Versatile Manufacturing Formats for Your Adherent Cell Therapy “Triathlon” & Its Bioprocess “Shoe Change”

Runner

Eureka! The glowing data speak for themselves; it’s time to graduate from HTS, petri dishes and mice, and time to help actual human patients in need. Your company or translational center wishes to join more than 1253 therapeutic developers across East Asia, Europe, or the USA, into which $23B has been invested to launch over 2000 clinical trials involved with advanced therapies (aka regenerative medicine). [1, 2] Given over 60 such therapies now on the market in nations monitored by the Alliance for Regenerative Medicine (ARM), your team would be entering a crowded race—not for the sprinter, but for the flexible and hardy endurance triathlete.

To cross the finish line, you’ll not only need sound strategy for years of trial design and management, but also for the cellular materials to manufacture (or directly comprise) the administered drug. Just as triathlon racers are seen in all shapes, sizes, and ages, your team will need a manufacturing process that best dovetails your capabilities and core strengths—and also, an idea of how you’ll need to “stretch.” [3, 4, 5] Bioprocess for advanced therapies involves many steps: tissue acquisition, cell isolation, cell expansion, harvest, volume reduction/washes, formulation & filling, QC & release, storage & inventory, and shipping/logistics. [6] Throughout these, testing of quality, potency, viability, and sterility is critical. [7] Although it’s important to control and document how each step is handled end-to-end, [8, 9] this blog will focus on cell expansion.

As any triathlete-in-training can tell you, “You get out of it what you put into it.” Like athletes, biomanufacturers also cross-train, breaking in their process “shoes” on liquids, machines, and solid surfaces. Hence, choice of versatile “upstream” cellular starting materials underlies an effective manufacturing plan. We’ve written extensively on the innate portability of hMSCs (human mesenchymal stromal/stem cells) into an assortment of expansion platforms from two-dimensional, planar growth (2D) to three-dimensional (3D) suspension growth on microcarriers. [10, 11, 12, 13]

MSCs are easy to grow and manipulate—and boast a proven track record of safety in more than 1500 posted clinical trials worldwide. [14] Regulatory-friendly, GMP-certified hMSC cell banks with defined donor characteristics can routinely and robustly expand from frozen vials into clinically relevant dose sizes when paired with a system of high-performance media, in less than 10 days. With some of these linked to Type II Drug Master Files (DMFs) from the FDA, [15, 16] simple, pre-validated bioprocess templates from standardized materials can allow a fast track to human doses, shortening development times from years to months. [17, 18]

Like a triathlete between two legs of the race, it’s often sensible—if not expected—for a bioprocess to “change shoes” after successfully completing one phase of competition. That is, some specialized cell expansion formats are more suited for manual and shorter duration needs (e.g., the “foot race”), whereas others are mechanically oriented and longer in length (e.g., the “bike race”).

To start the “foot race” with basic science or discovery phase R&D, hMSCs cells grow well in simple 2D tissue culture flasks for R&D purposes. We and our collaborators have found that they can be scaled up into 2D multi-stack flasks from Corning, [19] or scaled out into 96 well plates for bioassay under high content imaging systems. [20] Under our optimized growth conditions, no media change is required, and cells can be plated at low density to yield more than 100-fold expansion after a single passage. In terms of overall final cost, the media matters; high-performance media can achieve lower waste and substantially higher cells per liter. [21, 22, 23]

To zoom through the “cycling race” with clinically relevant doses of hMSCs for 1000s of patients with non-rare diseases and chronic health conditions, a trail to greatly increased CDMO capacity, cell volume, and operator bandwidth must be blazed. The transition from small scale, 2D flask-grown MSCs—e.g., for Phase I trials—to bioreactors for bigger patient cohorts is going to be all but necessary for full commercialization of some MSC therapeutics. [24, 25, 26] That is, when juggling batches of more than 10-20 billion cells, it becomes increasingly more practical and less costly to switch to a “3D” or bioreactor set-up.

