Uploaded by AtronsTube

Merged Vaccinology

advertisement
 “Vaccinology and Immunotherapy”
 Dr. Saddam M. (DVM,
Immunologist/Vaccinologist)
 University of Gondar
 June, 2023
Edward Jenner
Used principle of cross protection
Vaccine can be therapeutic for example BCG for TB administered after being diseased,
i.e it is given for sick animals or human.
Regarding vaccination advocacy people can be either of the three categories.
Pro, Hesitant, or anti.
French's are fist level vaccine resistant people in the world. They usually raise
vaccination scandal thinking that government is influenced by billion dollar
companies.
Addressing hesitant would add them to the pro group of Pro.
Hesitant should be addressed with trusted scientific community.
Infant mortality has remarkably decreased by vaccination.
Antivaxors are not supposed to be convinced at all.
Famouse anit vaxor: Andrew Jeremy Wakefield is a British anti-vaccine activist.
In 1998, he and 12 of his colleagues published the paper that the MMR vaccine is the
cause of autism.
Another example is : middle east. Resist the pork gelatin product product in the
vaccine.
In February 2021, the Peruvian 'vaccinegate' scandal broke when the media reported
that nearly 500 experimental doses of an ongoing COVID-19 trial were given to key
individuals not enrolled in the trial.
Scandal: an action or event regarded as morally or legally wrong and causing general
public outrage.
Covid: The first mRNA vaccine licensed
Nucleic acid vaccine have the disadvantage of: genotoxicty, oncogenesis, integrating
to Host DNA.
Vaccine safety study continues life long.
The aim of vaccination is Mimicking the invading pathogen
Malaria was licensed with 37% efficacy.
Purified Antigen can be given as a vaccine. E.g., Purified Human Prostate Specific
Antigen.
 What are Vaccines?
= vaccines are biological preparations that work by
stimulating the immune system and that are administered
to healthy (or sick) animals/humans for the purpose of
prevention (or treatment) of a disease respectively
Pro?
Anti
The discovery of vaccine adjuvants dates back to 1925, where Gaston Ramon showed that co-administration of his
newly invented diphtheria toxoid together with other compounds such as tapioca, lecithin, agar, starch oil,
saponin or breadcrumbs increased antitoxin responses to diphtheria. This was followed up by Glenny and coworkers, who in 1926 demonstrated that diphtheria toxoid precipitated with aluminum salts resulted in significant
increase of the immune response to the toxoid.
The most widely used adjuvant is aluminium salt which was first used by the immunologist, Alexander T. Glenny,
in 1926 at the Wellcome Physiological Research Laboratory in London. In an attempt to purify and concentrate
diphtheria toxoids (inactive toxin), Glenny and colleagues used potassium aluminium sulfate in the production of
the vaccine. Surprisingly, they found that vaccines developed using aluminium salt precipitation led to better
antibody responses in guinea pigs than the soluble toxoids — the first demonstration of aluminium salt
adjuvanticity.
Adjuvant: carrier proteins are not adjuvant, adjuvant have depot effect,
One of the proposed hypotheses of the mechanism behind the immunostimulatory effect obtained with the
cationic liposomal vaccine adjuvant is the ‘depot effect’ in which the liposomal carrier helps to retain the
antigen at the injection site thereby increasing the time of vaccine exposure to the immune cells.
Adjuvants trigger inflammation due to PRR engagement and activation of inflamasome.
Giving an antigen only leads to tolerogenic antigen and autimmunity will occur.
Live attenuated vaccines don’t need adjuvants. It has inherent adjuvanticity.
For immunologically non responsive vaccines(for old and young age) adjuvant will make them good.
Vaccines are the only scientific technology that we apply, or attempt to apply, to every singe human
being on the Earth. There is nothing like that in medicine”
Gregory A. Poland (M.D., MACP, FIDSA) Mayo Clinic – US
“With the exception of safe water, no other medicine or treatment, not even antibiotics, has had such a
major effect on mortality reduction...”
Impact of vaccination
1.
2.
3.
4.
Decreased mortality
Improve productivity
Decreased morbidity
Global financial burden
Immunology of spike protein is identifying the dominant antigen from the desired antigen. E.g., There are four
major structural proteins in the SARS-CoV, including the nucleocapsid, spike, membrane, and small envelope
proteins.
The wider safety margin of a vaccine is because we are giving it to healthy individuals not for sick as drugs.
Vaccines should be affordable
Easy safe and low cost of discovery.
Vaccines should have customized approach
Multiple discount projects are required for vaccine.
After prioritizing the disease vaccine should have product characterization.
Early de risking before production remove safety issues.
