Picture

Questions?

+1-866-353-3335

SEARCH
What are you looking for?
Need help finding what you are looking for? Contact Us
Compare

PUBLISHER: BioInformant | PRODUCT CODE: 1497893

Cover Image

PUBLISHER: BioInformant | PRODUCT CODE: 1497893

Global Induced Pluripotent Stem Cell (iPSC) Industry Report - Market Size, Trends, and Forecasts, 2024

PUBLISHED:
PAGES: 389 Pages
DELIVERY TIME: 1-2 business days
SELECT AN OPTION
PDF (Single User License)
USD 4197
PDF (Site License)
USD 4797
PDF (Global License)
USD 5197

Add to Cart

Since the discovery of induced pluripotent stem cell (iPSC) technology in 2006, significant progress has been made in stem cell biology and regenerative medicine. New pathological mechanisms have been identified and explained, new drugs identified by iPSC screens are in the pipeline, and the first clinical trials employing human iPSC-derived cell types have been initiated. iPSCs can be used to explore the causes of disease onset and progression, create and test new drugs and therapies, and treat previously incurable diseases.

Today, methods of commercializing induced pluripotent stem cells (iPSCs) include:

  • Cellular Therapy: iPSCs are being explored in a diverse range of cell therapy applications for the purpose of reversing injury or disease.
  • Disease Modelling: By generating iPSCs from patients with disorders of interest and differentiating them into disease-specific cells, iPSCs can effectively create disease models "in a dish".
  • Drug Development and Discovery: iPSCs have the potential to transform drug discovery by providing physiologically relevant cells for compound identification, target validation, compound screening, and tool discovery.
  • Personalized Medicine: The use of techniques such as CRISPR enable precise, directed creation of knock-outs and knock-ins (including single base changes) in many cell types. Pairing iPSCs with genome editing technologies is adding a new dimension to personalized medicine.
  • Toxicology Testing: iPSCs can be used for toxicology screening, which is the use of iPSCs or their derivatives (tissue-specific cells) to assess the safety of compounds or drugs within living cells.
  • Tissue Engineering: iPSCs can be seeded onto scaffolds made from biocompatible materials. These scaffolds mimic the structure and properties of the target tissue and can provide a supportive environment for cell growth and differentiation.
  • Organoid Production: iPS cells can be coaxed to self-organize into 3D structures called organoids, which mimic the structure and function of organs. Organoids can be used for studying organ development, modeling diseases, and testing drugs.
  • Gene Editing: iPS cells can be genetically modified using techniques like CRISPR-Cas9 to correct disease-causing mutations or introduce specific genetic changes. These edited iPS cells can then be differentiated into the desired cell type for transplantation or disease modeling.
  • Research Tools: iPSCs and iPSC-derived cell types are being widely used within a diverse range of basic and applied research applications.
  • Stem Cell Banking: iPSC repositories provide researchers with the opportunity to investigate a diverse range of conditions using iPSC-derived cell types produced from both healthy and diseased donors.
  • Cultured Meat Production: iPSCs are being utilized in clean meaat production by serving as the cellular foundation for the creation of lab-grown meat.
  • 3D Bioprinting: iPSCs can be directed to differentiate into cell types of interest, such as skin, heart, or liver cells, which are then incorporated into bioinks.
  • Wildlife Conservation and De-extinction Projects: iPSCs are being used in wildlife conservation and de-extinction projects. For example, Colossal Biosciences is using iPSC technology in an effort to achieve woolly mammoth de-extinction.

iPSC Market Dynamics

Since the discovery of iPSCs in 2006, it took only seven years for the first iPSC-derived cell product to be transplanted into a human patient in 2013. Since then, iPSC-derived cells have been used within a rapidly growing number of preclinical studies, physician-led studies, and clinical trials worldwide.

The discovery of iPSC has not only favorably transformed the field of drug discovery, toxicity testing and in-a-dish disease modeling, but also powerfully impacted the field of cell and gene therapy. The ability of iPSCs to multiply in vitro and then get differentiated into specialized cells makes iPSCs an ideal source of cells of different types for curative clinical cell replacement therapies and disease modeling.

Of course, 2013 was a landmark year because it saw the first cellular therapy involving the transplant of iPSCs into humans initiated at the RIKEN Center in Kobe, Japan. Led by Dr. Masayo Takahashi, it investigated the safety of iPSC-derived cell sheets in patients with macular degeneration. In another world first, Cynata Therapeutics received approval in 2016 to launch the first formal clinical trial of an allogeneic iPSC-derived cell product (CYP-001) for the treatment of GvHD. CYP-001 is an iPSC-derived MSC product. In this historic trial, CYP-001 met its clinical endpoints and produced positive safety and efficacy data for the treatment of steroid-resistant acute GvHD.

Today, at least 155 ongoing clinical trials are using iPSC-derived specialized cells to address various indications. iPSC-derived MSCs are being tested in the treatment of steroid-resistant acute graft versus host disease (GvHD). iPSC-derived dopaminergic progenitors are being evaluated in the treatment of Parkinson's disease. iNK cell-based cancer immunotherapy is being studied in the treatment of metastatic solid tumors. iPSC-derived retinal pigment epithelial cells have shown positive results in the treatment of age-related macular degeneration (AMD). Furthermore, iPSC derived insulin secreting beta cells are being tested for the treatment of Type 1 diabetes.

Although iPSCs have the potential to be used in both allogeneic and autologous applications, the development of allogeneic therapies using iPSC-derived products is outpacing the development of autologous therapies. Several allogeneic therapies utilizing iPSC-derived cells derived from healthy donors are being used to address diabetes, Parkinson's disease, and AMD, and these therapies are quickly progressing into early phase clinical trials.

Market competitors are also commercializing iPSC-derived products for use in drug development and discovery, disease modeling, and toxicology testing. Across the broader iPSC sector, FUJIFILM CDI (FCDI) is one of the largest and most dominant players. Cellular Dynamics International (CDI) was founded in 2004 by Dr. James Thomson at the University of Wisconsin-Madison, who in 2007 derived iPSC lines from human somatic cells for the first time. The feat was accomplished simultaneously by Dr. Shinya Yamanaka's lab in Japan. FUJIFILM acquired CDI in April 2015 for $307 million. Today, the combined company (FCDI) is the world's largest manufacturer of human cells created from iPSCs for use in research, drug discovery and regenerative medicine applications.

Another iPSC specialist is ReproCELL, a company that was established as a venture company originating from the University of Tokyo and Kyoto University in 2009. It became the first company worldwide to make iPSC products commercially available when it launched its ReproCardio product, which are human iPSC-derived cardiomyocytes. Within the European market, the dominant competitors are Evotec, Ncardia, and Axol Bioscience. Headquartered in Hamburg, Germany, Evotec is a drug discovery alliance and development partnership company. It is developing an iPSC platform with the goal to industrialize iPSC-based drug screening as it relates to throughput, reproducibility, and robustness. Today, Evotec's infrastructure represents one of the largest and most advanced iPSC platforms globally.