A proverbial change to “bike shoes” may be necessary to deal with gears, wheels, and spin for the longer-distance portion of the cell manufacturing “triathlon”—i.e., later-Phase clinical trials and post-BLA distribution to patients. Fortunately, our team repeatedly demonstrates that hMSCs not only grow prolifically in standard 2D flasks and plasticware but also in different bioreactor 3D culture contexts. These platforms often involve the “cycling” of media—combining impellers, agitators, or wheels with cell suspension tanks and microcarrier growth substrates. We observe close comparability between MSC product made by either 2D or in “3D” bioreactors using the RoosterBio cell and media system. [10]

With our company’s cell and media system, we consistently achieve densities of more than 0.5 million cells per mL between scales of 0.1L (PBSmini) to 50L (PBS80) via PBS Biotech’s Vertical Wheel® technology. [27] This can translate to 31 billion cells per run in a 50L format, or enough for dozens of average-sized MSC doses for humans in an early-Phase clinical trial.  [28]  Sartorius—long known as a gold standard in the bioprocess industry for full-scale manufacture of adherent and non-adherent cells—offers a graded approach of options from small (<1L) or pilot scale (>1L) to giant (1000L+). Applied to Sartorius’ 3D suspension stirred-tank bioreactors, our team’s product systems have shown successful scaling between the small, modular Ambr®250 [29] and the larger, Biostat STR® 50L systems. [12] With >25B cells per lot attainable, linear scalability to the Biostat STR® 2000L system could potentially yield one trillion cells per run, sufficient for thousands of MSC doses. To accommodate liter-scale, closed-system production volumes, we and others offer GMP media in bagged formats.

MSCs are, by definition, plastic-adherent cells. [30] Thus, when grown “3D” in bioreactors, they still require a surface on which to attach and spread out, whether “floating” or fixed within a complex, compact matrix. Perhaps extending the triathlete analogy a bit too far, other cell expansion systems are much like running and cycling on a single pair of “shoes”—no change of format required. That is, cell numbers required for autologous cell therapy batches tend to be lower than for allogeneics. These starting materials from autologous sources can readily “run” through hollow-fiber perfusion systems that allow the operator to carefully fine-tune critical process parameters in small-scale development mode or at full-scale.

Work with our collaborators demonstrate successful hMSCs growth in hollow-fiber (HFB) perfusion systems from FiberCell [19] and the Quantum® cell culture cartridges from Terumo. [31] These systems enable high cells per volume culture of up to 2.1m2 of surface area, closed system automation, and constant input/output flux of nutrients for stable growth conditions. Although the hollow fiber technology does not yet reach the large scale of other bioreactors, these devices are already outstanding for extracorporeal cell therapies for human patients, with cell numbers that may approach 1B per cartridge. [32] Further, intriguing options can be explored to harness cell-secreted products in a continuous mode, instead of via batch productions.

Other single-use bioreactor formats with demonstration in hMSCs include fixed bed, hollow fiber, rotary cell culture systems (RCCS), rotating bed, or rocking motion (e.g., WAVE™) configurations, as recently expertly reviewed by Nogeuira, et al. (2021; see Table 1, below). [33] These platforms vary in scalability, degree of adoption by large instrument makers, clinical readiness, reusable componentry, and range of stresses leveraged on the expanding cell populations. To best match hMSCs to a new bioreactor system, some empirical trial and error may be involved to best optimize mixing forces, composition/coating, and porosity of microcarriers, media constituents, and origin of cell by donor or tissue type.  [11]

Table 1, below, adapted from by Nogeuira, et al. (2021) [33]

