2018 Biotech Primer WEEKLY Compendium
2018 Biotech Primer WEEKLY Compendium
2018 Biotech Primer WEEKLY Compendium
B I O T E C H P R I M E R ’ S
W E E K LY
C O M P E N D I U M
Dear Reader,
The biopharma industry is moving at lightning speed and it can be a challenge
to keep pace. Here at Biotech Primer we spend hours each week researching,
writing, and editing original content for the Biotech Primer WEEKLY with one
goal: to help everyone better understand the latest science and technology
driving today’s healthcare industry.
This interactive eBook is a collection of Biotech Primer WEEKLYs from the last
year. The Biotech Primer WEEKLY is a one-page, easy-to-read newsletter
delivered each Thursday right to your inbox. If you are not already registered
to receive the Biotech Primer WEEKLY go to BiotechPrimer.com to register.
If you find value in the WEEKLY newsletter and this WEEKLY Compendium
ebook please send a copy to your friends and colleagues and encourage them
to pass on as well.
Thank you for downloading the Biotech Primer WEEKLY Compendium.
Stacey Hawkins, CEO Biotech Primer
[email protected]
Biotech Primer is a training company specializing in life science courses
for the non-scientist.
Go to BiotechPrimer.com
• Learn about our in-house, customized training
• Register for our open enrollment courses
• Enroll in our online learning
• View current WEEKLY issues not found in this eBook
• Register to subscribe to the WEEKLY
5 New Hope For Spinal Muscular Atrophy 65 The Multiple Myeloma Landscape
Approved to treat a variety of diseases including • This supplies the force needed to drive the
rheumatoid arthritis, diabetes, multiple sclerosis, drug-containing needle into the intestinal wall —
Crohn’s disease, and a whole range of cancers, these delivering the biologic to the bloodstream.
drugs include monoclonal antibody therapeutics, • The needle itself then dissolves, and the balloon
hormones, and immune system signaling molecules. is excreted.
Their safety and efficacy depend largely on the complex,
This may sound like the stuff of science fiction — and
three-dimensional structure of the protein product
indeed, the device is still in preclinical trials — but the
itself — which is incredibly delicate and time-consuming
robotic pill has attracted big name pharma partners,
to develop on a large scale. The majority of these are
including AstraZeneca (London, UK) and Novartis
administered via injection directly into the bloodstream
(Basel, Switzerland), as well as investment from
for maximum potency.
Google Ventures (Mountain View, CA) among other
So, why can’t we just swallow a biologic pill? The simple venture funds. If the device ultimately succeeds, it will
answer: biologics would not survive the acidic pH and revolutionize the delivery of biologic drugs.
digestive enzymes of the gut. Even if survival were
possible, the next issue to contend with is absorption;
ENGENE’S GUT CELL FACTORIES
if the protein is not broken down properly, it will not
be absorbed into the bloodstream. Companies seeking enGene (Vancouver, Canada) is bypassing the delivery
to develop oral delivery of biologics must overcome obstacle by attempting to turn the cells of the gut into
both hurdles. drug-producing factories. The trick is to deliver a gene —
the instructions for a specific therapeutic protein — to
Injectable delivery is cumbersome at best. It has a
those cells. enGene is using tiny carbohydrate-based
significant impact on a patient’s quality of life, which
nanoparticles to encase the gene. The carbohydrate
in turn affects compliance to drug therapy regimes.
coating protects the gene as it passes through the
In many cases, patients must make regular trips to an
stomach, yet allows it to be absorbed into the intestines.
infusion center to receive required treatments. Thus
The protein is then made in the intestines.
oral administration is the holy grail in terms of delivering
biologics. In this WEEKLY, we’ll take a look at the different This approach is especially appealing for diseases
paths being pursued to make this ambition a reality. affecting the colon and small intestine, such as
inflammatory bowel syndrome or Crohn’s disease.
enGene is currently using this platform in the preclinical
THE ROBOTS OF RANI THERAPEUTICS
development of the anti-inflammatory protein IL-10, for
One of the hottest companies in the oral delivery of the treatment of inflammatory bowel disease.
biologics space is Rani Therapeutics (San Jose, CA).
