The Scientific Observer Issue 36
Magazine
Published: June 28, 2024
Credit: Technology Networks
Cell and gene therapies are poised to change the landscape of medicine forever.
In this issue of The Scientific Observer, we hear from Victoria Gray, the first person to receive a CRISPR cell-based gene therapy for sickle cell disease (SCD). She shares her experiences of the life-changing therapy and talks about the struggles faced by SCD patients and the broader implications of gene therapy accessibility.
We also review the cell therapy landscape and learn more about the innovations in cell and gene therapy delivery that open new avenues for treating genetic disorders.
Also in issue 36:
- How To Enter a New Chapter in Academic Publishing
- Cell Therapy Targets, Clinical Applications, Manufacturing and Regulatory Considerations
- How Is CRISPR Gene Editing Being Used in Infectious Disease Research?
Electronic Pulses Could Reduce
the Need for High Doses in
Gene Therapy Delivery
How Is CRISPR Gene Editing
Being Used in Infectious
Disease Research?
ISSUE 36, JUNE 2024
HOW CRISPR GENE
THERAPY GAVE
A NEW LIFE
BREAKING
VICTORIA
GRAY
CHAINS: the
Sponsored by
2
CONTENT
FROM THE NEWSROOM 05
ARTICLE
Electronic Pulses Could Reduce
the Need for High Doses in Gene
Therapy Delivery 07
Kate Robinson
ARTICLE
Cell Therapy Targets, Clinical
Applications, Manufacturing and
Regulatory Considerations 11
Laura Lansdowne
FEATURE ARTICLE
Breaking The Chains: How
CRISPR Gene Therapy Gave
Victoria Gray A New Life 16
Molly Campbell
ARTICLE
How Is CRISPR Gene Editing
Being Used in Infectious Disease
Research? 26
Blake Forman
ARTICLE
How To Enter a New Chapter in
Academic Publishing 30
Molly Campbell
07 30
14
FEATURE
Breaking the
Chains: How
CRISPR Gene
Therapy Gave
Victoria Gray a
New Life
Molly Campbell
Victoria Gray modified,
Dear Readers,
Welcome to the 36th issue of The Scientific Observer. This
issue, we’re focusing on a field poised to change the
landscape of medicine forever: cell and gene therapy.
Our feature article tells the compelling story of
Victoria Gray, the first patient to receive a CRISPR
cell-based gene therapy for sickle cell disease (SCD).
Throughout her early years, Victoria’s dreams were
crushed by frequent, excruciatingly painful crises
caused by her disease. After years spent in and out of
hospital, she bravely chose to partake in a clinical trial
that completely changed her life. Through Victoria's
eyes, we learn about the daily struggles faced by SCD
patients, the historical mistreatment of minority patients in medical settings and the broader implications
of gene therapy accessibility.
Delivery is one aspect of cell and gene therapy that contributes to their high cost. In this issue, Kate Robinson
speaks with the research team behind a new technique
that utilizes electricity to enhance the body’s receptivity to gene therapy. This innovative approach has the
potential to improve the efficiency and effectiveness of
gene delivery, opening new avenues for the treatment
of a variety of genetic disorders.
Continuing on the theme of innovation in gene editing,
Blake Forman provides a summary of recent research
utilizing CRISPR technology in infectious disease
research. This summary captures how CRISPR-Cas
technology is being harnessed to combat a range of
infectious diseases, providing new hope for rapid and
precise treatment options.
We also delve into the intricacies of cell therapy with
an article focused on its targets, clinical applications, manufacturing and regulatory considerations.
Laura Lansdowne’s article provides a comprehensive
overview of the current landscape, addressing both
the promise and the challenges faced by this rapidly
evolving field.
Finally, we revisit the innovative world of academic
publishing through an interview with Alex Freeman,
the founder of Octopus. This novel publishing platform,
launched in 2022 with the support of UK Research
and Innovation (UKRI), represents a radical approach
to scholarly publication. In our follow-up interview,
Alex discusses the progress made since the platform's
launch and the ongoing challenges of establishing a
new paradigm in academic publishing.
We hope you enjoy our exploration into these pertinent topics – and many more – in issue 36 of The
Scientific Observer.
The Technology Networks Editorial Team
3
Kate Robinson
Kate is an Assistant Editor for
Technology Networks.
EDITORS’ NOTE
CONTRIBUTORS
Laura Elizabeth Lansdowne
Laura is the Managing Editor for
Technology Networks.
Molly Campbell
Molly is a Senior Science Writer
for Technology Networks.
Blake Forman
Blake is a Senior Science Writer
for Technology Networks.
4
iStock
5
Want to learn more?
Check out theTechnology Networks newsroom.
iStock, freestocks/ Umsplash,
Scientists from the Allen Institute applied BARseq to
interrogate gene expression patterns over four million cortical
neurons across nine mouse forebrain hemispheres, at cellular
resolution. They found that the transcriptomic signature of cortical neurons is highly predictive of their cortical area identity.
