Blog – Actelion Pharmaceuticals Deutschland GmbH https://cdmo.actelionpharmaceuticals.de Your Global CDMO Tue, 05 Aug 2025 14:17:52 +0000 en-US hourly 1 https://wordpress.org/?v=6.8.2 https://cdmo.actelionpharmaceuticals.de/wp-content/uploads/2024/04/cropped-actelion-Pharma-Symbol-2024-32x32.png Blog – Actelion Pharmaceuticals Deutschland GmbH https://cdmo.actelionpharmaceuticals.de 32 32 Biotechnology: Definition, Benefits, and Applications https://cdmo.actelionpharmaceuticals.de/biotechnology-definition-benefits-and-applications/ https://cdmo.actelionpharmaceuticals.de/biotechnology-definition-benefits-and-applications/#respond Tue, 05 Aug 2025 14:17:51 +0000 https://cdmo.actelionpharmaceuticals.de/?p=21463

Table of Contents

Biotechnology, a cornerstone of modern science, merges biological knowledge with engineering and technology to solve essential challenges across healthcare, agriculture, industry, and the environment. For pharmaceutical companies, understanding biotechnology is critical, whether for drug development, manufacturing, or leveraging innovative therapies. This blog post explains biotechnology in detail, covering its definition, benefits, main applications, operation processes, future, and key challenges, with examples tailored to both beginners and seasoned experts.

What Is Biotechnology? A Clear Definition

Biotechnology is the scientific use of living organisms, biological systems, or their components to produce products and develop technologies that improve human life. At its core, biotechnology uses biological processes such as cell cultivation, fermentation, and genetic engineering, often harnessing microorganisms in these processes to create new medicines, food items, and industrial materials.

Modern biotechnology often employs techniques such as recombinant DNA technology, molecular biology, and advanced engineering to manipulate the genetic makeup of organisms like plants, bacteria, and animals for specific benefits.

Benefits of Biotechnology for Pharmaceutical Companies

The biotechnology industry provides a wide range of benefits, especially for health care and medicine:

  • New Drug Development: Cutting-edge biotechnology processes enable the discovery and production of complex biomedical drugs, vaccines, and targeted therapies (such as gene therapy) for treating human diseases, including those previously untreatable diseases. Therapeutic proteins have been developed using genetic engineering, offering new treatment options for various conditions.
  • Personalized Medicine: By analyzing patient data and genetic makeup, pharmaceutical companies can develop medical treatments tailored to individuals, increasing efficacy and safety. Many new therapies and drugs have been developed through advances in biotechnology, including those based on engineered proteins.
  • Efficient Production: Advanced biological processes allow the pharmaceutical industry to produce large quantities of drugs cost-effectively, sometimes using genetically modified organisms (GMOs) like bacteria or yeast.
  • FDA Approved Therapies: Biotechnology companies pioneer many FDA approved biopharmaceuticals, providing new options for chronic and rare human diseases.
  • Support for Global Health: Vaccines, created through biotechnology applications, help control infectious diseases across the world.

How Does Biotechnology Work? Main Processes & Techniques

The processes of biotechnology center on understanding and harnessing biological systems at the cellular and molecular biology level. Here is how biotechnology generally operates:

  1. Research and Discovery: Scientists and researchers use basic research to unravel biological sciences and identify useful genes or molecular markers.
  2. Genetic Engineering & Recombinant DNA Technology: Through genetic engineering, molecular biology and recombinant DNA technology, students and professionals modify the genetic makeup of an organism, inserting or deleting genes to bestow new characteristics.
  3. Cell Cultivation: Laboratories grow cells in controlled environments to optimize production of antibiotics, enzymes, or vaccines.
  4. Fermentation & Bioprocessing: Traditional techniques such as fermentation are now enhanced with modern biotechnology to efficiently produce food and medical products.
  5. Scale-up & Commercialization: Finally, biotechnology firms and biotechnology companies transition these technologies from laboratory-scale to industrial-scale production, often under strict standards set by federal agencies and agencies like the FDA.

Main Applications of Biotechnology

Biotechnology has revolutionized several sectors essential for pharmaceutical companies and beyond. Here is an overview of its leading fields:

1. Health Care and Medicine

  • Drug Production: Biotechnology has contributed to health care by enabling scalable production of monoclonal antibodies, insulin, vaccines, and gene therapy products.
  • Diagnostics: Biotechnology companies develop precise diagnostic tools using molecular biology to detect human diseases at early stages.
  • Biomedical Devices: Innovations in biomedical science lead to smarter therapeutic devices and drug delivery systems.

2. Agricultural Biotechnology

  • Genetically Modified Crops: With agricultural biotechnology, scientists engineer crops (e.g., soybeans planted and corn) that have been developed to meet agricultural needs, such as higher yield, pest resistance, and enhanced nutrition. Genetically modified foods produced from these crops are assessed for safety and regulated by relevant government departments to ensure they meet health and safety standards.
  • Global Food Security: Advanced biotechnology helps increase food production, supporting global food security by making plants more resilient to environmental challenges such as drought or pests.
  • Reduce Pesticides and Chemicals: GM crops often require fewer chemicals or pesticides.

3. Industrial Biotechnology

  • Biodegradable Plastics: Industrial biotechnology enables the production of eco-friendly, biodegradable plastics, reducing waste and greenhouse gas emissions.
  • Enzymes for Food Production: New technologies help the food industry produce safer, more nutritious food products, such as helping to preserve dairy products.

4. Environmental Biotechnology

  • Pollution Control: Uses engineered organisms to degrade hazardous chemicals or clean up oil spills.
  • Waste Management: Advances in biotechnology allow for bio-processing of waste, turning biological processes into tools for conservation.

The Future of Biotechnology

As science advances, the future of biotechnology holds tremendous promise:

  • Synthetic Biology: The design and construction of new biological systems and organisms for unique purposes, beyond what natural biology provides.
  • Advanced Gene Therapies: More innovative genetic engineering and gene therapy products will treat inherited human diseases and cancer.
  • Personalized Medicine: Data from genomics and proteomics will enhance the ability to tailor medical care to individuals.
  • Sustainable Production: Biotechnology research will support greener production of food, fuels, and chemicals, addressing environmental challenges.
  • New Technologies: Ongoing development in science and engineering will bring more important techniques and more effective pharmaceuticals.

Key Challenges Facing Biotechnology

Despite its many benefits, biotechnology faces significant challenges:

  • Regulatory Hurdles: Gaining FDA approved status for biopharmaceuticals is rigorous and time-consuming, involving strict reviews by federal agencies and the National Academy of Sciences.
  • Cost of Research: High costs of basic research, clinical trials, and scaling up production can be prohibitive, especially for smaller biotechnology firms.
  • Public Perception: Concerns about biotechnology products, especially food, require transparent communication and education among students, scientists, and consumers.

Biotechnology continues to play a key role in advancing health care, agriculture, and the environment. With powerful techniques like genetic engineering and recombinant DNA technology, biotechnology companies and pharmaceutical professionals can produce innovative therapies, support global food security, and tackle pressing environmental challenges. As science, engineering, and medical research evolve, the biotechnology industry will keep shaping the future of health and well-being worldwide, while ensuring careful consideration of ethical, regulatory, and societal concerns.

FAQs about Biotechnology

What is the simple definition of biotechnology?

Biotechnology is the use of living organisms, biological processes, or their components to develop useful products, especially in fields like medicine, food, and agriculture.

What are the 4 types of biotechnology?

The main types are:

Blue Biotechnology: Applies to marine and aquatic biological sciences.

Red Biotechnology: Related to health care and medical applications.

Green Biotechnology: Focuses on agricultural biotechnology and crops.

White Biotechnology: Concerns industrial biotechnology and manufacturing.

What is a biotechnology job?

Jobs in biotechnology include researchers, scientists, and engineers who use science and engineering to develop and oversee the production of biomedical, agricultural, or industrial products and processes.

What does biotechnology do?

Biotechnology enables the development, improvement, and production of new medical, agricultural, and industrial products, using biological processes to tackle issues in health, food production, and the environment.

]]>
https://cdmo.actelionpharmaceuticals.de/biotechnology-definition-benefits-and-applications/feed/ 0
Excellence in Non-Sterile Liquids: Actelion Leipzig’s Journey https://cdmo.actelionpharmaceuticals.de/excellence-in-non-sterile-liquids-actelion-leipzig-journey/ https://cdmo.actelionpharmaceuticals.de/excellence-in-non-sterile-liquids-actelion-leipzig-journey/#respond Wed, 23 Jul 2025 08:44:06 +0000 https://cdmo.actelionpharmaceuticals.de/?p=21067

Table of Contents

For pharmaceutical companies with promising non-sterile liquid formulations, the path from the laboratory bench to commercial production can be both exhilarating and daunting. Whether you are a startup seeking your first commercial launch or an established pharma company expanding your portfolio, your choice of CDMO (Contract Development and Manufacturing Organization) is crucial. After all, you are not just looking for a manufacturer—you need a trusted partner committed to quality, flexibility, and your vision.

At Actelion Pharmaceuticals Deutschland GmbH Leipzig, we understand your unique challenges and ambitions. With a legacy of technical excellence, regulatory mastery, and a relentless focus on partnership, we deliver non-sterile liquids for the world’s most demanding markets, every single day.

What Are Non-sterile Liquids and Why Are They Important?

Non-sterile liquids encompass a diverse range of pharmaceutical dosage forms, solutions, drops, suspensions, and emulsions, that do not require sterile processing. These forms are administered orally or topically and are essential for many everyday therapies: think cough syrups, nasal drops, and oral suspensions for both adults and children. Oral suspensions are part of a broader group known as oral formulations, which include various types designed for ease of use across patient populations. Unlike injectables or certain ophthalmic products, these do not need to be entirely free from micro-organisms, but they must still meet strict safety and quality standards.

Moving a non-sterile liquid from development to production demands careful balance: the preparation of the formulation must maintain efficacy and stability, choose the right containers, ensure compliance, and ultimately safeguard patient trust. Non-sterile liquids offer advantages such as dosing accuracy, ease of use, and suitability for pediatric and geriatric populations, which help improve patient compliance and simplify administration.

Your Manufacturing Pathways: What Are the Options?

As you weigh your manufacturing choices, you will find three broad paths. Choosing the right manufacturing path can be beneficial for addressing formulation challenges and ensuring effective drug development.

1. Local Specialized CDMOs

Smaller, regional partners often offer personal service and expertise in niche batches, and can manufacture small-scale non-sterile liquid products tailored to client needs. Flexibility can be high, but you may outgrow their capacity as your product scales.

2. Large Multinational CDMOs

These bring robust infrastructure and deep regulatory experience, and typically provide integrated testing services to ensure product safety and compliance. However, you might find yourself lost in the crowd, and personalized support or custom solutions can be harder to secure.

