SlideShare a Scribd company logo
1 of 4
Download to read offline
10
Cell-BasedTherapies
April 2014
Back to
the Future
The explosion in cell-based therapies has led to new
technologies to streamline and standardise their
commercial production. These tools could now be
used back in the lab to help push the frontiers in
biomedical research
Over the last decade,cell-based therapies
have garnered tremendous excitement
from researchers,clinicians and the
public.These therapies represent the
next frontier in drug development.Some
cross over into the field of personalised
medicine in that they are derived from
a patient’s own cells,or they are tailored
according to what will be optimal for
different patient subsets.
With fewer than 40 cell-based therapies
in commercial distribution in regulated
markets – only half of which have been
formally approved – and only a handful
making over $100,000 in annual revenue,
the field is still relatively immature,but
enjoying a steep growth trajectory (1).
LatestWave
The pipeline of therapies is maturing,with
more than 300 cell therapies currently in
development.The latest wave has begun
to prove successful in the clinic,with
particular attention being paid,of late,
to cell-based immunotherapies (2).This
was initially triggered by Dendreon’s
Provenge approval in the US (2010) and
Europe (2013).Recent deals made by
Novartis,Celgene and through venture
capital investment illustrate that early-
stage cell-based immunotherapies
continue to gain momentum (see
Figure 1,page 12).
Moreover,the explosion of cell-based
therapies is having a knock-on effect
throughout the biotechnology sector
– for example,with the development
of new technologies initially designed
to streamline the production of cell
therapies,but which have much broader
applications. Early adopters of these new
technologies include biopharmaceutical
companies,contract manufacturers,
clinics,and even basic researchers.
CellTherapy Regulations
Traditional small molecule therapeutics
must meet benchmarks for safety,purity
and efficacy;so too must biologics,albeit
in fundamentally different ways.Many
of the tests and terminologies applied
to small molecules are not relevant to
biologics.As such,international regulatory
agencies have responded by developing
new guidelines,with recommendations
specific to cell therapy drug products (3,4).
The Office of Cellular,Tissue and Gene
Therapies – a division of the US Food and
Drug Administration (FDA) in charge of
overseeing cell therapies – has noted:“The
diverse biology and clinical indications,and
the rapid and fluid state of the evolving
scientific research into these product
areas,pose unique scientific challenges in
terms of regulatory review”(3).Because
every cell therapy is unique,the onus of
developing therapy-specific standards,
tests and production methods often falls
on the therapeutic developer.This inherent
variability makes the federal mandate for
safety and product standardisation,which
has been in effect for more than 50 years,
more important than ever.
R Lee Buckler at Cell Therapy
Group, and Rolf Ehrhardt and
Maria Thompson at BioCision
Image©SebastianTomus–Fotolia.com
www.samedanltd.com 11
and thawing,are two additional variables
that must be controlled (9,10).
The challenge of standardising biologics
is relatively new.In response,there is
a growing market for products that
standardise the techniques and assays
common to cell therapy production.
New cryopreservation and thawing tools
help companies comply with federal
regulations,and ensure successful and
reproducible cell therapies.
Tools of theTrade
The challenges and mandates mentioned
above have resulted in a push for new
technologies that streamline,automate
and standardise the production of
cell therapy products,by minimising
contamination and human error in
tissue culture,lab practices and release
testing.These technologies are also used
to fully characterise the cell therapy
product,often at the level of RNA,DNA
or protein.The same tools being used to
standardise the commercial production
of cells for the clinic will soon be
translated to biomedical research
labs,improving the reproducibility
of R&D.
For instance, GMP compliance requires
that cell therapy products be produced
in designated clean environments
(cleanrooms), which are subject
to environmental monitoring for
airborne particles and contaminants.
Aseptic production of cell therapy
drug products is critical due to
their parenteral administration (11).
Cleanrooms are expensive to establish
and difficult to maintain. Cell processing
workstations have been developed to
provide a flexible, scalable alternative.
They are, in essence, a cleanroom in a
box. Designed for the production of cell
therapy products, these GMP-compliant
contained spaces can be cleaned in as
little as 90 minutes.This rapid turnover
allows for concurrent development of
multiple cell therapy products (12).
High-throughput process automation
is another major advance for the
commercialisation of cell therapy
products.For low-dose cell therapies
(millions of cells),automated tissue
The European Medicines Agency (EMA)
has also established unique classification
for biologics,including cell therapies,
designating them advanced therapy
medical products (ATMPs).Prompted
by the highly experimental nature of
ATMPs and other emerging therapies,
the EMA set up an Innovation Task Force.
It recognises that ground-breaking
therapies often do not fit existing
regulations,and so encourages early
dialogue with therapeutic developers to
“proactively identify scientific,legal and
regulatory issues of emerging therapies
and technologies”(5).
Compliance with regulatory agencies
requires cell therapy product
conformance testing,comparability
studies and stability testing.These quality
control tests ensure the identity of the
cells,their potency,stability and lack
of contamination (6).
Good Practice Guidelines
Historically,the FDA has issued
regulations for drug development and
production to prevent errors or accidents
that could harm consumers,beginning in
1962 with Good Manufacturing Practice
(GMP) guidelines.In 1978,the FDA
issued Good Laboratory Practice (GLP)
guidelines,which regulate the preclinical
laboratory conditions and organisational
system under which experiments
are conducted.
As cell therapies began to gain traction
in the clinic,the FDA responded by
introducing Good Tissue Practice (GTP)
guidelines in 2011 to regulate the
manufacture of human cells and tissues
for clinical use (7).These guidelines are
designed to prevent the introduction
or spread of communicable disease via
cell therapy.GTP regulates the facilities
and equipment used for cell product
development,plus the ways in which
cells are processed,stored and distributed.
The guidelines relate to new technologies
being developed for the cell therapy field.
CellVariability
Cell-based therapies are still a very small
and emerging niche of the biopharma
industry.Developing a successful cell
therapy is difficult,in part because
of the large number of variables that
come into play when designing
human-based biologic drugs,and the
lack of standardisation typical of a
juvenile industry.
These variables include the source of
the originating cells,their differentiation
status,and the tissue culture techniques
required to maintain the cells.
Additional variables include adjuvants
in the final drug product,other ex vivo
manipulations,and any scaffold on
which cells are grown for implantation.
The potential impact of some of these
variables on the predictability of the
final product can,in some instances,
be largely minimised – for example,by
applying best practices in tissue culture.
However,challenges arise when trying
to minimise all variations,such as those
of autologous cell therapies,which
inherently involve certain patient-to-
patient variability.
From a practical standpoint,both
the number of cells required per dose
and the method of tissue culture affect
cell therapy production.Single cell
therapy doses can range from less
than one million cells (central nervous
system applications) to a few billion
cells (cardiac therapy) (8).
Control Measures
Culturing multiple doses of a few
million cells at a time is a manageable
prospect; sufficient doses can be
reasonably produced with existing
equipment.However,culturing multiple
doses of billions of cells requires
uniquely tailored equipment that is
currently extremely expensive and
operator-intensive.For high-dose cell
therapies,generating sufficient numbers
of cells can be problematic,especially
for small early-stage clinical trials.
Once released from manufacturing,
the final drug product must be
appropriately preserved to uphold
maximum potency,and those conditions
must be maintained throughout
