Cell and Gene Therapies: Innovations, Challenges and Clinical Breakthroughs
eBook
Published: December 17, 2024
Credit: Technology Networks
Cell and gene therapies are reshaping modern medicine, offering targeted solutions to complex diseases. These innovative approaches leverage biological mechanisms, delivering personalized treatments that surpass the limitations of traditional methods. However, challenges like scalability, regulatory compliance and manufacturing complexities often hinder broader adoption. Bridging these gaps is crucial to unlocking the full potential of these transformative therapies.
This eBook highlights groundbreaking innovations in cell- and gene-based treatments, delves into strategies to overcome barriers associated with their use and explores real-world clinical breakthroughs.
Download this eBook to discover:
- Cutting-edge developments in CRISPR, regenerative medicine and immune cell therapies
- Strategies to navigate manufacturing and regulatory hurdles for scalable solutions
- Insights into personalized medicine and its impact on conditions like cancer and inherited disorders
CELL AND GENE
THERAPIES:
Innovations, Challenges and Clinical Breakthroughs
Credit: iStock
SPONSORED BY
CHALLENGES AND
SOLUTIONS IN
CELL THERAPY
DEVELOPMENT
ADVANCES IN
BIOPHARMACEUTICAL
ANALYSIS
GENE THERAPY: A NEW
FRONTIER IN DISEASEMODIFYING THERAPIES
CONTENTS
04
Cell Therapy: Clinical Applications,
Manufacturing and Regulation
08
Microbial Monitoring for Biopharma
Manufacturing
12
Challenges and Solutions in Cell
Therapy Development
16
Removing the Glycocalyx Armor of
Cancer Cells for Treatment
20
Electronic Pulses Could Reduce the
Need for High Doses in Gene Therapy
Delivery
23
Gene Therapy: A New Frontier in
Disease-Modifying Therapies
27
Advances in Biopharmaceutical
Analysis
31
How To Run a Successful
CRISPR Experiment
35
Breaking the Chains: How CRISPR
Gene Therapy Gave Victoria Gray a
New Life
CELL AND GENE THERAPIES: INNOVATIONS, CHALLENGES AND CLINICAL BREAKTHROUGHS 3
TECHNOLOGYNETWORKS.COM
FOREWORD
The rapid advancement of cell and gene therapies is transforming the landscape of modern
medicine. These groundbreaking treatments harness the power of biological mechanisms,
offering innovative solutions for some of the most challenging diseases.
Covering a broad spectrum of topics, this eBook examines cutting-edge approaches like
CRISPR-driven gene editing, stem cell-based regenerative medicine and advancements in
cellular immunotherapies. It highlights personalized medicine’s growing role, demonstrating
how tailored treatments are revolutionizing care for conditions like cancer and inherited
disorders. Readers will also explore breakthroughs in manufacturing and regulatory
processes that are improving accessibility and scalability.
Whether you’re a researcher, healthcare professional or simply curious about the future of
medicine, this eBook offers an insightful exploration into the world of cell-based and genebased interventions.
The Technology Networks editorial team.
Cell therapy is a therapeutic strategy that involves the
transfer of autologous (patient-derived) or allogeneic
(donor-derived) cells into a patient’s body. Cell therapies can
be divided into two broad categories — stem-cell therapies
and non-stem-cell therapies. Within these two groups, a
variety of mechanisms of action are employed, tailored to the
specific type of cell therapy and the disease or condition it’s
designed to target.
For example, in regenerative medicine, stem cell therapies
work by replacing damaged or diseased cells with new,
functional ones to restore organ and/or tissue function.
Whereas adoptive cell therapies for cancer treatment exploit
immune cells by either expanding the number of cells, or by
genetically modifying them to boost their cancer-fighting
abilities, before being administered to the patient. The three
main cell therapy modalities are outlined in Figure 1.
In 2023, the global market value for cell therapy was
estimated to be USD 4.74 billion. This market is expected
to experience rapid growth due to increasing demand
for innovative cell therapies, which tend to have fewer
adverse effects compared to traditional modalities. This,
coupled with their wide range of clinical applications, is
anticipated to propel the market to approximately USD
20 billion by 2030.
In this listicle, we explore new therapeutic targets and
clinical applications, and discuss challenges and key
considerations related to the manufacture and regulation
of cell therapies.
Clinical applications and novel
research
While the oncology segment led the overall cell therapy
market in 2023, the potential of cell therapy in other
areas is increasingly evident. Here we highlight recent
research that illustrates the diverse potential of cell
therapy across different therapeutic areas.
WOMEN IN SCIENCE
Cell Therapy Targets, Clinical
Applications, Manufacturing
and Regulatory Considerations
Laura Elizabeth Lansdowne
4
Credit: iStock
TECHNOLOGYNETWORKS.COM
Neurology
UC San Diego Health became one of the first facilities
in the United States to administer the experimental
neural cell therapy, NRTX-1001, to subjects with
drug-resistant epilepsy. This cell therapy involves the
delivery of interneurons that secrete the inhibitory
neurotransmitter gamma-aminobutyric acid (GABA),
into the epileptic region of the brain. In December
2023, the biotherapeutics company developing NRTX1001 announced that the initial two trial subjects
consistently reported a decrease in seizure frequency (>
95% reduction from baseline), more than one year after
receiving treatment.
Regenerative medicine
Using mRNA technology encapsulated in nanoparticles,
researchers designed a novel stem cell therapy that
aims to stimulate the liver's natural repair processes.
The study was published in Cell Stem Cell. The therapy
works by delivering vascular endothelial growth factor A
(VEGFA) mRNA via lipid nanoparticles. This promotes
the conversion of biliary epithelial cells into hepatocytes,
the liver's functional cells. The research was conducted
using mouse and zebrafish disease models.
Immunology
Researchers developed a novel T-cell therapy to treat
a specific form of autoimmune encephalitis (NMDAR
encephalitis), an immune-mediated condition
whereby antibodies attack healthy brain cells, causing
inflammation and various neurological symptoms. The
team genetically modified T cells to selectively eliminate
anti-NMDAR B cells and autoantibodies against the
NMDA receptor. This preclinical study was published in
Cell. They now plan to test the therapy in human subjects
with NMDAR encephalitis.
Inherited blood disorders
The US Food and Drug Administration (FDA) approved
two cell-based gene therapies — Casgevy™ (exagamglogene
autotemcel) and Lyfgenia™ (lovotibeglogene autotemcel)
— to treat sickle cell disease in patients ≥ 12 years. Both
therapies are created using patients’ hematopoietic stem
cells, which are genetically modified and then reintroduced
as a single-dose infusion. Casgevy is the first FDAapproved therapy that exploits CRISPR-Cas9 gene
editing. CRISPR-Cas9 is used to reduce the expression
of BCL11A, which in turn, boosts the synthesis of
γ-globin and reactivates fetal hemoglobin production,
CELL AND GENE THERAPIES: INNOVATIONS, CHALLENGES AND CLINICAL BREAKTHROUGHS 5
Cell Therapies
Cells are administered to a patient as a
therapeutic modality.
Genetically modified cell therapies
Patient/donor cells are genetically modified
to perform a unique function they wouldn't
typically do that provides therapeutic
benefit to a patient.
Tissue-engineered products
Cells and/or biologically active substances
are designed to restore, maintain or replace
damaged tissues/organs.
Figure 1: Overview of cell therapy modalities. Credit: Technology Networks.
CELL AND GENE THERAPIES: INNOVATIONS, CHALLENGES AND CLINICAL BREAKTHROUGHS 6
TECHNOLOGYNETWORKS.COM
preventing sickling. Lyfgenia uses a lentiviral vector to
genetically modify the stem cells to produce a specific
hemoglobin called HbAT87Q. Cells containing this
hemoglobin have a reduced risk of sickling.
Oncology
A preclinical study published in Nature Communications
describes a novel allogeneic CAR T-cell therapy targeting
T-cell malignancies. This therapy focuses on eliminating
cancerous T cells that express a dominant T-cell receptor
called Vβ2. Unlike traditional treatments that risk
depleting all of a patient's T cells, this CAR T-cell therapy
selectively kills the diseased cells, preserving Vβ2-negative
healthy cells. CRISPR gene-editing technology was used
to specifically target the Vβ2-positive cancer cells.
The FDA approved Amtagvi™ (lifileucel), a tumor-derived
autologous T-cell immunotherapy, via its Accelerated
Approval Program. Amtagvi is indicated for use in adult
patients with a melanoma (unresectable or metastatic),
that has failed to respond to/stopped responding to
other specific therapies. A portion of the patient’s tumor
is surgically removed. Tumor-derived T cells are then
isolated from the excised tumor tissue, expanded at a
manufacturing site and then administered as a single-dose
intravenous infusion to the same patient.
Understanding the regulatory
landscape of cell therapy
The pharmaceutical industry is one of the most regulated
industries globally, and regulatory authorities play a crucial
role in overseeing various steps related to the development
of new therapies. Several different regulatory authorities
exist worldwide and each issues specific guidelines relating
to the development, registration, manufacturing, licensing,
marketing and labeling of medicines. As such, it is vital cell
therapy developers understand regulatory variations and
consider how these differences will impact the development
of a novel cell therapy, depending on where in the world they
are seeking marketing authorization. Here we take a closer
look at regulatory guidance in the UK, Europe and the US.
The European Medicines Agency (EMA) classes cell
therapies as a type of advanced therapy medicinal product
(ATMP). As such, they are governed by medicinal product
regulatory frameworks (Regulation (EC) No 1394/2007,
Directive 2001/83/EC). The manufacturing of these
products must comply with Good Manufacturing Practice
(GMP) principles (EudraLex Volume 4, Part IV).
Similar to the EMA, the UK’s Medicines and Healthcare
products Regulatory Agency (MHRA) classes cell therapies
as a type of ATMP. The donation, procurement and testing
of cells is covered by the EU Tissues and Cells Directive
(2004/23/EC). Under this directive there are two authorities
of note — the Human Fertilisation and Embryology
Authority (HFEA) and the Human Tissue Authority (HTA).
The HFEA oversees the use of gametes and embryos in the
development of ATMPs and the HTA is responsible for the
licensing and inspection for all other tissue and cell types.
In the US, cell therapies are regulated by the FDA’s Center
for Biologics Evaluation and Research (CBER). They fall
under Title 21 of the Code of Federal Regulations (CFR),
Part 127.3(d), and are defined as, “Articles containing or
consisting of human cells or tissues that are intended for
implantation, transplantation, infusion or transfer into a
human recipient".
The FDA recommends that “product testing for cellular
therapies include, but not be limited to, microbiological
testing (including sterility, mycoplasma and adventitious
viral agent testing) to ensure safety and assessments of other
product characteristics such as identity, purity (including
endotoxin), viability and potency.” The FDA has numerous
cellular and gene therapy guidance documents, addressing
specific aspects of cell and gene therapy development – for
example, potency assurance, manufacturing, trial design
and general regulatory considerations.
Addressing manufacturing and
scalability challenges
While the demand for cell therapies is undeniable, their
production comes with key challenges and regulators
require manufacturers to conduct extensive testing. Cell
therapy manufacturing methods range in complexity.
Some therapies require significant manipulation of cells
(e.g., genetic modification), while others may require
comprehensive cell cultivation steps. Here we discuss
several manufacturing and scalability considerations.
TECHNOLOGYNETWORKS.COM
Sourcing high-quality cells
The initial challenge in cell therapy production lies in
sourcing high-quality biological materials. The exact
geographical region and regulatory authority will
influence how starting material must be obtained, for
example collection/apheresis best practices.
Progress is being made to refine the cell extraction
and separation processes. For example, researchers
recently developed a technology capable of extracting
mesenchymal stem cells (MSCs) directly from bone
marrow – without the need for dilution. By continuously
sorting and isolating stem cells from blood cells using a
novel microfluidic platform, it was possible to double the
number of MSCs obtained from bone marrow samples and
reduce the extraction time to approximately 20 minutes.
In the case of allogenic (donor-derived) cell therapies,
specific donor eligibility requirements will also need to
be considered as these may differ depending on country.
Establishing an appropriate shelf life
Once obtained, the shelf life of fresh cells is often
short. For example, hemopoietic stem cells can be
stored unprocessed at 4 °C or room temperature for
approximately 72 hours post-collection. However,
after this time they begin to degrade, resulting in
compromised product quality and potency. To address
this, developers typically opt to cryogenically freeze
cells, increasing their shelf life to months or years,
but this relies on specialist facilities and procedures
to ensure the cells remain viable and stable. The
optimal cryopreservation and freeze—thawing process
will differ depending on the particular cell product.
Regulators require manufacturers to conduct stability
testing to confirm the product's integrity and efficacy
over time.
Addressing temperature sensitivity
Increased temperature sensitivity is another key challenge
faced by cell therapy manufacturers. Good distribution
practice guidelines stress that medicinal products must
not be exposed to conditions during transport, “that may
compromise their quality and integrity.” Temperature
changes must be tracked to confirm that products stay
within "defined limits" while in transit. The difficulty lies
in establishing these limits, as they must be broad enough
to allow transfer of the product, but not so broad they
jeopardize its quality.
Achieving quality bioproduction at
scale
A product’s critical quality attributes (CQAs) must
be well characterized early on in development before
scaling bioproduction to ensure the correct quality
control metrics are being monitored. Developers want
to avoid the need for major manufacturing changes late
in development as this could impact commercial viability
of the therapy. The term “technology transfer” describes
the transfer of a process from small scale (a laboratory
setting) to a commercial manufacturing facility. If
employing a contract manufacturing organization
(CMO) or contract development and manufacturing
organization (CDMO) to support the scale-up of a
therapy, it’s important to limit technology-transfer risk
by ensuring clear knowledge transfer, adequate training
and ensuring equipment/process commonalities.
Depending on the type of cell therapy, developers will
choose whether to scale-up (increase batch size) or
scale-out (increase the number of batches). Allogenic
therapies are typically scaled up whereas autologous
therapies are scaled out. Automated integration of
multiple manufacturing steps in a closed system
environment is vital to ensure GMP compliance and
process reproducibility, and reduce contamination risk.
Continued exploration and investment in cell therapies
is ushering in a new era of medical interventions. The
discovery of novel targets and mechanisms, refinement
of manufacturing processes and creation of detailed
regulatory guidance are helping to accelerate the approval
of cell therapies, offering hope and improved outcomes for
patients worldwide.
CELL AND GENE THERAPIES: INNOVATIONS, CHALLENGES AND CLINICAL BREAKTHROUGHS 7
Microbial contamination
Biomanufacturing relies on the use of living cells as factories
to produce biotherapeutics, or as therapeutics themselves.