3. In-house Manufacturing

While keeping everything internal offers maximum control, companies can produce non-sterile liquid products in-house, provided they invest in the necessary resources. However, the investment in equipment, validation, staff, and inspection readiness is daunting, particularly for novel or high-volume projects.

What Sets Actelion Leipzig Apart?

Actelion Leipzig combines the best of both worlds: the agility and expertise of a local partner, backed by the resources and rigor of a global CDMO. Our integrated supply chain management ensures seamless drug development, manufacturing, packaging, and distribution, supporting your project from start to finish. We make your success our mission, not just a transaction but a true collaboration.

Why Choose Actelion Leipzig? A Closer Look Inside

1. Comprehensive Dosage & Formulation Expertise

We handle a full spectrum of non-sterile liquid products. Whether you need solutions, drops, suspensions, or emulsions, our team can formulate and scale your project smoothly. The formulation process involves dissolving or suspending active pharmaceutical ingredients (APIs) and drug substances to ensure product stability and efficacy. With batch sizes up to 4,000 liters, you’ll never have to second-guess your CDMO’s capacity as your product grows from pilot to full market release.

Actelion Leipzig also has experience with semi solid dosage forms such as creams, gels, and lotions, as well as powders, to meet diverse client needs.

2. Flexible Packaging

Patients and healthcare providers rely on packaging that is safe, convenient, and suited to their needs. We fill both plastic and glass bottles, ranging from 20ml up to 700ml, offering both small-scale and bulk presentation for various markets. These products are carefully packaged to maintain stability and quality. Packaging components play an important role in preserving product integrity throughout the shelf life.

3. Leading Edge Technologies

Your product is in capable hands: our three specialized production lines, Dovema, Groninger, and Würschum, offer precision filling, efficient line speeds, and scalable output. Advanced automation means less risk of errors and consistent quality, batch after batch.

4. Relentless Commitment to Quality and Safety

At Actelion Leipzig, quality is never “good enough”: it is everything. We proudly operate under the most rigorous international standards:

  • EU GMP-certified for both human and animal healthcare products.
  • Strict adherence to German Arzneimittelgesetz (AMG) and serialization in line with EU FMD (to combat counterfeiting).
  • Certification by the local Veterinär- und Lebensmittelaufsichtsamt assures safety and compliance for cosmetics and food supplements too.
  • Regular monitoring and testing for bacteria to ensure product safety, efficacy, and regulatory compliance.

5. Thorough Quality Control and Analytical Capabilities

Every batch is subject to:

  • Comprehensive routine analytics, from raw materials to finished product.
  • Microbiological testing, ensuring bacterial counts stay within defined limits (TAMC & TYMC).
  • Stability & dissolution testing, residual solvents analysis, and method validation (using HPLC, GC, UV-VIS, titration, and more).
  • Full documentation for regulatory audits and transparent reporting for your team.

Non-sterile liquid manufacturing also supports individual dosing, which enhances versatility and patient compliance, especially for pediatric and geriatric populations.

6. Regulatory Know-How at Every Step

You do not have to be an expert in every regulation, we are. Our processes and permissions are validated by German and EU health authorities (including Landesdirektion Sachsen). We guide you through documentation, validation, and market submission, with a focus on prescription labeling and compliance to ensure accurate dispensing and proper product identification, so you can focus on your product and patients, not paperwork.

image of the dosage forms for non sterile liquids that we mnanufacture at Actelion Leipzig

Microbial Contamination Prevention: Safeguarding Product Integrity

Preventing microbial contamination is a cornerstone of non-sterile liquid manufacturing. Even though non-sterile drugs are not required to be completely free of microorganisms, strict adherence to regulatory guidelines ensures that any microbial presence remains well within safe limits. At every stage of the manufacturing process, from the selection of raw materials to the final packaging, robust controls are in place to minimize the risk of contamination.

Key strategies include sourcing high-quality raw materials, maintaining a clean and controlled production environment, and implementing rigorous cleaning and sanitation protocols for all equipment. Regular microbiological testing of both the environment and the product itself helps detect and address any potential issues before they impact product quality. Additionally, staff training and strict process documentation ensure that every batch of non-sterile oral liquids, solutions, and suspensions meets the highest standards for safety and efficacy.

By prioritizing contamination prevention, pharmaceutical manufacturers protect both their products and the patients who rely on them, reinforcing trust in the quality of non-sterile preparations.

Facility and Equipment: Where Innovation Meets Precision

The quality of non-sterile liquid pharmaceuticals depends not only on the formulation but also on the environment in which they are produced. At Actelion Leipzig, our facility is designed to support the highest standards of pharmaceutical manufacturing. Every aspect of our production environment, from advanced air filtration systems to precise temperature and humidity controls, is engineered to minimize contamination risks and ensure consistent product quality.

Our advanced equipment, including specialized filling lines and automated packaging systems, allows us to handle a wide variety of dosage forms and batch sizes with precision and efficiency. This flexibility enables us to accommodate everything from small-scale development batches to large-scale commercial manufacturing, all while maintaining strict adherence to regulatory requirements. By investing in the latest technologies and maintaining rigorous maintenance schedules, we ensure that every non-sterile liquid, oral solution, and suspension is manufactured to exacting standards.

Innovation and precision are at the heart of our manufacturing services, providing pharmaceutical companies with the confidence that their products are produced in an environment where quality is never compromised.

What Is It Like to Work with Us?

Pharmaceutical manufacturing is dynamic and sometimes unpredictable. At Actelion Leipzig, we work as an extension of your team, answering questions quickly, offering proactive solutions, and always seeking ways to help you succeed. Whether you’re launching your first product or scaling up an established brand, you’ll experience transparency, reliability, and real partnership.

Choosing the right partner for non-sterile liquid manufacturing is a decision that shapes the future of your pharmaceutical product. At Actelion Leipzig, we combine deep expertise, cutting-edge technology, and a steadfast commitment to quality to deliver manufacturing services that exceed industry standards. Our comprehensive approach, from contamination prevention to advanced facility management, ensures that every oral liquid, solution, and suspension is produced with the utmost care.

Whether you are developing a new medication or scaling up for commercial manufacturing, our team stands ready to support your vision with flexibility, transparency, and unwavering dedication. Trust Actelion Leipzig to be your partner in non-sterile liquid excellence, where your product’s quality and your patients’ safety are always our top priorities.

FAQs about Non-sterile Liquids

What are examples of non-sterile?

Non-sterile pharmaceutical products include oral syrups, cough mixtures, nasal drops, topical solutions, and some ophthalmic and veterinary liquids that do not require complete sterility. Many over-the-counter medications are also formulated as non-sterile liquids for consumer use without a prescription.

What are sterile liquids?

Sterile liquids are formulated to be completely free from any living microorganisms. These include injectable medications, certain ophthalmic solutions for direct eye injection, infusions, and intravenous fluids.

What does non-sterile mean?

Non-sterile means the product is not processed to eliminate all microbial life, though it must still be within tightly controlled microbiological limits. These products remain safe and effective for their intended use.

Which can be a non-sterile product?

Products such as oral suspensions, cough syrups, some eye or nasal drops (not injected), dermatological solutions, and certain animal health formulations are all examples of non-sterile products.

What is the difference between sterile and non-sterile?

Sterile products undergo processes designed to destroy all forms of microbial life and are used when there is risk of infection (such as injections). Non-sterile products do not require this level of control but must be manufactured under hygiene-controlled conditions with defined microbial limits.

What are examples of sterile products?

Examples of sterile pharmaceutical products include vaccines, injectable medications, infusion solutions, intraocular eye treatments, and sterile ointments for wounds.

]]>
https://cdmo.actelionpharmaceuticals.de/excellence-in-non-sterile-liquids-actelion-leipzig-journey/feed/ 0
Successful Out Licensing in Pharma: Best Practices https://cdmo.actelionpharmaceuticals.de/successful-out-licensing-in-pharma-best-practices/ https://cdmo.actelionpharmaceuticals.de/successful-out-licensing-in-pharma-best-practices/#respond Tue, 15 Jul 2025 10:38:11 +0000 https://cdmo.actelionpharmaceuticals.de/?p=20561

Table of Contents

The pharmaceutical industry thrives on innovation, collaboration, and the strategic exchange of assets and technologies. As companies seek to maximize their commercial potential, minimize development risks, and expand their market reach, out licensing has become an essential component of modern business strategy. Whether you are a small or large pharmaceutical company, understanding out licensing is crucial to making informed decisions and driving company success.

In this comprehensive guide, we will explore the meaning of out licensing, its key aspects, how it differs from in licensing, and practical steps for navigating the out licensing process. We will also highlight how Actelion Pharmaceuticals Deutschland GmbH’s Athens facility serves as an effective out licensing partner for companies seeking new pharma licensing opportunities.

What is Out Licensing? A Clear Definition

Out licensing is the process by which a company (the licensor) grants certain rights, often related to a drug, technology, or intellectual property, to another company (the licensee) to further develop, manufacture, market, or sell. Instead of commercializing or developing an asset themselves, licensors leverage the resources, expertise, and market reach of their partners. Companies often out license their assets to expand market access and form strategic partnerships, allowing them to enter new geographic regions or benefit from external expertise.

The out licensing model is prevalent among pharmaceutical companies, biotech firms, and technology providers looking to generate revenue, mitigate risks, and enable their assets to realize full commercial opportunity. Licenses are legal permissions granted to external partners, allowing them to develop, manufacture, or commercialize drugs or technologies under agreed terms.

Key Aspects of Out Licensing

Let’s break down the key aspects and benefits that make out licensing a strategic option for companies engaged in the pharmaceutical industry:

1. Revenue Generation

Out licensing enables a company to generate revenue from unused or underdeveloped assets, such as drug candidates, platform technologies, or proprietary processes. Payments can take various forms, including upfront payments, milestone payments, and royalties based on product portfolios performance.

2. Risk Mitigation and Sharing

One of the key aspects of out licensing is risk sharing. As development costs, regulatory approval, and market entry can be resource-intensive and uncertain, licensors can transfer much of the financial risk and development risks to the licensee. This arrangement supports mitigating risks while still capturing potential upside through deal terms like royalties and milestones.

3. Market Expansion

Out licensing opens doors for market expansion, allowing pharmaceutical companies to expand into new markets by leveraging the established market reach of the licensee. This is particularly valuable for entering diverse geographical regions or new therapeutic areas.

4. Strategic Partnerships and Focus

Forming pharma partnership deals through out licensing agreements creates strategic synergies and enables both parties to focus on their core competencies. For example, a biotech firm may specialize in early-stage development, while a larger pharma company can handle late stage trials, regulatory approval, and commercialization.