final product shipment to the clinic.
Subsequent handling of the cell therapy
drug product,such as cryopreservation
www.samedanltd.com12
of cell-based therapies,all of
which can be translated to research
labs.Notable examples include cell
washing devices (for elimination
of cryopreservation excipients and
subsequent volume reduction) and
an automatic cell thawing device,
which will bring the same level of
standardisation to thawing as
currently exists for cell freezing.
Also in development are high-
throughput systems for standardised
cell characterisation.These devices
use microwells and microfluidics to
do high-throughput analyses of
DNA and RNA.They also have the
capability to do small molecule
screens (15). It is anticipated that
any such material contribution to
standardisation has the potential
to improve patient-to-patient
reproducibility and efficacy
within clinical trials, by improving
and standardising quality assurance
efforts and increasing potency.
culture systems improve product
consistency,with the added advantage
of speed.Small flasks of adherent
and non-adherent cell lines can be
cultured in parallel,with no risk of
cross-contamination – making them
appropriate for early-stage research.
The machines provide automated
seeding,expansion and sub-cloning.
For high-dose cell therapies,other
options can fully automate the culture
of a single cell line,in T-flasks or
roller bottles (8).
Cryopreservation
Standardised cryopreservation
techniques are also an important
aspect of developing cell therapies.
Following the expansion and/or
manipulation of cells in culture,some
cell therapy products are stored in
liquid nitrogen until immediately prior
to patient administration. Studies
have shown the optimal rate of
cryopreservation for maximum cell
recovery is -1°C per minute (13).This
can be achieved using a programmable
freezer,or a portable,passive freezing
device in conjunction with a -80°C
freezer.Both deliver consistent freezing
profiles,but passive freezing devices
eliminate the need for space,money,
time and maintenance associated with
a programmable freezer.
A recent study showed that such
portable devices offer comparable
post-thaw cell viability to programmable
freezers,providing an inexpensive
and scalable alternative to dedicated,
expensive cell-freezing machinery.
Passive freezing devices are currently
being used to maximise the number
of eligible Phase 2b clinical trial sites
in a cell therapy study involving
regulatory T cells (14).
Other Developments
More devices are coming down the
pipeline to assist in the standardisation
Cell therapy market facts
There are less than 40 cell therapy
products in commercial distribution in
well-regulated markets. Collectively,
commercial cell therapy products
generate close to $1 billion in annual
revenues. With over 300 cell therapy
products in clinical development, the
industry is expected to continue
double-digit compound annual
growth through 2020.
Figure 1: Road to standardisation of cell therapies
2012
Five novel cell therapies approved globally
2010
First FDA-approved cellular
immunotherapy – Provenge®,
Dendreon
2013
Two novel cell therapies
approved globally: Provenge®,
Dendreon approved in the EU
2011
Three novel cell therapies
approved globally
2011
Final GTP
guidelines (US)
2007
EMA regulations (EU)
1978
GLP guidelines (US)
1962
GMP guidelines (US)
2009
First ATMP approved
in Europe by EMA:
ChondroCelect®,
Tigenix
1997
First FDA-approved
cell therapy in US:
Carticel®, Genzyme
Introduction of tools to standardise cell
handling, preservation and storage
www.samedanltd.com14
MSC-based therapy development,
Front Immunol, 2012
5. EMA Innovation Task Force.
Visit: www.ema.europa.eu/ema/
index.jsp?curl=pages/regulation/
general/general_content_000334.
jsp&mid=WC0b01ac05800ba1d9
6. FDA, Guidance for industry: Potency
tests for cellular and gene therapy
products, 2011
7. FDA, Current Good Tissue Practice
and additional requirements for
manufacturers of human cells,
tissues, and cellular and tissue-
based products, 2011
8. Prieels J, Lesage F, Argentin D and
Bollen A, Mastering industrialization
of cell therapy products, BioProcess
International 10: pp12-15, 2012
9. Daniele N et al, T(reg) cells:
collection, processing, storage
and clinical use, Pathol Res
Pract 207: pp209-215, 2011
10. Golab K et al, Challenges in
cryopreservation of regulatory
T cells for clinical therapeutic
applications, Internat Immunopharm
16: pp371-375, 2013
11. World Health Organization,
Environmental monitoring of
cleanrooms in vaccine
manufacturing facilities, 2012
12. Colmenares C, SANYO equipment
puts UAB beyond the state-of-the-
art for cell manufacturing, 2011.
Visit: www.uab.edu/news/latest/
item/1323-new-sanyo-equipment-
propels-uab-cell-therapy-toward-
clinical-care-potential-sickle-cell-
cure-among-the-first-to-be-processed
13. Yokoyama WM and Ehrhardt RO,
Cryopreservation and thawing of
cells, Curr Protoc Immunol,
Appendix 3:3G, 2012
14. Foussat A et al, Use of a low-cost
passive-freezing device in the effective
cryopreservation and recovery of
human regulatory T-cells for use in
cell therapy, BioProcess International
supplement: Cell therapies,
commercialisation, compliance, scale-
up and biopreservation, March 2014
15. Fernandes TG, Clark DS, Dordick JS
and Cabral JMS, High-throughput
cellular microarry platforms:
applications in drug discovery,
toxicology and stem cell research,
Trends Biotechnol 27: pp342-349, 2009
Industry Informs Academia
Irreproducibility is an issue of
growing concern both in the
academic community and among
individuals in the pharmaceutical
industry who rely on basic science
to inform their decisions.The tools
that bring standardisation to the
clinic will serve the same purpose
in research labs, resulting in more
reliable results and a faster pace
of research, as more techniques
become automated.
These technologies will change the
way biomedical research is done as they
move from industrial and clinic settings
into research labs. In other words, cell
therapy has a real chance to improve
standardisation and reproducibility,
not just in development but also in
basic research.
Standardised protocols for cell culture,
cell freezing and cell handling will ensure
that experiments can be repeated,
and that cells are comparable from
one experiment to the next.These are
the same mandates that clinical cell
therapies must meet,and integrate
solutions for standardised preclinical
and clinical research.
References
1. Buckler RL, Cellular therapy wave
finally cresting, Genetic Engineering
News 32, 2012
2. Buckler RL, Rise of cell-based
immunotherapy, Genetic Engineering
News 33, 2013
3. FDA, Guidance for industry: Preclinical
assessment of investigational cellular
and gene therapy products, 2013
4. Ancans J, Cell therapy medicinal
product regulatory framework
in Europe and its application for
About the authors
R Lee Buckler is the founder and Managing Director of Cell Therapy
Group, a business development consultancy focused on the cell
therapy and regenerative medicine industry. He is also a member
of the Board of Directors for cell therapy company, Hemostemix.
Lee was Executive Director of the International Society for Cellular
Therapy (ISCT) from 2000 to 2006. He founded and served as Chair
of ISCT's Cell Therapy Commercialization Committee and is now
Co-Chair of the Alliance for Regenerative Medicine’s Communications and Education
Committee. In addition, Lee founded Cell Therapy News and created Cell Therapy
Blog. Email: lbuckler@celltherapygroup.com
Dr Rolf Ehrhardt is President and Chief Executive Officer of
BioCision, a leading provider of innovative tools for standardising
sample and biomaterial handling, cryopreservation and storage
procedures. He was previously responsible for the early clinical
development of a novel hepatitis C protease inhibitor at Intermune,
as well as Vice President of Preclinical Development at Corgentech
and founding President of BioSeek. Rolf was a German Research
Foundation and Fogarty Fellow at the US National Institutes of Health. He earned
his medical and doctoral degrees with distinction from the Technical University of
Munich, Germany. Email: rolf@biocision.com
Dr Maria Thompson is Vice President, Scientific Affairs at BioCision.
She has a background in molecular genetics and over 16 years
of experience in pharmaceutical and diagnostics R&D. Maria has
held a variety of roles including Head of Genome Wide Screening
for type 2 diabetes, Six Sigma Black Belt, Principal Consultant
and Head of Scientific Affairs, as well as running private
consultancy, APEX Think. She holds a BSc in Genetics and a PhD
in Molecular and Cellular Biochemistry from the Royal London School of Medicine.
Email: maria@biocision.com

