Managed Access
Before you read this post: I’m about to weave a long story that might take 20 – 30 minutes to read. I start with recent good news before walking us back to 1953 and some phenomenal science that followed to allow this good news. Next, I add a ‘crash course’ that covers the steps from drug discovery and development through to clinical trials, regulatory submission and approval to commercial marketing. I’ll gloss over some things to simplify, but I do cover a lot, so read on at your own discretion.
Some good news

- share more about why the biochemist in me sees Vorasidenib as an exceptional example of science in action,
- put my Biochemistry B. Sc, post-grad diploma in Pharmaceutical Research & Development, and my 23-year career in Pharmaceutical Product Development to use in summarizing what it takes to develop, test, and submit new drugs for regulatory approval.
As we each care for ourselves and become caregivers for
those important to us, it’s likely that many of us will encounter discussions
about clinical trials and new therapies while also learning about a new condition,
disease, or illness we know little about.
My hope is that the latter part of this post might offer some broader
context – if this sounds interesting, read on!
Vorasidenib Managed Access Program / Special Access Program
I’ll start this section with a reference back to my January
2024 blog post where I described why it was an “exciting
time for Low Grade Gliomas”. In that
post, I shared my excitement as I learned about the science that had happened
in the years after I studied Biochemistry, where investments in foundational genetic
research led to targeted research in many forms of cancer, including brain
cancers. Scientists began to find
connections between some cancers and mutations in Isocitrate Dehydrogenase
(IDH), an enzyme that plays a key role in the Citric Acid Cycle which is key in
cellular energy metabolism and biosynthesis
By 2009, scientists had learned how mutated IDH alters the
Krebs cycle such that mutated versions of the enzyme begin producing a new
molecule, R(-)-2-hydroxyglutarate (2HG).
The mutated enzyme gave cells an energetic advantage, and as this new
2HG accumulated, it contributed to progression to more aggressive cancers:
“cancer-associated IDH1 mutations
result in a new ability of the enzyme to catalyse the NADPH-dependent reduction
of α-ketoglutarate to R-2-hydroxyglutarate (2HG). … Excess accumulation of 2HG
has been shown to lead to an elevated risk of malignant brain tumours in
patients with inborn errors of 2HG metabolism. … IDH1 mutations result in
production of the onco-metabolite 2HG, and indicate that the excess 2HG which
accumulates in vivo contributes to the formation and malignant progression of
gliomas.”
This research and more now gives me a clearer understanding
of how my brain cancer likely developed.
At some point, an ‘early’ precursor cell like a pluripotent cell, NG-2
glial precursor cell or Oligodendrocyte Progenitor cell randomly acquired the
394 C>T mutation in one copy of the gene encoding for IDH1, where the nucleotide
change from Cytosine to Thymine meant that the IDH enzyme had the amino acid
Cysteine instead of Arginine at position 132 – IDH1 mutant type “R132C”. This cell began to accumulate 2HG that over
time induced other changes such as the chromosomal 1p36 and 19q13 codeletions and
associated allelic losses.
I’ll step back quickly to talk about brain tumours and brain
cancer: brain tumours may form from cancers
elsewhere that have travelled to the brain through blood vessels, the lymphatic
system, or other means. Such metastatic brain
tumours are often referred to as secondary tumours and are the most common
causes of brain cancer
Around 350,000 people around the world receive a new brain cancer
diagnosis each year
With an understanding of the mechanism behind several types
of brain cancers, clinicians and scientists began work on identifying possible
ways that we could take advantage of the mutated enzyme for what it is
(considering its modified shape) and what it does (producing 2HG) and explored
what would have been a vast number of possible approaches to using this new
mechanistic understanding to find a treatment.
By 2017, Agios Pharmaceuticals had invested heavily such that the team
had a direction, captured in a patent describing a base molecular structure
which could be added to in multiple ways to form a large ‘family’ of related
molecules with the potential to effectively target the mutated enzyme in a way
that would inhibit it from working properly – using ‘what it is’ to stop it
from doing ‘what it does’.
