explained

Recombinant protein manufacturing has transformed biotechnology, enabling the production of therapeutic proteins, industrial enzymes, and bioengineered materials. The growing demand for recombinant proteins across regenerative medicine, disease diagnostics, and biopharmaceuticals has enhanced the importance of Contract Development and Manufacturing Organizations (CDMOs). These organizations bring essential expertise in process development, regulatory compliance, and large-scale production, ensuring the delivery of high-quality recombinant proteins. This review examines the critical role of CDMOs in optimizing recombinant protein manufacturing, exploring current challenges, innovations, and future advancements.

Bridging the gap between research and the patient

CDMOs serve as a crucial link between research institutions and commercial biomanufacturing by providing specialized services such as expression system optimization, purification strategies, and adherence to Good Manufacturing Practice (GMP) standards. CDMOs play a critical role in optimizing these expression platforms, ensuring high-yield, scalable, and regulatory-compliant production. These vital contributions in advancing recombinant protein production have a profound impact on the biopharmaceutical industry. As the demand for high-quality, scalable protein production escalates, CDMOs continue to be instrumental in providing dependable solutions.

Downstream process columns

Recombinant protein manufacturing

Recombinant protein expression is a cornerstone of scientific research and biopharmaceutical development, offering versatile solutions across various therapeutic applications. Bacterial and mammalian cell systems remain the most widely used platforms, each with distinct advantages.

Bacterial expression systems, particularly Escherichia coli, remain a cornerstone of recombinant protein production due to their rapid growth, ease of genetic manipulation, and cost-effectiveness. These systems offer high expression efficiency, making them ideal for large-scale manufacturing of simple proteins, enzymes, and non-glycosylated therapeutic proteins. However, bacterial hosts lack the intricate post-translational modification (PTM) machinery found in eukaryotic cells. This limitation can impact the correct folding, disulfide bond formation, and glycosylation of complex proteins, potentially affecting their functionality. Despite these challenges, bacterial systems continue to be a preferred choice for applications where process duration, scalability, and cost efficiency are paramount.

Mammalian cell systems, such as Chinese hamster ovary (CHO) and human embryonic kidney (HEK293) cells, are the gold standard for producing complex biopharmaceuticals. These platforms excel in generating proteins with native folding, proper post-translational modifications (PTMs), and human-like glycosylation—key factors in ensuring the efficacy and safety of monoclonal antibodies, recombinant vaccines, and other therapeutic proteins. With well-optimized expression technologies, mammalian systems offer a reliable and scalable solution for producing high-quality biologics, making them essential for advancing next-generation therapeutics.

A comprehensive approach to GMP-compliant large-scale recombinant protein production

Biopharmaceutical CDMO NorthX Biologics specializes in bacterial and mammalian cell culture processes for recombinant protein production. When embarking a new client project, a team of experts takes the gene sequence through gene cloning, cell bank generation, protein expression to final isolation and purification.

  1. Expression system selection
    Choosing the appropriate expression system is a critical factor in recombinant protein production. CDMOs assess various elements such as yield, scalability, and post-translational modifications to select the optimal system. Commonly used expression hosts include:
    • Bacterial systems (e.g., Escherichia coli): Offer high expression efficiency but are limited in terms of post-translational modifications
    • Mammalian cells (e.g., CHO, HEK293): Preferred for producing complex biopharmaceuticals due to their ability to perform human-like modifications
  1. Process development
    Co-funded by the Swedish Government, the Innovation Hub aims to advance infrastructure for advanced therapies and drive innovation in the biopharmaceutical sector. NorthX Biologics provides a clear distinction between non-GMP process development and GMP manufacturing, ensuring a seamless transition from early-stage development to full-scale, regulatory-compliant commercial production. An integrated approach across both upstream and downstream processes supports the production of high-quality recombinant proteins for advanced therapies.

    2.1 Upstream process development
    Efficient bacterial and mammalian cell culture processes are essential for maximizing recombinant protein production. Upstream process development focuses on optimizing media, fed-batch strategies, and bioreactor scale-up to improve protein yield and scalability. These processes are designed to ensure flexibility and efficiency in early-stage development.