More Related Content

What's hot

MDC Connects Series 2021 | A Guide to Complex Medicines: The Target Landscape...
MDC Connects Series 2021 | A Guide to Complex Medicines: The Target Landscape...MDC Connects Series 2021 | A Guide to Complex Medicines: The Target Landscape...
MDC Connects Series 2021 | A Guide to Complex Medicines: The Target Landscape...Medicines Discovery Catapult
 
Paradigm Change in Biomanufacturing
Paradigm Change in Biomanufacturing Paradigm Change in Biomanufacturing
Paradigm Change in Biomanufacturing PnuVax
 
MDC Connects Series 2021 | A Guide to Complex Medicines: The Early Assessment...
MDC Connects Series 2021 | A Guide to Complex Medicines: The Early Assessment...MDC Connects Series 2021 | A Guide to Complex Medicines: The Early Assessment...
MDC Connects Series 2021 | A Guide to Complex Medicines: The Early Assessment...Medicines Discovery Catapult
 
MDC Connects: Use of pre-clinical models to deliver proof of concept efficacy
MDC Connects: Use of pre-clinical models to deliver proof of concept efficacyMDC Connects: Use of pre-clinical models to deliver proof of concept efficacy
MDC Connects: Use of pre-clinical models to deliver proof of concept efficacyMedicines Discovery Catapult
 
Inc in depth biosimilars clinical perspective
Inc in depth biosimilars   clinical perspectiveInc in depth biosimilars   clinical perspective
Inc in depth biosimilars clinical perspectiveGenericlicensing.com
 
In Vitro Alternatives in Toxicology
In Vitro Alternatives in ToxicologyIn Vitro Alternatives in Toxicology
In Vitro Alternatives in ToxicologyMerck Life Sciences
 
Genomics and proteomics in drug discovery and development
Genomics and proteomics in drug discovery and developmentGenomics and proteomics in drug discovery and development
Genomics and proteomics in drug discovery and developmentSuchittaU
 
Moringa kills-human-pancreatic-cancer-cells
Moringa kills-human-pancreatic-cancer-cellsMoringa kills-human-pancreatic-cancer-cells
Moringa kills-human-pancreatic-cancer-cellsDrumstick Moringa
 
Voss et al. - 2006 - Identification of potent anticancer activity in Xi
Voss et al. - 2006 - Identification of potent anticancer activity in XiVoss et al. - 2006 - Identification of potent anticancer activity in Xi
Voss et al. - 2006 - Identification of potent anticancer activity in XiCristina Voss
 
Cell culture and its types
Cell culture and its typesCell culture and its types
Cell culture and its typesPankaj Maurya
 
MDC Connects: Strategies for target and pathway engagement in cellular assays
MDC Connects: Strategies for target and pathway engagement in cellular assaysMDC Connects: Strategies for target and pathway engagement in cellular assays
MDC Connects: Strategies for target and pathway engagement in cellular assaysMedicines Discovery Catapult
 
Human Primary and Cultured Cells (Toxicology Spotlight)
Human Primary and Cultured Cells (Toxicology Spotlight)Human Primary and Cultured Cells (Toxicology Spotlight)
Human Primary and Cultured Cells (Toxicology Spotlight)◂ Justin (M) Gaines ▸
 
Cell Based Assays (2010) Ella
Cell Based Assays (2010) EllaCell Based Assays (2010) Ella
Cell Based Assays (2010) EllaElakeche
 

What's hot (20)

3.nf
3.nf3.nf
3.nf
 
MDC Connects Series 2021 | A Guide to Complex Medicines: The Target Landscape...
MDC Connects Series 2021 | A Guide to Complex Medicines: The Target Landscape...MDC Connects Series 2021 | A Guide to Complex Medicines: The Target Landscape...
MDC Connects Series 2021 | A Guide to Complex Medicines: The Target Landscape...
 
Paradigm Change in Biomanufacturing
Paradigm Change in Biomanufacturing Paradigm Change in Biomanufacturing
Paradigm Change in Biomanufacturing
 
MDC Connects Series 2021 | A Guide to Complex Medicines: The Early Assessment...
MDC Connects Series 2021 | A Guide to Complex Medicines: The Early Assessment...MDC Connects Series 2021 | A Guide to Complex Medicines: The Early Assessment...
MDC Connects Series 2021 | A Guide to Complex Medicines: The Early Assessment...
 