By December 2020, the French-headquartered global pharmaceutical
company Les Laboratoires Servier invested their money ($2 billion, plus
royalties)
a) serial monitoring via MRI imaging in a “watch and wait”
phase (with or without surgery) in a low-grade brain tumour while tumour location,
size, growth, and progression are manageable
b) begin targeted radiation therapy to attack cancer cells
but with known harm to surrounding brain tissue
As an elegant approach to target only the mutated version of
the IDH enzyme, Vorasidenib can slow progression without causing new cancers or
brain damage. The Indigo clinical trial results
I described earlier – about Progression Free Survival (PFS) and Time To Next
Intervention (TTNI) – mean that this drug has already been shown to give
patients more time in the “watch and wait” period before needing another
intervention. Today, the next
intervention is still going to be radiation and chemotherapy, but this added
time will also allow patients to watch as the science progresses with other
innovative treatments, and to wait for access to new not-yet-available
therapeutic alternatives. The potential
for extra time offered by AG-881 / Vorasidenib is fantastic for its potential
to add years without significantly reducing quality of life.
I shared in an earlier post that “February
20, 2024 was an exciting day!” because that marked the day that Servier
submitted Vorasidenib for regulatory approval, with a breakthrough therapy
designation supported by a companion diagnostic test for IDH mutation, meaning
a regulatory decision is due by August 20, 2024. Less than four months after the February
submission, we’re now at the point where Servier has started expanding access
to this drug under their MAP and Health Canada’s SAP I described earlier. This reminds me again how my diagnosis came
at such an opportune moment with respect to the scientific and clinical
progress already underway. I’m
remarkably lucky to have had my cancer appear and have been diagnosed at a time
that now gives me access to this new therapy.
I’m excited also that my treatment under this extended access phase
comes with frequent blood testing and follow-ups that get reported back to
Servier and add data to their drug safety database. My experience will provide researchers more
data to grow understanding about the product’s potential adverse events and strengthen
the safety profile for future patients.
This concludes the first part of today’s post, describing the
fortuitous timing of decades of innovation overlapping with my personal
diagnosis. The rest of this post will be
my ‘crash course’ on drug discovery, clinical trials, drug applications and
approvals, and how this all comes together.
If it sounds interesting to you, read on!
Drug discovery
Drug discovery starts with understanding the disease,
illness, or condition that needs a new therapy such that the mechanism(s) can
be understood, and a drug can be targeted to interact with the mechanism in
some way. To frame this section, I’ll simplify
some main aspects of biochemistry by condensing an entire textbook
To put this in context of therapeutic drugs, I’ll briefly
explore processes that control blood pressure.
The heart pumps blood through blood vessels in our body and it feels intuitive
to understand the pressure in these blood vessels by looking at a) how hard the
heart pumps, b) how relaxed or constricted the blood vessels are, and c) the
volume of blood being pumped through the closed system. Years of research mean that we have a detailed
understanding of the role our kidneys play in regulating total blood volume by retaining
or releasing water, hormones that control blood vessel diameter and the heart’s
pumping action, and with local effect in specific tissues and organs balanced
with global whole-body effects.
Identifying and screening the right target
After finding the control mechanism(s), the next step is to
find / discover / design a molecule that might activate or deactivate the pedal
we’re interested in. This often starts
with looking at the 3-D structure of the protein(s) involved along with the 3-D
structure of the molecule(s) usually involved, then designing an alternate with
just the right shape. This alternate
often comes in the form of an adjustable template with a core molecule where subtle
changes in one or more functional group(s) can be changed to get just the right
fit to have the right effect. This
usually results in thousands of possible molecules that now need to be
researched and evaluated. It takes time
to develop synthetic routes capable of producing each molecule, and more time
to evaluate each one. Modern
computational tools help, but it is still a large task to evaluate thousands of
options.
In the case of Vorasidenib (AG-881), the naming might make
us think that this was ‘just’ the 881st compound evaluated but
history tells us that would be too quick an assumption. The drug Mifepristone has been in the news
recently based on US Supreme Court decisions, but some may remember the stories
in the late 1980s and early 1990s when many of us knew the research project
name RU-486.