Bioreactor Type

Major Manufacturers Culture volume or area for MSCs, L or m2

Advantages

Stirred tank Eppendorf, EMD Millipore, Satorius Stedim

0.01 – 2000*L+
*prospective for MSCs, 50L demo’d

• Long-established industry know-how across diverse cell systems, including hMSCs
•  Wide range of scale from R&D to pilot to commercial
•  Predictable scalability of cell yield per volume scale
Vertical-Wheel PBS Biotech 0.1 – 70L •  Low shear stress & efficient mixing
•  Wide range of mfg. scales
•  Unique design avoids cell entrapment under impeller
Hollow fiber Fibercell, Terumo to 2000m2 •  Low shear stress
•  Very small footprint with high surface growth area
•  Very compatible with perfusion, extracorporeal, and allogeneic clinical applications
•  Disposable, single-use cartridges for closed systems
Fixed bed Pall/iCELLIS®,or misc. DIY (“do-it-yourself”) 0.003 – 1000L+ •  Low shear stress
•  Small footprint with high surface growth area
Rotary cell culture system DIY to 0.01L •  Low shear stress
•  No air bubbles
•  Simulates low-gravity for NASA/space!
Rotating bed Zellwerk GmbH to 0.6m2 •  Low shear stress
•  Small footprint with high surface growth area
•  Intermittent contact with medium and headspace
Rocking motion (e.g., “WAVE™”) Cytiva (WAVE™ Cellbag™) 0.05 – 0.6L •  Low shear stress & efficient mixing
•  Wide range of mfg. scales, and popular among cell biomanufacturers
•  No air bubbles
•  Very compatible with autologous cell therapies

 

hMSCs don’t just comprise the main starting material as ready-to-inject cellular therapeutics. They also facilitate product systems that are portable into processes to harvest their prolific secretomes, i.e., EVs (extracellular vesicles)/exosomes. [19, 34] At 15L pilot scale using the PBS system, we recently reported EV yields of 3.31 trillion EVs from the culture, or about 3000+ EVs per cell. Similarly, using Sartorius’ Ambr®250 scalable model for stirred tank systems, we’re presently observing ~5000 EVs produced per cell. [13] Assuming a stringent purification scheme that discards ~90% of the EVs during DSP, this productivity level alone could supply tens of doses to human patients for diverse clinical indications. [35]

In downstream processing (DSP) of biologicals, it’s rare that you can “have your cake and eat it too;” although purity and potency are closely related, more stringent purification can negatively correlate with yield. [36] Considering the challenges of downstream production with EVs, it’s prudent to ask where standard 2D flask culture of MSCs can fit into the scheme of human EV trial material. Recent modeling studies have argued that “when lot size is sufficiently large, EV harvest can become cheaper than cell expansion.” [37] Our empirical data that evince higher cell densities via 3D tend to bear this out. [38] Comparing a 2×10-layer CellStack process (> 3L) to 3D bioreactors, we’ve reported about 500M cells and 9.8×1011 EVs of media via this 2D approach. This is less efficient than the 3L Vertical Wheel system from PBS, which cranks out 1.5B cells and 6.0×1012 EVs. Efficiency is also less than what we recently observed with a stirred tank model system. [13] Nevertheless, if we consider time to be a scarcity compounded by equally scarce funds for new bioreactor capex, it’s certainly feasible to proceed quickly via 2D flasks for EV/exosome preclinical work, IND-enabling studies, and small-scale Phase I trials. We and our collaborators continue to improve our EV processes by facilitating improved downstream processing—and also improved productivity of EVs on a per-cell basis. [39]

It’s not just how you grow hMSCs that matters, it’s what you do with them! Why not use them in a form that requires as little upstream effort as possible, e.g., for bioprinting? [40] Large, cost-efficient, post-expansion volumes of 50M cell vials may be prepared that are “ready to print” to use for various bioinks for tissue engineering applications, [24, 41] such as the CellInk BioX instrument series. [42, 43]

Much like bio-“3DP,” genetic engineering of hMSCs unlocks a whole new universe of preclinical and clinical possibilities. With advancing gene transfer and genome editing techs, each cell can be engineered into a micro-delivery system for a genetic medicine. To enhance transduction [44, 45] or transfection [46, 47] of these, complete media that easily “drops-in” one’s scalable bioprocess may be necessary, given that hMSCs and other primary cells are less amenable to conventional gene transfer modalities than transformed cell lines.

Asked publicly whether it’s masochism or motivation that inspires the rare triathlete or advanced therapeutics developer—and they’ll probably answer with something like “It’s just what we do…the easy road of mediocrity is simply more tedious to bear than the hard road toward excellence.” Privately, however, both know that it’s much easier to stay motivated (and less injured) by bringing the right tools into the right leg of the journey. Whether it’s the perfect shoe, or the lightest bike frame—or the best-fitting cell expansion bioprocess—the race can be open to nearly all, and there are hundreds of ways to win.