Their product is a “robotic pill” — a small device capable
APPLIED MOLECULAR TRANSPORT
of traversing the intestinal tract and injecting a biologic
drug directly into the body. It works in the following way: Applied Molecular Transport South San Francisco,
CA) is using a protein scaffold adapted from pathogenic
• After the pill is swallowed, it makes its way through
microbes such as salmonella, which colonize the gut by
the digestive process, keeping its fragile biologic
secreting immune-crippling proteins into our body.
drug cargo tucked safely inside. Its capsule is
pH-sensitive and stays intact at the low pH of Proteins from these “gut bugs” work by tricking the
the stomach. intestines into absorbing toxic proteins in the same way
NAKED DNA
WANTING THE WNT PATHWAY
Juventas (Cleveland, OH) is also developing a gene
Just last month, a new regenerative medicine company,
therapy for heart disease. However, instead of using
Surrozen, was launched in South San Francisco.
modified virus, Juventas relies on plasmids — small,
The company founders include scientists who have
circular pieces of DNA engineered to contain the
spent decades deciphering a cell signaling pathway
therapeutic gene. In this case, the plasmids are injected
known as the Wnt pathway, which plays a central role
directly into damaged cardiac tissue. Advantages to this
in stem cell maintenance and tissue regeneration.
“naked DNA” approach include reduced patient immune
Surrozen’s focus is on developing drugs that activate
response and lower manufacturing cost.
this pathway, potentially leading to treatments for
Dubbed JVS-100, the Juventas plasmid contains a degenerative diseases.
gene for stromal-cell derived factor-1 (SDF-1), which
If any of these companies are successful a shot of stem
recruits cardiac stem cells and promotes new blood
cells — not major surgery — may repair previously
vessel growth. Like Generx, JVS-100 does not integrate
unfixable tissue damage. We’ll keep a close eye on these
into the recipient’s own DNA, making the production
exciting developments and be sure to report them back
of SDF-1 temporary. JVS-100 has completed Phase I
to you.
clinical studies.
How does it work? • Infrared light is shone, activating the drug and
damaging the cancer cell membrane. This damaged
• The antibody binds to its target immunogen on the
membrane allows water to enter, causing the cancer
surface of the cancer cell.
cell to burst and die.
• The antibody-drug conjugate is then taken up or
• Upon bursting, the cancer cell releases tumor
internalized by the cancer cell.
immunogens that serve to activate the immune system
• Once inside the cancer cell, the linker is degraded to further recognize and target more tumor cells.
and the active drug released.
One caveat to using photoimmunotherapy is the
The ability to target only cancer cells allows drug tumor must be accessible to an infrared laser in
designers to use drugs that are much more toxic order to activate the photosensitive drug. Aspyrian
than traditional chemotherapy. The ADC has a higher Therapeutics (San Diego, CA) is in Phase II clinical
specificity and only attacks cancer cells, avoiding studies of photoimmunotherapy antibodies that target
nearby healthy tissue which is often destroyed head and neck cancer.
by chemotherapy.
Over the years, monoclonal antibodies have proven
to be safe and effective therapeutics in a number of
different indications, most notably various cancers
and autoimmune disorders. Innovations in antibody
technology ensure monoclonal antibodies will
remain one of the most powerful tools in the drug
development arsenal.
WHY MRNA?
You probably recognize DNA as the molecule of heredity,
and may recall that it provides cells with the instructions
for making proteins. Enter messenger RNA (mRNA) — the
literal messenger that relays the DNA instruction to the
ribosome where the protein-making process takes place.
