Cell & Gene Therapy Biomanufacturing CDMO Market by Service Type (Analytical Services, Clinical Manufacturing, Commercial Manufacturing), Therapy Type (Cell Therapy, Gene Therapy), Technology, Scale, Process Stage, Application, End User - Global Forecast
Description
The Cell & Gene Therapy Biomanufacturing CDMO Market was valued at USD 24.37 billion in 2024 and is projected to grow to USD 26.66 billion in 2025, with a CAGR of 9.45%, reaching USD 50.21 billion by 2032.
A concise orientation on how technical complexity, regulatory rigor, and commercial imperatives are reshaping third party manufacturing strategies for advanced therapies
The cell and gene therapy biomanufacturing contract development and manufacturing organization environment is undergoing rapid maturation driven by scientific progress, evolving regulatory expectations, and a growing commercial pipeline that demands scalable, high-quality manufacturing pathways. This introduction frames the report’s focus on the operational, technological, and strategic facets that matter most to organizations evaluating third-party manufacturing partnerships. It captures how manufacturing complexity, from vector production to downstream purification, increasingly shapes development timelines and long-term commercialization plans.
Transitioning from laboratory demonstration to regulated production requires disciplined process development, analytical rigor, and robust quality systems. Consequently, sponsors and CDMOs are investing in modular facility designs, advanced analytics, and workforce competency programs to reduce risk and accelerate transfer. The result is a market where technical differentiation, regulatory compliance, and commercial agility determine partnership value. This section sets the stage for deeper analysis by outlining the critical intersections among service offerings, therapy modalities, and technology choices that underpin strategic sourcing decisions across the value chain.
How technological advances, commercial pressures, and strategic outsourcing are converging to redefine partnerships and capability priorities across advanced therapy manufacturing
The landscape is shifting in ways that are both transformative and enduring. Advances in vector engineering, improvements in nonviral delivery modalities, and the emergence of standardized analytical platforms have collectively reduced certain development uncertainties, enabling more reproducible manufacturing outcomes. At the same time, commercial pressures and capital discipline are prompting sponsors to seek flexible manufacturing partnerships that can accommodate both personalized and scale-up production models. As a result, CDMOs are evolving from purely transactional producers into strategic partners that provide integrated development services, regulatory support, and supply chain resilience.
Moreover, the industry’s organizational models are changing. Companies are reallocating internal resources to focus on core discovery capabilities while outsourcing process development, clinical production, and certain commercial-scale activities. This redistribution accelerates time to clinic and mitigates fixed-cost exposure, but it also heightens the importance of due diligence around technology compatibility and platform transferability. Consequently, strategic alliances, co-investment in shared infrastructure, and the adoption of digital manufacturing tools are becoming common, fostering a collaborative ecosystem where operational excellence and strategic alignment drive long-term competitive advantage.
Assessing how evolving tariff regimes and trade policy dynamics are prompting resilience investments and reshaping procurement and supply chain choices in biomanufacturing
Recent policy developments and trade measures in the United States present a material set of considerations for global supply chains and capacity planning within biomanufacturing. Increased tariffs and shifting import/export regimes can affect the cost base of critical raw materials, single-use systems, and certain consumables that traverse international logistics networks. Consequently, organizations must evaluate supplier dispersion, local sourcing opportunities, and inventory strategies to mitigate exposure to tariff-driven cost volatility.
Furthermore, these tariff dynamics interact with regulatory and operational timelines. Sponsors moving components or intermediates across borders for fill–finish or analytical testing can face extended lead times and higher landed costs, which in turn influence batch scheduling and capital utilization. In response, many stakeholders are assessing nearshoring options, dual-sourcing arrangements, and inventory debottlenecking to maintain continuity. Ultimately, the cumulative impact of tariff changes is less about instantaneous disruption and more about reshaping procurement strategies, channeling capital toward supply-chain resilience, and encouraging investments that reduce dependency on vulnerable nodes in the global materials network.