 

References
  1. Regenerative Medicine, Alliance for, REGENERATIVE MEDICINE: DISRUPTING THE STATUS QUO. 2022: https://alliancerm.org/.
  2. Mason, C. and P. Dunnill, A brief definition of regenerative medicine. Regen Med, 2008. 3(1): p. 1-5. 10.2217/17460751.3.1.1
  3. Lembong, Josephine and Rowley, Jon. Building Effective Multi-Year Process Development Programs I: Estimating hMSC Lot Size Ranges for Clinical Manufacturing Through Commercial Demand. RoosterBio Blog 2021; Available from: https://www.roosterbio.com/blog/building-effective-multi-year-process-development-programs-i/.
  4. Lembong, Josephine and Rowley, Jon. Building Effective Multi-Year Process Development Programs II: Evolution of Technology Platform Decisions Based on Lot Size. RoosterBio Blog 2021; Available from: https://www.roosterbio.com/blog/building-effective-multi-year-process-development-programs-ii-evolution-of-technology-platform-decisions-based-on-lot-size/.
  5. RoosterBio. “One Does Not Simply Walk Into” a Therapeutic Cell Manufacturing Process… But the Long Road Need Not Be Perilous! RoosterBio Blog 2021; Available from: https://www.roosterbio.com/blog/one-does-not-simply-walk-into-a-therapeutic-cell-manufacturing-process-but-the-long-road-need-not-be-perilous/.
  6. Carmen, Jessica, et al., Cell therapy manufacturing: identifying and meeting demand, in Stem Cells in Regenerative Medicine. 2015. p. 49-68. 10.1002/9781118846193.ch4
  7. Guadix, J. A., et al., Principal Criteria for Evaluating the Quality, Safety and Efficacy of hMSC-Based Products in Clinical Practice: Current Approaches and Challenges. Pharmaceutics, 2019. 11(11). 10.3390/pharmaceutics11110552
  8. RoosterBio. Good Manufacturing Practice for Cell & Cell-Based Therapies: Facilities & Quality Control. RoosterBio Blog 2017; Available from: https://www.roosterbio.com/blog/good-manufacturing-practice-for-cell-and-cell-based-therapies-facilities-quality-control/.
  9. Williams, Kathy, and Hansen, Caitlin. Quality Begins at Inception. RoosterBio Blog 2020; Available from: https://www.roosterbio.com/blog/quality-begins-at-inception/.
  10. Adlerz, K., et al., A scalable and xeno-free bioreactor system for biomanufacturing of hUC-MSCs. Cytotherapy, 2020. 22(5, Supplement): p. S46-S47. https://doi.org/10.1016/j.jcyt.2020.03.052
  11. Kirian, Robert. Maintaining CQAs as Manufacturing Processes are Scaled from 2D to 3D Bioreactor Culture. RoosterBio Blog 2022; Available from: https://www.roosterbio.com/blog/maintaining-cqas-as-manufacturing-processes-are-scaled-from-2d-to-3d-bioreactor-culture/.
  12. Kirian, R. D., et al., SCALE-UP OF A XENO-FREE FED-BATCH STIRRED-TANK BIOREACTOR PROCESS FOR HMSC MANUFACTURING. Cytotherapy, 2022. 24(5): p. S71. 10.1016/S1465-3249(22)00219-5
  13. Lenzini, S., et al., DEVELOPING A MICROCARRIER STIRRED TANK PROCESS FOR LARGE-SCALE HMSC-EV PRODUCTION. Cytotherapy, 2022. 24(5): p. S91.
  14. MSC Trials 2011-2020. celltrials.org 2011-2022; Available from: https://celltrials.org/public-cells-data/msc-trials-2011-2020/65.
  15. PRWeb, CISION. RoosterBio Submits U.S. FDA Master Files for CliniControl™ Line of Human Mesenchymal Stem/Stromal Cell Products. prweb.com 2018; Available from: https://www.prweb.com/releases/roosterbio_submits_u_s_fda_master_files_for_clinicontrol_line_of_human_mesenchymal_stem_stromal_cell_products/prweb15909351.htm.
  16. PRWeb, CISION. First Patient Dosed in Phase I Clinical Trial using RoosterBio’s Cellular Starting Materials. 2020; Available from: https://www.prweb.com/releases/first_patient_dosed_in_phase_i_clinical_trial_using_roosterbios_cellular_starting_materials/prweb16955951.htm.