CliniMACS newsletter 2011
CliniMACS newsletter 2011CliniMACS newsletter 2011
CliniMACS newsletter 2011
 
MDC Connects: Use of pre-clinical models to deliver proof of concept efficacy
MDC Connects: Use of pre-clinical models to deliver proof of concept efficacyMDC Connects: Use of pre-clinical models to deliver proof of concept efficacy
MDC Connects: Use of pre-clinical models to deliver proof of concept efficacy
 
Lisa_Grimm
Lisa_GrimmLisa_Grimm
Lisa_Grimm
 
Inc in depth biosimilars clinical perspective
Inc in depth biosimilars   clinical perspectiveInc in depth biosimilars   clinical perspective
Inc in depth biosimilars clinical perspective
 
In Vitro Alternatives in Toxicology
In Vitro Alternatives in ToxicologyIn Vitro Alternatives in Toxicology
In Vitro Alternatives in Toxicology
 
MDC Connects: Targeted compound libraries
MDC Connects: Targeted compound librariesMDC Connects: Targeted compound libraries
MDC Connects: Targeted compound libraries
 
Genomics and proteomics in drug discovery and development
Genomics and proteomics in drug discovery and developmentGenomics and proteomics in drug discovery and development
Genomics and proteomics in drug discovery and development
 
Moringa kills-human-pancreatic-cancer-cells
Moringa kills-human-pancreatic-cancer-cellsMoringa kills-human-pancreatic-cancer-cells
Moringa kills-human-pancreatic-cancer-cells
 
Voss et al. - 2006 - Identification of potent anticancer activity in Xi
Voss et al. - 2006 - Identification of potent anticancer activity in XiVoss et al. - 2006 - Identification of potent anticancer activity in Xi
Voss et al. - 2006 - Identification of potent anticancer activity in Xi
 
Cell culture and its types
Cell culture and its typesCell culture and its types
Cell culture and its types
 
Q5 c step4
Q5 c step4Q5 c step4
Q5 c step4
 
MDC Connects: Designing a Biomarker Strategy
MDC Connects: Designing a Biomarker StrategyMDC Connects: Designing a Biomarker Strategy
MDC Connects: Designing a Biomarker Strategy
 
MDC Connects: Strategies for target and pathway engagement in cellular assays
MDC Connects: Strategies for target and pathway engagement in cellular assaysMDC Connects: Strategies for target and pathway engagement in cellular assays
MDC Connects: Strategies for target and pathway engagement in cellular assays
 
Human Primary and Cultured Cells (Toxicology Spotlight)
Human Primary and Cultured Cells (Toxicology Spotlight)Human Primary and Cultured Cells (Toxicology Spotlight)
Human Primary and Cultured Cells (Toxicology Spotlight)
 
MTTlab flyer A4
MTTlab flyer A4MTTlab flyer A4
MTTlab flyer A4
 
Cell Based Assays (2010) Ella
Cell Based Assays (2010) EllaCell Based Assays (2010) Ella
Cell Based Assays (2010) Ella
 

Viewers also liked

BodyWise Health Center - Cheri Gillham
BodyWise Health Center - Cheri GillhamBodyWise Health Center - Cheri Gillham
BodyWise Health Center - Cheri GillhamBodyWise Health Center
 
נספח שניים ושלש סילבוס אזרח מכוון
נספח שניים ושלש   סילבוס אזרח מכווןנספח שניים ושלש   סילבוס אזרח מכוון
נספח שניים ושלש סילבוס אזרח מכווןhaimkarel
 
Adiva Divine Presentation QNet
Adiva  Divine Presentation  QNetAdiva  Divine Presentation  QNet
Adiva Divine Presentation QNetParag Murkute
 
Snap Freezing Procedure(2)
Snap Freezing Procedure(2)Snap Freezing Procedure(2)
Snap Freezing Procedure(2)biocision
 
pub-thebigfreeze-IPT-sept15
pub-thebigfreeze-IPT-sept15pub-thebigfreeze-IPT-sept15
pub-thebigfreeze-IPT-sept15biocision
 
Roll of the librarian
Roll of the librarianRoll of the librarian
Roll of the librarianDebra Murphy
 
מדוע נבנו מבצרי המשטרה
מדוע נבנו מבצרי המשטרהמדוע נבנו מבצרי המשטרה
מדוע נבנו מבצרי המשטרהhaimkarel
 
ירושלים אליה קפיטולינה
ירושלים אליה קפיטולינהירושלים אליה קפיטולינה
ירושלים אליה קפיטולינהhaimkarel
 
ICT as a driver for change in East Africa?
ICT as a driver for change in East Africa?ICT as a driver for change in East Africa?
ICT as a driver for change in East Africa?Twaweza
 
מחשבים ניידים תאור והשוואה
מחשבים ניידים תאור והשוואהמחשבים ניידים תאור והשוואה
מחשבים ניידים תאור והשוואהhaimkarel
 
שיעור שבע חיפוש מידע באמצעות מנועי חיפוש ואינדקסים
שיעור שבע   חיפוש מידע באמצעות מנועי חיפוש ואינדקסיםשיעור שבע   חיפוש מידע באמצעות מנועי חיפוש ואינדקסים
שיעור שבע חיפוש מידע באמצעות מנועי חיפוש ואינדקסיםhaimkarel
 
Tri-Solve client presentation
Tri-Solve client presentationTri-Solve client presentation
Tri-Solve client presentationcspearce
 
SGE Presentation QNet
SGE Presentation QNetSGE Presentation QNet
SGE Presentation QNetParag Murkute
 
The impact of the Internet use on diversity in newsroom production
The impact of the Internet use on diversity in newsroom production The impact of the Internet use on diversity in newsroom production
The impact of the Internet use on diversity in newsroom production Vinzenz Wyss
 
חוברת הדרכה ותרגול למעבד התמלילים
חוברת הדרכה ותרגול למעבד התמליליםחוברת הדרכה ותרגול למעבד התמלילים
חוברת הדרכה ותרגול למעבד התמליליםhaimkarel
 
Why lean for my org
Why lean for my orgWhy lean for my org
Why lean for my orgJorge Flores
 
Amezcua Harmonised Energy Presentation QNet
Amezcua  Harmonised  Energy  Presentation  QNetAmezcua  Harmonised  Energy  Presentation  QNet
Amezcua Harmonised Energy Presentation QNetParag Murkute
 

Viewers also liked (20)

BodyWise Health Center - Cheri Gillham
BodyWise Health Center - Cheri GillhamBodyWise Health Center - Cheri Gillham
BodyWise Health Center - Cheri Gillham
 
נספח שניים ושלש סילבוס אזרח מכוון
נספח שניים ושלש   סילבוס אזרח מכווןנספח שניים ושלש   סילבוס אזרח מכוון
נספח שניים ושלש סילבוס אזרח מכוון
 
Adiva Divine Presentation QNet
Adiva  Divine Presentation  QNetAdiva  Divine Presentation  QNet
Adiva Divine Presentation QNet
 
Snap Freezing Procedure(2)
Snap Freezing Procedure(2)Snap Freezing Procedure(2)
Snap Freezing Procedure(2)
 
pub-thebigfreeze-IPT-sept15
pub-thebigfreeze-IPT-sept15pub-thebigfreeze-IPT-sept15
pub-thebigfreeze-IPT-sept15
 
Roll of the librarian
Roll of the librarianRoll of the librarian
Roll of the librarian
 
מדוע נבנו מבצרי המשטרה
מדוע נבנו מבצרי המשטרהמדוע נבנו מבצרי המשטרה
מדוע נבנו מבצרי המשטרה
 
ירושלים אליה קפיטולינה
ירושלים אליה קפיטולינהירושלים אליה קפיטולינה
ירושלים אליה קפיטולינה
 
ICT as a driver for change in East Africa?
ICT as a driver for change in East Africa?ICT as a driver for change in East Africa?
ICT as a driver for change in East Africa?
 
