Twitter Linkedin Youtube Campus Rubió
NEWS AND COMMUNICATIONS

History, Challenges and Opportunities in Biopharmaceuticals

What Are Biopharmaceuticals?

Biopharmaceuticals, also called biologics, are drugs developed using living cells or biological systems rather than chemical synthesis. Unlike small-molecule drugs, which are created through chemical reactions, biopharmaceuticals involve complex biological processes, including gene manipulation and cell cultivation. These innovative therapies include therapeutic proteins, monoclonal antibodies, gene therapies, and vaccines.

The biopharmaceutical definition extends beyond simple categorization. Biopharmaceuticals represent a leap forward in precision medicine, offering targeted mechanisms of action and the potential to treat previously incurable conditions. Their special ability to work with certain biological pathways makes them different from regular medicines. This quality is important for the future of healthcare.

The History and Development of Biopharmaceuticals

The journey of biopharmaceutical development began in the 1970s, with the creation of recombinant DNA technology. Scientists combined DNA from different organisms to produce human proteins in bacteria, a breakthrough that led to the FDA approval of recombinant human insulin in 1982. This became the world’s first marketed biopharmaceutical drug.

The use of Chinese Hamster Ovary (CHO) cells followed soon after, becoming the gold standard for producing therapeutic proteins due to their high productivity and regulatory acceptance (first recombinant expression in CHO cells reported in 1981, with widespread adoption in the 1980s).

Biotechnology companies like Genentech, Amgen, and Biogen pioneered this space, ushering in the modern age of biologics (first CHO-produced therapeutic approved in the 1980s).

Over the past four decades, biopharmaceuticals have evolved from basic protein replacement therapies to complex solutions like CAR-T cell therapy, mRNA vaccines, and CRISPR-based gene editing. Each stage in this development reflects the industry’s adaptability, driven by deeper biological insights and technological innovation.

  • • 1970s: Recombinant DNA developed. The first recombinant DNA molecules were engineered. Paul Berg created them in 1972, followed by Cohen & Boyer’s successful insertion into E. coli in 1973.
  • • 1982: First Approved Biopharmaceutical: Recombinant Insulin (Humulin)
    In 1978, Genentech engineered human insulin in E. coli; by 1982, the FDA had approved Humulin, the first biopharmaceutical and first genetically engineered therapeutic
  • • CHO cells: Enabled scalable protein manufacturing and trace back to the 1950s, but became central to biotech manufacturing later.
    • • The first CHO-derived biologic approved by the FDA was tissue plasminogen activator (Activase) in 1987 Danaher Life SciencesPMCBioMed CentralEuropean Pharmaceutical Manufacturer.
    • • Nowadays over 70% of approved recombinant proteins are produced using CHO cells.
  • • Biotechnology companies: Genentech, Amgen, and Biogen led early breakthroughs
    • – Genentech (founded 1976): Pioneered recombinant insulin and licensed it to Eli Lilly; went on to develop growth hormone, interferons, Activase, Rituxan, Herceptin, and more
    • – Amgen (founded 1980): Focused early on recombinant DNA, cloned the erythropoietin (EPO) gene, and developed Epogen (approved 1989). Later followed with Neupogen (G-CSF, approved 1991) (Amgen.)
    • – Biogen (founded 1978): Established in Geneva by molecular biology pioneers; early work included interferon-alpha production

Biopharmaceuticals in Modern Medicine

Today, biopharmaceuticals are transforming healthcare by offering highly targeted therapies that act at the molecular level, ensuring greater efficacy and fewer side effects compared to conventional drugs. From monoclonal antibodies like trastuzumab and rituximab to breakthrough treatments such as CAR-T cell therapies, mRNA vaccines, and gene therapies like Luxturna and Zolgensma, these innovations are redefining clinical practice across oncology, immunology, and rare diseases. As highlighted in Biopharmaceuticals: From Production to Application in Modern Healthcare biopharmaceuticals represent a rapidly expanding class of medicines that leverage biological insights for precision treatment. Similarly, research by Małgorzata Kesik-Brodacka in Biotechnology and Applied Biochemistry underscores their high specificity and lower risk of adverse effects compared to small-molecule drugs. This growing body of evidence confirms that biopharmaceuticals are not only revolutionizing patient outcomes but also setting the stage for the future of personalized medicine.