A structured segmentation perspective that clarifies capability alignment across services, therapy modalities, technology platforms, applications, scales, end users, and process stages
Meaningful segmentation reveals where capabilities and demand are concentrated across service types, therapy modalities, technology platforms, applications, scales, end users, and process stages, and it provides a framework to align commercial offers with sponsor needs. From the vantage of service type, providers that combine analytical services, clinical manufacturing, commercial manufacturing, and process development are better positioned to deliver end-to-end transferability and to reduce transactional friction. Sponsors often prioritize partners that can bridge early-stage development to clinical supply while scaling to commercial demands.
When viewed by therapy type, differentiators emerge between cell therapy and gene therapy programs, with cell therapy further split between allogeneic and autologous approaches that carry distinct manufacturing footprints and logistics requirements. Technology-based segmentation highlights divergent investments in non-viral vector approaches such as electroporation and lipid nanoparticles versus viral vector platforms including AAV, adenovirus, lentivirus, and retrovirus, each of which imposes unique process controls and facility design constraints. Application-focused segmentation across cardiovascular, neurology, oncology, and rare diseases underscores therapeutic-area specific assay needs and regulatory considerations, while scale distinctions among clinical, commercial, and preclinical activities clarify capacity and validation imperatives. In addition, end users ranging from biopharmaceutical companies to contract research organizations and research institutions bring differing procurement behaviors, timelines, and risk tolerances, and process-stage segmentation that separates upstream from downstream activities further refines operational priorities. Collectively, these lenses enable CDMOs and sponsors to align capabilities with strategic objectives and to prioritize investments that unlock the greatest operational leverage.
How regional regulatory nuance, investment patterns, and talent concentrations drive differentiated manufacturing strategies and influence global partnership footprints
Regional dynamics shape investment decisions, regulatory interactions, and partnership strategies across the global biomanufacturing landscape. In the Americas, strong developer ecosystems, concentrated capital pools, and established regulatory frameworks support a mature outsourcing environment that emphasizes scale-up and commercial readiness. Sponsors and providers in this region often prioritize capacity expansion and integration of quality systems that meet stringent regulatory expectations, which supports cross-border supply models and advanced analytics integration.
In contrast, Europe, Middle East & Africa exhibits a diverse regulatory tapestry and a mix of emerging centers of technical expertise. Collaborative consortia and public-private partnerships in parts of Europe drive innovation in manufacturing approaches and shared infrastructure models. Consequently, stakeholders must navigate heterogeneous approval pathways while leveraging localized talent and specialty service providers. Meanwhile, Asia-Pacific presents a rapidly evolving picture characterized by significant investment in manufacturing capacity, improving regulatory harmonization, and growing internal demand for advanced therapies. The region’s emphasis on cost-efficient production and shorter lead-time logistics is encouraging sponsors and CDMOs to evaluate regional manufacturing footprints, strategic alliances, and technology transfer models that balance speed to market with long-term supply security. Taken together, these regional insights inform how providers allocate capital, where sponsors seek redundancy, and how cross-regional partnerships can be structured for operational resilience.
Why specialization, platform integration, and operational excellence are the defining competitive levers that shape partnership selection and long-term supplier relationships
Competitive dynamics among leading firms emphasize specialization, platform breadth, and the capacity to co-develop scalable processes with sponsors. Key competitive moves include vertical integration of analytical and process development services, modularization of facility design to accelerate campaign changeover, and investment in automation and analytics that reduce variability and accelerate release testing. Some organizations differentiate through deep expertise in specific vector platforms or in autologous cell therapies, while others pursue a broader platform approach to capture a range of therapeutic programs.
Strategic partnerships and alliance structures are also prominent, enabling companies to share risk, co-invest in capacity, and accelerate technology transfer. Consequently, business models that combine laboratory-scale innovation with robust commercial-scale execution tend to command stronger strategic relationships with sponsors. Moreover, operational excellence in quality systems, regulatory support, and supply chain orchestration remains a consistent source of competitive advantage, influencing both client selection and long-term retention. For sponsors, selecting a partner with complementary capabilities and proven transfer pathways often proves more determinative of program success than selecting based solely on price or capacity alone.