JOURNAL: Nature
BARSeq Reveals the Brain Is
Like a Pointillism Painting
MOLLY CAMPBELL
Researchers have created the first panoramic view of infection
pathways in the human placenta using ex vivo explant models,
or “mini placentas” from human samples. This placenta map
could highlight potential drug targets to develop pregnancysafe therapies for diseases that can cause severe pregnancy
complications.
JOURNAL: Cell Systems
Placenta Map Reveals Source
of Infection-Related Pregnancy
Complications
BLAKE FORMAN
The first participant in a new psychedelic study has received a
dose of a synthetic formulation of 5-MeO-DMT. The study is set
to evaluate its neurophysiological effects on the human brain
and perceived “mystical experiences.”
First Patient Dosed in Study
To Unravel “Mystical Experiences”
of Psychedelic Derived From
Toad Skin
SARAH WHELAN
5 FROM THE NEWSROOM
From the Newsroom
6
From the Newsroom
FROM THE NEWSROOM
Want to learn more?
Check out theTechnology Networks newsroom.
Brita Seifert/ Pixabay, iStock, leezathomas099/ Pixabay
New analysis of two locks of hair belonging to Ludwig van
Beethoven proves that the composer did have high levels of lead
in his system when he died – but not high enough to be considered the sole cause of the great man’s death. The discovery rules
out one of the most popular theories explaining the composer’s
prolonged illness and eventual early death.
JOURNAL: Clinical Chemistry
Beethoven’s Hair Confirms He
Had Lead Poisoning – But It
Didn’t Kill Him
ALEXANDER BEADLE
After reassessing 48 previously published papers on the health
effects of plant-based diets, researchers from the University
of Bologna concluded that such lifestyles reduce the risk of
cardiovascular diseases and certain cancers.
JOURNAL: PLOS One
Yes, Plant-Based Diets Really
Are Better for Your Health,
Review Finds
LEO BEAR-MCGUINNESS
With interest in women's sports at an all-time high, researchers
from UCL and the University of Bath are investigating the
menstrual cycle's potential impact on injury risk among elite
footballers in the Women's Super League. The study monitored
injury risk in female footballers at different points of their
menstrual cycle.
JOURNAL: Medicine & Science in Sports & Exercise
Female Athletes Six Times More
Likely To Get Injured in the Days
Leading Up to Their Period
RHIANNA-LILY SMITH
7
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).
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 inElectronic Pulses Could
Reduce the Need for High
Doses in Gene Therapy
Delivery
KATE ROBINSON
iStock
8
activate 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 associated with 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 (A AV8) 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
iStock
9
iStock
problems in gene therapy: delivery,
delivery, delivery.”
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. ⚫
Direct delivery is a pathway to
reduce the doses required and
bypass the immune counterresponse, thereby reducing cost
and increasing safety.
10
iStock
From selecting instrumentation, building a team, managing day to day operations and helping
to shape the future of the organization you work for, running a lab presents many challenges.
Leadership experts will provide practical tips and guidance for current and aspiring lab
managers working in industry and academia.
REGISTER NOW
Brought to you by the publication
August 14–15, 2024
8AM PDT | 11AM EDT | 4PM BST
Sean Tucker
Director of Laboratory Services
North Kansas City Hospital
Antoni Lacinai
Workplace Communication Expert
Lacinai Communication
& Performance Development
Kabrena Rodda, PhD
Research Line Manager
Pacific Northwest National Laboratory
Rigoberto Advincula, PhD
Governor’s Chair and Professor
Oak Ridge National Laboratory
Oluwatoyin Asojo, PhD
Associate Director for Strategic Initiatives
Dartmouth Cancer Center
Bamidele Farinre
Chartered Biomedical Scientist
Institute of Biomedical Science
SPEAKERS
MORE SPEAKERS TO BE ANNOUNCED!
INTERESTED IN SPONSORING EVENTS LIKE THIS?
CONTACT OUR FRIENDLY SALES TEAM HERE
Modern Lab
Leader 2024
ONLINE SYMPOSIUM
11
iStock
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,
Cell Therapy Targets, Clinical
Applications, Manufacturing and
Regulatory Considerations
LAURA LANSDOWNE
From selecting instrumentation, building a team, managing day to day operations and helping
to shape the future of the organization you work for, running a lab presents many challenges.
Leadership experts will provide practical tips and guidance for current and aspiring lab
managers working in industry and academia.
