Cell and Gene Therapies: From Discovery to Delivery
eBook
Published: August 13, 2025

Credit: Technology Networks
Cell and gene therapies are changing what’s possible in modern medicine. But as these innovations accelerate, challenges around manufacturing, scaling up and ensuring consistent results are still major hurdles.
From dealing with unstable cell lines to figuring out how to manufacture therapies right at the point of care, scientists and developers are working through a complex and evolving field. Making these treatments more accessible, long-lasting and safe takes both technical know-how and big-picture thinking.
This eBook explores the latest solutions and expert insights that are helping to move advanced therapies forward.
Download this eBook to explore:
- How cell and gene therapies are evolving from experimental to mainstream
- Strategies to overcome manufacturing, resistance and delivery hurdles
- Insights into the future of scalable, personalized treatments across therapeutic areas
SPONSORED BY
Developments
in Point‑of‑Care
Manufacture of
Advanced Therapies
Current Challenges in
Cell Line Development
for Therapeutics
Why Are mRNA
Vaccines Not Giving
Long-Lasting Protection?
CELL AND GENE
THERAPIES:
From Discovery to Delivery
CONTENTS
5
Harnessing Cell Lines To Advance
Cancer Research and Treatment
9
Why Are mRNA Vaccines Not Giving
Long-Lasting Protection?
13
Improving the Efficacy and
Accessibility of CAR Therapies
17
Mass Spectrometry Imaging in
Pharmaceutical Development
21
Cell and Gene Therapy
Manufacturing
22
Current Challenges in Cell Line
Development for Therapeutics
27
New Purification Techniques in
Biopharmaceuticals
31
A Bright Future for RNA Therapeutics
34
Developments in Point‑of‑Care
Manufacture of Advanced
Therapies
CELL AND GENE THERAPIES: FROM DISCOVERY TO DELIVERY 3
TECHNOLOGYNETWORKS.COM
FOREWORD
Biotherapeutics are evolving rapidly, fueled by innovative science and collaborative ambition. This
eBook explores some of the most exciting frontiers in modern medicine, from the molecular precision
of RNA therapeutics to the logistical advancements enabling point-of-care manufacturing. These
topics highlight how diverse scientific breakthroughs are converging to reshape drug development
and patient care.
Inside, readers will discover the nuanced challenges of maintaining long-term vaccine efficacy, the
role of engineered cell lines in cancer research and how advances in CAR and NK cell therapies
are expanding the promise of immunotherapy. Articles also delve into enabling technologies like
mass spectrometry imaging and the latest purification techniques critical to scaling and refining
biopharmaceutical production.
Whether you’re an industry professional, academic researcher or healthcare innovator, this
eBook provides a valuable perspective on the current state and future direction of
biopharmaceutical development.
The Technology Networks editorial team
WHY CLINICAL RESEARCHERS TRUST ddPCR...Goals: Effective evaluation of cell and gene therapies in patientsClinical ResearchSerialMonitoringBiodistributionDose ResponseEvaluationKey Benefits Precision Accuracy Sensitivity Research Use OnlyWHY QA/QC SCIENTISTS TRUST ddPCR...Goals: Reliable assessments of purity, potency and safetyFinal QA/QCKey Benefits Precision Accuracy Sensitivity/ Specicity Time to resultsViral Titer and Empty-Full Capsid RatioMycoplasmaDetectionTransgeneCopy NumberResidual DNACell and gene therapies are continuing to gain popularity, with 431 now FDA approved and 4,000+ more in development.2 While these treatments are promising, managing safety and effectiveness in patients is complex. Around the world, groups ranging from drug discovery, development, and manufacturing to clinical laboratories are using Bio-Rad's Droplet Digital™ PCR (ddPCR™) technology as a reliable and scalable solution to myriad workflow challenges.Explore Bio-Rad's end-to-end solutions for developing and manufacturing cell and gene therapies at www.bio-rad.com/CGT1. https://bioinformant.com/u-s-fda-approved-cell-and-gene-therapies/2. https://www.asgct.org/global/documents/asgct-citeline-q1-2024-report.aspxAn End-to-End Cell and GeneTherapy Development PartnershipWHY CELL AND GENE THERAPY SCIENTISTS TRUST ddPCR....Goals: CMC submissions, efcient and scalable process developmentKey Benefits Precision Accuracy Inhibitor Tolerance/ Sensitivity MultiplexingViral Titer and Empty-Full Capsid RatioTransgene CopyNumberTransgeneExpressionIn Process TestingPlasmidIntegrity
5 CELL AND GENE THERAPIES
Credit: iStock/koto_feja
Harnessing Cell Lines To
Advance Cancer Research
and Treatment
Monica Hoyos Flight, PhD
Genetic variation affects the expression of genes and
function of proteins. DNA sequencing technologies
have uncovered millions of genetic changes associated
with various human traits and disease. However, it is
still largely unclear which of these changes are harmful
and how they cause problems. Efforts to understand the
effects of gene variants in cancer cell lines are shedding
light on cancer biology and drug resistance, and are
paving the way for personalized therapies.
This article examines some of the cell-based approaches
that researchers are using to study acquired drug
resistance and the effects of specific genetic variants
on cancer cell physiology. It also explores some of
the challenges associated with working with cells,
whether as model systems or for the production
of biotherapeutics, focussing on the biological
consequences of cell line engineering.
Developing cell lines to
understand acquired drug
resistance
Despite decades of medical advances, cancer is among
the leading causes of death worldwide. A significant
challenge in improving patient outcomes, particularly
of those with metastatic disease, is the development of
drug resistance.
Cancers that initially respond to treatment can later recur
in a resistant form.
TECHNOLOGYNETWORKS.COM
CELL AND GENE THERAPIES: FROM DISCOVERY TO DELIVERY 6
This is partly due to clonal evolution, a process through
which cancer cells expand and diversify by accumulating
mutations over time. As a result, even closely related
tumor cells may respond differently to the same therapy.
Moreover, anti-cancer therapies exert selective pressure,
accelerating clonal evolution processes and reducing the
efficacy of treatment.
Studying resistance mechanisms in tumor tissue is
challenging as it requires serial biopsies, which are
invasive and often not feasible. The use of liquid biopsies
has made monitoring cancer cell evolution easier, but
translating this information into treatment decisions
requires further research and clinical validation.
By growing cancer cells in the presence of anti-cancer
drugs, Martin Michaelis, professor of molecular
medicine at the University of Kent, aims to understand
what makes certain cells resistant to specific drugs and
what renders them vulnerable to others.
He has been looking after the Resistant Cancer Cell
Line (RCCL) collection since the 80s. The collection,
which originated in the laboratory of Jindrich Cinatl in
Frankfurt am Main, Germany, now comprises over
3,000 cell lines.
The cells are continuously exposed to increasing anticancer
drug concentrations to establish readily growing
sub-lines that show a clear resistance to the selection
agent. “Establishing a cell line resistant to classic cytotoxic
chemotherapeutics can take over a year,” Michaelis said,
“It is tough, expensive and labour intensive.”
Once the cells have acquired resistance to one drug, their
response to other drugs can be tested. “We often find
that cells that are resistant to one drug show collateral
vulnerability that makes them more sensitive to other
drugs,” he explained. Identifying biomarkers of these
cells could eventually help guide treatment decisions.
Over 100 research groups in academia and industry are
using the RCCL collection, applying omics methods to
characterize cells in depth and develop novel therapies
for patients who currently lack effective treatment
options. A lot of work is being done to understand the
rapid emergence of resistance to targeted therapies.
For example, patients with melanomas that harbor
the BRAF V600E mutation often respond very well
to BRAF inhibitors initially. But, resistance emerges
rapidly due to the development of compensatory
mechanisms within the cells.
Combining BRAF inhibitors with MEK inhibitors,
which target a related protein in the signaling pathway,
can mitigate this rapid resistance.1 This is reflected in
cell culture models where melanoma cells adapt to
BRAF inhibitor therapy much faster than to traditional
cytotoxic drugs.
Deciphering the mechanisms of resistance is no mean
feat. Michaelis and others have shown that every
resistance formation process is different. “If we take
one cell line and adapt it to the same drug 10–20
times, we get 10–20 different phenotypes and drug
resistance profiles,” he said. This diversity makes it
crucial to generate so many cell lines and support the
infrastructure required to maintain them.
High-throughput screening of cells
with cancer-associated mutations
Another way to study cancer cell biology and intrinsic
mechanisms of drug resistance, which are present before
any treatment is administered, involves systematically
knocking out or editing cancer-related genes. Geneediting
technologies are allowing researchers to
introduce these mutations into cell lines and examine
both their individual and combined effects on cancer
progression, metastasis and response to therapies.
Francisco Sánchez-Rivera, assistant professor of
biology at the Massachusetts Institute of Technology
(MIT) has been using prime editing, a refined version of
CRISPR technology, to examine the effects of genetic
variations on cancer cell growth and response to drugs.
Prime editing enables any kind of point mutation to
be introduced, as well as insertions and deletions, into
the DNA of living cells without creating double-strand
DNA breaks characteristic of traditional CRISPRCas9
methods. “With gene editing technologies it is
technically possible to understand the impact of every
single gene variant in every single coding or non-coding
TECHNOLOGYNETWORKS.COM
CELL AND GENE THERAPIES: FROM DISCOVERY TO DELIVERY 7
region in the human genome,” he said. “We are just
beginning to scratch the surface!”
His team has engineered over 1,000 variants of the TP53
tumor suppressor gene, the most frequently mutated
gene in cancer patients, to investigate their impact on
lung adenocarcinoma cells. By using a high-throughput
prime editing sensor strategy that couples prime editing
guide RNAs with synthetic versions of their cognate
target sites, they can quantitatively assess the effects
of genetic variants found in patients on cell function at
scale.2 Their work highlights the role of gene dosage in
influencing the natural proportions of proteins and their
interactions with each other.
Sanchez-Rivera is very enthusiastic about advances in
the field. “With CRISPR-based technologies we can
carry out experiments with remarkable precision and
efficiency; they are already allowing us to model and
dissect the aberrant circuitry of cancer cells.” Moreover,
as sequencing technologies increasingly become
integrated into clinical practice, this knowledge will
help tailor therapies to patients with tumors that have
a defined genetic makeup. “The next 5–10 years will be
truly transformative,” he said.
Challenges of working with
cell lines
In addition to the challenges associated with monitoring
the viability of cells, optimizing growth conditions and
avoiding contamination, the first major hurdles relate to
the efficiency of introducing genetic material into cells
(transfection) and controlling gene expression levels.
“We often have to test various delivery modalities,
because different cell types are more or less amenable
to transfection, and tweak and test different types of
functional editing reporters to ensure that our cell line
is capable of editing at very high efficiency,” Sanchez-
Rivera explained.
His team recently experienced significant challenges
trying to stably introduce cytosine and adenine base
editors into mouse B cell acute lymphoblastic leukemia
cells using lentiviruses. To overcome this issue, they
developed a strategy that couples stable viral guide RNA
delivery and transient mRNA electroporation. Using
this approach, they have produced single gene variantbased
cellular models as well as multiplexed scalable
mouse models in which thousands of genetic variants
can be examined in vivo.3
When working with genetically engineered cells, it
is important to consider the effect that the deleted,
modified or new gene will have on cellular resources as
this will influence the cells’ gene expression capacity.
“Every time you engineer cells with genetic constructs,
they draw on the cells’ enzymes, nucleotides and
ribosomes to express them,” said synthetic biologist
Francesca Ceroni. She and others have shown that
the introduction of genetic constructs can impose an
unintended “cost” or burden on mammalian cells.4, 5, 6
The diversion of cellular resources to transcribe and
translate new genes can lead to slower growth rates
and reduced expression, which can limit the production
of biotherapeutics. Ceroni’s group based at Imperial
College London focuses on characterizing the cellular
burden caused by recombinant protein production and
identifying design rules for synthetic systems to achieve
robust control of gene expression. They are working in
collaboration with several pharmaceutical companies to
optimize the production of biotherapeutics.
Addressing resource competition and cellular burden
is crucial for advancing cell engineering across various
applications, including disease modeling, bioproduction
and cell therapies. Ceroni calls for further research into
the implications of resource competition in primary cells.
“We need to further characterize the problem so that
we can then develop tools to address it.” Approaches to
precisely titrate the stoichiometry of gene expression
in mammalian cells, such as the host-independent and
programmable transcriptional system developed by Qin
and colleagues that can improve the production of viruslike
particles (VLPs),7 will contribute to improve the
production of biotherapeutics.
“The toolkit for controlling gene expression is limited,”
Ceroni said. By using machine learning to increase the
design space, her team are testing new components and
optimizing construct expression.
TECHNOLOGYNETWORKS.COM
CELL AND GENE THERAPIES: FROM DISCOVERY TO DELIVERY 8
When it comes to the production of biologics, this could
be key to reducing costs.
She concluded by emphasizing the importance of a
systems-thinking approach, especially when developing
biotherapeutics. “You can’t only look at the cells, the
interactions between different components of the
process (cells, growth conditions, cell engineering) need
to be carefully considered.”