Most biologic drugs are highly specific in their mechanistic
function, making them popular for the treatment of cancers,
autoimmune diseases and infections.
Production and characterization of these drugs are
exceedingly more complex than conventional small
molecule drugs that are chemically synthesized. Cells
must be grown and expanded under aseptic conditions.
The use of good manufacturing practices (GMP) and
strict quality control are applied to mitigate the risk of
product contamination.1
The World Health Organization (WHO) defines
adventitious agents as contaminating microorganisms
of cell culture or source material that have been
unintentionally introduced into the manufacturing
process of a biological product. This includes bacteria,
fungi, mycoplasma, parasites and viruses.2
These
contaminants can be introduced through starting
materials, such as cell substrates, or by environmental
exposure, such as equipment, handling or personnel.
Contamination is a significant issue for bioprocessing
as it results in loss of time, money and effort. Yearly
revenue losses are estimated between $100–300 million
due to contamination.3
Contaminated products that
are not detected during production must also be pulled
off shelves, which may result in shortages of life-saving
drugs and vaccines.4
The risk of contamination necessitates the need
to monitor for microbial contamination during
bioprocessing to mitigate both product lot failures as
well as adverse reactions to patients.
Detecting contamination during
bioprocessing
Bioprocess monitoring is a critical aspect of
manufacturing as it ensures the final product meets the
desired specifications, quality and safety requirements.
Monitoring of contamination is typically achieved by the
WOMEN IN SCIENCE
Microbial Monitoring for
Biopharma Manufacturing
Aron Gyorgypal, PhD
8
Credit: iStock
TECHNOLOGYNETWORKS.COM
analysis of process parameters such as pH, temperature,
dissolved oxygen, nutrient levels and cell density and
viability. These variables are already monitored during
processing as optimized conditions are required to
produce quality products.5
Multiple established methods exist to measure microbial
contamination:
• Enzyme-linked immunosorbent assay (ELISA):
assesses the presence and concentrations of specific
proteins or antibodies and can also be used to indicate
the presence of bacterial contamination.
• Polymerase chain reaction (PCR): can be used
to amplify specific DNA sequences of interest,
particularly for those of microbial contaminants.6
• Process Analytical technology (PAT): may be able
to detect contamination by statistical or mathematic
techniques if process deviations are detected.5,7
• Limulus Amebocyte Lysate (LAL): used to test
for endotoxins that are released by gram-negative
bacteria.8
Contamination’s influence on
product quality and safety
What are some of the key signs of cell culture
contamination? “Because microbes multiply much faster
than mammalian cells, the culture could quickly become
significantly more turbid with a high concentration of
bacteria. The microbes would also rapidly consume the
sugar and oxygen in the culture, effectively starving the
mammalian cells,” says Dr. Erica Berilla, senior research
scientist in the Office of Pharmaceutical Quality Research
(OPQR) within the Food and Drug Administration’s (FDA)
Center for Drug Evaluation and Research.
Berilla’s research focuses on new manufacturing
technologies used in the production of biologics and the
methods used to monitor those manufacturing processes.
Such projects include, but are not limited to, end-to-end
continuous manufacturing, automated in-process sampling
and microbial monitoring during biomanufacturing
processes. This research can help guide and inform the
FDA’s product quality assessors who may be encountering
these new technologies during their assessments of protein
drug substance products.
Berilla’s team wanted to understand microbial
contamination in terms of product quality while also
building an understanding of early contamination
detection. To do this, they intentionally contaminated a
Chinese hamster ovary (CHO) cell bioreactor that was
used to produce a model monoclonal antibody (mAb).
9
Near-infrared (NIR) spectroscopy was used to analyze
the culture in 15-minute intervals to generate a partial
least square (PLS) model. Though the model was not
sensitive to detect low-level contamination it could
detect contaminant in the later stages.
“We found that cell cultures contaminated with
Mycoplasma arginini (M. arginini) plateaued in their
growth once the mycoplasma concentration was maximal,
which usually took two to three days after mycoplasma
introduction… We noted significant increases in
ammonium and depletion of arginine that was not seen
in the control.” This result was perhaps to be expected,
given that there is a competition for nutrients within the
cell culture between the mammalian and the bacterial
cells. Also, because bacteria are more metabolically
efficient, there will be an increase in their proliferation
and production of metabolic byproducts that are toxic to
mammalian cells.
These experiments also gave insights into how the
quality of mAbs is affected by M. arginini contamination.
Principal component analysis revealed a correlation
CELL AND GENE THERAPIES: INNOVATIONS, CHALLENGES AND CLINICAL BREAKTHROUGHS 9
Methods to detect microbe contamination in a bioprocess. Credit:
Aron Gyorgypal, Adobe Illustrator.
TECHNOLOGYNETWORKS.COM
between contamination and increased antibody charge
variants and hypoglycosylation – qualities that influence
therapeutic efficacy and clearance.10
To mitigate the lost time and cost associated with
manufacturing, there needs to be more advances in
detection methods, Barilla says: “Technologies such as
next-generation sequencing (NGS) and spectroscopy
have the potential for use in earlier detection of
adventitious agent events during bioprocessing. Further
development and validation of these technologies could
lead to more rapid/real-time testing.”
NGS hold potential for contamination monitoring
as indicated by the 2024 ICH-Q5A(R2) guidance
document published by the FDA, which has encouraged
the use of the technology. In a recent study by Hiarai et
al., the performance of NGS was evaluated for in vitro
contaminant detection in biologic products.11 Here the
authors inoculated a model adenovirus into Vero cells and
checked for contamination with increasing dilutions to
find the limit of detection (LOD). The authors found that
the LOD for this assay was roughly 2.49x10-4 copies/ngRNA, or about 1:107
dilution from their original inoculate.
This proved to be extremely sensitive, however there
were some drawbacks such as the risk for false positives
that may arise when multiplexing the assay. While these
results are promising, the technology will need to be
validated for contaminant monitoring before it can be
implemented for routine use.
Emerging methods for detecting
microbial contaminations
There are many different methods currently used to detect
contamination. The method of choice will vary depending
on the type of culture that is being run, such as CHO-cellproducing mAbs, human-derived cells for cell therapies or
the production of viral vectors. “The first key factor to look
at is the unit operation along with the sensitivity required
of the assay,” says Dr. Richa Pandey, assistant professor of
biomedical engineering at the University of Calgary.
Pandey's research group is focused on point-of-care
diagnostics, but she has also taken an interest in using
her expertise in biosensor technology, applying it to
biomanufacturing. She recently published a review article
that highlights the current methods for the analysis of
biological contaminants within the biomanufacturing
industry.12 Pandey argues the need for continuous
monitoring systems for microbe detection that can be
enabled by optical and electrochemical sensors and
biosensors equipped with microfluidics.
“Optical sensors work by interacting light with thin films
that are functionalized with nanoparticles that, once
interacted with by the antigen of interest, produce a signal.
These sensors are quite advantageous as they are sensitive
and require low volumes of analyte,” says Pandey.
An example of this technology was showcased by
Zandieh et al., who demonstrated that the use of silver
nanocolumns functionalized with a polycationic peptide,
polymyxin B (PmB), to recognize and interact with
lipopolysaccharide (LPS) a highly toxic endotoxin
produced by gram native bacteria.13 The methodology
allowed sensitivity as low as 340 pg/mL of endotoxin.
Although the conventionally used LAL assay may be
more sensitive, it suffers from the need for sample
prep, which makes this optical sensor methodology
more attractive for a streamlined and fast bioprocess
contamination screening.
Electrochemical-based sensors are another methodology
that have been used to detect bioprocess contaminants.
“Unlike the optical sensor, an electrochemical sensor
measures binding events through either change in
conductance, resistance or capacitance on its surface,”
says Pandey.
CELL AND GENE THERAPIES: INNOVATIONS, CHALLENGES AND CLINICAL BREAKTHROUGHS 10
Representation of an optical (left) and electrochemical (right) biosensor for detecting contamination such as toxins produced by
microbes in a bioprocess. Credit: Aron Gyorgypal, Adobe Illustrator.
TECHNOLOGYNETWORKS.COM
The use of aptamer-based electrochemical sensors has
gained interest in recent years. Their popularity is derived
from their high affinity to a variety of targets, such as
proteins, small molecules and living cells. Kim et al. used an
endotoxin-specific aptamer to detect bioprocess microbial
contaminants.14 This methodology showed reduced
processing time against the conventional LAL, although
with less sensitivity. Studies like this one showcased the use
of aptamers for the detection of not only endotoxins, but
also whole pathogens, and can be used to measure different
compounds in a bioprocess culture.15,16
Indeed, the use of optical and electrochemical sensors does
have some advantages over current practices, given that
they allow for faster detection with little to no processing.
“Of course, there is merit to the current methods used for
contaminant detection, but there still needs to be advances
to make them more sensitive, as well as with automation to
allow for at-line or in-line monitoring,” says Pandey.
Moving towards automated
monitoring
Although the current gold standard technologies for
contamination detection are sensitive and provide specific
detection, they are still limited to batch monitoring. The
current priority is to detect contamination quickly and
accurately, without the need for off-line timely assays. The
production of a real-time monitoring system capable of
continuous contaminant monitoring would allow for early
intervention that would prevent the spread of adventitious
agents and reduce the risk for product recalls.
ABOUT THE INTERVIEWEES
Dr. Erica Berilla is a senior research scientist in the Office of
Pharmaceutical Quality Research (OPQR) within the Food and Drug
Administration’s (FDA) Center for Drug Evaluation and Research. She
obtained a PhD in Biomedical Sciences from the University of South
Florida in 2014.
Dr. Richa Pandey is an assistant professor of biomedical engineering
at the University of Calgary. She obtained a PhD in physical
electronics from Tel Aviv University in 2018. Her research is focused
on wearable biosensors, point of care technologies, biosensors and
lateral flow assays.
REFERENCES
1. Geigert J. Risk Management of the Minimum CMC Regulatory
Compliance Continuum. In: The Challenge of CMC Regulatory Compliance
for Biopharmaceuticals. Switzerland: Springer, Cham; 2023:77-119. doi:
10.1007/978-3-031-31909-9_4. Accessed April 10, 2024.
2. World Health Organization. Annex 2 WHO Good Manufacturing Practices
for Biological Products. https://www.who.int/publications/m/item/
annex-2-trs-no-999-WHO-gmp-for-biological-products. Published
August 19, 2016. Accessed April 10, 2024.
3. Shiratori M, Kiss R. Risk Mitigation in Preventing Adventitious Agent
Contamination of Mammalian Cell Cultures. In: New Bioprocessing
Strategies: Development and Manufacturing of Recombinant Antibodies
and Proteins. Springer, Cham; 2017:75-93. doi: 10.1007/10_2017_38.
Accessed April 10, 2024.
4. Ramanan S, Grampp G. Preventing shortages of biologic
medicines. Expert Rev Clin Pharmacol. 2014;7(2):151-159. doi:
10.1586/17512433.2014.874281.
5. Gomes J, Chopda VR, Rathore AS. Integrating systems analysis and
control for implementing process analytical technology in bioprocess
development. JCTB. 2015;90(4):583-589. doi: 10.1002/jctb.4591.
6. Morris C, Lee YS, Yoon S. Adventitious agent detection methods in
bio-pharmaceutical applications with a focus on viruses, bacteria,
and mycoplasma. Curr Opin Biotechnol. 2021;71:105-114. doi: 10.1016/j.
copbio.2021.06.027.
7. Wehbe K, Vezzalini M, Cinque G. Detection of mycoplasma in
contaminated mammalian cell culture using FTIR microspectroscopy.
Anal Bioanal Chem. 2018;410(12):3003-3016. doi: 10.1007/s00216-018-
0987-9.
8. Novitsky TJ. Biomedical Applications of Limulus Amebocyte Lysate. In:
Biology and Conservation of Horseshoe Crabs. Springer US; 2009:315-
329. doi: 10.1007/978-0-387-89959-6_20. Accessed April 10, 2024.
9. Fratz-Berilla EJ, Faison T, Kohnhorst CL, et al. Impacts of intentional
mycoplasma contamination on CHO cell bioreactor cultures.
Biotechnol Bioeng. 2019;116(12):3242-3252. doi: 10.1002/bit.27161.
10. Das TK, Narhi LO, Sreedhara A, et al. Stress factors in mab drug
substance production processes: critical assessment of impact on
product quality and control strategy. J Pharm Sci. 2020;109(1):116-133.
doi: 10.1016/j.xphs.2019.09.023.
11. Hirai T, Kataoka K, Yuan Y, et al. Evaluation of next-generation
sequencing performance for in vitro detection of viruses in
biological products. Biologicals. 2024;85:101739. doi: 10.1016/j.
biologicals.2023.101739.
12. Janghorban M, Kazemi S, Tormon R, Ngaju P, Pandey R. Methods and
analysis of biological contaminants in the biomanufacturing industry.
Chemosensors. 2023;11(5):298. doi: 10.3390/chemosensors11050298.
13. Zandieh M, Hosseini SN, Vossoughi M, Khatami M, Abbasian S, Moshaii
A. Label-free and simple detection of endotoxins using a sensitive LSPR
biosensor based on silver nanocolumns. Anal Biochem. 2018;548:96-101.
doi: 10.1016/j.ab.2018.02.023.
14. Kim SE, Su W, Cho M, Lee Y, Choe WS. Harnessing aptamers for
electrochemical detection of endotoxin. Anal Biochem. 2012;424(1):12-20.
doi: 10.1016/j.ab.2012.02.016.
15. Weaver S, Mohammadi MH, Nakatsuka N. Aptamer-functionalized
capacitive biosensors. Biosens Bioelectron. 2023;224:115014. doi:
10.1016/j.bios.2022.115014.
16. Robin P, Gerber-Lemaire S. Design and preparation of sensing
surfaces for capacitive biodetection. Biosensors. 2022;13(1):17. doi:
10.3390/bios13010017.
CELL AND GENE THERAPIES: INNOVATIONS, CHALLENGES AND CLINICAL BREAKTHROUGHS 11
Cell therapy involves the transplantation of live human
cells into a patient to repair damaged tissue or cure
disease. A variety of cell types either belonging to the
patient, in the case of autologous therapies, or from a
donor – known as allogeneic cell therapies – can be used.
Although cell therapies in the form of bone marrow
transplants have been used to treat rare forms of blood
cancer since the 1950s, most commercially available cellbased therapies today have been approved within the
past decade.