REGISTER NOW
Brought to you by the publication
August 14–15, 2024
8AM PDT | 11AM EDT | 4PM BST
Sean Tucker
Director of Laboratory Services
North Kansas City Hospital
Antoni Lacinai
Workplace Communication Expert
Lacinai Communication
& Performance Development
Kabrena Rodda, PhD
Research Line Manager
Pacific Northwest National Laboratory
Rigoberto Advincula, PhD
Governor’s Chair and Professor
Oak Ridge National Laboratory
Oluwatoyin Asojo, PhD
Associate Director for Strategic Initiatives
Dartmouth Cancer Center
Bamidele Farinre
Chartered Biomedical Scientist
Institute of Biomedical Science
SPEAKERS
MORE SPEAKERS TO BE ANNOUNCED!
INTERESTED IN SPONSORING EVENTS LIKE THIS?
CONTACT OUR FRIENDLY SALES TEAM HERE
Modern Lab
Leader 2024
ONLINE SYMPOSIUM
12
Technology Networks
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.
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 NRTX-1001 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 FDA-approved
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, 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.
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.
13
iStock
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.
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
14
iStock
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.
CONCLUSION
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. ⚫
15
EDITORS’ NOTE CONTRIBUTORS
Have an idea for a story?
If you would like to contribute to
The Scientific Observer, please feel free
to email our friendly editorial team.
Giving talks and posters at
conferences can be great but
can also be expensive and time
consuming. Your reach is also
limited to the people in the room.
What if you could get your
paper in front of
250,000 scientists
located across the globe, without leaving
your desk, for free and with minimal work?
Sound too good to be true?
Find out more
NEED TO DISSEMINATE
YOUR RESEARCH?
We want to help connect scientists with great research and have created an easyto-complete form and guidance to help distil the essence of your paper into a short,
punchy article that will then be shared:
on our site, which receives over
1 million page views
every month
with our
4 million + social
media followers
in our
18 newsletters
and may be included in our regular
digital magazine
The Scientific Observer
HOW CRISPR GENE THERAPY GAVE
A NEW LIFE
MOLLY CAMPBELL
BREAKING
VICTORIA GRAY
CHAINS: the
17
Veronica Gray modified, iStock
L
ike 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.
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
f low 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. W hen 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
“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.”
“I really gave up
on becoming
anything else.
I thought sickle
cell was just going
to be my life from
beginning to end.”
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”
FROM 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. W hile at his
clinic in 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 CR ISPR 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.”
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.
18
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.
19
In July 2019, she became the first
patient to receive CRISPR gene therapy for SCD.
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 wak ing up, and
the room was really bright. I didn’t
feel any thing, which was strange. I
didn’t have any shortness of breath
when I stood up, as had been the
case most mornings,” she says.
Having lived most of her life in excruciating pain and extreme fatigue,
the experience of wak ing up in its
absence was so downright bizarre,
Victoria had convinced herself
that she must be dead. Pinching
the sk in on her face, and her thighs,
she was reassured to feel the sharp,
physical sensations.
“I shouted to my k ids, ‘Hey y ’all,
come in here!’, and as they entered
the room, their faces lit up. I k new
in that moment, they could see me,
and I realized that I was very much
alive. I cried tears of joy, because
I k new 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 f lying
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 fulltime, 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 Casgev y. Upon
submitting the trial data, 31 patients
had sufficient follow-up time to be
Veronica Gray
“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’.”
20
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. Casgev y
became the first gene therapy
utilizing CR ISPR-Cas9 to receive
FDA approval, marking a historic
moment for the SCD community,
as lovotibeglogene autotemcel
(LyfgeniaT M) – another cell-based
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 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.”
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
A merican people were deprived of
jobs, educational opportunities,
marriage licenses and insurance
in the US if they carried the 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.
Veronica Gray
21
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 health-related
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. W hen 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.
W hat 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 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,
“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.”
“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.
22
iStock
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 Casgev y. 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 CR ISPR gene therapy
recipient, through advocacy work.
Once the news broke that she was
the first patient to receive CR ISPR
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. “ W hen I was f lown
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 CR ISPR 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.”
“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.”
23
iStock
Her advocacy work has most recently turned to the costs associated
with CR ISPR-based, and other
gene therapies, for SCD. A major
challenge for patients and clinicians
in accessing such emerging therapies will undoubtedly be their price.
W hile information on the exact cost
is currently limited, Casgev y 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 “oneand-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.
“ W hen 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 CR ISPR-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 A rea, 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
K liegman.
“The process of assembling the task
force and developing the report
24
iStock
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.
K liegman 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,” K liegman
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 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. ⚫
It’s simple, just choose the groups and
click the bubble.
Our Facebook groups allow you to keep up to
date with the latest news and research and share
content with like-minded professionals. With three
diverse groups there is something for everyone.
Hi. How are you? Have you seen our
Facebook groups?
Thanks! I’ll come join the conversation.
Come join the Neuroscience, News
and Research Group here.
Discover the Analytical Science
Network Group here.
Explore The Science Explorer Group here.
No I haven’t. Could you tell me more?
Sounds great! How do I join?
26
iStock
CRISPR gene editing
has enabled scientists
to rewrite the genetic
code of living organisms
and is revolutionizing medicine.