Practical strategic imperatives for executives to fortify manufacturing resilience, accelerate transfers, and align partnerships for long term competitive advantage
Industry leaders should take deliberate, actionable steps to build resilience, strengthen strategic partnerships, and accelerate time to clinic and commercialization. First, prioritize investments in modular facility designs and platform-agnostic process capabilities that allow rapid reconfiguration for different vector types and therapy modalities. This approach reduces capital intensity per program and improves responsiveness to sponsor needs.
Second, expand analytical and quality-by-design capabilities to enable earlier identification of critical process parameters and to shorten qualification cycles. Strong analytical platforms reduce release uncertainty and accelerate regulatory interactions. Third, diversify supplier networks for critical inputs and consider nearshoring or dual-sourcing arrangements to mitigate tariff exposure and logistics risk. Fourth, cultivate long-term strategic alliances with sponsors that include knowledge-transfer arrangements, shared investment frameworks, and performance-based milestones, thereby aligning incentives across development and commercial phases. Finally, invest in workforce development programs that combine technical training with cross-functional process knowledge to sustain operational excellence as throughput increases. These actions together enhance competitiveness, reduce program risk, and create durable value for both providers and sponsors.
A transparent multi-source methodology combining expert interviews, technical literature, and regulatory analysis to validate operational trends and strategic implications
This research synthesizes primary interviews, technical literature, operational case studies, and regulatory guidance to develop a robust assessment of manufacturing trends and strategic priorities. Primary inputs include structured interviews with manufacturing leaders, quality and regulatory experts, and senior development strategists who provided qualitative perspectives on process transfer, facility design, and partnership models. These insights were triangulated with publicly available regulatory documents, peer-reviewed publications on vector and nonviral delivery technologies, and company disclosures that describe facility investments and capability roadmaps.
Analytical rigor was applied through thematic coding of qualitative data, cross-referencing of technology-specific process constraints, and validation of operational practices against current regulatory expectations. Wherever possible, the methodology prioritized corroborated technical details over speculative assertions, and it emphasized replicable process characteristics that inform manufacturing decisions. Limitations included variability in proprietary process information and the rapidly evolving nature of platform technologies, which the research addresses by highlighting observable trends and validated practices rather than speculative performance claims. This methodological approach yields pragmatic insights designed to inform strategy, sourcing decisions, and operational planning.
A succinct synthesis of how technical, regulatory, and supply chain priorities converge to determine competitive positioning and partnership success in advanced therapy manufacturing
In conclusion, the cell and gene therapy biomanufacturing CDMO environment is maturing into a landscape where technical differentiation, regulatory readiness, and supply chain resilience determine competitive positioning. Sponsors increasingly demand partners who can provide integrated development services, scalable manufacturing, and strong analytical capabilities that reduce transfer risk and accelerate program timelines. At the same time, evolving trade and procurement dynamics are prompting stakeholders to reassess sourcing strategies and to prioritize investments that reduce exposure to external disruptions.
Moving forward, organizations that successfully combine platform flexibility, operational excellence, and strategic partnerships will be best positioned to capture program flow and to support the growing complexity of advanced therapies. The emphasis should be on measurable process robustness, validated transferability, and the ability to adapt to therapy-specific and region-specific demands. These priorities will underpin successful development and commercialization pathways as the sector continues its evolution.
Note: PDF & Excel + Online Access - 1 Year
A concise orientation on how technical complexity, regulatory rigor, and commercial imperatives are reshaping third party manufacturing strategies for advanced therapies
The cell and gene therapy biomanufacturing contract development and manufacturing organization environment is undergoing rapid maturation driven by scientific progress, evolving regulatory expectations, and a growing commercial pipeline that demands scalable, high-quality manufacturing pathways. This introduction frames the report’s focus on the operational, technological, and strategic facets that matter most to organizations evaluating third-party manufacturing partnerships. It captures how manufacturing complexity, from vector production to downstream purification, increasingly shapes development timelines and long-term commercialization plans.