    2.2 Downstream process development
    Downstream process development utilizes advanced chromatographic and filtration techniques to ensure high protein purity and yield. Chromatography resin screening, affinity chromatography, ion exchange, size exclusion chromatography and multimodal chromatography (MMC) are employed to refine and optimize protein purification for a variety of biopharmaceutical applications. This stage is critical in developing the optimal purification workflow before scaling up to GMP manufacturing.
  2. GMP Manufacturing
    3.1 Microbial protein expression
    NorthX Biologics specializes in microbial development and GMP-compliant protein expression, offering tailored solutions for the development and production of microbial proteins across a wide range of applications, including therapeutics, vaccines, and industrial uses. With advanced facilities and extensive expertise, flexible manufacturing options for both GMP and non-GMP applications are provided, ranging from fully closed single-use systems to cost-effective large-scale stainless-steel plants, supporting batch sizes from milligrams to kilograms. A long experience in producing extracellular, intracellular and periplasmic proteins ensures high-quality, soluble protein expression or inclusion body production, all while meeting the specific requirements of GMP manufacturing.

    3.2 Mammalian protein expression
    NorthX Biologics has deep expertise in the development and GMP-compliant manufacturing of proteins expressed in mammalian systems, with a strong focus on optimizing both upstream and downstream processes to ensure high yields and product quality. A comprehensive range of GMP-compliant mammalian protein production services is offered, including adherent and suspension-based manufacturing systems. Adherent manufacturing capabilities, using cell lines such as HEK293, are fully optimized for effective GMP translation, supporting scales from T-flasks to 500m² fixed-bed reactors for seamless scale-up. For suspension cell lines like CHO, HEK293, and PerC6, a variety of GMP-compliant options, including shake flasks, wave-mixing bioreactors, and single-use stirred tank bioreactors, are provided, all operated in batch, fed-batch, or perfusion modes.
  1. GMP compliance and quality control
    NorthX Biologics offers comprehensive biopharmaceutical testing services with customized solutions designed to meet the unique needs of each product, including analytics services. Batches produced meet stringent regulatory standards for clinical or commercial applications through quality control measures, including:
    • Process validation to ensure consistency in protein expression and purification
    • Analytical characterization using techniques such as ELISA and HPLC for functional validation
    • Adherence to GMP standards (Ph. Eur./USP), including the implementation of standard operating procedures (SOPs) for traceability
  1. Quality assurance and regulatory compliance
    NorthX Biologics ensures that all recombinant protein manufacturing adheres to regulatory requirements, ensuring compliance with FDA, EMA, and ICH guidelines. This can be achieved through:
    • Process validation
    • Analytical characterization and rigorous quality testing
    • Full GMP compliance to meet regulatory expectations
NorthX Biologics headquarters

Selective challenges in recombinant protein manufacturing

  • While significant progress has been made, CDMOs continue to face several challenges, including:
    • Process scalability: Managing the transition from lab-scale to commercial-scale production while maintaining consistency and quality
    • Protein aggregation: Overcoming stability issues that may affect protein formulation and storage
    • Regulatory variability: Adapting to the evolving landscape of global regulatory standards

Closing thoughts: The strategic value of CDMOs in protein production

Selecting the appropriate recombinant protein expression system is crucial for driving both scientific research and biopharmaceutical advancements. Whether utilizing bacterial or mammalian platforms, each offers unique benefits aligned with specific project needs. For CDMOs like NorthX Biologics, effectively applying their expertise in expression systems and advanced bioprocessing technologies is essential to deliver high-quality, scalable, and GMP-compliant solutions.

By collaborating with researchers and companies, NorthX Biologics plays a pivotal role in optimizing the development of therapeutic proteins, monoclonal antibodies, and other biologics, ensuring successful commercialization. Through their commitment to regulatory compliance and technical expertise, NorthX Biologics is a key partner in advancing next-generation therapies that improve patient outcomes globally.

Naresh Thatikonda, PhD
Scientist
NorthX Biologics

Naresh, a Scientist at NorthX Biologics and SME at our Matfors facility, holds a PhD in Biotechnology and a MBA in Industrial Management and Economics. He joined NorthX Biologics in 2019, where he has been providing process and CMC support for the GMP manufacturing of drug substance (DS) and drug product (DP), contributing to the production of biological drugs for clinical trials.

LinkedIn

References

Thatikonda, N. (2018). Functionalization of spider silk with affinity and bioactive domains via genetic engineering for in vitro disease diagnosis and tissue engineering (Doctoral dissertation, KTH Royal Institute of Technology).