Some times disease inhancement make us change the vaccine characterization after production. Eg.
The case of dungue virus
Trying to make vaccine without understanding pathogenesis is fisky
e.g., rota virus vaccine was the cause of intussusception
Standard of care won’t be stopped when we are doing clinical trial.
e.g., if we are giving a vaccine for HIV, we can’t sotp ART.
Introduction — The vaccine specificities
Prophylactic vaccines...
• Are given to healthy individuals
• Are usually administered to very large number of people
• Are needed across the globe and must be affordable to everyone
• May need to protect for a long period of time
• Need to be convenient to administer
• Take significant time and investment to be developed
The vaccine specificities require a customized approach, including at discovery stage
Introduction — Attrition rates in vaccine development
High attrition in the early stage of vaccine discovery
• Multiple Discovery Projects are required to ensure pipeline
sustainability
• Decisiveness is key:
— Asset level
— Portfolio level
• Adopt ways of working in Discovery that ensure flexibility and
sustainability
Introduction — Table of Content
Given the vaccine peculiarities, it is important to optimize the discovery phase:
• Identify & prioritize the right disease target
• Determine early on the mandatory vs desired product characteristics of the future vaccine
=> target product profile (TPP)
• Select the right vaccine approach/technological platform
• Validate the vaccine concept early on =>“ early DE-RISKING”
The TPP is essentially an organized “wish-list” of characteristics, features, and attributes that one would
liketo see in a newly developed medical product once it reaches the market.
IDENTIFICATION AND PRIORITIZATION OF THE DISEASE TARGET
Criteria for selecting & prioritizing new disease targets
1. Medical need, disease burden & economical burden:
a. Disease incidence and severity
b. Disease impact
 Individual vs society
 Short-term vs long-term
 Direct vs indirect
2. Pathogenesis & disease understanding
a. Causality between the pathogen & the disease
b. Factors driving disease severity
3. Protective immune response
4. Technical feasibility
5. Clinical feasibility
Ill. Protective immune response:
• Protective role of antibodies:
• Animal studies
• Protective monoclonal antibodies
• Protective role of maternally derived antibodies
• Protective role of T cells:
• Animal studies
• Disease in patients with allogenic bone marrow transplant
• Immune response associated with disease resolution
• Awareness of prior unsuccessful vaccine trials:
IV. Technical feasibility:
• Is it possible to induce a broad protection against all strains & serotypes of the pathogen
with only a few antigens/vaccine components?
• Is there a vaccine approach that can deliver the desired immune response?
=> Antibodies & CD8 T cell
• For the selected approach, do we think we will be able to scale-up the production process
to produce millions of dose per year?
• Will the cost of goods be compatible with the estimated vaccine price?
V. Clinical feasibility:
a. Is it possible to demonstrate a proof of concept early in the development? Is there a population with increased
disease incidence?
b. Can we show efficacy using a reasonable number of volunteers using an endpoint that would convince
both regulatory agencies & recommending bodies?
c. Is it possible to identify & enroll the necessary number of volunteers in clinical trials? how many countries &
centers?
d. Is there an immunological endpoint that can help us:
1. Measuring vaccine take?
2. Selecting the final dose/formulation?
3. Predicting vaccine efficacy?
What are endpoints when used in a clinical trial?
Biomarkers, are defined characteristics that are measured as indicators of health, disease, or a response to an
exposure or intervention, including therapeutic interventions .
Biomarkers can help diagnose a disease, or predict future disease severity or outcomes, like measurements of blood
pressure as an indicator of cardiovascular risks or measurements of blood sugar in diabetes. Biomarkers also are
used to identify the best treatment for a patient, to monitor the safety of a therapy, or to find out if a treatment is
having the desired effect on the body.
A clinical trial’s “endpoints” are measurements of what happens to people in the trial. When a trial is
intended to evaluate the efficacy and safety of a new medical product or a new use of an approved product, its
endpoints usually measure benefit. Investigators typically use either clinical or surrogate endpoints.
Surrogate endpoints are used when the clinical outcomes might take a very long time to study, or in cases where
the clinical benefit of improving the surrogate endpoint, such as controlling blood pressure, is well understood.
Clinical trials are needed to show that surrogate endpoints can be relied upon to predict, or correlate with, clinical