I don’t expect that AG-881 will appear in song lyrics soon, and
I don’t know whether this was the 2,881st compound Agios explored,
or the 13,881st or 27,881st or some other n,881st
compound but that it does represent a significant scientific
investment. For the thousands of candidate
compounds, project team members would have had to eliminate compounds known to
have potential harmful effects or potential for metabolic transformation into other
harmful molecules before even testing molecules for effectiveness. Scientists today have a suite of tools to
help with screening in advanced computer software (in-silico studies) before
moving to various traditional chemistry lab analyses (in-vitro studies) and
testing in living organisms (in-vivo studies).
In the case of Vorasidenib, inhibitors could first be evaluated with
in-vitro pre-clinical assays to assess whether a proposed compound might be
effective.
As a complicating factor in developing drugs to treat the
Central Nervous System, the blood vessels in the brain come with an added level
of protection. Blood vessels elsewhere in
the body are ‘leaky’, such that they allow liquid and components of the blood
to transfer into the tissues around the blood vessels, much like a soaker hose
in a garden that allows water to seep out to water the surrounding soil and
plants. Even ‘large’ things like the
immune system’s White Blood Cells can move from capillaries into surrounding
tissue to fight infection. In the brain
though, blood vessels are more tightly connected such that larger molecules can’t
leave the blood and gain access to the surrounding brain tissue. Blood vessels in the brain act more like a
typical garden hose that doesn’t leak water across its length. This Blood Brain Barrier protects the brain
from potential harm from undesirable molecules or infections but presents a
challenge for drug development teams who must now find a small molecule that
can gain access to the brain cells the molecule is intended to target.
After narrowing down the list from thousands of potential
candidates to a smaller group for testing, scientists will typically explore
the compound for potential adverse reactions (side effects) from known biochemistry
and finalize a list that can proceed to Clinical Trials as the next step, where
more information can be gathered about efficacy and other not-yet-known
potential adverse events. With a list of
potential Active Pharmaceutical Ingredients (APIs), work moves on for further
study, which is where we as patients and caregivers typically first encounter
them. In our Vorasidenib story, AG-881
was identified as a small brain penetrant molecule that had the potential to be
made into an oral drug that could be absorbed in the stomach, distributed
through the blood, could cross the Blood Brain Barrier and into cells throughout
the body. It could eventually come
across the mutated IDH enzymes in brain cancer cells where it would compete
with the active site of the enzyme and slow down the altered activity that
promotes tumour progression, all without blocking the ‘normal’ – Wild Type –
versions of IDH crucial for energy metabolism in all of our cells. More work was needed to find out how safe it
could be, and how well it might work, which brings us to clinical trials and
studies.
Clinical Trials and Studies
In this section, I focus mostly on the traditional ‘A vs. B’
clinical trial to figure out whether a new medicine / API can have a beneficial
outcome for patients. After, I’ll broadly
describe how this aligns with parallel work by teams who focus on 1) the drug
substance and the processes to scale up API synthesis from lab to commercial
scale, and 2) the drug product by developing the final tablet, capsule, injection,
or other dosage form that will eventually be made available to patients.
