Monoclonal Antibody Conjugation Technologies in 2025: Unleashing Precision Therapies and Market Expansion. Explore the Innovations, Key Players, and Future Trends Shaping the Next Era of Targeted Biologics.
- Executive Summary: 2025 Market Landscape and Key Insights
- Market Size, Growth Rate, and 2025–2030 Forecasts
- Core Technologies: Linker Chemistry, Payloads, and Conjugation Methods
- Emerging Applications: Oncology, Autoimmune, and Infectious Diseases
- Competitive Analysis: Leading Companies and Strategic Partnerships
- Regulatory Environment and Quality Standards (FDA, EMA, ICH)
- Manufacturing Advances: Scalability, Automation, and CMC Challenges
- Innovation Pipeline: Next-Gen Antibody-Drug Conjugates (ADCs) and Bispecifics
- Regional Trends: North America, Europe, Asia-Pacific, and Rest of World
- Future Outlook: Investment Hotspots, Unmet Needs, and Disruptive Technologies
- Sources & References
Executive Summary: 2025 Market Landscape and Key Insights
The monoclonal antibody (mAb) conjugation technologies sector is poised for significant growth and innovation in 2025, driven by the expanding clinical and commercial success of antibody-drug conjugates (ADCs), radioimmunoconjugates, and antibody-enzyme conjugates. These technologies enable the precise attachment of cytotoxic drugs, imaging agents, or enzymes to monoclonal antibodies, enhancing their therapeutic index and specificity. The market landscape in 2025 is characterized by a robust pipeline of next-generation conjugates, increased investment in site-specific conjugation methods, and a focus on improving manufacturing scalability and product homogeneity.
Key industry leaders such as Genentech (a member of the Roche Group), Pfizer, and Astellas Pharma are advancing proprietary conjugation platforms, with several late-stage ADCs and novel conjugates expected to reach regulatory milestones in 2025. Seagen (now part of Pfizer) continues to be a pioneer in ADC technology, leveraging its linker and payload innovations to expand its clinical portfolio. Meanwhile, Lonza and Catalent are strengthening their positions as leading contract development and manufacturing organizations (CDMOs), offering integrated conjugation services and scalable GMP manufacturing solutions to biopharma partners.
Recent years have seen a shift toward site-specific conjugation techniques, such as enzymatic and engineered cysteine approaches, which improve the consistency and safety profile of antibody conjugates. Companies like Sutro Biopharma and Ambrx are at the forefront of these innovations, utilizing cell-free protein synthesis and expanded genetic code technologies to enable precise payload attachment. These advances are expected to accelerate the development of next-generation ADCs with improved therapeutic windows and reduced off-target toxicity.
The regulatory environment in 2025 is increasingly supportive, with agencies such as the FDA and EMA providing clearer guidance on the characterization and quality control of antibody conjugates. This is facilitating faster clinical translation and market entry for novel conjugates. Additionally, the growing adoption of digital and automated process analytics is enhancing manufacturing efficiency and product quality, as seen in the offerings from Sartorius and Merck KGaA.
Looking ahead, the monoclonal antibody conjugation technologies market is expected to see continued expansion, with increasing numbers of approved ADCs, broader applications in oncology and beyond, and ongoing partnerships between biopharma innovators and CDMOs. The focus on precision, scalability, and regulatory compliance will remain central to the sector’s evolution through 2025 and the following years.
Market Size, Growth Rate, and 2025–2030 Forecasts
The global market for monoclonal antibody (mAb) conjugation technologies is experiencing robust growth, driven by the expanding pipeline of antibody-drug conjugates (ADCs), increasing approvals of targeted biologics, and ongoing innovation in site-specific conjugation methods. As of 2025, the market is estimated to be valued at approximately $3.5–4.0 billion, with a compound annual growth rate (CAGR) projected between 12% and 15% through 2030. This growth is underpinned by the rising demand for next-generation therapeutics in oncology, autoimmune diseases, and infectious diseases, as well as the need for improved drug delivery and reduced off-target toxicity.
Key industry players such as Genentech (a member of the Roche Group), Pfizer, Seagen (now part of Pfizer), Astellas Pharma, and Amgen are actively investing in the development and commercialization of advanced conjugation platforms. These companies are leveraging proprietary linker chemistries, site-specific conjugation techniques, and scalable manufacturing processes to enhance the efficacy and safety profiles of their ADC pipelines. For example, Seagen has pioneered the use of cleavable and non-cleavable linkers, which have become industry standards for ADC stability and controlled payload release.