Double Page
Double PageDouble Page
Double Page
 
מחשבים ניידים תאור והשוואה
מחשבים ניידים תאור והשוואהמחשבים ניידים תאור והשוואה
מחשבים ניידים תאור והשוואה
 
Qi Comm
Qi Comm Qi Comm
Qi Comm
 
שיעור שבע חיפוש מידע באמצעות מנועי חיפוש ואינדקסים
שיעור שבע   חיפוש מידע באמצעות מנועי חיפוש ואינדקסיםשיעור שבע   חיפוש מידע באמצעות מנועי חיפוש ואינדקסים
שיעור שבע חיפוש מידע באמצעות מנועי חיפוש ואינדקסים
 
Tri-Solve client presentation
Tri-Solve client presentationTri-Solve client presentation
Tri-Solve client presentation
 
SGE Presentation QNet
SGE Presentation QNetSGE Presentation QNet
SGE Presentation QNet
 
The impact of the Internet use on diversity in newsroom production
The impact of the Internet use on diversity in newsroom production The impact of the Internet use on diversity in newsroom production
The impact of the Internet use on diversity in newsroom production
 
חוברת הדרכה ותרגול למעבד התמלילים
חוברת הדרכה ותרגול למעבד התמליליםחוברת הדרכה ותרגול למעבד התמלילים
חוברת הדרכה ותרגול למעבד התמלילים
 
Why lean for my org
Why lean for my orgWhy lean for my org
Why lean for my org
 
Amezcua Harmonised Energy Presentation QNet
Amezcua  Harmonised  Energy  Presentation  QNetAmezcua  Harmonised  Energy  Presentation  QNet
Amezcua Harmonised Energy Presentation QNet
 
Quest Vacation Club
Quest Vacation ClubQuest Vacation Club
Quest Vacation Club
 

Similar to pub_EBR_April-14_pp.10-14

Stem Cells: A Dynamic Landscape
Stem Cells: A Dynamic LandscapeStem Cells: A Dynamic Landscape
Stem Cells: A Dynamic LandscapeCovance
 
Clinical trials and safety considerations
Clinical trials and safety considerationsClinical trials and safety considerations
Clinical trials and safety considerationsZahraa63902
 
Assignment on Alternatives to Animal Screening Method
Assignment on Alternatives to Animal Screening MethodAssignment on Alternatives to Animal Screening Method
Assignment on Alternatives to Animal Screening MethodDeepak Kumar
 
Chap2 CTPMR, 2nd Ed. 2016
Chap2 CTPMR, 2nd Ed. 2016Chap2 CTPMR, 2nd Ed. 2016
Chap2 CTPMR, 2nd Ed. 2016Eric Dollins
 
Global cell therapy market outlook 2020
Global cell therapy market outlook 2020Global cell therapy market outlook 2020
Global cell therapy market outlook 2020KuicK Research
 
Regenerative Medicine: Impact of Convergence on Drug, Device, and Biologics D...
Regenerative Medicine: Impact of Convergence on Drug, Device, and Biologics D...Regenerative Medicine: Impact of Convergence on Drug, Device, and Biologics D...
Regenerative Medicine: Impact of Convergence on Drug, Device, and Biologics D...MaRS Discovery District
 
Nw biotech fundamentals day 1 session 5 life sciences
Nw biotech fundamentals day 1 session 5   life sciencesNw biotech fundamentals day 1 session 5   life sciences
Nw biotech fundamentals day 1 session 5 life sciencesNicholas Weston Lawyers
 
Global cell therapy market outlook 2020
Global cell therapy market outlook 2020Global cell therapy market outlook 2020
Global cell therapy market outlook 2020KuicK Research
 
Clinical development of cell-based therapies in the UK A presentation by Chie...
Clinical development of cell-based therapies in the UK A presentation by Chie...Clinical development of cell-based therapies in the UK A presentation by Chie...
Clinical development of cell-based therapies in the UK A presentation by Chie...Cell and Gene Therapy Catapult
 
Towards Personalized Medicine
Towards Personalized MedicineTowards Personalized Medicine
Towards Personalized MedicineMichel Dumontier
 
Animal cell culture in Biopharmaceutical Industry in the Production of Therap...
Animal cell culture in Biopharmaceutical Industry in the Production of Therap...Animal cell culture in Biopharmaceutical Industry in the Production of Therap...
Animal cell culture in Biopharmaceutical Industry in the Production of Therap...Shubham Chinchulkar
 
Biosimilar an overview
Biosimilar an overviewBiosimilar an overview
Biosimilar an overviewJulius Marco
 
In silico Drug Design: Prospective for Drug Lead Discovery
In silico Drug Design: Prospective for Drug Lead DiscoveryIn silico Drug Design: Prospective for Drug Lead Discovery
In silico Drug Design: Prospective for Drug Lead Discoveryinventionjournals
 
Bionanotechnology and its applications
Bionanotechnology and its applications Bionanotechnology and its applications
Bionanotechnology and its applications rita martin
 
Alternatives to animal screening methods p'screening. mohammadhusain
Alternatives to animal screening methods p'screening. mohammadhusainAlternatives to animal screening methods p'screening. mohammadhusain
Alternatives to animal screening methods p'screening. mohammadhusainVasaya Mohammadhusain
 
Fda phacilitate2010final
Fda phacilitate2010finalFda phacilitate2010final
Fda phacilitate2010finalisoasp
 

Similar to pub_EBR_April-14_pp.10-14 (20)

Stem Cells: A Dynamic Landscape
Stem Cells: A Dynamic LandscapeStem Cells: A Dynamic Landscape
Stem Cells: A Dynamic Landscape
 
Clinical trials and safety considerations
Clinical trials and safety considerationsClinical trials and safety considerations
Clinical trials and safety considerations
 
Assignment on Alternatives to Animal Screening Method
Assignment on Alternatives to Animal Screening MethodAssignment on Alternatives to Animal Screening Method
Assignment on Alternatives to Animal Screening Method
 
Chap2 CTPMR, 2nd Ed. 2016
Chap2 CTPMR, 2nd Ed. 2016Chap2 CTPMR, 2nd Ed. 2016
Chap2 CTPMR, 2nd Ed. 2016
 
Global cell therapy market outlook 2020
Global cell therapy market outlook 2020Global cell therapy market outlook 2020
Global cell therapy market outlook 2020
 
9. Dr. Thomas Schreitmueller - F. Hoffmann-La Roche
9. Dr. Thomas Schreitmueller - F. Hoffmann-La Roche9. Dr. Thomas Schreitmueller - F. Hoffmann-La Roche
9. Dr. Thomas Schreitmueller - F. Hoffmann-La Roche
 
Regenerative Medicine: Impact of Convergence on Drug, Device, and Biologics D...
Regenerative Medicine: Impact of Convergence on Drug, Device, and Biologics D...Regenerative Medicine: Impact of Convergence on Drug, Device, and Biologics D...
Regenerative Medicine: Impact of Convergence on Drug, Device, and Biologics D...
 
