SlideShare una empresa de Scribd logo
1 de 4
Descargar para leer sin conexión
Cryopreservation Technologies
44 Innovations in Pharmaceutical Technology Issue 54
By Rolf Ehrhardt and
Maria Thompson at
BioCision
In order to maintain their peak functionality, therapeutic cells rely
on being effectively frozen and thawed. This calls for increasingly
optimised and standardised approaches to cryopreservation,
as improvements in consistency and reproducibility will have a
positive impact on patient outcomes
The Big Freeze
Keywords
Cellular therapy
Freezing devices
Cryoprotectants
Cell thawing
The goal of cell cryopreservation is to suppress
the natural processes of change and degradation
inherent within any biological system,and to preserve
viability,structure and function in as perfect a state
as possible.For therapeutic cells,maintaining their
peak achievable functionality is not merely a desirable
outcome,but an obligation subject to the legal and
ethical oversight of institutions such
as the FDA and EMA.However,the
sheer variety of cell types and their
diverse biological characteristics
defies a single method of culturing,
preserving and preparing cells for
clinical use.Nonetheless,standardising
how cells are handled during these
processes is important.
Cryopreservation
While an important part of the process,cryopreservation
has not always been given the same consideration as
other processing steps,like cell characterisation,potency
and safety.Given that pharmaceutical companies must
define their target patient population early on in drug
development,functional activity and toxicity assays
are understandably the focal point of due diligence
when dealing with cellular products.Cryopreservation
of products is often subject to somewhat less stringent
treatment,which can make it a weak link in the cellular
therapy manufacturing workflow.
Therapeutic cells respond to the stress of
cryopreservation in different ways,depending
Image:©roganjosh–morguefile.com
iptonline.com
45Innovations in Pharmaceutical Technology Issue 54
When cells are cooled below 0°C,extracellular ice
crystals begin to form and relative solute concentration
of the freezing media rises.Water flows out of the cell,
beginning the process of cell dehydration.The cooling
rate must be slow enough to allow sufficient egress
of water to minimise the chance of intracellular ice
crystal formation,while at the same time fast enough
to avoid serious dehydration effects.It is for this reason
that cryoprotectants are used:to lower the freezing
point of the extracellular medium,limiting both ice
crystal formation and the osmotic stress that results in
dehydration.Researchers should only use freezing media
that is both fully defined and validated to be optimal for
their cell type.While it is possible that optimal cooling
rates may vary by cell type,the current recommended
freezing procedure for many therapeutic cells is that
they are cooled gradually to approximately -80°C at a
controlled rate of -1°C per minute (2).This cooling rate is
proven to work well for peripheral blood mononuclear
cells,primary cells,stem cells and cell lines.
Freezing Devices
Active control of the freezing process is particularly
important in a regulated environment,where protocols
must comply with GMP standards to deliver a functional
product that is produced in controlled and predictable
conditions.Retention of good functionality requires a
reproducible,validated methodology.
Until recently,this generally meant cryopreservation
needed to be performed using large,expensive,
programmable controlled rate freezers.Such freezers
offer a precisely controlled cooling rate and are
well-validated, but their cost, large footprint and
maintenance issues are a prohibitive drawback,
especially to small or mid-sized companies.In the last
few years,passive freezing devices have gained in
popularity,largely due to their ability to offer equivalent
cooling rates and cell viability results,without the
exorbitant costs and laboratory space required for
controlled rate freezers.Passive cell freezing containers
– particularly those that lower contamination risks
and operating costs by being alcohol-free – are
easily transferrable from the development phase to
the manufacturing suite,and are increasingly being
validated for use in GMP environments (3).
Maintaining the Cold Chain
Once cells and other biological products are safely
frozen,there will inevitably come a time when they
must be transported – either within a facility or to an
end-user.Therapeutic cells,like any other biological
material,are subject to the risk of temperature
fluctuation during transport and,while regulatory
on variables such as cell type,length of storage,
cryoprotectants used,or freezing and thawing
conditions.Optimising cryopreservation to meet the
needs of cellular therapy can be a complicated process.
For example,therapeutic cell viability is often assessed
immediately post-thaw,without taking delayed-
onset cell death into account.The same is true of cell
function.The assays themselves may be well-defined,
but checking functional activity immediately post-
thaw may not give the best indication of what will
occur in the patient.Some cell types require a post-
thaw recovery period of several hours or even days
before functionality is restored; others may be more
dramatically affected by cryogenic storage conditions,
and prone to changes in phenotype or sub-population
frequency (1).
The scientist must determine if cryopreserved cells
maintain the same potency and function at the point
of infusion into a patient as they did before they
were frozen.Freezing and recovery media should be
carefully chosen to match the cell type in question,and
researchers need to ensure that the components of the
media are fully defined.Perhaps the most important
consideration for cryopreserving cells,particularly
in a Good Manufacturing Practice (GMP) regulated
environment,is to confirm that the cryopreservation
methodology itself – and the technology supporting
it – is fully optimised and standardised.
Given the natural proclivity for cells to respond and
adapt to their environment,there is an ongoing need
for cryopreservation techniques to adapt in a way
that minimises variability and subjectivity at every
possible point when the process is transferred from
development to manufacturing.The complete process
includes freezing,cold chain management during
transportation and thawing.Luckily,necessity informs
invention,and as cellular therapy gains momentum,
we are beginning to see a paradigm shift towards
more optimised,standardised and translatable
cryopreservation technologies.
Cell Freezing
This is the starting point of any cryopreservation
workflow,and while pharma R&D teams may not always
have the time and resources to devote to optimising
freezing rates and media for each cell type,these factors
will have an important influence on ultimate cell survival.
Fortunately,a great deal of scientific effort has been
devoted over the years to understanding the effects of
various freezing parameters on cell survival and recovery.
Generally speaking,freezing protocols are based on
avoiding the physiological effects of dehydration and
ice crystal formation.
46 Innovations in Pharmaceutical Technology Issue 54
iptonline.com
containers,and are capable of maintaining temperatures
below -50°C for over 24 hours.Liquid nitrogen dry
vapour shippers can now be used in place of dry ice
shipping,and can provide cooling for 10 days or more
while fragile biospecimens are shipped.Other liquid
nitrogen-based solutions exist that are more suitable
for short-distance transport.
The International Society for Biological and
Environmental Repositories recognised a hand-portable
liquid nitrogen-based carrier as their‘Outstanding
New Product’of 2015.The CryoPod™ Carrier holds
samples at below -150°C for up to four hours,while
providing automatic temperature alarms,monitoring
and logging,so that users will immediately be alerted if
the temperature has been compromised at any point in
the journey.Such products point towards a shift to more