REFERENCES:
1. Subbiah V, Baik C, Kirkwood JM. Clinical development of BRAF
plus MEK inhibitor combinations. Trends Cancer. 2020;6(9):797–
810. doi: 10.1016/j.trecan.2020.05.009
2. Gould SI, Wuest AN, Dong K et al. High-throughput evaluation
of genetic variants with prime editing sensor libraries. Nat
Biotechnol. 2024:1–15. doi: 10.1038/s41587-024-02172-9
3. Acosta J, Johnson GA, Gould SI et al. Multiplexed in vivo base
editing identifies functional gene-variant-context interactions.
bioRxiv. 2025. doi: 10.1101/2025.02.23.639770
4. Gabrielli J, Di Blasi R, Kontoravdi C et al. Degradation bottlenecks
and resource competition in transiently and stably engineered
mammalian cells. Nat Commun. 2025;16(1):328. doi: 10.1038/
s41467-024-55311-w
5. Di Blasi R, Gabrielli J, Shabestary K et al. Understanding resource
competition to achieve predictable synthetic gene expression
in eukaryotes. Nat Rev Bioeng. 2024;2(9):721–732 . doi: 10.1038/
s44222-024-00206-0
6. Jones RD, Qian Y, Ilia K et al. Robust and tunable signal processing
in mammalian cells via engineered covalent modification cycles.
Nat Commun. 2022;13(1):1720. doi: 10.1038/s41467-022-29338-w
7. Qin C, Xiang Y, Liu J et al. Precise programming of multigene
expression stoichiometry in mammalian cells by a modular
and programmable transcriptional system. Nat Commun.
2023;14(1):1500. doi: 10.1038/s41467-023-37244-y
MEET THE INTERVIEWEES:
Martin Michaelis is a professor of molecular medicine at the
University of Kent, Canterbury, UK, and a research group leader
at the Dr Petra Joh Research Institute in Frankfurt am Main,
Germany. His research focuses on the investigation of acquired
drug resistance in cancer using the Resistant Cancer Cell Line
(RCCL) collection, a unique resource consisting of more than 3,000
drug-adapted cancer cell lines. Additional research interests
include the identification of virulence mechanisms and therapeutic
targets in viruses, and meta-research on practices in the life
sciences, with a focus on reproducibility and ethical standards.
Francisco Sánchez-Rivera, is an assistant professor of biology
at the Massachusetts Institute of Technology (MIT). His group
studies the cellular and molecular mechanisms by which genes
and disease-predisposing mutations influence the development
of diseases like cancer. To do so, they are applying CRISPR-based
methods to engineer mutations with high efficiency and precision
in cells and tissues of living animals to quantitatively interrogate
cancer variants at scale and pave the way for novel therapeutic
strategies.
Francesca Ceroni is a senior lecturer in the Department of
Chemical Engineering at Imperial College London, UK. She sits
on the Imperial College Centre for Synthetic Biology Management
Board and between 2018 and 2021 she was a member of the
World Economic Forum Expert Network. Her research focuses on
understanding the complexity of genetic regulation by building
synthetic systems from the bottom up. Her team works on
genetically engineered bacterial and mammalian cells to develop
innovative approaches to mitigate resource competition and
burden, and improve construct performance.
9 CELL AND GENE THERAPIES
Credit: iStock/MicroStockHub
Why Are mRNA Vaccines
Not Giving Long-Lasting
Protection?
Aron Gyorgypal, PhD
Why do mRNA vaccines struggle to provide lasting
protection, and could the absence of bone marrow longlived
plasma cells explain the waning antibody levels and
provide insight for durable vaccine development?
Viral infections, reinfections
and immunity – SARS-CoV-2 as
an example
Viral infections can spread through human-to-human
interaction when viral particles enter the body via
mucous membranes, such as those in the nose, mouth
or eyes, as well as through open wounds. Briefly, the
cascade of events for respiratory viruses goes as follows:
viruses infect the airway by binding to specific receptors
on our cells within the respiratory tract; for example, the
ACE2 receptors on the pneumocytes within the alveoli
are targeted by SARS-CoV-2. The subsequent targeting
allows the viral pathogen to enter the cell and release
its genetic material, hijacking the cell’s machinery to
cause viral replication. The immune system recognizes
the immunogenic viral RNA produced through pattern
recognition receptors and arms the innate immune
system. Antigen-presenting cells (APCs), such as
dendritic cells, take in antigens and present them
to CD4+ T cells, priming them; likewise, B cells also
recognize antigens, internalizing them and presenting
them on MHC II receptors on their surface. The B and
T cells then undergo linked recognition, producing
memory B and T cells and plasma cells, leading to
adaptive immunity.
TECHNOLOGYNETWORKS.COM
CELL AND GENE THERAPIES: FROM DISCOVERY TO DELIVERY 10
These plasma cells are required to produce antibodies
against the virus that produce protection against
reinfection.
It is known that long-lived bone marrow plasma
cells are needed to produce protective antibodies
against reinfection. It is also known that patients
who have recovered from COVID-19 initially have
a substantially lower risk of reinfection with SARSCoV-
2.1 Nonetheless, anti-SARS-CoV-2 antibodies
decrease rapidly within the first few months after
infection. Literature has shown that the anti-spike
proteins of SARS-CoV-2 diminish rapidly from
circulation within the first four months and then
more gradually in the subsequent seven months postinfection.
2 A major concern during the pandemic was
that this loss in humoral antibodies would correlate to
a loss in protection against the virus. However, Turner
and colleagues showed that COVID-19-recovered
individuals indeed had circulating memory B cells
directed against SAR-CoV-2 and that the longevity of
serum antibodies is not the only factor in determining
durable immunity against reinfection as memory B cells
could rapidly differentiate into antibody-secreting cells
during virus re-exposure.3 This provided evidence that
natural infection to SARS-CoV-2 indeed establishes
two armed humoral immune memory through long-lived
plasma B cells and memory B cells.
Protecting against infections with
vaccination
However, what about protecting against a viral infection
even before the first exposure?
The first commercially produced and FDA-approved
mRNA vaccine was against the SARS-CoV-2 within
the United States. While the usual production of a
conventional vaccine takes 10–15 years, the COVID-19
vaccine was developed in less than one year, raising
concerns about safety and efficacy. A meta-analysis
from 2024 concluded that of the vaccines tested,
mRNA and inactivated-based vaccines had the greatest
effect after the first and second dose, with mild local
and systemic adverse effects and a very rare rate of
adverse effects.4
What has been seen, however, is that without mRNA
vaccine boosters, the rate of reinfection increases as these
vaccines do not protect against new variants.5 Another
concern is the efficacy of the vaccines to produce longterm
durable protection, like natural immunity.
How mRNA vaccines interact with
the immune system
There is a concern about the lack of durable protection
from mRNA vaccination. To understand why, we need
to delve into the engineering of the mRNA vaccine as
well as the hypothesized method of immune response
that produces protection.
The mRNA vaccines consist of a 5’cap attached to
the 5’UTR, followed by the coding sequence for the
SARS-CoV-2 spike protein, terminated by a 3’UTR
and a poly-A tail. This mRNA is encapsulated in a lipid
nanoparticle (LNP) and stabilized by polyethylene
glycol (PEG). After intramuscular injection of the
vaccine, the mRNA-LNP is taken up by APCs, such
as dendritic cells, and trafficked to the lymph nodes.
Here, they prime both CD4+ and CD8+ T cells. CD8+
induces the production of cytotoxic T cells, which
destroy infected cells, while the antigen-primed CD4+
T cell differentiates into Th1 cells or T follicular helper
(Tfh) cells. The Tfh cells help to initiate a germinal
center reaction, which results in the formation of
affinity matured memory B cells and antibody-secreted
long-lived plasma cells.6 The desired outcome of the
vaccination regimen is to produce long-lived plasma B
cells that can survive for years, producing neutralizing
antibodies against the viral antigen as well as the
memory B cells, which, when re-exposed to the antigen,
give rise to new high-affinity antibody-secreting cells
capable of neutralizing the antigen.
Nevertheless, this is not the case. In recent years, during
the COVID-19 pandemic, protective antibodies for
immunized individuals waned over the months after the
completion of the vaccination regimen. Should the longlived
plasma B cells not be active and re-confer protection
TECHNOLOGYNETWORKS.COM
CELL AND GENE THERAPIES: FROM DISCOVERY TO DELIVERY 11
against the virus? Yes, that is the function of the adaptive
immune system, yet this seems not to be happening.
Why mRNA does not produce
durable protection, a hypothesis
The mechanism by which mRNA vaccination generates
an adaptive immune response and subsequent immunity
is unknown. Giannotta and Giannotta7 argue that there
is an immune memory problem in which long-lived
plasma B cells are absent.
What is known is that the mRNA-LNP is taken up
by APCs and is found at the site of injection and the
draining lymph nodes, showing the translation of the
mRNA cargo into spike protein. This confers that
the mRNA is reaching the cytosol within the cells.
Monocytes will express the antigen on their surface,
typically done by bone marrow-derived APCs to prime
CD8+ T cells using MHC class I molecules. However,
mRNA-LNP is also taken up by non-immune cells as an
off-target effect; these non-immune cells then generate
proinflammatory cytokines and chemokines at the site
of immunization, which can trigger antigen-specific
antibodies and induction of CD8+ T-cell responses
through cross-presentation. Antigen-specific CD8+ T
cells are the main determinant for immune protection.
These CD8+ cells, along with memory B cells, block
the progression of infection. However, one major issue
with vaccination is that CD8+ T cells produce a low
concentration of CD38 versus that of natural infection.8
As CD38 regulates the infection-induced regulatory
process to induce an adaptive immune response, this
could be a reason for a differential response and lack of
long-lived protection.
Moreover, it is not known exactly how CD4+ T cells
are activated. The SARS-CoV-2 mRNA-LNP induces
proinflammatory and functional CD4+ and CD8+ T cells
and a Th1 response. However, these mRNA-LNPs only
encode a truncated version of the spike protein. T cells
do not recognize the virus. CD8+ T cells recognize only
virus-infected cells, and CD4+ T cells recognize viral
antigens of infected cells. Conceptually, only long-lived
plasma B cells can recognize the virus. However, there
may be some reasons long-lived plasma B cells are not
present after vaccination, in the same way as they are
from natural infection:
∙ Cross-reactive T-cell immunity – SARS-CoV-2
shares a broad CD4+ and 8+ T-cell cross-reactivity
with human endemic coronaviruses and evokes
a secondary response to cross-reactive epitopes.
This could eradicate the mRNA-LNP SARS-CoV-2
infection immediately.
∙ CD4+ cell-mediated memory – Cross-reactive T cells
against common coronavirus may become activated,
limiting the mRNA-LNP SARS-CoV-2 infection.
In other words, pre-existing spike-cross- reactive
T cells may be activated, limiting the infection. And
as T-cell activation is required for germinal center
development, this may intervene with the vaccination.
∙ Cross-reactive memory B cells and re-exposure –
Memory B cells can defend against infection but
not prevent it, and upon re-exposure to the virus/
antigen, if no long-lived plasma cells are present, the
memory B cells are recalled. Cross-reactive memory
B cells may become activated upon mRNA-LNP
SARS-CoV-2 infection.9
∙ Waning levels of anti-spike antibody levels – Although
waning levels of antibody-mediated protection
is expected. During the outbreak of the delta
variant, highly vaccinated populations did not
see protection, causing serious illness and, in
some, death.10
mRNA vaccines do not produce
long-lived plasma B Cells
Their hypothesis was correct – Recently, Nguyen and
colleagues published in Nature Medicine that the waning
of SARS-CoV-2 specific antibodies could be accounted
for by the absence of bone marrow long-lived plasma
cells after vaccination.11 To do this, the team recruited 19
healthy adults between 2.5–33 months after receiving a
SARS-CoV-2 vaccine and measured influenza, tetanus
and SARS-CoV-2 antibody-secreting cells within the
bone marrow. While the influenza and tetanus-specific
TECHNOLOGYNETWORKS.COM
CELL AND GENE THERAPIES: FROM DISCOVERY TO DELIVERY 12
IgG titers correlated with long-lived plasma cells, serum
levels for SARS-CoV-2 did not. It is widely assumed that
long-lived plasma cells come from memory B cells, yet
SARS-CoV-2 vaccination fails to imprint the required B
cell phenotype even 33 months after vaccination.
However, it is important to note that the study only
looked at 19 healthy individuals who self-reported their
vaccination status. It is also important to note that bone
marrow antibody-secreting cells are rare, and bone
marrow aspirates may be a better sample for this type
of interrogation.
Nonetheless, the reasoning behind the failure to
produce long-lived plasma cell response is unknown.
mRNA Vaccines, what is next?
There must be a way to produce durable protection
using mRNA vaccines, specifically producing longlived
plasma cells within the bone marrow. That may
be done by altering the vaccination regimen. Studies
looking at increasing the mRNA dose within the LNP to
overcome the limits of anti-LNP antibodies and crossreactive
memory B cells may be an option. The addition
of adjuvants to augment the immune response could
also be tried. Alternatively, the engineering of different
spike proteins could be tried to alter the response. For
the foreseeable future, it may be necessary to administer
mRNA vaccine boosters for high-risk individuals every
4–6 months to provide protection.