5. Monetizing Intellectual Property

For companies with valuable but unexploited intellectual property, out licensing is a practical way to unlock its commercial potential and boost company value using multiple valuation methods such as risk adjusted net present value.

Understanding the Difference: Out Licensing vs. In Licensing

While out licensing is about granting rights, in licensing is about acquiring them. Here is a quick summary:

  • Out licensing: The company (licensor) grants rights to another party to exploit their technologies or products.
  • In licensing: The company (licensee) obtains rights from another party to use their intellectual property or assets.

Both strategies help pharmaceutical companies achieve their strategic goals, such as accessing innovative technologies (via in licensing) or extracting value from their existing pipeline (through out licensing).

Step-by-Step: The Out Licensing Process

Successfully navigating the out licensing process involves several critical stages. Here’s a practical breakdown:

1. Asset Identification and Valuation

  • Companies must identify promising assets, such as a biopharmaceutical asset or proprietary technology in house, for out licensing.
  • Accurate asset valuation using multiple valuation methods like risk adjusted net present value is essential to understand the worth of the offering.

2. Prospecting and Establishing Contacts

  • Identifying potential partners and prospective partners is a critical step. These could include regional companies, large pharmaceutical firms, or innovative startups.
  • Establishing contacts and leveraging networks or platforms is crucial for presenting your asset to suitable potential licensees.

3. Data Room Preparation and Due Diligence

  • Preparing a virtual data room or data room, a secure repository for confidential documents, enables prospective partners to perform due diligence on your asset, development, clinical data, and intellectual property status.

4. Partnering Process and Deal Negotiation

  • The partnering process involves making introductions, technical meetings, and presenting the asset’s commercial opportunity.
  • Negotiating deal terms, such as upfront payments, milestone payments, royalties, and responsibilities for regulatory approval, is central to achieving a mutually beneficial licensing deal.

5. Finalizing Licensing Agreements

  • Once terms are settled, both parties sign binding licensing agreements that define rights, obligations, product portfolios, and financial arrangements.

6. Ongoing Business Development and Relationship Management

  • Business development teams from both sides continue to collaborate, monitor progress, and adjust to challenges as development and commercialization proceed.
image showing the 6 steps of the out licensing process

Benefits of Out Licensing for Pharmaceutical Companies

Pharmaceutical companies, irrespective of their scale, can benefit in several ways by adopting an out licensing strategy:

1. Generate Revenue from Underutilized Assets

Vacant technologies and assets can be transformed into reliable income streams, especially when internal development or marketing is not feasible.

2. Reduce Development and Financial Risk

By collaborating with a licensing partner, companies can share or offload the burden of high development costs, regulatory approval hurdles, and other development risks.

3. Accelerate Access to New Markets and Patients

Through market expansion with established external partners, assets are more likely to enter new markets quickly, helping expand company impact and support global health.

4. Focus on Core Strengths and Complementary Capabilities

Companies can focus on their strengths, such as discovery or technology development, while external partners with complementary capabilities handle later development and commercialization.

5. Foster Industry Partnerships and Strategic Synergies

Entering out licensing agreements builds lasting relationships, encourages pharma partnership deals, and drives collective progress within the pharmaceutical industry.

How Actelion Pharmaceuticals Deutschland GmbH Can Help You Out Licensing

Actelion Pharmaceuticals Deutschland GmbH’s facility in Athens is dedicated to helping pharmaceutical companies unlock the benefits of out licensing. Here is how our out licensing model empowers our partners:

  • Access to Our Generic and Value Added Medicines Product Portfolios: We offer an extensive and growing selection of finished dosage forms ready for global and regional expansion.
  • Accelerate Entry into New Markets: Our robust network, regulatory know-how, and established presence help partners effectively expand their market reach.
  • Mutually Beneficial Out Licensing Agreements: We utilize a collaborative approach to optimize financial returns
  • Dedicated Support from Out Licensing Experts: Our seasoned business development team assists with every aspect of the out licensing process, from data room preparation and due diligence to deal negotiation and ongoing support.
  • Shared Mission for Global Health: By partnering with Actelion Pharmaceuticals Deutschland GmbH, you join us in delivering vital healthcare solutions to more markets, faster.

Out licensing is more than a financial transaction; it is a strategic approach allowing pharmaceutical companies to expand their market presence, focus on core strengths, and build enduring industry partnerships. By leveraging the out licensing process, firms can unlock hidden value, drive innovation, and collaborate for mutual benefit.

Whether you are a startup with groundbreaking technologies or a global leader managing a vast product portfolio, informed decisions about out licensing can accelerate your journey toward success in the ever-evolving pharmaceutical industry.

FAQs About Out Licensing

What is the meaning of out-licensing?

Out licensing refers to the process where a company grants permission to another party (licensee) to use, develop, produce, or commercialize its technologies, products, or intellectual property. This usually involves formal licensing agreements and is a common practice in the pharmaceutical industry to generate revenue and expand market presence.

What is an example of out-licensing?

An example would be a company with a new drug candidate out licensing it to a larger pharmaceutical company that has the resources to conduct late stage trials, navigate regulatory approval, and launch the drug in new markets. In this arrangement, the licensee typically pays upfront payments, milestone payments, and royalties to the original developer.

What is the out-licensing strategy?

The out-licensing strategy is a strategic approach by which a company leverages its own assets or technologies by partnering with licensees who can best maximize their potential. This strategy allows companies to focus on their strengths, mitigate risks, enter new markets efficiently, and foster strong partnerships.

What is the difference between Inlicense and Outlicense?

  • In licensing: A company acquires rights from another party to use their intellectual property or products, thus expanding its product portfolios.
  • Out licensing: A company grants another party the rights to use or commercialize its own assets or technologies.

Both forms of licensing are used strategically to meet business development goals, access complementary capabilities, and optimize commercial potential.

]]>
https://cdmo.actelionpharmaceuticals.de/successful-out-licensing-in-pharma-best-practices/feed/ 0
Autologous vs Allogeneic: Tailoring Biologic Manufacturing https://cdmo.actelionpharmaceuticals.de/autologous-vs-allogeneic-tailoring-biologic-manufacturing/ https://cdmo.actelionpharmaceuticals.de/autologous-vs-allogeneic-tailoring-biologic-manufacturing/#respond Tue, 08 Jul 2025 11:15:38 +0000 https://cdmo.actelionpharmaceuticals.de/?p=19802

Table of Contents

In advanced medicine, the distinction between autologous and allogeneic sources is foundational—not just in clinical care, but throughout drug development and pharmaceutical manufacturing. While most commonly referenced in cell and gene therapy, these concepts impact a broad array of biologic products, blood-based medicines, and tissue-derived therapies. For contract development and manufacturing organizations (CDMOs) such as Actelion Pharmaceuticals Deutschland GmbH, understanding these terms is critical to supporting client innovations from bench to patient.

Decoding the Basics: What Are Autologous and Allogeneic Approaches?

Simply put, autologous means “from yourself—back to yourself.” When a treatment uses cells, tissues, or biological materials collected from a patient and then gives them back after processing, that process is called autologous. This option offers a high level of personalization and is typically less likely to trigger the body’s defense systems.

On the other hand, allogeneic means “from a donor to a recipient.” Here, materials come from another person (often carefully matched and screened). This approach is fundamental for creating standardized, “off-the-shelf” therapies that can serve many patients with a single batch.

Both meet unique and rapidly growing needs in pharmaceutical development—but they also come with their own set of challenges.

image showing the differences between autologous and allogeneic cell therapies

Common Therapeutic Uses of Autologous and Allogeneic

Biologics & Plasma Therapies

  • Autologous Examples: Think of niche treatments like platelet-rich plasma (PRP) therapy, where a patient’s own blood is processed and returned for wound healing or joint regeneration.
  • Allogeneic Examples: Most blood plasma products on the market—like immunoglobulins or clotting factors—are pooled from several healthy donors, purified, and delivered to patients in need worldwide.

Tissue Engineering and Implants

  • Autologous Grafts: Here, a patient may have a piece of bone, skin, or tendon removed, treated, and re-implanted in another part of their body. This approach significantly reduces the risk of rejection or complications.
  • Allogeneic Grafts: Alternatively, donor tissue (skin, bone, valves) from tissue banks is standardized and used in reconstructive surgeries, trauma care, or transplants for patients lacking healthy autologous material.

Next-Generation Vaccines and Personalized Medicine

  • Autologous Vaccines: The future holds vaccines made from a patient’s own tumor antigens, potentially leading to hyper-targeted cancer therapies.
  • Allogeneic Vaccines: Everyday flu shots or COVID-19 vaccines are allogeneic—engineered for the masses, with broad spectrum effectiveness.

Advantages and Disadvantages: Autologous vs Allogeneic

Autologous Approaches

Advantages:

  • Unmatched compatibility: Since it’s your own material, risk of immune rejection or graft-versus-host disease is minimal.
  • No long wait times: No need to search for a matching donor—everything starts with you.

Disadvantages:

  • Personalized means complex: Every treatment batch is unique to each patient, requiring meticulous tracking and customization.
  • Time & scalability constraints: This process can be slower and may not be ideal for large urgent patient populations.

Allogeneic Approaches

Advantages:

  • Wider access: One donor’s contribution can help dozens (or hundreds) of recipients.
  • Faster response: Standardized batches mean therapies can be made in advance and ready as soon as they’re needed.

Disadvantages:

  • Immune risks present: Even with careful matching, there’s still a potential for immune response or rejection.
  • Safety and screening: Rigorous donor screening and regulatory controls are always essential.

A Partner for Flexible and High-Quality Manufacturing

Whether a pharmaceutical program relies on autologous or allogeneic principles, the path from development to delivery requires adaptable, high-quality manufacturing. For projects demanding agility—such as small-batch, patient-specific runs—or those requiring the highest compliance for broader-scale, standardized production, robust expertise and advanced infrastructure are essential.

With over 20 years of experience, Actelion Pharmaceuticals Deutschland GmbH’s Swiss Jura facility offers comprehensive support tailored to both clinical and commercial manufacturing. Specializing in the aseptic production of lyophilized and liquid sterile (bio)pharmaceutical products through fill and finish, the facility is ideally suited for small clinical and commercial batches. Certified by both GMP and FDA, Jura upholds the highest quality and safety standards.

Clients benefit from end-to-end solutions:

  • Scale-up and Technology Transfer: Seamless transition from development to production, backed by strong technical expertise.
  • Flexible Batch Production: No minimum batch sizes—ensuring the ultimate flexibility for unique, complex, or limited-quantity projects.
  • Aseptic Process Simulation (APS): Fully compliant APS services reinforce product sterility and efficacy.
  • GMP & FDA Regulatory Support: Comprehensive compliance guidance, making even the most complex clinical projects manageable.
  • API Management and Specialized Formulations: Skilled handling of sensitive or valuable materials, including peptides, biologicals, controlled substances, and ophthalmic products.