The market outlook for 2025–2030 is shaped by several factors:
- Pipeline Expansion: Over 100 ADCs and other mAb conjugates are in clinical development globally, with a significant number expected to reach late-stage trials or receive regulatory approval by 2030. This is expected to drive demand for conjugation technologies and contract manufacturing services.
- Technological Advancements: Innovations such as enzymatic conjugation, site-specific modification, and novel payload-linker combinations are being adopted by leading manufacturers, including Lonza and Catalent, to improve product consistency and therapeutic index.
- Manufacturing Scale-Up: The need for large-scale, GMP-compliant production is prompting investments in dedicated ADC manufacturing facilities by companies like Lonza and Samsung Biologics.
- Regulatory Momentum: Recent approvals of ADCs by regulatory agencies in the US, EU, and Asia are accelerating market adoption and encouraging further R&D investment.
Looking ahead, the monoclonal antibody conjugation technologies market is expected to surpass $7 billion by 2030, with oncology remaining the dominant therapeutic area. However, expansion into non-oncological indications and the emergence of novel conjugate formats (e.g., bispecifics, radioimmunoconjugates) are anticipated to further diversify the market landscape. Strategic collaborations between biopharma innovators and specialized CDMOs will likely play a pivotal role in meeting the growing demand for high-quality, scalable conjugation solutions.
Core Technologies: Linker Chemistry, Payloads, and Conjugation Methods
Monoclonal antibody conjugation technologies are at the heart of next-generation targeted therapeutics, particularly antibody-drug conjugates (ADCs). As of 2025, the field is characterized by rapid innovation in three core areas: linker chemistry, payload selection, and conjugation methods. These advances are driven by the need for improved efficacy, safety, and manufacturability of ADCs and related bioconjugates.
Linker Chemistry: The stability and release profile of the linker connecting the antibody to its payload are critical for therapeutic performance. In 2025, cleavable linkers—such as hydrazone, disulfide, and peptide-based linkers—remain widely used, enabling payload release in response to specific intracellular conditions. However, non-cleavable linkers, which require complete antibody degradation for payload release, are gaining traction for their enhanced plasma stability and reduced off-target toxicity. Companies like Genentech and Seagen are at the forefront, developing proprietary linker technologies that balance stability in circulation with efficient intracellular release. Additionally, site-specific conjugation strategies are being integrated with novel linkers to further improve homogeneity and pharmacokinetics.
Payloads: The choice of cytotoxic payload is expanding beyond traditional microtubule inhibitors (e.g., auristatins, maytansinoids) to include DNA-damaging agents, topoisomerase inhibitors, and even immune-modulating molecules. Pfizer and Astellas Pharma are notable for advancing ADCs with novel payloads, aiming to overcome resistance mechanisms and broaden the therapeutic window. The next few years are expected to see further diversification, including the use of protein degraders and RNA-based payloads, as companies seek to address previously undruggable targets.
Conjugation Methods: Traditional random conjugation methods, such as lysine or cysteine modification, often result in heterogeneous products with variable drug-to-antibody ratios (DAR). In 2025, site-specific conjugation technologies are increasingly adopted to produce more uniform ADCs. Amgen and Roche are investing in enzymatic and engineered amino acid approaches, such as transglutaminase-mediated conjugation and incorporation of non-natural amino acids, to achieve precise payload attachment. These methods improve batch-to-batch consistency and therapeutic index, and are expected to become industry standards in the coming years.
Looking ahead, the convergence of advanced linker chemistries, novel payloads, and precise conjugation methods is poised to accelerate the development of safer and more effective monoclonal antibody conjugates. Industry leaders are collaborating with technology providers and contract manufacturers to scale up these innovations, setting the stage for a new wave of targeted therapies by the late 2020s.
Emerging Applications: Oncology, Autoimmune, and Infectious Diseases
Monoclonal antibody (mAb) conjugation technologies are rapidly advancing, driving innovation in the treatment of oncology, autoimmune, and infectious diseases. These technologies enable the precise attachment of cytotoxic drugs, radionuclides, or imaging agents to antibodies, enhancing their therapeutic efficacy and specificity. In 2025 and the coming years, the field is witnessing a surge in next-generation antibody-drug conjugates (ADCs), bispecific antibodies, and novel linker chemistries, with a strong focus on improving safety profiles and expanding clinical applications.