U.S. Food and Drug Administration. (2023). A Quick-Start Guide to Biologics Manufacturing. Retrieved from https://www.fda.gov/media/170955/download 

BiologicsCorp. (2013). A Guide to the Production of Recombinant Proteins. Retrieved from https://biologicscorp.com/wp-content/uploads/2013/05/A-Guide-to-the-Production-of-Recombinant-Protein.pdf 

Personalized medicine and the CDMO: Adapting to a new era of healthcare

Neoantigen cancer vaccines represent a groundbreaking advancement in personalized medicine, offering tailored cancer treatments designed for each patient’s unique tumor profile. These therapies require rapid turnaround from tumor identification to clinical delivery — a critical factor when days can mean the difference between life and death for cancer patients. NorthX Biologics is uniquely positioned to meet these demands, providing agile, small-volume, multi-batch production, in-house rapid analytics, and robust supply chain solutions to bring lifesaving therapies to patients faster.

Authors: Ola Tuvesson, Chief Technology Officer, and Isa Lindgren, Ph.D., Head of Analytics, NorthX Biologics

Neoantigen cancer vaccines: a personalized approach gaining momentum

Personalized medicines are revolutionizing treatment paradigms by tailoring therapies to each patient’s unique genetic and biological characteristics. A particularly promising area is neoantigen-based cancer vaccines, which target antigens specific to an individual’s tumor microenvironment, offering a precise and highly individualized approach to oncology.

Although no neoantigen cancer vaccines have received regulatory approval yet, the field is advancing rapidly, with several candidates progressing to phase II and later-stage clinical trials. Both established pharmaceutical companies and agile startups are actively developing these therapies. Messenger RNA (mRNA) remains the most common modality due to its versatility and rapid production capabilities, though DNA-based vaccines are also gaining traction.

While the science behind neoantigen cancer vaccines is robust, critical challenges remain, particularly in the realm of analytics, process development, and chemistry, manufacturing, and control (CMC). For these therapies to advance through clinical trials and reach commercialization, comprehensive characterization and validated processes are essential. Developers must overcome hurdles such as the need for rapid analytics, platform processes, and stringent sterility testing under accelerated timelines. For a patient battling cancer, delays in treatment can mean the difference between life and death, underscoring the urgency of streamlined processes from tumor identification to clinical delivery.

Overcoming regulatory hurdles for neoantigen therapies

The evolution of personalized medicine, including neoantigen-based cancer vaccines, depends on innovative companies willing to pave the way and progressive contract development and manufacturing organizations (CDMOs) capable of supporting these groundbreaking therapies. However, regulatory uncertainty remains a significant hurdle. Current regulatory frameworks, designed for traditional therapies like small molecules and biologics, are not adapted to the needs for personalized medicines, where each product is tailored to an individual patient.

To address this challenge, a paradigm shift in regulatory evaluation is required. Reviewing every individualized therapy on a case-by-case basis is impractical. Instead, a platform-based approach — validating the overall manufacturing process rather than individual batches — is the most feasible path forward. Such a shift would allow regulators to focus on standardizing key processes while permitting minor, patient-specific variations in raw materials.

Critical to this transition is the development of robust, reliable platform analytical methods. These methods must be qualified using a bracketing principle, ensuring that key attributes remain consistent across therapies, regardless of genetic sequence variations. This platform approach can be supplemented with sequence-dependent analytical techniques to confirm the product’s identity, balancing regulatory rigor with the flexibility required for personalized treatments.

Scaling down for personalization

Personalized medicines, such as neoantigen cancer vaccines, require a fundamental shift in biomanufacturing approaches. For decades, the industry has focused on scaling up — boosting titers and increasing batch sizes to efficiently produce biologics for large patient populations. Personalized therapies, however, demand the exact opposite: scaling down to produce one batch per patient. This shift introduces new complexities, requiring innovative solutions to maintain productivity and efficiency and keep cost of goods (COGs) manageable.

Isa Lindgren, Head of Analytics

With each batch tailored to an individual patient, large-scale bioreactors and processes become impractical and cost-prohibitive. Instead, manufacturers must embrace smaller, highly efficient systems capable of running multiple batches with short product turnovers or even in parallel under GMP conditions. These systems must also integrate streamlined supply chains and pre-positioned raw materials to meet accelerated production timelines.

Orchestrating supply chains for individualized therapies

In personalized medicines like neoantigen cancer vaccines, organizational precision across supply chains and manufacturing operations is paramount. Unlike therapies produced in bulk for large patient populations, personalized treatments require multiple small batches to be manufactured simultaneously, each tailored to a specific patient. This introduces significant logistical challenges, demanding seamless coordination of material inflows, production processes, and product deliveries.