REFERENCES:
1. Lumley SF, O’Donnell D, Stoesser NE, et al. Antibody status and
incidence of SARS-CoV-2 infection in health care workers. N Engl J
Med. 2021;384(6):533-540. doi: 10.1056/nejmoa2034545
2. Seow J, Graham C, Merrick B, et al. Longitudinal observation
and decline of neutralizing antibody responses in the three
months following SARS-CoV-2 infection in humans. Nat Microbiol.
2020;5(12):1598-1607. doi: 10.1038/s41564-020-00813-8
3. Turner JS, Kim W, Kalaidina E, et al. SARS-CoV-2 infection
induces long-lived bone marrow plasma cells in humans. Nature.
2021;595(7867):421-425. doi: 10.1038/s41586-021-03647-4
4. Beladiya J, Kumar A, Vasava Y, et al. Safety and efficacy of
COVID-19 vaccines: a systematic review and meta-analysis
of controlled and randomized clinical trials. Rev Med Virol.
2024;34(1). doi: 10.1002/rmv.2507
5. Tartof SY, Slezak JM, Puzniak L, et al. Effectiveness of BNT162b2
BA.4/5 bivalent mRNA vaccine against a range of COVID-19
outcomes in a large health system in the USA: a test-negative
case–control study. Lancet Respir Med. 2023;11(12):1089-1100. doi:
10.1016/S2213-2600(23)00306-5
6. Bettini E, Locci M. SARS-CoV-2 mRNA vaccines: immunological
mechanism and beyond. Vaccines. 2021;9(2):1-20. doi: 10.3390/
vaccines9020147
7. Giannotta G, Giannotta N. mRNA COVID-19 vaccines and long-lived
plasma cells: a complicated relationship. Vaccines. 2021;9(12). doi:
10.3390/vaccines9121503
8. Oberhardt V, Luxenburger H, Kemming J, et al. Rapid and stable
mobilization of CD8+ T cells by SARS-CoV-2 mRNA vaccine.
Nature. 2021;597(7875):268-273. doi: 10.1038/s41586-021-03841-4
9. Song G, He W ting, Callaghan S, et al. Cross-reactive serum and
memory B-cell responses to spike protein in SARS-CoV-2 and
endemic coronavirus infection. Nat Commun. 2021;12(1). doi:
10.1038/s41467-021-23074-3
10. Shitrit P, Zuckerman NS, Mor O, Gottesman BS, Chowers M.
Nosocomial outbreak caused by the SARS-CoV-2 Delta variant in
a highly vaccinated population, Israel, July 2021. Eurosurveillance.
2021;26(39). doi: 10.2807/1560-7917.ES.2021.26.39.2100822
11. Nguyen DC, Hentenaar IT, Morrison-Porter A, et al. SARS-CoV-
2-specific plasma cells are not durably established in the bone
marrow long-lived compartment after mRNA vaccination. Nat
Med. 2025;31(1):235-244. doi: 10.1038/s41591-024-03278-y
13 CELL AND GENE THERAPIES
Credit: iStock/artacet
Improving the Efficacy and
Accessibility of CAR Therapies
Kate Harrison, PhD
Chimeric antigen receptor (CAR) T cells have seen
enormous success in cancer therapy, with seven
therapies now approved for clinical use. In CAR
T-cell therapy, T cells are harvested from patients
and genetically altered to express CARs that strongly
recognize cancer cell or tumor antigens. Enhanced cells
are then expanded in vivo and transfused back into the
patient to attack the cancerous cells.
However, despite the successes of CAR T cell therapy, it
still poses multiple challenges to widespread adoption,
including potentially severe side effects such as
cytokine release syndrome and neurotoxicity, limited
efficacy for solid tumors and complex ex vivo processes.
As a result of these challenges, CAR T-cell therapy can
also be prohibitively expensive, often costing hundreds
of thousands of dollars per patient and requiring
hours of care.
Technology Networks recently had the pleasure
of speaking with Dr. Neil Sheppard, director of
the Therapeutic Innovation in Natural Killer cells
(THINK) lab at the Perelman School of Medicine,
University of Pennsylvania, whose research focuses
on the development of CAR NK-cell therapies and the
improvement of CAR T-cell therapies. He discussed
the challenges involved in developing new CAR-based
treatments, what the future holds for the field and how
effective and safe immunotherapeutics can be made
accessible and affordable to everyone.
Q: Can you give us a brief overview of your
current research and where your focus lies?
A: My lab is called the Therapeutic Innovation and NK
cells (THINK) lab. We mostly work on natural killer
cells and their application in cancer immunotherapy,
although we also do still work on T cells – they’re both
very interesting tools, suitable for different jobs.
TECHNOLOGYNETWORKS.COM
CELL AND GENE THERAPIES: FROM DISCOVERY TO DELIVERY 14
Within our focus on NK cells, we have several project
categories. Our biggest project focuses on optimizing
the NK cell “chassis” – for example, removing
checkpoints or factors involved in exhaustion, to
improve them as therapeutics. To do this, we’re using a
form of gene editing called base editing, which allows
you to do multiplex edits to a cell without damaging
it, by changing one base into another. In comparison,
older tools like CRISPR can break the backbone of
DNA, causing extreme translocations or inducing death
by apoptosis. We can also achieve the same end by
programming the cell as it grows, for example with small
molecule compounds that push the cells down a certain
path or lock them into a certain beneficial state. We
hope that with some programming with small molecules
or cytokines and some gene editing, we can create an
ideal NK cell that can deliver a therapeutic transgene,
such as a CAR.
We’ve also spent a lot of time working on NK
cryopreservation. Unlike T cells, NK cells don’t
cryopreserve very well – but if you can’t cryopreserve
them, you don’t have a product. A lot of the trials that
have been done so far with CAR NK cells have been
single-center trials, where altered NK cells have been
infused right away, as a fresh product, which can’t really
be commercialized.
Beyond that, I think successful immunotherapy is
rarely going to be single agent. I see a lot of benefits
for combination immunotherapy: CAR cells and other
agents. We’re just wrapping up a clinical trial, the first
in the world to use CAR T cells with oncolytic viruses
together, and now we’re designing our own oncolytic
viruses to use with NK cells. Cancer has a wound
healing phenotype that doesn’t usually attract NK cells.
Instead, tumors tend to collect macrophages, then
reprogram them to make the tumor worse. They also
collect T regulatory cells, which can create a barrier to
successful immunotherapy. But if you can infect the
tumor with an oncolytic virus, you’ve created the perfect
context to attract CAR T, or CAR NK cells, maximizing
exposure to the target tissue.
Our final angle is radiotherapy. Radiotherapy kills
cancer cells, but it also causes stress and DNA damage
in the cells it doesn’t kill. Stress and DNA damage
attract NK cells, so we think that that could increase
engagement with CAR NK cells. Those are all the
things my lab is working on, and we’re also working
towards putting a CAR NK cell product in the clinic for
glioblastoma in a few years.
Q: What are the advantages of NK cells as
therapeutics compared to CAR T cells?
A: CAR T cells are potentially very powerful cells,
because they’re all very highly antigen-specific, but
they’re almost too good at doing their job. They
produce high levels of cytokines, replicate rapidly
and can cause a lot of damage, resulting in potentially
life-threatening side effects. The two most common are
cytokine release syndrome (when too many cytokines
are produced, causing an immune cascade) and immune
cell-associated neurotoxicity syndrome (ICANS)
(excess central nervous system inflammation and
disturbance caused by excessive cytokine production).
Unfortunately, this means that CAR T-cell therapy can
only be given in tertiary medical centers (of which there
are only around 30 approved centers in the US), to an
admitted patient with available space in an emergency
room. This limits the accessibility and availability
of CAR T cell treatments, as does the cost of these
treatments, and the fact that the T cells must be taken
from the patient’s own body.
NK cells provide an alternative in several ways. Since NK
cells don’t undergo large clonal expansions and produce
a different cytokine profile from T cells, they are less
likely to trigger such severe side effects. Additionally, NK
cells don’t cause graft-versus-host disease, so they could
be made into an allogenic therapeutic. There have been
several ongoing projects using CAR NK cells against
CD19 that have shown great results, but also show that
they can be used as a safe allogenic therapeutic, without
causing CRS or ICANS. Therefore they could potentially
be given in outpatient centers or community hospitals, at
a lower cost, making them far more accessible than CAR
T-cell therapies.
In addition, T cells are so antigen-specific (either via
their own T-cell receptor or a CAR) that if they don’t
see their specific antigen, or the cancer mutates, tumor
escape can occur.
TECHNOLOGYNETWORKS.COM
CELL AND GENE THERAPIES: FROM DISCOVERY TO DELIVERY 15
In comparison, NK cells have evolved to recognize
dozens of different stress signals, such as the
consequences of DNA damage and metabolic stress,
which are almost universal in cancer. This potentially
gives CAR-NK cells a very broad activity that could
help prevent tumor escape.
Q: What are some of the challenges
involved in developing novel CAR -based
therapies?
A: One of the shared challenges (though less relevant
to NK cells because of their broad activity) is antigen
selection, especially for solid tumors. We were lucky
with the hematological malignancies, there were
obvious targets in the B cell markers. In solid tumors,
we’re targeting tumor-associated antigens that are
over-expressed on tumors but are still present in healthy
tissues. This leads to untargeted off-tumor toxicity,
which can limit the dose of cell therapies.
We also have to deal with antigenic heterogeneity
and antigen escape. Whatever your target antigen,
expression won’t be uniform and the likelihood is
that a percentage of tumor cells won’t be destroyed
and can potentially repopulate. You need to engage
multiple antigens to solve this problem, but that also
potentially means stacking toxicities. We haven’t seen
many CAR NK cells for solid tumors enter clinical
testing yet, but one antigen that’s received attention
recently is claudin18.2 (CLDN18.2) in pancreatic and
esophageal cancers. CLDN18.2 is usually expressed
in tight junctions, not really visible to the immune
system. Tumors depolarize membranes, so they’re no
longer neatly stacked, and CLDN18.2 becomes visible
– making it a great antigen to target. However, even this
isn’t foolproof; high doses of CLDN18.2 CAR T cells
can cause gastric bleeds – an on-target toxicity.
We certainly have our work cut out for us to design
effective CARs that don’t cause toxicity. We’re trying
to make these CARs smarter, but we also need to make
sure that once they’re inserted into the target cell, they
don’t overload it, or domain swap with each other to
cause tonic signaling (low level activation in the absence
of target antigen), or cause any secondary malignancies.
Q: How do you consider challenges like
toxicity when moving your therapies
towards a clinical trial? How do you think
these safety concerns can be overcome
entirely in future therapies?
A: There’s a lot that the regulators expect you to do
pre-clinically in order to get an investigational drug
application approved in the US (or a clinical trial
application in the UK and elsewhere). One very
important thing is to screen your CAR for off-target
toxicity, to make sure it isn’t going to cross react with a
second tissue or antigen and cause damage.
The clinical trial design provides a lot of levers to control
safety – for example, starting with a low dose of CAR
without lymphodepletion, with staggered administration
and dose escalation. Lymphodepletion is usually needed
to give physical space for your CAR cells to occupy, and
to encourage the body to return to homeostatic immune
cell levels by producing cytokines that encourage
immune cell expansion. Without lymphodepletion,
expansion of CAR T or CAR NK cells is very limited,
reducing the chance of damaging side effects or
toxicities. It’s also important to have treatment for
toxicities readily available to mitigate any issues. Overall,
patients are very carefully managed during trials.
One potential way of overcoming safety issues like
toxicity is to design solutions into the cell therapy.
CARs can be designed with on- or off-switches that
can be manipulated using small molecule drugs or
monoclonal antibodies. Kill-switches are also possible,
which – when the controlling small molecule is
administered – will wipe out any CAR cells. However,
there is then no chance of patient benefit then because
the cell therapy is eliminated. It's much better to have
safety by design, rather than an emergency off-button.
There are a lot of options for safety management, but
some of them are more precise than others. If our goal is
to make these therapies accessible in outpatient clinics,
we need something that is very smart, very safe and
hopefully manages itself, without the need for a lot of
expert physicians to control it.
TECHNOLOGYNETWORKS.COM
CELL AND GENE THERAPIES: FROM DISCOVERY TO DELIVERY 16
Q: What does the future hold for CAR
therapeutics?
A: Like any class of drugs, CAR therapies don’t have
a right to exist, they could be swept away by future
innovations. We’ll always do what's simple and what
works best, and some things are just too complicated.
Recently, we've seen a big pullback from gene therapy,
despite the curative promises, because the drugs were
prohibitively expensive. So, we need to fix the financial
toxicity of CAR cells as well as the medical toxicities.
One of the most exciting things happening at the
moment is programming the patient’s own cells in vivo,
by delivering CARs using viral vectors or mRNA and
lipid nanoparticles. There are already multiple trials
in progress that are looking very promising for B cell
malignancies. For successful use of CAR therapries
against solid tumors we need to really work on
improving the cell chassis – like we’re doing in our lab
– to make the immune cells more robust, more resilient
to the immunosuppressive tumor microenvironment.
This would be ex vivo cell therapy, but it could still be
allogenic and off-the-shelf, which would greatly improve
the cost and the safety.
There are still some challenges to overcome, but I really
see in vivo CAR therapeutics as the future. Engineered,
gene-edited therapies for solid tumors are going to be
necessary in order to get the kind of successful, longterm
results we need.
