December 15, 2018

Building Effective Multi-Year Process Development Programs: Evolution of Technology Platform Decisions Based on Lot Size

Authored by Josephine Lembong, Ph.D., Scientist, Product & Process Development & Jon Rowley, Ph.D., Founder and Chief Product Officer, RoosterBio Inc.


Previously, we published a blog post on estimating hMSC lot sizes for clinical manufacturing, with the goal of outlining a development program that could tailor accordingly. This exercise is crucial because the calculated target cell lot size dictates the final production platform needed for your therapeutic product. The next step would be to determine the appropriate manufacturing platform, for each unit operation, that will meet the calculated hMSC lot sizes throughout clinical development. Having a solid, multi-year plan will help your company succeed at navigating this complex maze that is the path to market success.

“It is the technologist’s and engineer’s jobs to drive the technology platform decision making process”

The final decision of production platforms can be overwhelming; even though there is a certain goal in mind for the present time, you do want to keep it flexible and scalable for other potential applications in the future. There is also the goal of managing through the “Comparability Challenges” as these changes are implemented. Adding to the complexity is that as the RegenMed industry grows, the technology providers of cell processing platforms across the various unit operations seems to be increasing in a fractal nature, with little standardization across devices. These technologies (e.g. bioreactors, continuous centrifuges, fill finish/controlled freezing, and other automation platforms) are significant investments to the company in the form of cost and time. It is the technologist’s and engineer’s jobs to drive the technology platform decision making process by derisking these technologies and establishing a multi-year development program, all while determining the costs associated with the program, and communicating these needs to the company’s business team so that they can raise the needed capital for these programs over time.

The goal of this post is to lay out the various scales of hMSC production and highlight the existing technology platforms for the different unit operations involved in the manufacturing process. This will help define the requirements that will guide the company’s multi-year process development program to meet projected future lot sizes.

Each phase of a clinical trial is associated with a specific production scale, which dictates the production platform

At the end of the last post, we arrived at the following estimated lot sizes based on a set of assumptions: 525 billion viable hMSCs per final commercial manufacturing lot, assuming a mid-range dose for a cardiac indication aimed to treat 100,000 patients per year, with a relatively safe, conservative assumptions regarding losses in cell viability and recovery during every step of the production process. Assuming a go-to-market lot size of 20% of the full commercial scale, we estimated that one could target a 100B cell lot size for Phase III, a 25B cell lot size for Phase II, and potentially a 10B cell lot size for a Phase I trial. These are simply guidelines that will change based on assumptions, but we recommend everyone go through this exercise (as outlined here) for each therapeutic program.




November 17, 2018

RoosterBio Products Continue to Expand Presence in Major Mesenchymal Stem/Stromal Cell (MSC)-related Regenerative Medicine Peer Reviewed Publications

Authored by Joseph Candiello, Ph.D., Technical Application Specialist

Figure 1. Current and emerging applications using hMSCs as a critical raw material.

Human mesenchymal stem/stromal cells (hMSCs) are a critical starting material in a growing variety of established and emerging applications spanning the Regenerative Medicine space (Fig1). Their unique balance of functional bioactive attributes, expansion potential, and established safety profile have resulted in a steady increase in both scientific publications and a parallel increased presence in clinical applications over the past 10+ years. Specific to scientific publications, from 2011 to 2015 there were over 17,000 MSC related publications, with over 50% pertaining to human MSC use. This trend has continued with over 9,600 MSC related publications in 2016-2017 and with over 5,000 articles using human hMSCs.

We are excited that over the past 4 years RoosterBio’s core technology, hMSC cell banks and associated Bioprocess Media Systems have been a used in an increasing number of these peer reviewed scientific publications or what we affectionally refer to as our customer’s “brainy stuff”. The initial articles using RoosterBio technology were published in Biomacromolecules and Cytotherapy in the 2nd half of 2015; less than 2 years from when our first products left RoosterBio’s doors. During 2016 and 2017, RoosterBio products have appeared in leading academic journals such as PNAS (twice), Biomaterials, Stem Cells and Translational Medicine, American Journal of Respiratory and Critical Care Medicine, and Nucleic Acids Research.
Figure 2: Key Metrics of Publications using RoosterBio products.