stringent,standardised temperature-control solutions.
Re-Thinking CellThawing
In the field of regenerative medicine,the final step in
the cryopreservation workflow involves thawing the
cryopreserved cells so they can be administered to the
patient.Controlling the rate of thawing is as important
as freezing,and optimising both is crucial to cell survival
and function.Only recently,however,has cell thawing
been given the same serious consideration that cell
freezing has been subject to (7).
During thawing,many of the internal processes of cell
freezing are reversed.As cells are warmed,extracellular
ice melts and water can rush into the cell,causing it to
swell or even burst.If thawing is done too slowly,or heat
application is uneven,tiny ice crystals formed during the
freezing process can grow and become large enough
to cause damage.The shock of transit back to a viable
state can also cause oxidative stress,triggering apoptotic
pathways and delayed onset cell death.
Water Bath Technology
By far the most common method of thawing cells
is through the use of a heated water bath.When
agencies promote guidelines for maintaining optimal
temperatures during clinical studies and manufacturing
processes,temperature control during transport is still
poorly upheld.A recent US study found that the top
six globally administered vaccines (including those for
polio,diphtheria and measles) lose up to 36% of their
effective shelf-life due to failures in meeting cold chain
requirements,as measured by vaccine vial monitoring
(4).And this is not an isolated report; another study
found that most,if not all,vaccines are exposed to
freeze damage risk at some point throughout the
shipping process (5).
Temperature fluctuation problems are not only limited
to long-distance shipping;they can occur during cross-
campus or cross-town transport,during transfer to
and from liquid nitrogen storage,or even simply when
cryostorage boxes or liquid nitrogen canes are shifted
around to access specific samples.But while transport
of temperature-sensitive biologicals may not be
standardised,scientists are beginning to recognise that
even short-term increases in temperature can expose cells
to the dangers of thermal cycling and ice recrystallisation,
lowering cell viability,recovery and,in the case of stem
cells,even the ability to be pluripotent (6).The good news
is,in recent years,several transportation and storage
solutions have come to market that are aimed at more
stringently controlling the cold chain,whether it be during
long-distance transport or movement within a medical,
research or biopharma manufacturing facility.
NovelTransport Systems
For a long time,homemade dry ice containers were
the only method for shipping temperature-sensitive
biomaterials.With the emergence of the cellular
therapy industry,it became recognised that this type
of container was too unreliable to entrust products
representing a company’s major investment in time and
money – not to mention what might be a patient’s best
hope for survival.Recently,more robust dry ice-based
transport systems – and even mobile dry ice-based
workstations – have emerged to deliver more reliable
temperature stability than conventional Styrofoam
Temperature fluctuation problems are not only limited
to long-distance shipping; they can occur during cross-
campus or cross-town transport, during transfer to
and from liquid nitrogen storage, or even simply when
cryostorage boxes or liquid nitrogen canes are shifted
around to access specific samples
iptonline.com
47Innovations in Pharmaceutical Technology Issue 54
References
1. McKenna KC, Beatty KM, Vicetti Miguel R and
Bilonick RA, Delayed processing of blood
increases the frequency of activated CD11b+
CD15+
granulocytes which inhibit T cell function,
J Immunol Methods 341: pp68-75, 2009
2. Bissoyi A, Nayak B, Pramanik K and Sarangi SK,
Targeting cryopreservation-induced cell death:
A review, Biopreservation and Biobanking
12(1): pp23-34, 2014
3. Stone M et al, Maximizing PMBC recovery and
viability: A method to optimize and streamline
peripheral blood mononuclear cell isolation,
cryopreservation and thawing, BioProcess
International, April 2015
4. Kartoglu U et al, Use of cool water packs to
prevent freezing during vaccine transportation
at the country level, PDA Journal of
Pharmaceutical Science and Technology 63(1):
pp11-26, 2009
5. Kumru O et al, Vaccine instability in the cold
chain: Mechanisms, analysis, and formulation
strategy, Biologicals 42: pp237-259, 2014
6. Deller RC, Vatish M, Mitchell DA and Gibson MI,
Synthetic polymers enable non-vitreous cellular
cryopreservation by reducing ice crystal growth
during thawing, Nature Communications 5:
p3,244, 2014
7. Thompson M, Kunkel E and Ehrhardt R,
Balancing the Cryopreservation Equation,
Cold Facts, January 2015
examined in the context of a GMP-regulated
environment, this method is fraught with problems.
Due to the fact that water is an efficient carrier of
contaminating organisms, GMP regulations either
prohibit water bath use, or specify that a water bath
be cleaned and refilled after each use.This method
of thawing can also be very subjective.The thawing
endpoint is based on each operator’s observation –
often determined by repeatedly lifting the vial out of
the bath to peer at it – of when the last ice crystal is
ready to melt.
Often,no set times are applied and the researcher’s
attention may be momentarily called away to another
task, adding to the risk of variable endpoints and
potential overcooking of cells.The problem is
compounded when more than one sample is involved.
In a clinical setting, cryobags are often used to freeze
large volumes of cells.When these bags are thawed,
the first bag may take just four minutes. However, the
first bag cools the temperature of the water bath and
then, when the second bag is added, it may take much
longer. Again, the process is inconsistent and will lead
to variable results.
A more standardised approach would greatly benefit
researchers, in-process manufacturing personnel and
clinicians alike. An automated cell thawing system
is now on the market that is designed to detect the
solid-to-liquid phase change within a cryovial.
The system is water free, small enough to fit inside
a tissue culture hood and has already been validated
in several labs that follow GMP regulations (3).The
technology is adaptable to other vessel formats – for
example, bags, bigger vials and multiple vials – giving
it the versatility to be adopted into many different
clinical models.
Better Prospects
Today, we are at a stage where the cellular therapy
industry is just beginning to develop. As with all
new endeavours, there are growing pains to be
overcome before the infrastructure can be put
into place. In addition, the creation of guidelines
for the standardisation of workflow methods is
still ongoing.
Cryopreservation is one of the lynchpins of cellular
therapy, and improvements in consistency and
reproducibility will no doubt benefit patient outcomes
down the line. Optimising cryopreservation will
improve the safety and efficacy of cellular drug
products. It is therefore important that we adopt
tools and procedures that both reduce variability
and streamline the process as we move forward.
Dr Rolf Ehrhardt is the President and Chief
Executive Officer of BioCision, a leading
provider of innovative tools for standardising
sample and biomaterial handling,
cryopreservation and storage procedures.
Previously, he was responsible for the early
clinical development of a novel hepatitis C virus protease
inhibitor at Intermune, was Vice President of Preclinical
Development at Corgentech and founding President of
BioSeek. Rolf 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 of
Scientific Affairs at BioCision. She has a
background in molecular genetics, and over
18 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, in addition to running APEX Think, a
private consulting company. She has a BSc in Genetics and a
PhD in Molecular and Cellular Biochemistry from the Royal
London School of Medicine, UK. Email: maria@biocision.com