ABOUT THE INTERVIEWEE
Neil C. Sheppard, D.Phil. (Oxon), leads a lab focused on natural
killer (NK) cell biology and cancer therapies at the University of
Pennsylvania’s Center for Cellular Immunotherapies (CCI). Educated
in the UK at the Universities of Bristol and Oxford, Neil has over
20 years of international experience with immunotherapies and
vaccines in academia and industry. He has led programs, team,
and labs in the UK, Australia, California and Pennsylvania. He has
advanced multiple CAR-T and TCR-T programs through successful
INDs into Ph1 FTIH or Ph1b combination studies. Neil was
commended by SITC in 2020 and Marquis Who's Who in 2022 for
Innovation in Immunotherapy.
17 CELL AND GENE THERAPIES
Credit: iStock/Suriphon Singha
Mass Spectrometry Imaging in
Pharmaceutical Development
Joanna Owens, PhD
Mass spectrometry imaging (MSI) technology has
existed in simple form for decades, but recent advances in
MS sensitivity and data analysis means it’s finally coming
of age.1 In this article, we explore how MSI is evolving
into a high-resolution spatial biology toolset to transform
the traditional model of drug discovery and development.
The evolving role of MS in drug
development
MS technologies play a crucial role in drug discovery
and development, serving as effective tools for the swift
identification and quantification of complex molecules.2
Modern MS methods have greatly enhanced the ability
to analyze low levels of potential drug molecules
– from oligonucleotides, to peptides, proteins and
small molecules, and are now used across the drug
development pipeline from target discovery, compound
screening, and toxicity and quality control testing2, as
outlined in Table 1.
Many of these applications have traditionally involved
bulk analysis of cells or tissues, but advances in MSI
technologies mean it’s now possible to carry out
spatial chemical analysis at the single-cell level. This
advancement is paving the way for novel applications in
drug discovery and development.
What is MS imaging?
“Mass spectrometry imaging is a way to look at
metabolism in a spatial manner, which gives you much
more information than just analyzing the bulk tissue,”
explained Professor Brent Stockwell of Columbia
University, USA. “Traditionally, you would administer a
drug to an animal, analyze the bulk liver tissue and get a
concentration of the drug at a particular timepoint, and
TECHNOLOGYNETWORKS.COM
CELL AND GENE THERAPIES: FROM DISCOVERY TO DELIVERY 18
this gives you useful information, but this cannot tell
you which cells the drug accumulates in and what the
impact of the drug was in terms of other metabolites and
measures of the cell state.”
MSI achieves this by chemically analyzing snapshots,
called a pixel, at a single point within a tissue. For each
pixel, molecules are extracted and introduced into
the ionization source of the MS instrument, which
separates every molecule by its mass-to-charge ratio
and determines their relative abundance in the sample.
From this, you can visualize the abundance of different
molecules across the tissue.
“There’s an inherent trade-off in the MS imaging
approach: if you use a bigger pixel size, you cover more
tissue but you get less spatial resolution to be able to see
how patterns vary,” said Stockwell.
“However, more sensitive MS instruments together
with better ionization methods are making it possible
to reliably detect minute amounts of sample. This,
combined with improved software for data analysis and
annotation, is expanding the uses for MS imaging.”
The evolution of MS imaging
MSI was first developed more than 50 years ago using
secondary ion mass spectrometry (SIMS), and the most
widely used MSI method today is MALDI.1 But despite
its widespread use, MALDI-based MSI is not without its
limitations – the fragmentation of biological molecules
and careful consideration of the matrix for each
study are a few examples.1 Other ionization methods
have been used for MS imaging, such as desorption
electrospray ionization (DESI) and nanoDESI which
make it possible to analyze non-volatile molecules
Table 1: Applications of mass spectrometry imaging across different stages of R&D.
Stage of drug R&D Example applications
Discovery ∙ MS-based proteomics and metabolomics on clinical tissue samples are used
for target identification/validation and to gain insights into a drug’s mechanism
of action.3
∙ MS plays a pivotal role in determining the structure and characteristics of potential
drug compounds, many of which are combinatorial-chemistry synthesis products.2,3
∙ MS has increasingly become the method of choice in high-throughput or ultra-highthroughput
screening.3,4
Development ∙ In drug development, liquid chromatography-mass spectrometry (LC-MS) is
an essential workhorse technology used to identify and characterize the active
drug and its metabolites in different tissue samples to establish the absorption,
distribution, metabolism and excretion (ADME) profile of a drug candidate.5,6,7
∙ MS is also important for identifying, monitoring and validating biomarkers
throughout drug development.8
Quality control ∙ MS technologies are critical tools for ensuring the quality of traditional smallmolecule
drugs during formulation and manufacturing.
∙ MS is increasingly used to confirm the identity, biophysical properties and purity of
biopharmaceuticals such as monoclonal antibodies.9,10
TECHNOLOGYNETWORKS.COM
CELL AND GENE THERAPIES: FROM DISCOVERY TO DELIVERY 19
without fragmentation.1 Still, it was not until recently
that the technology matured to a point that allows for
high-impact studies.
“One downside to conventional MALDI methods is the
chemical preparation you need to use to be able to detect
certain molecules,” explained Professor David Muddiman,
of North Carolina State University, USA. Muddiman has
spent nearly two decades developing a less destructive
ionization approach called matrix-assisted laser desorption
electrospray ionization (MALDESI) which makes it
possible to directly analyze a diverse range of biological
molecules without requiring chemical derivatization in a
tissue.1 “For example, neurotransmitters are tiny molecules
that the matrix in MALDI interferes with, so you need
to treat them with a reagent to bring them into a higher
mass range. With MALDESI-based MS imaging, you can
analyze the endogenous neurotransmitters in the tissue
without any pre-treatment.”
The same applies to glycans – carbohydrate groups
added co- and post-translationally to proteins that play
crucial roles in biology - which are easily fragmented
during conventional MALDI-MS. “If you look in the
literature at MS imaging using MALDI, there are
very few instances where analysis has been able to
identify sialic acids – terminal monosaccharides on
carbohydrates – and there’s no method to recover that
information through bioinformatics. This is because
there’s no way of knowing what the original molecule in
the sample was. With our method, we are reading back
authentically the biology that’s been presented to us.”
MS imaging opportunities in drug
development
The ability to track the single-cell pharmacokinetics
and pharmacodynamics of a drug using MS imaging has
been coined by Stockwell as “spatial pharmacology”.11
“Spatial biology methods such as spatial transcriptomics
or proteomics can give you information about the cell
types and cell communities and the state of individual
cells,” he explained. “But right now, there’s a layer of
information you can’t get directly from those methods
– and that’s the small-molecule products of those
metabolic reactions. But with MS imaging you can see
the abundance of those metabolites across the tissue in
a spatially defined manner.”
In the long term, there could be a potential application
for MSI in late-stage drug development, where
MALDI-based MS is currently the gold standard for
characterizing the distribution of drug candidates in
development. In the short term, there is a clear role for
MSI in the drug discovery pipeline. “I think the demand
right now for MS imaging is in looking at disease models
versus healthy normal tissues,” noted Stockwell. “It
can help to understand disease mechanisms, find new
targets and validate drug candidates from screening at
an early stage.”
As the technology becomes automated and higher
throughput, it becomes a more feasible choice for use in
In the long term,
there could be a
potential application
for MSI in late-stage
drug development,
where MALDI-based
MS is currently
the gold standard
for characterizing
the distribution of
drug candidates in
development.
TECHNOLOGYNETWORKS.COM
CELL AND GENE THERAPIES: FROM DISCOVERY TO DELIVERY 20
drug screening. “Scientists in biopharma have adapted
IR-MALDESI to measure around 22 wells per second
for screens involving a simple enzymatic metabolic
readout12, and around 1-3 wells per second for more
complex high-content screens,” said Muddiman. “It’s
potentially turning the drug discovery pipeline on
its head – by starting with the experiment that really
matters – the phenotype of a drug candidate, whether it
hits its target and identifying any off-target effects.”
Only in the past few years has it become possible to
look at single-cell resolution using MSI and it’s still a
relatively specialized field. “As companies start to adopt
these latest advances that give single-cell resolution,
they will gain more useful information and find new
applications that will be commonly implemented into
their workflows,” said Stockwell.
“The cell is the fundamental unit of biology, it’s the
building block of organisms. If you can’t see the individual
cells, you’re always going to be somewhat in the dark.”
REFERENCES
1. Caleb Bagley M, Garrard KP, Muddiman DC. The development
and application of matrix assisted laser desorption electrospray
ionization: The teenage years. Mass Spectrom Rev. 2023;42(1):35-
66. doi: 10.1002/mas.21696
2. Michnowicz J. Mass spectrometry in drug discovery and
development. Nat Rev Drug Discov. 2002;1(8):651. doi: 10.1038/
nrd886
3. Dueñas ME, Peltier-Heap RE, Leveridge M, Annan RS, Büttner
FH, Trost M. Advances in high-throughput mass spectrometry in
drug discovery. EMBO Mol Med. 2023;15(1):e14850. doi: 10.15252/
emmm.202114850
4. Williams JD, Pu F, Sawicki JW, Elsen NL. Ultra-high-throughput
mass spectrometry in drug discovery: fundamentals and recent
advances. Expert Opin Drug Discov. 2024;19(3):291-301. doi:
10.1080/17460441.2023.2293153
5. Chu I, Nomeir AA. Utility of mass spectrometry for in-vitro
ADME assays. Curr Drug Metab. 2006;7(5):467-477. doi:
10.2174/138920006777697954
6. Ahire D, Kruger L, Sharma S, Mettu VS, Basit A, Prasad B.
Quantitative proteomics in translational absorption, distribution,
metabolism, and excretion and precision medicine. Pharmacol Rev.
2022;74(3):769-796. doi:10.1124/pharmrev.121.000449
7. Santiago BG, Eisennagel SH, Peckham GE, et al. Perspective on
high-throughput bioanalysis to support in vitro assays in early
drug discovery. Bioanalysis. 2023;15(3):177-191. doi: 10.4155/bio-
2022-0207
8. Robinson MR, Miller RA, Spellman DS. Mass spectrometrybased
biomarkers in drug development. Adv Exp Med Biol.
2019;1140:435-449. doi: 10.1007/978-3-030-15950-4_25
9. Deschamps E, Calabrese V, Schmitz I, Hubert-Roux M,
Castagnos D, Afonso C. Advances in ultra-high-resolution
mass spectrometry for pharmaceutical analysis. Molecules.
2023;28(5):2061. doi: 10.3390/molecules28052061
10. Zhang H, Cui W, Gross ML. Mass spectrometry for the biophysical
characterization of therapeutic monoclonal antibodies. FEBS Lett.
2014;588(2):308-317. doi: 10.1016/j.febslet.2013.11.027
11. Rajbhandari P, Neelakantan TV, Hosny N, Stockwell BR. Spatial
pharmacology using mass spectrometry imaging. Trends
Pharmacol Sci. 2024;45(1):67-80. doi: 10.1016/j.tips.2023.11.003
12. Radosevich AJ, Pu F, Chang-Yen D, et al. Ultra-high-throughput
ambient MS: Direct analysis at 22 samples per second by infrared
matrix-assisted laser desorption electrospray ionization mass
spectrometry. Anal Chem. 2022;94(12):4913-4918. doi: 10.1021/acs.
analchem.1c04605
MEET THE INTERVIEWEES
Brent R. Stockwell is chair of the department of biological sciences
and a professor at Columbia University in the departments of
biological science and chemistry. His research involves the
discovery of small molecules that can be used to understand and
treat cancer and neurodegeneration, with a focus on biochemical
mechanisms governing cell death.
Dave Muddiman is the Jacob and Betty Belin Distinguished
Professor of Chemistry at NC State University. His group
focuses on the development of innovative mass spectrometry
measurements to solve important biological problems.
Cell and Gene Therapy
Manufacturing
Getting Products to Patients
Source material acquisition
The quality of the initial material fundamentally
impacts the entire manufacturing process and final
product efficacy.
Samples must be handled with care, preserved and
tracked to ensure strict quality control.
Genetic modification/editing
Once the source material arrives at the manufacturing
facility, genetic modification begins.
Quality control at this stage involves
multiple checks to verify the accuracy of genetic
modifications and absence of off-target effects.
Cell expansion & culture
The cell expansion phase transforms small populations of
modified cells into therapeutic quantities.
This scaling process takes place in specialized bioreactors
that precisely control temperature, pH, oxygen levels and
nutrient concentrations.
Purification & quality control
Therapeutic cells must be separated from process‑related
impurities while maintaining cell viability and functionality.
This stage employs a combination of advanced separation
technologies, including magnetic-activated cell sorting
(MACS), fluorescence-activated cell sorting (FACS) and
tangential flow filtration.
Final product preparation
The final stage focuses on preparing the therapy for patient
administration, requiring careful consideration of product
stability and delivery requirements.
1
2
3
4
5
A multi-step manufacturing process
The CGT manufacturing process combines cutting-edge biotechnology with rigorous quality controls.
Each step presents unique challenges and opportunities for innovation.