In general, the design of pre-clinical and clinical trials begins
with an approach to minimize potential harm to participants while growing
knowledge about the efficacy of an API and its final dosage form and learning
about the safety profile through appropriate strength and by collecting
information about potential adverse reactions and events that most of us refer
to as side effects. With a knowledge of
the condition being targeted and the mechanisms involved, researchers adjust study
design at each phase to best achieve research goals while minimizing risk to
any trial participants. Generally, trial
designs fall into the four (or five) phases I summarize in the table below,
where researchers will customize the trial approach and phases to meet the
unique needs of the patient population and condition intended to be treated by
the drug being developed:
Phase |
Purpose and Description |
Typical sizing,
comments |
Preclinical |
Target screening,
described above where teams use in-silico and in-vitro studies to screen
targets. |
Thousands of
molecules, no human participants but potential for cellular or animal
testing. |
0 |
Initial
subtherapeutic evaluation of in-vivo Pharmacokinetics (PK)* and
Pharmacodynamics (PD)* |
Few
participants, probably a number close to 10 or so. |
1 / I |
Dose-ranging studies
to find a broad tolerable range, starting at a low dose before increasing
while monitoring for any progressive or new adverse reactions. This phase often evaluates possible dosage
forms (IV, injectables, tablets, capsules, creams, or other forms) and
focuses on dose tolerability. |
Still small, but more
likely to be tens of people (e.g., perhaps 20, 30, 50 or more), focusing on
healthy volunteers screened appropriately to maximize safety while learning
about adverse reactions. |
2 / II |
Initial efficacy
studies, evaluating how well the drug works, what dosing provides the optimal
balance of therapeutic benefit while reducing risk of adverse events, and any
special considerations around how to take the drug (e.g., with or without
food, time of day, and similar items that later guide how your doctor or
pharmacist tells you to use the medicine) |
Slightly
larger, typically with 100 or more patients with the condition to be treated,
and with screening criteria to limit the potential of harm to those enrolled,
such that some patients will be excluded from participation at this phase. |
3 / III |
Larger scale clinical
efficacy trials, comparing the drug against other established standard
treatment(s) and/or placebo, typically in a blinded fashion where some study
participants receive the established treatment or placebo while others
receive the new drug under study. The
study is designed to allow for conclusive statistical evaluation comparing
the two groups, with a variety of study designs available, and gathers more
information about safety and adverse reactions by including more participants. The final biostatistical analysis will
determine whether the new drug has a risk/benefit profile that can support
its approval as a new therapy. |
Larger, typically with
many hundreds of patients involved, often exceeding 1,000 participants in
multi-center trials. This stage often
includes more exclusion criteria to ensure the study can reach a
statistically valid conclusion without ‘noise’ from extra variables, but
usually allows more patients to participate than in a Phase II study. |
4 / IV |
Post-market surveillance
to further monitor safety through adverse events and reactions. This may include active and ongoing surveillance
of earlier study participants through structured and solicited feedback after
moving to the commercially available approved drug, spontaneous reporting by
other patients or caregivers through established drug safety /
pharmacovigilance reporting channels, and more. |
Any patients receiving
treatment after the drug receives regulatory approval and is launched for
commercial availability in approved markets. |
5 / V |
Real World Evidence
and ongoing research. While
traditional clinical trial phase descriptions end at phase IV, extended
research and meta-analysis is sometimes referred to as Phase V. Instead of focusing on specific study
participants, researchers review data and reports from many available sources
to learn more about ‘real world’ use. |
All use of the drug,
including reports of off-label use, experience in different settings (e.g.,
at home vs. clinical or long-term-care settings), and other published
literature. |
* Think of PK
as the motion (kinetics) of the drug in the body – what your body does to the
drug, typically in terms of Absorption, Distribution, Metabolism and Excretion
(ADME), and PD as the power (dynamics) of the drug in the body – what the drug does
to your body in terms of its biochemical activity and role as an agonist or
antagonist for the appropriate ‘gas and brake pedals’, and the final result in
a measurable outcome.
Researchers must design studies to consider impact on
participants, the ability to include enough participants in studies to reach
meaningful statistical conclusions while accounting for early participation
attrition and other factors, all while ensuring that the studies uphold
established ethical standards of care for participants.