Ncardia was formed through the merger of Axiogenesis and Pluriomics in 2017. Its predecessor, Axiogenesis, was founded in 2011 with an initial focus on mouse embryonic stem cell-derived cells and assays. When Yamanaka's iPSC technology became available, Axiogenesis became the first European company to license it in 2010. Today, the combined company (Ncardia) is a global authority in cardiac and neural applications of human iPSCs. Founded in 2012, Axol Bioscience is a smaller but noteworthy competitor that specializes in iPSC-derived products. Headquartered in Cambridge, UK, it specializes in human cell culture, providing iPSC-derived cells and iPSC-specific cell culture products.

Of course, the world's largest research supply companies are also commercializing a diverse range of iPSC-derived products and services. Examples of these companies include Lonza, BD Biosciences, Thermo Fisher Scientific, Merck, Takara Bio, and countless others. In total, at least 90 market competitors now offer a diverse range of iPSC products, services, technologies, and therapeutics.

This global strategic report reveals all major market competitors worldwide, including their core technologies, strategic partnerships, and products under development. It covers the current status of iPSC research, biomedical applications, manufacturing technologies, patents, and funding events, as well as all known trials for the development of iPSC-derived cell therapeutics worldwide. Importantly, it profiles leading market competitors worldwide and presents a comprehensive market size breakdown for iPSCs by Application, Technology, Cell Type, and Geography (North America, Europe, Asia/Pacific, and Rest of World). It also presents total market size figures with projected growth rates through 2030.

TABLE OF CONTENTS

1. REPORT OVERVIEW

  • 1.1. Statement of the Report
  • 1.2. Executive Summary

2. INTRODUCTION

3. . CURRENT STATUS OF IPSC INDUSTRY

  • 3.1. Progress made in Autologous Cell Therapy using iPSCs
    • 3.1.1. Examples of Autologous iPSC-derived Cell Therapies in Development
    • 3.1.2. Manufacturing Timeline for Autologous iPSC-derived Cell Products
    • 3.1.3. Cost of iPSC Production
    • 3.1.4. Automation in iPSC Production
  • 3.2. Allogeneic iPSC-based Cell Therapies
  • 3.3. Share of iPSC-based Research within the Overall Stem Cell Industry
  • 3.4. Major Focus Areas of iPSC Companies
  • 3.5. Commercially Available iPSC-derived Cell Types
  • 3.6. Relative use of iPSC-derived Cell Types in Toxicology Testing Assays
  • 3.7. iPSC-derived Cell Types used in Clinical Trials
  • 3.8. Currently Available iPSC Technologies
    • 3.8.1. Brief Descriptions of some iPSC-related Technologies
      • 3.8.1.1. Nucleofector Technology
      • 3.8.1.2. Opti-ox Technology
      • 3.8.1.3. MOGRIFY Technology
      • 3.8.1.4. Transcription Factor-based iPSC Differentiation Technology
      • 3.8.1.5. Flowfect Technology
      • 3.8.1.6. Technology for Mass Production of Platelets
      • 3.8.1.7. SynFire Technology

4. HISTORY OF INDUCED PLURIPOTENT STEM CELLS (IPSCS)

  • 4.1. First iPSC Generation from Mouse Fibroblasts, 2006
  • 4.2. First Human iPSC Generation, 2007
  • 4.3. Creation of CiRA, 2010
  • 4.4. First High-Throughput Screening using iPSCs, 2012
  • 4.5. First iPSC Clinical Trial Approved in Japan, 2013
  • 4.6. First iPSC-RPE Cell Sheet Transplantation for AMD, 2014
  • 4.7. EBiSC Founded, 2014
  • 4.8. First Clinical Trial using Allogeneic iPSCs for AMD, 2017
  • 4.9. Clinical Trial for Parkinson's Disease using Allogeneic iPSCs, 2018
  • 4.10. Commercial iPSC Plant SMaRT Established, 2018
  • 4.11. First iPSC Therapy Center in Japan, 2019
  • 4.12. First U.S.-based NIH-Sponsored Clinical Trial using iPSCs, 2019
  • 4.13. Cynata Therapeutics' World's Largest Phase III Clinical Trial, 2020
  • 4.14. Tools and Know-how to Manufacture iPSCs in Clinical Trials, 2021
  • 4.15. Production of in-house iPSCs using Peripheral Blood Cells, 2022

5. RESEARCH PUBLICATIONS ON IPSCS

  • 5.1. Rapid Growth in iPSC Publications
    • 5.1.1. PubMd Publications on Pathophysiological Research using iPSCs
    • 5.1.2. PubMed Papers on Reprogramming
    • 5.1.3. PubMed Papers on iPSC Differentiation
    • 5.1.4. PubMed Papers on the use of iPSCs in Drug Discovery
    • 5.1.5. PubMed Papers on iPSC-based Cell Therapy
      • 5.1.5.1. Percent Share of Published Articles by Disease Type
      • 5.1.5.2. Percent Share of Articles by Country

6. IPSC: PATENT LANDSCAPE ANALYSIS

  • 6.1. iPSC Patent Applications by Jurisdiction
  • 6.2. iPSC Patent Applicants
  • 6.3. Inventors of iPSC Patents
  • 6.4. iPSC Patent Owners
  • 6.5. Legal Status of iPSC Patents

7. IPSC: CLINICAL TRIAL LANDSCAPE

  • 7.1. Number of iPSC Clinical Trials
  • 7.1. Recruitment Status of iPSC Clinical Trials
  • 7.3. iPSC Clinical Trials Stydy Designs
  • 7.4. Therapeutic & Non-Therapeutic iPSC Clinical Trials
    • 7.4.1. Non-Therapeutic Clinical Studies by Use
    • 7.4.2. Diseases Targeted by Therapeutic Studies
      • 7.4.2.1. Therapeutic Clinical Studies by Autologous & Allogeneic Sources of iPSCs
    • 7.4.3. Examples of iPSC-based Therapeutic Studies
  • 7.5. iPSC-based Trials by Phase of Study
  • 7.6. iPSC Clinical Trials by Funder Type
  • 7.7. Geographic Distribution of iPSC-based Clinical Trials
  • 7.8. Promising iPSC Product Candidates
    • 7.8.1. CYP-001, CYP-004 & CYP-006 from Cynata Therapeutics
    • 7.8.2. BioVAT-HF from Repairon GmbH
    • 7.8.3. HS-001 from Heartseed
    • 7.8.4. CNTY-101 from Century Therapeutics
    • 7.8.5. FT-576 & FT-819 from Fate Therapeutics
    • 7.8.6. RPE from National Eye Institute
    • 7.8.7. QN-019a from Qihan Biotech
    • 7.8.8. iPSC-CL from Heartworks, Inc.
  • 7.9. Companies having Preclinical iPSC Assets
    • 7.9.1. Aspen Neuroscience
    • 7.9.2. Ryne Biotech
    • 7.9.2. Bluerock Therapeutics
    • 7.9.4. Vita Therapeutics
    • 7.9.5. Hopstem Biotechnology
    • 7.9.6. Res Nova Bio, Inc.
    • 7.9.7. Cytovia Therapeutics
    • 7.9.8. Hebecell Corporation
    • 7.9.9. Sana Biotechnology
    • 7.9.10. SCG Cell Therapy Pte
    • 7.9.11. Cytomed
    • 7.9.12. Shoreline Biosciences
    • 7.9.13. Neukio Biotherapeutics
    • 7.9.14. Exacis Biotherapeutics
    • 7.9.15. CellOrigin Biotech