benefit. Surrogate endpoints that have undergone this testing are called validated surrogate endpoints and these
are accepted by the regulatory body as evidence of benefit.
Found diseased, dead, survival, measuring critical viral load between vaccinated and unvaccinated subjects are the
good end points.
Toxins need antibody mediated vaccine. Subunit vaccine induce antibody response.
Prior unsuccessful vaccine is important to the coming generation of vaccines optimization factors
Identifying T cell epitop.
Bactericidal antibody is
is the immunologic
surrogate of protection
against meningococcal
disease!
Vaccine feasibility check
Volunteer
Reasonable endpoint : to show efficalcy and get license
In meningococcus licensing
For protection and guarantee
•Immunologic endpoint is done in the laboratory
•Clinical endpoint : severity
Bactericidal antibody: bactericidal assay
Virus : cell culture and lesion induction and or lysis
Sweet spot : inclusive
Selecting the right vaccine approach
The RSV Paediatric example
GSK’s approach for RSV paediatric vaccine candidate = Recombinant adenovector
coding for RSV F, N and M2-1 antigens
=> Early control of infection & virus load by:
Neutralizing antibodies directed against the F protein
CD8 T cells directed against F, N & M2.1 to clear infected cells
=> Good quality immune response induced by the adenovector:
Intra-cellular expression of the RSV antigens, as with live RSV virus
Induction of a Th1 or balanced Th1/Th2 immune response
=> Scalability of the platforms demonstrated
Early Project De-Risking
Why?
• Avoid exposing volunteers to an investigational vaccine that is unlikely to offer
significant benefit
• Limit at-risk investment
• Opportunity cost
How?
• Identify what are the key risks of the project
• Design & prioritize experiments assessing those risks
• Stop unsuccessful projects. Accelerate/invest in those for which the major nsk have been
discharged
The RSV Paediatric example
Key risk/question:
• Could the RSV vaccine promote disease enhancement?
Will the vaccine offer protection against the disease?
1.
Existence of multiple serotypes (e.g. rhinovirus, S. suis, FMD, BTV, Pasteurella):
induction of cross-protection.
2.
Search for stronger adjuvant systems
3.
New immune correlates of protection(systems biology approaches)
4.
Antigenic variation(e.g. influenza, Borrelia, HIV)
5.
Failure to induce sufficient CD4 T helper/CTL activity (Tuberculosis)
6.
Complex life cycle of pathogen e.g. Malaria parasite
7.
Virus escape mechanisms (herpesviruses)
8.
Antigenic competition- purely empirical; some vaccines work fine together (e.g.
DPT), others don’t.
9.
Passive (maternal) antibody interference during first weeks/months of newborn
Therapeutic vaccines for cancer and HIV
Infectiou
s
challeng
e
experime
nts
In vivo
product
ion of
organis
ms
Small
market
volum
e
Veterinar
y
Vaccinolo
gy
Less
rigorou
s
regulati
on
Limite
d
Resear
ch
fundin
g
 “Vaccinology and Immunotherapy”
 Dr. Saddam M. (DVM, Vaccinologist)
 University of Gondar
 College of Veterinary Medicine and Animal Sciences
 June, 2023
 Preclinical
 Antigenicity
 Immunogenicity
• Antigen & Adjuvant Justification
Requirements for regulatory Authorities Prior to
First Time in Human Clinical Trials
 Efficacy
 Safety (toxicology)
*preclinical efficacy can be tested when relevant animal model exists for that particular disease!
 Nature of the pathogen in question
 Life cycle, structure, replication…
 Epidemiology of the disease
 Affected age group, geographical distribution (local, regional, pandemic)
 Intended users
 Pregnant, pediatrics, geriatrics…
 In case of animal vaccines–species of animals, purpose of the animal kept
 Technical feasibility
 Clinical feasibility
 Economical feasibility
First lecture
Impact of Pathogen Life cycle on Vaccine Design
Extracellular vs Intracellular
 Extracellular & intracellular pathogens: targeting antigens
on surface of pathogen
 Prevent infection & dissemination by vaccine design
that induces antibodies and CD4 T cells.
• Inactivated pathogen , recombinant proteins etc...
 Intracellular pathogen:
• Eliminate infected cells & prevent dissemination by
vaccine design that induces CD8 and CD4 T cells.
 Attenuated pathogens, live vaccine vectors etc.....
 Conventionally attenuated vaccines
 Genetically/rationally attenuated vaccines
 Whole inactivated vaccines
 (Recombinant) subunit vaccines
 DNA vaccine
 RNA vaccines
Antigen discovery:
• Sequence
based
approach
• HT proteomics
• Peptide libraries
 Virus Like Particles (VLP) based vaccines
 Vectored vaccines: DC-based, viral-vectored
computational
Antigen Selection and Discovery
Antigen Discovery
Reverse vaccinology
• Entire pathogen genome screened using bioinformatics to identify
appropriate vaccine antigen targets.
• Antigen candidates identified in the genome with the specific
attributes required can then be screened in animal models.
Example: Identification of the vaccine candidate for group B
meningococcus vaccine (Pizza et al. science 287 2000)