In oncology, ADCs remain at the forefront of monoclonal antibody conjugation applications. Companies such as Genentech (a member of the Roche Group) and Seagen are leading the development and commercialization of ADCs for various cancers. Seagen’s ADC technology, which utilizes proprietary linker and cytotoxic payload systems, has resulted in multiple FDA-approved therapies, including those for breast and urothelial cancers. The company continues to expand its pipeline, with several candidates in late-stage clinical trials targeting solid tumors and hematologic malignancies. Similarly, Astellas Pharma is advancing its ADC portfolio, focusing on novel payloads and site-specific conjugation to improve therapeutic indices.
Autoimmune diseases are emerging as a significant area for mAb conjugation technologies. AbbVie and Janssen Pharmaceuticals are exploring antibody conjugates that deliver immunomodulatory agents directly to affected tissues, aiming to reduce systemic toxicity and enhance efficacy. These approaches are in early clinical development, with expectations for proof-of-concept data in the next few years. The use of antibody-cytokine conjugates and targeted delivery of small molecules is anticipated to broaden the therapeutic landscape for conditions such as rheumatoid arthritis and inflammatory bowel disease.
Infectious diseases represent a growing frontier for mAb conjugation. GSK and Sanofi are investing in antibody-antibiotic conjugates and antibody-antiviral conjugates to address antimicrobial resistance and emerging viral threats. These conjugates are designed to deliver potent agents directly to pathogens or infected cells, minimizing off-target effects. Early-stage clinical trials are underway, with initial results expected to inform further development strategies by 2026.
Looking ahead, the outlook for monoclonal antibody conjugation technologies is robust. Advances in site-specific conjugation, novel payloads, and improved linker stability are expected to yield safer and more effective therapeutics. The integration of artificial intelligence and high-throughput screening is accelerating candidate selection and optimization. As more products enter late-stage development and regulatory review, the next few years are poised to see a significant expansion of approved therapies across oncology, autoimmune, and infectious disease indications, solidifying the role of mAb conjugation as a cornerstone of precision medicine.
Competitive Analysis: Leading Companies and Strategic Partnerships
The monoclonal antibody (mAb) conjugation technologies sector is witnessing rapid evolution, driven by the demand for more precise, potent, and safer antibody-drug conjugates (ADCs) and related bioconjugates. As of 2025, the competitive landscape is shaped by a mix of established pharmaceutical giants, specialized biotechnology firms, and strategic collaborations aimed at advancing next-generation conjugation platforms.
Among the global leaders, F. Hoffmann-La Roche Ltd continues to leverage its deep expertise in oncology and biologics, with a robust pipeline of ADCs and investments in site-specific conjugation technologies. Roche’s focus on improving linker stability and payload diversity has positioned it at the forefront of clinical-stage ADC development. Similarly, Pfizer Inc. maintains a strong presence, particularly following its acquisition of Seagen, a pioneer in ADC technology, which has expanded Pfizer’s capabilities in both cytotoxic payloads and novel linker chemistries.
Japanese pharmaceutical leader Takeda Pharmaceutical Company Limited is actively advancing its proprietary conjugation platforms, emphasizing improved therapeutic index and manufacturability. Takeda’s partnerships with technology innovators have enabled access to novel payloads and site-specific conjugation methods, supporting its expanding oncology portfolio.
Specialized biotech firms are also critical drivers of innovation. Sutro Biopharma, Inc. is recognized for its cell-free protein synthesis and precise conjugation technologies, enabling the development of homogeneous ADCs with defined drug-to-antibody ratios. ADC Therapeutics SA focuses on pyrrolobenzodiazepine (PBD)-based payloads and has established multiple partnerships to accelerate clinical development and commercialization.
Strategic alliances are a hallmark of the sector’s competitive dynamics. For example, Genmab A/S and Seagen Inc. (now part of Pfizer) have collaborated to combine Genmab’s antibody engineering with Seagen’s linker-payload expertise, resulting in several promising ADC candidates. Lonza Group AG, a leading contract development and manufacturing organization (CDMO), has formed partnerships with both large pharma and emerging biotechs to provide scalable, GMP-compliant conjugation services, reflecting the growing trend toward outsourcing complex biomanufacturing.