In addition to a presence in these high-profile publications, RoosterBio's hMSC manufacturing-focused products, have had a steady yearly increase in overall total peer reviewed articles in print (Fig. 2). In 2018 to date a total of 35 articles have included RBI products, surpassing 2017’s total and doubling 2016’s output. In addition to total publications, we have tracked articles using a few key metrics – Journal’s Impact Factor (IF), an index of citations per publication, and studies which contain an in vivo, or animal model component (Fig 2). With respect to IF, articles using RBI product have had an increase in average IF from 4.4 in 2016 to 5.2 in 2018 and total number of publications with an IF >4 from 5 articles in 2015 to 17 to date in 2018, which is almost half of the total articles this year. Original research articles with an in vivo component have had similar increases, from 2 articles (12%) in 2015 to 10 articles (28%) so far in 2018.

The breadth of topics included in these publications is as diverse as current applications across the hMSC Regenerative Medicine space – and is called out in our databaseof current RoosterBio product publications. As frequency of publications increases, we will highlight some of these – starting with some recent exciting work being reported across the industry:
As RoosterBio continues to provide hMSC Bioprocess Systems to support Regenerative Medicine research and product development efforts; the central focus is on how we can radically simplify our customers workflow. To this end, across the life sciences industry, timeframes from project initiation to first publication continue to increase; with a current average of 3 to 4 years. At the same time, it is critical for researchers and product developers to shorten both publication and product development timeframes. RoosterBio high volume hMSCs and bioprocess media systems are uniquely designed to accelerate these timeframes by providing well characterized, consistent raw materials designed for simplicity, reduced cost, and shortened time to generate the necessary cellular components for research, development, or clinical translation.

Highlighted Publications:

Defining Hydrogel Properties to Instruct Lineage- and Cell-Specific Mesenchymal Differentiation. Hung BP, Harvestine JN, Saiz AM, Gonzalez-Fernandez T, Sahar DE, Weiss ML, Leach JK, Biomaterials, 2019.

IFN-γ and TNF-α Pre-licensing Protects Mesenchymal Stromal Cells from the Pro-inflammatory Effects of Palmitate. Boland L, Burand AJ, Brown AJ, Boyt D, Lira VA, Ankrum JA. Mol Ther. 2018.

Deciphering the role of substrate stiffness in enhancing the internalization efficiency of plasmid DNA in stem cells using lipid-based nanocarriers. Modaresi S, Pacelli S, Whitlow J, Paul A. Nanoscale. 2018.

Acoustophoretic printing. Foresti D, Kroll KT, Amissah R, Sillani F, Homan KA, Poulikakos D, Lewis JA. Sci Adv. 2018.

3D printed biofunctionalized scaffolds for microfracture repair of cartilage defects. Guo T, Noshin M, Baker HB, Taskoy E, Meredith SJ, Tang Q, Ringel JP, Lerman MJ, Chen Y, Packer JD, Fisher JP. Biomaterials. 2018.

Mesenchymal stem cell-derived extracellular vesicles attenuate pulmonary vascular permeability and lung injury induced by hemorrhagic shock and trauma. Potter DR, Miyazawa BY, Gibb SL, Deng X, Togaratti PP, Croze RH, Srivastava AK, Trivedi A, Matthay M, Holcomb JB, Schreiber MA, Pati S. J Trauma Acute Care Surg. 2018.

If you are interested in having a conversation about how RoosterBio can shorten your gaps between experiments and accelerate your time to publication, contact us at info@roosterbio.com.


September 12, 2018

Building Effective Multi-Year Process Development Programs: Estimating hMSC Lot Size Ranges for Clinical Manufacturing through Commercial Demand – it is all about the assumptions

Authored by Josephine Lembong, Ph.D., Scientist, Product & Process Development & Jon A. Rowley, Ph.D., Founder and Chief Product Officer, RoosterBio Inc.