Conventional laboratory-based, experimental microbiological approaches to antigen
identification typically starts with

the
cultivation of the target pathogenic microorganism under laboratory conditions,
 Attenuate or inactivate the agent OR dissecting

them into their component proteins,
assaying these in some cascade of in vitro and in vivo assays, leading ultimately to the identification of proteins
which display requisite protective immunity

However, it is not always possible to cultivate a particular pathogen outside of the
host organism e.g.????

Many proteins are only
expressed transiently during the course of infection.

Nor are all proteins easily expressed in

Thus, many potential candidate vaccines may be missed
sufficient quantities in vitro
•
Figure 7.1. Scheme representing the classical approach for antigens production
•
(a) and more advanced approaches (b—d) (a) The whole pathogen is isolated and inactivated,
or it is produced in a attenuated version.
•
(b) Recombinant subunit antigens: The molecule responsible for immunogenic response is
identified, its gene is cloned, and the protein is overexpressed and purified.
•
(c) Reverse Vaccinology: Starting from a pathogen's sequenced genome, many potential
antigen candidates are identified, cloned, expressed, and purified.
•
(d) 'tructurg (Vaccinology: This approach is aimed at improving known antigens [obtained
through (b)or following rational design based on the three-dimensional structure of the protein.
REVERSE VACCINOLOGY
The ultimate goal of RV and other discovery techs: to deliver a shortlist of
antigens which can be validated through subsequent laboratory examinations!
• In 2000, a new resource for meningococcal vaccine development
became available with completion of the genome of the virulent MenB
strain MC58, giving in silico access to 2158 predicted ORFs to screen
for novel vaccine antigens.
• Assuming that surface-exposed antigens are the most suitable vaccine
candidates, due to their potential to be recognized by the immune
system, the draft MC58 genome was screened using bioinformatics
tools, leading to the identification of 570 ORFs predicted to encode
either surface-exposed or secreted proteins.
• Antigen selection then continued based on a number of criteria:
• the ability of candidates to be cloned and expressed in E. coli as
recombinant proteins (350 candidates);
• the confirmation of surface exposure by ELISA and FACS analysis (91
candidates);
•
•
the ability of induced antibodies to elicit protective immunity, as measured by
serum bactericidal assay and/or passive protection in infant rats (28
candidates); and
screening to determine the conservation of antigens within a panel of diverse
meningococcal strains, primarily containing disease-associated MenB strains.
Selection of Antigen from Pathogen
Based on existing preclinical or clinical data
•
Different parts of the pathogen can be the target of antibodies or T cells
•
An immune response to 1 or multiple antigens may be needed for
protection
•
The protection against different types or subtypes of pathogens requires
a well conserved antigen.
Identification of protective epitopes by screening a peptide library against sera or PBL from infected
subjects!
 GenOMICS
 TranscriptOMICS
 ProteOMICS
Antigen Discovery
Using proteomics approaches to screen for "new' antigens
• Stage specific expression
• Use infected cells to find vaccine epitopes presented on infected cells
Data mining for vaccine candidates
Advantages
• Access to every antigen
• Non-cultivable can be approached
• Non abundant antigens can be identified
• Antigens not expressed in vitro can be identified
Disadvantages
• Non protein antigens like polysaccharides, glycolipids cannot be used
Vaccine design
Antigen Target +/- Adjuvant?
 Live vaccines
Usually do not require
adjuvants as they mimic
natural infection and have
high immunogenicity
(naturally adjuvanted)
 Inactivated vaccines
May require adjuvants, as
they can lack pathogenic
features of the
microorganism
responsible for triggering
the immune response
Vaccines with purified
or recombinant antigens
Usually require adjuvants,
as they often have low
immunogenicity owing
to a lack of natural
innate immune triggers
Examples of Vaccines with intrinsic Adjuvants
Give examples of Live attenuated veterinary vaccines produced by the NVI
Examples of vaccines combined with adjuvants
Give examples of inactivated or subunit veterinary vaccines
Adjuvant Selection
Considerations
 Vaccine Approach
• Added value , compatible
 Type of immunity required
• Antibodies and/or CD4 and/or CD8
 Population to vaccinate
• Infants, adolescents, elderly
 Healthy or diseased ?
Vaccine design and characterization
In Vitro
Preclinical
RO development
Correlate of Protection (COP) ?
Immunological Parameters that Correlate with Protection
•
COP: infection, disease, transmission
 Antibodies and/or T cells
•
If the immunologic COP is validated: Preclinical development focused on demonstrating the
type of immunity required
•
If no COP identified: Preclinical development required to evaluate multiple immunological
parameters.
 An ICP is a variable immune response that is statistically
associated with protection

Mechanistic correlate of protection (mCoP): directly responsible for protection and

Non-mechanistic correlate of protection (nCoP): that is easy to measure but
which may only be a substitute for an mCoP that is unknown or difficult to measure.
 If an infection is uncommon or deadly, such that an efficacy trial is
not feasible or ethical, a CoP may yet enable licensure of a
vaccine, and once established, a CoP will allow bridging from one
vaccine preparation to another.
 In addition, a CoP may be absolute, i.e. there is a threshold value
above which protection is certain, or
 Relative i.e. higher values are quantitatively more protective than
lower values but there are occasional failures even at high levels and
occasional protection at lower levels.
• Development of Read-outs
 Immunogenicity
 Antibodies (serum, mucosal....)
• Functional: neutralization, opsonization etc..
• Binding antibodies: ELISA
 T cells
• ICS (intracellular cytokine staining), ELISPOT
 Efficacy
 Titration of viral or bacterial load (culture or molecular assays)
 If relevant animal model exists recapitulating the human
pathogen of infection
• Characterization of disease symptoms or pathology.
adaptive immunity
CD8+ T-cell
B-cell
Antibodies
mustration courtesy of GSK
Preclinical
In Vivo Expermentaion
Preclinical Studies
☂ Experimental designs require:
 Justification of immunological read-outs & sample size based
on statistical criteria
 Approval by animal ethics committee
☂ Requirement for good scientific (GSP) or good laboratory
practices (GLP), defined in medical product development
regulations, for preclinical laboratory studies which includes
 Study conduct, personnel, facilities
 Equipment, written protocols, operating procedures, study
reports
 System of quality assurance oversight
Immunogenicity Studies
☂ Characterization and quantification of the immune response induced by the
candidate vaccine antigen.
 Choice of antigen to be justified
• Humoral responses (antibodies).
• Cellular immune ( T cells).
 Choice and justification for the need of an adjuvant
• At minimum compared to unadjuvanted & Alum formulations
☂ Selection of multiple parameters to improve immunogenicity:
 Route of administration
 Number of doses
 Demonstration of dose range effect
 "Relative" dose of vaccine components
☂ Insure consistency of vaccine lots.
Pre-clinical Evidence for HPV Vaccine Design