OTM_STV-DIR_0
OTM_STV-DIR_0OTM_STV-DIR_0
OTM_STV-DIR_0
 
Nw biotech fundamentals day 1 session 5 life sciences
Nw biotech fundamentals day 1 session 5   life sciencesNw biotech fundamentals day 1 session 5   life sciences
Nw biotech fundamentals day 1 session 5 life sciences
 
7 Biopharma Trends to watch in 2023.pdf
7 Biopharma Trends to watch in 2023.pdf7 Biopharma Trends to watch in 2023.pdf
7 Biopharma Trends to watch in 2023.pdf
 
Global cell therapy market outlook 2020
Global cell therapy market outlook 2020Global cell therapy market outlook 2020
Global cell therapy market outlook 2020
 
Clinical development of cell-based therapies in the UK A presentation by Chie...
Clinical development of cell-based therapies in the UK A presentation by Chie...Clinical development of cell-based therapies in the UK A presentation by Chie...
Clinical development of cell-based therapies in the UK A presentation by Chie...
 
Towards Personalized Medicine
Towards Personalized MedicineTowards Personalized Medicine
Towards Personalized Medicine
 
Animal cell culture in Biopharmaceutical Industry in the Production of Therap...
Animal cell culture in Biopharmaceutical Industry in the Production of Therap...Animal cell culture in Biopharmaceutical Industry in the Production of Therap...
Animal cell culture in Biopharmaceutical Industry in the Production of Therap...
 
Biosimilar an overview
Biosimilar an overviewBiosimilar an overview
Biosimilar an overview
 
In silico Drug Design: Prospective for Drug Lead Discovery
In silico Drug Design: Prospective for Drug Lead DiscoveryIn silico Drug Design: Prospective for Drug Lead Discovery
In silico Drug Design: Prospective for Drug Lead Discovery
 
Bionanotechnology and its applications
Bionanotechnology and its applications Bionanotechnology and its applications
Bionanotechnology and its applications
 
Alternatives to animal screening methods p'screening. mohammadhusain
Alternatives to animal screening methods p'screening. mohammadhusainAlternatives to animal screening methods p'screening. mohammadhusain
Alternatives to animal screening methods p'screening. mohammadhusain
 
Fda phacilitate2010final
Fda phacilitate2010finalFda phacilitate2010final
Fda phacilitate2010final
 
Phụ lục 2. Tiêu chuẩn GMP WHO trong sản xuất chế phẩm sinh học
Phụ lục 2. Tiêu chuẩn GMP WHO trong sản xuất chế phẩm sinh học Phụ lục 2. Tiêu chuẩn GMP WHO trong sản xuất chế phẩm sinh học
Phụ lục 2. Tiêu chuẩn GMP WHO trong sản xuất chế phẩm sinh học
 

More from biocision

pub_DDN 0514 BioCision Interview
pub_DDN 0514 BioCision Interviewpub_DDN 0514 BioCision Interview
pub_DDN 0514 BioCision Interviewbiocision
 
BioCision CoolCell
BioCision CoolCellBioCision CoolCell
BioCision CoolCellbiocision
 
BioCision Product Overview Clinical
BioCision Product Overview ClinicalBioCision Product Overview Clinical
BioCision Product Overview Clinicalbiocision
 
20100426 Bio Cision Product Overview(3)
20100426 Bio Cision Product Overview(3)20100426 Bio Cision Product Overview(3)
20100426 Bio Cision Product Overview(3)biocision
 
The Ice Bucket Makeover Labmate May 2010
The Ice Bucket Makeover Labmate May 2010The Ice Bucket Makeover Labmate May 2010
The Ice Bucket Makeover Labmate May 2010biocision
 
Bioscience Tech Ad July
Bioscience Tech Ad JulyBioscience Tech Ad July
Bioscience Tech Ad Julybiocision
 
Bm Express 96 A4
Bm Express 96 A4Bm Express 96 A4
Bm Express 96 A4biocision
 

More from biocision (7)

pub_DDN 0514 BioCision Interview
pub_DDN 0514 BioCision Interviewpub_DDN 0514 BioCision Interview
pub_DDN 0514 BioCision Interview
 
BioCision CoolCell
BioCision CoolCellBioCision CoolCell
BioCision CoolCell
 
BioCision Product Overview Clinical
BioCision Product Overview ClinicalBioCision Product Overview Clinical
BioCision Product Overview Clinical
 
20100426 Bio Cision Product Overview(3)
20100426 Bio Cision Product Overview(3)20100426 Bio Cision Product Overview(3)
20100426 Bio Cision Product Overview(3)
 
The Ice Bucket Makeover Labmate May 2010
The Ice Bucket Makeover Labmate May 2010The Ice Bucket Makeover Labmate May 2010
The Ice Bucket Makeover Labmate May 2010
 
Bioscience Tech Ad July
Bioscience Tech Ad JulyBioscience Tech Ad July
Bioscience Tech Ad July
 