Jon Rowley, RoosterBio’s Founder and Chief Product Officer, gave a talk at ISCT 2018 in May and had a set of slides about estimating lot size that had a lot of people scrambling to take notes. We thought we would share the content here at the RoosterBio blog with more discussion and make the content broadly available and open for discussion.  

The concept is consistent with all good strategic planning; 
  1. Understand what the future looks like and work backwards,
  2. Create a model using reasonable to conservative assumptions along the way to estimate a range of needs,
  3. Use this model to lay out what the next several years will look like assuming successful achievement of intermediate milestones, and create multi-year programs that are right-sized, with the right technology platform, for the stage of product / clinical development of your company.
This is the type of consultation that we provide in our Process Design and Acceleration business unit at RoosterBio, which we offer to our customers that are incorporating RoosterBio hMSC cell banks and bioprocess media systems into their product and process development efforts. However, we love to share our knowledge with the broader RegenMed Industry and are offering it up here.

Notes to readers: this exercise has the base assumption that this is an Allogenic, Universal Donor manufacturing process [1] and is not applicable to Autologous or patient-specific products. The core logic would still hold when applied to autologous, but at a much smaller scale.

Lot Size Targets Dictate the Production Platform 

The goal of any process development program is to create a right-sized manufacturing process for your immediate business goals, but with future scalability in mind. Understanding the future lot size requirements will help strategically align manufacturing requirements today with commercial scalability while laying a platform foundation to minimize comparability risks.

First Start with the End in Mind – Engage with Marketing

Get with the business team within your company (usually Strategic Marketing and/or Commercial Operations) and request a numbers-driven discussion on which of the multiple indications that a process development program should be built for. There are usually several therapeutic indications of interest, but it is important to realize that a manufacturing process for an ocular indication that has a dose of 1 million cells/dose is very different than a Crohn's disease indication that has a dose size of greater than 1 billion cells/dose [2]. Each therapeutic indication from a company must be treated as a distinct product and target, with a distinct process. Just because the same cell type is used in both therapeutic products does not mean they share a manufacturing process. This is critical to bring clarity to each program.

“A common pitfall that many cell therapy companies fall into is that each therapeutic indication is not addressed as a unique product with a unique manufacturing process”

Once the team is aligned on the target indication (or set of prioritized target indications), then you need to understand what a reasonable peak commercial market demand would be – this is likely to be found in the business model projections that are built for investors and is a good place to start. If there are 500,000 patients a year in your target indication and the business is aiming to capture 20% of them (or treat 100,000 patients/year) at peak commercial demand, then you need a process that will scale with that over time, assuming both clinical and market success. It is a good idea to establish a range. We often like to perform this exercise with a low, middle and high assumptions (such as 30,000 patients as low, 100,000 as the target, and 250,000 patients treated/year if wildly successful). For this article, we will simply focus on the '100,000 patients treated assumption' for our calculations.

Dosing Assumptions: this is one of the hardest parts

Estimating dosing is sometimes the most difficult part. Does the patient need a single dose or multiple doses, and how many cells per dose? In many cases a dose escalation trial has not yet been performed, so some educated guesstimates are required. Taking a low/mid/high range of estimates also works here, and there is sufficient published work related to hMSCs in different indications that you can get close enough for these purposes [2,3]. For this example, we will assume a cardiac indication. There is a good amount of published work on hMSCs for cardiac indication and we can assume a conservative low dose of 25 million cells, a mid-range dose of 50 million, and a high dose of 100 million [4,5]. In this blog post we will perform all calculations with an assumption of 50 million cells per dose.