From pilot and scale-up batches for preclinical and early-phase clinical studies to full clinical or commercial manufacturing, release analytics, packaging, stability studies, and GMP storage, Actelion Jura delivers Swiss precision and reliability every step of the way.

By integrating technology, technical know-how, and proactive project management, the facility empowers clients to confidently advance their products—whether highly-personalized (autologous) or broadly-distributed (allogeneic)—from early development through to market launch.

In conclusion, the division between autologous and allogeneic is about much more than biology—it shapes every stage of the pharmaceutical journey, from research and regulatory planning to manufacturing and logistics. As the world moves toward more personalized and precise therapies, making an informed choice is essential for patients and developers alike.

At Actelion Pharmaceuticals Deutschland GmbH, we help our partners navigate these complexities, ensuring every product—whether made for one or many—meets the highest standards of safety, efficacy, and quality.

FAQs: Autologous vs Allogeneic

What’s the difference between autologous and allogeneic?

Autologous utilizes the patient’s own cells, minimizing rejection risk; allogeneic uses genetically matched donor cells, requiring tissue compatibility and often delivering immune-mediated therapeutic benefits.

What is the difference between autologous and allogeneic transfusions?

Autologous transfusion involves a patient’s own blood collected ahead of time. Allogeneic transfusion uses blood from another person and is subject to immune matching protocols.

What is an example of allogeneic?

A bone marrow transplant from an unrelated donor for treatment of relapsed leukemia.

What is the purpose of autologous?

To enable safe transplantation and cell therapies without the risk of immune-mediated rejection or graft versus host disease.

Is CAR-T allogeneic or autologous?

Most current CAR-T cell therapies are autologous, using the patient’s own immune cells. Next-generation products are exploring allogeneic CAR-T using healthy donor sources.

What is meant by allogeneic?

A biological product or tissue derived from a genetically different individual of the same species.

What is an autologous transplant?

A therapeutic procedure in which a patient receives an infusion of their previously harvested and stored bone marrow, blood stem cells, or blood after high-dose therapy.

]]>
https://cdmo.actelionpharmaceuticals.de/autologous-vs-allogeneic-tailoring-biologic-manufacturing/feed/ 0
Connecting Development Expertise with Global Manufacturing https://cdmo.actelionpharmaceuticals.de/connecting-development-expertise-with-global-manufacturing/ https://cdmo.actelionpharmaceuticals.de/connecting-development-expertise-with-global-manufacturing/#respond Wed, 02 Jul 2025 14:17:31 +0000 https://cdmo.actelionpharmaceuticals.de/?p=19391 In today’s competitive CDMO landscape, the ability to offer integrated, flexible, and high-quality services across development and manufacturing is more important than ever. At actelion, our development site in Athens plays a central role in enabling such synergy, acting as a strategic hub that connects innovation, adaptability, and global execution. 

Our expertise is not limited to troubleshooting or process optimization; it spans the entire lifecycle of a medicine. Actelion Athens is a pharmaceutical development site. Here, we go beyond generics. We design and develop value-added medicines tailored to our customers’ strategies and market demands, from concept to commercial. This capability positions us as a true partner, not just a service provider, in product development. 

Our Integrated Network  

One of our strongest assets is the integration between our dedicated development hub in Athens and our network of manufacturing facilities across Europe and Japan. This alignment allows us to transfer projects seamlessly while maintaining full control over quality, timelines, and regulatory compliance. 

Having Athens as a base for development and then transferring it to other sites in Europe is a great competitive advantage. We are able to use the expertise from development directly during the most critical phase: from tech transfer to commercial scale. 

This end-to-end coordination enhances tech transfer, scalability, and minimizes risks, ensuring our partners can move from lab to market with confidence and speed. 

Flexibility is essential in the CDMO space. Often, customers come to us with an existing process that doesn’t fully align with available manufacturing setups. Thanks to early-stage alignment between Athens and our commercial sites, we can anticipate and solve compatibility challenges. 

Supporting Multi-Market and Complex Projects 

Our network isn’t just European. It’s global, with manufacturing capabilities extending to Japan. This global presence is a unique advantage for companies looking to commercialize across strict regulatory markets. 

For example, Japan is known for its rigorous pharmaceutical standards. Through our Kawaoge site, actelion can serve as a quality, regulatory, and operational gateway into the Japanese market. We help our customers launch in strict markets like Japan using the knowledge and expertise we have locally. That’s a true differentiator. 

Additionally, our sites have a legacy of over 300 years combined in pharmaceutical excellence, from development to clinical trial supplies and commercial launches. This extensive expertise allows us to support biotech innovators and established pharma alike, in everything from new chemical entities to global product launches. 

A Synergistic Vision 

What truly sets Actelion apart is the synergy between development and manufacturing, between European precision and global ambition, and between our customer-first mindset and operational execution. Athens is not just a facility; it’s pharmaceutical innovation, powered by integration, flexibility, and trust. 

]]>
https://cdmo.actelionpharmaceuticals.de/connecting-development-expertise-with-global-manufacturing/feed/ 0
Understanding cGMP: Key Insights for Effective Manufacturing Practices https://cdmo.actelionpharmaceuticals.de/understanding-cgmp-key-insights-for-effective-manufacturing-practices/ https://cdmo.actelionpharmaceuticals.de/understanding-cgmp-key-insights-for-effective-manufacturing-practices/#respond Tue, 01 Jul 2025 15:04:43 +0000 https://cdmo.actelionpharmaceuticals.de/?p=19427

Table of Contents

When it comes to pharmaceutical quality, nothing is more important than ensuring the products you manufacture or develop meet the strictest standards for safety, efficacy, and purity. In the pharmaceutical industry and life sciences sectors, especially in the United States, this means adhering to current Good Manufacturing Practice—commonly written as cGMP. But what exactly does cGMP mean? How is it implemented, and why is it crucial for pharmaceutical companies, medical device companies, or even dietary supplements manufacturers?

This comprehensive guide will provide you with an expert understanding of cGMP, including its definition, regulatory framework, certification process, procedures, core functions, practical examples, and the foundational Five Ps. Whether you’re new to drug development or an experienced quality management system specialist looking to refresh your knowledge, you’ll find detailed, actionable information here.

What is cGMP? A Clear Definition

cGMP stands for current Good Manufacturing Practice. These are regulations enforced by regulatory agencies such as the Food and Drug Administration (FDA) in the United States and adopted globally through organizations like the World Health Organization (WHO).

The term “current” means the regulations are always evolving—manufacturing practices must stay up-to-date with the latest science, technologies, and process innovations. cGMP guidelines provide the minimum requirements that pharmaceutical companies, medical device manufacturers, and life sciences companies must meet to ensure product quality, ensure safety, and prevent contamination during the manufacturing process.

cGMP encompasses everything from raw materials, facilities equipment, and staff training, to quality control, product quality, and standard operating procedures (SOPs). The ultimate goal is quality assurance throughout the entire production process and manufacturing processes.

The cGMP Regulatory Landscape

In the United States, the FDA regulations relating to cGMP are found in the Code of Federal Regulations (CFR), specifically under 21 CFR Part 210 and 211 for human drugs. The federal register routinely publishes updates, further interpretations, and enforcement decisions regarding gmp regulations.

cGMP regulations are the basis for inspection by regulatory authorities not only in the U.S. but around the world. Other key global regulations include WHO GMP guidelines and the European Union’s EudraLex Volume 4.

The Certification Process: How Do You Get cGMP Certified?

1. Establish a Quality Management System (QMS):

Start by ensuring you have a robust quality management system (QMS) or quality management systems (QMSs) in place. This includes total quality management, documentation controls, and ongoing review.

2. Conduct a Gap Analysis:

Evaluate your facilities, personnel, operating procedures, manufacturing processes, and management systems to find any gaps with current gmp requirements.

3. Address Deficiencies:

Implement corrective actions, update standard operating procedures, enhance staff training (gmp training), and upgrade facilities equipment as necessary.

4. Internal Audit:
Run a mock inspection mimicking FDA or other regulatory agencies to ensure readiness.

5. Apply for Certification:

Apply to an official auditing organization, which could be the FDA itself for U.S. companies, or an accredited certification body for other markets.

6. Undergo Inspection:

Prepare for a thorough evaluation covering everything from documentation and process controls to manufacturing processing and product quality.

7. Achieve Certification:

Once you address all findings, you’ll obtain your cgmp certification and be listed in the federal register as a compliant manufacturer.

cGMP Certification Requirements

What Are the Core Requirements?

  • Proper design of manufacturing processes and facilities equipment
  • Thorough documentation of all operating procedures
  • Use of qualified raw materials and validated manufacturing processes
  • Well-trained personnel, proven through gmp training
  • Efficient quality control and ongoing quality assurance
  • Systems to prevent contamination and ensure product quality
  • Active monitoring of process controls, product quality, and record keeping
  • Quality management practices that ensure ongoing compliance with gmp standards
  • Immediate corrective actions in response to deviations or issues
  • Regular internal and external audits

These requirements are universal across pharmaceutical products, dietary supplements, medical devices, and even new modalities like positron emission tomography.

Practice Examples of Current Good Manufacturing Practices

Here are practical examples of current good manufacturing practices (cGMP) in action:

  • Environmental Monitoring: Regular microbial and particulate checks in cleanrooms used for drug manufacturing.
  • Validation of Cleaning Procedures: Lab-tested cleaning protocols to assure there’s no cross-contamination in the production process.
  • Supplier Qualification: Vetting and monitoring suppliers of raw materials to ensure only high-quality components enter the manufacturing process.
  • Batch Record Review: Reviewing detailed production logs for each batch of pharmaceutical products to verify conformance to quality standards.

These practices ensure that quality products are reliably produced and distributed.

Four Critical cGMP Procedures

The four key procedures integral to current good manufacturing practice are:

  1. 1. Documentation Management: Accurate creation, storage, and retrieval of all manufacturing and quality assurance documents.
  2. 2. Personnel Training: Ongoing gmp training and competency assessments for all staff.
  3. 3. Sanitation and Hygiene: Strict procedures for cleaning facilities and monitoring staff hygiene.
  4. 4. Process Validation: Regular testing and verification that all manufacturing processes and product quality standards are achieved.

Each of these procedures is core to achieving gmp compliance and cgmp compliance.

The 5 Key Elements of cGMP: Understanding the Five Ps

Modern quality management systems in pharmaceutical manufacturing often refer to the “Five Ps of cGMP.” These five foundational pillars—People, Premises, Processes, Products (or Materials), and Procedures—create a cohesive framework for ensuring cgmp compliance throughout the entire manufacturing process. Let’s break down how each contributes to quality assurance and product quality.