Animal models (COPV, ROPV...): demonstrate importance of anti-LI antibodies for
protection against infection.
 "Virus Like Particles" VLP shown to be immunogenic
 Important target for viral neutralization
 Induce an efficient immune response capable of protecting mucosa from infection,
following systemic immunization
 Protection against virus challenge following vaccination with L1
• Passive transfer of anti-L1 Ab can protect PV challenged animals
Recombinant L1 protein:
• Major surface protein of HPV
• Self-assemble into Virus Like Particles: Resemble intact viruses
Established a key role for antibodies:
• Induction of neutralizing antibodies against the structural capsid protein L1 should
prevent/reduce HPV infection and associated lesions.
Characteristics of Good Animal Model
•
Prediction the protection in humans
•
Reproduce infection, pathology and/or clinical symptoms
•
Ideally use the same strain of pathogen that infections humans, if not
species specific pathogens.
Model Attributes and Limitations
☂ Human pathogen infection & disease symptoms
• Ex :HBV / chimpanzee, HSV / guinea pig , RSV / cotton rats,
influenza / ferrets, rotavirus /gnotobiotic pis...
☂ Human pathogen infection without disease symptoms
• Ex :B. pertussis / mice, RSV / mouse, influenza / mouse & dengue
virus/ non-human primates....
☂ Non-Human Pathogen (clinical symptoms similar to humans)
• Ex :SHIV or SIV / non-human primates, gpCMV /guinea pig, bRSV
/cow-calves...
Influenza
•
•
•
The main correlate of influenza vaccine efficacy is HI titer: antibodies that block
influenza agglutination.
An HI titer of 1:40 is associated with reduction in the risk of contracting influenza
infection or influenza disease (De Jong 2003)
Both antibody and T cell responses contribute to protection against seasonal influenza
(McElhaney etal., 2009).
Influenza Challenge Models
 Animal models (mice, ferrets, and guinea pigs) provide important
information about vaccine immunogenicity & correlates of
protection.
• Vaccine effects included reduced viral loads in URT and lungs,
lower morbidity and less lung pathology.
 Ferrets most appropriate model for efficacy studies:
 Susceptibility to infection with unadapted human influenza virus
isolates
 Efficiency in transmitting influenza virus
 Clinical signs of disease similar to human influenza disease.
Toxicology Evaluation : Why ?
 Tox studies evaluate the local tolerance and potential systemic toxic effects
following vaccination
 Vaccine = combination of agents (antigen, adjuvants, carrier): potentially
toxic
 Local effects: pain, swelling & redness at the injection site
 Systemic effects: fever, body weight, ophthalmology, clinical and
morphological pathology.
Objective of Toxicology Studies
☂ Demonstrate the candidate has an acceptable safety profile
• Antigen & Adjuvant components
☂ Demonstrate reversibility of any potential effects.
☂ Key Evaluations
• Macroscopic
• Organ weight
• Microscopic observations: Histopathology
• Serological anomalies
• Vaccine Immunogenicity
☂ Adapt clinical study monitoring, if warranted.
• Reproductive Toxicity studies
• Key Evaluations
— Female fertility
— Development of fetus (teratogenic effects)
— Post-natal development
— Vaccine Take
• Required for vaccines intended for women of
childbearing age.
• When:
— Parallel with Phase Ill clinical trials.
— Conducted prior to studies enrolling pregnant women
Thanks!
Questions?
“Vaccinology and Immunotherapy”
Sadam M. (DVM, vaccinologist)
University of Gondar
June, 2023
Early
phase
s:
R&D
Postlicensure
studies:
impact
studies,
safety
studies,
new
indications
Vaccines
Clinical
vaccine
developme
nt:
Phase-I,
Phase-II,
and
Phase-III
Preclinical
developmen
ts: safety,
immunogen
icity &
efficacy
studies

Preliminary trial: a clinical trial that is not intended to serve as a pivotal trial.

They are usually conducted to obtain information on the safety and immunogenicity of candidate
vaccine formulations and to select the formulation(s) and regimen(s) for evaluation in pivotal trials.
They may also serve to inform the design of pivotal trials (e.g. by identifying the most appropriate
populations and endpoints for further study). On occasion, a preliminary trial may provide an initial
evaluation of vaccine efficacy.