Más contenido relacionado

Destacado

Does information work? Presentation by Rakesh Rajani
Does information work? Presentation by Rakesh RajaniDoes information work? Presentation by Rakesh Rajani
Does information work? Presentation by Rakesh RajaniTwaweza
 
How transparency and accountability can make development work
How transparency and accountability can make development work How transparency and accountability can make development work
How transparency and accountability can make development work Twaweza
 
BodyWise Health Center - Cheri Gillham
BodyWise Health Center - Cheri GillhamBodyWise Health Center - Cheri Gillham
BodyWise Health Center - Cheri GillhamBodyWise Health Center
 
pub_EBR_April-14_pp.10-14
pub_EBR_April-14_pp.10-14pub_EBR_April-14_pp.10-14
pub_EBR_April-14_pp.10-14biocision
 
Cyber Attacks
Cyber AttacksCyber Attacks
Cyber Attackshaimkarel
 
איחוד הרשויות
איחוד הרשויותאיחוד הרשויות
איחוד הרשויותhaimkarel
 
Presentation by Rachel Steinacher, on IPA and RCTs
Presentation by Rachel Steinacher, on IPA and RCTsPresentation by Rachel Steinacher, on IPA and RCTs
Presentation by Rachel Steinacher, on IPA and RCTsTwaweza
 