In 2023, Casgevy became the first
US Food and Drug Administration
(FDA)-approved therapy that utilizes CR ISPR-Cas9, offering new hope
for patients with sickle cell disease.
Alongside its cell and gene therapy
applications, CR ISPR technology
is becoming an increasingly popular
tool for infectious disease research.
It has allowed scientists to improve
their understanding of the biology
and genetics of human pathogens,
and is being explored as a technique
for diagnosing and treating diseases
such as human immunodeficiency
virus (HIV). Here, we highlight
some of the latest applications of
CR ISPR gene editing in infectious
disease research.
COULD CRISPR GENE
EDITING PROVIDE A CURE
FOR HIV?
Retroviruses like HIV cleverly integrate their genetic material into host
genomes and are notoriously difficult
to treat. Even with effective treatment,
some immune cells go into a resting
state but still contain HIV DNA.
Infection with HIV is currently treatable with lifelong antiviral therapy to
reduce viral load to undetectable levels, but it is not curable. CRISPR has
provided new hope in the search for a
HIV cure, and researchers are working
towards using this technology to completely excise the viral DNA from the
genome of host cells.
Scientists at Temple University published evidence last year showing that
a single injection of a novel CRISPR
gene-editing treatment safely and
efficiently removes simian immunodeficiency virus (SIV) from the genomes
of rhesus macaque monkeys.
The outcomes of this study set the
stage for an ongoing Phase 1/2 clinical
trial of EBT-101, a HIV-specific CRISPR-Cas9 gene-editing therapy, which
was granted FDA Fast Track Designation in July 2023. The preclinical study,
published in the journal Gene Therapy,
represented a significant advance
in the generation of a cure for HIV
in humans.
How Is CRISPR Gene Editing Being
Used in Infectious Disease Research?
BLAKE FORMAN
27
In a proof-of-concept study (the results
of which are yet to be peer reviewed)
presented at the 2024 European
Congress of Clinical Microbiology
and Infectious Diseases (ECCMID),
researchers claimed they removed
HIV from lab-cultured cells using
CRISPR-Cas9 gene editing.*
According to the scientists, they
adopted a broad-spectrum approach,
using CRISPR technology to edit two
regions of the HIV genome that are
conserved across all known strains
of the virus. They found that the size
of the vector used to transport the
cassette encoding the therapeutic
CRISPR-Cas reagents into the cells
was too large. Another challenge was
reaching the HIV reservoir cells that
rebound when HIV antiretroviral
treatment is stopped.
To overcome these challenges, the
authors tried various techniques to
reduce the size of the cassette and
therefore the vector system itself.
They successfully minimized the size
of the vector, enhancing its delivery
to HIV-infected cells, and were able
to target HIV reservoir cells by focusing on specific proteins found on
the surfaces of these cells.
The researchers hope to advance to
preclinical models to study the safety
and efficacy of a therapeutic strategy
combining CRISPR therapeutics and
receptor-targeting reagents.
CRISPR ENZYMES SUPPORT
PROACTIVE PLANNING
AGAINST FUTURE PANDEMICS
Searching for ways to improve
CRISPR-based solutions to RNA
viruses, which could help combat
future pandemics, is an active area
of research.
CRISPR-Cas13 systems have become
indispensable tools for various RNA
targeting applications, including antiviral development to combat viruses
such as SARS-CoV-2.
Within the Cas13 family, Cas13d is the
most active subtype in mammalian
cells. However, it is inefficient in the
cytoplasm of cells, where many RNA
viruses replicate.
What is EBT-101?
EBT-101 is an in
vivo CRISPR-based
therapeutic candidate
designed to excise HIV
pro-viral DNA from
HIV-infected cells. The
treatment employs
CRISPR-Cas9 and two
guide RNAs that target
three sites within the
HIV genome, thereby
excising large portions
of the HIV genome.
Examples of Cas enzymes
Many CRISPR associated proteins (Cas) possess nuclease
activity and play a vital part in the bacterial and archaeal
defense system. In 2005, the first Cas protein with
nuclease activity was discovered while studying the
genome of Streptococcus thermophilus. The protein is
now known as Cas9 and is one of the most common
Cas proteins used in CRISPR gene-editing of DNA. Other
popular systems include the CRISPR-Cas13 system, which
is used for precise RNA manipulation without permanent
changes to the genome. Cas12 and Cas14 enzymes are
also being explored in genome editing technologies.
The authors stated, “We
have developed an efficient
combinatorial CRISPR attack
on the HIV virus in various cells
and the locations where it can
be hidden in reservoirs, and
demonstrated that therapeutics
can be specifically delivered to
the cells of interest.”
28
iStock
Researchers from Helmholtz Munich
and the Technical University of Munich overcame this obstacle by engineering nucleocytoplasmic shuttling
Cas13d (Cas13d-NCS). This system
can transfer nuclear CRISPR RNA
into the cytosol.