RNA RELAY
Messenger RNA (mRNA) is a molecule that carries a
copy of the protein-making instructions from DNA in the
MEET INTRONS & EXONS
nucleus of a cell to the ribosomes located just outside
of the nucleus. The information contained in mRNA is Exondys 51 (Sarepta Therapeutics; Cambridge, MA)
used by the ribosomes to assemble a protein. Without and Spinraza (Ionis and Biogen; Cambridge, MA) are
mRNA relaying the instructions, there would be no recently-approved antisense drugs that work by altering
protein produced. how the cell processes pre-mRNA — an immature
template of mRNA. Pre-mRNA is a long strand of RNA
from which specific pieces are cut out or added in before
ANTISENSE EXPLAINED
becoming the mature mRNA that is able to relay the
Antisense drugs are short (between 10 and 30 protein-making instructions from DNA to the ribosomes.
nucleotides), synthetic pieces of nucleic acid whose
Think of pre-RNA as a master grocery list that includes
sequence is complementary to the mRNA that codes for
all of the most common foods you buy over the year.
a disease-associated protein. When the antisense drug
Envision mRNA as your adjusted grocery list that
enters a patient’s cells, it binds to the disease-causing
includes only what you need to buy for this week.
mRNA. This binding triggers an enzyme called RNAse H
to destroy the double-stranded antisense-mRNA hybrid. When pre-mRNA is first produced, it contains two types
Our bodies recognize that double-stranded RNA as a of sections that are either cut out or left in depending on
mistake and destroys it. Without the mRNA, the disease- the instructions:
associated protein simply is not made — stopping • Introns: regions cut out of the final mRNA; grocery
disease in its tracks. items you delete from your master list because you
The history of antisense in drug development is quite don’t need them this week.
fickle. Over the years the inability to get antisense • Exons: regions included in the final mRNA; grocery
drugs to accumulate at therapeutically effective items you keep from your master list because you
concentrations in the target tissue was the undoing of need them this week.
many clinical trials. But the love affair with antisense
The company has two more Nanobody products in As Nanobodies continue to be tested for safety and
Phase II clinical testing: Vobarilizumab, which reduces efficacy, a whole new kingdom of potential antibody
the activity of interleukin-6 (IL-6). IL-6 is a protein targets may begin to emerge.
that stimulates the immune response; inhibiting the
Amicus Therapeutics (Cranbury, NJ) found itself in The only Fabry’s disease treatment on the market in
the news earlier this month when the FDA agreed to the U.S. is Fabrazyme, which is made by Genzyme
review the company’s new drug application for their (Cambridge, MA). Fabrazyme is an enzyme-replacement
investigational therapy to treat Fabry’s disease. The therapy: since the patients don’t make enough functional
drug under consideration, migalastat, has already galactosidase enzyme, scientists produce it in the lab
been approved by the European Medicines Agency. using cells that have been genetically engineered to
It belongs to a small, but growing class of therapeutics produce the enzyme, which is then purified and injected
known as pharmacological chaperones that properly fold into patients.
improperly folded proteins that cause disease.
MECHANISM OF ACTION: MIGALASTAT
Let’s take a look at which chaperones are on the dance
floor and find out the steps they are taking to treat Amicus’ drug migalastat, if approved, would be the first
disease caused by proteins. small molecule treatment for Fabry’s. The potential
availability of swallowing a drug (vs. injecting) would give
TERM OF THE WEEK: CHAPERONE PROTEIN those with Fabry’s another drug delivery option.