New Candidate Vaccine: is a vaccine that is taken in national regulations to be separate and
distinct from other candidate and licensed vaccines. Examples of new candidate vaccines include but
are not limited to:




a vaccine that contains a new antigenic component (i.e. not previously used in a licensed vaccine);
a vaccine that contains a new adjuvant;
a vaccine that contains antigen(s) ± adjuvant(s) not previously combined together in a vaccine;
a vaccine with the same antigenic component(s) ± adjuvant as a licensed vaccine that is PRODUCED
BY A DIFFERENT MANUFACTURER (including situations in which seed lots or bulk antigenic
components used to make a licensed vaccine are supplied to other manufacturers for their own vaccine
production).
 Non-inferiority trial: aim to demonstrate that the test
intervention is not worse than the reference intervention
by more than a small pre-specified amount known as
the non-inferiority margin.
 In non-inferiority trials it is assumed that the reference
intervention has been established to have a significant
clinical effect (against placebo).
 Pharmacovigilance: encompasses the science and
activities relating to the detection, assessment,
understanding and prevention of adverse effects or any
other possible drug-related problems
 Pivotal trials: pivotal clinical trials provide the major
evidence in support of licensure.
 Posology: the vaccine posology for a specific route of
administration and target population includes:
 the dose content and volume delivered per dose;
 the dose regimen (i.e. the number of doses to be given
in the primary series and, if applicable, after the primary
series);
 the dose schedule (i.e. the dose intervals to be adhered
to within the primary series and between the primary
series and any further doses).
 Post-licensure safety surveillance: a system for
monitoring AEFIs in the post-licensure period.

Protocol: a document that states the background, rationale and objectives of the
clinical trial and describes its design, methodology and organization, including
statistical considerations and the conditions under which it is to be performed and
managed. The protocol should be signed and dated by the investigator, the
institution involved and the sponsor.
 Randomization: In its simplest form, randomization is a process by which n
individuals are assigned to test (nT) or control (nC) treatment(s) so that all
possible groups of size n = nT + nC have equal probability of occurring. Thus,
randomization avoids systematic bias in the assignment of treatment.

Responder: a trial subject who develops an immune response (humoral or
cellular) that meets or exceeds A PREDEFINED THRESHOLD VALUE USING A
SPECIFIC ASSAY.

This term may be applied whether or not there is an established ICP and when the
clinical relevance of achieving or exceeding the predefined response is unknown.
 Sponsor: the individual, company, institution or
organization that takes responsibility for the initiation,
management and conduct of a clinical trial.
 The sponsor of a clinical trial may not be the entity that applies
for a license to place the same product on the market and/or
the entity that holds the license (i.e. is responsible for postlicensing safety reporting) in any one jurisdiction.
Superiority trial: a trial with the primary objective of demonstrating that a test group is
superior to a reference group on the basis of the primary endpoint.
 In the context of vaccine development the primary endpoint may be a safety parameter
(e.g. occurrence of a specific type of AE), a clinical condition (e.g. occurrence of a
specific infectious disease) or an immunological parameter (e.g. a measure of the
immune response to one or more antigenic components of the vaccine).