  17. Alvaro, David and Patel, Ashish. Accelerating the Development of a Novel Cell Therapy for COVID-19–Associated Lung Fibrosis. Pharma’s Almanac 2021; Available from: https://www.pharmasalmanac.com/articles/accelerating-the-development-of-a-novel-cell-therapy-for-covid-19-associated-lung-fibrosis.
  18. Patel, Ashish and Candiello, Joseph. Rapid translation of a cellular therapeutic: Choosing product & process development solutions for clinical manufacturing readiness. Cell & Gene Therapy Insights 2022; Available from: https://www.insights.bio/cell-and-gene-therapy-insights/webinars/291/Rapid-translation-of-a-cellular-therapeutic-Choosing-product-process-development-solutions-for-clinical-manufacturing-readiness.
  19. Gobin, J., et al., Hollow-fiber bioreactor production of extracellular vesicles from human bone marrow mesenchymal stromal cells yields nanovesicles that mirrors the immuno-modulatory antigenic signature of the producer cell. Stem Cell Res Ther, 2021. 12(1): p. 127. 10.1186/s13287-021-02190-3
  20. Andrews, Seth H., et al., Morphological Landscapes from High Content Imaging Identify Optimal Priming Strategies that Enhance MSC Immunosuppression. bioRxiv, 2021: p. 2021.02.23.432501. 10.1101/2021.02.23.432501
  21. Rowley, Jon, et al., Meeting lot-size challenges of manufacturing adherent cells for therapy.
  22. Pereira Chilima, Tania D., Fabien Moncaubeig, and Suzanne S. Farid, Impact of allogeneic stem cell manufacturing decisions on cost of goods, process robustness and reimbursement. Biochemical Engineering Journal, 2018. 137: p. 132-151. https://doi.org/10.1016/j.bej.2018.04.017
  23. Omokhowa Agbojo, Maya Lim, & Josephine Lembong. Environmental Analysis of Therapeutic hMSC Manufacturing: A Comparison of Multiple Bioprocess Systems. RoosterBio Blog 2021; Available from: https://www.roosterbio.com/blog/environmental-analysis-of-therapeutic-hmsc-manufacturing-a-comparison-of-multiple-bioprocess-systems/.
  24. Lim, Mayasari. Cells as Bioinks for 3D Bioprinting. RoosterBio Blog 2019; Available from: https://www.roosterbio.com/blog/cells-as-bioinks-for-3d-bioprinting/.
  25. Carson, Jon; Farrance, Ian; Getz, John; Rowley, Jon. Scalable MSC Manufacturing Matters in a Rapid Response to COVID-19. RoosterBio Blog 2020; Available from: https://www.roosterbio.com/blog/scalable-msc-manufacturing-matters-in-a-rapid-response-to-covid-19/.
  26. Olsen, T. R., et al., Peak MSC-Are We There Yet? Front Med (Lausanne), 2018. 5: p. 178. 10.3389/fmed.2018.00178
  27. Lembong, J., et al., Bioreactor Parameters for Microcarrier-Based Human MSC Expansion under Xeno-Free Conditions in a Vertical-Wheel System. Bioengineering (Basel), 2020. 7(3). 10.3390/bioengineering7030073
  28. Kirian, RD, et al., Scaling a xeno-free fed-batch microcarrier suspension bioreactor system from development to production scale for manufacturing XF hMSCs. Cytotherapy, 2019. 21(5): p. S71-S72.
  29. Rotondi, M., et al., Design and development of a new ambr250(R) bioreactor vessel for improved cell and gene therapy applications. Biotechnol Lett, 2021. 43(5): p. 1103-1116. 10.1007/s10529-021-03076-3
  30. Dominici, M., et al., Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy, 2006. 8(4): p. 315-7. 10.1080/14653240600855905
  31. Potter, D. R., et al., Mesenchymal stem cell-derived extracellular vesicles attenuate pulmonary vascular permeability and lung injury induced by hemorrhagic shock and trauma. J Trauma Acute Care Surg, 2018. 84(2): p. 245-256. 10.1097/TA.0000000000001744
  32. Allen, A., et al., Mesenchymal Stromal Cell Bioreactor for Ex Vivo Reprogramming of Human Immune Cells. Sci Rep, 2020. 10(1): p. 10142. 10.1038/s41598-020-67039-w