Oracle Financial Services - VN Customer Appreciation - Solutions Brief
Oracle Financial Services - VN Customer Appreciation - Solutions BriefOracle Financial Services - VN Customer Appreciation - Solutions Brief
Oracle Financial Services - VN Customer Appreciation - Solutions Briefguest25a062
 
אתרים נוצריים מסביב לכנרת
אתרים נוצריים מסביב לכנרתאתרים נוצריים מסביב לכנרת
אתרים נוצריים מסביב לכנרתhaimkarel
 
Snap Freezing Procedure(2)
Snap Freezing Procedure(2)Snap Freezing Procedure(2)
Snap Freezing Procedure(2)biocision
 
שיעור שלישי מעבד תמלילים וגלישה ראשונה באינטרנט
שיעור שלישי   מעבד תמלילים וגלישה ראשונה באינטרנטשיעור שלישי   מעבד תמלילים וגלישה ראשונה באינטרנט
שיעור שלישי מעבד תמלילים וגלישה ראשונה באינטרנטhaimkarel
 
נתיבה בן יהודה
נתיבה בן יהודהנתיבה בן יהודה
נתיבה בן יהודהhaimkarel
 
ירושלים איליה קפיטולינה
ירושלים איליה קפיטולינהירושלים איליה קפיטולינה
ירושלים איליה קפיטולינהhaimkarel
 
חוות דעת מומחה משלימה 29 1 07
חוות דעת מומחה משלימה 29 1 07חוות דעת מומחה משלימה 29 1 07
חוות דעת מומחה משלימה 29 1 07haimkarel
 
דרום מזרח טורקיה
דרום מזרח טורקיהדרום מזרח טורקיה
דרום מזרח טורקיהhaimkarel
 
BioCision CoolCell
BioCision CoolCellBioCision CoolCell
BioCision CoolCellbiocision
 
Karthik Muralidharan on research on achieving universal quality primary educa...
Karthik Muralidharan on research on achieving universal quality primary educa...Karthik Muralidharan on research on achieving universal quality primary educa...
Karthik Muralidharan on research on achieving universal quality primary educa...Twaweza
 
מפות בגולן
מפות בגולןמפות בגולן
מפות בגולןhaimkarel
 

Destacado (20)

Does information work? Presentation by Rakesh Rajani
Does information work? Presentation by Rakesh RajaniDoes information work? Presentation by Rakesh Rajani
Does information work? Presentation by Rakesh Rajani
 
How transparency and accountability can make development work
How transparency and accountability can make development work How transparency and accountability can make development work
How transparency and accountability can make development work
 
BodyWise Health Center - Cheri Gillham
BodyWise Health Center - Cheri GillhamBodyWise Health Center - Cheri Gillham
BodyWise Health Center - Cheri Gillham
 
pub_EBR_April-14_pp.10-14
pub_EBR_April-14_pp.10-14pub_EBR_April-14_pp.10-14
pub_EBR_April-14_pp.10-14
 
Cyber Attacks
Cyber AttacksCyber Attacks
Cyber Attacks
 
איחוד הרשויות
איחוד הרשויותאיחוד הרשויות
איחוד הרשויות
 
Presentation by Rachel Steinacher, on IPA and RCTs
Presentation by Rachel Steinacher, on IPA and RCTsPresentation by Rachel Steinacher, on IPA and RCTs
Presentation by Rachel Steinacher, on IPA and RCTs
 
Oracle Financial Services - VN Customer Appreciation - Solutions Brief
Oracle Financial Services - VN Customer Appreciation - Solutions BriefOracle Financial Services - VN Customer Appreciation - Solutions Brief
Oracle Financial Services - VN Customer Appreciation - Solutions Brief
 
אתרים נוצריים מסביב לכנרת
אתרים נוצריים מסביב לכנרתאתרים נוצריים מסביב לכנרת
אתרים נוצריים מסביב לכנרת
 
Snap Freezing Procedure(2)
Snap Freezing Procedure(2)Snap Freezing Procedure(2)
Snap Freezing Procedure(2)
 
שיעור שלישי מעבד תמלילים וגלישה ראשונה באינטרנט
שיעור שלישי   מעבד תמלילים וגלישה ראשונה באינטרנטשיעור שלישי   מעבד תמלילים וגלישה ראשונה באינטרנט
שיעור שלישי מעבד תמלילים וגלישה ראשונה באינטרנט
 
נתיבה בן יהודה
נתיבה בן יהודהנתיבה בן יהודה
נתיבה בן יהודה
 
Netikecija
NetikecijaNetikecija
Netikecija
 
ירושלים איליה קפיטולינה
ירושלים איליה קפיטולינהירושלים איליה קפיטולינה
ירושלים איליה קפיטולינה
 
חוות דעת מומחה משלימה 29 1 07
חוות דעת מומחה משלימה 29 1 07חוות דעת מומחה משלימה 29 1 07
חוות דעת מומחה משלימה 29 1 07
 
דרום מזרח טורקיה
דרום מזרח טורקיהדרום מזרח טורקיה
דרום מזרח טורקיה
 
BioCision CoolCell
BioCision CoolCellBioCision CoolCell
BioCision CoolCell
 
Quest Net Profile
Quest Net ProfileQuest Net Profile
Quest Net Profile
 
Karthik Muralidharan on research on achieving universal quality primary educa...
Karthik Muralidharan on research on achieving universal quality primary educa...Karthik Muralidharan on research on achieving universal quality primary educa...
Karthik Muralidharan on research on achieving universal quality primary educa...
 