Autoimmune Diseases

One of the most significant breakthroughs has been the use of cytokine inhibitors to treat chronic autoimmune conditions like rheumatoid arthritis (RA), Crohn’s disease, and psoriasis. These drugs selectively block key immune system components, providing relief while reducing systemic immune suppression.
A landmark example is adalimumab (Humira), a monoclonal antibody that targets TNF-α, a pro-inflammatory cytokine. In the STAR clinical trial, 52.8% of RA patients treated with adalimumab plus conventional therapy achieved a clinical response (ACR20) at week 24, compared to only 34.9% in the placebo group. Importantly, improvements were also observed in higher response thresholds (ACR50 and ACR70), while safety outcomes were similar to placebo.
Similarly, biosimilar development has confirmed long-term safety and efficacy. For instance, the DE019 Phase III study demonstrated equivalent clinical outcomes between the adalimumab biosimilar SB5 and the reference drug, further expanding access to this life-changing therapy.

 Cancer Treatment

Biopharmaceuticals are also at the forefront of cancer care, especially through monoclonal antibodies and immunotherapies. These treatments are designed to target tumor-specific markers, minimizing damage to healthy cells, while some approaches actively stimulate the immune system to fight cancer.
For example, cancer vaccines are gaining traction. The experimental mRNA-4157/V940 vaccine developed by Moderna, in combination with pembrolizumab, reduced the risk of recurrence by 44% in high-risk melanoma patients compared to pembrolizumab alone, according to the KEYNOTE-942 Phase 2b trial.

Rare Genetic Disorders

Biopharmaceutical innovation is also transforming outcomes for patients with rare genetic disorders—conditions that historically had very limited treatment options. Modern approaches include:

  • • Enzyme Replacement Therapies (ERTs), which supply missing or deficient enzymes (e.g., in Gaucher disease or Fabry disease).
  • • Gene Therapies, which correct faulty genetic sequences at the source of disease.

A landmark case is onasemnogene abeparvovec (Zolgensma), a gene therapy for spinal muscular atrophy (SMA). In the pivotal Phase I trial published in the New England Journal of Medicine, infants treated with this one-time intravenous therapy showed dramatic motor improvements and a survival rate far higher than expected, offering what is effectively a potential cure for a condition that was once uniformly fatal.

COVID-19 Response

mRNA vaccines, developed by Moderna and Pfizer-BioNTech, showcased the speed and precision of biopharmaceuticals in global health emergencies. These vaccines use tiny particles to send genetic code into cells. This triggers a protective immune response. This is a big step forward in drug delivery and preventing infectious diseases.

Challenges in Biopharmaceuticals Production

Despite the medical benefits, producing biopharmaceuticals remains a highly complex task that involves both technical and regulatory challenges.

• Upstream Processing

This stage involves cultivating living cells (mostly CHO cells) in bioreactors. Factors like oxygen levels, pH, and nutrient supply must be meticulously controlled to ensure consistent protein expression. Scale-up from lab to commercial volumes introduces risks related to cell growth variability andcontamination.

•  Downstream Processing

After protein production, the biologic must be separated and purified. Techniques like ultrafiltration, chromatography, and viral inactivation are used to achieve high purity levels. Any inconsistency during downstream processing may compromise drug quality and patient safety.

• Regulatory Approval

Unlike small molecules, biologics require extensive safety and efficacy validation due to their complexity. FDA approval involves multiple clinical trials and adherence to GMP compliance. Similarly, the European Medicines Agency (EMA) evaluates biologics through centralized procedures, ensuring uniform approval across EU member states. Regulatory agencies worldwide require detailed data on manufacturing, quality control, and product stability, leading to longer development timelines.

Disadvantages of Biopharmaceuticals

Biopharmaceuticals provide advanced treatment options, but they present notable disadvantages that affect accessibility, scalability, and patient outcomes. Their complexity translates into higher costs, longer development timelines, potential safety concerns, and distribution challenges compared to traditional small-molecule drugs.

1. High Production Costs.

Biologics are expensive to manufacture due to cell-based production systems, stringent quality controls, and high batch failure rates. According to BioProcess International, production costs can elevated manufacturing costs of biologics directly translate into high treatment prices, which can place a heavy strain on health system budgets.

2. Long Development Cycles.

Developing a biopharmaceutical typically takes 10 to 15 years. This extended timeline stems from complex clinical trials, regulatory hurdles, and the need for large-scale biologic validation (Source: Nature Reviews Drug Discovery).

3. Immunogenicity Risks.

Biologics can trigger immune responses in patients. Anti-drug antibodies (ADAs) may neutralize therapeutic effects or cause serious side effects. A study in Frontiers in Immunology highlights that immunogenicity remains a key concern for monoclonal antibody therapies.

4. Cold-Chain Dependency.

Most biologics require storage and transportation at controlled temperatures—typically between 2 °C and 8 °C—to maintain their stability and efficacy. This cold-chain dependency significantly increases distribution costs and presents logistical challenges, particularly in low- and middle-income countries where consistent refrigeration is not always feasible. According to recommendations from the World Health Organization (WHO), addressing cold-chain limitations is essential to ensuring equitable global access to life-saving biopharmaceuticals.