1. People

Highly skilled and properly trained personnel are at the core of every quality-driven company. In cgmp manufacturing, all staff involved in manufacturing processes—from operators to quality specialists—must have well-defined roles and responsibilities. Ongoing gmp training ensures staff understand their tasks, can follow standard operating procedures, and are prepared to handle deviations or unexpected events. This focus on people safeguards the integrity of the production process and helps ensure product quality.

2. Premises

Facilities equipment and the overall environment where drug manufacturing or medical device production occurs play a significant role in quality design. The physical premises must be suitable for the type of products made, easy to clean, and organized to prevent cross-contamination. Regular maintenance, cleaning routines, and documented inspections are essential for keeping the workspace compliant with cgmp guidelines and avoiding any risk to product quality or safety.

3. Processes

Every step in the manufacturing process must be clearly defined, validated, and carefully controlled. This includes mapping out each phase—from raw materials receipt, through in-process checks, to finished product release—to ensure consistent output. Robust process controls enable traceability and help regulatory agencies and internal auditors track down the root of any issues. Well-established processes are critical for achieving consistent results and meeting quality standards every time.

4. Products (Materials)

Raw materials and components, as well as finished pharmaceutical products, must adhere to strict quality and safety specifications. Each lot is tested and verified to ensure it matches defined characteristics. Whether your facility is handling active pharmaceutical ingredients, excipients, or packaging, meticulous testing and documented results ensure every batch meets good manufacturing practice expectations and fda regulations.

5. Procedures

Clearly outlined and thoroughly documented procedures are the backbone of every quality management system. Each operation—from cleaning protocols to documentation standards—must have written standard operating procedures (SOPs). Continuous adherence to and regular review of these procedures reduces the risk of human error, facilitates gmp compliance, and supports a culture of total quality management.

Why Are the 5 Ps Important?

The integration of these five elements into the daily routines of pharmaceutical companies and medical device manufacturers ensures a holistic approach to quality. The Five Ps represent comprehensive protection—helping to prevent contamination, guarantee batch consistency, and advance quality safety for patients and end-users. By focusing equally on personnel, environment, processes, materials, and procedures, companies set themselves up for success in both regulatory compliance and the delivery of reliable, high-quality medicinal products.

For organizations that strive for cgmp certification, mastering the Five Ps isn’t just about meeting regulatory requirements but establishing a foundation for operational excellence and long-term trust in the competitive pharmaceutical industry.

The Function of Good Manufacturing Practice

The primary function of good manufacturing practice (GMP)—and its enhanced version, cGMP—is to ensure product quality, ensure safety, and maintain consistency for every batch produced. In broader terms:

  • They protect consumers by verifying that products are safe, effective, and free from contamination.
  • They reduce the risk of product recalls, regulatory penalties, and brand damage for pharmaceutical companies, medical device companies, and the wider pharmaceutical industry.
  • They help companies meet gmp requirements, cgmp regulations, and gmp guidelines—hallmarks of responsible quality design and manufacturing process management.

cGMP in the Real World: Why It Matters

In the fast-evolving life sciences landscape, cGMP has moved from a compliance requirement to a strategic differentiator. For pharmaceutical companies developing active pharmaceutical ingredients or drug administration systems, for medical device manufacturers, and for dietary supplements brands, cGMP standards bolster market access, win consumer trust, and streamline global supply chains.

Modern quality management systems extend beyond simply meeting fda cgmp standards. They incorporate the principles of total quality management, quality safety, robust monitoring control and real-time data analytics to continuously ensure products meet or exceed international quality standards.

cGMPcurrent Good Manufacturing Practice—is the essential foundation that enables pharmaceutical companies, medical device manufacturers, and all life sciences organizations to deliver quality products with safety, efficacy, and integrity. By understanding the cgmp certification process, meeting routine audit requirements, applying good manufacturing practices daily (including the Five Ps), and leveraging modern quality management systems, your organization can not only achieve compliance but also foster continuous improvement.

While cGMP regulations are ever-evolving, a commitment to excellence in manufacturing processes and quality assurance will always be the best strategy for pharmaceutical industry leaders and innovators. At Actelion Pharmaceuticals Deutschland GmbH, we are deeply committed to pharmaceutical quality and safety. We rigorously adhere to cGMP guidelines across all of our facilities and operations, ensuring that our clients and partners always receive products manufactured to the highest standards of compliance, consistency, and care. Through ongoing investment in quality management systems, robust training, and continuous improvement, Actelion Pharmaceuticals Deutschland GmbH remains dedicated to upholding the trust our customers and regulatory authorities place in us.

FAQs About cGMP

What is the meaning of cGMP?

cGMP stands for current Good Manufacturing Practice. It refers to the latest and most up-to-date set of regulations and practices that ensure the quality, safety, and efficacy of products manufactured in the pharmaceutical industry, medical devices, and related sectors.

What does cGMP do in the body?

Scientifically, cGMP can also refer to cyclic guanosine monophosphate, a molecule involved in signaling processes like vasodilation. In manufacturing, however, cGMP ensures that medicines and devices are made to the highest standards and are safe for human use.

Why is the “C” small in cGMP?

The “c” stands for “current”, emphasizing that companies must use the current (most recent and updated) good manufacturing practices.

What does cGMP mean for supplements?

For dietary supplements, cGMP ensures that they’re manufactured under controlled processes, use approved raw materials, and meet stringent quality standards to protect consumer safety.

Are GMP supplements safe?

GMP supplements are considered much safer than non-GMP marked products, provided that the manufacturer maintains gmp compliance. Quality management systems and gmp guidelines help ensure that these supplements do not contain contaminants and are consistently formulated.

Does cGMP apply to food?

Yes, in many cases, food drug administration authorities enforce cgmp regulations for food, beverages, and certain cosmetics to maintain good manufacturing and quality assurance practices.

What are the 10 principles of cGMP?

The 10 principles are:

Continuous improvement and review

Production under controlled conditions

Equipment and environment cleaning

Clear, executable standard operating procedures

Documentation of all activities

Hygiene of personnel and environment

Validation and verification of manufacturing processes

Systematic handling of complaints and recalls

Traceability of products and processes

Well-defined roles and responsibilities

What is the difference between cGMP and non-cGMP?

cGMP manufacturers adhere to the current and most stringent gmp standards and regulatory expectations, ensuring top-grade product quality and regulatory compliance. Non-cGMP manufacturers may lack these rigorous systems, increasing the risk of unsafe or inconsistent products.

]]>
https://cdmo.actelionpharmaceuticals.de/understanding-cgmp-key-insights-for-effective-manufacturing-practices/feed/ 0
Lyophilization: Guide to Freeze Drying in Pharmaceuticals https://cdmo.actelionpharmaceuticals.de/lyophilization-guide-to-freeze-drying-in-pharmaceuticals/ https://cdmo.actelionpharmaceuticals.de/lyophilization-guide-to-freeze-drying-in-pharmaceuticals/#respond Wed, 18 Jun 2025 14:56:26 +0000 https://cdmo.actelionpharmaceuticals.de/?p=18682

Table of Contents

Lyophilization—often called freeze drying—has become an indispensable technology in the pharmaceutical industry. But what is lyophilization exactly, and why do pharmaceutical companies depend on it to safeguard their pharmaceutical and biological products? This blog post presents a deep dive into the lyophilization process, including its definition, advantages, disadvantages, function, stages, and the science behind each step. Whether a pharmaceutical beginner or an expert, you’ll gain a comprehensive understanding of the freeze drying process and its many facets.

What is Lyophilization? A Definition

Lyophilization is a sophisticated low temperature dehydration process used to remove water from sensitive materials by freezing them and then sublimating the ice under low pressure. The process occurs at pressures much lower than atmospheric pressure, which is essential for sublimation to take place. Unlike traditional drying methods, freeze drying rapidly converts water from the solid phase directly to the vapor phase—bypassing the liquid phase—through a process called sublimation. Sublimation is driven by differences in vapor pressure between the ice and the surrounding environment. This technique preserves the material’s physical form, activity, and structure by targeting water molecules, which are removed from the material during lyophilization.

The lyophilization process is particularly crucial for pharmaceutical and biological products that are sensitive to higher temperatures. This process ensures that biologically active ingredients remain stable and potent throughout manufacturing, shipping, and storage.

Key Concepts in the Lyophilization and Freeze Drying Process

The freeze drying process involves a precise sequence of well-controlled steps. Key parameters such as shelf temperature, chamber pressure, and eutectic temperature must be carefully monitored for optimal results. Special equipment like a vacuum pump, refrigeration system, shelf fluid (often silicone oil), and accurate product temperature sensors are integral to achieve both primary drying and secondary drying phases.

The aim is to maintain the integrity of biological materials and extend the shelf life of the final product, which may include vaccines, injectable drugs, or other sensitive dosage forms such as tablets, wafers, powders, pellets, and injectables produced by lyophilization. By producing a freeze dried material with a porous structure, the process allows for rapid and easy rehydration at the point of use. Freeze dried products offer advantages such as improved shelf stability, retention of nutritional and organoleptic qualities, and suitability for specialized applications.

Careful process control and optimization are essential to ensure consistent results. Optimizing freeze drying cycles, including both primary and secondary drying cycles, is crucial for achieving desired product quality, efficiency, and stability.

What Makes It Essential and Difficult in Lyophilization

Lyophilization is widely used in pharmaceutical manufacturing due to its exceptional ability to preserve drug efficacy, stabilize sensitive products, and extend shelf life. Despite these significant benefits, the process also poses challenges that require careful consideration and expert control.

Advantages of lyophilization:

  • Superior preservation of drug efficacy and quality
  • Extended shelf life for sensitive products
  • Maintains the stability and structure of delicate drug substances
  • Allows for easy and safe reconstitution before use
  • Facilitates transport and long-term storage of pharmaceuticals

Disadvantages of lyophilization:

May complicate reconstitution if the process is not well controlled

The cycle is considerably longer than traditional drying methods

High capital investment and operational costs are required for specialized equipment

Process control is critical, improper optimization can reduce product activity or damage structure

Risks of instability or degradation if not carefully monitored

Potential formation of large ice crystals during slow freezing, which can harm product quality.

Is Lyophilization Healthy? Impact on Pharmaceuticals

For the pharmaceutical industry, a key question is whether freeze drying affects the safety and effectiveness of products. Scientific evidence and quality control standards show that lyophilization is a safe process. By maintaining low temperature during the freezing phase and subsequent primary drying sublimation, the original activity and stability of most active components are preserved.

Unlike some drying methods that may use harmful chemicals or excessively high temperatures, freeze drying uses physical means—namely cold, vacuum pump action, and controlled heat transfer—to gently dehydrate the product. The result is a freeze dried product that can safely and easily be reconstituted without compromising patient health or the drug’s efficacy.