For neoantigen cancer vaccines, the process begins with the collection and analysis of a patient’s tumor sample to identify relevant neoantigens. This analysis triggers a narrow production window, during which the drug product must be manufactured and delivered. For patients with life-threatening cancers, every day matters. Personalized therapies must transition from concept to clinic with unprecedented speed, necessitating CDMOs that excel in agile, efficient production, and robust in-house analytics.

The patient’s journey demands a biomanufacturing process that prioritizes speed without compromising quality. Ensuring the availability of raw materials — pre-positioned and ready for use — is critical. Likewise, platform-based manufacturing processes that allow for rapid initiation and parallel execution are essential to meeting these time-sensitive demands.

From tumor analysis to delivery: breaking down analytical roadblocks

For neoantigen cancer vaccines, the journey begins with identifying the specific genetic mutations within each patient’s tumor — a highly individualized process that forms the foundation for these personalized therapies. This critical first step relies on next-generation sequencing (NGS) and machine learning algorithms to pinpoint relevant neoantigens. While this analysis typically falls under the responsibility of the therapy developer, the subsequent steps in the process require efficient manufacturing and rigorous testing to ensure the final drug product meets quality standards and can be delivered to the patient on time.

Once the relevant antigens have been defined, the focus shifts to ensuring rapid and reliable production and testing. At this critical stage, NorthX Biologics provides comprehensive support, offering streamlined CMC processes and rapid product release testing to minimize delays. NorthX Biologics’ integrated in-house analytics capabilities, including advanced sterility testing solutions, enable efficient product release, ensuring therapies move swiftly from manufacturing to the clinic.

Ola Tuvesson, Chief Technology Officer

In the case of highly personalized, short-lifespan therapies like neoantigen cancer vaccines, sterility testing can present a significant challenge. While rapid sterility tests have been developed, the process remains time-consuming. NorthX Biologics addresses this challenge by employing closed systems and stringent aseptic controls, reducing the risk of contamination and ensuring product quality. Additionally, the company collaborates with innovators in advanced testing solutions to remain at the forefront of analytical capabilities, minimizing delays that could impact patient outcomes.

A trusted partner for neoantigen cancer vaccine development

NorthX Biologics is uniquely positioned to support the development and manufacturing of personalized neoantigen-based cancer vaccines. The company’s fully integrated in-house capabilities — including specialized analytics for process development and product release — enable efficient, end-to-end support. Recognizing the importance of rapid turnaround, NorthX Biologics collaborates closely with experts in advanced testing solutions, such as accelerated sterility testing, to minimize delays and keep timelines on track.

With decades of experience manufacturing both technical and therapeutic proteins, NorthX Biologics combines its proven expertise with the scientific innovation of its Innovation Hub. This powerful combination has enabled the establishment of a highly agile manufacturing organization and a streamlined, adaptable supply chain. Effective quality control, quality assurance, and product release processes ensure NorthX Biologics can meet the rigorous demands of personalized medicine while maintaining the highest standards.

NorthX Biologics stands out through its Beyond CDMO approach, extending beyond traditional manufacturing services to act as an innovation partner, enabler, and strategic guide for its clients. By fostering strong collaborations with suppliers and customers, the company provides customized solutions that address the specific needs of each project. This forward-thinking philosophy reflects a commitment to advancing therapeutic development and delivering personalized medicines faster. Guided by the principle small enough to care and big enough to deliver,” NorthX Biologics delivers decisions quickly, adapts readily to change, and leverages its deep expertise to help drug developers bring life-changing therapies to patients in need. By combining agility, flexibility, and strategic insight, NorthX Biologics empowers its clients to navigate the complexities of this evolving landscape, ensuring that innovation reaches patients without delay.

Ola Tuvesson
Chief Technology Officer
NorthX Biologics

As CTO, Ola is leading NorthX Biologic’s development and project organization, focusing on delivering technologies and strategies to ensure high-end services within bioprocessing and analytics. He has more than 20 years’ experience from the pharma and biotech industry, ranging from early development to commercial GMP manufacturing. Ola has worked in several fields, including ATMP products, vaccines, and other biologicals. He has the essential knowledge to take a product from early pre-clinical development into clinical trials and to the market.

Isa Lindgren, Ph.D.
Head of Analytics
NorthX Biologics

Isa Lindgren, Ph.D., is Head of Analytics at NorthX Biologics, leading the QC and Analytical Development teams across the Matfors and Stockholm sites. With a background of 15+ years in life sciences research and experience from preclinical work at Chiesi Pharma in biologics and ATMPs, Isa has extensive expertise in analytics. Six years in the US have equipped her with valuable international experience for global communication and high-level customer care. Known for her technological acumen, she ensures NorthX Biologics remains a front-runner in analytics to deliver biologics at the highest quality. 