Transitioning from laboratory demonstration to regulated production requires disciplined process development, analytical rigor, and robust quality systems. Consequently, sponsors and CDMOs are investing in modular facility designs, advanced analytics, and workforce competency programs to reduce risk and accelerate transfer. The result is a market where technical differentiation, regulatory compliance, and commercial agility determine partnership value. This section sets the stage for deeper analysis by outlining the critical intersections among service offerings, therapy modalities, and technology choices that underpin strategic sourcing decisions across the value chain.
How technological advances, commercial pressures, and strategic outsourcing are converging to redefine partnerships and capability priorities across advanced therapy manufacturing
The landscape is shifting in ways that are both transformative and enduring. Advances in vector engineering, improvements in nonviral delivery modalities, and the emergence of standardized analytical platforms have collectively reduced certain development uncertainties, enabling more reproducible manufacturing outcomes. At the same time, commercial pressures and capital discipline are prompting sponsors to seek flexible manufacturing partnerships that can accommodate both personalized and scale-up production models. As a result, CDMOs are evolving from purely transactional producers into strategic partners that provide integrated development services, regulatory support, and supply chain resilience.
Moreover, the industry’s organizational models are changing. Companies are reallocating internal resources to focus on core discovery capabilities while outsourcing process development, clinical production, and certain commercial-scale activities. This redistribution accelerates time to clinic and mitigates fixed-cost exposure, but it also heightens the importance of due diligence around technology compatibility and platform transferability. Consequently, strategic alliances, co-investment in shared infrastructure, and the adoption of digital manufacturing tools are becoming common, fostering a collaborative ecosystem where operational excellence and strategic alignment drive long-term competitive advantage.
Assessing how evolving tariff regimes and trade policy dynamics are prompting resilience investments and reshaping procurement and supply chain choices in biomanufacturing
Recent policy developments and trade measures in the United States present a material set of considerations for global supply chains and capacity planning within biomanufacturing. Increased tariffs and shifting import/export regimes can affect the cost base of critical raw materials, single-use systems, and certain consumables that traverse international logistics networks. Consequently, organizations must evaluate supplier dispersion, local sourcing opportunities, and inventory strategies to mitigate exposure to tariff-driven cost volatility.
Furthermore, these tariff dynamics interact with regulatory and operational timelines. Sponsors moving components or intermediates across borders for fill–finish or analytical testing can face extended lead times and higher landed costs, which in turn influence batch scheduling and capital utilization. In response, many stakeholders are assessing nearshoring options, dual-sourcing arrangements, and inventory debottlenecking to maintain continuity. Ultimately, the cumulative impact of tariff changes is less about instantaneous disruption and more about reshaping procurement strategies, channeling capital toward supply-chain resilience, and encouraging investments that reduce dependency on vulnerable nodes in the global materials network.
A structured segmentation perspective that clarifies capability alignment across services, therapy modalities, technology platforms, applications, scales, end users, and process stages
Meaningful segmentation reveals where capabilities and demand are concentrated across service types, therapy modalities, technology platforms, applications, scales, end users, and process stages, and it provides a framework to align commercial offers with sponsor needs. From the vantage of service type, providers that combine analytical services, clinical manufacturing, commercial manufacturing, and process development are better positioned to deliver end-to-end transferability and to reduce transactional friction. Sponsors often prioritize partners that can bridge early-stage development to clinical supply while scaling to commercial demands.
When viewed by therapy type, differentiators emerge between cell therapy and gene therapy programs, with cell therapy further split between allogeneic and autologous approaches that carry distinct manufacturing footprints and logistics requirements. Technology-based segmentation highlights divergent investments in non-viral vector approaches such as electroporation and lipid nanoparticles versus viral vector platforms including AAV, adenovirus, lentivirus, and retrovirus, each of which imposes unique process controls and facility design constraints. Application-focused segmentation across cardiovascular, neurology, oncology, and rare diseases underscores therapeutic-area specific assay needs and regulatory considerations, while scale distinctions among clinical, commercial, and preclinical activities clarify capacity and validation imperatives. In addition, end users ranging from biopharmaceutical companies to contract research organizations and research institutions bring differing procurement behaviors, timelines, and risk tolerances, and process-stage segmentation that separates upstream from downstream activities further refines operational priorities. Collectively, these lenses enable CDMOs and sponsors to align capabilities with strategic objectives and to prioritize investments that unlock the greatest operational leverage.