The Freeze Drying Method: How Lyophilization Works

The lyophilization process is characterized by three main steps:

A. Freezing Step

The freezing process is the initial and critical step in lyophilization. The product is cooled below its freezing temperature (often with liquid nitrogen) so water forms ice crystals. This freezing stage determines the size and distribution of ice crystals, which impacts product stability and the efficiency of the drying cycle. Ice formation is influenced by the rate of cooling and atmospheric pressure conditions during the freezing process. Both slow freezing and rapid freezing methods can be used; slow freezing often results in large ice crystals. Manipulating atmospheric pressure during the freezing process can influence ice formation and the structure of the frozen product. During this stage, water within the product matrix becomes frozen water, which will be removed during subsequent drying. Monitoring of eutectic temperature and freezing phase is crucial to avoid unwanted transitions.

B. Primary Drying (Sublimation)

At reduced chamber pressure and controlled shelf temperature, the primary drying phase begins. Here, the vacuum pump lowers the pressure below the triple point, and water vapor is removed from the frozen material by sublimation. Fine precise control over parameters such as product temperature and shelf fluid temperature—often managed with silicone oil—ensures that only the ice (not the solid matrix) is removed. Successful primary drying yields up to 95% water removal.

C. Secondary Drying (Desorption)

After sublimation, some water remains adsorbed. The secondary drying phase involves raising the shelf temperature (still under low pressure) to drive off this bound water. This is critical for achieving optimal shelf life and product stability.

Infographic explaining the steps in the lyophilization process

Temperature Control in Lyophilization

Temperature control is fundamental to the success of the lyophilization process, directly impacting the quality, stability, and efficacy of freeze dried pharmaceuticals. During the freeze drying process, the product is first cooled below its triple point to ensure complete solidification. In the primary drying phase, the shelf temperature is carefully maintained—typically between -30°C and -50°C—to promote sublimation without exceeding the product’s eutectic temperature. The condenser temperature is set even lower, often between -60°C and -80°C, to efficiently capture water vapor as it leaves the frozen material.

Precise control of product temperature is essential to prevent melt back or collapse, which can occur if the product is exposed to too much heat. Advanced temperature control systems, sometimes utilizing liquid nitrogen or other cryogenic fluids, enable rapid freezing and maintain the low temperature required throughout the process. This careful management ensures that both primary drying and secondary drying phases proceed efficiently, preserving the porous structure and potency of the final product. By keeping the product temperature below critical thresholds—such as the eutectic point—manufacturers safeguard the integrity of sensitive pharmaceutical products during every stage of the drying process.

Refrigeration and Equipment Used in Lyophilization

The effectiveness of the freeze drying process relies heavily on specialized equipment designed for precise control of temperature and pressure. At the heart of any freeze dryer is a robust refrigeration system, which cools both the condenser and the shelf fluid to the low temperatures necessary for successful sublimation. The condenser temperature typically operates between -60°C and -80°C, efficiently trapping water vapor, while the shelf fluid—often silicone oil—maintains the shelves at -30°C to -50°C to keep the product frozen during primary drying.

A powerful vacuum system is also essential, creating a low pressure environment (usually between 50-200 mTorr) within the vacuum chamber. This reduced pressure enables the direct transition of ice from the solid phase to vapor phase, bypassing the liquid phase and ensuring gentle dehydration of the frozen water in the product. The freeze dryer’s control system allows for precise adjustment of all parameters, including shelf temperature, chamber pressure, and drying time, ensuring optimal results for each batch. Together, these components provide the precise control needed to produce high-quality freeze dried materials in the pharmaceutical industry.

How Long Does Lyophilization Take?

The length of the freeze drying process varies based on product type, chamber pressure, and desired moisture level. The number and duration of freeze drying cycles, including both primary and secondary drying cycles, can significantly impact the total processing time and product quality. Most lyophilization cycles take from several to many hours, sometimes even days, depending on freezing step, primary drying, and secondary drying duration. Rapid heat transfer and efficient vacuum pump operation facilitate shorter cycles, but too much heat can damage the product. Hence, precise control of product temperature, shelf temperature, and eutectic temperature is key.

Why Do Pharmaceutical Companies Rely on Lyophilization?

The ability to extend shelf life, maintain the physical form, and provide products in convenient final containers makes lyophilization invaluable for the pharmaceutical industry. Pharmaceutical applications that require long-term stability at room temperature, rapid dissolution, and preserved potency benefit most from this technology. Lyophilized formulations safeguard labile products such as vaccines, blood plasma, and biologics, serving both commercial and emergency needs.

The Function and Importance of Lyophilization

The main function of freeze drying is to offer unmatched stability for temperature-sensitive biological materials and pharmaceutical products. By removing water while preserving a stable physical form and active structure, the lyophilization process ensures quality control for sensitive dosage forms. This results in pharmaceutical products that have a significantly extended shelf life, easier transport logistics, and lower risk of degradation.

Science and Optimization in the Freeze Drying Process

Process optimization plays a vital role in every lyophilization cycle. Specialists monitor parameters like eutectic point, condenser temperature, shelf fluid flow, vacuum system performance, and triple point stability. Advanced PAT (Process Analytical Technology) tools, including infrared radiation thermography, help finely tune drying kinetics, ensuring reliable product outcomes batch after batch. This ongoing optimization is essential to minimize risks of too much heat exposure, large ice crystals formation, and inefficacies in primary and secondary drying. Optimizing freeze drying cycles, including primary and secondary drying cycles and parameters such as shelf temperature, is crucial not only in pharmaceuticals but also in other industries such as the food industry.

Lyophilization—the gold standard in freeze drying—has transformed the pharmaceutical industry by enabling safe, effective, and long-lasting pharmaceutical and biological products. The food industry also uses freeze drying to preserve high-value products like seasonal fruits, vegetables, instant soups, and coffee, extending shelf-life and maintaining quality. With stringent quality control and a deep understanding of the complex lyophilization process, pharmaceutical companies can deliver innovative, life-saving therapies worldwide. A robust command of parameters like eutectic temperature, primary drying phase, and secondary drying is imperative for optimizing every lyophilization cycle and ensuring the best patient outcomes. After lyophilization, an inert gas such as nitrogen is often used to break the vacuum and preserve product sterility.

Strategic Benefits of Working with a CDMO Partner

Partnering with a contract development and manufacturing organization (CDMO) offers pharmaceutical companies significant strategic advantages in the development and production of lyophilized products. CDMOs bring specialized expertise in formulation development, process optimization, and scale-up, ensuring that each step of the freeze drying process is executed with precise control and efficiency. Their access to advanced freeze drying equipment and analytical instrumentation allows for rigorous quality control and compliance with regulatory standards such as Good Manufacturing Practice (GMP).

Swiss Excellence in Lyophilization: Our Jura Facility at Your Service

Located in the heart of Switzerland, our Jura facility combines over 20 years of specialized expertise with GMP and FDA certification to offer exceptional lyophilization and aseptic fill-finish services for (bio)pharmaceutical products. We support both clinical and commercial manufacturing, with a strong focus on small and flexible batch production—and no minimum batch size requirements. Our lyophilization capabilities include everything from process scale-up and technology transfer to robust project management, aseptic process simulation (APS), and comprehensive stability studies. This ensures your sensitive formulations preserve their activity, potency, and structure at every stage, whether at pilot, clinical, or commercial scale. Our skilled team optimizes each freeze drying cycle and provides dedicated regulatory support, enabling efficient transitions from development to GMP storage and global supply. By integrating cutting-edge technology, meticulous quality control, and tailor-made solutions, our Jura facility stands out as your trusted partner for high-quality, reliable, and fully compliant lyophilized product manufacturing—empowering your success at every step of pharmaceutical innovation.

By leveraging a CDMO’s experience and resources, pharmaceutical companies can accelerate product development timelines, extend shelf life, reduce capital investment in specialized equipment like vacuum systems, and focus on their core strengths such as drug discovery and commercialization. CDMOs also help navigate complex regulatory requirements, ensuring that freeze dried pharmaceuticals are developed and manufactured to the highest standards of quality and efficacy. This collaborative approach supports successful product launches, robust lyophilization cycles, and long-term market success in the pharmaceutical industry.

FAQs about Lyophilization and Freeze Drying

What is lyophilization and why is it used?

Lyophilization, or freeze drying, is a low temperature dehydration process used to preserve sensitive pharmaceutical and biological products. It works by freezing the product and sublimating ice under low pressure, effectively removing water while maintaining product stability and efficacy. This process increases shelf life and maintains potency during storage and transport.

Why are drugs lyophilized?

Many drugs are lyophilized to extend shelf life, stabilize dosage forms, and protect biological materials from degradation during transportation and storage. It is especially crucial for labile injectables, vaccines, and certain antibiotics.

What are the disadvantages of lyophilization?

The main disadvantages are long lyophilization cycle times, high capital and operational costs, risk of too much heat or improper heat transfer damaging sensitive products, and possible formation of large ice crystals that may affect the material’s cell walls and porous structure. Stringent quality control is essential to overcome these limitations.

What are the 4 steps of freeze-drying?

The freeze drying process includes:

Stoppering/final containers: Sealing the freeze dried product to maintain stability and maximize shelf

Freezing step: Cooling the product below its freezing temperature to form ice.

Primary drying phase: Subliming ice under vacuum pump at carefully controlled shelf temperature.

Secondary drying: Removing adsorbed water by increasing temperature under continued low pressure.

What temperature is lyophilization?

Lyophilization (freeze-drying) typically occurs at temperatures between -40°C and -80°C during the initial freezing stage.

  • Secondary drying (desorption) stage: The temperature is increased further (often 0°C to +30°C, sometimes up to +40°C) to remove residual moisture.
  • Freezing stage: The product is frozen at very low temperatures, usually between -40°C and -80°C.
  • Primary drying (sublimation) stage: While under vacuum, heat is gradually applied to raise the temperature, but it generally remains below 0°C (often from -20°C to -5°C) to ensure sublimation of ice.
]]>
https://cdmo.actelionpharmaceuticals.de/lyophilization-guide-to-freeze-drying-in-pharmaceuticals/feed/ 0
Flexibility: Key to Success in Contract Fill & Finish Operations https://cdmo.actelionpharmaceuticals.de/flexibility-key-to-success-in-contract-fill-finish-operations/ https://cdmo.actelionpharmaceuticals.de/flexibility-key-to-success-in-contract-fill-finish-operations/#respond Mon, 16 Jun 2025 12:28:24 +0000 https://cdmo.actelionpharmaceuticals.de/?p=18612 At actelion, we always put our customers first. And here in Jura, we have invested in flexible filling capacity so we can continue to meet their needs as they change and evolve over time.  