מפות בגולן
מפות בגולןמפות בגולן
מפות בגולן
 

Similar a pub-thebigfreeze-IPT-sept15

Cryopreservation of gamtes by bhawan.pptx
Cryopreservation of gamtes by bhawan.pptxCryopreservation of gamtes by bhawan.pptx
Cryopreservation of gamtes by bhawan.pptxBhawanpreetkaurahluw
 
Unit-IV Germplasm Storage and Cryopreservation.pptx
Unit-IV Germplasm Storage and Cryopreservation.pptxUnit-IV Germplasm Storage and Cryopreservation.pptx
Unit-IV Germplasm Storage and Cryopreservation.pptxVikrantPawar37
 
Sperm cryoperservation
Sperm cryoperservationSperm cryoperservation
Sperm cryoperservationYasminmagdi
 
Cryopreservation.pptx
Cryopreservation.pptxCryopreservation.pptx
Cryopreservation.pptxAlthamishAzzu
 
Spermcryoperservation by Dr.Chandan
Spermcryoperservation by Dr.Chandan Spermcryoperservation by Dr.Chandan
Spermcryoperservation by Dr.Chandan Morris Jawahar
 
Cryo 2014 CellSeal Presentation - Final without video
Cryo 2014 CellSeal Presentation - Final without videoCryo 2014 CellSeal Presentation - Final without video
Cryo 2014 CellSeal Presentation - Final without videoEvonne R. Fearnot, MSBME
 
Cryopreservation
CryopreservationCryopreservation
Cryopreservationmegon94
 
Cryop ppt
Cryop pptCryop ppt
Cryop pptmegon94
 
Cryopreservation, germplasm storage
Cryopreservation, germplasm storageCryopreservation, germplasm storage
Cryopreservation, germplasm storageKAUSHAL SAHU
 
Cryopreservation of plant tissues.ppt
Cryopreservation of plant tissues.pptCryopreservation of plant tissues.ppt
Cryopreservation of plant tissues.pptPudhuvai Baveesh
 
Source of Microbes & Enzyme Immobilization
Source of Microbes & Enzyme ImmobilizationSource of Microbes & Enzyme Immobilization
Source of Microbes & Enzyme ImmobilizationAhmed Abdellatif
 
Dylan MacPhail Hounours Report
Dylan MacPhail Hounours ReportDylan MacPhail Hounours Report
Dylan MacPhail Hounours ReportDylan MacPhail
 
Cryopreservation and its application to aquaculture.pptx
Cryopreservation and its application to aquaculture.pptxCryopreservation and its application to aquaculture.pptx
Cryopreservation and its application to aquaculture.pptxNarsingh Kashyap
 
Cryopreservation, germplasm storage 2
Cryopreservation, germplasm storage 2Cryopreservation, germplasm storage 2
Cryopreservation, germplasm storage 2KAUSHAL SAHU
 

Similar a pub-thebigfreeze-IPT-sept15 (20)

Cryopreservation src
Cryopreservation srcCryopreservation src
Cryopreservation src
 
Cryopreservation
CryopreservationCryopreservation
Cryopreservation
 
Cryopreservation
CryopreservationCryopreservation
Cryopreservation
 
Cryopreservation of gamtes by bhawan.pptx
Cryopreservation of gamtes by bhawan.pptxCryopreservation of gamtes by bhawan.pptx
Cryopreservation of gamtes by bhawan.pptx
 
Cryopreservation
CryopreservationCryopreservation
Cryopreservation
 
Unit-IV Germplasm Storage and Cryopreservation.pptx
Unit-IV Germplasm Storage and Cryopreservation.pptxUnit-IV Germplasm Storage and Cryopreservation.pptx
Unit-IV Germplasm Storage and Cryopreservation.pptx
 
Sperm cryoperservation
Sperm cryoperservationSperm cryoperservation
Sperm cryoperservation
 
Cryopreservation.pptx
Cryopreservation.pptxCryopreservation.pptx
Cryopreservation.pptx
 
Spermcryoperservation by Dr.Chandan
Spermcryoperservation by Dr.Chandan Spermcryoperservation by Dr.Chandan
Spermcryoperservation by Dr.Chandan
 