How regional regulatory nuance, investment patterns, and talent concentrations drive differentiated manufacturing strategies and influence global partnership footprints
Regional dynamics shape investment decisions, regulatory interactions, and partnership strategies across the global biomanufacturing landscape. In the Americas, strong developer ecosystems, concentrated capital pools, and established regulatory frameworks support a mature outsourcing environment that emphasizes scale-up and commercial readiness. Sponsors and providers in this region often prioritize capacity expansion and integration of quality systems that meet stringent regulatory expectations, which supports cross-border supply models and advanced analytics integration.
In contrast, Europe, Middle East & Africa exhibits a diverse regulatory tapestry and a mix of emerging centers of technical expertise. Collaborative consortia and public-private partnerships in parts of Europe drive innovation in manufacturing approaches and shared infrastructure models. Consequently, stakeholders must navigate heterogeneous approval pathways while leveraging localized talent and specialty service providers. Meanwhile, Asia-Pacific presents a rapidly evolving picture characterized by significant investment in manufacturing capacity, improving regulatory harmonization, and growing internal demand for advanced therapies. The region’s emphasis on cost-efficient production and shorter lead-time logistics is encouraging sponsors and CDMOs to evaluate regional manufacturing footprints, strategic alliances, and technology transfer models that balance speed to market with long-term supply security. Taken together, these regional insights inform how providers allocate capital, where sponsors seek redundancy, and how cross-regional partnerships can be structured for operational resilience.
Why specialization, platform integration, and operational excellence are the defining competitive levers that shape partnership selection and long-term supplier relationships
Competitive dynamics among leading firms emphasize specialization, platform breadth, and the capacity to co-develop scalable processes with sponsors. Key competitive moves include vertical integration of analytical and process development services, modularization of facility design to accelerate campaign changeover, and investment in automation and analytics that reduce variability and accelerate release testing. Some organizations differentiate through deep expertise in specific vector platforms or in autologous cell therapies, while others pursue a broader platform approach to capture a range of therapeutic programs.
Strategic partnerships and alliance structures are also prominent, enabling companies to share risk, co-invest in capacity, and accelerate technology transfer. Consequently, business models that combine laboratory-scale innovation with robust commercial-scale execution tend to command stronger strategic relationships with sponsors. Moreover, operational excellence in quality systems, regulatory support, and supply chain orchestration remains a consistent source of competitive advantage, influencing both client selection and long-term retention. For sponsors, selecting a partner with complementary capabilities and proven transfer pathways often proves more determinative of program success than selecting based solely on price or capacity alone.
Practical strategic imperatives for executives to fortify manufacturing resilience, accelerate transfers, and align partnerships for long term competitive advantage
Industry leaders should take deliberate, actionable steps to build resilience, strengthen strategic partnerships, and accelerate time to clinic and commercialization. First, prioritize investments in modular facility designs and platform-agnostic process capabilities that allow rapid reconfiguration for different vector types and therapy modalities. This approach reduces capital intensity per program and improves responsiveness to sponsor needs.
Second, expand analytical and quality-by-design capabilities to enable earlier identification of critical process parameters and to shorten qualification cycles. Strong analytical platforms reduce release uncertainty and accelerate regulatory interactions. Third, diversify supplier networks for critical inputs and consider nearshoring or dual-sourcing arrangements to mitigate tariff exposure and logistics risk. Fourth, cultivate long-term strategic alliances with sponsors that include knowledge-transfer arrangements, shared investment frameworks, and performance-based milestones, thereby aligning incentives across development and commercial phases. Finally, invest in workforce development programs that combine technical training with cross-functional process knowledge to sustain operational excellence as throughput increases. These actions together enhance competitiveness, reduce program risk, and create durable value for both providers and sponsors.