Calculating Yearly Product Needs at Peak Commercial Demand
  • At peak demand, we aim to produce 100,000 doses per year, with a size of 50 million cells per dose.
  • 100,000 doses per year ≈ 2,000 doses per week
    • Weekly production is not recommended – it is simply unsustainable, so let’s assume one (1) lot production every two (2) weeks, or 25 lots total in a year (which is still a lot for any manufacturing site)
    • Not all lots will be successful – assume a 10% scrap rate, thus 2-3 lots are lost per year, giving us 22-23 lots per year
    • 100,000 doses per year / ~23 lots per year = 4,450 doses/lot 
    • Assume 10% of lot goes to testing (~450 dose), so a final target lot size of 4,900 doses/lot will be needed to be manufactured at peak commercial demand
It is important to remember that it typically takes multiple years for a successful therapeutic to reach peak demand, so it is not necessary to go to market with a manufacturing process capable of meeting peak demand – especially for an early stage field like Regenerative Medicine. We will create estimates for peak demand, but then focus on building a reasonable “go-to-market” lot size and manufacturing process.

Calculating Total Cells to Manufacture that are Required to Fill into the Final Container
  • We will assume 1 dose goes into one container.
  • We assumed a mid-range dose size of 50 million (i.e. 50 million viable cells per vial, post-thaw).
  • Cell Count is a strict QC parameter with a straight forward specification. It is important to point out that often there is a solid safety factor, or overfill, applied to this metric. You never want to fail a lot on cell count after you spent hundreds of thousands of dollars creating the units – so the risk averse will overfill vials until sampling and cell enumeration is an accurate, consistent, and precise method.
    • Assume 15% (worst case) viability drop and 20% recovery loss during cryopreservation (these assumptions should be data driven from your process, and constantly trended and updated during manufacturing).
    • Based on the above assumptions, to consistently achieve 50 million viable cells after thaw, it is required to target filling at 75 million viable cells into the final container:
      • 75 million viable cells – 15 million (20% total cell loss on recovery) = 60 million viable cells 
      • 60 million – 9 million (15% viability drop) = 51 million viable cell target specification
  • NOTE: some programs will only overfill by 10-20%, but we find that with this range it is very difficult to routinely obtain successful cell counts in a manufacturing/QC environment, so we recommend 30-50% overfill for calculations.

Calculating Lot Size Requirements at Peak Commercial Demand

One unfortunate fact of cell manufacturing is that you need to manufacture more cells than you need to fill, since there will be losses associated with the post-harvest processing steps. Many biological manufacturing processes have losses in the 40%-60% range in this “downstream” processing.  For our process, we go through the following math:
  • 75 million cells/dose × 4900 doses per manufacturing lot = 368 billion viable cells needed to target 4900 doses from every manufacturing lot.
  • We assume 30% post-harvest cell loss during processing for this exercise – thus at least 525 billion cells need to be manufactured in order to have 368 billion cells during the fill unit operation.
    • 525 billion viable cells – 157 billion (30% post-harvest cell loss) = 368 billion viable cells 
  • 30% cell loss does seem significant, however every process development group will have a team working to optimize their process to minimize this number.  It is likely that recoveries from earlier processes will be much greater than 30%, and with optimization at large scale it is possible to improve.  For modeling, it is always worth using reasonable to conservative estimates.
In conclusion, we arrived at the following numbers:
  • At peak commercial demand, 23 successful lots of 525 billion cells per lot need to be manufactured in order to meet the demand, assuming a mid-range dose for a cardiac indication aimed to treat 100,000 patients per year. 
This calculation does not take into account further downstream losses associated with stability, multiple distribution sites, safety stocks – which will just make the numbers worse. For this exercise, we will stop here. The 525 billion viable cells per lot is the number we were looking for, and it is this number that will drive the production platform decisions for streamlined hMSC clinical manufacturing.

For an early-stage RegenMed company, a go-to-market lot size of ~20% of the “peak commercial scale” is a reasonable target to plan for, so a ~100 billion cells/lot would be a good target number to develop and run your Phase III trial at.

We’d like to summarize this post with a few number recommendations:


Phase III/Go-to-Market: ~100B cells

Phase II (100-200 patients over 3-4 lots): 15-25B cells – this is an intermediate step up from Phase I and within a log of the Phase III/go-to-market process.

Phase I (small scale): 5-10B cells

The next stage is to lay out the technology platforms for the various manufacturing unit operations that are capable of processing these cell numbers. This will be the topic of our next blog post.