To date, 27 cell therapies have been approved by the
United States Food and Drug Administration (FDA)
for the treatment of multiple indications, including
various forms of cancer, inherited metabolic disorders
and immune system disorders.1
Many more are likely
to be submitted for approval soon, given the over 1,600
ongoing clinical trials for cell therapies registered with
ClinicalTrials.gov.2
In this article, we take a look at how researchers are
tackling some of the challenges that are preventing
cell therapies from being more widely used, paying
particular attention to cellular immunotherapies for
cancer and induced pluripotent stem cells (iPSC) for
regenerative medicine. We also outline some advances
in manufacturing and regulatory approval processes
that will help expand patient access to innovative, lifechanging treatments.
Non-stem cell-based therapies
These types of therapies involve somatic cells from a
patient or a donor, such as fibroblasts, pancreatic islet
cells or immune cells. Examples include allogeneic
cultured keratinocytes for the treatment of adults with
thermal burns and allogeneic pancreatic islet cells for
people with type 1 diabetes who are unable to maintain
healthy blood sugar levels despite thorough diabetes
management and education.
WOMEN IN SCIENCE
Challenges and Solutions in
Cell Therapy Development
Monica Hoyos Flight, PhD
12
Credit: iStock
TECHNOLOGYNETWORKS.COM
CELL AND GENE THERAPIES: INNOVATIONS, CHALLENGES AND CLINICAL BREAKTHROUGHS 13
The most commonly used somatic cells are T cells, white
blood cells that are critical for triggering an immune
response to pathogens, allergens and tumors. The first
chimeric antigen receptor (CAR) T cell to be approved by
the FDA in 2017 was invented by Professor Carl June at
the University of Pennsylvania. This CAR T-cell therapy
– known as tisagenlecleucel (Kymriah®) – can be used
for the treatment of certain pediatric and young adult
patients with advanced chronic lymphocytic leukemia.
Currently, there are six FDA-approved CAR T-cell
therapies for the treatment of various lymphomas and
myeloma. These cellular immunotherapies involve
collecting T cells from the patient’s blood and modifying
them genetically in the laboratory so that they make
CARs that track down cancer-associated antigens.
When re-infused into the patient, the modified T cells
attach to and kill the cancer cells, thereby helping to clear
the cancer from the body.
Dr. Neil Sheppard, a director in Prof. Carl June’s laboratory,
highlights the advantages of cellular immunotherapies for
cancer: “We know it is possible for the immune system to
completely cure us of even advanced diseases, including
cancer, under the right circumstances and potentially
without damaging healthy tissues,” he explains.
While he acknowledges that a lot of work remains to
be done to realize the full potential of these therapies,
he is optimistic about progress. “With various genetic,
epigenetic and RNA-editing tools the promise of cellular
immunotherapies is within our grasp,” he says.
Next-generation cellular
immunotherapies
Despite the remarkable efficacy of CAR T-cell therapies
against B-cell malignancies, patients can experience
recurrence if the cancer cells stop expressing the targeted
antigen, and CAR T-cell therapies have also shown
limited efficacy against solid tumors.3,4
To address these challenges, Sheppard and others are
pursuing several approaches. These include developing
CAR T cells that target multiple antigens to prevent
immune escape and using alternative cell types such
as natural killer (NK) cells, which do not mediate graftversus-host disease and might be easier to use in an
allogeneic setting.5
“Solid tumors remain a challenge,” says Sheppard. “To
achieve CAR T efficacy in solid tumors, target antigens
such as claudin 6 and claudin 18.2 are being utilized,
sometimes in conjunction with vaccination with the
antigen to boost CAR T-cell proliferation and activity.”
There is growing evidence that intratumoral injection
of CAR T cells can have a greater effect compared to
intravenous infusion. Moreover, combining CAR T cells
with oncolytic viruses that attract CAR T cells to the
tumor aids tumor infiltration and strengthens resistance
to the immunosuppressive tumor microenvironment.6
The recent approval of the first tumor-infiltrating
lymphocyte (TIL) therapy raises new hope for cell
therapies in solid cancers.7
TILs have an innate ability
to seek out cancer neoantigens, fragments of mutated
intracellular proteins that are presented on the surface
of cancer cells. One such treatment for unresectable
or metastatic melanoma – lifileucel (Amtagvi™) –
involves the removal of TILs from patients’ tumor
tissue, growing them in bioreactors and re-infusing
them into patients to destroy cancer cells expressing
these patient-specific antigens.
Stem cell-based therapies
Stem cell treatments play a key role in regenerative
medicine as their ability to self-renew and differentiate
into specialized cells can be used to replace damaged or
diseased cells and restore normal function.
Dr. Shinya Yamanaka’s groundbreaking discovery that
adult cells can be reprogrammed into pluripotent stem
cells through the expression of embryonic transcription
factors offers researchers the opportunity to generate
patient-specific regenerative cell therapies without
having to rely on controversial embryonic stem cells or
worry about immune rejection.8
Since then, great progress has been made in generating
and differentiating iPSCs. “iPSCs are remarkably useful,
and in a class by themselves,” says Professor Jeanne
Loring, founding director of the Center for Regenerative
Medicine and emeritus professor at the Scripps Research
Institute in La Jolla, California. “They are the only noncancer cells that can continue to divide indefinitely in a
culture dish, and retain the ability to give rise to every
cell type in the body.”
TECHNOLOGYNETWORKS.COM
Loring’s research focuses on developing iPSC-based
therapies for Parkinson’s disease, multiple sclerosis and
autism. She believes these therapies hold great promise for
conditions known to be caused by the loss of specific cell
types. However, there are still several obstacles associated
with iPSC-based therapies that need to be addressed before
they can enter clinical trials.
“The development of iPSC-derived cell therapies requires
cutting-edge technologies in multiple areas – from making
the stem cells and differentiating them into the right cell type
in culture, to determining the best stage of differentiation
for transplanting the cells and sophisticated genomics and
bioinformatics, with machine learning to make sure the cells
are at the right stage and that they don’t contain dangerous
mutations,” Loring explains.
In her opinion, technology is key to overcoming these
challenges. “This is a field where all of the work has been
hands-on with highly skilled people, which makes it very
expensive,“ she says. “The use of AI to make decisions
about the culture of the cells and new genomics methods
that ensure the integrity of the genomes of cells before they
are transplanted, are just a couple of examples of ways to
advance cell therapies to the clinic.”
Manufacturing innovations
In addition to sharing the main challenges of drug
development, such as the need to demonstrate safety, target
engagement and pharmacological activity, cell therapies face
unique challenges. “The manufacturing process for CAR
T-cell therapy greatly affects the potency of cell therapy
product,” says Sheppard. His team is exploring how to scale
up their cell manufacturing process in the laboratory into a
large-scale good manufacturing process (GMP) needed for
clinical trials or even a commercial scale process designed
to serve thousands of patients each year.
So far, they have learned that, with patient-specific immune
cells, the longer the duration of the cell manufacturing
process and the more manipulation steps involved, the
lower the potency of the emerging cell. “The evidence
shows that CAR T-cell products made in manufacturing
processes as short as one to three days have superior
activity once inside the human body compared to those
made over nine days or more.”
In the not-too-distant future, Sheppard envisages the
creation of self-contained GMP machines where a patient’s
T cells are loaded and the chosen type of CAR T-cell therapy
is produced at the press of a button in a hospital setting.
“I imagine the future of CAR T-cell therapy production
being as simple from the prescriber’s perspective as using
the office coffee machine – load the desired sachet (gene
vector), water (reagents) and a cup (cells), press the button
and your work is done.”
Speeding up production of stem
cell-based therapeutics
Since 2003, the UK’s Stem Cell Bank (UKSCB) has
been supporting the development of pluripotent stem
cell-derived therapies and driving improvements in the
manufacturing process to produce quality-controlled
pluripotent stem cell lines (iPSCs and human embryonic
stem cells, or hESCs) that can be differentiated into
specialized cell types for tissue regeneration. At the 20th
anniversary of the cell repository last October, Dr. Lee
Carpenter, the head of the UKSCB, highlighted automation
and machine learning as being key to accelerating the safe
manufacture of stem cell-derived therapies and giving
patients faster access to potentially life-saving treatments.9
The UKSCB has demonstrated, using a robotic system, that
clinical-grade pluripotent stem cell production is possible
and is currently comparing the automated process to manual
production. The use of automation, robotics and modular
production platforms will not just help reduce reliance on
manual operations and variability in process performance, it
will also enable scalability and drive down costs. “There is a
race to develop the instrumentation to automate stem cellderived cell therapy manufacturing,” says Loring.
Streamlining the approval process
The final hurdle when developing cell therapies is
obtaining regulatory approval. The review of a New
Drug Application (NDA) by the FDA can take up to a
year, significantly delaying the commercialization of a
new therapy.10
Cell therapies are evolving quickly, and so too must the
regulations. Regulatory agencies are adapting to the
unique challenges of cell therapies by implementing
CELL AND GENE THERAPIES: INNOVATIONS, CHALLENGES AND CLINICAL BREAKTHROUGHS 14
TECHNOLOGYNETWORKS.COM
pathways aimed at expediting assessments, such as
the FDA's Regenerative Medicine Advanced Therapy
(RMAT) designation and the European Medicines
Agency (EMA)'s Priority Medicines (PRIME) scheme.11
In addition, the FDA is on a recruitment drive to hire
more staff in its Office of Therapeutic Products to tackle
the increasing workload.12
Efforts to increase the dialog between regulators,
industry and academia to harmonize regulatory
standards are also starting to bear fruit. The benefits
of international programs to build regulatory capacity
and share information are evidenced by multinational
approvals of CAR T-cell therapies.13 As the cell therapy
pipeline continues to expand, increasing the alignment of
regulatory pathways across countries will be crucial to
facilitating access to new transformative treatments to
patients around the world.
ABOUT THE INTERVIEWEES
Dr. Neil Sheppard is the director of the Therapeutic Innovation in
Natural Killer cells (THINK) lab, which focuses on improving the
therapeutic application of NK cells in cancer. As part of Prof. Carl
June's team, he is also overseeing the development of combination
strategies for clinical-stage CAR T-cell therapies. Sheppard joined
Penn Medicine’s Center for Cellular Immunotherapies in 2019 and
has over 20 years experience of working on immunotherapies in
academia, pharma and biotechnology companies.
Dr. Jeanne Loring is professor emeritus at The Scripps Research
Institute, where she was the founding director of the Center for
Regenerative Medicine. She is an internationally recognized
authority in stem cell research and has received multiple awards
for her accomplishments as a scientist, educator and advocate
for evidence-based patient therapies. In 2018 she co-founded
the biotechnology company Aspen Neuroscience to develop
personalized cell therapies for Parkinson’s disease.
REFERENCES
1. Approved Cellular and Gene Therapy Products. FDA. Published
online March 18, 2024. Accessed April 26, 2024. https://www.fda.
gov/vaccines-blood-biologics/cellular-gene-therapy-products/
approved-cellular-and-gene-therapy-products
2. Active clinical trials of cellular therapies. clinicaltrials.
gov. https://clinicaltrials.gov/search?intr=cell%20
therapy&aggFilters=status:act
3. Cappell KM, Kochenderfer JN. Long-term outcomes following
CAR T cell therapy: what we know so far. Nat Rev Clin Oncol.
2023;20(6):359-371. doi: 10.1038/s41571-023-00754-1
4. Hou AJ, Chen LC, Chen YY. Navigating CAR-T cells through
the solid-tumour microenvironment. Nat Rev Drug Discov.
2021;20(7):531-550. doi: 10.1038/s41573-021-00189-2
5. Myers JA, Miller JS. Exploring the NK cell platform for cancer
immunotherapy. Nat Rev Clin Oncol. 2021;18(2):85-100. doi:
10.1038/s41571-020-0426-7
6. Daei Sorkhabi A, Mohamed Khosroshahi L, Sarkesh A, et
al. The current landscape of CAR T-cell therapy for solid
tumors: Mechanisms, research progress, challenges, and
counterstrategies. Front Immunol. 2023;14. doi: 10.3389/
fimmu.2023.1113882
7. Mullard A. FDA approves first tumour-infiltrating lymphocyte
(TIL) therapy, bolstering hopes for cell therapies in solid cancers.
Nat Rev Drug Discov. 2024;23(4):238-238. doi: 10.1038/d41573-
024-00035-1
8. Takahashi K, Yamanaka S. Induction of pluripotent stem cells
from mouse embryonic and adult fibroblast cultures by defined
factors. Cell. 2006;126(4):663-676. doi: 10.1016/j.cell.2006.07.024
9. Patients could have faster access to ground-breaking stem cell
treatment with manufacturing innovation. GOV.UK. Published
online October 11, 2023. Accessed April 29, 2024. https://www.
gov.uk/government/news/patients-could-have-faster-accessto-ground-breaking-stem-cell-treatment-with-manufacturinginnovation
10. Rotolo A, Chabannon C, Gramignoli R. Identification of hurdles
in the development of cell-based therapies. Cytotherapy.
2020;22(2):53-56. doi: 10.1016/j.jcyt.2019.12.009
11. Drago D, Foss-Campbell B, Wonnacott K, Barrett D, Ndu A.
Global regulatory progress in delivering on the promise of gene
therapies for unmet medical needs. Mol Ther Methods Clin Dev.
2021;21:524-529. doi: 10.1016/j.omtm.2021.04.001
12. Establishment of the Office of Therapeutic Products. FDA.
Published online March 16, 2023. Accessed April 29, 2024.
https://www.fda.gov/vaccines-blood-biologics/cellular-genetherapy-products/establishment-office-therapeutic-products
13. Arcidiacono J. International Harmonization for Cell and Gene
Therapy Products. In: Galli MC, ed. Regulatory Aspects of Gene
Therapy and Cell Therapy Products: A Global Perspective.
Springer International Publishing; 2023:235-240. doi:
10.1007/978-3-031-34567-8_14
CELL AND GENE THERAPIES: INNOVATIONS, CHALLENGES AND CLINICAL BREAKTHROUGHS 15
Cancer has claimed more than eight million lives
worldwide each year since 2012.1
While mortality rates
of major cancer types like lung, breast and colorectal have
decreased, the overall number of cancer patients is not
seeing a decline. Cell-based immunotherapy is an emerging
and promising treatment using engineered immune cells
such as cytotoxic T cells and natural killer (NK) cells to
eliminate cancer cells.2
However, there are challenges for
effective use of immune cells against solid tumors including
the immunosuppressive tumor microenvironment and
antigen escape by cancer cells. In addition, cancer cells
can also construct a glycocalyx barrier against attack by
patrolling immune cells and engineered cell therapies.
A glycocalyx barrier is a carbohydrate or sugar coating
that can biochemically downregulate the activities of
immune cells and physically shield molecular interactions
between immune cells and their target cancer cells. One
can consider the glycocalyx as an armor of cancer cells.