Future demand can be unpredictable, and the characteristics of products entering development are not static. A key consideration when we were designing our new fill and finish line in Jura was therefore to ensure it had the flexibility to accommodate a wide range of different products. 

As part of the design process, we looked for some insight into the products customers might be making in the coming years, and therefore what assets and capabilities we are likely to need in future. If, for example, a new modality is in the early stages of research, a filling line specifically tailored to current product types may not suffice. As well as the drug product itself, these predictions extended to the packaging materials and components we might need to use to meet future demand.  

Whenever a customer comes to us with a new project, the first step is a technical assessment of the product to determine whether it is compatible with our assets. Greater flexibility, of course, means we are more likely to be able to accommodate it here, and with the experience we have on site. It’s a good idea to run feasibility studies early on to ensure it will indeed fit in with our capabilities and equipment, and whether any adaptations to our current assets might be required. 

Fine-tuning operations 

Constant discussion and communication with customers is also important. This feedback will help us understand what modifications to the line might be required and allow us to implement them more quickly. We also need to be transparent about project progress, reinforcing the trust built up between us. This feedback loop helps us fine-tune operations to meet their needs, and allows us pass on our expertise to them. For example, we are used to managing complex formulations, including suspensions and highly viscous products. It allows us to suggest the most effective technical solutions if their product proves challenging.  

We also want to maximise the use of single-use components in our processes. It is usually faster, as it reduces both changeover times and validation requirements, while also cutting the chances of cross-contamination. It also increases our flexibility. It is possible to make much of the line single use – from the product tank at the start right through to the filling needles, tailored to the product and perfectly adapted to our equipment. Even something as simple as using pre-sterilised stoppers can save time and reduce risk.  

Careful coordination of all functions is essential. Whether using an existing line, revamping a line or implementing a new line, these are really complex projects involving many different companies and competencies. At the start of each shift, we meet to determine what tasks are scheduled that day, and what the priorities are. The more activities we can carry out safely in parallel, the faster the whole process will be.  

While it’s a small site here at Jura, we have all the necessary competencies needed on-site – and our small size makes us fast and efficient. We monitor progress of commissioning and equipment qualification on a daily basis, constantly adapting and challenging the schedule. We try to debottleneck as far as is practical, and anticipate what problems could arise in the future, whether from a technical perspective or in terms of resources. We always try to be one step ahead.  

]]>
https://cdmo.actelionpharmaceuticals.de/flexibility-key-to-success-in-contract-fill-finish-operations/feed/ 0
Parenteral Route: Types, Benefits, & Uses in Pharma Industry https://cdmo.actelionpharmaceuticals.de/parenteral-route-types-benefits-and-uses-in-pharma-industry/ https://cdmo.actelionpharmaceuticals.de/parenteral-route-types-benefits-and-uses-in-pharma-industry/#respond Tue, 10 Jun 2025 15:26:15 +0000 https://cdmo.actelionpharmaceuticals.de/?p=18410 In the pharmaceutical industry, understanding various drug administration routes is crucial for optimizing therapeutic outcomes. The main routes of administration include oral (administered orally), parenteral, topical, and others.

Among these, the parenteral route stands out for its ability to deliver medications directly into the body, bypassing the gastrointestinal tract. Some drugs cannot be administered orally because they are poorly absorbed through the gastrointestinal tract. Drugs administered orally must pass through the digestive system, which can limit their effectiveness for certain medications. This method is particularly valuable when rapid drug action is required or when oral administration is not feasible.

Understanding the Parenteral Route

The term “parenteral” originates from Greek, meaning “outside the intestine.” In medical practice, the parenteral route refers to delivering substances directly into the body, typically through injections, thereby circumventing the gastrointestinal tract. Parenteral routes include various injection techniques, such as intradermal, subcutaneous, and intramuscular, which are used for the safe and effective delivery of parenteral medications. This approach enables systemic administration of drugs, allowing them to enter the systemic circulation promptly for rapid therapeutic effects.

Advantages of Parenteral Administration and Drug Absorption

Parenteral administration offers several benefits:

  • Rapid Onset of Action: By introducing drugs directly into the bloodstream, the parenteral route ensures swift therapeutic effects, essential in emergencies, due to rapid absorption.
  • Precise Dosage: This method allows for accurate dosing, minimizing variability in drug absorption.
  • Bypassing First-Pass Metabolism: Unlike oral medications, parenteral drugs avoid degradation in the liver, enhancing bioavailability.
  • Suitable for Unconscious or Uncooperative Patients: When patients cannot take medications orally, parenteral administration becomes indispensable.

Injectable medications are commonly used when rapid absorption and precise dosing are required, ensuring safe injection practices and reducing the risk of infection transmission.

Types of Parenteral Injections

There are four primary types of parenteral injections:

1. Intravenous (IV)

Intravenous injections are used to inject medication directly into the veins, ensuring immediate entry into the systemic circulation. This method is ideal for rapid drug delivery and is commonly used in emergency settings.

2. Intramuscular (IM)

Intramuscular injections involve administering medication into muscle tissue. Selecting the appropriate injection site, such as the deltoid or vastus lateralis, is crucial for safe and effective intramuscular injections. Proper identification and preparation of injection sites help prevent complications and ensure effective medication delivery. This route allows for moderate absorption speed and is often used for vaccines and hormonal therapies.

3. Subcutaneous (SC)

Subcutaneous administration involves delivering drugs via subcutaneous injection into the fatty tissue beneath the skin. The subcutaneous route is characterized by slower absorption because subcutaneous tissue contains fewer blood vessels than muscle. When selecting a subcutaneous injection site, it is important to avoid major blood vessels to ensure safe and effective medication delivery. This method provides slower, sustained drug absorption, suitable for medications like insulin.

4. Intradermal (ID)

Intradermal injections are given just beneath the skin’s surface, primarily for diagnostic purposes like allergy testing.

Equipment and Devices Used in Parenteral Administration

The success and safety of parenteral administration rely heavily on the selection and proper use of specialized equipment. Each route of administration—whether intravenous, intramuscular, subcutaneous, or intradermal—requires specific devices designed to optimize drug delivery and minimize risks for both patients and health care workers.

Key equipment and devices include:

  • Syringes and Needles: Available in various sizes and gauges, syringes and needles are chosen based on the type of medication, volume to be administered, and the selected route of administration. Using the correct combination ensures accurate dosing and reduces the risk of tissue damage or medication errors.
  • Infusion Sets: For intravenous administration, infusion sets—including IV catheters, tubing, and connectors—facilitate the controlled delivery of fluids and medications directly into the bloodstream. These sets are essential for continuous or large-volume infusions.
  • Multi Dose Vials and Plastic Containers: Multi dose vials and multi dose plastic containers allow for the withdrawal of multiple doses from a single container, supporting efficient medication administration in clinical settings. Proper aseptic technique is critical to prevent contamination.
  • Engineered Safety Devices: To enhance safe medication administration and reduce the risk of sharp related injuries immediately, many facilities use engineered safety devices such as retractable needles, needleless systems, and safety syringes. These innovations are vital for injury prevention strategies related to blood borne illness and body fluid exposure management.
  • Sterile Preparation Tools: Maintaining sterility is paramount in parenteral medication administration. Equipment such as sterile gloves, alcohol swabs, and syringe sterile packaging help ensure infection control throughout the injection procedure.
  • Specialized Devices: Certain medications or patient populations may require specialized devices, such as infusion pumps for precise dosing, or auto-injectors for self-administration of injectable medicines.

Selecting the appropriate equipment for each route of administration not only supports rapid and effective absorption but also aligns with national patient safety agency guidelines and best practices for safe medication administration. Proper training in the use of these devices is essential for health care providers to prevent medication errors, ensure patient safety, and maintain the integrity of parenteral drug administration.

Safety and Infection Control Considerations in Parenteral Administration

Ensuring patient safety during parenteral medication administration involves:

  • Proper Technique: Using the correct needle length and ensuring the needle does not touch unsterile surfaces such as vial edges, needle caps, or counters to prevent contamination.
  • Infection Control: Adhering to strict infection control protocols to prevent blood borne illness.
  • Increased Risk and Adverse Reactions: Recognizing the increased risk of infection and adverse reactions associated with parenteral administration due to the breach in the skin barrier, and emphasizing the need for strict aseptic technique.
  • Disease Control: Implementing disease control measures, such as safe injection practices, to prevent healthcare-associated infections.
  • Accurate Documentation: Maintaining a detailed medication administration record to track dosages and prevent medication errors.
  • Patient Identification: Verifying patient identifiers before administering medication to ensure the correct medication is given.

Addressing Patient Concerns

Administering medication via injections can cause patient’s anxiety heightened pain. Strategies to reduce muscle tension and help the patient relax include:

  • Clear Communication: Explaining the procedure to alleviate fears.
  • Comfort Measures: Using distraction techniques or topical anesthetics.
  • Professionalism: Ensuring swift and skilled administration to minimize discomfort.

The parenteral route is a vital component in modern medicine, offering rapid and effective drug delivery, especially when oral routes are unsuitable. At Actelion Pharmaceuticals Deutschland GmbH, our expertise in parenteral administration ensures high-quality, sterile products tailored to meet diverse therapeutic requirements.

Frequently Asked Questions (FAQs) about Parenteral Route

What are the 4 types of parenteral injections?

The main types are intravenous (IV)—straight into the vein; intramuscular (IM)—into muscle; subcutaneous (SC)—under the skin; and intradermal (ID)—just beneath the skin surface.

What is called parenteral?

“Parenteral” covers any medication administration that avoids the digestive tract, relying on direct injection into body tissues or the circulatory system.

What is the difference between enteral route and parenteral route?

Enteral (like oral administration or rectal drug administration) uses the GI tract; parenteral goes outside it, most often via injection.

Is oral a parenteral route?

No, the oral route is not a parenteral route. It involves administering medication through the mouth, utilizing the gastrointestinal tract for drug absorption.

Are inhaled medications considered a route of administration?

Yes, inhaled medications are a recognized route of administration. Drugs are delivered directly to the lungs using inhalers or nebulizers. This method allows for rapid absorption, can provide local effects in the respiratory tract, and minimizes systemic side effects. Proper inhaler technique and particle size are important for effective lung deposition.

How are drugs delivered to the central nervous system?

Certain parenteral routes and specialized techniques, such as intranasal or intrathecal administration, can deliver drugs directly or indirectly to the central nervous system (CNS). These methods bypass traditional barriers, allowing for rapid and targeted therapeutic effects within the brain and spinal cord while reducing systemic exposure.