Click here to view the full infographic
22 CELL AND GENE THERAPIES
Credit: iStock/luismmolina
Current Challenges in Cell Line
Development for Therapeutics
Kerry Taylor-Smith
Cell lines are a crucial tool in scientific research; without
them, many experiments would be impossible.
Cell lines are “a culture of mammalian cells that can
grow indefinitely in certain cultivation conditions in
a laboratory setting, and retain a distinct phenotype
and function, and can be stable over many population
doublings,” explains Dr. Paula Meleady, associate
professor in the School of Biotechnology at Dublin
City University.
Numerous cell lines have been derived over the past 70
years and are used extensively in biomedical research to
help scientists understand many fundamental biological
processes. “For example, a cancer cell line is derived
from cancer cells from a specific tissue (e.g., lung,
breast) and is used in research to study the biology of
cancer and to evaluate drugs used in cancer treatments,”
Meleady adds.
This article explores some of the challenges in
developing cell lines for therapeutics, such as stability of
the cell line, and how they are being addressed.
A vital part of the drug
development process
Cell lines are essential to drug development, allowing
researchers to test the efficacy of their therapeutics
in vitro before moving onto in vivo studies. Cell lines
are used to test drug metabolism and cytotoxicity,
study gene function and to generate a wide range of
biological medicines, from proteins such as antibodies,
recombinant protein products like insulin, to vaccines.1
Cell lines derive from a single original cell and are
genetically identical, so they “offer a more uniform
product, meaning that every molecule in a therapy is the
same; or as close as can be,” explains Dr. James Budge,
TECHNOLOGYNETWORKS.COM
CELL AND GENE THERAPIES: FROM DISCOVERY TO DELIVERY 23
a postdoctoral research fellow with a focus on Chinese
Hamster Ovary (CHO) cell lines at the University of
Kent. “The biology of each individual cell can have an
impact on the product’s biotherapeutic properties such
as the glycan profile, which refers to sugars added to the
molecule in the cell which work to prevent recognition
by a patient’s immune system and improve its stability
and half-life post administration.”
“Over the past 30 years, the CHO cell line has
been used for the production of biotherapeutics,
particularly monoclonal antibodies,” says Meleady.
“CHO cells are the most commonly used host cell
line for biotherapeutic production, with over 70% of
biopharmaceuticals produced from these cells.”
“CHO cells were initially used to study molecular
cell genetics; the reduced number of chromosomes
compared to human cells was an appealing property
for these studies,” adds Budge. These cells can be
cultured for long periods and showed robust genetic
stability, while also allowing for genetic manipulation.
“It is this robustness, ease of culture and the ability to
produce complex protein-based materials with humanlike
post-translational modifications which have seen
these cells become the dominant host for production of
biotherapeutic materials,” he adds.
Stability of cell lines
The production of high amounts of biotherapeutics
relies on the generation of stable cell lines. “Cell
line development begins with the transfection of a
suitable host cell line with the gene of interest, leading
to the random integration of DNA from the gene of
interest into the host genome,” explains Meleady. “The
engineered cell line will start to grow and produce
the product of interest, e.g., monoclonal antibody
biotherapeutic, usually under selection pressure.”
“Stability of cell lines presents a major challenge,”
says Budge. Cell lines need to display and maintain
functional features as similar as possible to primary
cells, but those which have undergone many passages
may harbor spontaneous mutations that affect their
phenotype, native functions and responsiveness to
stimuli, which could lead to unwanted variations in the
end product.1
“Phenotypic instability is a very common challenge
in biopharmaceutical production and is due to the
‘plasticity’ of CHO cells, which have a high propensity
for genomic rearrangements, such as deletions or
translocations,” explains Meleady. “This can be a
source of cell line instability during the bioproduction
process, resulting in the cells growing slower, a
reduction in viability and a fall-off in productivity of the
biotherapeutic of interest.”
There is a lot of research focusing on trying to find
solutions to this problem, Meleady adds. “This is
required to ensure stable, long-term production of
the biotherapeutic of interest, to ensure bioprocess
consistency and to assure acceptable product quality for
the regulatory authorities so that the product is of the
highest quality and safe for patient administration.”
Targeted integration techniques, including transponbased
technologies, which “enable the knock-in of
recombinant protein coding genes into well-defined and
transcriptionally active genomic sites is an active area of
research at the moment,” adds Meleady.
Genetic engineering may minimize the risk of
instability by knocking in or out specific genes.
The production of
high amounts of
biotherapeutics relies
on the generation of
stable cell lines.
TECHNOLOGYNETWORKS.COM
CELL AND GENE THERAPIES: FROM DISCOVERY TO DELIVERY 24
“Gene editing techniques could be used to modify the
genetic landscape of the host cell to ensure stable and
consistent expression of the desired genes, for example,
through the use of CRISPR-based genome editing
tools,” explains Meleady. “This technology offers new
opportunities to engineer the host cell line to produce
proteins with specific and desirable characteristics, such
as high cell densities, high viability, high productivity
and highest product quality.”
“For example, knocking out apoptosis-related genes
has the potential to increase cell survival and culture
longevity,” Meleady continues. “Knocking out host cell
proteins has the potential to eliminate the production
of proteins naturally produced by the host cell line that
could co-purify with the desired product. By knocking
out genes encoding these host cell proteins, the risk of
contamination can be reduced, simplifying downstream
processing and improving product purity.”
Single cell cloning and
monoclonality
“One challenge surrounding the development of cell
lines is cloning out individual cells and selecting the one
with the desired properties,” says Budge. “Scientists
need to ensure that the cell line is generating high titers
but also producing molecules with the best product
quality to meet its therapeutic potential.”
Prior to cloning, cells are incubated individually and
screened for the desired characteristics. Clones can
be isolated using limiting dilution cloning, which is
straightforward but time-consuming, with months spent
obtaining results.2 Automation can speed up clone
development and colony screening, reduces the chances
of contamination and can be a useful in ensuring
stability and product quality.
“Another challenge is that regulatory authorities require
substantial documentation of the methods used during
cell line development, to ensure clonality which is
crucial for product quality and safety,” says Meleady. “A
monoclonal cell line originates from one parent cell, to try
to eliminate the risk of genetic heterogeneity that could
affect the characteristics of the protein being produced.”
Growing cells can be subjected to genetic drifts,
mutations or loss of a plasmid. By testing and
documenting cells on day zero, scientists can prove the
cell line came from a single cell, thus minimizing risk and
ensuring a consistent production process and product
quality. Emerging techniques include fluorescenceactivated
cell sorting (FACS), the sorting of cells based
on fluorescent characteristics; and microfluidic dropbased
single-cell printers with the ability to image
newly-plated wells containing single cells. 3,4
Productivity
“The majority of significant improvements over
recent years has been the ability to achieve higher cell
densities in culture,” explains Budge. “More often than
not, more cells mean more product, and improvements
to culture media and feeding strategies have been
central to this. Advancements in perfusion culture
techniques and continuous culturing has also played a
key role more recently.”
Budge has had success in improving product titers by
manipulating the lipid content of CHO cell membranes;
“By overexpressing certain lipid modifying genes,
Cell lines are vital to
the drug development
process and without
them, we wouldn’t be
able to test the efficacy
of many important
medicines.
TECHNOLOGYNETWORKS.COM
CELL AND GENE THERAPIES: FROM DISCOVERY TO DELIVERY 25
we’ve been able to change cellular properties such as
membrane fluidity and/or endoplasmic reticulum size,
which improves the capacity of our CHO hosts to
produce biotherapeutic material.”
“More recently we have been working on transposon
‘jumping gene’ technology, which is now commonly
used in industry to produce cell pools,” says Budge.
“These technologies allow you to ‘cut and paste’ DNA
cargo into a host cell genome and ultimately can reduce
the time taken to generate cell pools (leading to quicker
cell line selection) and increase product yields by
increasing the frequency of integration events at desired
positions in the CHO genome. Furthermore, since the
cell pools produced using transposons have higher
titers, an increased percentage of ‘high expressers’ are
present in a pool. This can reduce the requirement for
intense selection processes.”
Quality control
Product quality is of key importance for the production
of biotherapeutics, as Meleady explains: “Correct
posttranslational modifications (PTMs), especially glycan
structures, are crucial for the potency and control of
pharmacokinetic and pharmacodynamic properties of
biotherapeutics. Heterogeneity of N-glycans can be an
issue during cell line development and bioproduction.
This heterogeneity, which can arise due to the variability
of N-glycan processing (and often linked to the ‘plasticity’
of CHO cells), may compromise the activity and safety of
biotherapeutics, particularly monoclonal antibodies.”
Meleady says control measures include the
establishment and confirmation of clonality to ensure
the stability of the host cell line. “There is also extensive
stability testing carried out during cell line development,
including monitoring the product for any changes to
sequence and PTM modifications (e.g., glycosylation),
i.e., using extensive quality control analytics.”
Artificial intelligence (AI) and machine learning will
also have an important role going forward in cell line
development, Meleady believes. AI tools can automate
cell culture experiments from start to finish, while AIpowered
software could generate 2D and 3D cellular
models and supervise automated feeding and media
changing, for example. This could help researchers
discover novel drug targets and investigate toxicity
more quickly.
Machine learning can make protocol development
more efficient, flexible and powerful; their algorithms
can prompt decision-making and allow for real time
customization. It also allows for fully automated
workflows to generate reproducible results across
multiple experiments, expediting milestones and
improving confidence in results.
Other challenges
Cell lines are cost effective, easy to grow and provide
an unlimited supply of material, but they don’t truly
represent how cells operate and interact within the body,
particularly when used in biomedical research and drug
development. The two-dimensional nature of cell culture
systems means they lack some aspects of life within an
organism, such as contact with tissue surfaces. Threedimensional
biology such as organoids or spheroids
grown in a scaffold would better represent the in vivo
environment and give more physiologically relevant
information, but these cultures can be inconsistent as
there is no standardized way to grow them.1,5
Potential contamination is also a concern; some are
obvious, like bacterial overgrowth, while others, such
as microbial mycoplasma, are less so. Mycoplasma
contamination can remain undetected in cell lines for
long periods of time, highlighting the importance of
scientists testing their cell lines frequently.1
“CHO and human embryonic kidney cells have been
the workhorses behind the production of complex,
multichain and multidomain molecules such as
monoclonal antibodies and the bioprocesses and
technologies surrounding them have been exquisitely
optimized to produce such molecules,” Budge explains.
“Newer molecules such as bispecific antibodies present
new challenges, and the tried and tested bioprocesses
require some redesign to tackle these.”
TECHNOLOGYNETWORKS.COM
CELL AND GENE THERAPIES: FROM DISCOVERY TO DELIVERY 26
These more complex therapeutic proteins can be used to
increase the frequency of correct pairings and therefore
the yield of the required protein but can be difficult to
express in CHO cells.
Cell lines are vital to the drug development process and
without them, we wouldn’t be able to test the efficacy
of many important medicines. But developing cell lines
isn’t without challenges. Over the years, much effort
has gone into improving and streamlining the process
to improve the stability, purity and quality of cell lines.
Technology is increasingly being used to automate
processes and improve decision making, saving time
and improving productivity – and it’s likely its use will
increase, particularly as new molecules are developed.
REFERENCES
1. Kaur G, Dufour JM. Cell lines. Spermatogenesis. 2012;2(1):1-5. doi:
10.4161/spmg.19885
2. Ye M, Wilhelm M, Gentschev I, Szalay A. a modified limiting
dilution method for monoclonal stable cell line selection using a
real-time fluorescence imaging system: a practical workflow and
advanced applications. Methods Protoc. 2021;4(1):16. doi:10.3390/
mps4010016
3. Yeh CF, Lin CH, Chang HC, Tang CY, Lai PT, Hsu CH. A microfluidic
single-cell cloning (SCC) device for the generation of monoclonal
cells. Cells. 2020;9(6):1482. doi:10.3390/cells9061482
4. Liao X, Makris M, Luo XM. Fluorescence-activated cell sorting for
purification of plasmacytoid dendritic cells from the mouse bone
marrow. J Vis Exp. 2016;(117):54641. doi: 10.3791/54641
5. Cacciamali A, Villa R, Dotti S. 3D cell cultures: Evolution of an
ancient tool for new applications. Front Physiol. 2022;13:836480.
doi: 10.3389/fphys.2022.836480
MEET THE INTERVIEWEES:
Dr. Paula Meleady is an associate professor in the School of
Biotechnology at Dublin City University. Her research focuses
on the use of proteomic and mass spectrometry technologies to
characterize recombinant mammalian cells to gain insights to
improving productivity and quality of biopharmaceuticals.
Dr. James Budge is a postdoctoral research fellow at the
University of Kent. His research focuses on engineering Chinese
Hamster Ovary (CHO) cell lines and the development of the
processes used to manipulate them for improved production of
biotherapeutic materials.
27 CELL AND GENE THERAPIES
Credit: iStock/Toshe_O
New Purification Techniques in
Biopharmaceuticals
Aron Gyorgypal, PhD
Biotherapeutics are produced using a multitude of
technologies and vary depending on the indication.