References:
  1. Simaria et al., 2013. Allogeneic cell therapy bioprocess economics and optimization: Single‐use cell expansion technologies. Biotechnol Bioeng 111(1): 69-83. doi: 10.1002/bit.25008 
  2. Olsen et al., 2018. Peak MSC—Are We There Yet? Front Med 5:178. doi: 10.3389/fmed.2018.00178 
  3. Squillaro et al., 2016. Clinical Trials With Mesenchymal Stem Cells: An Update. Cell Transplant 25(5):829-848. doi: 10.3727/096368915X689622 
  4. Majka et al., 2017. Concise Review: Mesenchymal Stem Cells in Cardiovascular Regeneration: Emerging Research Directions and Clinical Applications. Stem Cells Transl Med 6(10):1859-1867. doi: 10.1002/sctm.16-0484 
  5. Golpanian et al., 2016. Concise Review: Review and Perspective of Cell Dosage and Routes of Administration From Preclinical and Clinical Studies of Stem Cell Therapy for Heart Disease. Stem Cells Transl Med 5(2):186-191. doi: 10.5966/sctm.2015-0101




May 16, 2018

ISCT 2018: MSC Biomanufacturing, Bioprocessing, Scale-Up, Analytics and Exosome Production Take Center Stage



Authored by Katrina Adlerz, Ph.D. Scientist, Analytical Development, RoosterBio Inc.

RoosterBio attended the International Society for Cell Therapy annual meeting, held May 2-5, 2018 in Montreal, which brought together leaders in the field including academic researchers, industry scientists, regulators, and clinicians. The society and conference focus on three key areas of translation: Academia, Regulatory, and Commercialization.

Six Roosters attended to hear the latest research and translational innovations at the scientific talks, present three different posters, and man the booth in the exhibit hall. Jon Rowley, founder and CTO of RoosterBio, also gave two talks discussing innovations to accelerate MSC Biomanufacturing, Bioprocessing and Scale-Up. He shared insights from RoosterBio’s path to GMP-manufactured cells and media as well as strategies for overcoming obstacles in his talk “Technologies for Radically Reducing Development Timelines of hMSC-based Therapeutic Products” during the Strategies for Commercialization Session. (New to MSCs? Read more about MSCs and biomanufacturing here, herehere and here.) For a copy of Jon's talk, email us.

RoosterBio Analytical, Process & Product Development efforts were well-represented with three posters.

- “A Xeno-Free Fed-Batch Microcarrier Suspension Bioreactor System for the Scalable and Economic Expansion of hBM-MSCs” showed that MSC critical quality attributes were maintained in 0.1L and 3L bioreactor culture. Poster.
- “Scalable Xeno-Free Manufacturing of Extracellular Vesicles Derived from Human Mesenchymal/Stromal Stem Cells” explained a process for generating a high yield of EVs/Exosomes from MSCs in a shortened time frame using RoosterNourishTM-MSC-XF Media. Poster.
- “Development & Technology Transfer of a cGMP Potency Assay: Testing of an Ancillary Material for Stem Cell Manufacturing” outlined the steps in assay development, assay qualification/validation, and tech transfer for a custom potency assay based on cell expansion. Poster.
- In addition, the RoosterBio , BioLife Solutions and Brooks Life Science teams collaboratively presented a poster on MSC cryopreservation: "The Effect of Cryomedia Selection and Transient Warming Events on Post-Cryopreservation Human MSC Function". Poster. 

The posters gave presenting scientists the opportunity to talk with those doing similar work in process or assay development. It allowed us to learn from and share our expertise with the community.

The conference kicked off with a RoosterBio-sponsored workshop: Improving Mesenchymal Stem Cell Potency and Survival. Steven Bauer, Branch Chief of the Cellular and Tissue Therapy Branch  of the US FDA, and Head of the FDA's MSC Consortium, discussed his innovative work developing predictive assays for MSC potency by analyzing cell morphology in his talk “High Throughput Approaches to Assess MSC Function”.  You can find his blog here.  There were also a number of talks discussing clinical trials results. A common theme of the session was the need to develop analytical methods and assays that can predict MSC-based treatment efficacy in patients.