8. M&A, COLLABORATIONS & FUNDING ACTIVITIES IN IPSC SECTOR

  • 8.1. Mergers and Acquisitions (M&A) Sector
    • 8.1.1. Century Therapeutics & Clade Therapeutics
    • 8.1.2. Evotech & Rigenerand
    • 8.1.3. Fujifilm Corporation & Atara Biotherapeutics
    • 8.1.4. Catalent & RheinCell Therapeutics
    • 8.1.5. Axol Biosciences & Censo Biotechnologies
    • 8.1.6. Bayer AG & Bluerock Therapeutics
    • 8.1.7. Pluriomix & Axiogenesis
  • 8.2. Partnership/Collaboration & Licensing Deals in iPSC Sector
    • 8.2.1. Shinobi Therapeutics & Panasonic
    • 8.2.2. SCG Cell Therapy and A*STAR
    • 8.2.3. Charles River Laboratories & Pluristyx, Inc.
    • 8.2.4. Pluristyx, Inc. & National Resilience, Inc.
    • 8.2.5. University of Texas & GeneCure
    • 8.2.6. Heartseed, Inc. & Undisclosed Biotech
    • 8.2.7. Bluerock Therapeutics & Bit.bio
    • 8.2.8. Applied Stem Cell, Inc. & CIRM
    • 8.2.9. Resolution Therapeutics & OmniaBio, Inc.
    • 8.2.10. REPROCELL, Inc. & CIRM
    • 8.2.11. REPROCELL, Inc. & BioBridge Global
    • 8.2.12. Elevate Bio & CIRM
    • 8.2.13. Evotec & Sernova
    • 8.2.14. Evotec & Almiral
    • 8.2.15. Quell Therapeutics & Cellistic
    • 8.2.16. MDimmune & YiPSCELL
    • 8.2.17. Edigene & Neukio Biotherapeutics
    • 8.2.18. Matricelf & Ramot
    • 8.2.19. Evotec & Boehringer Ingelheim
    • 8.2.20. Pluristyx, Pancella & Implant Therapeutics
    • 8.2.21. Century Therapeutics & Bristol Myers Squibb
    • 8.2.22. Fujifilm Cellular Dynamics & Pheno Vista Biosciences
    • 8.2.23. Metrion Biosciences & Bioqube Ventures
    • 8.2.24. Cytovia Therapeutics & Cellectis
    • 8.2.25. Exacis Biotherapeutics & CCRM
    • 8.2.26. Cynata Therapeutics & Fujifilm Corporation
    • 8.2.27. Bone Therapeutics & Implant Therapeutics
    • 8.2.28. REPROCELL & TEXCELL
    • 8.2.29. Jacobio & Herbecell
    • 8.2.30. NeuCyte & KIF1A.ORG
    • 8.2.31. Kite & Shoreline Biosciences
    • 8.2.32. Neuropath Therapeutics & Hopstem Biotechnology
    • 8.2.33. Allele Biotech & Cellatoz
    • 8.2.34. Bluerock Therapeutics, Fujifilm Cellular Dynamics & Opsis Therapeutics
    • 8.2.35. Newcells & Takeda
    • 8.2.36. Biocentriq & Kytopen
    • 8.2.37. Fujifilm Cellular Dynamics & Sana Biotechnology
    • 8.2.38. Evotec & Medical Center Hamburg-Eppdorf (UKE)
    • 8.2.39. NeuCyte & Seaver Autism Center for Research and Treatment
    • 8.2.40. Cytovia Therapeutics & National Cancer Institute
    • 8.2.41. Mogrify & MRC Laboratory of Molecular Biology
  • 8.3. Venture Capital Funding in iPSC Sector
    • 8.3.1. Asgard Therapeutics
    • 8.3.2. Kenai Therapeutics
    • 8.3.3. Pluristyx
    • 8.3.4. Fujifilm Cellular Dynamics
    • 8.3.5. Mogrify Ltd.
    • 8.3.6. Heartseed, Inc.
    • 8.3.7. Elevate Bio
    • 8.3.9. Aspen Neurosciences
    • 8.3.10. Axol Biosciences
    • 8.3.11. Thyas, Co. Ltd
    • 8.3.12. Synthego
    • 8.3.13. Cellino Biotech, Inc
    • 8.3.14. Curi Bio
    • 8.3.15. Ncardia
    • 8.3.16. Evotec SE
    • 8.3.17. bit.bio
    • 8.3.18. Clade Therapeutics
    • 8.3.19. Shoreline Biosciences
    • 8.3.20. Kytopen
    • 8.3.21. Cytovia Therapeutics & CytoLynx
    • 8.3.22. TreeFrog Therapeutics
    • 8.3.23. HebeCell Corporation
    • 8.3.24. Neukio Biotherapeutics
    • 8.3.25. Stemson Therapeutics
    • 8.3.26. Vita Therapeutics
    • 8.3.27. Century Therapeutics
    • 8.3.28. Heartseed
    • 8.3.29. Mogrify
    • 8.3.30. Metrion Biosciences
    • 8.3.31. Elevate Bio
    • 8.3.32. Vita Therapeutics

9. GENERATION OF INDUCED PLURIPOTENT STEM CELLS (IPSCS)

  • 9.1. OSKM Cocktail
    • 9.1.1. Octamer-binding Transcription Factor 4 (Oct4)
    • 9.1.2. Sry-related Box (SOX) Factor 2
    • 9.1.3. Kruppel-like Factors (Klf4)
    • 9.1.4. C-Myc
  • 9.2. Pluripotency-Associated Transcription Factors and their Functions
    • 9.2.1. Different Combinations of Factors for Different Cell Sources
  • 9.3. Delivery of Reprogramming Factors
    • 9.3.1. Integrating Systems
      • 9.3.1.1. Retroviral Vectors
      • 9.3.1.2. Lentiviral Vectors
      • 9.3.1.3. piggyBack (PB) Transposon Method
    • 9.3.2. Non-Integrative Delivery Systems
      • 9.3.2.1. Adenoviral Vectors
      • 9.3.2.2. Sendai Viral Vectors
      • 9.3.2.3. Plasmid Vectors
      • 9.3.2.4. Minicircles
      • 9.3.2.5. oriP/Epstein-Barr Nuclear Antigen-1 (EBNA1)-based Episomes
      • 9.3.2.6. RNA Delivery Approach
      • 9.3.2.7. Proteins
    • 9.3.3. Comparison of Delivery Methods
  • 9.4. Genome Editing Technologies in iPSC Generation
    • 9.4.1. CRISPR/Cas9
  • 9.5. Available iPSC Lines and their applications