Monoclonal antibodies (mAbs) have been on the market
for decades and have successfully treated cancers and
autoimmune diseases. The use of viral vectors as gene
therapies has recently gained attraction through new
techniques, allowing the treatment of genetic disorders,
infectious diseases and cancers. The use of new vaccine
technology, such as messenger RNA (mRNA), has
allowed the production of potent immune responses
to mitigate infections, as seen with the COVID-19
pandemic.
Purification methodologies between different
biotherapeutics vary drastically and, depending on the
product, require a nuanced approach to enable or more
efficiently allow for the purification of the product
after upstream production. New technology and
workflows are looking to enable the more efficient or
cheaper purification of these products for downstream
processing.
Purification and processing of
mAbs
The purification and downstream processing of mAbs is
now relatively mature. "The platform technology is quite
developed for mAb manufacturing. After production
of the antibody by cell culture, the supernatant is
captured by protein A chromatography and held at low
pH for activation. Then follows a series of polishing
steps through cation exchange or ionic exchange before
the formulation of the final product," said Dr. Lukas
Gerstweiler, lecturer in bioprocess engineering at the
University of Adelaide, Australia. "I think the major
challenges of today are to integrate the processes to
TECHNOLOGYNETWORKS.COM
CELL AND GENE THERAPIES: FROM DISCOVERY TO DELIVERY 28
work continuously as well as maybe finding ways to
bring down the costs for production."
Continuous bioprocessing poses unique challenges
for antibody purification as a truly integrated line
between upstream and downstream processing is
still being investigated. "The past couple of years
have seen the use of rapid cycling technology, which
bolsters productivity," said Gerstweiler. Indeed, the
use of protein A membrane adsorbers, however, has
shown promise as an alternative to resin for process
intensification. These absorbers eliminate the need
for column packing, which lowers equipment cost,
decreases fouling, pressure drops, challenging and
stationary phase compression and has even been
shown to decrease host cell protein contaminants.1,2
However, it does come at the cost of increased buffer
volumes. However, this technology can help move mAb
processing from batch-based purification to semicontinuous
purification.
Finding methodologies to decrease purification costs
would also be helpful for less developed countries or
companies looking to decrease the costs associated
with downstream purification. One option is through
precipitation in a coiled flow inverter reactor, leading
to mAb capture by cation exchange multimodal
chromatography that then polishes the protein in an ion
exchange membrane.3,4
While mAb-based downstream processing has been
around for decades and the purification modalities have
matured, there is still a need for a better understanding
of the processes to enable continuous processing and
the nuances within the process to allow for better
manufacturing economics.
Viral vectors for gene therapy
Downstream purification of viral vectors poses tough
challenges due to technology limitations. While many
different practices exist for their purification, they suffer
from drawbacks such as poor scalability or inefficiencies
in early purification steps, which make them costprohibitive
due to yield losses.5
"The current state of the art for viral vector purification
starts with density gradient centrifugation. The
advantage of doing this is that not only can you
purify your viral vector from everything else in your
cell culture, but you can also separate empty and
full capsids," said Dr. Caryn Heldt, director of the
Health Research Institute, the James and Lorna Mack
Chair in Continuous Processing and professor in the
Department of Chemical Engineering at Michigan
Technological University. "But the issue with this is
that it is not feasible for commercial scale as it requires
scaling out, meaning that more ultracentrifuges
would be required to increase throughput. So, what
can be done? Well, another approach is through
chromatography, which works well on a lab scale but
on the commercial scale, besides costs, it can cause
issues as these chromatography columns produce large
pressure drops which can affect purification."
However, there are new advances in purification
technology that could alter the current purification
paradigm. "The vast majority of the newer techniques
are now in chromatography. A major focus is on new
ligand development and resin design," said Heldt.
Affinity chromatography has not yet been developed
for large-scale manufacturing. There currently exists
only a handful of targeted affinity resins that are specific
for only one type of virus serotype. To make the resins
economically feasible, they must be reusable and bind
to multiple viral serotypes. Some proprietary resins
are currently being investigated and found to bind to
multiple adeno-associated virus (AAV) serotypes.6
However, they still struggle with the copurification of
empty capsids along with the product.
A recent approach being investigated by multiple groups
is to functionalize peptides to chromatography columns
to allow for universal binding of virus serotypes, namely
"serotype-agnostic" resins.7 The production of these
protein ligands is based on mimicking the anti-AAV
antibody A20 by abstracting target peptide sequences
that target regions of the capsid, which are highly
conserved across stereotypes of various clades.8 These
peptide-ligands have comparable binding capacity with
commercial adsorbents but also reduce host cell protein
TECHNOLOGYNETWORKS.COM
CELL AND GENE THERAPIES: FROM DISCOVERY TO DELIVERY 29
contaminants and increase stability as they allow for
AAV elution at physiological pHs instead of acidic pHs.9
Where is the future of viral vector gene therapy going?
"The two big things I would like to see in the future are
continuous manufacturing and a better understanding of
the viral vector when it is loaded with gene versus when
it is not," said Heldt. Indeed, understanding the process
in which these capsids bind to chromatography columns
between full and empty columns would allow for more
rational engineering of materials that would bias against
the empty capsid products, whether this is done by
engineering the resin material or the capsid itself.
Purification technology for mRNA
vaccines
mRNA-based biotherapeutics are emerging as a
promising technology for vaccine development. On both
laboratory and manufacturing scale production, the
use of chromatographic separation has been selected
as a primary technique for purification based on
selectivity, adaptability and scalability.10 The shift from
more conventional protein-based therapeutic products
towards a nucleic acid-based platform brings other
challenges towards purification as the physiological
properties, structure and even impurity composition
vary drastically, requiring a selection of nuanced
chromatographic resins. This also accounts for large
inefficiencies that are present today in the downstream
processing of mRNA vaccines.11 Such inefficiencies are
due to limitations in the stationary phases used to purify
the therapeutic. Current methodologies rely on the
use of proprietary polymer resin materials and the use
of monolithic columns, typically based on size, charge,
hydrophilicity and affinity.12
As mRNA is negatively charged, anion exchange
chromatography is typically utilized to purify mRNA
from impurities. The selectivity on the column is
based on molecular weight and sequence and is prone
to produce aggregates, which can be mitigated with
denaturing agents or organic reagents – although this
is not favorable for large-scale manufacture due to
safety concerns. The approach to limit this concern
is through multimodal ion exchange/hydrogen
bonding chromatography. An approach by modifying
a monolithic column, can enable a high yield of
mRNA from in vitro transcription products, enabling
purification independent of construct size or the poly-A
tail of the vaccine.13
Affinity chromatography is another attractive
option for purification. The use of oligo-dT affinity
chromatography can capture the poly-A tail of
mRNA transition through A-T pairing. This can
effectively remove impurities such as the DNA
template, nucleotide substrates, enzymes and buffer
components. The immobilization of oligo-dT to an
electrospun polymer nanofiber adsorbent has been
shown to increase both the yield and allow for higher
flow rates to decrease processing time.14 The largest
criticism of oligo-dT-based chromatography is that it
cannot distinguish single- and double-stranded RNA.
Nevertheless, this technology has been used for the
purification of SARS-CoV-2 mRNA.15 In reality, the
processing of mRNA may require the use of both
chromatography and membrane filtration. Membrane
systems may be investigated further to enable
continuous processing in the future.16
The processing of mRNA-based therapeutics is still
inefficient, and a better understanding of the impurities
and separation modalities is required for more efficient
purification technology. This could be through size
exclusion, ion exchange, hydrophilic interaction, affinity
interactions or another modality.
REFERENCES:
1. Schmitz F, Minceva M, Kampmann M. Comparison of batch and
continuous multi-column capture of monoclonal antibodies
with convective diffusive membrane adsorbers. J Chromatogr A.
2024;1732:465201. doi:10.1016/j.chroma.2024.465201
2. Trnovec H, Doles T, Hribar G, Furlan N, Podgornik A.
Characterization of membrane adsorbers used for impurity
removal during the continuous purification of monoclonal
antibodies. J Chromatogr A. 2020;1609. doi:10.1016/j.
chroma.2019.460518
3. Kateja N, Kumar D, Sethi S, Rathore AS. Non-protein A purification
platform for continuous processing of monoclonal antibody
therapeutics. J Chromatogr A. 2018;1579:60-72. doi:10.1016/j.
chroma.2018.10.031
4. Arakawa T, Tomioka Y, Nakagawa M, et al. Non-Affinity Purification
of Antibodies. Antibodies. 2023;12(1). doi:10.3390/antib12010015
TECHNOLOGYNETWORKS.COM
CELL AND GENE THERAPIES: FROM DISCOVERY TO DELIVERY 30
5. Singh N, Heldt CL. Challenges in downstream purification of gene
therapy viral vectors. Curr Opin Chem Eng. 2022;35. doi:10.1016/j.
coche.2021.100780
6. Florea M, Nicolaou F, Pacouret S, et al. High-efficiency purification
of divergent AAV serotypes using AAVX affinity chromatography.
Mol Ther Methods Clin Dev. 2023;28:146-159. doi:10.1016/j.
omtm.2022.12.009
7. Shastry S, Barbieri E, Minzoni A, et al. Serotype-agnostic
affinity purification of adeno-associated virus (AAV) via peptidefunctionalized
chromatographic resins. J Chromatogr A.
2024;1734. doi:10.1016/j.chroma.2024.465320
8. Shastry S, Chu W, Barbieri E, et al. Rational design and
experimental evaluation of peptide ligands for the purification of
adeno-associated viruses via affinity chromatography. Biotechnol
J. 2024;19(1). doi:10.1002/biot.202300230
9. Mietzsch M, Smith JK, Yu JC, et al. Characterization of AAVSpecific
Affinity Ligands: Consequences for Vector Purification
and Development Strategies. Mol Ther Methods Clin Dev.
2020;19:362-373. doi:10.1016/j.omtm.2020.10.001
10. Keulen D, Geldhof G, Bussy O Le, Pabst M, Ottens M. Recent
advances to accelerate purification process development: A
review with a focus on vaccines. J Chromatogr A. 2022;1676.
doi:10.1016/j.chroma.2022.463195
11. Rosa SS, Prazeres DMF, Azevedo AM, Marques MPC. mRNA
vaccines manufacturing: Challenges and bottlenecks. Vaccine.
2021;39(16):2190-2200. doi:10.1016/j.vaccine.2021.03.038
12. Feng X, Su Z, Cheng Y, Ma G, Zhang S. Messenger RNA
chromatographic purification: advances and challenges. J
Chromatogr A. 2023;1707. doi:10.1016/j.chroma.2023.464321
13. Megušar P, Miklavčič R, Korenč M, et al. Scalable multimodal
weak anion exchange chromatographic purification for stable
mRNA drug substance. Electrophoresis. 2023;44(24):1978-1988.
doi:10.1002/elps.202300106
14. Dewar EA, Guterstam P, Holland D, et al. Improved mRNA affinity
chromatography binding capacity and throughput using an oligodT
immobilized electrospun polymer nanofiber adsorbent. J
Chromatogr A. 2024;1717. doi:10.1016/j.chroma.2024.464670
15. Corbett KS, Edwards DK, Leist SR, et al. SARS-CoV-2 mRNA
vaccine design enabled by prototype pathogen preparedness.
Nature. 2020;586(7830):567-571. doi:10.1038/s41586-020-2622-0
16. Javidanbardan A, Messerian KO, Zydney AL. Membrane
technology for the purification of RNA and DNA therapeutics.
Trends Biotechnol. 2024;42(6):714-727. doi:10.1016/j.
tibtech.2023.11.016
MEET THE INTERVIEWEES
Lukas Gerstweiler is a lecturer in Bioprocess Engineering at
the School of Chemical Engineering, University of Adelaide.
His research interests are within improving biopharmaceutical
processes using continuous manufacturing and model-based
process optimization.
Caryn Heldt is the Director of the Health Research Institute, the
James and Lorna Mack Chair in Continuous Processing, Professor
in the Department of Chemical Engineering, and an Affiliate
Professor in Biological Sciences at Michigan Technological
University. Her lab is focused on the purification, removal,
inactivation and stabilization of viruses and gene therapy vectors.
31 CELL AND GENE THERAPIES
Credit: iStock/ nopparit
A Bright Future for RNA
Therapeutics
RJ Mackenzie
The COVID-19 pandemic changed our society, and
the technology that powers it, in an instant. Suddenly,
remote working, telehealth and online learning boomed.
Other changes took longer to set in – governments
are only starting to appreciate the impact of a surge
in chronic health conditions. One rapid change was
in RNA biology – developing mRNA vaccines against
COVID-19 helped us beat the virus into retreat faster
than many could have hoped.
The advances in RNA biology since the pandemic could
have an enduring impact as well. They promise rapid,
personalized and flexible treatments against multiple
diseases. But these changes will only be realized if the
technology can overcome new hurdles. In this article,
we will explore whether RNA therapeutics can realize
their full potential. If they do, their impact against
COVID-19 will be only an overture.
RNA therapeutics: a promising
technology
RNA therapeutics exploit the nucleic acid’s position
as a middleman in the central dogma of biology.
RNA is required to turn genetic DNA instructions
into functional proteins. By introducing custom
RNA instructions, incorrect DNA blueprints can be
overwritten and new protein products can be generated.
Classical drug development techniques that produce
small-molecule therapeutics are time-consuming
processes. The molecules’ surface structure needs
to match their intended target receptors carefully.