The expanding biologics CDMO market: Innovative modalities and the role of NorthX Biologics

The pharmaceutical industry is experiencing a rapid transformation as advanced biologics – viral vectors, recombinant proteins, plasmid DNA, and cell and gene therapies – take center stage. With over 14,800 active biologics innovation programs in development, including monoclonal antibodies, viral vaccines, and gene therapy vectors like AAVs and adenoviruses, the demand for specialized biologics contract development and manufacturing organizations (CDMOs) is soaring. Plasmid manufacturing remains integral to these innovations, underpinning gene therapies, DNA vaccines, and mRNA manufacturing.

Manufacturing these advanced therapies is a highly complex process that requires careful control over production platforms and scale-up strategies. For viral vectors, the process begins with a master cell bank (MCB), containing cells such as HEK293 or Vero that have been optimized for viral vector production. The viral seed stock, a small, well-characterized batch of virus, is used to infect the MCB during upstream processing. The viral particles are then expanded in either suspension bioreactors or adherent systems like the iCellis500 platform. This is followed by cell lysis, endonuclease treatment, and depth filtration to remove debris. Tangential flow filtration (TFF) further concentrates the viral product, while chromatography ensures the removal of impurities during purification. The process culminates in formulation and aseptic fill-finish, ensuring the product meets regulatory and safety requirements.

Recombinant protein services and plasmid manufacturing

For recombinant proteins, microbial systems such as E. coli or mammalian cell lines like CHO are commonly used. Microbial production often leverages transient expression, where plasmids containing the gene of interest are introduced into bacterial cells for rapid protein synthesis. In mammalian systems, cell banks provide stable production platforms where cells are expanded, transfected with the target gene, and induced to produce recombinant proteins. After fermentation or cell culture, purification involves ultrafiltration, diafiltration, and chromatography to isolate the desired protein, followed by formulation and fill-finish.

Man in production clothes working on filling machine


Plasmid DNA, essential for gene therapies and mRNA vaccines, is manufactured using microbial fermentation systems. A carefully selected host strain of E. coli is used to amplify plasmid DNA during fermentation. After cell harvest and lysis, the plasmid is separated from host DNA and proteins through filtration, chromatography, and buffer exchange processes. The purified plasmid DNA is then formulated and filled aseptically under GMP conditions. NorthX Biologics has excelled in this space, offering scalable plasmid DNA production, ensuring a seamless transition from research-grade material to clinical GMP batches.

Biologics CDMO services

NorthX Biologics has emerged as a leader in the advanced biologics space by integrating these complex manufacturing processes into streamlined, end-to-end solutions. Our capabilities span viral vectors, recombinant protein services, plasmid manufacturing, and cell therapy services. In cell therapy manufacturing, we leverage allogeneic or autologous cell banks, expanding cells in controlled GMP cleanrooms. Cells undergo activation, differentiation, and harvest, followed by aseptic filling and cryopreservation to ensure product viability.

These manufacturing capabilities are supported by NorthX Biologics’s expertise in process development, regulatory compliance, and analytical testing. By offering upstream process optimization, purification, and aseptic fill-finish under one roof, NorthX Biologics significantly reduces timelines while ensuring product quality.

Women i production clothes working in cell lab

The success of NorthX Biologics’s approach is demonstrated in real-world collaborations. During the OPENCORONA project, NorthX Biologics rapidly produced GMP-grade plasmid DNA for a SARS-CoV-2 vaccine, meeting strict quality and regulatory standards. In viral vector manufacturing, NorthX Biologics enabled HOOKIPA Pharma to scale a novel immunotherapy product to 200L using their transient expression processes and robust purification techniques. For Mendus, NorthX Biologics established GMP cell therapy manufacturing capabilities within just eight months, ensuring a smooth tech transfer and rapid scale-up for clinical production. Similarly, in collaboration with Abera Bioscience, NorthX Biologics supports the GMP manufacturing of outer membrane vesicles (OMVs), leveraging microbial processes to produce these naturally occurring particles for innovative vaccines.

Aseptic fill-finish and analytical expertise

To meet growing market demands, NorthX Biologics has invested in two European facilities equipped for microbial and mammalian production. These sites include BSL2 and BSL3 capabilities, supporting a range of biologics platforms. Our aseptic fill-finish services adhere to Annex 1 regulations, providing the highest level of assurance for clinical and commercial products. Additionally, NorthX Biologics’s in-house analytical expertise ensures rigorous quality control, from biologics process development and in-process monitoring to release testing and stability studies.