Chaperone proteins are proteins that assist in the correct In the lab, migalastat binds to and inhibits galactosidase.
folding and assembly of other proteins. Many of the In the body, this high affinity is taken advantage of by
proteins produced by our cells require chaperone migalastat binding to mutated galactosidase during
proteins to ensure their correct molecular structure. the process of folding, where it then shifts the folding
towards the correct conformation. The now correctly
A pharmacological chaperone is a small molecule drug folded protein makes its way to a cellular compartment
that targets specific misfolded proteins and encourages known as the lysosome, where it carries out its job of
them to fold correctly. digesting lipids. The inside of the lysosome has an acidic
Protein misfolding plays a role in many different rare pH, which causes migalastat to disassociate, leaving
diseases, including enzyme deficiencies like Fabry’s behind a functional galactosidase enzyme for the body
and the related Niemann-Pick disease, as well as to pick up and use.
Huntington’s disease, and some cases of amyotrophic Fabry’s is caused by a variety of different mutations
lateral sclerosis (ALS). Some of the mutations in the within the galactosidase gene; not all of them are
genetic disease cystic fibrosis (CF) involve misfolded amenable to treatment with migalastat. Amicus
proteins. Diseases caused by misfolded proteins scientists estimate that between 35% to 50% of patients
that disrupt cellular function are sometimes called will be responsive to migalastat.
proteopathies, where proteo = protein, pathy = disease.
TERM OF THE WEEK: KILLER T-CELLS So what’s a scientist to do?! Figure out a way to train
killer T-cells to ALWAYS recognize and destroy cancer
CAR-T therapy is modeled after a cell in the immune
cells… enter CAR-T.
system known as the killer T-cell. The job of a killer T-cell
is exactly what the name implies — to kill dangerous
cells. They target diseased cells in the body via their HOW TO TRAIN AN IMMUNE SYSTEM
receptors: each one has a uniquely shaped receptor, Killer T-cells are “trained” to go after early and late stage
and will recognize its intended target because the shape cancer by having their physical structure altered. This
of its receptor “matches” or fits into a uniquely shaped alteration is accomplished by fusing an antibody with the
surface protein found only on diseased cells. Once the receptor of a killer T-cell to create a chimeric molecule —
Killer T-cell “docks” onto its target, it injects an enzyme or the “C” in CAR-T.
which triggers death. The result: no more bad cells. Training day begins by having killer T-cells drawn out of
a patient’s body and isolated in the lab. Next, scientists
deliver a gene to the T-cells that encodes the chimeric
receptor. This receptor consists of two parts:
• A targeting domain: This is the part of the chimeric
receptor that will be outside of the T-cell. It is
composed of an antibody that will recognize
and dock onto a unique surface protein of the
patient’s cancer.
• An activation domain: This part of the receptor will be
triggered once the targeting domain is engaged. It
will signal to the killer T-cell to:
1. Stay alive.
2. Make copies of itself.
3. Release signaling molecules called cytokines.
Cytokines are chemical signals that activate
other white blood cells to join the fight against
the tumor.
4. Kill the target cell.
NON-SPECIFIC IMMUNITY:
MACROPHAGES AND NEUTROPHILS
Non-specific (or innate) immunity fends off pathogens at
the cellular level. The troops are specialized white blood
cells (WBC). Most WBC in the body are non-specific,
meaning these foot soldiers attack in the same fashion Activated T-cells divide rapidly and produce three types
without stopping to consider the specific characteristics of descendants: killer T-cells, helper T-cells, and memory
of the enemy. T-cells. All recognize the same target as the originally
Macrophages are one type of non-specific defender that activated T-cell.
freely circulate in the bloodstream. When they encounter • Killer T-cells roam the body in search of their pre-
a bug, they engulf it – essentially eating it. Macrophages programmed epitope, and seal the deal by injecting
have the ability to differentiate between good and bad granzyme B, triggering cell death.