Cryo 2014 CellSeal Presentation - Final without video
Cryo 2014 CellSeal Presentation - Final without videoCryo 2014 CellSeal Presentation - Final without video
Cryo 2014 CellSeal Presentation - Final without video
 
Cryopreservation
CryopreservationCryopreservation
Cryopreservation
 
Cryop ppt
Cryop pptCryop ppt
Cryop ppt
 
Surya kant agrawal
Surya kant agrawalSurya kant agrawal
Surya kant agrawal
 
Cryopreservation, germplasm storage
Cryopreservation, germplasm storageCryopreservation, germplasm storage
Cryopreservation, germplasm storage
 
Cryopreservation of plant tissues.ppt
Cryopreservation of plant tissues.pptCryopreservation of plant tissues.ppt
Cryopreservation of plant tissues.ppt
 
Source of Microbes & Enzyme Immobilization
Source of Microbes & Enzyme ImmobilizationSource of Microbes & Enzyme Immobilization
Source of Microbes & Enzyme Immobilization
 
Dylan MacPhail Hounours Report
Dylan MacPhail Hounours ReportDylan MacPhail Hounours Report
Dylan MacPhail Hounours Report
 
Cryopreservation
CryopreservationCryopreservation
Cryopreservation
 
Cryopreservation and its application to aquaculture.pptx
Cryopreservation and its application to aquaculture.pptxCryopreservation and its application to aquaculture.pptx
Cryopreservation and its application to aquaculture.pptx
 
Cryopreservation, germplasm storage 2
Cryopreservation, germplasm storage 2Cryopreservation, germplasm storage 2
Cryopreservation, germplasm storage 2
 

Más de biocision

pub_DDN 0514 BioCision Interview
pub_DDN 0514 BioCision Interviewpub_DDN 0514 BioCision Interview
pub_DDN 0514 BioCision Interviewbiocision
 
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
 

Más de biocision (6)

pub_DDN 0514 BioCision Interview
pub_DDN 0514 BioCision Interviewpub_DDN 0514 BioCision Interview
pub_DDN 0514 BioCision Interview
 