Mucins are an important component of the glycocalyx
that, at high cell surface densities, can form a thick and
brush-like layer to protect immune cells from being
recognized by immune cells.3
Park and colleagues recently showed that enhanced
stimulation of NK and T cells with chimeric antigen
receptors (CARs) can improve cytotoxicity against
mucin-bearing cancer cells.4 CARs are engineered
proteins that enable immune cells to recognize specific
cancer antigens and enable cancer cell destruction. In
addition, by endowing the surfaces of effector immune
cells with glycocalyx-editing enzymes such as mucinases,
such immunoengineering approaches can overcome the
glycocalyx armor of cancer cells.
A model of glycocalyx armor
To understand how the thickness of the glycocalyx layer
protects cancer cells from destruction by immune cells,
the authors created a breast cancer cell line that stably
WOMEN IN SCIENCE
Removing the Glycocalyx Armor
of Cancer Cells for Treatment
Andy Tay, PhD
16
Credit: iStock
TECHNOLOGYNETWORKS.COM
expresses a green fluorescent protein-mucin (GFP-Muc1)
protein under the control of a tetracycline-inducible
promoter. This meant that at higher doses of doxycycline
(an antibiotic), there was stronger induction of mucin
expression. The authors also made sure there was
minimal cell-to-cell variability in Muc1 surface levels on
their breast cancer cells.
The higher the expression of GFP-Muc1, the weaker the
killing efficacy by NK-92, a NK cell line which is being
tested for anti-cancer therapy. However, removal of Muc1
ectodomain with StcE, a mucin-specific metalloprotease,
and genetic deletion revoked the protection. Ectodomain
is the domain of a membrane protein that extends into the
extracellular space. They are usually the parts of proteins
that initiate contact with surfaces, which leads to signal
transduction. A change of glycocalyx thickness from ~100
nm to ~60 nm made a huge difference to whether NK-92
cells were able to eliminate the breast cancer cells.
The authors further proposed that the mucin-rich glycocalyx
can be modelled as a polymer brush whose properties can
be tuned by glycosylation and crosslinking. For instance,
breast cancer cells with mucins that are truncated and
contain less glycosylation (at the O-glycan sites) were more
vulnerable to NK-92-medicated cytotoxicity.
Synergy between chimeric antigen
receptors and glycocalyx-editing
enzymes
To assess the utility of engineered immune cells to
eliminate cancer cells, the authors introduced NK-92 cells
and primary T cells with CARs against human epidermal
growth factor receptor 2 (HER2) and CD19. Interestingly,
even at high GFP-Muc1 induction and expression levels,
CAR NK and T cells were able to eliminate the cancer
cells, suggesting that protection mediated by a thicker
glycocalyx layer can be at least partially overcome with
stronger receptor-mediated stimulation between CARs
and their targets.
To further enhance the cancer killing efficacy of
CAR immune cells, the authors considered other
immunoengineering approaches to enable immune
cells to kill target cancer cells that express high levels of
mucin. They first tethered the surfaces of NK-92 cells with
StcE mucinase that is known to break down gut mucosal
mucin barriers and found that surprisingly, StcE would
readily and stably adhere to the surfaces of NK-92 cells for a
prolonged period even after stringent buffer washing. Further
experiments with a mutated mucinase i.e. StcE ΔX409, which
was catalytically active but did not tether to the NK-92 cell
surface after rinsing, found that there was no enhanced
cytolytic ability, indicating the importance of tethering.
When the authors performed brightfield imaging, they
found that while both unmodified and StcE-modified
NK-92 cells could form viable mechanical connections
with cancer cells, a high density of GFP-Muc1 and cancerassociated microvilli persisted in the contact zone. On the
other hand, with StcE-modified NK-92 cells, there were
lower GFP-Muc1 levels and fewer microvilli, suggesting
that surface-tethered StcE locally cleaved the mucin and
disrupted cancer-associated microvilli.
The synergistic effects of StcE-modifications with HER2
CAR NK-92 cells were demonstrated as the treated cancer
cells had 50% (120 nm to 60 nm) reduction in glycocalyx
thickness and there was close to 300% and 40% better
cytolytic outcomes compared to unmodified NK-92 cells
and HER2 CAR NK-92 cells, respectively.
Facile method to equip immune
cells with mucinase
To improve the conjugation of StcE to the cell surfaces of
NK cells, the authors finally developed a modular strategy
by anchoring glycocalyx-editing enzymes onto surfaces of
NK-92 cells through leucine-zipper-mediated coupling.
First, the RR leucine zipper was genetically expressed
onto the surfaces of NK-92 cells as a fusion protein with
an ALFA-tag and sfGFP fluorescent marker to form ZIPNK-92 cells. StcE was also modified with an EE leucine
zipper, enabling it to bind strongly to ZIP-NK-92 cells and
cleave Muc1 from surfaces of cancer cells.
"This is an interesting study that suggests a new angle for
attacking cancer cells by trying to attack its the protective
glycocalyx," Dr. Pakorn Kanchanawong, associate
professor at the National University of Singapore, who
is not involved in this paper said. "In terms of future
therapeutic research, especially in vivo, I would imagine
that as the glycocalyx is probably easily replenishable by
CELL AND GENE THERAPIES: INNOVATIONS, CHALLENGES AND CLINICAL BREAKTHROUGHS 17
TECHNOLOGYNETWORKS.COM
cells, very likely this will have to be pursued in conjunction
with perturbing glycosylation mechanisms," he added.
In conclusion, immunoengineering of immune cells with
surface-displayed or secreted enzymes that can disrupt the
glycocalyx armor of cancer cells is an attractive strategy for
cancer treatment. This approach is particularly powerful
if it can be used to specifically cleave cancer-associated
mucins. Additionally, pharmacological and metabolic
agents can be employed to disrupt the synthesis of
glycocalyx in cancer cells, for synergy with CAR immune
cell therapies. Such a combined strategy effectively
leverages on advances in biomolecular and biophysical
understanding of interactions between effector immune
cells and their target cancer cells.
ABOUT THE INTERVIEWEE
Dr. Pakorn Kanchanawong is an associate professor at the National
University of Singapore. His research interests include superresolution microscopy, optical bioimaging, force generation, force
transmission and force sensing, and mechanobiology of cell–cell
and cell–matrix interaction.
REFERENCES
1. Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A.
Global cancer statistics, 2012. CA Cancer J Clin. 2015;65(2):87-
108. doi: 10.3322/caac.21262
2. Mellman I, Coukos G, Dranoff G. Cancer immunotherapy comes of
age. Nature. 2011;480(7378):480-489. doi: 10.1038/nature10673
3. Paszek MJ, DuFort CC, Rossier O, et al. The cancer glycocalyx
mechanically primes integrin-mediated growth and survival.
Nature. 2014;511(7509):319-325. doi: 10.1038/nature13535
4. Park S, Colville MJ, Paek JH, et al. Immunoengineering can
overcome the glycocalyx armour of cancer cells. Nat Mater.
2024;23(3):429-438. doi: 10.1038/s41563-024-01808-0
CELL AND GENE THERAPIES: INNOVATIONS, CHALLENGES AND CLINICAL BREAKTHROUGHS 18
While gene therapy has proven to be promising
for diseases ranging from cancer to diabetes, the
challenge of getting the right dose of genetic material
into target cells has caused a bottleneck in the
application of such therapies.
In new research published in PLOS ONE, researchers
from the University of Wisconsin–Madison have
reported on the development of a technique employing
electric pulses to make the human body more receptive
to certain gene therapies.
We spoke to two of the study authors, Professors Susan
Hagness and John Booske, to learn more about the
benefits of direct delivery of gene therapy materials,
the challenges associated with gene therapy delivery
and the use of electronic pulses to encourage uptake of
genetic material.
Q: What are the benefits of direct delivery
of gene therapy material?
A: Direct delivery may reduce the total dose needed
for treatment because it eliminates the attrition of
material during circulation through the body and other
organs prior to arrival in the targeted tissue/organ
(compared to, say, systemic, peripheral injection into a
remote blood vessel).
WOMEN IN SCIENCE
Electronic Pulses Could Reduce
the Need for High Doses in
Gene Therapy Delivery
Kate Robinson
20
Credit: iStock
TECHNOLOGYNETWORKS.COM
Systemic delivery typically requires large(r) doses
to compensate for losses during passage through the
circulatory system and other organs. Manufacturing the
genetic material delivered via virus vectors is expensive
– prohibitively so for many treatments of inherited
metabolic diseases (such as hemophilia, diabetes, etc.).
Reducing the required dose is critical to practical,
affordable treatments.
Direct delivery may also minimize the time the material
spends in the circulatory system before uptake in the
liver cells. This reduces the likelihood that the immune
system will mount an attack on the gene therapy “foreign”
material and destroy or inactivate it. The larger doses
required with systemic, peripheral injection, along with
the associated immunological response (e.g., cytokine
storms) present a heightened risk.
Direct delivery is a pathway to reduce the doses required
and bypass the immune counter-response, thereby
reducing cost and increasing safety.
Q: What are the challenges of gene
therapy delivery, and how do these affect
patients?
A: The challenges of conventional gene therapy delivery
approaches (peripheral injection, e.g., in a remote blood
vessel) include the requirements for large doses (to ensure
enough of the dose finally arrives at the targeted site
and is taken up by the targeted tissue cells) and the risk
of immune counter-response to the injection of foreign
genetic material.
The former leads to a prohibitively expensive cost of
treatment, making it impractical as a widespread treatment
option. The latter includes loss of genetic material that may
result in ineffective treatment or a dangerous overreaction
that can threaten the health of the patient.
Q: How does the application of electronic
pulses increase the uptake of gene therapy
material into hepatocytes?
A: We are not certain what the specific underlying
physical mechanisms are that result in enhanced uptake of
the gene therapy material. We have some hypotheses, but
no definitive identification of a mechanism currently.
Our findings, reported in the PLOS ONE article, help to
rule out some of the possible mechanisms. For example,
we know that electric pulses do not modify the genetic
material directly before it is absorbed into the cells,
and we know that the electric pulses do not modify the
culture medium or environment that the cells reside in.
Future experiments are being designed and conducted
to pin down the mechanism(s) that are at play. We
know that electric pulses induce (nano)pores into the
cell membranes, and this effect has been exploited in
other investigations to permeabilize the membranes
and allow small(er) molecules to be taken up by those
cells. However, the larger size of the virus vector (AAV8)
capsids used in our experiments lends doubt to the
likelihood that this is the mechanism responsible for
enhanced uptake in our experiments. It is a subject for
further investigation.
Q: What impact could efficient delivery
of gene therapies have on patients in the
future?
A: Efficient delivery could make many gene therapy
cures affordable and safe for a large population
of patients. To elaborate, genetic mutation-based
metabolic diseases significantly reduce the quality of
life for hundreds of millions of people in the world.
There are 100s of such diseases, including diabetes,
cystic fibrosis, sickle cell anemia and hemophilia.
Many of them involve the liver due to its central role
in metabolism. Developed countries spend trillions of
dollars each year on patient care, with nearly a trillion
dollars spent annually on type 1 diabetes (T1D) alone.
Cures for many of these diseases could be attainable
if practical, cost-effective methods existed to modify
the gene(s) of the liver cells, sufficient to correct the
inherited metabolic discrepancy. Some success with
systemic injection gene therapy has been reported, but
only in small mammals or with prohibitive costs (~$1
M/treatment in the case of hemophilia). In other words,
a famous statement in 1999 by Salk Institute Professor
Inder Verma, one of the foremost recognized leaders
in gene therapy, still remains relevant today: “There
are only three problems in gene therapy: delivery,
delivery, delivery.”
CELL AND GENE THERAPIES: INNOVATIONS, CHALLENGES AND CLINICAL BREAKTHROUGHS 21
TECHNOLOGYNETWORKS.COM
Q: What are the next steps in translating
this research into clinical trials?
A: Next steps include in vitro investigations of optimal
electric pulse parameters and in vivo studies to determine
how the phenomenon (which we have observed in vitro)
manifests in living tissues. To date, we have only had the
opportunity to investigate electric pulsing with a single
choice of electric field strength and pulse length, and
only with single pulses.
An important question to answer is whether other
treatment parameter combinations–i.e., different electric
field strengths, different pulse lengths, and/or multiple
pulses–produce a stronger effect. Of course, it is
important to identify the maximum treatment parameter
thresholds above which cell or tissue damage occurs, to
ensure that parameter choices are always safely below
those thresholds. Meanwhile, our experiments to date
were conducted on cells in well culture plates (i.e., in
vitro). Although research has shown that cells in vivo
(in living tissues) can have similar responses to electric
pulsing as cells in vitro, the magnitude of the responses
and the parameter values that produce the maximum safe
response can be expected to be different. So, experiments
with animal models are the next critical step, especially
with larger mammals.
Q: Do you have plans to explore the use
of electronic pulses in the delivery of gene
therapies to other cell types?
A: The potential impact of translating this to clinical
practice in liver/hepatocytes has considerable impact
value, so that is our primary focus for now. But certainly,
expanding the scope of these investigations is of interest
in the longer term.
ABOUT THE INTERVIEWEES
John Booske is the Keith and Jane Morgan Nosbusch emeritus
professor in electrical and computer engineering at University
of Wisconsin–Madison. His research focuses on plasmas,
metamaterials, metasurfaces and media that have a strong
interaction with electromagnetic radiation, electromagnetic field
effects and microwave vacuum electronics. John holds a PhD in
Nuclear Engineering from the University of Michigan.
Susan Hagness is the Philip Dunham Reed Professor and department
chair of electrical and computer engineering at the University of
Wisconsin–Madison. Her group’s research spans computational
and experimental applied electromagnetics, with an emphasis on
bioelectromagnetics. Susan golds a PhD in Electrical Engineering
from Northwestern University.
CELL AND GENE THERAPIES: INNOVATIONS, CHALLENGES AND CLINICAL BREAKTHROUGHS 22
There are over 6,000 genetic disorders and the list grows
continually. Gene therapy works to treat or prevent disease
of monogenic cause by modifying an individual's genes. In
2012, alipogene tiparvovec was the first to receive European
regulatory approval for treating lipoprotein lipase deficiency
by delivering a functional copy of the enzyme to muscle cells
using an adeno-associated viral (AAV) vector. This approval
(now withdrawn due to low demand) was followed closely
in 2017, in the US, by voretigene neparvovec, for treating
Leber congenital amaurosis, an inherited eye disease of
progressive blindness, in biallelic carriers of mutations to
the RPE65 gene.