OXYGEN DEPRIVED
Earlier this year, Cellectis (Paris, France) published a The decoy peptides reduce cytokine production by
paper describing work they’ve done to engineer CAR-Ts ~70%, which is likely enough to prevent the immune
with an oxygen-sensitive domain. response from spinning out of control. In the words of
Laura Savatski of the Blood Research Institute, “By
Under normal cellular conditions, this domain signals the
reducing cytokine production by CAR-T cells, you prevent
CAR-T to remain inactive. Under low oxygen, or hypoxic
a cytokine storm from happening. So instead of dealing
conditions, this domain sends an activation signal to
with a problem at the back end of the therapy, you solve
the CAR-T. Since most solid tumors have a hypoxic
it at the front end through intelligent design.” Decoy
environment, an oxygen-sensitive CAR-T should be
molecules are currently in preclinical development.
activated within the tumor but not outside of it.
With some of the best minds in the biopharma industry
working on CAR-T design, this landmark FDA approval is
INTELLIGENT DESIGN THROUGH DECOYS
likely to be just the first shot in a treatment revolution.
It turns out that CAR-T activation results in cytokine
production because part of the CAR-T is actually inside
A few key challenges have prevented xenografts from • After several rounds of cell division, an early-stage
solving the problem of organ shortages for patients in embryo is formed, which can then be transferred
need. They include: into a surrogate mother to complete development.
• Determining the correct animal donor, taking into This was the first time that as many as 25 genes had
account both anatomical and ethical considerations. been simultaneously and precisely deactivated with
• Targeting domain: This part of the CAR exists outside Many immunologists believe that prompt action by NK
the white blood cell. It is composed of an antibody cells helps eliminate cancer cells early on– before they
that recognizes and docks onto a specific cancer grow into a serious problem. However, in the early stages
surface protein. of tumor development, there are often not enough red
flags – abnormal proteins on the cancer cell surface – to
• Activation domain: This component kicks into gear tag them as dangerous. Engineering NK cells to display
once the targeting domain locks onto the intended a CAR “trains” them to recognize and respond to tumor
cancer surface protein. In CAR-T cells, the activation cells. Once activated, the CAR-NKs behave much like
domain signals T-cells to do three things: 1) make killer T-cells, releasing cytokines that bolster the immune
copies of themselves; 2) release signaling molecules response to the cancer cells--killing the nasty cells by
called cytokines (proteins that prompt other white injecting even nastier toxins.
blood cells to attack the tumor); and 3) finally—the
really good bit-- kill cancer cells.
HOMEGROWN ISN’T ALWAYS BEST
CAR-NK cells have two important advantages over
CAR-T cells: safety and accessibility. CAR-T cells must
come from the patient’s own T-cells to avoid triggering
graft-versus-host-disease (GVHD). This potentially
deadly illness occurs when the patient’s immune system
responds badly to foreign tissue. Donor NK cells, in
contrast, don’t appear to cause GVHD.
THERAPEUTIC CHOICES
There are small molecule drugs and large molecule
drugs. Small molecule drugs include medicines most
of us know, such as aspirin and over-the-counter cold
remedies. In contrast, large molecule drugs, also known
as biologics, are often specialty drugs, delivered through
injection by a healthcare professional.
NO GLOW
Therapeutic choice hinges on a disease’s origins. If an
illness originates outside the cell, a biologic is a logical Scientists use similar colorimetric tests for large
choice since it is too big to enter the cell. Though more molecule drugs such as antibodies. Let’s say that
expensive to produce, biologics typically cause fewer activated “Receptor X” turns on a particular gene that
adverse reactions than their smaller counterparts. in turn causes cancer. Scientists can engineer cells so
when Receptor X is activated, the cell produces a green
Small molecule drugs attack disease from within the
fluorescence and when it is not activated no green
cell. The downside? Their small size means they have
appears. Researchers are hoping for “no glow” or in lab-
the potential to also activate many off-target molecules.
speak, a “blank.” Blanks are the winners that go on to the
This collateral damage increases the chance of
next stage of drug development.
adverse reactions.
ASSAY DEVELOPMENT:
GLOW AND NO GLOW
To find the best drug candidate, scientists need to
design easy-to-perform, large-scale, fast, and accurate
assays. Think automation. Think high throughput
screening. Scientists often work with tests that produce
a fluorescent signal or color change because color is easy
to measure, relatively inexpensive to design, and safe.