The scientists showed that Cas13dNCS outperforms its predecessors in
degrading mRNA targets and neutralizing self-replicating RNA, including
replicating sequences of several variants of SARS-CoV-2.
This achievement represents a significant step in strengthening our
defenses against future outbreaks of
RNA viruses.
DISEASE VECTORS: CRISPR
ADDRESSES THE ROOT OF
THE PROBLEM
Many human pathogens such as
malaria are vector-borne. Vectors
are living organisms that can transmit infectious pathogens between
humans. Vector-borne diseases are
human illnesses caused by parasites,
viruses and bacteria that are transmitted by vectors. These diseases are
often transmitted from blood-feeding
arthropods like mosquitoes. CRISPR
gene editing provides an opportunity
to control the spread of these animal
vectors, thus preventing the transmission of the pathogens they carry.
Chagas disease can be transferred to
humans by insects such as triatomine
bugs, also known as kissing bugs. As
treatment options are limited, strategies for Chagas disease control have
focused on ways to manipulate the
organisms that carry the parasite.
The application of CRISPR technology in kissing bugs has proven
difficult. Traditional gene editing
methods involve injecting the CRISPR gene-editing material directly
into embryos, which, due to the hardness of kissing bugs' eggs, has proven
challenging.
In a recent paper published in The CRISPR Journal, researchers demonstrated
the application of CRISPR-Cas9 gene
editing in kissing bugs for the first
time, creating new possibilities for
using genetic technologies to control
vector-borne Chagas disease.
The research team from Penn State
College of Agricultural Sciences have
developed an approach called Receptor-Mediated Ovary Transduction
of Cargo or “ReMOT Control”. This
technology enables the injection of
materials directly into the mother's
circulatory system and guides that
material to the developing eggs.
CRISPR IMPROVES THE
TIME TO RESULT IN DISEASE
DIAGNOSIS
In addition to treating and preventing
infectious diseases, CRISPR technol-
“Here, we showed that you could genetically
modify this vector insect. Our technology has
the potential to make gene editing more efficient,
easier and cheaper in a wide range of animals,”
said Dr. Jason Rasgon, Dorothy Foehr Huck
and J. Lloyd Huck Endowed Chair in Disease
Epidemiology and Biotechnology at Penn State
College of Agricultural Sciences.
29
iStock
ogy has been widely used in research
developing novel diagnostic tools for
diseases such as SARS-CoV-2.
A rapid test for diagnosing melioidosis, a rare tropical disease, was recently described in a study published in
The Lancet Microbe.
Melioidosis is caused by the bacterium Burkholderia pseudomallei. Present
in soil and water in tropical and subtropical regions, the bacterium enters
humans via inoculation through skin
abrasions, ingestion or inhalation.
“Melioidosis has been neglected despite its high mortality rate and high
incidence in many parts of Asia. Early
diagnosis is essential so that the specific treatment required can be started
as soon as possible,” said Professor
Nick Day, senior author and director
of the Mahidol-Oxford Tropical Medicine Research Unit .
Diagnosis of melioidosis requires culturing bacterial samples, which takes
three to four days. In this study, the
team set out to develop a new rapid
test to reduce patient diagnosis time.
Their test, called CRISPR-BP34,
involves rupturing bacterial cells
and using a recombinase polymerase
amplification reaction to amplify the
bacterial target DNA. Additionally, a
CRISPR reaction is used to provide
specificity, and a simple lateral flow
read-out is employed to confirm cases
of melioidosis.
The team collected clinical samples
from 114 patients with melioidosis
and 216 patients without the disease
at a hospital in northeast Thailand,
where the disease is endemic. The
CRISPR-BP34 test was then applied
to these samples.
The new test showed enhanced sensitivity at 93%, compared to 66.7% in the
current gold-standard culture-based
method. It also delivered results in
less than four hours for urine, pus and
sputum samples, and within one day
for blood samples.
ELIMINATING
ANTIMICROBIAL-RESISTANT
BACTERIA WITH GENE
EDITING
Widespread misuse and overuse of
antimicrobials have led to antimicrobial resistance (AMR) being declared
one of the top global public health
and development threats. Scientists
across the globe are now rapidly
searching for viable alternatives to
antibiotics.
CRISPR has not only been used to
identify AMR genes but has potential
as a therapeutic tool to treat antibiotic-resistant bacteria and other
pathogens.
At North Carolina State University, researchers showed that the
CRISPR-Cas system can target and
eliminate the gut bacteria Clostridioides difficile (C. difficile) in vivo.
The results were published in the
journal mBio.
Antibiotic use is a major risk factor
for C. difficile infection because
broad-spectrum antimicrobials disrupt the indigenous gut microbiota,
decreasing colonization resistance
against C. difficile.