  33. Nogueira, D. E. S., J. M. S. Cabral, and C. A. V. Rodrigues, Single-Use Bioreactors for Human Pluripotent and Adult Stem Cells: Towards Regenerative Medicine Applications. Bioengineering (Basel), 2021. 8(5). 10.3390/bioengineering8050068
  34. Adlerz, Katrina, et al., Comparison of msc-evs manufatured in 2D versus scalable 3D bioreactor systems. Cytotherapy, 2019. 21(5): p. S58.
  35. Lenzini, Stephen. Establishing a Working Range for Effective MSC-EV Dose. RoosterBio Blog 2021; Available from: https://www.roosterbio.com/blog/establishing-a-working-range-for-effective-msc-ev-dose/.
  36. Mateescu, B., et al., Obstacles and opportunities in the functional analysis of extracellular vesicle RNA – an ISEV position paper. J Extracell Vesicles, 2017. 6(1): p. 1286095. 10.1080/20013078.2017.1286095
  37. Ng, K. S., et al., Bioprocess decision support tool for scalable manufacture of extracellular vesicles. Biotechnol Bioeng, 2019. 116(2): p. 307-319. 10.1002/bit.26809
  38. Lenzini, Stephen. Extracellular Vesicle/Exosome Upstream Process Development: Maximizing Productivity to Accelerate Clinical Adoption. RoosterBio Blog 2022; Available from: https://www.roosterbio.com/blog/extracellular-vesicle-exosome-upstream-process-development-maximizing-productivity-to-accelerate-clinical-adoption/.
  39. Scheiber, A. L., et al., Culture Condition of Bone Marrow Stromal Cells Affects Quantity and Quality of the Extracellular Vesicles. Int J Mol Sci, 2022. 23(3). 10.3390/ijms23031017
  40. Ayan, B., et al., Aspiration-assisted bioprinting for precise positioning of biologics. Sci Adv, 2020. 6(10): p. eaaw5111. 10.1126/sciadv.aaw5111
  41. Soliman, B. G., et al., Development and Characterization of Gelatin-Norbornene Bioink to Understand the Interplay between Physical Architecture and Micro-Capillary Formation in Biofabricated Vascularized Constructs. Adv Healthc Mater, 2022. 11(2): p. e2101873. 10.1002/adhm.202101873
  42. PRWeb, CISION. RoosterBio Inc. Launches Industry’s First Ready-to-Print Stem Cell Product. 2015; Available from: http://www.prweb.com/releases/2015/10/prweb13027227.htm.
  43. Apelgren, P., et al., Chondrocytes and stem cells in 3D-bioprinted structures create human cartilage in vivo. PLoS One, 2017. 12(12): p. e0189428. 10.1371/journal.pone.0189428
  44. Willstaedt, Terri. Genetically Engineered Mesenchymal Stromal Cells (MSCs): A Promising Tool for the Successful Delivery of Therapeutic Genes. RoosterBio Blog 2021; Available from: https://www.roosterbio.com/blog/genetically-engineered-mesenchymal-stromal-cells-a-promising-tool-for-the-successful-delivery-of-therapeutic-genes/.
  45. Willstaedt, TM, et al., Development of an optimized lentiviral transduction medium and process to manufacture genetically modified MSC working cell banks. Cytotherapy, 2021. 23(5): p. S45.
  46. Doolin, M., Willstaedt, T., Rowley, J. Optimization of Transient Transfection of Mesenchymal Stromal Cells with Plasmid DNA or Messenger RNA. 2022; Available from: https://annualmeeting.asgct.org/abstracts/abstract-details?abstractId=6086.
  47. RoosterBio. RoosterGEM™ Expands into New Fields of “Cell-Grow-Culture” for mRNA Transfection. RoosterBio Blog 2022; Available from: https://www.roosterbio.com/blog/roostergem-expands-into-new-fields-of-cell-grow-culture-for-mrna-transfection/.

RoosterBio is fueling the rapid implementation of scalable advanced therapies. Contact us to discuss how we can accelerate your product & process development. Follow us on LinkedIn for more educational resources just like this.