Additionally, only a tiny fraction of the human genome
codes for small-molecule drug targets.1 What gives RNA
therapeutics an edge is that their base ingredients are
so well understood. “We know exactly how to build a
piece of DNA or RNA to encode whatever we want,”
says Brian Brown, director of the Icahn Genomics
TECHNOLOGYNETWORKS.COM
CELL AND GENE THERAPIES: FROM DISCOVERY TO DELIVERY 32
Institute at Mount Sinai. RNA can target non-coding
genes and transcripts, meaning the druggable space
for these compounds is vast. Extensive toolboxes
for building RNA means new therapeutics can be
designed rapidly.
These are powerful tools, but they once seemed a
scientific dead end. In early experiments, cells exposed
to external RNA constructs reacted with a powerful
and potentially lethal immune response.2 These issues
seemed insurmountable – to the extent that mRNA
researcher Katalin Karikó was academically blackballed
by the University of Pennsylvania for pursuing her
research in the area. Karikó would go on to find an
innovative solution to the problem. By swapping out a
nucleotide from mRNA’s structure for a modified analog
that produced no inflammation, she created a molecule
that didn’t set off immune alarms. Karikó would go on to
share the 2023 Nobel Prize for Physiology and Medicine
for her work. In addition to being a tale about the value
of sticking to your guns, Karikó’s discovery was also the
breakthrough that helped RNA biology enter a golden
age, says John Cooke, medical director of the Center for
RNA Therapeutics at Houston Methodist.
A silver lining to COVID-19
After this immune roadblock was overcome, the
technology blossomed. A variety of RNA therapeutics
have since been designed. These include RNA
interference (RNAi) molecules, which can block
transcription of target genes; antisense oligonucleotides,
which interfere with mRNA production; and RNA
aptamers, which bind to intra- and extracellular
targets with affinity and specificity matching that of
antibodies.3,4,5
Alongside mRNA therapeutics, these technologies
have been iteratively improved over the 20 years since
Karikó cracked the code of safe RNA therapy. But these
advancements went unheralded by the wider public
until the COVID-19 pandemic. When the time came
for the rapid development of a novel vaccine, RNA
therapeutics stepped up to the task.
Now, the technology is firmly in the public spotlight. This
higher profile has turbocharged progress in the field.
Of the 17 RNA therapeutics approved by the FDA, 9
have been given the green light in the last 5 years.6 It’s “a
silver lining to the COVID crisis,” says Cooke. The new
challenge is to maximize RNA therapeutics’ potential.
A safe delivery
Samir Mitragotri, a professor of bioengineering
at Harvard University, says that delivering RNA
therapeutics to their intended targets is now the field’s
biggest challenge. “These molecules are not super
stable,” he adds. “They are susceptible to degradation.
They are large compared to other therapeutics.”
These factors become even more important when you
consider the impact of a misdirected therapeutic. Smallmolecule
drugs, like aspirin, will leave the body rapidly,
so their effects on off-target tissues are also short-lived.
RNA therapeutics, by kickstarting or stopping gene
transcription, can have longer durations of action. “The
burden of making sure that it goes to the right place is
really high,” says Mitragotri.
But before they act on their targets, RNA therapeutics have
to reach them in the first place. That’s not straightforward.
The molecules are at the mercy of proteins called nucleases
in the bloodstream that easily break them down. A solution
for the problem came from nanoscience.
Lipid nanoparticle (LNP) technology isn’t new. The first
FDA-approved drug that utilized the technology – the
antifungal amphotericin B – is over 30 years old. But
it was only in 2018 that the RNAi-based compound
patisiran, the first such drug to be encapsulated in
LNPs, was approved to treat the neurodegenerative
condition transthyretin-related hereditary amyloidosis.7
LNP technology traps therapeutic RNA inside lipid
shells. This protects the RNA from nucleases, stabilizes
the molecule and helps it pass through lipophilic
membranes.8 The technology was used in Pfizer and
Moderna’s COVID-19 mRNA vaccines. “I think that the
unsung hero, at least of the [mRNA] vaccines, is really
this delivery system,” says Brown.
TECHNOLOGYNETWORKS.COM
CELL AND GENE THERAPIES: FROM DISCOVERY TO DELIVERY 33
Washed away
Other innovations have taken different routes toward
protecting RNA. One approach is to change the linear
structure of therapeutic RNA molecules, instead making
them circular, as the nucleases that chew up RNA bind
onto the molecule 5’ or 3’ end.9 “If you have a circular
RNA, there's no end. So the exonucleases can't gain
purchase and the RNA lasts longer,” says Cooke, whose
lab has been experimenting with this new structure.
Though technologies like LNPs have improved RNA
therapeutics’ durability, they can’t bypass other
challenges in drug delivery. Mitragotri says the challenge
facing RNA–LNP complexes in the bloodstream is
similar to that facing someone washed away down
a mighty river. The complex’s target is on the blood
vessel’s “bank”, but they lack an anchor that will get
them close enough to bind. Mitragotri highlights a
strategy in which LNPs “hitch a ride” on passing red
blood cells, which bump against blood vessel walls when
they diffuse oxygen into tissues.10 “We have designed
ways to allow the nanoparticles to attach to red blood
cells and circulate in the blood for a longer time and be
able to target the tissue,” he explains.
If these technologies improve RNA therapeutics’
stability and targeting, there could be rapid benefits to
the health system. Cooke outlines a new program at
Houston Methodist using mRNA vaccines to target
cancer. This approach can create new vaccines quickly,
and Cooke says that the team intend to develop,
generate and administer cancer vaccines within the
same hospital – something he thinks is a world first.
Cooke hopes that this approach could become
commonplace within a decade. “You'll have universitybased
programs where you can have these deployable
manufacturing units that can generate clinical-grade
RNA on a desktop. [...] These desktop apparatuses will
generate personalized RNA therapeutics,” he says.
Mitragotri is similarly optimistic. He points out that having
many different RNA tools – be it siRNA, mRNA, circular
constructs or hitchhiking molecules – will be essential
if the field is to tackle the grand challenges of human
disease. “We need a relatively wide basket of technologies
to support that variety of applications,” he concludes.
REFERENCES:
1. Falese JP, Donlic A, Hargrove AE. Targeting RNA with small
molecules: from fundamental principles towards the clinic. Chem
Soc Rev. 2021;50(4):2224-2243. doi: 10.1039/D0CS01261K
2. Kim YK. RNA therapy: rich history, various applications and
unlimited future prospects. Exp Mol Med. 2022;54(4):455-465. doi:
10.1038/s12276-022-00757-5
3. Traber GM, Yu AM. RNAi-Based Therapeutics and Novel
RNA Bioengineering Technologies. J Pharmacol Exp Ther.
2023;384(1):133-154. doi: 10.1124/jpet.122.001234
4. Rinaldi C, Wood MJA. Antisense oligonucleotides: the next
frontier for treatment of neurological disorders. Nat Rev Neurol.
2018;14(1):9-21. doi: 10.1038/nrneurol.2017.148
5. Keefe AD, Pai S, Ellington A. Aptamers as therapeutics. Nat Rev
Drug Discov. 2010;9(7):537-550. doi: 10.1038/nrd3141
6. Curreri A, Sankholkar D, Mitragotri S, Zhao Z. RNA therapeutics
in the clinic. Bioeng Transl Med. 2022;8(1):e10374. doi: 10.1002/
btm2.10374
7. Akinc A, Maier MA, Manoharan M, et al. The Onpattro story and
the clinical translation of nanomedicines containing nucleic acidbased
drugs. Nat Nanotechnol. 2019;14(12):1084-1087. doi: 10.1038/
s41565-019-0591-y
8. Jung HN, Lee SY, Lee S, Youn H, Im HJ. Lipid nanoparticles for
delivery of RNA therapeutics: Current status and the role of in
vivo imaging. Theranostics. 2022;12(17):7509-7531. doi: 10.7150/
thno.77259
9. Liu X, Zhang Y, Zhou S, Dain L, Mei L, Zhu G. Circular RNA: An
emerging frontier in RNA therapeutic targets, RNA therapeutics,
and mRNA vaccines. J Control Release. 2022;348:84-94. doi:
10.1016/j.jconrel.2022.05.043
10. Brenner J, Mitragotri S, Muzykantov V. Red Blood Cell Hitchhiking:
A Novel Approach for Vascular Delivery of Nanocarriers.
Annu Rev Biomed Eng. 2021;23:225-248. doi: 10.1146/annurevbioeng-
121219-024239
MEET THE INTERVIEWEES:
Dr. Brian Brown is a molecular biologist and immunologist. He is
director of the Icahn Genomics Institute (IGI).
Dr. Samir Mitragotri is a bioengineer and Hiller Professor of
Bioengineering and Hansjörg Wyss Professor of Biologically
Inspired Engineering at Harvard University.
Dr. John Cooke is director of the Center for Cardiovascular
Regeneration and medical director of the Center for RNA
Therapeutics at Houston Methodist.
34 CELL AND GENE THERAPIES
Credit: iStock/Rafa Jodar
Developments in
Point‑of‑Care Manufacture
of Advanced Therapies
Isabel Ely, PhD
Advanced therapy medicinal products (ATMPs)
represent a frontier in personalized medicine and
regenerative healthcare. This innovative medical
treatment utilizes gene therapy, somatic-cell therapy or
tissue engineering – sometimes designed based on each
patient's clinical or even genetic features – aiming for
long-lasting or permanent effects to treat disease.
Currently, ATMPs are manufactured at a few centralized
sites, meaning the starting materials and/or final
products are generally frozen for transportation – often
resulting in reduced therapy potency. Therefore, the
therapy must be delivered to the patient some minutes
or seconds after manufacture.
Point-of-care (POC) manufacturing aims to overcome
this limitation by producing ATMPs in hospitals. This
is especially important when it needs to be delivered to
the patient without delay and there is no time for storing
the medicine. POC manufacturing enables hospitals to
be in contact with cutting-edge products, refining the
therapeutic skills that they hold and increasing the range
of therapies made available to patients.
POC ATMPs remain at the cutting edge of medical
innovation, with ongoing advancements enhancing the
ATMPs and streamlining the administration processes
involved in POC manufacturing. In this listicle, we
explore some of the latest trends and innovations
shaping the field of POC manufacturing for ATMPs.
TECHNOLOGYNETWORKS.COM
CELL AND GENE THERAPIES: FROM DISCOVERY TO DELIVERY 35
POC manufacturing readiness
and decentralization of
manufacturing systems
In 1974, NASA proposed the idea of technology readiness
levels to assess the maturity of certain technologies.1
Adopted by the US Department of Defense, manufacturing
readiness levels were then introduced, with the most
mature stage (i.e., highest readiness level) detailing all
materials, equipment, facilities and personnel are in place
and have met full-rate production requirements.2
Manufacturing readiness levels
The manufacturing readiness level serves as a metric
to evaluate the maturity of manufacturing processes,
analogous to technology readiness levels used to gauge
technological development. Manufacturing readiness
levels provide quantitative measures to assess the
readiness of a technology, component or system
specifically from a manufacturing standpoint.
Now, manufacturing readiness is being applied to a range
of technology fields – with recent applications in the
biomedical field and POC manufacturing of ATMPs.3
Over the past few decades, UK hospitals have
produced ATMPs in limited quantities. In the future,
this production is expected to become more seamlessly
integrated into routine clinical practices, with the
technical and regulatory expertise hospitals have
gained forming the basis for scaling up and increasing
production frequency in the coming years – known as
POC manufacturing readiness.
Achieving POC manufacturing readiness is necessary to
produce POC ATMPs, in which several resources, skills
and institutional processes must be present for their
success. The three main aspects of POC manufacturing
readiness include staff and institutional procedures,
infrastructure and transportation.3
Staff and institutional procedures
The adverse effects of ATMPs can be severe, meaning
their quality must be strictly monitored and individuals
who conduct ATMP POC manufacturing need to be
highly skilled. Often, this central role is played by a
“qualified person” (QP) – a professional indicated on the
manufacturer’s license as being legally responsible for
ensuring that the product has been manufactured in line
with quality and efficacy parameters.
It is generally believed there is an insufficient number
of QPs in the UK, especially in hospitals, with only
21 hospital-based QPs being registered since 2011.3
Hospitals have, therefore, attempted to advance
other staffing aspects to improve manufacturing
readiness, such as the creation and refinement of
ATMP committees.3 These committees play a liaising
role in ATMP manufacturing processes due to the
various departments and facilities involved (i.e., testing
laboratories, pharmacies and clinical wards). When
ATMP POC manufacture becomes more frequent, these
ATMP committees will play a decisive role, either by
having their responsibilities expanded or by serving
as a model for the creation of even more specialized
committees in hospitals.
Infrastructure
Manufacturing cell and gene therapies undoubtedly
incur high costs for infrastructure changes, which could
ATMPs ideally need to
be produced in a closed
system where starting
materials and reagents
are processed within
machines, with little
human manipulation.
TECHNOLOGYNETWORKS.COM
CELL AND GENE THERAPIES: FROM DISCOVERY TO DELIVERY 36
be even more apparent for hospitals, as they were
not originally designed to host ATMP manufacturing
activities. Therefore, today’s hospital staff need to find
ways of accommodating POC manufacturing activities
in a clinical setting.