Depending on the assay, a color change may or may not
indicate a drug candidate works.
LOOKING AHEAD
Luxturna’s success raises hopes of cures for numerous
other diseases caused by a defect in a single gene,
including hemophilia, sickle cell anemia, Huntington’s
disease, and other types of hereditary blindness. As
the table below indicates, many companies have gene
therapy treatments in clinical trials.
Biotech Primer Weekly wrapped up last year by exploring • Maximum concentration (Cmax) of a drug in plasma
the first two stages of drug discovery. We looked at and target tissues;
how pharmaceutical companies identify drug targets, • Bioavailability (where in the body and at what
or the molecules (usually proteins) involved in an illness concentration the drug ends up);
that an ‘as yet undeveloped drug’ will hopefully act on.
• Half-life, or how long a drug takes to reach half its
Next, we examined how researchers develop those
maximum concentration in the body;
pharmaceutical candidates. Now we turn to what comes
next for fledgling drugs on the arduous journey to FDA • Clearance, or the time for a drug to reach
approval. First, though, a smidgeon of Latin. undetectable levels through excretion.
Pharmacodynamic analyses involve observing the
PRIMUM NON NOCERE biological repercussions of increasing amounts of a drug.
This maxim from Hippocrates means “First, do no harm.” Negative side effects include nausea, loss of appetite,
Before testing a drug on people, researchers must make fatigue, skin sensitivity, and changes in blood pressure
sure it’s safe in two important ways. First, they assess a or heart rate. Animal PK and PD studies give scientists an
substance’s safety in vitro. This Latin phrase means “in idea of what a safe and effective dose of a drug might be
glass”—that is, lab-grown cells. The cell type varies, but in people.
if possible, researchers use one that is relevant to the
disease in question. For example, they may use lung cells
to test a drug for respiratory syncytial virus (RSV).
In vivo testing (“in a living thing”) comes next. This area
of preclinical testing assesses a drug candidate’s toxicity
in at least two different species of animals, such as mice
and guinea pigs. The animals receive more of the drug
for longer than would human volunteers. Meanwhile,
lab technicians watch the animals for adverse effects.
Preclinical testing must follow the FDA’s Good
Laboratory Practice (GLP) guidelines. These regulations
help ensure scientific integrity and humane treatment of
laboratory animals.
HUNTING FOR MUTANTS
HOMING IN ON THE SWEET SPOT Most drug candidates also undergo mutagenicity
testing, which determines their likelihood of triggering
Almost any substance–even water!–can be toxic in
mutations. Causing mutations indicates that a fledgling
very high amounts. Consequently, drug developers
drug may be carcinogenic, hence often consigning it to
aim for just the right dose–one that gives the desired
the graveyard of pharmaceutical failure. One common
effect with minimal unwanted consequences. Finding
screening for mutagenicity is the Ames test, which
the sweet spot is the domain of pharmacokinetic
identifies chemicals that cause increased rates of
(PK) and pharmacodynamic (PD) analyses. Think of
mutations in bacteria.
pharmacokinetics as “what a body does to a drug” and
pharmacodynamics as “what a drug does to a body.” Researchers can also assess a drug’s carcinogenic
potential by examining test animals for tumors.
PK analyses typically measure:
COCKTAIL FODDER: SAME FAMILY, Today’s technology makes it possible for the curious
DIFFERENT ANCESTOR? to shed light on their DNA. The databases of genetic
markers are growing apace, making it easier to uncover
Can siblings’ ancestral origins be different? The long-lost relatives, for better or for worse. So how
surprising answer is yes! Imagine that a small about it? Are you ready to find out who you really are,
percentage, say 10 percent, of a woman’s DNA contained biologically speaking?
genetic markers indicating Mongolian ancestry. Because