Looking ahead, the next few years are expected to see intensified competition as companies race to commercialize ADCs with novel payloads, improved safety profiles, and expanded indications beyond oncology. The convergence of advanced conjugation chemistries, automation, and AI-driven design is likely to further differentiate market leaders. Strategic partnerships—particularly those combining proprietary antibody platforms with innovative conjugation and payload technologies—will remain central to maintaining competitive advantage in this dynamic field.
Regulatory Environment and Quality Standards (FDA, EMA, ICH)
The regulatory environment for monoclonal antibody (mAb) conjugation technologies is rapidly evolving, reflecting the increasing complexity and clinical significance of antibody-drug conjugates (ADCs) and related bioconjugates. In 2025, both the U.S. Food and Drug Administration (FDA) and the European Medicines Agency (EMA) continue to refine their frameworks to address the unique challenges posed by these advanced therapeutics. The International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH) also plays a pivotal role in harmonizing global standards.
The FDA maintains a risk-based approach to the regulation of mAb conjugates, emphasizing the need for robust characterization of both the antibody and the conjugated payload, as well as the linker chemistry. The agency’s guidance documents, such as those on the development of ADCs, stress the importance of demonstrating product consistency, stability, and safety throughout the manufacturing process. In 2024 and 2025, the FDA has increased its scrutiny of site-specific conjugation methods and the control of drug-to-antibody ratio (DAR), which are critical for product efficacy and safety. The agency also requires detailed analytical data on process-related impurities and potential immunogenicity, reflecting the growing complexity of next-generation ADCs.
The EMA, similarly, has updated its guidelines to address the specificities of mAb conjugates, particularly in the context of quality by design (QbD) and lifecycle management. The agency emphasizes the need for comprehensive comparability studies when changes are made to conjugation processes or manufacturing sites. In 2025, the EMA is expected to further align its requirements with those of the FDA, particularly regarding the control of critical quality attributes (CQAs) such as conjugation site heterogeneity and payload distribution.
The ICH, through its Q5E and Q6B guidelines, provides a harmonized framework for the characterization and quality control of biotechnological products, including mAb conjugates. Ongoing discussions within ICH working groups in 2025 are focused on updating guidance to reflect advances in site-specific conjugation technologies and analytical methods. These updates aim to facilitate global regulatory convergence and streamline the approval process for innovative ADCs.
Major industry players such as Roche, Pfizer, and Astellas Pharma are actively engaging with regulators to shape the evolving standards, leveraging their experience in bringing multiple ADCs to market. These companies are also investing in advanced analytical platforms and digital quality management systems to ensure compliance with the latest regulatory expectations.
Looking ahead, the regulatory landscape for mAb conjugation technologies in 2025 and beyond will likely see increased emphasis on real-time release testing, digitalization of quality systems, and the integration of advanced analytics to support continuous manufacturing and lifecycle management. This dynamic environment underscores the need for ongoing collaboration between industry, regulators, and standard-setting bodies to ensure the safe and effective development of next-generation antibody conjugates.
Manufacturing Advances: Scalability, Automation, and CMC Challenges
Monoclonal antibody (mAb) conjugation technologies are undergoing significant transformation as the biopharmaceutical industry seeks to address the growing demand for antibody-drug conjugates (ADCs) and other next-generation therapeutics. In 2025 and the coming years, manufacturing advances are focused on improving scalability, automation, and Chemistry, Manufacturing, and Controls (CMC) processes to support robust, reproducible, and cost-effective production.
A key trend is the shift from traditional batch processes to continuous and semi-continuous manufacturing platforms. This transition is driven by the need for higher throughput and consistent product quality, especially as more ADCs progress through clinical pipelines. Companies such as Lonza and Catalent are investing in modular, flexible manufacturing suites that can accommodate a range of conjugation chemistries and payloads. These facilities are designed to support both clinical and commercial-scale production, with integrated automation to minimize manual interventions and reduce the risk of contamination.
Automation is increasingly being adopted across conjugation workflows, from payload attachment to purification and fill-finish operations. Advanced process analytical technologies (PAT) and real-time monitoring systems are being implemented to ensure critical quality attributes (CQAs) are maintained throughout the manufacturing process. Sartorius and Merck KGaA are notable for their development of automated systems and single-use technologies that streamline conjugation steps and facilitate rapid changeovers between products.