As advanced therapies continue to evolve, the role of biologics CDMOs in enabling efficient and scalable manufacturing becomes increasingly critical. NorthX Biologics stands out as a true partner in innovation, seamlessly integrating cell banking, seed stock preparation, transient expression systems, and robust purification and analytical technologies to support pharmaceutical innovators. By combining decades of GMP experience with cutting-edge manufacturing capabilities, NorthX Biologics is well-positioned to deliver life-saving therapies faster and more efficiently, helping patients worldwide benefit from the next generation of biologics.

Protein science and the future of biotherapeutics

The study of proteins is transforming our understanding of human biology and disease, enabling breakthroughs in diagnostics, drug development, and advanced therapies. In this article, Mathias Uhlén, Professor of Microbiology at KTH Royal Institute of Technology and member of NorthX Biologics’ Board of Directors, explores: 

  • The Human Protein Atlas: A cornerstone for drug discovery and development. 
  • AI’s role in accelerating protein structure prediction and molecular interactions. 
  • Next-generation technologies revolutionizing diagnostics and therapeutic monitoring. 
  • NorthX Biologics’ innovation hub driving scalable, efficient protein production. 

Join the journey into the future of protein science and learn how these advancements are shaping the next generation of precision medicine and therapeutic development. 

Fill out the form to read more.

Read about our Privacy Policy

Navigating the complexities of microbial and mammalian manufacturing

With a rich historical legacy and decades of expertise at its two sites, NorthX Biologics has long mastered the complexities of microbial and mammalian manufacturing. By offering capabilities and differentiating experience in both expression systems, NorthX Biologics provides tailored, cost-effective solutions that optimize processes and meet the diverse needs of recombinant protein production.

Authors: Peter Boman, Chief Operations Officer, and Helena Pettersson, Chief Production Officer, Head of USP, NorthX Biologics

E.coli

Distinct approaches in biomanufacturing: mammalian cell culture vs. microbial fermentation

In biopharmaceutical manufacturing, mammalian cell culture remains a dominant method, commonly utilizing Chinese hamster ovary (CHO) cells for producing protein-based drugs and human embryonic kidney (HEK) cells for viral vectors. However, microbial fermentation, which employs bacteria like Escherichia coli and Pseudomonas fluorescens, as well as yeasts like Saccharomyces cerevisiae and Pichia pastoris, is gaining traction, especially with the rise of new modalities, such as nucleic acids, single-domain antibodies, peptidbodies, and antibody fragments.

One of the main advantages of microbial fermentation is speed. Whereas mammalian cell culture processes can take six weeks or more from start to purified drug substance, fermentation processes are typically completed within a few days. Additionally, mammalian cells require complex and costly media formulations — often containing up to 100 ingredients — while bacteria and yeast need only simple glucose-based feeds. Microbial systems also tend to express target substances at higher concentrations than mammalian cells, further enhancing their appeal for specific applications.

However, bacteria and yeast have limitations. They are generally inefficient at producing larger recombinant proteins and monoclonal antibodies, making them better suited for smaller biologics, such as peptides, cytokines, and growth factors. Furthermore, microbial systems lack the capacity for extensive post-translational modifications (PTMs), such as glycosylation, which restricts their use in applications that require these modifications, including the prevalent monoclonal antibodies. Mammalian cell culture, therefore, is often the preferred option for these more complex requirements, as it provides the capabilities that microbial systems cannot.

The capability to offer both mammalian cell culture and microbial fermentation allows a comprehensive contract development and manufacturing organization (CDMO) like NorthX Biologics to better tailor production processes to the unique needs of each biomolecule. Advances in fermentation technology enable the efficient and cost-effective production of smaller and less complex biologics, while innovations in mammalian cell culture, such as enhanced titers and improved process control, support the manufacture of more complex biomolecules requiring intricate post-translational modifications. By leveraging expertise in both systems, forward-thinking CDMOs can provide optimized, high-quality solutions across a broad range of biologics, ultimately delivering greater value to their clients.

Matching biomolecules to manufacturing systems

Choosing between mammalian cell culture and microbial fermentation for biopharmaceutical production requires a balance of financial, technical, and quality considerations. The optimal manufacturing system for a drug candidate depends primarily on the unique characteristics of the biomolecule, including its size, complexity, and need for PTMs, as well as the intended use of the final product.