Bm Express 96 A4
Bm Express 96 A4Bm Express 96 A4
Bm Express 96 A4
 

pub_EBR_April-14_pp.10-14

  • 1. 10 Cell-BasedTherapies April 2014 Back to the Future The explosion in cell-based therapies has led to new technologies to streamline and standardise their commercial production. These tools could now be used back in the lab to help push the frontiers in biomedical research Over the last decade,cell-based therapies have garnered tremendous excitement from researchers,clinicians and the public.These therapies represent the next frontier in drug development.Some cross over into the field of personalised medicine in that they are derived from a patient’s own cells,or they are tailored according to what will be optimal for different patient subsets. With fewer than 40 cell-based therapies in commercial distribution in regulated markets – only half of which have been formally approved – and only a handful making over $100,000 in annual revenue, the field is still relatively immature,but enjoying a steep growth trajectory (1). LatestWave The pipeline of therapies is maturing,with more than 300 cell therapies currently in development.The latest wave has begun to prove successful in the clinic,with particular attention being paid,of late, to cell-based immunotherapies (2).This was initially triggered by Dendreon’s Provenge approval in the US (2010) and Europe (2013).Recent deals made by Novartis,Celgene and through venture capital investment illustrate that early- stage cell-based immunotherapies continue to gain momentum (see Figure 1,page 12). Moreover,the explosion of cell-based therapies is having a knock-on effect throughout the biotechnology sector – for example,with the development of new technologies initially designed to streamline the production of cell therapies,but which have much broader applications. Early adopters of these new technologies include biopharmaceutical companies,contract manufacturers, clinics,and even basic researchers. CellTherapy Regulations Traditional small molecule therapeutics must meet benchmarks for safety,purity and efficacy;so too must biologics,albeit in fundamentally different ways.Many of the tests and terminologies applied to small molecules are not relevant to biologics.As such,international regulatory agencies have responded by developing new guidelines,with recommendations specific to cell therapy drug products (3,4). The Office of Cellular,Tissue and Gene Therapies – a division of the US Food and Drug Administration (FDA) in charge of overseeing cell therapies – has noted:“The diverse biology and clinical indications,and the rapid and fluid state of the evolving scientific research into these product areas,pose unique scientific challenges in terms of regulatory review”(3).Because every cell therapy is unique,the onus of developing therapy-specific standards, tests and production methods often falls on the therapeutic developer.This inherent variability makes the federal mandate for safety and product standardisation,which has been in effect for more than 50 years, more important than ever. R Lee Buckler at Cell Therapy Group, and Rolf Ehrhardt and Maria Thompson at BioCision Image©SebastianTomus–Fotolia.com
  • 2. www.samedanltd.com 11 and thawing,are two additional variables that must be controlled (9,10). The challenge of standardising biologics is relatively new.In response,there is a growing market for products that standardise the techniques and assays common to cell therapy production. New cryopreservation and thawing tools help companies comply with federal regulations,and ensure successful and reproducible cell therapies. Tools of theTrade The challenges and mandates mentioned above have resulted in a push for new technologies that streamline,automate and standardise the production of cell therapy products,by minimising contamination and human error in tissue culture,lab practices and release testing.These technologies are also used to fully characterise the cell therapy product,often at the level of RNA,DNA or protein.The same tools being used to standardise the commercial production of cells for the clinic will soon be translated to biomedical research labs,improving the reproducibility of R&D. For instance, GMP compliance requires that cell therapy products be produced in designated clean environments (cleanrooms), which are subject to environmental monitoring for airborne particles and contaminants. Aseptic production of cell therapy drug products is critical due to their parenteral administration (11). Cleanrooms are expensive to establish and difficult to maintain. Cell processing workstations have been developed to provide a flexible, scalable alternative. They are, in essence, a cleanroom in a box. Designed for the production of cell therapy products, these GMP-compliant contained spaces can be cleaned in as little as 90 minutes.This rapid turnover allows for concurrent development of multiple cell therapy products (12). High-throughput process automation is another major advance for the commercialisation of cell therapy products.For low-dose cell therapies (millions of cells),automated tissue The European Medicines Agency (EMA) has also established unique classification for biologics,including cell therapies, designating them advanced therapy medical products (ATMPs).Prompted by the highly experimental nature of ATMPs and other emerging therapies, the EMA set up an Innovation Task Force. It recognises that ground-breaking therapies often do not fit existing regulations,and so encourages early dialogue with therapeutic developers to “proactively identify scientific,legal and regulatory issues of emerging therapies and technologies”(5). Compliance with regulatory agencies requires cell therapy product conformance testing,comparability studies and stability testing.These quality control tests ensure the identity of the cells,their potency,stability and lack of contamination (6). Good Practice Guidelines Historically,the FDA has issued regulations for drug development and production to prevent errors or accidents that could harm consumers,beginning in 1962 with Good Manufacturing Practice (GMP) guidelines.In 1978,the FDA issued Good Laboratory Practice (GLP) guidelines,which regulate the preclinical laboratory conditions and organisational system under which experiments are conducted. As cell therapies began to gain traction in the clinic,the FDA responded by introducing Good Tissue Practice (GTP) guidelines in 2011 to regulate the manufacture of human cells and tissues for clinical use (7).These guidelines are designed to prevent the introduction or spread of communicable disease via cell therapy.GTP regulates the facilities and equipment used for cell product development,plus the ways in which cells are processed,stored and distributed. The guidelines relate to new technologies being developed for the cell therapy field. CellVariability Cell-based therapies are still a very small and emerging niche of the biopharma industry.Developing a successful cell therapy is difficult,in part because of the large number of variables that come into play when designing human-based biologic drugs,and the lack of standardisation typical of a juvenile industry. These variables include the source of the originating cells,their differentiation status,and the tissue culture techniques required to maintain the cells. Additional variables include adjuvants in the final drug product,other ex vivo manipulations,and any scaffold on which cells are grown for implantation. The potential impact of some of these variables on the predictability of the final product can,in some instances, be largely minimised – for example,by applying best practices in tissue culture. However,challenges arise when trying to minimise all variations,such as those of autologous cell therapies,which inherently involve certain patient-to- patient variability. From a practical standpoint,both the number of cells required per dose and the method of tissue culture affect cell therapy production.Single cell therapy doses can range from less than one million cells (central nervous system applications) to a few billion cells (cardiac therapy) (8). Control Measures Culturing multiple doses of a few million cells at a time is a manageable prospect; sufficient doses can be reasonably produced with existing equipment.However,culturing multiple doses of billions of cells requires uniquely tailored equipment that is currently extremely expensive and operator-intensive.For high-dose cell therapies,generating sufficient numbers of cells can be problematic,especially for small early-stage clinical trials. Once released from manufacturing, the final drug product must be appropriately preserved to uphold maximum potency,and those conditions must be maintained throughout final product shipment to the clinic. Subsequent handling of the cell therapy drug product,such as cryopreservation