The study showed that the CRISPR-Cas system in C. difficile can
be repurposed as an antimicrobial
agent through the expression of a
self-targeting CRISPR that redirects
endogenous CRISPR-Cas3 activity
against the bacterial chromosome.
The researchers tested this approach
in mice infected with C. difficile. Two
days after the CRISPR treatment,
the mice showed reduced C. difficile
levels, however, those levels began to
increase again two days later.
The researchers explained that future
work will include retooling the phage
to prevent C. difficile from returning
after the initial effective killing.
THE FUTURE OF CRISPR
IN INFECTIOUS DISEASE
RESEARCH
The versatility of CRISPR gene
editing has resulted in its application
in many facets of infectious disease
research. As CRISPR technology
continues to undergo technical
improvements, the prospects for
its application in treating incurable
diseases such as HIV are becoming
increasingly promising. The notable
applications discussed here merely
offer a glimpse into the evolving
landscape of how CRISPR gene
editing can be harnessed to improve
human health. ⚫
* These research findings are yet to be
peer-reviewed. Results are therefore
regarded as preliminary and should be
interpreted as such. Find out about the
role of the peer review process in research
here. For further information, please
contact the cited source.
30
iStock
I
n 2016, Dr. Alexandra Freeman
returned to a career in academic research after several successful years
working in media.
Shortly after this transition, she observed concerning parallels between
the industry that she had left behind,
and that which she had re-joined. It
seemed as though, all around her, academics held a firm desire to tell what
Freeman describes as “neat, short,
easy-to-read and persuasive” stories
in their papers. But good research isn’t
about storytelling, she thought – it’s
about evidence communication.
Inspired to create change, Freeman
sat down one evening and got to work
developing Octopus, a novel and
radical publishing platform for scholarly research. She penned the idea in
a single night, and with funding from
UK Research and Innovation (UKRI),
officially launched Octopus in the
summer of 2022.
“In Octopus, there are eight types of
publication, each reflecting a part of
the research process,” Freeman told
Technology Networks in an interview
ahead of Octopus’ launch. “This allows
researchers to specialize in one or two
of these parts of the process (with their
very different skills) and to publish
their work literally in collaboration
with the rest of the research community – in time, as well as in space!
Someone might publish a theoretical
idea now, which someone else, 20
years in the future, will collect data
to test. Then, another researcher, 20
How To Enter a New Chapter in
Academic Publishing
MOLLY CAMPBELL
RADICALLY RETHINKING
SCIENTIFIC PUBLICATION:
THE "OCTOPUS" MODEL
READ MORE
31
Alexandra Freeman
years further in the future, might again
choose to analyze that data using new
techniques.”
As Octopus approaches its second
birthday, Technology Networks reconnected with Freeman to learn about
the academic community’s response
to the platform, how it has evolved
and her current stance on the publishing landscape.
Molly Campbell (MC): Can you
provide our readers with an
update on how the launch of
Octopus went, and how the scientific community has reacted
to it?
Alexandra Freeman (AF): The
launch itself was a great event, but
it was just the lighting of the fuse.
Since then, we’ve been continuing
to develop all the features that we
need to in order to fulfil the vision
that I originally had for Octopus –
and that will continue for another
year or more.
The reception has been great –
we’ve got over 1,000 users, and publications on Octopus get viewed a
lot – far more than I had anticipated!
W hen we show Octopus to people,
they are almost entirely positive
about it too – but that isn’t to say
that everything’s plain sailing, as I
knew it wouldn’t be.
MC: Octopus evolved from your
feelings of frustration towards
researchers’ desire to tell “neat,
short, easy-to-read persuasive
stories in academic papers.”
Has anything changed?
AF: In the big picture, no – not yet.
The incentive structure for researchers is still mostly geared towards a
paper in a journal (or a monograph).
To get the biggest readership (and
hence market) for those, the pressure
is for brevity, simplicity and readability. Very few “casual” readers
want highly detailed methods, complete results or analytical code etc.,
and it’s not commercially viable to
cater for those who do, (even though
that is what is needed to be able to
assess and build on research that’s
been done).
However, the ground is shifting
– funders are beginning to make
moves that could change the entire
landscape. Funders really care about
Figure 1: A branching chain of research publications on Octopus.ac.
The pressures that researchers feel come from
institutions and funders – the people who will
employ them and pay them.
32
the quality of research work and
have very few competing interests,
so it’s them that needed to take the
first steps – and that is happening. I
think we’re going to see the pace of
change pick up now.
MC: Almost two years after
launch, is Octopus having the
impact that you hoped for?
Have there been any challenges? If so, how did you overcome
them?
AF: Not yet. It is having an effect,
in many different ways, but I’m very
ambitious for the change needed, so
there’s a long way to go.
W hen we introduce the platform
and explain why it is designed the
way it is, researchers understand
it. A lmost all say they would like
to try it and support its aims. Most,
though, say that they don’t feel able
to use it regularly because of the
pressures they’re under – or feel
they’re under – to publish “the traditional way.”