Advancements and Innovation in Biopharma Technology

To address production challenges, improve efficiency, and reduce overall costs, biopharmaceutical companies are embracing a new era of technological advancement. From single-use systems to AI-powered automation, innovation is driving a fundamental transformation in how medicines are developed, manufactured, and delivered.
At the core of this transformation is innovation management, the structured planning, development, and implementation of new ideas, processes, and products. In the biopharma and healthcare sectors, effective innovation management ensures that cutting-edge technologies are translated into real-world solutions that are both scalable and impactful. This strategic approach not only supports technological adoption but also fosters collaboration among academia, industry, and regulatory stakeholders.
By fostering an innovation-friendly ecosystem, companies can improve patient outcomes, streamline clinical and production workflows, and achieve sustainable growth.

Single-Use Technologies

Disposable bioreactors and tubing systems significantly cut down on cleaning times, lower contamination risks, and minimize downtime between production batches. These single-use components also contribute to sustainability by reducing water and energy consumption, making biomanufacturing greener and more efficient.

Continuous Manufacturing

Unlike traditional batch processing, continuous manufacturing operates nonstop, which enhances production speed and reduces variability. With integrated real-time monitoring, companies can maintain strict quality control standards while minimizing raw material waste—leading to higher efficiency and cost savings.

Automation and AI Integration

The incorporation of automation, robotics, and artificial intelligence (AI) in biomanufacturing is transforming precision and reproducibility across processes. Real-time analytics and machine learning enable predictive maintenance, risk assessment, and rapid troubleshooting, optimizing overall production workflows.

Advanced Drug Delivery Systems

Cutting-edge delivery technologies such as lipid nanoparticles, microneedle patches, and hydrogel-based implants are revolutionizing how medications are administered. These innovations improve drug targeting and bioavailability, ensuring effective treatment with fewer side effects.

Opportunities in Precision and Personalized Medicine

Biopharmaceuticals play a pivotal role in advancing precision and personalized medicine, which customizes treatments based on an individual patient’s genetic makeup and physiological profile.

Applications Include:

These applications demonstrate how precision and personalized medicine are transforming healthcare—making treatments more targeted, effective, and patient-centered:

• Personalized Oncology: Targeted cancer therapies designed around specific genetic mutations to improve treatment outcomes.
For example, In breast cancer, therapies like Trastuzumab (Herceptin®) target tumors with specific HER2 protein overexpression, improving treatment effectiveness. Recent studies show patients who receive therapies matched to their tumor’s genetic mutations have significantly better survival outcomes. (Sources: Precision or Personalized Medicine, Personalized Cancer Therapy Improves Outcomes in Advanced Disease, Says Study).

• Customized Biologics: Tailoring biologic drugs using pharmacokinetic data to optimize dosing for each patient.
Pharmacokinetic profiling for biologic drugs such as monoclonal antibodies (e.g., infliximab, used in rheumatoid arthritis) enables clinicians to tailor dosing for each patient based on how quickly each individual metabolizes the drug, minimizing side effects and maximizing efficacy. Ongoing clinical trials use personalized monitoring and AI to continuously adjust biologic therapy for best individual responses. (Sources: Enabling technologies for personalized and precision medicine).

• Data-Driven Therapy Selection: Leveraging metabolomic and genomic information to guide the most effective treatment choices. Metabolomic and genomic data are integrated to select therapies for non–small-cell lung cancer (NSCLC). For instance, genetic testing for mutations in the EGFR or ALK genes allows optimal selection of targeted drugs, which have higher success rates and fewer adverse effects than standard chemotherapy. However, studies show only about 65-75% of patients with actionable mutations currently access these treatments, highlighting the need for broader implementation. (Sources: Advances in personalized medicine).

What is precision and personalized medicine?

Precision and personalized medicine are innovative approaches to healthcare that use detailed information—such as a person’s genetic makeup, environment, and lifestyle—to guide prevention, diagnosis, and treatment of diseases. Unlike the traditional “one-size-fits-all” model, these approaches allow doctors to predict which treatments and strategies will be most effective for specific groups or individual patients.

Precision medicine often refers to grouping patients based on molecular profiles (like genomics or metabolomics) and targeting treatments to those most likely to benefit. For example, in cancer treatment, specific therapies are tailored to the genetic mutations found in a patient’s tumor.
Personalized medicine is used interchangeably with precision medicine, but sometimes emphasizes broader individual factors, including environmental and lifestyle data, to ensure that prevention and therapy are optimally suited for each person.
Both are transforming healthcare by increasing the accuracy and effectiveness of medical care, reducing trial-and-error treatments, and moving toward patient-centered solutions.