In the case of the Indigo clinical trial for Vorasidenib,
the study was designed by enrolling patients who were already in a “watch and
wait” stage either after or before surgical removal, who had sufficient tumour
volume to be able to monitor through standard MRI imaging, and who had not
already had radiation or chemotherapy that could impact their progression. This allowed researchers to ethically assign
participants to either the Placebo or Treatment arm of the study without adding
risk to patients. Under the study,
participants were monitored by the same serial MRI imaging they would expect,
and those images were used to decide when participants’ cancer progressed to a
point requiring further treatment. Study
researchers collaborated with patient care teams such that patients on the
placebo arm could be moved to the treatment arm or an alternative therapy if
their scans showed the need for further intervention. This design allowed researchers to monitor
PFS and TTNI as primary measurable targets for statistical evaluation, while
also collecting information about patients’ reported quality of life. I remain thankful for the patients before me
who participated in earlier clinical trial phases, including the Phase III
trial that allowed researchers to gather data that demonstrated efficacy
Scaling things up
At the early stages of clinical trial, researchers are
typically working with small-scale trials of new APIs, often developed on lab
benches in things that look like beakers, flasks, and test tubes that many
people recognize as typical in a chemistry lab, and with batch sizes that might
be measured in grams. To scale up to the
larger quantities needed for future commercial manufacturing, industrial
chemists begin the work of designing large scale processes using industrial
sized reactors while evaluating and reducing potential impurities like related
compounds that might be produced as side products or residual solvents or
reactants used in the complex synthetic routes, such that they can scale up to
large batches (many kilograms) of high quality, high purity drug substance to
be later turned into the final drug product to be dispensed by pharmacies upon
approval.
This means that there is one team working on the API, the drug
substance that supplies the pharmaceutical effect – the Ibuprofen in
Advil, and a second team working on the final dosage form, the drug product
that patients will eventually take – the Advil tablet that contains Ibuprofen. Ideally, the teams work in tandem, and adjust
their work as they learn more during progressing clinical trial phases. The drug product development team finds
potential ingredients and formulations typically used for the desired dosage
form, evaluates for any compatibility issues where an ingredient might degrade
the drug substance, and ideally gives feedback to the industrial chemists to
describe the optimal characteristics like particle size, crystal structure, bulk
powder density and flowability and more.
As the drug substance team scales up their synthetic process, it may
change the bulk qualities of the finished batch that impact the drug product team
as they also scale up from something like a ‘test kitchen’ scale batch measured
in grams to the future industrial scale batch in hundreds of kilograms. Both the drug substance development and drug
product development identify the critical processing controls and parameters
and the quality attributes that can be tested for to confirm the final product
is suitable for its intended use. Both
teams work on the Chemistry and Manufacturing Controls (CMC) that identify the
design space in an approach commonly referred to as Quality by Design (QbD) with
a series of experiments and trial batches prepared to fully understand the
processes in making both Drug Substance and Drug Product so that variations in
raw materials and processing steps are fully characterized to ensure repeatable
future full-scale production.
New drug submissions / applications
Historically, a New Drug
Application (NDA) to the US FDA, a New Drug Submission (NDS) to Health Canada, and
similar applications to other global Health Agencies (HAs) were filed as large
document collections typically sent in bankers’ boxes with over 100,000 pages,
but now filed digitally in an electronic Common Technical Document (eCTD)
structure with a similar ‘page count’ now interconnected and managed for regulatory
review.
The submissions are formatted with an overall summary in Module 2 to guide reviewers with a high-level review of what’s ahead in the detailed sections that follow. Module 3 covers all Quality aspects and CMC details for both the Drug Substance and Drug Product, including the development process
from initial trials to future intended commercial production. Module 4 focuses on safety, including non-clinical reports capturing pharmacology details, PK information, Toxicology and more. Module 5 includes detailed reports from the clinical studies demonstrating efficacy and product suitability for its intended use. The structure is detailed by the International Council for Harmonisation (ICH) and summarized in the triangle image product development and regulatory teams around the world are familiar with.When the submission is made to a Health
Agency, the HA assigns resources with the skills necessary to review the
applicable sections and often collaborates with other multidisciplinary groups
for specialized reviews. Health Canada,
for example, works with a pan-Canadian health organization now known as Canada’s
Drug Agency / L’Agence des médicaments du Canada (CDA-AMC). Formerly the Canadian Agency for Drugs and
Technologies in Health (CADTH), it was set up to “[coordinate and align] public
value within Canada’s drug and health technology landscape.”
During review, the Health Agency shares
any identified deficiencies with the company sponsoring the application so that
the Sponsor and the HA can evaluate the science supporting the application and
to either correct deficiencies or supplement the application with new
information where needed. It is a
comprehensive review often with multiple ‘back-and-forth’ interactions with a
goal of assuring patient safety while giving patients access to new therapies
that offer benefit and hope.