10. HUMAN IPSC BANKING

  • 10.1. Major Biobanks Storing iPSCs & iPSC Lines
    • 10.1.1. RIKEN
      • 10.1.1.1. Human iPS Cells offered by RIKEN
    • 10.1.2. WiCell
      • 10.1.2.1. WiCell's iPSC Lines
    • 10.1.3. Fujifilm Cellular Dynamics, Inc.
      • 10.1.3.1. iPSC Generation
    • 10.1.4. Sampled
      • 10.1.4.1. Biobanking Services
      • 10.1.4.2. Sampled's iPSC Services
    • 10.1.5. Coriell Institute for Medical Research
      • 10.1.5.1. iPSCs at Coriell
      • 10.1.5.2. Coriell's Biobank
        • 10.1.5.2.1. National Institute of General Medical Sciences (NIGMS)
        • 10.1.5.2.2. National Institute on Aging (NIA)
        • 10.1.5.2.3. Allen Cell Collection
        • 10.1.5.2.4. iPSC Collection from Rett Syndrome Research Trust
        • 10.1.5.2.5. Autism Research Resource
        • 10.1.5.2.6. HD Community BioRepository
        • 10.1.5.2.7. CDC Cell and DNA Repository
        • 10.1.5.2.8. NEI-AREDS Genetic Repository
    • 10.1.6. European Bank for Induced Pluripotent Stem Cells (EBiSC)
      • 10.1.6.1. EBiSC Catalogue
      • 10.1.6.2. EBiSC's iPSC Banking Service
  • 10.2. Cell Sources for iPSC Banks
  • 10.3. Reprogramming Methods in iPSC Banks
  • 10.4. Ownership and Investments made in iPSC Banks

11. BIOMEDICAL APPLICATIONS OF IPSCs

  • 11.1. iPSCs in Basic Research
    • 11.1.1. To Understand Cell Fate Control
    • 11.1.2. To Understand Cell Rejuvenation
    • 11.1.3. To Understand Pluripotency
    • 11.1.4. To Study Tissue & Organ Development
    • 11.1.5. To Produce Human Gametes from iPSCs
    • 11.1.6. Providers of iPSC-Related Services for Researchers
  • 11.2. Applications of iPSCs in Drug Discovery
    • 11.2.1. Drugs Tested for Cardiovascular Diseases using iPSCs
    • 11.2.2. Drugs Tested for Neurological Diseases using iPSC Lines
    • 11.2.3. Drugs Tested for Rare Diseases using iPSC Lines
  • 11.3. Applications of iPSCs in Toxicology Studies
    • 11.3.1. Examples of Drugs Tested for their Toxicity using iPSCs
    • 11.3.2. Relative Use of iPSC-Derived Cell Types used in Toxicity Testing Studies
  • 11.4. Applications of iPSCs in Disease Modeling
    • 11.4.1. Cardiovascular Diseases Modeled with iPSC-Derived Cells
      • 11.4.1.1. Percent Utilization of iPSCs for Cardiovascular Disease Modeling
    • 11.4.2. Modeling Liver Diseases using iPSC-Derived Hepatocytes
    • 11.4.3. iPSCs in Neurodegenerative Disease Modeling
    • 11.4.4. iPSC-derived Organoids for Disease Modeling
    • 11.4.5. Cancer-Derived iPSCs
  • 11.5. Applications of iPSCs in Cell-Based Therapies
    • 11.5.2. Companies Focusing only on iPSC-based Therapies
  • 11.6. Other Novel Applications of iPSCs
    • 11.6.1. Applications of iPSCs in Tissue Engineering
      • 11.6.1.1. 3D Bioprinting Techniques
      • 11.6.1.2. Biomaterials
      • 11.6.1.3. 3D Bioprinting Strategies
      • 11.6.1.4. Bioprinting iPSC-Derived Cells
    • 11.6.2. iPSCs from Farm Animals
      • 11.6.2.1. iPSCs Generated from Cattle
      • 11.6.2.2. iPSCs from Sheep
      • 11.6.2.3. iPSCs from Goat
      • 11.6.2.4. iPSCs Generated from Buffalo
      • 11.6.2.5. iPSC Generation from Avians
    • 11.6.3. iPSC Lines for the Preservation of Endangered Species of Animals
    • 11.6.4. iPSCs in Cultured Meat

12. MARKET ANALYSIS

  • 12.1. Global Market for iPSCs by Geography
  • 12.2. Global Market for iPSCs by Technology
  • 12.3. Global Market for iPSCs by Biomedical Application
  • 12.4. Global Market for iPSCs by Derived Cell Type
  • 12.5. Market Drivers
    • 12.5.1. Current Drivers Impacting the iPSC Market Place
  • 12.6. Market Restraints
    • 12.6.1. Economic Issues
    • 12.6.2. Genomic Instability
    • 12.6.3. Immunogenicity
    • 12.6.4. Biobanking