Until fairly recently, there weren’t
that many alternatives to the traditional article formats, and it was
understandable for the research assessment system to be built around
these formats. But now, there are
better alternatives for sharing work
in enough detail for it to be fully
assessed and built on (Octopus being one example) and so the whole
landscape can change.
We need to make funders and institutions aware of these new alternative publishing platforms and their
advantages (to them, to researchers
and to the whole research landscape) so that they can change the
incentive system that researchers
feel trapped within. That is something I’m very much working on.
MC: Do you feel that the research community is aware of
Octopus and how they can use
it?
AF: “The research community” is
huge! A lot of people have heard about
it, although we know from research
that many have some misconceptions
about it – it is quite a different way of
working. I have published on it myself
and know some of the questions that
arise, like “what should be in a Results
publication, as opposed to an Analysis publication?”, so I think there’s
quite a lot of work still to do helping
people with their first publications.
But there are also going to be huge
numbers of people – around the
world – who haven’t yet even heard
of it. Many of those will, I think, be
mid-career researchers who feel there
is a set career path that they are on.
They likely feel very under pressure
to publish in certain ways, and don’t
even have the time and energy to
look at anything other than getting
the next publication in the specific
journal that they think they have to,
in order to secure their next contract
or grant.
The only way these researchers can be
freed to think about the quality of the
work they’re doing – and how useful
what they’re sharing is for people
other than themselves – is if institutions and funders make it very clear
indeed that “the system” has changed
and demonstrate that the old rules no
longer apply.
MC: There have been many notable changes to the publishing
landscape since our last conversation, such as the introduction of large language models
(LLMs). What are your thoughts
on the current scientific publishing landscape?
AF: There have been some big
changes. As you say, generative
A I is a huge one. We were dealing
with a system where people were
incentivised to get their names on
as many publications as possible
– that was bad enough. Now those
publications can even be generated
artificially within minutes, at scale,
by computers.
I can’t see any way in which the old
system, with a small group of volunteer peer-reviewers and editors,
can deal with sifting through an
almost infinite volume of papers,
to try and recognize the ones that
are created by humans and based on
actual research, and then pick out
the good research.
Rather than resorting to “easy” cues
that inevitably lead to bias (such as
previous reputation of authors or
institutions), I think we’ll need to
move to systems that demand greater evidence of the work done and
its trustworthiness – such as open
data, analytical code, etc.
I’ve always thought that the old
system of editorial approval is unsustainable: that we cannot rely on
“peer reviewed” as being a stamp of
trustworthiness. I think that has
become increasingly obvious as the
volume of published A I-generated
papers and A I-generated reviews
has been revealed. As readers, we
The old system of editorial
checking was already breaking
down under the volume of
publication, and that pressure
hose of publications has just been
turned right up.
33
are going to have to judge things for
ourselves more carefully and not
take things “on trust”, outsourcing the judgement to anonymous
reviewers; just as we have to with
any information we read online
these days.
On a more positive note, the Gates
Foundation has signaled the start
of another major change in the publishing landscape – it has announced
that it will no longer pay publishers
to publish papers. Instead, it will be
supporting more free, alternative
ways of sharing work, such as the
use of pre-print servers. I think this
is exactly the kind of leadership that
funders need to take. They have the
power to change the status quo, and
someone needs to!
MC: UKRI provided the funding
to launch Octopus, and for several years thereafter. Has funding been secured for the future?
AF: UKRI, through Research England, has released two more years
of funding for Octopus. However,
we do need to look ahead to the
future. Most importantly we want
to keep our costs minimal – it’s only
a tiny staff and enough to cover
the technical costs of keeping the
platform running. We want to make
the back-end database distributed
so that institutions can volunteer
to host a portion of it. This will
keep our hosting costs low, and will
ensure that all the data is safely mirrored across different geographical
locations.
MC: Are there any misunderstandings or misconceptions
about Octopus that you would
like to address?
AF: A few have come up, and it’d be
great to set the record straight.
1. Octopus is like a pre-print server
in that you can publish work on it
– and get peer reviews – and then
submit it to a journal. However,
since Octopus doesn’t publish
papers, you need to format your
work differently.
2. It is like a pre-registration
platform, in that you can publish your research questions,
hypotheses and methods before
collecting any data (and there
is a marker to specifically
highlight that you are pre-registering). But you do have to
make these public before they
get DOIs and dates. On the plus
side, like a registered report,
you can get peer review of these
before you go and collect data.
3. It is a bit like using GitHub, in
that you can “fork” a chain of research and take it in a different
direction, but unlike GitHub
(or ResearchEquals, or Jupyter
notebooks) it’s not a place for
day-to-day work, which is constantly changing or being updated. It’s designed to be where you
publish finished work (this can
be a smaller chunk of work than
you might think of when you’re
used to publishing papers).