Opportunities in Rare and Orphan Diseases

Biopharmaceuticals are transforming the treatment landscape for rare and orphan diseases—conditions often neglected by traditional drug development due to small patient populations and high research costs. These innovative therapies focus on addressing unmet medical needs and improving quality of life for patients with limited options.

What is a rare disease?

A rare disease is a condition that affects a small percentage of the population—commonly defined in Europe as affecting fewer than 1 in 2,000 people, and in the United States as fewer than 200,000 individuals. There are over 7,000–10,000 rare diseases known worldwide, from genetic conditions to rare cancers and infections.

What is an orphan disease?

An orphan disease is typically a rare disease for which little or no treatment exists. Because so few people are affected, pharmaceutical companies and researchers have historically lacked incentives to develop therapies, leaving these diseases “orphaned” by the industry.

How common are rare and orphan diseases?

While each rare disease affects very few people, together they impact about 300 million people worldwide—nearly 4% of the global population. In some regions like Quebec, hundreds of thousands are affected.

The applications and use case of biosimilars for rare and orphan diseases

Biosimilars are increasingly important in treating rare and orphan diseases, offering affordable alternatives to original biopharmaceuticals and enabling wider patient access. Their introduction helps break down barriers to innovative therapies, making disease-modifying treatments more available for underserved patient populations:

1. Targeting Molecular Causes

Unlike conventional treatments that primarily manage symptoms, biopharmaceuticals aim to tackle the underlying molecular or genetic causes of rare diseases. This approach enables more effective, long-lasting therapies that can halt or reverse disease progression.

2. Regulatory Incentives and Fast-Track Approvals

The biopharma industry benefits from orphan drug incentives offered by regulatory agencies worldwide. These include market exclusivity, tax credits, and accelerated approval pathways, which encourage investment and innovation in rare disease treatments.

3. Collaboration and Patient Advocacy

Strong partnerships between regulatory bodies, biotech companies, and patient advocacy groups are crucial to advancing orphan drug development. These collaborative efforts improve clinical trial designs, facilitate patient recruitment, and expand access to life-changing therapies.

Why Are Biopharmaceuticals in High Demand?

Biopharmaceuticals are experiencing unprecedented demand driven by increasing rates of chronic diseases, a growing need for targeted and personalized therapies, and significant investment in biotechnology innovation. Their unique ability to provide precise, patient-specific treatments fuels both market growth and continuous advancements in the healthcare industry.
Funding in Europe is robust, with over €5 billion invested in early-stage biotech startups in 2024, supporting fast-track approvals and the integration of precision medicine for chronic and rare disease populations.

Conclusion: The Future of Biopharmaceuticals

Biopharmaceuticals are fundamentally reshaping modern healthcare by delivering disease-targeted, personalized solutions that improve patient outcomes. While challenges remain in manufacturing and regulatory compliance, emerging technologies such as gene editing, continuous manufacturing processes, and advanced drug delivery systems are making therapies more scalable and accessible. The future of medicine will increasingly depend on biopharmaceutical innovation to address unmet medical needs and expand global access to life-changing treatments.

 

 

 

Scroll up
Our site uses cookies to collect information about your device and browsing activity. We use this data to improve the site, ensure security and deliver personalized content. You can manage your cookie preferences by clicking here.
Accept cookies Configure Decline cookies
Basic cookie information
This website uses cookies and/or similar technologies that store and retrieve information when you browse. In general, these technologies can serve very different purposes, such as, for example, recognizing you as a user, obtaining information about your browsing habits or personalizing the way in which the content is displayed. The specific uses we make of these technologies are described below. By default, all cookies are disabled, except for technical ones, which are necessary for the website to function. If you wish to obtain more information or exercise your data protection rights, you can consult our "Política de cookies".
Accept cookies Configure
Technical cookies needed Always active
Technical cookies are strictly necessary for our website to work and for you to navigate through it. These types of cookies are those that, for example, allow us to identify you, give you access to certain restricted parts of the page if necessary, or remember different options or services already selected by you, such as your privacy preferences. Therefore, they are activated by default, your authorization is not necessary. Through the configuration of your browser, you can block or alert the presence of this type of cookies, although such blocking will affect the proper functioning of the different functionalities of our website.
Analysis cookies
Analysis cookies are those used to carry out anonymous analysis of the behavior of web users and allow measuring user activity and creating navigation profiles in order to improve the websites.
Confirm preferences
We want everyone to have access to our medicines wherever they are, regardless of prevalence.
OUR PRODUCTS