During this back-and-forth, the Sponsor
will prepare internally for the commercial production activities they have
already described in Module 3. They will
assess raw materials against their established specifications, dispense and
manufacture initial batches and conduct detailed testing that includes final
specifications and added tests designed to evaluate the success of scaled up
production batches. They time this work
to a predicted positive regulatory decision (approval by the HA) and prepare their
production teams to package and label the final drug product, then to transport
to distribution centers and pharmacies to align with a final approval date.
Specifics of the final approval
will vary by country and Health Agency but may include conditions or
limitations for use by specifying or excluding identified populations through
the indications and contraindications for use, and typically set up pricing and
reimbursement criteria. In Canada, for
example, the Patented Medicine Prices Review Board (PMPRB) reviews and monitors
prices charged by patentees, may pre-approve pricing though pre-approval is not
a requirement, and investigate when prices appear to exceed guidelines.
Bringing this back to Vorasidenib,
Servier submitted their application on February 20th, which began activity
toward an August 20, 2024 decision given the assigned breakthrough therapy
designation. Having completed Phase III clinical
trials, Servier engaged their Managed Access Program allowing expanded access
to patients like me, before full approval, and as part of early data gathering
under Phase IV studies. I see this Expanded
Access under Health Canada’s Special Access Program as a clear demonstration of
how the executive team uses the combined resources at Servier to the benefit of
patients.
The team at Servier is now
preparing their full-scale manufacturing process to make PRVoranigo™
tablets as the drug product containing Vorasidenib as the API / Drug
Substance. Teams are testing the incoming
ingredients, sampling, and testing at various steps in their manufacturing process,
and are packaging the tablets with the necessary protection to maintain shelf
life and the printed material to help patients as they begin therapy, and with
additional information to assist clinicians and pharmacists as this new product
comes to market. This drug is more than
just a new treatment for patients, it is hope for more time and a better
quality of life in the face of a terminal diagnosis. It offers an opportunity to delay the harmful
effects of existing therapy and time to wait for science to progress to the
next new treatment option. I remain
fully optimistic, and with a sense of just how fortunate those of us with Astrocytomas
and/or Oligodendrogliomas are to have this new possibility about to be
available for use, and excitement for what the coming years will offer.
Works Cited
1. Vorasidenib in IDH1- or IDH2-Mutant Low-Grade
Glioma. Mellinghoff, Ingo K., et al. 7, s.l. : Massachusetts
Medical Society, August 17, 2023, New England Journal of Medicine, Vol. 389,
pp. 589 - 601.
2. Servier US | Les
Laboratoires Servier. Expanded Access. Servier US. [Online] 2024.
https://servier.us/expanded-access/.
3. Servier | Les Laboratoires
Servier. Servier - Medicinal Product Managed Access Programs. servier.com.
[Online] 2024.
https://servier.com/en/research-innovation/medicinal-product-managed-access-programs/.
4. Government of
Canada. Health Canada. Health Canada's special access programs:
Overview. [Online] 02 15, 2023.
https://www.canada.ca/en/health-canada/services/drugs-health-products/special-access.html.
5. Government of
Canada . Food and Drug Regulations (C.R.C., c. 870). Justice Laws
Website. [Online] 06 11, 2024. https://laws-lois.justice.gc.ca/eng/regulations/c.r.c.,_c._870/index.html.
6. Pelley, John W.
Citric acid cycle – an overview. Elsevier's Integrated Review Biochemistry.
2nd. Philadelphia : Elsevier, 2012, 7, pp. 57-65.
7. Nobel Prize
Outreach. The Nobel Prize in Physiology or Medicine 1953. The Nobel
Foundation. [Online] 2024. [Cited: 6 17, 2024.]
http://nobelprize.org/nobel_prizes/medicine/laureates/1953/.
8. Cancer-associated
IDH1 mutations produce 2-hydroxyglutarate. Dang, L., White, D., Gross,
S. et al. November 22, 2009, Nature, Vol. 462, pp. 739 - 744.
9. The role of the
NG2 proteoglycan in OPC and CNS network function. Sakry D, Trotter J.