4. It is like a repository, in that you
can put work on Octopus that
has been published in journals,
but again, the format is different
so you will need to do a bit of
work to turn it into Octopus
publications. The benefit, of
course, is that Octopus is open
for others to read, so your work
can get a broader readership
than a paywalled article, and
you don’t have word or format
limits so you can go into more
detail too.
MC: You run Octopus in your
spare time – you must be busy!
How are you balancing everything?
AF: I’m used to being busy! But all the
day-to-day work is done by a team
based at Jisc now, so although I keep
across everything with meetings –
and I still give quite a few talks – I
can fit it in. Octopus is so important
– I’ll make as much time as it needs
for as long as I possibly can.
MC: Is there anything else that
you would like our readers to
know about Octopus?
AF: If you agree with the principles
of Octopus, the best way that you
can help support us as a researcher
is to publish on it. It will probably
take you about an hour or two to take
one of your published papers and put
the work up on Octopus, depending
on how quick your co-authors are to
approve publications If you have less
time than that, see whether there is
a publication you can write a peer
review of – these take a much shorter
time than reviews of whole papers.
If you have the power to change
policy at an institution or a funder,
check whether your policies support
people who use these alternative
publishing platforms. W hat can you
do to help change the tidal stream
that good, careful researchers currently feel is against them? We all
have our parts to play. We can all
make a positive difference. ⚫
Keeping our costs minimal means
we shouldn’t need too much to
keep going. But it’s not nothing,
so I am going to be doing a lot of
talking to funders, philanthropists
and institutions.
iStock, Alexandra Freeman, Victoria Gray
34
Meet the interviewees whose insights featured in issue 35 of The Scientific Observer:
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.
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.
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.
Alexandra Freeman, PhD started with a 16-year
career at the BBC, working on various television series. Her work
won a number of awards, from a BAFTA to a AAAS Kavli gold
award for science journalism. She then joined the University of
Cambridge to lead the Winton Centre, where she had a particular
interest in helping professionals such as doctors, journalists or
legal professionals communicate numbers and uncertainty better,
and in whether narrative can
be used as a tool to inform but
not persuade. In 2024 she was
chosen to be a crossbench peer
in the House of Lords through
a process of selection. She is
an advocate of Open Research
practices and the reform of
the science publishing system,
and in her spare time leads the
Octopus platform for primary
research publication.
Melinda Kliegman, PhD, 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.
Meet the Interviewees
How a CRISPR-Like System
Was Discovered
in Eukaryotes
Cicadas Could Help Us
Develop Future Self-Cleaning
Everyday Surfaces
PSYCHEDELIC PSYCHEDELIC
ISSUE 29, AUGUST 2023
THE POWER OF PLACEBO IN THE POWER OF PLACEBO IN
TRIALS TRIALS
The Power and Potential of
Epigenetic Aging Clocks
Could Psychedelics Change
How We Treat Migraine and
Cluster Headache?
ISSUE 27, JUNE 2023
PRECISION
MICROBIOME EDITING
To Tackle Methane
Emissions
The Future of Food: Five Key
Considerations in Alternative
Protein Food Production
The Ultimate Guide
to Avoiding Predatory
Conferences
ISSUE 30, SEPTEMBER 2023
Exploring the Next Frontier in Food
Sponsored by
A Recipe for Richer
Metabolomics
Time To Say Goodbye to
the Traditional BMI?
HowCollaboration and
CuriosityMake for a
Successful Scientist
Towards theLab
of theFuture
Sponsored by
ISSUE 26, MAY 2023
NEURODEGENERATION
Are We Closer to a Cure, or Still a World Apart?
The Environmental Impact
of Biotech and Pharma
Operations
Immune Cartographers
With Professor Emma Lundberg
ISSUE 28
Half-Synthetic Yeast
Engineered for the First Time
How Body-on-a-Chip Is
Transforming Research
ISSUE 32, NOVEMBER 2023
Biology
NATURE'S TOOLS, REDESIGNED
How Do You Make a Water
Purifier From Fruit Waste?
Building an Atlas
of the Brain
Sponsored by
ISSUE 24, MARCH 2023
A Green Approach
To Upcycle Vegetable Waste
HPV Is Not Just a Women's
Health Issue
ISSUE 31, OCTOBER 2023
Little
Things
AN INSPIRED SCIENTIST'S PURSUIT FOR A CANCER CURE
The
Sponsored by
How Collaboration and
Curiosity Make for a
Successful Scientist
Towards the Lab
of the Future
Sponsored by
ISSUE 25, APRIL 2023
The Landscape
of Agricultural
Biotechnology
Strategies for Sustainable
Science at CU Boulder
and Beyond
ISSUE 35, APRIL 2024
Make sure you never miss an issue
of The Scientific Observer.
Subscribe Now
Make sure you never miss an issue
of The Scientific Observer.
Subscribe Now
Sponsored by
Download the Magazine for FREE Now!