13. COMPANY PROFILES

  • 13.1. AcceGen
    • 13.1.1. ASC-CRISPR iPSC Gene Editing Technology Service
  • 13.2. Acellta, Ltd.
    • 13.2.1. Technology
      • 13.2.1.1. Maxells
      • 13.2.1.2. Singles
      • 13.2.1.3. Differentiation
      • 13.2.1.4. Manufacturing Facility
      • 13.2.1.5. Services
  • 13.3. AddGene, Inc.
    • 13.3.1. Viral Plasmids
  • 13.4. Allele Biotechnology, Inc.
    • 13.4.1. Technologies
      • 13.4.1.1. mRNA Genome Editing
      • 13.4.1.2. Single Cell Cloning
  • 13.5. ALSTEM, Inc.
    • 13.5.1. Cell Line Generation Tools
    • 13.5.2. Cell Immortalization Kits
    • 13.5.3. iPSC Kits
    • 13.5.4. Cell Lines
    • 13.5.5. Gene Editing
    • 13.5.6. iPS Cell Lines
    • 13.5.7. Virus Packaging Tools
  • 13.6. Altos Labs
    • 13.6.1. Altos' Science
  • 13.7. AMS Biotechnology, Ltd. (AMSBIO)
    • 13.7.1. Cell Line Products
      • 13.7.1.1. Disease Models
      • 13.7.1.2. Viral Production Services
  • 13.8. Applied StemCell (ASC)
    • 13.8.1. iPSC-Based Preclinical CRO Services
      • 13.8.1.1. Reprogramming to Differentiation
      • 13.8.1.2. Neurotoxicity Screening
    • 13.8.2. GMP Grade iPSC Services & Products
      • 13.8.2.1. GMP iPSC
      • 13.8.2.2. Knock-In Ready GMP TARGATT iPSCs
    • 13.8.3. GMP TARGATT iPSC-iNK Platform
    • 13.8.4. CRISPR iPSC Genome Editing Service
      • 13.8.4.1. CRISPR Knock-In & Point Matation iPS Cell Generation
      • 13.8.4.2. CRISPR iPSC Gene Knockout
      • 13.8.4.3. TARGATT Knock-In iPS Cells
    • 13.8.5. iPSC Generation Services
    • 13.8.6. iPSC Differentiation Service
    • 13.8.7. Stem Cell Products
  • 13.9. Asgard Therapeutics
  • 13.10. Aspen Neurosciences, Inc.
    • 13.10.1. Aspen's Clinical Pipeline
  • 13.11. Astellas Pharma, Inc.
    • 13.11.1. Allogeneic Cell Therapy
    • 13.11.2. Universal Donor Cell Technology
    • 13.11.3. Astella's Robust Pipeline
  • 13.12. Axol Biosciences, Ltd.
    • 13.12.1. Axol's Genetically Engineered Disease Lines
    • 13.12.2. Custom Human iPSC iPSC Services
    • 13.12.3. Axol's Products
  • 13.13. BioCentriq
    • 13.13.1. LEAP Advanced Therapy Platform
  • 13.14. Bit.bio
    • 13.14.1. Therapeutics
    • 13.14.2. Opti-Ox Reprogramming Technology
      • 13.14.2.1. ioCells
      • 13.14.2.2. ioWild Type Cells
      • 13.14.2.3. ioGlutamatergic Neurons
      • 13.14.2.4. ioSkeletal Myocytes
      • 13.14.2.5. ioGABAergic Neurons
      • 13.14.2.6. ioDisease Models
      • 13.14.2.7. ioGlutamatergic Neurons50CAGWT
  • 13.15. BlueRock Therapeutics LP
    • 13.15.1. BlueRock's Cell Therapy
    • 13.15.2. CELL + GENE Platform
    • 13.15.3. BlueRock's Cell Therapy Programs
  • 13.16. BrainXell
    • 13.16.1. Products
    • 13.16.2. Custom Service Projects
    • 13.16.3. In-House Assay Services
  • 13.17. Cartherics Pty, Ltd.
    • 13.17.1. Allogeneic CAR Immune Cells
  • 13.18. Catalent Biologics
    • 13.18.1. OneBio Integrated Suite
    • 13.18.2. Drug Substance Development
    • 13.18.3. Drug Product Development
    • 13.18.4. Analytical Services
    • 13.18.5. Catalent's iPSC Services
  • 13.19. Cellistic
    • 13.19.1. Pulse Platform
    • 13.19.2. Echo Platform
    • 13.19.3. iPSC-based Allogeneic Approach
      • 13.19.3.1. Model 1
      • 13.19.3.2. Model 2
  • 13.20. CellOrigin Biotech (Hangzhou), Co., Ltd.
  • 13.21. Celogics, Inc.
    • 13.21.1. Celo-Cardiomyocytes
  • 13.22. Cellular Engineering Technologies (CET)
    • 13.22.1. iPS Cell Reprogramming Methods
    • 13.22.2. Applications of CET's Stem Cells
      • 13.22.2.1. Hypoimmune Cell Lines
      • 13.22.2.2. Cell Therapy Development
      • 13.22.2.3. Disease Modeling
      • 13.22.2.4. Drug Development & Discovery
      • 13.22.2.5. Regenerative Medicine
      • 13.22.2.6. Toxicology Studies
    • 13.22.3. Products
  • 13.23. Cellusion, Inc.
    • 13.23.1. Orphan Drug Designation
    • 13.23.2. Bullous Keratopathy
  • 13.24. Century Therapeutics, Inc.
    • 13.24.1. Cell Therapy Platform
    • 13.24.2. Century's Product Pipeline
  • 13.25. Citius Pharmaceuticals, Inc.
    • 13.25.1. Stem Cell Platform
  • 13.26. Creative Bioarray
    • 13.26.1. Pluripotent Stem Cells
    • 13.26.2. iPSC-Derived Cells
    • 13.26.3. Services
  • 13.27. Curi Bio
    • 13.27.1. Disease Model Development Services
  • 13.28. Cynata Therapeutics, Ltd.
    • 13.28.1. Cymerus Platform
    • 13.28.2. Clinical Development for GvHD
    • 13.28.3. Osteoarthritis
    • 13.28.4. ARDS
    • 13.28.5. Diabetic Wounds
  • 13.29. Cytovia Therapeutics
    • 13.29.1. iPSC-derived NK & CAR-NK Cells
  • 13.30. DefiniGEN
    • 13.30.1. DefiniGEN's Platform
    • 13.30.2. Efficacy Screening Services
    • 13.30.3. Toxicology Screening
    • 13.30.4. Disease Models
    • 13.30.5. iPSC Cell Products
  • 13.31. Editas Medicine
    • 13.31.1. SLEEK Gene Editing
    • 13.31.2. iPSC-Derived NK Cells
  • 13.32. Editco Bio., Inc.
    • 13.32.1. Knockout iPS Cell Lines
    • 13.32.2. Knock-in iPS Cell Lines
  • 13.33. ElevateBio
    • 13.33.1. iPSC Technology
  • 13.34. Elixirgen Scientific, Inc.
    • 13.34.1. Technology
    • 13.34.2. Service Offerings
    • 13.34.3. iPSC Products
  • 13.35. Eterna Therapeutics
    • 13.35.1. Gene Editing
    • 13.35.2. Gene Delivery
  • 13.36. Evotec AG
    • 13.36.1. iPS Cell Therapies
    • 13.36.2. Drug Discovery Services
    • 13.36.3. Therapeutic Areas
  • 13.37. Eyestem
    • 13.37.1. Eyecyte-RPE
    • 13.37.2. Eyecyte-PRP
    • 13.37.3. Aircyte-AEC
  • 13.38. Fate Therapeutics
    • 13.38.1. iPSC Platform
    • 13.38.2. iPSC Manufacturing
    • 13.38.3. Product Pipeline
      • 13.38.3.1. FT576
      • 13.38.3.2. FT522
      • 13.38.3.3. FT819
      • 13.38.3.4. FT825
    • 13.38.4. Fate Therapeutics' Collaborations
      • 13.38.4.1. ONO Pharmaceutical, Co., Ltd.
      • 13.38.4.2. Masonic Cancer Center, University of Minnesota
      • 13.38.4.3. Memorial Sloan-Kettering Cancer Center
      • 13.38.4.4. Oslo University Hospital
  • 13.39. FUJIFILM Cellular Dynamics, Inc.
    • 13.39.1. Products
    • 13.39.2. FUJIFILM's Custom Services
    • 13.39.3. iPSC Disease Modeling
    • 13.39.4. Safety Pharmacology/Toxicology Testing
  • 13.40. Gameto
    • 13.40.1. Fertilo
  • 13.41. Greenstone Biosciences
  • 13.42. Heartseed, Inc.
    • 13.42.1. HS-001: The Lead Product Candidate
    • 13.42.2. Technologies
      • 13.42.2.1. Remuscularization
      • 13.42.2.2. Patented iPSC Production
      • 13.42.2.3. Differentiation
      • 13.42.2.4. Purification
      • 13.42.2.5. Spheroid
  • 13.43. HebeCell
    • 13.43.1. ProtoNK
    • 13.43.2. Retinal Photoreceptor Progenitors
    • 13.43.3. Nanoproteins
  • 13.44. Helios K.K.
    • 13.44.1. Research Activities
  • 13.45. Hera BioLabs
    • 13.45.1. Proprietary SRG Rat
    • 13.45.2. Cas-CLOVER Gene Editing Platform
    • 13.45.3. The piggyback Transposon System Platform
    • 13.45.4. Cell Line Development Services
    • 13.45.5. Custom Cell Line Engineering Services
    • 13.45.6. Animal Model Creation
    • 13.45.7. In vivo Research Services
      • 13.45.7.1. Custom Research Models
      • 13.45.7.2. Metabolic Disease Models
      • 13.45.7.3. Xenograft & PDX Services
      • 13.45.7.4. Pharmacology & Toxicology Services
  • 13.46. Hopstem Biotechnology