Since these landmark approvals, there has been several
additional approved gene therapies. Aside from treating
monogenic disorders, AAV-based gene therapies have
also been approved for treating certain cancers, through
engineered chimeric antigen receptor T cells, and they are
being explored against infectious diseases via delivered
recombinant antibodies.
The gene therapy landscape is dynamic and constantly
shifting. The technology holds the promise of potential
disease modification, providing life-altering benefits for
patients that otherwise would have faced dire outcomes. In
the midst of this shifting landscape, the gene therapy field
has celebrated victories and faced obstacles. Herein, we will
review major advances in disease modification for illnesses
that presently still lack effective treatments, as well as some
challenges and approaches to overcome them.
Gene therapy: Panacea
for neuromuscular and
neurodegenerative disease?
Neuromuscular diseases (NMDs) and neurodegenerative
diseases (NDDs) are generally progressive illnesses that
often shorten lifespan. Unfortunately, many also lack
disease-modifying therapies. This has made NMDs and
NDDs of genetic etiology prime candidates for gene
therapy, by addressing the root cause of disease, i.e., the
WOMEN IN SCIENCE
Gene Therapy: A New Frontier
in Disease-Modifying Therapies
Masha G. Savelieff, PhD
23
Credit: iStock
TECHNOLOGYNETWORKS.COM
defective gene. The 2019 approval by the US Food and
Drug Administration for onasemnogene abeparvovec, an
AAV therapy for spinal muscle atrophy (SMA), marked a
turning point in the treatment of NMDs.
“SMA is a NMD caused by mutations to the gene SMN1,
which codes for survival of motor neuron, a protein
essential to neuronal health,” explained Kathrin Meyer,
assistant professor at the University of Missouri, who was
involved in the preclinical and clinical studies that led to
the approval of onasemnogene abeparvovec.
SMA patients exhibit gradual loss of motor neurons,
leading to progressive muscle weakness, although the
rate of decline varies across individuals. In the most
severe cases, respiratory failure occurs. “The gene therapy
treatment for SMA involves delivering a functional copy
of SMN1 using an AAV9 carrier by intravenous injection.
In the clinical trial, recipients experienced improved
functional and survival outcomes compared to natural
history data of the disease progression,” Meyer elaborated.
Meyer is presently assessing the feasibility of gene therapy
for additional NMDs and NDDs. One focus is Batten
disease and the family of neuronal ceroid lipofuscinoses,
which are NDDs caused by mutations to lysosome storage
genes called ceroid-lipofuscinosis neuronal (CLN). “We
found that delivery of AAVs carrying a functional CLN
copy improves lifespan and brain and behavioral pathology
in CLN6 and CLN8 animal models of Batten disease,”
Meyer summarized of this line of work.
Guangping Gao, director of the Horae Gene Therapy
Center at the University of Massachusetts Medical
School, is also actively researching gene therapy as a
potential treatment avenue for NMDs and NDDs. “We
are especially interested in Canavan disease, a fatal rare
recessive NDD caused by loss-of-function mutations to
aspartoacylase (ASPA),” Gao described of his research.
Loss of aspartoacylase activity prevents breakdown
of N-acetylaspartate, which accumulates in the central
nervous system and interferes with neuron myelin
sheaths. Disease onset occurs in infancy and manifests
with poor motor function with reduced head control, lack
of visual fixation and muscular hypotonia, which develops
to include spasticity, ataxia and seizure. “We have shown
that AAV-mediated delivery of a functional ASPA copy
to ASPA–/– mice drastically improves neuropathology,
myelination and motor function. Moreover, using our
Canavan disease model and gene therapy, we have been
able to shed insight into disease pathophysiology, such as
peripheral involvement, would could help further refine
our therapeutic approach,” Gao explained.
Gene therapy can replace genes with defective loss-offunction mutations with a functional copy of the gene.
Alternatively, the AAV vehicle can be used to deliver short
hairpin (shRNA) or interfering RNAs (RNAi) to knock down
expression of a gene with toxic gain-of-function mutation.
This is a viable approach, for example, for cases of the motor
neuron disease amyotrophic lateral sclerosis caused by
mutations to the gene superoxide dismutase 1 (SOD1). In
animal models of ALS, AAV-mediated delivery of shRNA or
RNAi slows disease progression in SOD1G93A mice.
Rather than targeting a specific mutation, gene therapy can
also be used to support damaged tissues, e.g., delivering a
copy of gene that strengthens muscles to treat NMDs with
muscle weakness, or a growth factor to regenerate injured
nerves. Ultimately, a gene therapy with a base-editing
CRISPR-Cas cargo could correct genes, with either lossor gain-of-function mutations.
Gene therapy has already celebrated some successes,
with approved therapies that tangibly improve outcomes
for patients, and potential developments in place for
additional diseases. Like all technologies, however, it also
faces some obstacles.
Overcoming the immune response
A prominent challenge facing gene therapies is that AAVs
and their transgene cargo can elicit an immune response,
innate or adaptive, which diminishes transduction efficiency
and long-term gene expression and increases the chances
of adverse events. “Ideally, gene therapy was envisioned as
a one-shot treatment, but in practicality, re-dosing is likely
needed depending on the target tissue and age of treatment.
Moreover, although AAVs are quite effective, high systemic
doses are sometimes needed. Both these factors potentially
provoke adverse events associated with AAV and transgene
immunogenicity,” Gao discussed.
CELL AND GENE THERAPIES: INNOVATIONS, CHALLENGES AND CLINICAL BREAKTHROUGHS 24
TECHNOLOGYNETWORKS.COM
One route is to change the injection method for AAV
administration. “It is possible to reduce a systemic immune
response by employing intrathecal injection. Although
this delivery route is safer, local immune responses in
highly targeted regions, so called “hotspots” can still be of
concern,” Meyer explained. “However, this injection method
preferably targets central nervous system tissue, such as the
brain and spinal cord, which will not be the optimal tissue
target for many diseases.”
Another option is to eliminate preexisting AAV antibodies,
resulting either from prior AAV gene therapy doses or
from natural infection to wild-type AAV, which is endemic
to humans. “There are several ways to accomplish this,”
Gao outlined. “Pharmacological agents, for instance
rituximab to deplete B cells, or physical methods, such as
plasmapheresis and AAV serotype-specific IgG absorption
to remove circulating antibodies. We can also block the
innate response through complement deactivation with
antibodies against complement components, such as
anti-C5 eculizumab.”
Moreover, it is possible to engineer the AAV capsid, vector
genome and transgene, e.g., by “cloaking”, to lower their
immunogenicity. The transgene expression from AAV
vector can also be engineered to evade immune cells, such
as antigen presenting cells (APCs), an arm of the adaptive
immune response. Gao developed a microRNA (miR)-
based strategy to evade APCs.
In a proof-of-concept study using an AAV expressing
the highly immunogenic ovalbumin, Gao demonstrated
that incorporating the target sites of the APC-specific
miR142 and other miRNAs into the transgene detargeted
ovalbumin expression from APCs, i.e., they did not display
ovalbumin antigens, blunting their immune response
against it. In tandem, the cytotoxic T-cell response against
ovalbumin was also blunted. More recently, Gao extended
this approach by evaluating combinatorial microRNAs to
better block the APC and cytotoxic T-cell response.
Towards tailored tissue targeting
and transduction
Another major goal of gene therapy research is improving
AAV tissue specificity and augmenting tissue transduction,
i.e., that the AAVs target the intended tissue and disseminate
widely across it. Wild-type AAVs occur in several serotypes,
distinct variations in the capsid, that differ in their tissue
specificity and ability to cross the blood–brain barrier,
impacting their tissue targeting and transduction efficiency.
“Widespread transgene delivery is a major challenge,”
Meyer elaborated. “There are AAVs, such as AAV9, with
good ability to cross the blood–brain barrier and access
the brain and spinal cord, which are optimal target tissues
for NMDs and NDDs.” Using intrathecal AAV9 injection,
Meyer successfully showed widespread transgene
expression in mouse and nonhuman primate animal models,
which achieves up to 80% targeting to spinal motor neurons
as well as across various brain areas. Transduction can be
further enhanced by placing the animal in the Trendelenburg
position – lain flat on the back with the head tilted 15 to 30
degrees below the feet. “Although we’ve developed injection
methods that enhance transgene distribution across neural
tissue, penetration to the deep layers remains relatively
limited, but we are actively researching better methods,”
Meyer added.
An alternative to wild-type AAVs is to rationally engineer
or evolve novel capsids with distinct or improved tissue
targeting and blood–brain barrier and trans-vascular
penetrance. “Improved capsid specificity and transduction
efficiency will mean lower doses, which will mean fewer
possible complications arising from immune responses
to high dose AAV gene therapy,” Gao explained. “Further
refinement may be achieved by tissue-specific codonoptimized and CpG motif-reduced transgenes, to enhance
translation within the target tissue versus other tissues
and blunt innate immune response.”
Gene expression in moderation
and in context
Another facet of gene therapy research is modulating
transgene expression. The overarching goal is stable
and durable expression, but to levels commensurate
with physiological expression. “You need to be able to
physiologically regulate transgene expression because
uncontrolled expression, even of endogenous proteins,
becomes toxic at excessive levels,” Gao elaborated.
“Furthermore, you want to be able to turn some gene
therapies off. For example, if using an AAV vehicle to deliver
CRISPR-Cas cargo, you don’t want the Cas nuclease
CELL AND GENE THERAPIES: INNOVATIONS, CHALLENGES AND CLINICAL BREAKTHROUGHS 25
TECHNOLOGYNETWORKS.COM
expressed in perpetuity because it will increase off-target
cleavage and cause unintended genomic damage.”
Meyer suggested that the field needs to understand
expression of the target gene in its native environment
better. “Animal models are great and have helped us test
the efficacy of potential gene therapies for rescuing disease
phenotypes. However, oftentimes we don’t understand the
disease mechanisms in detail. On paper it looks simple; it
seems like you might need to deliver a functional gene copy
for diseases with loss-of-function mutations, and deliver
a shRNA or RNAi for diseases with gain-of-function
mutations. In practice, it isn’t always this straightforward.”
In amyotrophic lateral sclerosis, for example, the most
common known monogenic cause are repeat expansions
to the gene C9orf72, which cause both toxic gain-offunction as well as haploinsufficiency and loss-offunction. “Generating models using patient-derived cells
allows us to investigate disease pathophysiology and gene
expression patterns of genetic mutations in context, and to
dissect contributions from various cellular compartments.
Failing to understand the complex mechanisms in context
in patient cells could lead to patients not responding to a
gene therapy.”
A hefty price tag
Although gene therapies have profoundly improved
outcomes for some patients, their hefty price tags limit
access. Moreover, some NMDs and NDDs are caused by
rare mutations, so patients are not always diagnosed early.
“There are three major hurdles that are currently impacting
translation of gene therapies to patients,” Meyer explained.
“On the first front, we need to streamline manufacturing,
which is currently expensive and variable. On the second
front, regulatory aspects are difficult to navigate because
methods to assess gene therapies, such as dosage, are not
standardized, making it problematic to compare efficacy
between different gene therapy products. This was the
scenario with therapeutic antibodies a while back, which
has since been streamlined, so we should be able to achieve
the same for gene therapies. On the third front, we need to
make sequencing technologies more accessible to patients
with rare diseases to identify mutations and diagnose
patients earlier. We will need to be able to treat patients
earlier, pre-emptively, before too much tissue damage has
already occurred to truly modify the disease course.”
Gao echoed similar sentiments regarding manufacturing
when asked about future directions for gene therapy. “I
think if we look to the future, we have to identify what
the current problems are, the challenges we are presently
facing,” he replied. “If we are to bring gene therapies
to patients more readily down the drug development
pipeline, we will need to develop better manufacturing
platform technologies and lower costs.”
ABOUT THE INTERVIEWEES
Kathrin Meyer is an assistant professor in the department of
physical medicine and rehabilitation at the University of Missouri.
She studied molecular and cellular biology at the University
of Bern, Switzerland followed by a post-doctoral fellowship at
Nationwide Children’s Hospital, Columbus Ohio, USA developing
different therapeutic approaches for spinal muscular atrophy.
Guangping Goa is a professor in microbiology and physiological
systems, the director of the Horae Gene Therapy Center and the
Viral Vector Core and the Penelope Booth Rockwell Professor
in Biomedical Research at the University of Massachusetts
Medical School.
CELL AND GENE THERAPIES: INNOVATIONS, CHALLENGES AND CLINICAL BREAKTHROUGHS 26
Biopharmaceuticals – also known as biological therapies
or biologics – are usually derived from some form of living
system, be it cell culture, animal products or human cell or
tissue samples. They are complex and expensive to produce
and highly vulnerable to degradation and contamination,
requiring careful analysis during research, development
and manufacturing stages to ensure quality and integrity. In
this article we highlight some of the analytical techniques
employed in biopharmaceutical analysis, and how the field
is evolving to meet new challenges. .
Biopharmaceutical
characterization and quality
The field of biopharmaceuticals not only includes
antibody-based drugs, but also the expanding fields of cell
and gene therapies. Across the spectrum of biologics, the
most common analytical technologies for characterizing
biopharmaceuticals are mass spectrometry (MS) and
liquid chromatography (LC), either alone or in tandem.
“Some of the most important properties you’re testing for
with biologics are post-translational modifications (PTMs)
and protein aggregation, said Jared Auclair, director of
bioinnovation at the Biopharmaceutical Analysis and
Training Laboratory (BATL), Northeastern University,
USA. “For PTMs, you might use reverse-phase LC in
tandem with MS, whereas size exclusion chromatography
might be used to detect aggregation in the development of
antibody-based drugs.”
However, although there exists an entire toolbox of
methods required to characterize protein and antibodybased products, method development for cell and gene
therapy is lagging, says Auclair. “I liken it to different
stages of construction of a house: for antibodies, the
house is built, albeit not 100% complete, and you can live
in it. For gene therapies, we have a basic structure, but
for cell therapies, we’ve only just got the building blocks.
Cell-based therapies are about 5–10 years behind where
antibody-based biologics are in terms of analytics.”
WOMEN IN SCIENCE
Advances in
Biopharmaceutical Analysis
Joanna Owens, PhD
27
Credit: iStock
TECHNOLOGYNETWORKS.COM
What’s universal to all biopharmaceuticals, though, is
that whichever characterization method is used, it needs
to be carried through the entire development process
from research to commercialization. “This doesn’t
only include collecting the same data, if you’re going to
monitor PTMs, for example, you should be using as close
to the same instrumentation as possible,” said Auclair.
“And if the quality of the product is paramount to its
efficacy, then continued monitoring is key and we need
to be looking at opportunities to monitor in real-time, so
we can kill products that are below quality before they
advance too far towards commercialization.”