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-thebigfreeze-IPT-sept15

  • 1. Cryopreservation Technologies 44 Innovations in Pharmaceutical Technology Issue 54 By Rolf Ehrhardt and Maria Thompson at BioCision In order to maintain their peak functionality, therapeutic cells rely on being effectively frozen and thawed. This calls for increasingly optimised and standardised approaches to cryopreservation, as improvements in consistency and reproducibility will have a positive impact on patient outcomes The Big Freeze Keywords Cellular therapy Freezing devices Cryoprotectants Cell thawing The goal of cell cryopreservation is to suppress the natural processes of change and degradation inherent within any biological system,and to preserve viability,structure and function in as perfect a state as possible.For therapeutic cells,maintaining their peak achievable functionality is not merely a desirable outcome,but an obligation subject to the legal and ethical oversight of institutions such as the FDA and EMA.However,the sheer variety of cell types and their diverse biological characteristics defies a single method of culturing, preserving and preparing cells for clinical use.Nonetheless,standardising how cells are handled during these processes is important. Cryopreservation While an important part of the process,cryopreservation has not always been given the same consideration as other processing steps,like cell characterisation,potency and safety.Given that pharmaceutical companies must define their target patient population early on in drug development,functional activity and toxicity assays are understandably the focal point of due diligence when dealing with cellular products.Cryopreservation of products is often subject to somewhat less stringent treatment,which can make it a weak link in the cellular therapy manufacturing workflow. Therapeutic cells respond to the stress of cryopreservation in different ways,depending Image:©roganjosh–morguefile.com
  • 2. iptonline.com 45Innovations in Pharmaceutical Technology Issue 54 When cells are cooled below 0°C,extracellular ice crystals begin to form and relative solute concentration of the freezing media rises.Water flows out of the cell, beginning the process of cell dehydration.The cooling rate must be slow enough to allow sufficient egress of water to minimise the chance of intracellular ice crystal formation,while at the same time fast enough to avoid serious dehydration effects.It is for this reason that cryoprotectants are used:to lower the freezing point of the extracellular medium,limiting both ice crystal formation and the osmotic stress that results in dehydration.Researchers should only use freezing media that is both fully defined and validated to be optimal for their cell type.While it is possible that optimal cooling rates may vary by cell type,the current recommended freezing procedure for many therapeutic cells is that they are cooled gradually to approximately -80°C at a controlled rate of -1°C per minute (2).This cooling rate is proven to work well for peripheral blood mononuclear cells,primary cells,stem cells and cell lines. Freezing Devices Active control of the freezing process is particularly important in a regulated environment,where protocols must comply with GMP standards to deliver a functional product that is produced in controlled and predictable conditions.Retention of good functionality requires a reproducible,validated methodology. Until recently,this generally meant cryopreservation needed to be performed using large,expensive, programmable controlled rate freezers.Such freezers offer a precisely controlled cooling rate and are well-validated, but their cost, large footprint and maintenance issues are a prohibitive drawback, especially to small or mid-sized companies.In the last few years,passive freezing devices have gained in popularity,largely due to their ability to offer equivalent cooling rates and cell viability results,without the exorbitant costs and laboratory space required for controlled rate freezers.Passive cell freezing containers – particularly those that lower contamination risks and operating costs by being alcohol-free – are easily transferrable from the development phase to the manufacturing suite,and are increasingly being validated for use in GMP environments (3). Maintaining the Cold Chain Once cells and other biological products are safely frozen,there will inevitably come a time when they must be transported – either within a facility or to an end-user.Therapeutic cells,like any other biological material,are subject to the risk of temperature fluctuation during transport and,while regulatory on variables such as cell type,length of storage, cryoprotectants used,or freezing and thawing conditions.Optimising cryopreservation to meet the needs of cellular therapy can be a complicated process. For example,therapeutic cell viability is often assessed immediately post-thaw,without taking delayed- onset cell death into account.The same is true of cell function.The assays themselves may be well-defined, but checking functional activity immediately post- thaw may not give the best indication of what will occur in the patient.Some cell types require a post- thaw recovery period of several hours or even days before functionality is restored; others may be more dramatically affected by cryogenic storage conditions, and prone to changes in phenotype or sub-population frequency (1). The scientist must determine if cryopreserved cells maintain the same potency and function at the point of infusion into a patient as they did before they were frozen.Freezing and recovery media should be carefully chosen to match the cell type in question,and researchers need to ensure that the components of the media are fully defined.Perhaps the most important consideration for cryopreserving cells,particularly in a Good Manufacturing Practice (GMP) regulated environment,is to confirm that the cryopreservation methodology itself – and the technology supporting it – is fully optimised and standardised. Given the natural proclivity for cells to respond and adapt to their environment,there is an ongoing need for cryopreservation techniques to adapt in a way that minimises variability and subjectivity at every possible point when the process is transferred from development to manufacturing.The complete process includes freezing,cold chain management during transportation and thawing.Luckily,necessity informs invention,and as cellular therapy gains momentum, we are beginning to see a paradigm shift towards more optimised,standardised and translatable cryopreservation technologies. Cell Freezing This is the starting point of any cryopreservation workflow,and while pharma R&D teams may not always have the time and resources to devote to optimising freezing rates and media for each cell type,these factors will have an important influence on ultimate cell survival. Fortunately,a great deal of scientific effort has been devoted over the years to understanding the effects of various freezing parameters on cell survival and recovery. Generally speaking,freezing protocols are based on avoiding the physiological effects of dehydration and ice crystal formation.