    • 13.46.1. Pipeline
  • 13.47. Implant Therapeutics, Inc.
    • 13.47.1. Services
  • 13.48. IN8bio
    • 13.48.1. The DeltEx Platform
    • 13.48.2. iPSC Gamma-Delta T Cells
  • 13.49. I Peace, Inc.
    • 13.49.1. GMP Products
    • 13.49.2. Custom Manufacturing Services
  • 13.50. IPS HEART
    • 13.50.1. IPS HEART's Approach
    • 13.50.2. ISX-9 CPC
    • 13.50.3. GIVI-MPC
  • 13.51. iPS Portal, Inc.
    • 13.51.1. Services
      • 13.51.1.1. Development Services
      • 13.51.1.2. Business Support Services
  • 13.52. iPSirius
    • 13.52.1. iPSirius' Platform
  • 13.53. iXCells Biotechnologies
    • 13.53.1. iPS Cell Products
    • 13.53.2. Preclinical Services
  • 13.54. Kenai Therapeutics, Inc.
  • 13.55. Khloris Biosciences, Inc.
  • 13.56. Kytopen
    • 13.56.1. Products
      • 13.56.1.1. Flowfect Discover
      • 13.56.1.2. Flowfect TX
      • 13.56.1.3. Flowfect Connect
  • 13.57. Laverock Therapeutics
    • 13.57.1. GEiGS and iPSCs
    • 13.57.2. Ex Vivo GEiGS-Enabled Cell Therapies
  • 13.58. Lindville Bio, Ltd.
    • 13.58.1. Services
  • 13.59. Lonza Group, Ltd.
    • 13.59.1. iPSC Manufacturing Expertise
    • 13.59.2. Nucleofector Technology
  • 13.60. Matricelf
    • 13.60.1. Solution to Spinal Cord Injury
  • 13.61. Megakaryon Corporation
    • 13.61.1. Production of Platelets from iPSCs
    • 13.61.2. Development of Megakaryocytes from iPSCs
    • 13.61.3. Safe Production of Platelets
    • 13.61.4. Research & Development Pipeline
  • 13.62. Metrion Biosciences, Ltd.
    • 13.62.1. Ion Channel High-Throughput Screening
    • 13.62.2. Clinical QTc/QRS Prediction using hiPSC-Derived Cardiomyocytes
  • 13.63. Mogrify
    • 13.63.1. MOGRIFY Platform
    • 13.63.2. epiMOGRIFY Platform
  • 13.64. Ncardia Services B.V.
    • 13.64.1. Ncyte Astrocytes
    • 13.64.2. Ncyte Endothelial Cells
    • 13.64.3. Ncyte Neural Mix
    • 13.64.4. Ncyte Smooth Muscle Cells
    • 13.64.5. Ncyte vCardiomyocytes
    • 13.64.6. Custom Disease Modeling Services
    • 13.64.7. High-Throughput Screening Services
    • 13.64.8. iPSC-Based Efficacy Assay Services
    • 13.64.9. iPSC-Based Safety & Toxicity Assays
  • 13.65. NeuCyte
    • 13.65.1. Technology
    • 13.65.2. Drug Discovery
  • 13.66. Neukio Biotherapeutics
    • 13.66.1. Allogeneic Immunotherapy Platform
  • 13.67. Newcells Biotech
    • 13.67.1. Retina Models
    • 13.67.2. Retinal Organoids
    • 13.67.3. Retinal Pigment Epithelium (RPE)
    • 13.67.4. Kidney Proximal Tubule Cell Model
    • 13.67.5. Assay-Ready aProximate
    • 13.67.6. Glomerular Toxicity and Disease Modeling
    • 13.67.7. Lung Airway Models
    • 13.67.8. Disease Modeling Services
      • 13.67.8.1. In vitro Retinal Disease Modeling for Retinal Therapy
      • 13.67.8.2. in vitro Evaluation of Retinal Toxicity Services
      • 13.67.8.3. Gene Therapy Services
      • 13.67.8.4. Drug Transporter Interactions & DDI Services
      • 13.67.8.5. Cross Species Comparison Services
      • 13.67.8.6. Kidney Toxicity Services
      • 13.67.8.7. Kidney Disease Modeling Services
      • 13.67.8.8. Fibroblast Assay Services
      • 13.67.8.9. Lung Toxicity Study Services
  • 13.68. NEXEL, Co., Ltd.
    • 13.68.1. Products
      • 13.68.1.1. Cardiosight-S
      • 13.68.1.2. Hepatosight-S
      • 13.68.1.3. Neurosight-S
    • 13.68.2. Curi Bio Systems
      • 13.68.2.1. Mantarray
      • 13.68.2.2. Cytostretcher
      • 13.68.2.3. NanoSurface Plates
    • 13.68.3. Services
      • 13.68.3.1. NeXST (Next Xight Screening Test)
      • 13.68.3.2. Curi Engine SVC
  • 13.69. Notch Therapeutics
    • 13.69.1. Technology
    • 13.69.2. Product Development
  • 13.70. Orizuru Therapeutics, Inc.
    • 13.70.1. iCM Project
  • 13.71. Phenocell SAS
    • 13.71.1. iPSC-derived RPE Cells for Age-related Macular Degeneration (AMD)
    • 13.71.2. R&D Solutions for Acne & Hyperseborrhea
    • 13.71.3. Skin Pigmentation Research & Testing Platform
    • 13.71.4. Cells & Kits
  • 13.72. Pluristyx
    • 13.72.1. The panCELLa Platform
    • 13.72.2. RTD iPSC & GMP Cell Banks
    • 13.72.3. Development Services
    • 13.72.4. Custom Gene Editing
    • 13.72.5. iPSC GMP Manufacturing Expertise
    • 13.72.6. Custom Gene Editing
    • 13.72.7. FailSafe
    • 13.72.8. iACT Stealth Cells
    • 13.72.9. Products
      • 13.72.9.1. PluriBank PSCs
      • 13.72.9.2. ESI Pluripotent Stem Cells
      • 13.72.9.3. Wild Type & Disease Affected PSCs
    • 13.72.10. Differentiated Cells
  • 13.73. ReNeuron
    • 13.73.1. Technology Platform
  • 13.74. Repairon GmbH
    • 13.74.1. Technology
      • 13.74.1.1. Engineered Heart Muscle (EHM)
  • 13.75. REPROCELL USA, Inc.
    • 13.75.1. Services
      • 13.75.1.1. Donor Recruitment and Patient-Derived Cells
      • 13.75.1.2. Example Case Study
      • 13.75.1.3. Target Cell Isolation
      • 13.75.1.4. iPSC Reprograming Service
      • 13.75.1.5. iPSC Expansion, Characterization and Banking Services
      • 13.75.1.6. Neuronal Differentiation Services
      • 13.75.1.7. Gene Editing Services
    • 13.75.2. REPROCELL's iPSC Products
      • 13.75.2.1. Stemgent
  • 13.76. Res Nova Bio, Inc.
    • 13.76.1. Preclinical Study
  • 13.77. Sartorius CellGenix GmbH
    • 13.77.1. Products
  • 13.78. Shinobi Therapeutics
  • 13.79. Shoreline Biosciences
    • 13.79.1. iMACs
  • 13.80. StemSight
    • 13.80.1. Technology
  • 13.81. Stemson Therapeutics
    • 13.81.1. iPSCs for Hair Follicles
  • 13.82. Stemina Biomarker Discovery
    • 13.82.1. Cardio quickPREDICT
    • 13.82.2. devTOX quickPREDICT
  • 13.83. Tempo Bioscience, Inc.
    • 13.83.1. Tempo-iAstro
    • 13.83.2. Tempo-iBMEC
    • 13.83.3. Tempo-iCardio
    • 13.83.4. Tempo-iCort
    • 13.83.5. Tempo-iDopaNer
    • 13.83.6. Tempo-iLSEC
    • 13.83.7. Tempo-iKupffer
    • 13.83.8. Tempo-iHepStellate
    • 13.83.9. Tempo-iHep3D
    • 13.83.10. Tempo-iKer
    • 13.83.11. Tempo-iKidneyPod
    • 13.83.12. Tempo-iMel
    • 13.83.13. Tempo-iMG
    • 13.83.14. Tempo-iMono
    • 13.83.15. Tempo-iMotorNer
    • 13.83.16. Tempo-iMSC
    • 13.83.17. Tempo-iNStem
    • 13.83.18. Tempo-iOligo
    • 13.83.19. Tempo-iOsteo
    • 13.83.20. Tempo-iPeri
    • 13.83.21. Tempo-iPhago
    • 13.83.22. Tempo-iRPE
    • 13.83.23. Tempo-iSchwann
    • 13.83.24. Tempo-iSenso
    • 13.83.25. Tempo StemBank
  • 13.84. Uncommon (Higher Steaks)
    • 13.84.1. iPSC-Based Cultured Pork
  • 13.85. Universal Cells
    • 13.85.1. Technologies
      • 13.85.1.1. Recombinant Adeno-Associated Virus
      • 13.85.1.2. PSCs for Every Organ
      • 13.85.1.3. Universal Donor Cells
      • 13.85.1.4. HLA Engineering
  • 13.86. VCCT, Inc.
    • 13.86.1. Regenerating RPE Cells
  • 13.87. ViaCyte, Inc.
    • 13.87.1. Technology
      • 13.87.1.1. Autologous Approach
      • 13.87.1.2. Allogeneic Approach
    • 13.87.2. Pipeline
  • 13.88. Vita Therapeutics
    • 13.88.1. Technology
  • 13.89. XCell Science
    • 13.89.1. Control Lines
      • 13.89.1.1. XCL-1
      • 13.89.1.2. XCL-6
    • 13.89.2. Cell Products
      • 13.89.2.1. Control Lines
      • 13.89.2.2. Knock-out Lines
      • 13.89.2.3. Reporter Lines
    • 13.89.3. Services
  • 13.90. Yashraj Biotechnology, Ltd.
    • 13.90.1. iPSC Products
    • 13.90.2. Contract Research Services