Process innovation for
biopharmaceuticals
While developing and improving methods to monitor
impurities is a major focus of the research at BATL, it
sits alongside a focus on process analytics. Here, method
development goes hand-in-hand with engineering
to determine the pain points of biopharmaceutical
production processes and find solutions, such as new
in-line sensors that can monitor different aspects of the
process. But there are challenges.
“One of the major challenges is integration, especially
where different equipment manufacturers and software
providers are involved. The technologies are there, but
it’s just a matter of implementation and automation,” said
Auclair. “In our lab, we have instrumentation that pulls
samples from the bioreactor so we can test different
quality checks and try to make that process better and
more robust.”
Another headache is data analysis. “The hardware is
always ahead of the data analysis tools, in terms of
capability, speed and ease of use,” says Auclair. “In the
research space, it’s OK to have a really complex process,
but as you move down the commercialization space, you
need technologies and processes that don’t require 30
years’ experience in informatics to run.”
One area that is gaining momentum is the multi-attribute
method (MAM), an MS-based protocol that monitors
several properties, such as PTMs, sequence coverage
and new peak detection, in the same experiment.1
Auclair’s team is looking at how they can optimize MAM
as development processes change, how to integrate it inline with bioreactors and how to apply the approach to
gene and cell therapies.
“The ultimate goal of our work is to innovate these
product development and commercialization processes,
so that more good medicines are discovered and don’t
fail because the quality of the product wasn’t considered
throughout the process.”
Microbial monitoring in
biopharmaceutical development
Another major area of biopharmaceutical analysis is
monitoring for contamination by microbes and viruses.
It is essential to ensure that biopharmaceuticals are not
contaminated with pathogenic or environmental bacteria,
CELL AND GENE THERAPIES: INNOVATIONS, CHALLENGES AND CLINICAL BREAKTHROUGHS 28
“In drug development,
especially academic
drug development,
many people don’t think
about product quality
from the beginning. And
if you don’t think about
that from the beginning
then your product
doesn’t get to market,
it dies in the valley of
death,“ says Auclair.
TECHNOLOGYNETWORKS.COM
fungi or viruses, which, if introduced even in small amounts,
can potentially multiply to high levels in the bulk material
during manufacturing or in the final drug product.
Pathogens can potentially be introduced via cell lines, raw
materials, use of contaminated human or animal products
during manufacture or from the environment – such as
human contact during sampling.2
Many recombinant
protein therapeutics, vaccines and plasma products
are produced using cell culture, which is susceptible to
contamination. Although a rare occurrence, this can reduce
the yield of the drug, affect the quality of the end product
and can have considerable financial costs for industry and
potentially serious safety implications for patients.3
“As the manufacture of biomedicines is a complex, multi-step
process, typically involving potentially non-sterile biological
ingredients, a sound microbial and viral safety strategy is
required,” said Dr. Oleg Krut, head of the microbial safety
section at the Paul-Ehrlich-Institut, Federal Institute for
Vaccines and Biomedicines, Germany. “The use of sensitive
methods to detect bacteria or fungi themselves is an integral
part of this strategy. All ingredients must be tested using
the most sensitive assays to ensure that any microbes and
viruses are detected before use and that contaminated
material will not be used during manufacture.”
Bacteria and fungi are usually detected by growth-based
methods, that is, detection of turbidity in liquid microbial
culture or the enumeration of colonies on agar plates. “The
main methodological challenges are the time required to
obtain a valid result (up to 14 days), and the high sensitivity
required to detect a single microorganism,” explains Krut.
“A further challenge is that introducing more advanced
microbiological techniques that maintain the highest
sensitivity with shorter time-to-result is slow, because of the
necessary validation requirements.”
Viruses have traditionally been detected by inoculating
samples into animals or indicator cell cultures that are
monitored for signs of infection.2,3
However, only a limited
range of viruses can be detected through animal testing.3
“Replacement of animal testing is being considered through
the revision of regulatory guidelines such as ICH Guideline
Q5A(R2) on viral safety evaluation of biotechnology
products derived from cell lines of human or animal origin,
in line with the 3R principles for animal welfare,” said Dr.
Johannes Blümel, head of the viral safety section, at the
Paul-Ehrlich-Institut, Federal Institute for Vaccines and
Biomedicines, Germany. “The use of indicator cells is still
a very valuable method for testing cell cultures for viral
contamination as it has an excellent sensitivity for many
viruses. However, the range of viruses that can be detected is
still limited by the ability of the indicator cells to support viral
replication and to show signs of infection that are easily visible
under a light microscope.” Virus testing using indicator cells
is complemented by nucleic acid amplification technologies
such as polymerase chain reaction (PCR). However, viral
variants or newly emerging viruses may pose a threat.
“Viruses can be specifically detected by very sensitive
methods if their genomes or host cells are known,” said
Blümel. “However, although PCR has excellent sensitivity,
the technology remains limited towards detection of specific
viruses. A battery of several different PCR tests therefore
needs to be designed according to a risk assessment of the
virus species that could contaminate.”
Advances in microbial monitoring
methods
There have been two major trends in microbial safety
in recent years, says Krut. “The first is the automation
of classical cell culture methods, such as automated
liquid culture and automated colony counting. These
improvements shorten the time to results, reduce handson time and operator bias and increase throughput and
data integrity. As they are modifications of classical
methods, they are easier to validate.”
The second trend is the emergence of rapid
microbiological methods (RMMs), such as nucleic
acid-based techniques, adenosine triphosphate
(ATP)-based contamination detection, solid-state
cytometry and MS. “Unlike classical microbial culture,
which takes two weeks, RMMs can detect microbial
contamination within days or even hours,” says Krut.
“Such methods are needed for biomedicines with short
shelf-lives where classical methods are simply too slow.
However, they require extensive validation, which is
often seen as a major barrier to their implementation in
biopharmaceutical production.”
CELL AND GENE THERAPIES: INNOVATIONS, CHALLENGES AND CLINICAL BREAKTHROUGHS 29
TECHNOLOGYNETWORKS.COM
Dr. Krut’s team is evaluating culture-independent
flow or solid-state cytometry for bacterial detection
and enumeration, which it is hoped will allow faster
time to results, more rapid Official Medicines Control
Laboratories (OMCLs) provision of certificates and
earlier governmental batch release of biomedicine
batches to the market.
In viral safety, using next-generation sequencing (NGS)
or high-throughput sequencing (HTS) in a metagenomic
approach (i.e. detecting all viral sequences in a sample)
has the potential to overcome the limitations of current
PCR-based assays.4 Research at the Paul-EhrlichInstitut is focused on optimizing HTS technology for the
sensitive detection of human viruses in blood samples.
“Our multidisciplinary team of medical virologists,
molecular biologists and bioinformaticians is
particularly interested in reducing host cell background
and optimizing library construction in order to increase
the sensitivity of the method, as well as in applying
artificial intelligence and machine learning methods
to advance the bioinformatics pipeline further,” says
Blümel. “Our method allows us to address any type of
emerging agent, using technology that meets regulatory
requirements, and will ensure we keep pace with
emerging viruses and are prepared.”
ABOUT THE INTERVIEWEES
Jared Auclair is the director of bioinnovation and the Vice Provost
Research Economic Development at Northeastern University.
He holds a PhD in Biomedical Sciences from the University of
Massachusetts.
Oleg Krut is head of the microbial safety section at the PaulEhrlich-Institut, Federal Institute for Vaccines and Biomedicines,
Germany.
REFERENCES
1. Millán-Martín S, Jakes C, Carillo S, et al. Multi-attribute method
(MAM): An emerging analytical workflow for biopharmaceutical
characterization, batch release and cGMP purity testing at the
peptide and intact protein level. Crit Rev Anal Chem. 2023;1-18. doi
:10.1080/10408347.2023.2238058
2. Valiant WG, Cai K, Vallone PM. A history of adventitious agent
contamination and the current methods to detect and remove
them from pharmaceutical products. Biologicals. 2022;80:6-17.
doi:10.1016/j.biologicals.2022.10.002
3. Barone PW, Wiebe ME, Leung JC, et al. Viral contamination in
biologic manufacture and implications for emerging therapies.
Nat Biotechnol. 2020;38(5):563-572. doi:10.1038/s41587-020-
0507-2
4. Khan AS, Mallet L, Blümel J, et al. Report of the third conference
on next-generation sequencing for adventitious virus detection
in biologics for humans and animals [published online ahead of
print, 2023 Jul 19]. Biologicals. 2023;83:101696. doi:10.1016/j.
biologicals.2023.101696
CELL AND GENE THERAPIES: INNOVATIONS, CHALLENGES AND CLINICAL BREAKTHROUGHS 30
CRISPR is a powerful technique that can enhance your
research. If you are getting to grips with a CRISPR protocol
for the first time, optimizing and planning your experiment
can make things easier and can be the key to a successful
CRISPR experiment.
Optimize CRISPR delivery in your
chosen cell line
With every new cell line that we use in the lab, we first do an
optimization experiment. We trial a few different transfection
conditions (lipofection, nucleofection, etc.) delivering Cas9
and a control guide RNA that we know consistently work in
different cell types. We routinely use a guide targeting the
AAVS1 locus for human cells and tubulin beta for mouse
cells. Then, we quantify the editing efficiency using PCR,
Sanger sequencing and inference of CRISPR edits (ICE)
analysis and determine the best possible delivery method
for the cell type. Surprisingly, there is a lot of variation in
CRISPR efficiency between delivery methods and cell lines.
You might think that this is too time-consuming, but
ultimately you will be saving time and money. At the
end of a CRISPR optimization experiment, you will
have a protocol specific for your cell line that you can
use to CRISPR different regions of the genome. Most
labs work only with a handful of cell lines, usually
derived from similar tissues making this step a valuable
resource for future work.
Design your experiment - CRISPR
knockout
Once you have established the best CRISPR delivery
for your cell line of interest, you can select your desired
modification. CRISPR knockouts are the easiest
experiments, and they require two components: Cas9
protein and guide RNA.
When I first started doing CRISPR cell lines, we used
to deliver Cas9 and guide RNAs as plasmids. Now, we
WOMEN IN SCIENCE
How To Run a Successful
CRISPR Experiment
Samantha Carrera, PhD
31
Credit: iStock
TECHNOLOGYNETWORKS.COM
know that the most efficient CRISPR method is using
Cas9 proteins and chemically synthesized guide RNAs,
which are preassembled into a ribonucleoprotein
(RNP) and delivered into the cells.1
Even though they
are more expensive to buy, it will be more cost-effective
in the end as you will save time and effort not having to
repeat your experiment.
The most important part to design is the guide RNA; there
are numerous websites where you can input your sequence
of interest, and they will give you a list of guide RNAs that
you can use. The tricky part is deciding which one to choose.
First you must decide which exon of your gene you want to
target. I recommend avoiding the first exon if possible. When
you create insertions/deletions (indels) here, the splicing
machinery can skip the exon and still produce a truncated
version of the protein.2
We usually pick exons that are as
upstream as possible and that are common to all isoforms.
The most common modification introduced by Cas9 is +/- 1
bp so directing a guide to exons with an asymmetric splicing
phase makes it more likely for the transcript to be knocked
out of frame.
With every CRISPR design you should pick guides
that have low off-targeting potential. Even though some
guides are labelled as inefficient, it is difficult to predict
the efficiency of the guide RNA. For this reason, I would
recommend designing three guides and delivering them
in parallel alongside the control guide RNAs (either
AAVS1 or tubulin).
Once you have transfected cells for a gene knockout, you
can isolate genomic DNA and use primers designed around
the cutting area to determine the percentage of indels you
have in your cell population using Sanger sequencing and
ICE/TIDE analysis. Once you have determined this, you
will have to do some functional validations to determine
whether your protein of interest has been knocked down;
this can be done by western blotting.
Design your experiment - CRISPR
knock-in
I believe that CRISPR knock-ins are where you really
realize how powerful this technique is. You can add
functional tags to your gene of interest, including
fluorescent proteins, luminescent reporters, proximity
labelling enzymes, degron tags and pull-down tags. You
can also insert transgenes in safe harbor loci, so-called
because they aren’t easily silenced. These transgenes
are used in technology platforms that allow you to
control the expression of your protein of interest by
adding small molecules. These include degron systems
and tetOn/off compatible cells. You can also introduce
point mutations into your gene of interest or into
enhancer sequences to study gene regulation.
A CRISPR knock-in experiment is harder to plan than
a knockout experiment and decisions need to be made
about the site of insertion and the need for additional
tags and linker sequences. If you are introducing
functional tags you might need to first assess if N- or
C- terminal tagging is better for preserving the function
and expression of your protein. Once you have decided
this, you must design a guide that is close to the site
(within 20 bp) where you want your insertion.
The next step is to design your homology directed
repair (HDR) template, which will contain your insert
of interest flanked by homology arms. The homology
arms are sequences of DNA that correspond to your
gene of interest on either side of your insertion site. It is
worth remembering that the larger the insert, the more
inefficient the HDR process is going to be and the longer
homology arms you will need.
If your insertion is small (<100 bp) the best thing to use is a
single-stranded DNA template with ~80 bp-long homology
arms on each side. These are commercially available and
usually have modifications that make them more likely to
get integrated in the genome. For larger insertions we prefer
using plasmids where we clone the homology arms and
our insert. The homology arms in this case are around 800
bp. It is important to make sure to mutate the protospacer
adjacent motif (PAM) site in the HDR template to avoid
Cas9 cutting it (causing insertions or deletions).
Once you have designed your guides and your template,
you can use your optimized protocol to deliver them into
your cells and create a polyclonal population. At this point,
you will have many cells that have not been modified and
it is worth spending some time considering how to enrich
your pool of cells with your desired modification.
CELL AND GENE THERAPIES: INNOVATIONS, CHALLENGES AND CLINICAL BREAKTHROUGHS 32
TECHNOLOGYNETWORKS.COM
Enrich your modification
The next thing that you need to consider is if a polyclonal
cell line is sufficient for your experiments or if you need
to go through the labour-intensive process of generating
a monoclonal cell line.
If your desired edit contains a sortable marker, like a
fluorophore, you can first enrich your population of cells
by fluorescence-activated cell sorting (FACS). This will
result in a pool of cells that are slightly different from
each other, but which all express your fluorophore. It is
good to bear in mind that the levels of fluorescence will
depend on the expression of your gene of interest, so it
might be hard to distinguish a positive population if the
levels are too low.
Another way of enriching your population is by
introducing an antibiotic selection cassette and treating
the cells with that antibiotic. This will result in the
elimination of the cells without the insert, leaving
behind a positive pool of cells.