Pt B, Brain Research, Vol. 1638, pp. 161–166.
10. Transformation
of quiescent adult oligodendrocyte precursor cells into malignant glioma
through a multistep reactivation process. Galvao, Rui P., et al.
40, 2014, Proceedings of the National Academy of Sciences of the United States
of America, Vol. 111, p. 201414389.
11. NG2-expressing
glial precursor cells are a new potential oligodendroglioma cell initiating
population in N-ethyl-N-nitrosourea-induced gliomagenesis. Briançon-Marjollet,
Anne, et al. 10, 2010, Carcinogenesis, Vol. 31, pp. 1718 - 1725.
12. The 2021 WHO
Classification of Tumors of the Central Nervous System: a summary. Louis
DN, Perry A, Wesseling P, Brat DJ, Cree IA, Figarella-Branger D, Hawkins C, Ng
HK, Pfister SM, Reifenberger G, Soffietti R, von Deimling A, Ellison DW.
8, Aug 2, 2021, Neuro Oncol., Vol. 23, pp. 1231-1251.
13. Metastatic
brain tumors: diagnosis, treatment, and nursing interventions. Armstrong,
T. S. and Gilbert, M. R. 5, 2000, Clinical Journal of Oncology Nursing,
Vol. 4, pp. 217-225.
14. The 2021 World
Health Organization Classification of Tumors of the Central Nervous System:
What Neuroradiologists Need to Know. A.G. Osborn, D.N. Louis, T.Y.
Poussaint, L.L. Linscott, K.L. Salzman. Jun 2022.
15. International
patterns and trends in the brain cancer incidence and mortality: An
observational study based on the global burden of disease. Ilic, Irena
and Ilic, Milena. 7, s.l. : Cell Press, July 13, 2023, Heliyon, Vol.
9.
16. Cancer Research
UK. Cancer Research UK. Types of brain tumours. [Online] 2024.
https://www.cancerresearchuk.org/about-cancer/brain-tumours/types.
17. American
Association of Neurological Surgeons. Brain Tumors. AANS.org. [Online]
2024.
https://www.aans.org/en/Patients/Neurosurgical-Conditions-and-Treatments/Brain-Tumors.
18. Cleveland
Clinic. ClevelandClinic.org. Oligodendroglioma. [Online] 2024.
https://my.clevelandclinic.org/health/diseases/21191-oligodendroglioma.
19. Zhang, Shijie.
United States Patent. 9,751,863 B2 United States, September 5, 2017.
Therapeutically Active Compounds and their Methods of Use.
20. Servier
Pharmaceuticals, LLC. Press Release. Servier home page. [Online]
Dec 21, 2020.
https://servier.com/en/newsroom/servier-to-acquire-agios-pharmaceuticals-oncology-business/.
21. Brain Tumour
Foundation of Canada. Oligodendroglioma. Brain Tumour Foundation of
Canada. [Online] 2024. https://www.braintumour.ca/brain_tumour_types/oligodendroglioma/.
22. Radiation-induced
brain injury: a review. Greene-Schloesser, Dana, et al. 2012,
Frontiers in Oncology, Vol. 2, pp. 73-73.
23. Alkylating
Agents and DNA Polymerases. Bignold, Leon P. 2, 2006, Anticancer
Research, Vol. 26, pp. 1327-1336.
24. Stryer L, Berg
JM, Tymoczko JL. Biochemistry. s.l. : W.H. Freeman, 2002.
25. Neuro-endocrine
regulation of blood pressure. Chopra, Sandeep, Baby, Chris and Jacob,
Jubbin J. 8, 2011, Indian Journal of Endocrinology and Metabolism, Vol.
15, p. 281.
26. Thyroid hormone
and blood pressure regulation. Danzi, Sara and Klein, Irwin. 6,
2003, Current Hypertension Reports, Vol. 5, pp. 513-520.