]]>
https://cdmo.actelionpharmaceuticals.de/parenteral-route-types-benefits-and-uses-in-pharma-industry/feed/ 0
Non-Sterile Liquids in Pharma: Options & CDMO Solutions https://cdmo.actelionpharmaceuticals.de/non-sterile-liquids-in-pharma-options-cdmo-solutions/ https://cdmo.actelionpharmaceuticals.de/non-sterile-liquids-in-pharma-options-cdmo-solutions/#respond Wed, 04 Jun 2025 14:41:29 +0000 https://cdmo.actelionpharmaceuticals.de/?p=18333 In today’s pharmaceutical landscape, non-sterile liquids are more than just simple formulations. They are precision-crafted medical products with demanding regulatory, therapeutic, and commercial requirements. Whether you are launching an oral solution, expanding into disinfection products, or adapting a formulation for pediatric or geriatric use, the manufacturer you choose can either fast-track your success or become a bottleneck. Selecting the right CDMO is crucial for ensuring product quality, safety, and compliance.

So, what exactly are your options when it comes to non-sterile liquids? And how do you decide which CDMO is best equipped to handle the complexity of your project?

Let’s break down the major categories, challenges, and decision points and why more healthcare companies are trusting Actelion Pharmaceuticals Deutschland GmbH to bring their pharmaceutical products to life and serve their customers with high-quality non-sterile liquids.

The Role of Non-Sterile Liquids in Drug Development

Non-sterile liquids are used across a wide range of medical procedures, especially where full sterilization is not required but microbial safety still matters. Controlling bacteria is essential to prevent contamination in non-sterile liquids. Unlike sterile manufacturing, these formats are typically used for oral or topical drug administration, and must still meet stringent quality controls to prevent contamination and cross contamination.

They are also a core part of healthcare professionals’ everyday toolkit, from pediatric antibiotics and dermatology emulsions to antiseptic sprays and wound treatments.

How Non-Sterile Liquids Compare to Sterile Products

Non-sterile liquids and sterile products serve distinct roles in healthcare, each designed for specific applications and risk profiles. While sterile products are manufactured to be completely free of microorganisms, making them essential for clinical settings where the risk of infection is critical, such as injections, infusions, or direct contact with the bloodstream, non-sterile liquids are intended for situations where this same level of sterility is not required.

Non-sterile liquids, such as oral drops, nasal sprays, and certain wound care solutions, are formulated for use on the skin, in the mouth, or in other areas where the body’s natural barriers help prevent infection. These products are not manufactured to the same level of sterility as injectables or ophthalmic solutions, but they are still carefully controlled to minimize the presence of harmful microorganisms and avoid contamination. For example, oral drops and nasal sprays are effective for treating common conditions like coughs, colds, or minor wounds, where the risk of infection is lower and the product’s intended use does not demand absolute sterility.

In contrast, sterile products are designed for high-risk environments and applications, such as surgical procedures or direct administration into the bloodstream, where even minimal contamination could lead to serious infection. The manufacturing process for sterile products is far more stringent, involving aseptic technique and specialized facilities to ensure complete sterility.

Ultimately, non-sterile liquids are a practical and effective choice for many medical and consumer health applications. They provide reliable treatment options for skin care, wound management, and oral administration, without the added complexity and cost of sterile manufacturing, making them ideal for a wide range of products and situations.

The Main Types of Non-Sterile Liquid Formulations

Choosing the right format for your product is essential not just for therapeutic efficacy, but for market success. Here is a breakdown of the main types:

1. Oral Solutions: Clean, Clear, and Patient-Friendly

Oral solutions are liquid medications with dissolved APIs, favored for their ease of swallowing and quick absorption. They are common in pediatrics, geriatrics, and chronic care.

✅ Best for: Taste-masked formulations, precise dosing
✅ Challenges: Flavor integration, long-term stability, microbial control
✅ Packaging: Plastic or glass bottles, often with measuring caps or spoons. The choice of material for bottles and closures is crucial, as it affects product stability, compatibility, and safety.

This is where manufacturing precision matters. Consistency, homogeneity, and filtration systems must work flawlessly.

2. Oral Drops: Micro-Dosing with Maximum Control

Oral drops require low-volume, high-precision dosing, typically for infants or niche therapeutic areas. Because they are so concentrated, there is a fine line between efficacy and risk, making microbial integrity and dosing accuracy critical.

✅ Best for: Sensitive APIs, infant formulations, niche indications
✅ Packaging: Dropper bottles with calibrated closures
✅ Considerations: Avoiding leaks, ensuring stability, and selecting the right method to ensure both dosing accuracy and effective microbial control

For healthcare professionals, oral drops are essential tools but for manufacturers, they’re one of the most technically challenging formats in the production process.

3. Suspensions: When Solubility Is a Challenge

Sometimes, a drug doesn’t dissolve, so you suspend it. Suspensions are heterogeneous formulations where particles float in a liquid base. They are commonly used in wound care products or pediatric antibiotics. In many cases, suspensions require wetting agents to ensure that the particles are properly dispersed and do not clump together, highlighting the importance of the wet nature of these formulations.

✅ Best for: Poorly soluble drugs, taste-masked applications
✅ Risks: Sedimentation, inconsistent dosing
✅ Solution: Controlled particle sizing, homogenization, “shake-before-use” labeling

This is where compounding expertise shines: the margin for error is slim, and poorly handled suspensions can lead to dosing inaccuracies or microbial growth. Different methods for controlling particle size and preventing microbial contamination are essential to ensure the safety and effectiveness of suspensions.

4. Emulsions: Blending Oil and Water, the Right Way

Emulsions combine oil and water with stabilizers, but keeping the mix stable over months (or years) is no small feat. If an emulsion loses its stability, it becomes compromised, which can affect safety and efficacy. Therefore, it is essential to formulate durable emulsions that can withstand storage and handling. Used in topical or oral formats, emulsions are ideal for delivering lipophilic or hydrophilic APIs in a smooth, patient-friendly product.

✅ Use cases: Dermatology, oral lipophilic drugs, medical device adjuncts
✅ Formats: O/W and W/O emulsions, glass or plastic bottles
✅ Key concerns: Droplet size, phase separation, preservative compatibility

Perfecting emulsions involves real science, from emulsifier selection to sterile compounding techniques (where applicable) and understanding how they behave under stress.

5. Disinfectants: Safety, Compliance, and Scale

Disinfection products are a world of their own. Often alcohol-based and flammable, they require special facilities, ATEX-compliant processes, and packaging expertise to ensure both efficacy and safety.

✅ Formats: Liquids or sprays in 25 ml to 1,000 ml bottles
✅ Considerations: Volatility, regulatory classification (drug or biocide)
✅ Applications: Surface sterilization, wound cleaning, healthcare use

Used by healthcare workers, patients, and even institutions aligned with World Health Organization protocols, these products need high throughput without sacrificing GMP compliance. Maintaining a reliable supply of disinfectants is essential for healthcare and institutional needs to ensure safety and operational continuity.

The Manufacturing Process for Non-Sterile Liquids

The production process of non-sterile liquids is a carefully controlled procedure designed to ensure product safety, stability, and quality without the need for full sterility. It begins with precise compounding, where active ingredients are blended to achieve the desired consistency, therapeutic effect, and shelf-life. This step is critical for ensuring that oral solutions, drops, emulsions, and ointments deliver consistent results with every dose.

Once formulated, non-sterile liquids are packaged in containers such as glass bottles or specialized plastic bottles, chosen for their ability to maintain product stability and prevent contamination. The packaging process is engineered to minimize the risk of cross contamination, with facilities and equipment designed to keep the product free from unwanted microorganisms and environmental contaminants.

Quality control is a cornerstone of non-sterile liquid manufacturing. Each batch undergoes rigorous testing to confirm that it meets established safety and efficacy standards. This includes checks for microbial content, physical stability, and packaging integrity. Facilities like those operated by actelion in Leipzig and Halden are purpose-built for the production of non-sterile liquids, following EU-GMP guidelines to ensure every product is manufactured to the highest standards for medical and healthcare applications.

Non-sterile liquids are versatile and practical alternatives to sterile products in many situations. They are used not only as oral solutions and drops, but also as wound dressings, skin cleansers, and topical ointments, offering flexible solutions for a variety of medical and consumer needs. By focusing on robust formulation, secure medical device packaging, and stringent quality control, manufacturers can deliver non-sterile liquids that are safe, effective, and ideally suited to their intended applications in both healthcare and pharmaceutical products.

Why Actelion Pharmaceuticals Deutschland GmbH Is the Partner You’re Looking For

With the two manufacturing facilities in Germany (Leipzig) and Norway (Halden), Actelion Pharmaceuticals Deutschland GmbH offers full-service CDMO capabilities for non-sterile liquids, from R&D and pharmacy compounding to commercial-scale production.

Here’s what makes us different:

Specialized Facilities

  • Leipzig: Oral solutions, drops, suspensions, emulsions (up to 4,000L)
  • Halden: High-volume disinfectants (up to 5,300L, ATEX-compliant)

Regulatory Confidence

  • EU-GMP certified
  • Compliant with medical device and biocide regulations
  • End-to-end packaging and serialization

Whether you are launching a pediatric suspension or scaling up a portfolio of hospital disinfectants, we make the complex simple — and the difficult achievable.

Every format, API, and use case in the non-sterile liquid space presents a unique challenge. Choosing a partner that understands the different types of formulations, the medical context, and the production process, from medical grade packaging to pharmacy compounding needs, is the only way to ensure long-term success.

At actelion, we do not just manufacture. We collaborate, solve problems, and help your product make the leap from idea to shelf safely, efficiently, and at scale.

FAQs about Non-Sterile Liquids

What are non-sterile liquids?

Non-sterile liquids are pharmaceutical or healthcare formulations that do not require complete sterility but must meet strict microbial quality standards to ensure safety during use.

What are examples of non-sterile?

Examples include oral solutions, suspensions, emulsions, oral drops, and disinfectants. They are commonly used in medical settings, consumer health, and daily wound care. Non-sterile dressings are often used for covering minor wounds where sterility is not critical. In wound care, non-sterile swabs may be used to absorb blood and exudate during cleaning and dressing changes.

What are sterile liquids?

Sterile liquids are formulations that must be completely free from microorganisms, typically used in injectables, ophthalmic solutions, or surgical procedures. Their production involves sterile manufacturing, aseptic technique, and specialized environments. Sterile liquids must be free from contaminants such as bacteria, fungi, and viruses to ensure patient safety.

What are non-sterile items?

Non-sterile items include non sterile gloves, face masks, topical medications, and some medical devices. These are typically used in lower-risk settings or where sterilization is not essential to function or safety. When using non-sterile items, proper handling and storage are important to prevent cross contamination.

]]>
https://cdmo.actelionpharmaceuticals.de/non-sterile-liquids-in-pharma-options-cdmo-solutions/feed/ 0