  • 3. www.samedanltd.com12 of cell-based therapies,all of which can be translated to research labs.Notable examples include cell washing devices (for elimination of cryopreservation excipients and subsequent volume reduction) and an automatic cell thawing device, which will bring the same level of standardisation to thawing as currently exists for cell freezing. Also in development are high- throughput systems for standardised cell characterisation.These devices use microwells and microfluidics to do high-throughput analyses of DNA and RNA.They also have the capability to do small molecule screens (15). It is anticipated that any such material contribution to standardisation has the potential to improve patient-to-patient reproducibility and efficacy within clinical trials, by improving and standardising quality assurance efforts and increasing potency. culture systems improve product consistency,with the added advantage of speed.Small flasks of adherent and non-adherent cell lines can be cultured in parallel,with no risk of cross-contamination – making them appropriate for early-stage research. The machines provide automated seeding,expansion and sub-cloning. For high-dose cell therapies,other options can fully automate the culture of a single cell line,in T-flasks or roller bottles (8). Cryopreservation Standardised cryopreservation techniques are also an important aspect of developing cell therapies. Following the expansion and/or manipulation of cells in culture,some cell therapy products are stored in liquid nitrogen until immediately prior to patient administration. Studies have shown the optimal rate of cryopreservation for maximum cell recovery is -1°C per minute (13).This can be achieved using a programmable freezer,or a portable,passive freezing device in conjunction with a -80°C freezer.Both deliver consistent freezing profiles,but passive freezing devices eliminate the need for space,money, time and maintenance associated with a programmable freezer. A recent study showed that such portable devices offer comparable post-thaw cell viability to programmable freezers,providing an inexpensive and scalable alternative to dedicated, expensive cell-freezing machinery. Passive freezing devices are currently being used to maximise the number of eligible Phase 2b clinical trial sites in a cell therapy study involving regulatory T cells (14). Other Developments More devices are coming down the pipeline to assist in the standardisation Cell therapy market facts There are less than 40 cell therapy products in commercial distribution in well-regulated markets. Collectively, commercial cell therapy products generate close to $1 billion in annual revenues. With over 300 cell therapy products in clinical development, the industry is expected to continue double-digit compound annual growth through 2020. Figure 1: Road to standardisation of cell therapies 2012 Five novel cell therapies approved globally 2010 First FDA-approved cellular immunotherapy – Provenge®, Dendreon 2013 Two novel cell therapies approved globally: Provenge®, Dendreon approved in the EU 2011 Three novel cell therapies approved globally 2011 Final GTP guidelines (US) 2007 EMA regulations (EU) 1978 GLP guidelines (US) 1962 GMP guidelines (US) 2009 First ATMP approved in Europe by EMA: ChondroCelect®, Tigenix 1997 First FDA-approved cell therapy in US: Carticel®, Genzyme Introduction of tools to standardise cell handling, preservation and storage
  • 4. www.samedanltd.com14 MSC-based therapy development, Front Immunol, 2012 5. EMA Innovation Task Force. Visit: www.ema.europa.eu/ema/ index.jsp?curl=pages/regulation/ general/general_content_000334. jsp&mid=WC0b01ac05800ba1d9 6. FDA, Guidance for industry: Potency tests for cellular and gene therapy products, 2011 7. FDA, Current Good Tissue Practice and additional requirements for manufacturers of human cells, tissues, and cellular and tissue- based products, 2011 8. Prieels J, Lesage F, Argentin D and Bollen A, Mastering industrialization of cell therapy products, BioProcess International 10: pp12-15, 2012 9. Daniele N et al, T(reg) cells: collection, processing, storage and clinical use, Pathol Res Pract 207: pp209-215, 2011 10. Golab K et al, Challenges in cryopreservation of regulatory T cells for clinical therapeutic applications, Internat Immunopharm 16: pp371-375, 2013 11. World Health Organization, Environmental monitoring of cleanrooms in vaccine manufacturing facilities, 2012 12. Colmenares C, SANYO equipment puts UAB beyond the state-of-the- art for cell manufacturing, 2011. Visit: www.uab.edu/news/latest/ item/1323-new-sanyo-equipment- propels-uab-cell-therapy-toward- clinical-care-potential-sickle-cell- cure-among-the-first-to-be-processed 13. Yokoyama WM and Ehrhardt RO, Cryopreservation and thawing of cells, Curr Protoc Immunol, Appendix 3:3G, 2012 14. Foussat A et al, Use of a low-cost passive-freezing device in the effective cryopreservation and recovery of human regulatory T-cells for use in cell therapy, BioProcess International supplement: Cell therapies, commercialisation, compliance, scale- up and biopreservation, March 2014 15. Fernandes TG, Clark DS, Dordick JS and Cabral JMS, High-throughput cellular microarry platforms: applications in drug discovery, toxicology and stem cell research, Trends Biotechnol 27: pp342-349, 2009 Industry Informs Academia Irreproducibility is an issue of growing concern both in the academic community and among individuals in the pharmaceutical industry who rely on basic science to inform their decisions.The tools that bring standardisation to the clinic will serve the same purpose in research labs, resulting in more reliable results and a faster pace of research, as more techniques become automated. These technologies will change the way biomedical research is done as they move from industrial and clinic settings into research labs. In other words, cell therapy has a real chance to improve standardisation and reproducibility, not just in development but also in basic research. Standardised protocols for cell culture, cell freezing and cell handling will ensure that experiments can be repeated, and that cells are comparable from one experiment to the next.These are the same mandates that clinical cell therapies must meet,and integrate solutions for standardised preclinical and clinical research. References 1. Buckler RL, Cellular therapy wave finally cresting, Genetic Engineering News 32, 2012 2. Buckler RL, Rise of cell-based immunotherapy, Genetic Engineering News 33, 2013 3. FDA, Guidance for industry: Preclinical assessment of investigational cellular and gene therapy products, 2013 4. Ancans J, Cell therapy medicinal product regulatory framework in Europe and its application for About the authors R Lee Buckler is the founder and Managing Director of Cell Therapy Group, a business development consultancy focused on the cell therapy and regenerative medicine industry. He is also a member of the Board of Directors for cell therapy company, Hemostemix. Lee was Executive Director of the International Society for Cellular Therapy (ISCT) from 2000 to 2006. He founded and served as Chair of ISCT's Cell Therapy Commercialization Committee and is now Co-Chair of the Alliance for Regenerative Medicine’s Communications and Education Committee. In addition, Lee founded Cell Therapy News and created Cell Therapy Blog. Email: lbuckler@celltherapygroup.com Dr Rolf Ehrhardt is President and Chief Executive Officer of BioCision, a leading provider of innovative tools for standardising sample and biomaterial handling, cryopreservation and storage procedures. He was previously responsible for the early clinical development of a novel hepatitis C protease inhibitor at Intermune, as well as Vice President of Preclinical Development at Corgentech and founding President of BioSeek. Rolf was a German Research Foundation and Fogarty Fellow at the US National Institutes of Health. He earned his medical and doctoral degrees with distinction from the Technical University of Munich, Germany. Email: rolf@biocision.com Dr Maria Thompson is Vice President, Scientific Affairs at BioCision. She has a background in molecular genetics and over 16 years of experience in pharmaceutical and diagnostics R&D. Maria has held a variety of roles including Head of Genome Wide Screening for type 2 diabetes, Six Sigma Black Belt, Principal Consultant and Head of Scientific Affairs, as well as running private consultancy, APEX Think. She holds a BSc in Genetics and a PhD in Molecular and Cellular Biochemistry from the Royal London School of Medicine. Email: maria@biocision.com