For some biomolecules, production in mammalian cells is necessary due to their capacity to perform complex modifications, while other substances are more efficiently produced through microbial fermentation. In certain cases, a highly sensitive protein may benefit from fermentation’s shorter production timeline, which can reduce the risk of degradation. Quality standards also vary across products, with some requiring exceptionally high purity that may be easier to achieve with one system over the other.

Ultimately, understanding these specific requirements early in the development process helps ensure that the chosen manufacturing path is both efficient and cost-effective.

COO Peter Boman

Advances in mammalian bioproduction: higher titers and enhanced control

Mammalian cell culture has seen remarkable advancements over the past two decades, most notably a significant increase in production titers. While titers of 0.1 grams per liter were typical when biologics first entered the market, today’s processes commonly achieve titers of 10 grams per liter or higher. This leap has been driven by a deeper understanding of cellular mechanisms and the adoption of automation and digitalization in biomanufacturing.

Process analytical technologies now enable real-time monitoring and control, providing insights that have propelled improvements in cell-line engineering and media formulation. These innovations allow biopharma companies to better optimize mammalian cell culture processes, enhancing both productivity and efficiency.

Enhancing fermentation efficiency: optimizing strains and streamlining processes

Recent advancements in bacterial and yeast expression systems have focused on increasing efficiency and reducing production costs. New microbial strains now allow for more effective expression across a broader range of proteins, expanding the applicability of fermentation. Additionally, significant improvements in downstream processing have been achieved, as this stage traditionally accounts for a large portion of the cost of goods in biologics manufacturing.

One of the primary goals in downstream optimization has been to minimize the number of costly chromatography steps. Techniques such as heat inactivation and precipitation are now used to simplify protein purification, offering a more cost-effective and efficient approach to removing undesired proteins. These developments make microbial fermentation a more competitive option for producing biologics.

Accelerating bioprocessing with AI-driven efficiency

Optimizing bioprocesses, whether through mammalian cell culture or microbial fermentation, involves extensive and costly screening of cell lines and process parameters. Today, digitalization — particularly through artificial intelligence (AI) and machine learning (ML) — is transforming this process by enabling biopharmaceutical scientists to work more efficiently and effectively.

With adequate data, AI and ML algorithms can streamline the design of complex studies, such as design-of-experiment (DoE) frameworks, helping researchers reduce preparation time and gain richer, more actionable insights. These digital tools allow for smarter process development, accelerating the pathway to optimized production for both traditional and emerging biomanufacturing systems.

Trusted expertise in mammalian expression at NorthX Biologics

Although formally established in 2021, NorthX Biologics draws on decades of biological manufacturing expertise, emerging from a combination of legacy entities. The company’s mammalian cell culture capabilities expanded significantly with the acquisition of a specialized development and manufacturing facility from Valneva in Stockholm, Sweden, bringing a team of experts in mammalian process development, GMP manufacturing, and state-of-the-art equipment.

This site has a rich history, including the production of Sweden’s first inactivated polio vaccine. Later, it contributed to global eradication efforts through collaborations with the World Health Organization and the Bill & Melinda Gates Foundation. In addition to its robust protein and antibody production capabilities, NorthX Biologics has demonstrated expertise in handling high-risk viruses. The Stockholm facility features high-containment, multipurpose biosafety infrastructure, enabling the safe production of drug substances for pandemic-related viruses, including the highly contagious SARS-CoV-2.

NorthX’s clients benefit from close collaboration with scientists who bring deep process knowledge from development through to commercial production. With extensive experience in process development and scale-up, the team at NorthX Biologics ensures that manufacturability is built into each project from the start, helping clients avoid potential challenges during GMP implementation.

Furthermore, NorthX Biologics is well equipped to support comparability studies for products transitioning from early to late-stage clinical trials, ensuring continuity and consistency. Transparency and open communication are foundational to NorthX’s approach, fostering trust both within the team and with clients. NorthX’s clients know they can rely on the company for timely, complete, and quality-driven delivery.

Helena Pettersson

Microbial fermentation expertise at NorthX Biologics

NorthX Biologics’ Matfors, Sweden site has been manufacturing proteins through microbial fermentation since the early 1990s. While the site has a long history in biologics production, it transitioned to a multiproduct contract manufacturing organization in 2005 and has since undergone continuous investment to expand its capabilities. These include fill/finish services, high-quality plasmid manufacturing, enhanced quality control capabilities, and large-scale protein purification with single-use fermentation suites.