Sometimes you don’t want your protein to have
a fluorophore or an antibiotic cassette that could
potentially interfere with its function. In this case, we
co-integrate a removable antibiotic selection cassette
which allows us to select for HDR-positive cells. A
promoter and the desired antibiotic are flanked by loxP
sites that allow their removal once the cells have been
verified by expressing Cre-recombinase (Figure 1). With
the fluorophores you can introduce a cleavable signal
like T2A, in that way your protein of interest and the
CELL AND GENE THERAPIES: INNOVATIONS, CHALLENGES AND CLINICAL BREAKTHROUGHS 33
HOW TO RUN A SUCCESSFUL CRISPR EXPERIMENT 3
ppysites that allow their removal once the cells have been verified by expressing Cre-recombinase (Figure 1).
With the fluorophores you can introduce a cleavable signal like T2A, in that way your protein of interest
and the fluorophore will be cleaved when transduced, making it less likely that the insertion interferes
with the function of the protein of interest.
Gene of interest
gRNA_1
5'H 3'H
gRNA_2
Cas9 HDR template
Gene of interest
5'H with PAM mutations mScarlet-I 3'H
5'H with PAM mutations mScarlet-I
PGK promoter bGH poly(A) signal
PuroR 3'H
Cre recombinase
3'H
IoxP IoxP
T2A
7500
IoxP
7500 10,000
2000 3000
Figure 1: CRISPR knock-in enrichment by removable antibiotic selection. Cas9 is delivered into the cells along with
two guide RNAs (gRNA) and an HDR template. The HDR template includes a Puromycin resistance cassette that
will allow the selection of cells that had integrated the fluorophore (mScarlet-I). Once the cells have been selected,
the antibiotic selection cassette can be removed by the addition of Cre recombinase. Credit: Technology Networks
TECHNOLOGYNETWORKS.COM
fluorophore will be cleaved when transduced making it
less likely that the insertion interferes with the function
of the protein of interest.
If you want to produce monoclonal cell lines you can
use your enriched population and either FACS sort
into single cells or use the serial dilution method. This
method consists in diluting your cells until you have one
cell for each 100 µl and seed this volume into a single
well on a 96-well plate. It is important to note that not
all cell lines are happy growing as single cells and you
will struggle to create monoclonal populations out of
these cell lines. In my experience with these cells, it is
worth seeding the single cells into conditional media –
this is media taken from a growing population of cells
that has been filtered to avoid carrying any cells but
with the advantage of carrying any secreted proteins.
Finally, you need to verify that your cells have the
desired modification. For this, there are a few things
you must consider. First, designing your primers
outside of the homology arms to discard the possibility
that your template has integrated in another area of
the genome. You also must remember that your PCR
will preferentially amplify any unmodified alleles, as it
requires less time to copy a smaller DNA product than a
bigger one. I usually design a handful of primers including
internal ones and insert-specific ones that I can use in
different combinations. This will give a good indication
that the cells have the desired insert in them. I will always
send these PCR products for sequencing to check that
there are no insertions, deletions or mutations that will
interfere with the expression of my gene. You will also
need to do some functional validations, for example using
western blotting to see if your protein has a size shift that
corresponds to your insert.
Conclusion
Creating CRISPR cell lines doesn’t have to be difficult
process. If you take advantage of the tips above you can
create all the tools you need to advance your research
opening different possibilities for studying your
prefered cellular pathway. With all these things in mind,
enjoy CRISPRing!
REFERENCES
1. Kim S, Kim D, Cho SW, Kim J, Kim JS. Highly efficient RNAguided genome editing in human cells via delivery of purified
Cas9 ribonucleoproteins. Genome Res. 2014;24(6):1012-1019. doi:
10.1101/gr.171322.113
2. Sharpe JJ, Cooper TA. Unexpected consequences: exon
skipping caused by CRISPR-generated mutations. Genome Biol.
2017;18(1):109. doi:10.1186/s13059-017-1240-0
CELL AND GENE THERAPIES: INNOVATIONS, CHALLENGES AND CLINICAL BREAKTHROUGHS 34
Like many young girls in elementary school, Victoria
Gray wanted to be a cheerleader, until she was told by
her doctor that this simply wouldn’t be possible; the
exertion placed on her body by the training regimen
could have devastating consequences. “That was my
first disappointment as a kid,” she recalls.
Sadly, it wouldn’t be her last.
Tall for her age, Victoria later thought about trying
out for a spot on the school basketball team. Her
uncle had a great love for the game, which inspired
her. Plus, everyone around her said she would
probably have a natural knack for shooting hoops.
Why not give it a go? She thought. But once again,
her enthusiasm was shot down by another firm “no”
from her pediatrician.
WOMEN IN SCIENCE
Breaking the Chains: How
CRISPR Gene Therapy Gave
Victoria Gray a New Life
Molly Coddington
35
“My life was constantly
full of ‘noes’, and people
telling me ‘you can’t do
this’ – it was limitation
after limitation,” she says.
“I had to alter everything
I dreamed about, every
step of the way from
childhood to adulthood.”
TECHNOLOGYNETWORKS.COM
If you’re lucky enough to grow up having a healthy
childhood, it’s hard to imagine the cruel realities of
experiencing a sick one. Victoria describes the motions
of her life as though it was a distressing movie being
played out in front of her, and someone – somewhere
– kept hitting the pause button. Her days were shaped
not by the whimsical imagination of a young child, but
by her illness. That’s because at just three months of age
she had been diagnosed with sickle cell disease (SCD).
SCD is a group of inherited blood disorders that cause
red blood cells to become hard and sticky. In healthy
individuals, red blood cells are a disc shape, allowing
them to flow easily through our blood vessels. In SCD,
they form a “C” shape known as a sickle, which causes
the blood cells to die quickly or cause blockages in blood
vessels. According to the National Institutes of Health,
over 20 million people worldwide are affected by SCD.
SCD stole Victoria’s childhood, including her right to
an education. “I had to alter everything I dreamed about
every step of the way from childhood to adulthood.
When I started college, I wanted to be a cardiologist.
But doctors explained that the stress involved with
studying medicine wouldn’t be good for me. So, I put
myself on pause – once again,” she says.
Undeterred and eager to help care for others, Victoria
started to explore a nursing degree, until “I had one the
worst pain crises of my life,” she says.
Crises refer to acute conditions, such as the blockage
of a blood vessel by sickled cells, caused by SCD. They
are the main clinical hallmark of the disease, causing
severe, debilitating pain and extreme fatigue, among
other symptoms. Oftentimes, they strike at random, and
can persist for long periods of time requiring extended
stays in hospital.
“That crisis put me in the hospital for three months. I
lost the ability to use my arms and my legs,” Victoria
describes. Perhaps most devastatingly, she says, “I also
lost my ability to dream.”
After undergoing comprehensive rehabilitation,
Victoria eventually regained her strength and her
ability to walk. But despite the physical improvements,
mentally, she had to resign herself to the fact that a
career maybe wasn’t on the cards for her. It was yet
another blow, but one that she handled with the grace
and determination she carried from a young age to find
fulfillment in life. Victoria chose to focus her time, and
the energy she could muster between crises, on being
a wonderful mom to her four children, and a strong
partner to her husband, Earl.
Then, in 2018, an opportunity came around that finally
awarded Victoria the opportunity to say “yes” for the
first time in her life. It was, she would come to learn, an
opportunity that marked not only a major milestone in
the history of medicine, but one that freed her from the
chains of SCD – most likely, forever.
Finally, a “yes” for Victoria
Patients with SCD are often prescribed drugs, such as
painkillers, to try and curb the symptoms associated
with crises. These medications fail to target or cure the
underlying cause of the disease, and while they offer
symptomatic relief, they can have harmful side effects.
There are few authorized therapeutics on the market
for SCD patients. In 2018, Victoria, who lives in Forest,
Mississippi, was under the care of Dr. Haydar Frangoul,
waiting to learn of her eligibility for one such treatment
– a haploidentical “haplo” stem cell transplant using
cells donated from her brother.
Dr. Frangroul is the director of the Pediatric Stem Cell
Transplant program at Tristar Centennial Children’s
Hospital, and an investigator at the Sarah Cannon
Research Institute in Nashville. While at his clinic in
CELL AND GENE THERAPIES: INNOVATIONS, CHALLENGES AND CLINICAL BREAKTHROUGHS 36
“I really gave up on
becoming anything else.
I thought sickle cell was
just going to be my life
from beginning to end.”
TECHNOLOGYNETWORKS.COM
Nashville, Victoria experienced a crisis that would ultimately
change her life. “I had to be admitted, and during that hospital
stay Dr. Frangoul approached me at my bedside. He knew
that I was feeling really down, and he offered me a second
option besides the Haplo transplant,” she recalls.
Victoria had some reservations about the Haplo transplant,
largely due to the risk of graft versus host disease, a
complication that can occur when a donor’s cells attack the
recipient’s: “Dr. Frangoul told me that they were starting a
new trial soon using CRISPR gene therapy. I hadn’t heard
about it, so he showed me a small video on his phone and
sent me a link so I could review it later.”
The clinical trial that Victoria participated in was testing a
cell-based gene therapy known as CasgevyTM. The therapy
utilizes CRISPR-Cas9 gene-editing technology, which is
directed to cut DNA in specific locations and enable the
removal, addition or replacement of DNA. During treatment,
the SCD patient’s blood stem cells are extracted from their
body and edited in a laboratory. CRISPR-Cas9 is used to
create an edit in the BCL11A gene within the patient’s cells,
which triggers the production of fetal hemoglobin. Once
these cells are re-inserted into the patient, they settle back
into the bone marrow and the increased fetal hemoglobin
facilitates the delivery of oxygen around the body.
As with any clinical trial, there were risks involved. Given
that Victoria would be the first patient to ever receive this
experimental therapy, these risks felt somewhat heightened.
She had a difficult decision to make.
“I'm a woman of faith – I pray a lot. I went to God, in private,
about graft versus host disease, because I really didn’t want
to experience that. So, when gene therapy came along, I felt
like it was my answer from God, as though he was saying to
me ‘I remove your fears now. This opportunity is for you’,”
Victoria says.
Ultimately, the possibility of a life that would not be plagued
by pain outweighed any doubts she might have had about
being patient one. Within 24 hours of speaking to Dr.
Frangoul, Victoria took the courageous decision to volunteer
for the trial.
In July 2019, she became the first patient to receive CRISPR
gene therapy for SCD.
“The trial was a different experience to anything I’ve had before,
because for the first time, I felt hopeful. I was fighting for my life,
and for my family.” Sure, I had to travel back to Nashville a lot
for testing, and there were times [during the trial], especially
after the chemotherapy, that it was hard. But my dad was with
me, and he kept reminding me, ‘Vicky, you’ve seen worse, you
are strong enough to get through this’. It helped to lift me back
up and remind me of why I was doing this.
Am I dead?
Eight months after the therapy was administered, Victoria
woke one morning and felt “different”, though she couldn’t
quite place what that meant. “I remember waking up, and
the room was really bright. I didn’t feel anything, which was
strange. I didn’t have any shortness of breath when I stood up,
as had been the case most mornings,” she says.
CELL AND GENE THERAPIES: INNOVATIONS, CHALLENGES AND CLINICAL BREAKTHROUGHS 37
“My“Dr. Frangoul
explained CRISPR
therapy to me like this:
he said, ‘just imagine a
textbook with thousands
and thousands of words,
and there are a few words
in there that are incorrect.
The CRISPR technology
could go into the cells,
find the incorrect word
and edit it without
changing the story’.”
TECHNOLOGYNETWORKS.COM
Having lived most of her life in excruciating pain
and extreme fatigue, the experience of waking up in
its absence was so downright bizarre, Victoria had
convinced herself that she must be dead. Pinching the
skin on her face, and her thighs, she was reassured to
feel the sharp, physical sensations.
“I shouted to my kids, ‘Hey y’all, come in here!’, and as they
entered the room, their faces lit up. I knew in that moment,
they could see me, and I realized that I was very much
alive. I cried tears of joy, because I knew then that the gene
therapy had worked,” Victoria describes, visibly emotional.
That bright, beautiful morning was four years ago, and it
marked Victoria’s new beginning.
She describes her life now as one of freedom. She is free
from constant pain and exhaustion. Free from having to
stare at a hospital room’s four walls while experiencing a
crisis episode. Free from relying on medication just to make
it through the day. Free from all the ways that SCD stole her
autonomy and chained her to a life burdened by illness.
Victoria can now play with her children and embody the
parent she always dreamed of being. She can immerse
herself in typical mom activities, often taken for granted as
mundane, but that were once out of her reach. She can make
choices. She can travel abroad – even flying to London last
year to speak at the Third International Summit on Human
Genome Editing.
Eventually, Victoria’s health improved to the extent
that she could realize her ambition of working full-time,
securing a position as a cashier at her local Walmart.
It was during a particularly busy shift on December 8,
2023, when she received news that the US Food and
Drug Administration (FDA) had decided to approve
Casgevy for the treatment of SCD.
Casgevy’s approval was based on data submitted from
the trial that Victoria herself had bravely participated in.
The trial’s primary outcome had been freedom from severe
vaso-occlusive crises for at least 12 months during the
study’s 24-month follow-up period. In total, 44 patients
were dosed with Casgevy. Upon submitting the trial data,
31 patients had sufficient follow-up time to be evaluated. Of
these, 29 patients reached the primary efficacy outcome,
with 0 patients suffering from graft failure or graft rejection –
the trial had proven a success. Casgevy became the first gene
therapy utilizing CRISPR-Cas9 to receive FDA approval,
marking a historic moment for the SCD community, as
lovotibeglogene autotemcel (LyfgeniaTM) – another cellbased gene therapy for SCD, developed by Bluebird Bio
Inc. – also received a green light from the agency.
SCD patients face medical and
racial discrimination, hindering
clinical research prospects
Victoria received many phone calls that day, including one
from a nurse who treated her at Dr. Frangoul’s clinic in
Nashville. “We both cried,” she says, “It was tears of pure
CELL AND GENE THERAPIES: INNOVATIONS, CHALLENGES AND CLINICAL BREAKTHROUGHS 38
“When I saw the text
[from my husband], that
the FDA had approved
Casgevy, I had to rush
off the floor at Walmart
because I felt the tears
coming,” she says. “I
got in my car, and I just
cried. I was so, so happy
because I knew what a
relief this would be for
other sickle cell patients
that were in a dark place
like I had once been.
Now, there was hope.”
TECHNOLOGYNETWORKS.COM
joy, because all of the pain, disappointment and judgement
that I had faced from childhood now felt worth it. This was
going to bring about change for other people who feel alone
and feel overlooked.”