INDEX OF FIGURES

  • FIGURE 3.1: Development of iPSC-based Autologous Cell Therapy for Canavan Disease
  • FIGURE 3.2: Manufacturing Timeline for Autologous iPSC-derived Cell Products
  • FIGURE 3.3: Cost of iPSC Production
  • FIGURE 3.4: Technical Set Up of the Stem Cell Factory (SCF)
  • FIGURE 3.5: Development of iPSC-based Allogeneic Cell Therapy
  • FIGURE 3.6: Share of iPSC-based Research within the Overall Stem Cell Industry
  • FIGURE 3.7: Major Focus Areas of iPSC Companies
  • FIGURE 3.8: Relative use of iPSC-derived Cell Types in Toxicology Studies
  • FIGURE 3.9: Comparison of Lipofection and Nucleofection Technologies
  • FIGURE 5.1: No. of Research Publications on iPSC in PubMed.gov, 2010-May 29, 2024
  • FIGURE 5.2: Pubmed Publications on Pathophysiological Research using iPSCs
  • FIGURE 5.3: PubMed Publications on Reprogramming Somatic Cells
  • FIGURE 5.4: No. of PubMed Papers on iPSC Differentiation
  • FIGURE 5.5: PubMed Papers on the use of iPSCs in Drug Discovery
  • FIGURE 5.6: PubMed Papers on iPSC-based Cell Therapy
  • FIGURE 5.7: Percent Share of Published Articles by Disease Type
  • FIGURE 5.8: Percent Share of Articles by Country
  • FIGURE 6.1: Number of iPSC Patents Filed by Year, 2000-May 5, 2024
  • FIGURE 7.1: Number of Clinical Trials by Year
  • FIGURE 7.2: iPSC Clinical Trials by Design, May 2024
  • FIGURE 7.3: Therapeutic & Non-Therapeutic iPSC Clinical Trials
  • FIGURE 7.4: Non-Therapeutic Clinical Trials by Use
  • FIGURE 7.5: Percent Share of Diseases Targeted by Therapeutic Studies
  • FIGURE 7.6: Share of Autologous & Allogeneic iPSCs in Clinical Studies
  • FIGURE 7.7: iPSC Clinical Trials by Phase of Study
  • FIGURE 7.8: iPSC Clinical Trials by Funder Type
  • FIGURE 9.1: The Roles of OSKM Factors in the Induction of iPSCs
  • FIGURE 9.2: Delivery Methods for iPSC Induction
  • FIGURE 9.3: Schematic of Retroviral Delivery Method
  • FIGURE 9.4: Schematic of Lentiviral Delivery Method
  • FIGURE 9.5: Schematic of piggyBack Transposon Delivery Method
  • FIGURE 9.6: Shematic of Adenoviral Vector Delivery
  • FIGURE 9.7: oriP/Epstein-Barr Nuclear Antigen-1 (EBNA1)-based Episomes
  • FIGURE 9.8: RNA Delivery Approach
  • FIGURE 9.9: Protein Delivery
  • FIGURE 10.1: PubMed Citations for iPSCs and iPSC Lines registered in hPSCreg
  • FIGURE 10.1: Disease States represented by NIGMS Cell Lines
  • FIGURE 10.2: Subject Age Range in Collections
  • FIGURE 11.1: Biomedical Applications of iPSCs
  • FIGURE 11.1: Advantages of iPSC usage in Drug Discovery
  • FIGURE 11.2: iPSCs and their Potential for Toxicity Testing and Drug Screening
  • FIGURE 11.3: Relative Use of iPSC-Derived Cell Types used in Toxicity Testing Studies
  • FIGURE 11.4: Percent Share Utilization of iPSCs for Cardiovascular Disease Modeling
  • FIGURE 11.5: Techniques used for iPSC Bioprinting
  • FIGURE 12.1: Estimated Global Market for iPSCs by Geography, 2023-2030
  • FIGURE 12.2: Estimated Global Market for iPSCs by Technology, 2023-2030
  • FIGURE 12.3: Estimated Global Market for iPSCs by Biomedical Application, 2023-2030
  • FIGURE 12.4: Global Market for iPSCs by Derived Cell Type, 2023
  • FIGURE 13.1: dCas9-VPR System
  • FIGURE 13.2: Universal Donor Cell Technology
  • FIGURE 13.3: Century's Approach to iPSC Therapy
  • FIGURE 13.4: FT576
  • FIGURE 13.5: FT522
  • FIGURE 13.6: FT819
  • FIGURE 13.7: FT825
  • FIGURE 13.8: Developing iPSC Neurons by SynFire Technology
  • FIGURE 13.9: Mantarray Instrument
  • FIGURE 13.10: Cytostretcher
  • FIGURE 13.11: NanoSurface Plate
  • FIGURE 13.12: Repairon's Engineered Heart Muscle (EHM)
  • FIGURE 13.13: REPROCELL's Example Case Study: Alzheimer's Disese
  • FIGURE 13.14: Cardio quickPREDICT Process
  • FIGURE 13.15: devTOX quickPREDICT Process