A transparent multi-source methodology combining expert interviews, technical literature, and regulatory analysis to validate operational trends and strategic implications
This research synthesizes primary interviews, technical literature, operational case studies, and regulatory guidance to develop a robust assessment of manufacturing trends and strategic priorities. Primary inputs include structured interviews with manufacturing leaders, quality and regulatory experts, and senior development strategists who provided qualitative perspectives on process transfer, facility design, and partnership models. These insights were triangulated with publicly available regulatory documents, peer-reviewed publications on vector and nonviral delivery technologies, and company disclosures that describe facility investments and capability roadmaps.
Analytical rigor was applied through thematic coding of qualitative data, cross-referencing of technology-specific process constraints, and validation of operational practices against current regulatory expectations. Wherever possible, the methodology prioritized corroborated technical details over speculative assertions, and it emphasized replicable process characteristics that inform manufacturing decisions. Limitations included variability in proprietary process information and the rapidly evolving nature of platform technologies, which the research addresses by highlighting observable trends and validated practices rather than speculative performance claims. This methodological approach yields pragmatic insights designed to inform strategy, sourcing decisions, and operational planning.
A succinct synthesis of how technical, regulatory, and supply chain priorities converge to determine competitive positioning and partnership success in advanced therapy manufacturing
In conclusion, the cell and gene therapy biomanufacturing CDMO environment is maturing into a landscape where technical differentiation, regulatory readiness, and supply chain resilience determine competitive positioning. Sponsors increasingly demand partners who can provide integrated development services, scalable manufacturing, and strong analytical capabilities that reduce transfer risk and accelerate program timelines. At the same time, evolving trade and procurement dynamics are prompting stakeholders to reassess sourcing strategies and to prioritize investments that reduce exposure to external disruptions.
Moving forward, organizations that successfully combine platform flexibility, operational excellence, and strategic partnerships will be best positioned to capture program flow and to support the growing complexity of advanced therapies. The emphasis should be on measurable process robustness, validated transferability, and the ability to adapt to therapy-specific and region-specific demands. These priorities will underpin successful development and commercialization pathways as the sector continues its evolution.
Note: PDF & Excel + Online Access - 1 Year
Table of Contents
187 Pages
- 1. Preface
- 1.1. Objectives of the Study
- 1.2. Market Segmentation & Coverage
- 1.3. Years Considered for the Study
- 1.4. Currency
- 1.5. Language
- 1.6. Stakeholders
- 2. Research Methodology
- 3. Executive Summary
- 4. Market Overview
- 5. Market Insights
- 5.1. Integration of automated closed system bioreactors to enhance cell therapy scalability and reduce contamination risk
- 5.2. Adoption of digital twin modeling and AI-driven process analytics for real-time bioprocess optimization in cell and gene therapy manufacturing
- 5.3. Expansion of allogeneic cell therapy contract manufacturing capabilities to meet increasing global commercial demand
- 5.4. Implementation of single-use sensor technologies for continuous monitoring and control of critical quality attributes in viral vector production