Key Principles of Vaccine Clinical Development
 Common principles guide the development of any
vaccine candidate
 But each vaccine follows a unique developmental path
depending on characteristics such as:
– the type of vaccine (live/killed/subunit/DNA/peptide)
– disease epidemiology
– target population
– and the availability of a pre-existing vaccine
No harm principle
 Vaccines target usually healthy population, often infants or children
 Target population may have specific characteristics
 Vaccine effect can vary e.g. high malaria prevalence, nutrition, health
status
 Vaccine effect can vary because of variation in prevailing disease —
different vaccine may be needed (eg Malaria caused by Plasmodium
falciparum - Plasmodium vivax)
 Vaccine schedule is adapted to population: concomitant medicines or
vaccines, disease pressure
Good Clinical Practice (GCP) guides development
"Good Clinical Practice (GCP) is an international ethical and scientific quality
standard for designing, conducting, recording and reporting trials that involve the
participation of human subjects.
Compliance with this standard provides public assurance that the rights, safety
and well-being of trial subjects are protected, consistent with the principles that
have their origin in the Declaration of Helsinki, and that the clinical trial data are
credible" ICH.org
The 13 principles of GCP
1. Clinical trials should be conducted in accordance with the ethical principles that have
their origin in the Declaration of Helsinki, and that are consistent with GCP and the
applicable regulatory requirement(s).
2. Before a trial is initiated, foreseeable risks and inconveniences should be weighed against
the anticipated benefit for the individual trial subject and society. A trial should be initiated
and continued only if the anticipated benefits justify the risks.
3. The rights, safety, and well-being of the trial subjects are the most important
considerations and should prevail over interests of science and society.
4. The available nonclinical and clinical information on an investigational product should be
adequate to support the proposed clinical trial.
5. Clinical trials should be scientifically sound, and described in a clear, detailed protocol.
6. A trial should be conducted in compliance with the protocol that has received prior
institutional review board (IRB)/independent ehics commtittee (IEC) approval/favourable
opinion.
7. The medical care given to, and medical decisions made on behalf of, subjects
should always be the responsibility of a qualified physician or, when appropriate, of
a qualified dentist.
8. Each individual involved in conducting a trial should be qualified by education,
training, and experience to perform his or her respective task(s).
9. Freely given informed consent should be obtained from every subject prior to
clinical trial participation.
10. All clinical trial information should be recorded, handled, and stored in a way
that allows its accurate reporting, interpretation and verification.
11 . The confidentiality of records that could identify subjects should be protected,
respecting the privacy and confidentiality rules in accordance with the applicable
regulatory requirement(s).
12. Investigational products should be manufactured, handled, and stored in
accordance with applicable good manufacturing practice (GMP). They should be
used in accordance with the approved protocol.
13. Systems with procedures that assure the quality of every aspect of the trial
should bé implemented.
Phases of Vaccine Development
•
•
•
•
Implemented
License
d
Phase 3
Phase 2
Phase 1
Pre-clinical
On average 10-15 years
Highly pyramidal
For each success there are many failures
Failures could occur at any stage:
preclinical, P1, P2, P3, and postlicensure (e.g. Rotashield licensed in
1998 in the US, withdrawn in 1999 for
being associated with intussusception
leading to recollection of all distributed
doses)
This postulate was the first framework for identifying a cause of a disease.
Koch’s postulates are based on inductive reasoning and state that an organism is causal
if:
It is present in abundance in all cases of the disease but not in the absence of the
disease.
It does not occur in another disease as a fortuitous and non-pathogenic parasite,
It is isolated in pure culture from diseased animal, is repeatedly passaged, and induces
the same disease in other animals
Limitations of Koch’s postulates
•Koch’s postulates were too difficult to satisfy because it is a rigid framework for testing
the causal importance of a microorganism.
•It was based on imperfect knowledge of Causation:
MO were assumed to be the sole causes of diseases
Not applicable for non infectious Diseases
Ignored multifactorial etiology & the influence of environmental factors,
As a result, A more cosmopolitan theory of cause was needed.
Steps in Koch’s Postulates
1) The suspected pathogen is found in sick animals but not in healthy ones.
2) The pathogen is able to be grown in a pure culture in the laboratory.
3) The laboratory cultured pathogen is injected into the body of a healthy animal. The
healthy animal develops the same disease that was seen in the first host animal.
4) The pathogen is recovered from the second animal and is the same as the original
pathogen.
This method is the standard for determining the identity of a disease-causing pathogen.
Koch discovered the cause of anthrax (Bacillus anthracis), tuberculosis (Mycobacterium
tuberculosis), and cholera (Vibrio cholerae) using this approach.
 Phase I, first-in-man studies refer to the first administration of a
vaccine candidate to humans.
 The primary objective is to
reactogenicity
evaluate
the
safety
and
 Injection site reaction
 Pain, erythema, swelling
 Systemic reaction
 Fever, anorexia, fatigue, headache, muscle ache, joint pain
 The secondary objective is collection of immune response.
 Often times, the dose, immunization schedule and mode of
vaccine administration (DI, MI, ...) are also assessed.
 First-in-man Phase I studies are usually small trials in healthy,
immunocompetent naïve adults who are at low risk of acquiring a
vaccine-relevant infection (determined by serology, exposure, and
travel history).
 Based on results of adult studies (referred to as Phase Ia trials),
 Subsequent Phase I studies may be conducted in different age or
population groups closer to the target population
 To assess possible differences in dose, safety, vaccine schedule, or
route of administration.
 Such subsequent studies in different geographies and populations are
referred to as Phase Ib.

RTS, S/GSK [The WHO Regional Office for Africa (WHO/AFRO) Malaria Vaccine
Implementation Programme (MVIP)] underwent Phase
I testing first in
malaria-naïve (USA) adults

And then in malaria-exposed
adults (Mozambique, Kenya, Tanzania Africa),

Followed by trials in 1-11-year-olds and finally
in infants residing in malaria-
endemic regions.

Cervarix was tested in 18-30-year-olds
in USA who were seronegative for
HPV DNA to assess the monovalent and bivalent formulations, while another Phase
I/II study was conducted in 18-30- year-old women positive for HPV 16 or HPV 18 DNA.
 Phase I trials are usually
open-label and non-
randomized
 But, it is possible to conduct randomized controlled
trials (RCTs) in which a placebo or a vaccine against a
different disease is used as a comparator.
 To control for bias, such a study can be single-blinded
or double-blinded.
 The practice of using bedside formulations wherein
the vaccine antigen and adjuvant are mixed just prior
to immunization is frequently followed in Phase I trials.
 This can allow the vaccine developer to test more than
one adjuvant with the same vaccine antigen without
having too many vaccine formulations.
 It is recommended that the Phase I study site be located
within or in the vicinity of a tertiary care hospital for
safety reasons
 With Medical doctors trained to GCP in Clinical Trials
 Able to take care of an anaphylactic shock
 After immunization, the need for day-care observation is
guided by the need for monitoring adverse events.
 Tolerability and reactogenicity due to the vaccine or the process of
vaccination is the major safety outcome evaluated in a Phase I
trial.
 To ensure comparability of safety data within and across clinical
trials, it is recommended to follow a standardized approach of
data collection, analysis, and reporting.
 For healthy volunteer vaccine studies, the toxicity grading scales
provided by the USFDA and the case definitions developed by the
Brighton Collaboration for specific solicited events are
recommended as standard references.
 Clinical safety laboratory testing (e.g., hematology, biochemistry,
urinalysis) also forms a part of the safety data that are collected at
baseline, at defined intervals, and at the end of the trial.
 The immunogenicity assays should preferably be validated and