INDEX OF TABLES

  • TABLE 3.1: Examples of Autologous iPSC-derived Cell Therapies in Development
  • TABLE 3.2: Examples of Clinical Trials involving Allogeneic iPSCs
  • TABLE 3.3: Commercially Available iPSC-derived Cell Types
  • TABLE 3.4: iPSC-derived Cell Types used in Clinical Trials
  • TABLE 4.1: Timeline of Important Milestones Reached in iPSC Industry
  • TABLE 5.1: No. of Research Publications on iPSC in PubMed.gov, 2006-June 1, 2024
  • TABLE 6.1: iPSC Patent Applications by Jurisdiction as of May 5, 2024
  • TABLE 6.2: Patent Applicants as of May 5, 2024
  • TABLE 6.3: iPSC Patent Inventors
  • TABLE 6.4: iPSC Patent Owners
  • TABLE 6.5: Legal Status of iPSC Patents
  • TABLE 7.1: Recruitment Status of iPSC Clinical Trials, May 2, 2024
  • TABLE 7.2: Examples of iPSC-based Therapeutic Interventional Studies
  • TABLE 7.3: The Promising iPSC-based Product Candidates Developed across the World
  • TABLE 7.4: Examples of Key iPSC-based Preclinical Studies
  • TABLE 8.1: M&A in iPSC Sector
  • TABLE 8.2: Partnership/Collaboration & Licensing Deals in iPSC Sector, 2021-May 2024
  • TABLE 8.3: Venture Capital Funding in iPSC Sector, 2021-May 2024
  • TABLE 9.1: Pluripotency-Associated Transcription Factors and their Functions
  • TABLE 9.2: Diffewrent Combinations of Factors for Different Cell Sources
  • TABLE 9.3: Comparison of Delivery Methods of Reprogramming Factors
  • TABLE 9.4: iPSC Disease Models Generated by CRISPR/Cas9
  • TABLE 9.5: Available iPSC lines and their Major Applications
  • TABLE 10.1: Major Biobanks Storing iPSCs & iPSC Lines
  • TABLE 10.2: Disease-Specific iPSCs offered by RIKEN
  • TABLE 10.3: Types of iPS Cell Lines available with WiCell - a Sample
  • TABLE 10.4: The Four California Institutions recruiting Tissue Donors
  • TABLE 10.5: iPSC Disease Samples with FCDI
  • TABLE 10.6: Examples of Allen's Fluorescently Tagged hiPSC lines
  • TABLE 10.7: Rett Syndrome Trust's iPSC Collection
  • TABLE 10.8: Cell Sources & Reprogramming Methods for iPSC Banks
  • TABLE 10.9: Ownership of iPSC Banks and the Investments Made
  • TABLE 11.1: Providers of iPSC-Related Services and Products for Researchers
  • TABLE 11.2: Drugs Tested for Cardiovascular Diseases using iPSCs
  • TABLE 11.3: Drugs Tested for Neurological Diseases using iPSC Lines
  • TABLE 11.4: Drugs Tested for Rare Diseases using iPSC Lines
  • TABLE 11.5: Examples of Drugs Tested for their Toxicity using iPSC-Derved Cell Lines
  • TABLE 11.6: Published Human iPSC Models
  • TABLE 11.7: Partial List of Cardiovascular & other Diseases Modeled using iPSCs
  • TABLE 11.8: Liver Diseases Modeled using iPSCs
  • TABLE 11.9: Examples of iPSC-Based Neurodegenerative Diseae Modeling
  • TABLE 11.10: Organoid Types and Diseae Modeling Applications
  • TABLE 11.11: Examples of Cancer-Derived iPSCs
  • TABLE 11.12: Major Sponsors of iPSC-based Cell Therapies
  • TABLE 11.13: Selected Interventional Clinical Trials of iPSC-Based Cell Therapy
  • TABLE 11.14: Companies focusing only on iPSC-based Therapies
  • TABLE 11.15: Features of Different iPSC Bioprinting Techniques
  • TABLE 11.16: Bioprinting of iPSC-Derived Cells
  • TABLE 11.17: iPSCs Generation from Cattle
  • TABLE 11.18: iPSCs Generation from Sheep
  • TABLE 11.19: iPSCs Generation from Goat
  • TABLE 11.20: iPSCs Generation from Buffalo
  • TABLE 11.21: iPSC Generation from Avians
  • TABLE 11.22: Timeline of Development of iPSCs Generated from Domestic & Wild Animals
  • TABLE 12.1: Estimated Global Market for iPSCs by Geography, 2023-2030
  • TABLE 12.2: Estimated Global Market for iPSCs by Technology, 2023-2030
  • TABLE 12.3: Estimated Global Market for iPSCs by Biomedical Application, 2023-2030
  • TABLE 12.4: Global Market for iPSCs by Derived Cell Type, 2023-2030
  • TABLE 13.1: Aspen's Clinical Pipeline
  • TABLE 13.2: Astella's Robust & Competitive Pipeline
  • TABLE 13.3: Bit.bio's Cell Therapy Pipeline
  • TABLE 13.4: BlueRock's Pipeline of Cell Therapy Products
  • TABLE 13.5: Cartheric's R&D Pipeline
  • TABLE 13.6: CellOrigin's R&D Pipeline
  • TABLE 13.7: Cellusion's Pipeline
  • TABLE 13.8: Century's Pipeline Products
  • TABLE 13.9: Cytovia's iPSC-Derived CAR-iNK Product Pipeline
  • TABLE 13.10: Eterna's R&D Pipeline
  • TABLE 13.11: Eyestem's Product Pipeline
  • TABLE 13.12: Fate Therapeutic's Product Pipeline
  • TABLE 13.13: Examples of Greenstone's iPSC Line Collections
  • TABLE 13.14: HebeCell's Product Pipeline
  • TABLE 13.15: Helio's Research & Development Status
  • TABLE 13.16: Hopstem's Product Pipeline
  • TABLE 13.17: IPS HEART's R&D Pipeline
  • TABLE 13.18: iPSirius' R&D Pipeline
  • TABLE 13.19: Kenai Therapeutic's Pipeline
  • TABLE 13.20: Khloris Biosciences' iPSC-Based Clinical Programs
  • TABLE 13.21: Laverock's R&D Pipeline
  • TABLE 13.22: Megakaryon's Research & Development Pipeline
  • TABLE 13.23: NEXEL Pipeline
  • TABLE 13.24: Notch Therapeutic's R&D Pipeline
  • TABLE 13.25: Available Stemgent iPSCs with REPROCELL
  • TABLE 13.26: Shinobi Therapeutics' Product Pipeline
  • TABLE 13.27: ViaCyte's Product Pipeline
  • TABLE 13.28: Vita Therapeutic's R&D Pipeline
Have a question?
Picture

Jeroen Van Heghe

Manager - EMEA

+32-2-535-7543

Picture

Christine Sirois

Manager - Americas

+1-860-674-8796

Questions? Please give us a call or visit the contact form.
Hi, how can we help?
Contact us!