“It had felt as though nobody was coming to save us. Then
suddenly we had a novel therapy for the SCD community. I
felt so happy and grateful,” she adds.
While reminiscing, Victoria takes a moment to pause. It’s
clear that the emotions attached to these memories are a
complex mixture of elation and pain. “I want to emphasize
that, had it not been for the positive way in which I was
treated by Dr. Frangoul and his team, I might not have
accepted the opportunity to take part in this trial that has
saved my life,” she says.
Discrimination is an issue encountered by many SCD
patients across the globe during their lifetime. In a
perspective piece published in 2020, Power-Hays and
McGann expressed that there may be “no population of
patients whose health care and outcomes are more affected
by racism than those with SCD.”
During the 1970s, many African American people were
deprived of jobs, educational opportunities, marriage
licenses and insurance in the US if they carried the SCD
trait. This grim picture was mirrored across the pond in
the UK. “In the 20th century, I'd say that racism showed
itself in the lack of willingness on the part of statutory
services, such as education, social services and housing,
to take account of the needs of those living with SCD,”
says Professor Simon Dyson, director of the Unit for
the Social Study of Thalassaemia and Sickle Cell at De
Montfort University in the UK. “In terms of sickle-cell
screening, SCD had to meet prevalence thresholds not
demanded of other rarer conditions, before newborn
screening to save the lives of black infants was
eventually made universal in England in 2004.”
A 2018 systematic review by Dr. Dominique Bulgin and
colleagues explored the extent of health-related stigma
in adolescents and adults living with SCD, analyzing
data from 27 studies published between 2004–2017.
They found that people with SCD experience healthrelated stigma based not only on their race, but on
their disease status, socioeconomic status, delayed
growth and puberty and having chronic and acute pain,
requiring opioid treatment.
“Individuals with SCD reported being stigmatized as drug
seeking or drug addicts and having their experiences of
pain discredited by healthcare providers,” the review states.
Sadly, this experience is one that resonates with Victoria.
“I was once in the emergency room when a nurse said to
me, ‘You know, I feel so sorry for you sicklers’ – this was
a term he used – ‘because you guys just get addicted to
these pain meds, and then you can’t tell the difference
between withdrawal and a real crisis’,” she says.
“He then said that it’s just inevitable that all SCD
patients become drug addicts. I couldn’t believe it. I was
in crisis. I was in pain, and I was crying out to a person
who was supposed to help me. Instead, he judged me,”
she continues. This, sadly, was not an isolated incident.
Victoria recounts another crisis episode, where she
felt as though her symptoms of pain were starting to
improve. She asked the nurse treating her to avoid
administering any excessive pain medication that might
make her feel drowsy, as she wanted to sit up and try to
move around the room. Instead, the nurse pressed the
button on her medication dispenser, and Victoria fell
asleep. When she woke, she learned that instead of pain
medication, the nurse had given her a different type of
drug because they “wanted to see what it would do”.
Unfortunately, there was little consequence for
the nurse, but the ramifications for Victoria were
heartbreaking, she explains, and led her to question her
worth as a SCD patient: “I felt as though the nurse had
been given the right to experiment on me without my
consent. What if that drug had taken my life? I couldn’t
trust the staff that was treating me, I felt as though I was
a burden, like they were trying to get rid of me when I
was coming to them for help.”
“There is no way,” Victoria adds, “that I would have
accepted an experimental treatment like gene therapy,
if I had been offered it at this facility.” Research shows
that she is not alone in feeling this way.
In 2020, Cho et al. conducted a study examining
CELL AND GENE THERAPIES: INNOVATIONS, CHALLENGES AND CLINICAL BREAKTHROUGHS 39
TECHNOLOGYNETWORKS.COM
the motivations and decision-making processes of
enrollees and decliners of high-risk trials for SCD. Of
the 26 SCD patients interviewed, the majority reported
negative interactions with health care providers. These
experiences were so bad that many individuals had
chosen to avoid hospitals during significant pain crises.
If SCD patients are afraid to even ask for help when
they are in pain, how can they be expected to partake
in high-risk clinical research? It’s an almost impossible
decision, but it’s one that Victoria chose to make for
herself, for her family and for the SCD community. It’s
a decision that, now, in hindsight, she is thrilled about,
but she remains eager to warn the medical community
of the risks they pose to the health of SCD patients –
and the future of SCD research – by forgetting that the
patient in front of them is a human being.
Patient advocacy and increasing
access to gene therapies
Now, five years after the trial commenced, Victoria is
undertaking her follow-up appointments – which last 15
years from the study enrollment date – as per the study
protocol. These appointments monitor her health and
assess the long-term efficacy and safety of Casgevy. As
gene therapies are emerging drugs, whether or not they
are effective for the duration of a patient’s life is yet to
be determined. “I pray that it is a forever change, and I
believe that it is,” Victoria says.
She is enjoying applying her newfound energy to spread
the word about her experience as an SCD patient, and a
CRISPR gene therapy recipient, through advocacy work.
Once the news broke that she was the first patient to
receive CRISPR gene therapy for SCD, Victoria was
invited to speak with SCD organizations, meet other
patients – or “warriors”, as she refers to them – and
attend international events to share her story. “When I
was flown to London to speak at the summit on human
genome editing, I was amazed, because people really
cared about my experience,” she says.
A recent highlight, she says, was her appearance on
Good Morning America, where Victoria was able to
meet a fellow SCD patient – Jamie – who had been
inspired by her story and made the decision to receive
CRISPR gene therapy. “It was truly a fulfilling moment,”
she recalls, “because it was one thing that I hoped for
– to be able to affect someone else's life in a positive
way. To see [Jamie] looking so healthy after not being
able to leave the house, or struggling to take care of his
children, it was just incredible.”
Victoria also had the opportunity to visit the production
facility that created her CRISPR gene therapy. “I was
like a kid, looking at the machines and listening to how
everything works. I know that this [therapy] was many
years in the making, and it was an honor to meet the
scientists that were working so hard when I thought no
one cared.”
Her advocacy work has most recently turned to the
costs associated with CRISPR-based, and other gene
therapies, for SCD. A major challenge for patients and
clinicians in accessing such emerging therapies will
CELL AND GENE THERAPIES: INNOVATIONS, CHALLENGES AND CLINICAL BREAKTHROUGHS 40
“Please, just treat us how
you want to be treated,”
Victoria asks. “We did
not choose to have SCD.
We did not choose the
amount of medication
we require to ease our
pain. We just want to be
cared for, and we want to
feel normal. Please, treat
use with respect.”
TECHNOLOGYNETWORKS.COM
undoubtedly be their price. While information on the
exact cost is currently limited, Casgevy will reportedly
be priced at $2.2 million per patient in the US, and over
£1.5 million per patient in the UK. Granted, it’s designed
to be a “one-and-done” treatment, which could override
economic burden of a lifetime of prescriptions, hospital
stays and other associated costs with SCD management.
But very few people – especially marginalized patients
– have $2.2 million at their disposal, and in the US
particularly, there’s lingering uncertainty as to whether
most insurance companies will cover the therapies.
“When I heard the price after the approval, it made the
moment bittersweet,” Victoria says. “I knew that, for me,
if I would have had to pay for it, I wouldn't have been
able to afford it. Because I couldn't even work. Speaking
to SCD patients through my work, it’s clear that other
patients and their families feel the same way.”
There is major research work going on across the globe
in an attempt to reduce the costs associated with gene
therapies, including those that are CRISPR-based.
One example is a proposed movement towards in vivo
delivery of gene therapies, which could reduce the
costs associated with extracting cells, editing them in a
laboratory and then infusing them back into the patient.
At present, this research is in the laboratory stage, rather
than clinical testing.
The Innovative Genomics Institute (IGI) is a non-profit
academic institution that was founded by Professor
Jennifer Doudna, who became a Nobel Laureate in 2020
in recognition of her research discovering CRISPR
gene editing technology. It’s a joint effort between
leading research institutes in the Bay Area, including
the University of California (UC) Berkeley and UC
San Francisco, with affiliates at UC Davis, Lawrence
Berkeley National Laboratory, Lawrence Livermore
National Laboratory, Gladstone Institutes and and
other institutions.
Dr. Melinda Kliegman, director of the Public Impact at
the IGI, recently led the IGI’s Affordability Task Force
in generating a report, titled “Making Genetic Therapies
Affordable and Accessible”. The task force assembled in
2021 to start work on the report, which explores the key
drivers of the high prices associated with gene therapies
and identifies approaches to increase their accessibility.
“The IGI develops the underlying technology used to
make these [gene] therapies, but we are not involved
in commercialization and pricing. We wanted to
understand more about what goes into setting these
prices, and what, if anything, we could do to lower
them,” says Kliegman.
“The process of assembling the task force and
developing the report took over a year and involved
35 task force members. The breadth of expertise of
task force members helped us cover the many different
complex issues that lead to high prices of gene therapies.
It was a huge effort, but interesting, since we were all
learning new things,” Kliegman adds.
The report is 75 pages long and provides a comprehensive
overview of the various factors contributing to the
current pricing of gene therapy. Kliegman summarizes
the key “take home points” of the task force’s findings:
“The issues affecting affordability are multifaceted and
system wide. I would like to acknowledge that these
therapies are expensive and difficult to manufacture,
and there are small patient populations from which to
recover costs,” she says.
“Companies also need to make a profit and adequate
returns for investors. Given this, a for-profit company
may not be the best business model for developing
bespoke gene therapies. We need a paradigm shift in the
way these therapies are commercialized, for example
utilizing non-profit medical research organizations and
public benefit corporations running on moderate-cost
capital from social impact investors and government and
philanthropic grants,” Kliegman continues.
As part of the report, the IGI team built a model that
shows it could be possible to commercialize a gene
therapy for 10x less than they are currently marketed
at today, roughly ~$250,000 per patient for a therapy
that could treat 2000 patients per year. This model is
a departure away from the traditional methods used to
develop gene therapies, and so, responses to the report
have been mixed, Kliegman says. “Our proposal is
oriented towards access, not profit maximization. Many
people think it's naive to expect anyone to ‘leave money
CELL AND GENE THERAPIES: INNOVATIONS, CHALLENGES AND CLINICAL BREAKTHROUGHS 41
TECHNOLOGYNETWORKS.COM
on the table’, while others agree with the need to improve
access and affordability.”
There is change on the horizon, though: “We have had
the privilege of meeting with organizations aligned with
the suggestions in the report. Unsurprisingly, some of
them had representatives on our task force,” Kliegman
emphasizes. “The 90-10 Institute recently launched,
which is a nonprofit working to establish an impact
investment fund for public benefit pharmaceutical
companies. There are also organizations like Odylia
Therapeutics and Caring Cross, which are nonprofits
developing and delivering gene therapies and public
benefit manufacturing organizations like Landmark Bio
and Vector BioMed.”
In Victoria’s mind, the high price of gene therapy is “just
another hurdle to overcome”. She remains optimistic
that the industry will reach a collective decision on how
to ensure all patients can access the therapies they so
desperately need. One day, she hopes that there will be
no SCD patients having to attend emergency rooms,
receive transfusions or rely on pain medication.
“I can live with the title of being a sickle cell warrior – I
think we all can. But I want everyone in this community
to be free from the hold that the disease places on
our lives.” That is her dream, she says, and unlike the
dreams that SCD denied her in childhood, hopefully,
science can make this one come true.
ABOUT THE INTERVIEWEES
Victoria Gray was the first sickle cell anemia patient in the world
to be treated with CRISPR gene editing in 2019. After a lifetime
of pain, treatments and hospitalizations for sickle cell disease,
she is now symptom-free and working as a patient advocate and
international speaker to spread the word about CRISPR and rare
disease to clinicians, scientists, patients and students.
Melinda Kliegman is director of public impact at the Innovative
Genomics Institute (IGI). In this role, she leads the Public
Impact team, which works to align IGI’s genome-engineering
innovations with societal values by engaging in public dialogue,
original research, and policy creation through outreach to key
stakeholders to ensure that genome-editing technology benefits
everyone equitably. Melinda holds a PhD in Biology from Stanford
University. Before joining the IGI, she worked at the Bill & Melinda
Gates Foundation, the world’s largest philanthropic organization.
CELL AND GENE THERAPIES: INNOVATIONS, CHALLENGES AND CLINICAL BREAKTHROUGHS 42
TECHNOLOGYNETWORKS.COM
CELL AND GENE THERAPIES: INNOVATIONS, CHALLENGES AND CLINICAL BREAKTHROUGHS 43
CONTRIBUTORS
Andy Tay, PhD
Andy Tay received his PhD from the University
of California, Los Angeles, focusing on
neuromodulation and engineering. He
subsequently completed his postdoctoral training
at Stanford University where he developed
nanotechnologies for immuno-engineering. Andy
Tay is currently a Presidential Young Professor at
the National University of Singapore.
Aron Gyorgypal, PhD
Aron Gyorgypal is a postdoctoral research fellow
at Harvard Medical School and Massachusetts
General Hospital in Boston, Massachusetts.
Traditionally trained as an engineer, Aron
received his PhD in chemical and biochemical
engineering from Rutgers University in New
Jersey, where he studied antibody glycosylation
in the context of bioprocessing to produce more
homogeneous biologics.
Kate Robinson
Kate Robinson is a science editor for
Technology Networks. She joined the team in
2021 after obtaining a bachelor's degree in
biomedical sciences
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.
Laura Lansdowne
Laura Lansdowne is the managing editor at
Technology Networks, she holds a first-class
honors degree in biology. Before her move
into scientific publishing, Laura worked at the
Wellcome Sanger Institute and GW Pharma.
Masha Savelieff, PhD
Masha Savelieff is a professional freelance
scientific writer. She began her science career
in chemistry at the American University of Beirut
in Lebanon, where she earned her BSc and
MSc degrees. She then went on to receive her
PhD, also in chemistry, from the University of
Illinois in 2008.
TECHNOLOGYNETWORKS.COM
CELL AND GENE THERAPIES: INNOVATIONS, CHALLENGES AND CLINICAL BREAKTHROUGHS 44
CONTRIBUTORS
Molly Coddington
Molly Coddington is a Senior Writer and
Newsroom Team Lead for Technology Networks.
She holds a first-class honors degree in
neuroscience. In 2021 Molly was shortlisted for the
Women in Journalism Georgina Henry Award.
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 and extensive experience as an editor
for Springer Nature journals and research
communications manager at the University of
Edinburgh.
Samantha Carrera, PhD
Samantha Carrera is an experimental officer
in the Genome Editing Unit at the University
of Manchester. Originally from Mexico, she
completed her PhD in Biochemistry at the
University of Leicester in the UK where she studied
the effect of the microenvironment on the
response to apoptosis.
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