- 5.5. Strategic partnerships between CDMOs and academic labs to accelerate cGMP translation of novel gene editing platforms
- 5.6. Development of modular fill-finish solutions for decentralized cell therapy administration and local hospital-based manufacturing
- 5.7. Regulatory alignment efforts toward harmonized global standards for potency assays in CAR-T and gene therapy products
- 5.8. Adoption of scalable plasmid DNA supply chains to support rapid scale-up of next-generation viral vector production
- 5.9. Advances in upstream perfusion culture processes to increase yields of viral vectors and reduce production costs
- 5.10. Incorporation of sustainability initiatives such as green solvent processes and renewable energy use in cell and gene therapy CDMO operations
- 6. Cumulative Impact of United States Tariffs 2025
- 7. Cumulative Impact of Artificial Intelligence 2025
- 8. Cell & Gene Therapy Biomanufacturing CDMO Market, by Service Type
- 8.1. Analytical Services
- 8.2. Clinical Manufacturing
- 8.3. Commercial Manufacturing
- 8.4. Process Development
- 9. Cell & Gene Therapy Biomanufacturing CDMO Market, by Therapy Type
- 9.1. Cell Therapy
- 9.1.1. Allogeneic
- 9.1.2. Autologous
- 9.2. Gene Therapy
- 10. Cell & Gene Therapy Biomanufacturing CDMO Market, by Technology
- 10.1. Non-Viral Vector
- 10.1.1. Electroporation
- 10.1.2. Lipid Nanoparticles
- 10.2. Viral Vector
- 10.2.1. Aav
- 10.2.2. Adenovirus
- 10.2.3. Lentivirus
- 10.2.4. Retrovirus
- 11. Cell & Gene Therapy Biomanufacturing CDMO Market, by Scale
- 11.1. Clinical Scale
- 11.2. Commercial Scale
- 11.3. Preclinical Scale
- 12. Cell & Gene Therapy Biomanufacturing CDMO Market, by Process Stage
- 12.1. Downstream
- 12.2. Upstream
- 13. Cell & Gene Therapy Biomanufacturing CDMO Market, by Application
- 13.1. Cardiovascular
- 13.2. Neurology
- 13.3. Oncology
- 13.4. Rare Diseases
- 14. Cell & Gene Therapy Biomanufacturing CDMO Market, by End User
- 14.1. Biopharmaceutical Companies
- 14.2. Contract Research Organizations
- 14.3. Research Institutions
- 15. Cell & Gene Therapy Biomanufacturing CDMO Market, by Region
- 15.1. Americas
- 15.1.1. North America
- 15.1.2. Latin America
- 15.2. Europe, Middle East & Africa
- 15.2.1. Europe
- 15.2.2. Middle East
- 15.2.3. Africa
- 15.3. Asia-Pacific
- 16. Cell & Gene Therapy Biomanufacturing CDMO Market, by Group
- 16.1. ASEAN
- 16.2. GCC
- 16.3. European Union
- 16.4. BRICS
- 16.5. G7
- 16.6. NATO
- 17. Cell & Gene Therapy Biomanufacturing CDMO Market, by Country
- 17.1. United States
- 17.2. Canada
- 17.3. Mexico
- 17.4. Brazil
- 17.5. United Kingdom
- 17.6. Germany
- 17.7. France
- 17.8. Russia
- 17.9. Italy
- 17.10. Spain
- 17.11. China
- 17.12. India
- 17.13. Japan
- 17.14. Australia
- 17.15. South Korea
- 18. Competitive Landscape
- 18.1. Market Share Analysis, 2024
- 18.2. FPNV Positioning Matrix, 2024
- 18.3. Competitive Analysis
- 18.3.1. AGC Biologics, Inc.
- 18.3.2. Ajinomoto Bio-Pharma Services, Inc.
- 18.3.3. Alvotech A/S
- 18.3.4. BioNTech SE
- 18.3.5. BioReliance
- 18.3.6. Catalent, Inc.
- 18.3.7. Charles River Laboratories International, Inc.
- 18.3.8. Cobra Biologics Holdings Ltd.
- 18.3.9. Cobra Therapeutics
- 18.3.10. Fujifilm Diosynth Biotechnologies U.S.A., Inc.
- 18.3.11. Genezen Laboratories, Inc.
- 18.3.12. Lonza Group AG
- 18.3.13. Merck KGaA
- 18.3.14. Minaris Regenerative Medicine GmbH
- 18.3.15. Novasep SAS
- 18.3.16. Paragon Bioservices, Inc.
- 18.3.17. Recipharm AB
- 18.3.18. Rentschler Biopharma SE
- 18.3.19. Repligen Corporation
- 18.3.20. Samsung Biologics Co., Ltd.
- 18.3.21. Thermo Fisher Scientific, Inc.
- 18.3.22. Thermo Genesis Holdings, Inc.
- 18.3.23. Vironova AB
- 18.3.24. WuXi AppTec Co., Ltd.
Pricing
Currency Rates
Questions or Comments?
Our team has the ability to search within reports to verify it suits your needs. We can also help maximize your budget by finding sections of reports you can purchase.