Hospital laboratories will also play a key role in ATMP
POC manufacture, as they may need to be mobilized
for performing tests on tissues and cells as part of the
quality control associated with therapy manufacture.
There is also the obvious need for equipment. ATMPs
ideally need to be produced in a closed system where
starting materials and reagents are processed within
machines, with little human manipulation, so the risk of
contamination and human errors is reduced.4 Further,
it is sometimes possible to freeze starting materials and
final products, which requires equipment for accurate
temperature control so that living structures are
not damaged.5
Transportation
The production of ATMPs in hospitals entails procuring
a quite long list of products, be it for collection of starting
materials, quality control or manufacture itself – meaning
hospitals need to engage in a series of external relations.
Starting materials and final products cannot be
transported easily due to the presence of fragile living
structures such as cells, meaning factors such as
temperature and vibration need to be controlled if it is
necessary to transport cells and tissues.
Currently, hospitals procuring or shipping biological
materials use commercial courier services. Evolving
airline service strategies and security policies have
complicated the model of product transport, so other
methods, such as overnight shipping services, have been
explored to assess their feasibility.6
These courier companies have designed specialized
package tracking and monitoring programs developed
specifically for healthcare and scientific businesses,
which hospitals have also used. Further, hospitals
have tried to ensure quality in their relations with
companies transporting materials and samples by using
ISO certifications.
Automation and digital integration
Currently, manufacturing processes for ATMPs are
largely manual and performed in planar culture systems
– processes that are highly laborious and difficult to
scale up. Resultingly, such processes are prone to
human error and can result in batch-to-batch variability,
high manufacturing costs, high risk of contamination
and batch loss.7
Manufacturing products following good manufacturing
practice (GMP) guidelines will minimize process
variability and variation in factors such as cell quality.
However, full GMP compliance in the ATMP realm is
currently challenging – particularly due to the increased
difficulty in sourcing compliant starting material.
Automation has been increasingly integrated into
biopharmaceutical industries, particularly cell and gene
therapies.8 Automation reduces human error, increases
scalability and reduces production time. Digital tools
such as real-time monitoring and artificial intelligence
for process optimization are also increasingly
being adopted.
POC manufacturing can experience in-process
variation from human handling which can subsequently
impact product quality. Even when following
stringent protocols, variation is observed between
different handlers resulting from minor procedure
imprecisions, such as variation in pipetting technique
Automation has been
increasingly integrated
into biopharmaceutical
industries, particularly
cell and gene therapies.
TECHNOLOGYNETWORKS.COM
CELL AND GENE THERAPIES: FROM DISCOVERY TO DELIVERY 37
or slight deviations in incubation times. Automation
can eliminate such in-process variation in multiple
ways, such as through robotic arms to repeatedly and
consistently perform pipetting or a mixing action. Even
cell culture sub-processes (e.g., medium changes) can
be automated, allowing for consistent speed, force
and accuracy – thus leading to reduced variability and
increased process reliability.
Integration and automation of process analytics can
remove the subjectivity of processing decisions, making
more sophisticated processing rules possible. An example
of this is the development of pattern recognition and
image processing software that can be used to objectively
determine confluence.8 Cell culture protocols rely on
passaging adherent cells when they reach a confluence
level of 80-90%. Confluency is typically estimated
through microscopic visualization with the percentage
estimation entirely subjective and dependent on the
individual. Employing automated image acquisition and
processing can allow for adaptive processing based on
objective and comprehensible criteria.9
Automation integration in POC manufacturing
of ATMPs faces several challenges including the
development of technologically advanced interfaces,
compliance with good manufacturing practices, the need
to extensively validate automated systems and how to
limit the effects on ATMP production if there is ever
system downtime. Addressing these challenges requires
a combination of advanced engineering, robust software
solutions, regulatory foresight and interdisciplinary
collaboration to realize the full potential of automation
in POC ATMP manufacturing.
Advancing ATMPs techniques:
innovations in hydrogels
Innovations of new techniques and products continue
to emerge in ATMPs. Hydrogels – which are highly
hydrated three-dimensional polymeric matrices – are
one of these recent innovations that hold substantial
promise in biomedical fields due to their biocompatible,
chemically modifiable and physically tunable nature.10
For example, hydrogels have demonstrated the potential
to support cell viability and functionalities and facilitate
targeted delivery and controlled release of therapeutic
agents.11,12,13
Synthesis and modification technologies used today
have matured enough to advance hydrogel material
significantly, moving away from the simplistic structures
exhibited by early-generation hydrogels. Now, hydrogels
have been developed to react to specific biological and
pathological stimuli such as pH, temperature and reactive
oxygen species, allowing the intricate requirements of
specific diseases and heightened clinical demands to be
met.14,15,16 Furthermore, the stable structure hydrogels
experience upon water absorption acts as a delivery
platform for bioactive substances and pharmaceuticals.
Hydrogels can also mimic the extracellular matrix
environment, promoting the growth and survival of
encapsulated cells.17 The development of biodegradable
variants of hydrogels, either allowing degradation over
time or under specific stimuli, enables the release of
their contents without eliciting toxic side effects to
surrounding tissues.18 Injectable hydrogels further
ensure sustained and controlled drug release at
the targeted site, significantly reducing the adverse
reactions associated with systemic drug exposure.19
Although significant advancements have been made
in the use of hydrogels, which contribute to enhancing
POC ATMPs, challenges remain.
Many studies focused on hydrogel-based cell therapies
remain in the nascent stages, with research progression
hindered by technical complexities, biological intricacies,
safety concerns, funding limitations and challenges
of interdisciplinary collaboration.20 Importantly,
extrapolation of results from in vitro and animal models
to humans is complicated by biological variability,
necessitating a comprehensive safety assessment of
hydrogels to prevent adverse effects on patients.
Injectable hydrogels are viewed as the most promising
candidates for clinical translation. However, challenges
in understanding biological degradation mechanisms and
injection timing require further clarification. The misinjection
of hydrogels into critical areas, such as major
blood vessels therefore increasing the risk of embolism,
TECHNOLOGYNETWORKS.COM
CELL AND GENE THERAPIES: FROM DISCOVERY TO DELIVERY 38
warrants the need for protocols to mitigate such risks
and highlights the complexity of clinical complexity.
REFERENCES
1. Sadin SR, Povinelli FP, Rosen R. The NASA technology push
towards future space mission systems. Acta Astronautica.
1989;20:73-77. doi: 10.1016/0094-5765(89)90054-4
2. Manufacturing Readiness Level (MRL) Deskbook. 2018. Available
at: https://www.dodmrl.com/MRL_Deskbook_2018.pdf. Accessed
February 10, 2025.
3. Bicudo E, Brass I. Institutional and infrastructure challenges for
hospitals producing advanced therapies in the UK: The concept of
‘point-of-care manufacturing readiness.’ Regenerative Medicine.
2022. doi: 10.2217/rme-2022-0064
4. Gannon PO, Harari A, Auger A, et al. Development of an optimized
closed and semi-automatic protocol for Good Manufacturing
Practice manufacturing of tumor-infiltrating lymphocytes in
a hospital environment. Cytotherapy. 2020;22(12):780-791. doi:
10.1016/j.jcyt.2020.07.011
5. Petrenko Y, Chudickova M, Vackova I, et al. Clinically Relevant
Solution for the Hypothermic Storage and Transportation of
Human Multipotent Mesenchymal Stromal Cells. Stem Cells
International. 2019;2019(1):5909524. doi: 10.1155/2019/5909524
6. Howard A, Chitphakdithai P, Waller EK, et al. Evaluation of
peripheral blood stem cell quality in products transported by
traditional courier or commercial overnight shipping services.
Transfusion. 2014;54(6):1501-1507. doi: 10.1111/trf.12533
7. Heathman TRJ, Rafiq QA, Chan AKC, et al. Characterization of
human mesenchymal stem cells from multiple donors and the
implications for large scale bioprocess development. Biochemical
Engineering Journal. 2016;108:14-23. doi: 10.1016/j.bej.2015.06.018
8. Moutsatsou P, Ochs J, Schmitt RH, Hewitt CJ, Hanga MP.
Automation in cell and gene therapy manufacturing: from past to
future. Biotechnol Lett. 2019;41(11):1245-1253. doi: 10.1007/s10529-
019-02732-z
9. Schenk FW, Kulik M, Schmitt R. Metrology-based quality
and process control in automated stem cell production. tm -
Technisches Messen. 2015;82(6):309-316. doi: 10.1515/teme-2015-
0036
10. Lu P, Ruan D, Huang M, et al. Harnessing the potential of hydrogels
for advanced therapeutic applications: current achievements and
future directions. Sig Transduct Target Ther. 2024;9(1):1-66. doi:
10.1038/s41392-024-01852-x
11. Zhang R, Zhang D, Sun X, Song X, Yan KC, Liang H. Polyvinyl
alcohol/gelatin hydrogels regulate cell adhesion and chromatin
accessibility. International Journal of Biological Macromolecules.
2022;219:672-684. doi: 10.1016/j.ijbiomac.2022.08.025
12. Zhou J, Du X, Xu B. Regulating the rate of molecular self-assembly
for targeting cancer cells. Angewandte Chemie International Edition.
2016;55(19):5770-5775. doi:10.1002/anie.201600753
13. Shi W, Ji Y, Zhang X, Shu S, Wu Z. Characterization of pH- and
thermosensitive hydrogel as a vehicle for controlled protein
delivery. JPharmSci. 2011;100(3):886-895. doi: 10.1002/jps.22328
14. Zhou J, Wang H, Chen H, et al. pH-responsive nanocomposite
hydrogel for simultaneous prevention of postoperative adhesion
and tumor recurrence. Acta Biomaterialia. 2023;158:228-238. doi:
10.1016/j.actbio.2022.12.025
15. Zhang JT, Huang SW, Zhuo RX. Temperature-sensitive
polyamidoamine dendrimer/poly(N-isopropylacrylamide)
hydrogels with improved responsive properties. Macromolecular
Bioscience. 2004;4(6):575-578. doi: 10.1002/mabi.200400003
16. Chen SX, Zhang J, Xue F, et al. In situ forming oxygen/ROSresponsive
niche-like hydrogel enabling gelation-triggered
chemotherapy and inhibition of metastasis. Bioactive Materials.
2023;21:86-96. doi: 10.1016/j.bioactmat.2022.08.002
17. Zimmermann U, Mimietz S, Zimmermann H, et al. Hydrogelbased
non-autologous cell and tissue therapy. BioTechniques.
2000;29(3):564-581. doi: 10.2144/00293rv01
18. Gholamali I, Yadollahi M. Doxorubicin-loaded carboxymethyl
cellulose/Starch/ZnO nanocomposite hydrogel beads as
an anticancer drug carrier agent. International Journal of
Biological Macromolecules. 2020;160:724-735. doi: 10.1016/j.
ijbiomac.2020.05.232
19. Jin H, Wan C, Zou Z, et al. Tumor ablation and therapeutic
immunity induction by an injectable peptide hydrogel. ACS Nano.
2018;12(4):3295-3310. doi: 10.1021/acsnano.7b08148
20. Ansar R, Saqib S, Mukhtar A, et al. Challenges and recent
trends with the development of hydrogel fiber for biomedical
applications. Chemosphere. 2022;287:131956. doi: 10.1016/j.
chemosphere.2021.131956
CELL AND GENE THERAPIES: FROM DISCOVERY TO DELIVERY 39
TECHNOLOGYNETWORKS.COM
CONTRIBUTORS
Aron Gyorgypal, PhD
Aron is a postdoctoral research fellow at Harvard Medical
School and Massachusetts General Hospital in Boston,
Massachusetts. Following the completion of his PhD,
he joined the Anthony Lab at Harvard Medical School
and Massachusetts General Hospital in the immunology
department.
Isabel Ely, PhD
Isabel is a Science Writer and Editor at Technology
Networks. She holds a BSc in exercise and sport science
from the University of Exeter, a MRes in medicine and
health and a PhD in medicine from the University of
Nottingham.
Joanna Owens, PhD
Joanna Owens holds a PhD in molecular toxicology
from the University of Surrey. She has over 20 years’
experience writing about a wide range of scientific topics
in biosciences, pharmaceuticals and biotechnology.
Kate Harrison, PhD
Kate Harrison is a senior science writer and is responsible
for the creation of custom-written projects. She holds a
PhD in virology from the University of Edinburgh.
Kerry Taylor-Smith
Kerry Taylor-Smith is a science writer who covers
everything from climate change and the environment, to
health and wellness, to material science, astronomy and
space.
Monica Hoyos Flight, PhD
Monica is a freelance science communications consultant
providing writing and editing services for researchers,
research organizations and companies. She has a PhD in
neuroscience from Imperial College London.
RJ Mackenzie
RJ is a freelance science writer based in Glasgow. He
covers biological and biomedical science, with a focus on
the complexities and curiosities of the brain and emerging
AI technologies.
Sponsored by

Download the eBook for FREE Now!
Information you provide will be shared with the sponsors for this content. Technology Networks or its sponsors may contact you to offer you content or products based on your interest in this topic. You may opt-out at any time.
Experiencing issues viewing the form? Click here to access an alternate version