27. First-Line
Treatment for Hypertension. Bui, Quynh. 11, 2010, American Family
Physician, Vol. 81, p. 1333.
28. Mifepristone
(RU486) and therapeutic late pregnancy termination: a double-blind study of
two different doses. Frydman, René, et al. 6, 1988, Human
Reproduction, Vol. 3, pp. 803-806.
29. Mifepristone
(RU486): A review. Mahajan, Damodar K. and London, Steve N. 6,
1997, Fertility and Sterility, Vol. 68, pp. 967-976.
30. The many lives
of mifepristone: Multi-glandular exaptation of an endocrine molecule. Magon
N, Chauhan M, Goel P, Ruprai RK. 4, Jul-Aug 2012, Indian J Endocrinol
Metab., Vol. 16, pp. 670-671.
31. Assessing
inhibitors of mutant isocitrate dehydrogenase using a suite of pre-clinical
discovery assays. Urban, Daniel J., et al. 1, 2017, Scientific
Reports, Vol. 7, p. 12758.
32. Laterra, John
J, et al. Blood—Brain Barrier. [Online] 1999. [Cited: 6 19, 2024.]
https://ncbi.nlm.nih.gov/books/nbk28180.
33. Characteristics
of compounds that cross the blood-brain barrier. Banks, William A.
1, 2009, BMC Neurology, Vol. 9, pp. 1-5.
34. Health Canada.
Clinical trials and drug safety. Government of Canada - Canada.ca. [Online]
05 22, 2020. https://www.canada.ca/en/health-canada/services/clinical-trials.html.
35. Friedman,
Lawrence M., et al. Fundamentals of Clinical Trials. 5. s.l. :
Springer, 2015.
36. Early
participant attrition from clinical trials: role of trial design and
logistics:. Siddiqi, Azfar-e-Alam, et al. 4, 2008, Clinical Trials,
Vol. 5, pp. 328-335.
37. Schmuff, Norman
R. and Lin, David T. Chemistry, Manufacturing and Controls (CMC). Wiley
Online Library. [Online] January 10, 2014.
https://onlinelibrary.wiley.com/doi/abs/10.1002/0471667196.ess7246.
38. Barrett, Jeffrey
S. Chemistry, Manufacturing, and Controls. Fundamentals of Drug
Development. s.l. : Wiley, 2022, 16.
39. The Electronic
Common Technical Document. Cartwright, Anthony C. 3, 2006,
International Journal of Pharmaceutical Medicine, Vol. 20, pp. 149-158.
40. Computers in
the new drug application process. JF, Studebaker. 1, Jan - Feb
1993, J Chem Inf Comput Sci, Vol. 33, pp. 86-94.
41. U.S. Food &
Drug Administration (FDA). Electronic Common Technical Document (eCTD). Development
& Approval Process | Drugs. [Online] 03 22, 2023.
https://www.fda.gov/drugs/electronic-regulatory-submission-and-review/electronic-common-technical-document-ectd.
42. International
Council for Harmonisation (ICH). CTD - M4 : The Common Technical Document.
ICH Standards. [Online] 2021. https://www.ich.org/page/ctd.
43. CDA-AMC.
CADTH Is Now Canada’s Drug Agency. Canada's Drug Agency. [Online] May
1, 2024. [Cited: June 19, 2024.]
https://www.cadth.ca/news/cadth-now-canadas-drug-agency.
44. —. The pCODR
Expert Review Committee (pERC). Canada's Drug Agency. [Online] June 17,
2024. [Cited: June 19, 2024.]
https://www.cadth.ca/pcodr-expert-review-committee-perc-0.
45. PMPRB.
Patented Medicine Prices Review Board. Canada.ca | Government of Canada. [Online]
May 22, 2024. [Cited: June 19, 2024.]
https://www.canada.ca/en/patented-medicine-prices-review.html.
46. Patented
Medicine Prices Review Board. Alignment Among Public Formularies In
Canada. National Prescription Drug Utilization Information System
(NPDUIS). Ottawa : PMPRB, 2017. General Overview. ISBN: 978-0-660-09799-2
.
Comments
Post a Comment