Scalability remains a central challenge, particularly as novel conjugation chemistries—such as site-specific and enzymatic approaches—are adopted to improve product homogeneity and therapeutic index. These methods often require specialized reagents and process conditions, necessitating close collaboration between technology providers and contract development and manufacturing organizations (CDMOs). Abzena and Pfizer are among the companies advancing scalable site-specific conjugation platforms, with a focus on ensuring compatibility with large-scale bioreactor and downstream processing systems.
CMC challenges are also evolving, as regulatory expectations for ADCs and other mAb conjugates become more stringent. There is a growing emphasis on comprehensive characterization of conjugation sites, drug-to-antibody ratio (DAR), and impurity profiles. Industry leaders are investing in high-resolution analytical tools and digital data management systems to support regulatory submissions and lifecycle management. The outlook for 2025 and beyond suggests continued innovation in manufacturing technologies, with a focus on digitalization, modularity, and end-to-end integration to meet the needs of a rapidly expanding mAb conjugate market.
Innovation Pipeline: Next-Gen Antibody-Drug Conjugates (ADCs) and Bispecifics
Monoclonal antibody conjugation technologies are at the heart of the innovation pipeline for next-generation antibody-drug conjugates (ADCs) and bispecific antibodies, with 2025 poised to see significant advances in both precision and scalability. The core challenge in ADC development remains the creation of stable, site-specific linkages between monoclonal antibodies and cytotoxic payloads, ensuring optimal therapeutic index and minimizing off-target effects. Recent years have seen a shift from traditional random conjugation methods to more controlled, site-specific approaches, and this trend is accelerating as new candidates enter clinical pipelines.
One of the most prominent players, Genentech (a member of the Roche Group), continues to refine its THIOMAB™ technology, which enables precise engineering of cysteine residues for site-specific conjugation. This platform underpins several late-stage ADCs and is being adapted for next-generation payloads and bispecific formats. Similarly, Seagen (formerly Seattle Genetics), a pioneer in ADCs, is advancing its proprietary linker and conjugation technologies, including the use of engineered cysteines and novel cleavable linkers, to improve payload delivery and reduce systemic toxicity.
In 2025, the field is also witnessing the emergence of enzymatic conjugation methods, such as those developed by Sutro Biopharma, which leverages cell-free protein synthesis to incorporate non-natural amino acids at defined positions, enabling highly uniform ADCs. This approach is gaining traction for its scalability and ability to support complex bispecific and multi-specific antibody formats. Lonza, a leading contract development and manufacturing organization (CDMO), is investing in next-generation conjugation platforms, including site-specific enzymatic and click chemistry-based methods, to support the growing demand for clinical and commercial ADC manufacturing.
Another notable advance is the use of glycan engineering, as seen in the work of Samsung Biologics and Catalent, which allows for conjugation at defined glycosylation sites, further enhancing homogeneity and pharmacokinetics. These technologies are being rapidly adopted as the industry moves toward more complex ADCs and bispecifics, with several candidates expected to enter pivotal trials in 2025 and beyond.
Looking ahead, the outlook for monoclonal antibody conjugation technologies is robust, with ongoing investments in automation, high-throughput screening, and analytical characterization. As regulatory expectations for product consistency and safety rise, the adoption of site-specific and modular conjugation platforms is expected to become standard practice. The convergence of these innovations is set to accelerate the development of safer, more effective ADCs and bispecifics, expanding their therapeutic reach across oncology and beyond.
Regional Trends: North America, Europe, Asia-Pacific, and Rest of World
The global landscape for monoclonal antibody (mAb) conjugation technologies is marked by dynamic regional trends, with North America, Europe, Asia-Pacific, and the Rest of World (RoW) each contributing distinctively to innovation, manufacturing, and market adoption. As of 2025 and looking ahead, these regions are shaping the future of antibody-drug conjugates (ADCs), radioimmunoconjugates, and other mAb-based therapeutics through investments in R&D, regulatory evolution, and strategic partnerships.
- North America remains the epicenter of mAb conjugation technology development, driven by a robust biopharmaceutical ecosystem and a concentration of leading companies. The United States, in particular, is home to pioneers such as Genentech (a member of the Roche Group), which continues to advance ADCs and next-generation conjugation chemistries. Seagen (formerly Seattle Genetics) is another key player, with multiple FDA-approved ADCs and ongoing pipeline expansion. The region benefits from strong regulatory support and a mature clinical trial infrastructure, fostering rapid translation of novel conjugation technologies into clinical candidates.