  • 3. 46 Innovations in Pharmaceutical Technology Issue 54 iptonline.com containers,and are capable of maintaining temperatures below -50°C for over 24 hours.Liquid nitrogen dry vapour shippers can now be used in place of dry ice shipping,and can provide cooling for 10 days or more while fragile biospecimens are shipped.Other liquid nitrogen-based solutions exist that are more suitable for short-distance transport. The International Society for Biological and Environmental Repositories recognised a hand-portable liquid nitrogen-based carrier as their‘Outstanding New Product’of 2015.The CryoPod™ Carrier holds samples at below -150°C for up to four hours,while providing automatic temperature alarms,monitoring and logging,so that users will immediately be alerted if the temperature has been compromised at any point in the journey.Such products point towards a shift to more stringent,standardised temperature-control solutions. Re-Thinking CellThawing In the field of regenerative medicine,the final step in the cryopreservation workflow involves thawing the cryopreserved cells so they can be administered to the patient.Controlling the rate of thawing is as important as freezing,and optimising both is crucial to cell survival and function.Only recently,however,has cell thawing been given the same serious consideration that cell freezing has been subject to (7). During thawing,many of the internal processes of cell freezing are reversed.As cells are warmed,extracellular ice melts and water can rush into the cell,causing it to swell or even burst.If thawing is done too slowly,or heat application is uneven,tiny ice crystals formed during the freezing process can grow and become large enough to cause damage.The shock of transit back to a viable state can also cause oxidative stress,triggering apoptotic pathways and delayed onset cell death. Water Bath Technology By far the most common method of thawing cells is through the use of a heated water bath.When agencies promote guidelines for maintaining optimal temperatures during clinical studies and manufacturing processes,temperature control during transport is still poorly upheld.A recent US study found that the top six globally administered vaccines (including those for polio,diphtheria and measles) lose up to 36% of their effective shelf-life due to failures in meeting cold chain requirements,as measured by vaccine vial monitoring (4).And this is not an isolated report; another study found that most,if not all,vaccines are exposed to freeze damage risk at some point throughout the shipping process (5). Temperature fluctuation problems are not only limited to long-distance shipping;they can occur during cross- campus or cross-town transport,during transfer to and from liquid nitrogen storage,or even simply when cryostorage boxes or liquid nitrogen canes are shifted around to access specific samples.But while transport of temperature-sensitive biologicals may not be standardised,scientists are beginning to recognise that even short-term increases in temperature can expose cells to the dangers of thermal cycling and ice recrystallisation, lowering cell viability,recovery and,in the case of stem cells,even the ability to be pluripotent (6).The good news is,in recent years,several transportation and storage solutions have come to market that are aimed at more stringently controlling the cold chain,whether it be during long-distance transport or movement within a medical, research or biopharma manufacturing facility. NovelTransport Systems For a long time,homemade dry ice containers were the only method for shipping temperature-sensitive biomaterials.With the emergence of the cellular therapy industry,it became recognised that this type of container was too unreliable to entrust products representing a company’s major investment in time and money – not to mention what might be a patient’s best hope for survival.Recently,more robust dry ice-based transport systems – and even mobile dry ice-based workstations – have emerged to deliver more reliable temperature stability than conventional Styrofoam Temperature fluctuation problems are not only limited to long-distance shipping; they can occur during cross- campus or cross-town transport, during transfer to and from liquid nitrogen storage, or even simply when cryostorage boxes or liquid nitrogen canes are shifted around to access specific samples
  • 4. iptonline.com 47Innovations in Pharmaceutical Technology Issue 54 References 1. McKenna KC, Beatty KM, Vicetti Miguel R and Bilonick RA, Delayed processing of blood increases the frequency of activated CD11b+ CD15+ granulocytes which inhibit T cell function, J Immunol Methods 341: pp68-75, 2009 2. Bissoyi A, Nayak B, Pramanik K and Sarangi SK, Targeting cryopreservation-induced cell death: A review, Biopreservation and Biobanking 12(1): pp23-34, 2014 3. Stone M et al, Maximizing PMBC recovery and viability: A method to optimize and streamline peripheral blood mononuclear cell isolation, cryopreservation and thawing, BioProcess International, April 2015 4. Kartoglu U et al, Use of cool water packs to prevent freezing during vaccine transportation at the country level, PDA Journal of Pharmaceutical Science and Technology 63(1): pp11-26, 2009 5. Kumru O et al, Vaccine instability in the cold chain: Mechanisms, analysis, and formulation strategy, Biologicals 42: pp237-259, 2014 6. Deller RC, Vatish M, Mitchell DA and Gibson MI, Synthetic polymers enable non-vitreous cellular cryopreservation by reducing ice crystal growth during thawing, Nature Communications 5: p3,244, 2014 7. Thompson M, Kunkel E and Ehrhardt R, Balancing the Cryopreservation Equation, Cold Facts, January 2015 examined in the context of a GMP-regulated environment, this method is fraught with problems. Due to the fact that water is an efficient carrier of contaminating organisms, GMP regulations either prohibit water bath use, or specify that a water bath be cleaned and refilled after each use.This method of thawing can also be very subjective.The thawing endpoint is based on each operator’s observation – often determined by repeatedly lifting the vial out of the bath to peer at it – of when the last ice crystal is ready to melt. Often,no set times are applied and the researcher’s attention may be momentarily called away to another task, adding to the risk of variable endpoints and potential overcooking of cells.The problem is compounded when more than one sample is involved. In a clinical setting, cryobags are often used to freeze large volumes of cells.When these bags are thawed, the first bag may take just four minutes. However, the first bag cools the temperature of the water bath and then, when the second bag is added, it may take much longer. Again, the process is inconsistent and will lead to variable results. A more standardised approach would greatly benefit researchers, in-process manufacturing personnel and clinicians alike. An automated cell thawing system is now on the market that is designed to detect the solid-to-liquid phase change within a cryovial. The system is water free, small enough to fit inside a tissue culture hood and has already been validated in several labs that follow GMP regulations (3).The technology is adaptable to other vessel formats – for example, bags, bigger vials and multiple vials – giving it the versatility to be adopted into many different clinical models. Better Prospects Today, we are at a stage where the cellular therapy industry is just beginning to develop. As with all new endeavours, there are growing pains to be overcome before the infrastructure can be put into place. In addition, the creation of guidelines for the standardisation of workflow methods is still ongoing. Cryopreservation is one of the lynchpins of cellular therapy, and improvements in consistency and reproducibility will no doubt benefit patient outcomes down the line. Optimising cryopreservation will improve the safety and efficacy of cellular drug products. It is therefore important that we adopt tools and procedures that both reduce variability and streamline the process as we move forward. Dr Rolf Ehrhardt is the President and Chief Executive Officer of BioCision, a leading provider of innovative tools for standardising sample and biomaterial handling, cryopreservation and storage procedures. Previously, he was responsible for the early clinical development of a novel hepatitis C virus protease inhibitor at Intermune, was Vice President of Preclinical Development at Corgentech and founding President of BioSeek. Rolf 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 of Scientific Affairs at BioCision. She has a background in molecular genetics, and over 18 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, in addition to running APEX Think, a private consulting company. She has a BSc in Genetics and a PhD in Molecular and Cellular Biochemistry from the Royal London School of Medicine, UK. Email: maria@biocision.com