The facility supports a wide range of bacterial fermentation using both anaerobic and aerobic strains and is equipped to handle risk group Level 1 and 2 organisms in Matfors, as well as up to risk group Level 3 organisms at the Stockholm biosafety facility. With extensive in-house analytical and quality assurance capabilities, including QA/QP services and analytical development expertise, NorthX Biologics ensures rigorous oversight across all production stages.

Different strains present unique challenges—some produce lower yields due to limited growth density, while others, like cholera strains, can complicate product purification. With decades of experience as a service provider, NorthX’s team excels at addressing these complexities. They are adept at identifying the ideal strain for each project and tailoring fermentation and lysis methods to achieve optimal results.

NorthX’s facility is designed to accommodate both small-volume production for early-phase studies and large-scale manufacturing for late-stage trials. In high-demand situations, the team is capable of operating on a 24/7 schedule to meet client needs. Supported by experienced project managers with deep expertise in multiproduct facility operations, NorthX Biologics ensures seamless execution across diverse biomanufacturing demands.

Who benefits from partnering with NorthX Biologics?

At NorthX Biologics, our commitment goes beyond being a trustworthy CDMO; we strive to be a true partner in innovation, sustainability, and excellence. Our Innovation Hub in Matfors showcases our dedication to advancing cutting-edge technologies that not only benefit our customers but ultimately improve outcomes for patients worldwide. With a problem-solving mindset and customer-first approach, we support clients at every stage of their journey — from grant writing to navigating the complexities of biologics manufacturing.

NorthX Biologics offers a unique value proposition as a multipurpose, one-stop-shop CDMO. We combine our dual expertise in mammalian and microbial manufacturing with extensive in-house capabilities in analytical development, quality assurance, and large-scale production. This comprehensive service portfolio allows us to streamline drug development and manufacturing, enabling faster, more efficient, and cost-effective solutions for pharmaceutical and biotech companies.

Sustainability is deeply embedded in our operations. We actively collaborate with dedicated sustainability personnel and adhere to guidelines from Swedish authorities to become an increasingly eco-friendly organization. Powered by green electricity and sustainable practices, we are working toward reducing our environmental footprint while maintaining the highest levels of quality and compliance.

Sweden’s reputation for excellence extends to our highly educated workforce, whose expertise ensures the delivery of superior results. Clients benefit from our commitment to delivering products on time, in full, and with exceptional quality. This dedication applies whether you are a small or medium-sized drug developer working on complex, niche, high-value therapeutic candidates or a large biopharma company seeking a reliable CDMO partner for scalable production.

Choosing NorthX Biologics means choosing an experienced, innovative, and sustainable partner that is ready to meet your needs at every stage, from early development to commercialization. With Swedish precision and a proven track record, we deliver high-value solutions that inspire confidence and enable success.

Peter Boman
Chief Operations Officer
NorthX Biologics

Peter is the COO at NorthX Biologics, and as such responsible for our manufacturing facilities in Matfors. He holds a master’s degree in engineering, biotechnology, and business administration. Peter has worked in research and development with complex sample analytics, analytical development, and cell culture process development. He joined NorthX in 2013 and has since continuously improved his talented team, resulting in the production of a large number of biological drugs for clinical trials.

LinkedIn

Helena Pettersson
Chief Production Officer, Head of USP
NorthX Biologics

Helena is CPO at NorthX Biologics as well as Head of Upstream at our Stockholm site. She holds a master’s degree in chemical engineering, biotechnology from Mälardalens University. With former roles as scientist in process development at Pharmacia, CMC development at SOBI, and leadership in process development at Crucell, Helena brings more than 25 years of experience. She manages a highly skilled and experienced team, making sure we are on top of the advanced therapeutics sector.

LinkedIn

Advancing the science of technical and therapeutic proteins

NorthX Biologics is at the forefront of technical and therapeutic protein development, playing a critical role in supporting the production of advanced therapies. With over three decades of expertise, NorthX provides scalable solutions from early-stage research to full-scale commercial production, driving innovation in the biopharmaceutical industry.

In our latest article, Eva-karin Gidlund, Ph.D., MBA, Chief Business Officer, explores:

  • NorthX’s expertise in both technical and therapeutic protein manufacturing
  • Our scalable production capabilities, from concept to commercialization
  • The company’s commitment to sustainability and eco-friendly operations in Sweden
  • Our cutting-edge Innovation Hub as a center for collaborative development

Ready to explore the future of protein production? Download the full article to learn how NorthX Biologics can help advance your biopharma projects.

Fill out the form to read more.

Read about our Privacy Policy