- Europe is characterized by a collaborative research environment and a growing number of biotech firms specializing in site-specific conjugation and linker technologies. Companies such as Sphere Fluidics and Lonza are actively involved in developing scalable manufacturing solutions and innovative payload-linker systems. The European Medicines Agency (EMA) is increasingly harmonizing regulatory pathways for complex biologics, which is expected to accelerate approvals and market entry for new mAb conjugates in the coming years.
- Asia-Pacific is emerging as a significant hub for both contract development and manufacturing (CDMO) and original innovation. China, in particular, has seen rapid growth in mAb conjugation capabilities, with companies like WuXi AppTec and Biocytogen investing in state-of-the-art conjugation platforms and expanding their global reach. Japan and South Korea are also increasing their presence, supported by government initiatives and partnerships with Western firms. The region is expected to see double-digit growth in ADC clinical trials and manufacturing capacity through 2025 and beyond.
- Rest of World (RoW) regions, including Latin America and the Middle East, are gradually entering the mAb conjugation space, primarily through technology transfer agreements and collaborations with established global players. While local innovation is still nascent, increasing demand for advanced oncology therapeutics is prompting investments in infrastructure and regulatory alignment.
Looking forward, regional specialization and cross-border collaborations are anticipated to intensify, with North America and Europe leading in innovation and regulatory frameworks, while Asia-Pacific rapidly scales up manufacturing and clinical development. This global interplay is expected to drive broader access to next-generation mAb conjugates and fuel continued growth in the sector through the late 2020s.
Future Outlook: Investment Hotspots, Unmet Needs, and Disruptive Technologies
Monoclonal antibody (mAb) conjugation technologies are poised for significant evolution in 2025 and the coming years, driven by the need for more precise, potent, and safer therapeutics. The sector is witnessing robust investment, particularly in next-generation antibody-drug conjugates (ADCs), site-specific conjugation methods, and novel linker chemistries. These advances are addressing longstanding challenges such as heterogeneous drug-to-antibody ratios, off-target toxicity, and limited payload diversity.
A major investment hotspot is the development of site-specific conjugation platforms, which enable uniform attachment of payloads to defined antibody sites, improving therapeutic index and manufacturing consistency. Companies like Genentech and Amgen are actively advancing engineered cysteine and non-natural amino acid technologies to achieve this precision. Meanwhile, Seagen (formerly Seattle Genetics) continues to lead in linker and payload innovation, with its proprietary technologies underpinning several approved and late-stage ADCs.
Another area of unmet need is the expansion of payload classes beyond traditional cytotoxics. The next wave of conjugates is expected to incorporate immune modulators, protein degraders, and oligonucleotides, broadening the therapeutic scope. Roche and Astellas Pharma are investing in these modalities, aiming to address cancers with limited treatment options and to reduce systemic toxicity.
Disruptive technologies are also emerging in the form of bioconjugation automation and continuous manufacturing, which promise to streamline production and reduce costs. Lonza, a leading contract manufacturer, is scaling up integrated solutions for ADC production, including high-throughput conjugation and purification systems. This is expected to accelerate the translation of novel conjugates from bench to clinic.
Looking ahead, regulatory agencies are signaling support for innovative conjugation approaches, provided they demonstrate clear safety and efficacy benefits. The U.S. FDA’s recent approvals of next-generation ADCs underscore the agency’s openness to novel chemistries and manufacturing paradigms. As a result, the competitive landscape is intensifying, with established pharmaceutical companies and specialized biotech firms vying for leadership in this space.
In summary, the future of monoclonal antibody conjugation technologies will be shaped by investments in site-specific conjugation, diversification of payloads, and disruptive manufacturing solutions. These advances are expected to unlock new therapeutic opportunities, address unmet clinical needs, and drive the next phase of growth in the biopharmaceutical sector.
Sources & References
- Catalent
- Ambrx
- Sartorius
- Samsung Biologics
- Roche
- Janssen Pharmaceuticals
- GSK
- Takeda Pharmaceutical Company Limited
- Sutro Biopharma, Inc.
- Genmab A/S
- Abzena
- Sphere Fluidics
- WuXi AppTec