performed under Good Clinical Laboratory Practice (GCLP).
The immunological data can be presented as recommended in
EMA guidelines:
The percentage of “responders” or individuals who “seroconvert” [with 95% confidence interval (CI)].
Responders are either individuals developing an immune
response above a certain threshold level or those who reach a certain
minimum increment in antibody concentration/titer after
vaccination.
These increments may or may not indicate protection.
These criteria should be defined in the protocol prior to study
initiation.

Live attenuated/killed vaccines pose concerns about possible shedding of
infectious agents, transmission to contacts, and a possible reversion to a more
virulent state.

Therefore, volunteers of such Phase I trials require intensive investigations in
closely monitored clinical settings, including evaluation for any clinical signs of
infection.

The extent, route, and duration of shedding vary with the type of vaccine and route
of administration.

Immunocompromised persons should avoid contact with such vaccinees for
a certain time period to avoid contracting an indirect infection.
 A candidate vaccine should proceed to Phase II clinical evaluation after
achieving a satisfactory outcome in Phase I studies in terms of both
safety and immunogenicity.
 The transition from a controlled clinical setting to field evaluation incurs
much greater monetary investment, hence stringent go/no-go
criteria are observed by the developers.
•
•
•
•
•
•
Clinical Trials: Phase Il
POC (Proof of Concept /FeasibiIity)
Age de-escalation: adults adolescents /children /toddlers
/infants
Sample size: 50 — 200 individuals /group
Select and justify final dose and formulation
Further evaluate the safety profile
Might evaluate the most efficient vaccination schedule (Idose, 2-doses ...)
Might evaluate most efficient route of vaccination
The objective is to identify the vaccine preparation, optimal dose, and schedule
to be taken up for confirmatory Phase III trials.
 These studies have the desired statistical power and a defined sample size, to
provide a clinically meaningful outcome on the safety, immunogenicity, and efficacy
endpoints.
 Phase II studies assess the impact of multiple variables on immune response, such
as age, ethnicity, gender, and presence of maternal or pre-existing antibodies (in
infants) and evaluate:
 Age of first administration of vaccine [e.g., during the development of RotarixTM,
the age of first administration varied between countries depending on factors such
as nutrition status, influence of maternal antibodies, and Expanded Program on
Immunization (EPI) schedule].
 Number of vaccine doses (For RotarixTM, two and three doses were studied in
Phase II studies, while for GardasilTM the dosing schedule remained standard
throughout clinical development).
 Sequence or interval between vaccine doses, route of administration, duration of
immunity, potential need for booster immunizations, and qualitative aspects of the
immune response.

 In a Phase II trial of GardasilTM, incidence of persistent
infection associated with HPV 6, HPV 11, HPV 16, or
HPV 18 decreased by 90% in women allocated active
vaccine compared with those allocated placebo over 36
months follow-up.
 During Phase II trials, if an immune correlate of
protection is identified, it facilitates the interpretation of
results in future clinical studies with immune response as
end points.
 Phase II studies recruit hundreds to thousands of subjects
 A large population allows to conclude with confidence that the
vaccine candidate is safe, sufficiently immunogenic, and maybe
protective
 The study population can comprise adults, adolescents, children, infants,
or even pregnant women, depending on the study objective
 However, for a vaccine being developed for infants a step-down
approach is usually followed wherein trials are conducted sequentially in
adults, adolescents, children, and infants.
 Additionally, different populations can be enrolled in different countries
to reduce costs, save time, and still collect meaningful data to be able to
proceed to the next phase of development (The most recent example is of
the vaccine developed for cervical cancer (GardasilTM), in which case
Phase II studies were conducted in both female and male populations
aged 9-25 years.)

In randomized controlled designs, the investigational vaccine is tested against
either a placebo or another vaccine.

These studies are usually conducted in
community-based study sites
where controlled trials are feasible, i.e., in places where information about the
population (demography, migration, sex ratio, disease patterns, etc. and the
pathogen/disease of interest (different strains of pathogen, disease severity and pattern,
seasonality) is available.

Depending on the type of vaccine being studied, the study area can differ (e.g., for
GardasilTM the target population was young adults and adolescents, therefore the
sources of the study population were colleges, universities, and their surrounding
communities, while rotavirus vaccine studies enrolled children and infants from
communities, hospitals, and polyclinics.)
 Phase II studies designed to report partial efficacy of a
vaccine candidate and are conducted in settings of
high incidence of the infectious disease so as to be
able to provide a good readout of the end point.
 For example: two similar Phase IIb studies were
conducted with RotarixTM in Latin America and
Singapore to evaluate the safety, immunogenicity, and
efficacy of different dose concentrations.
 Both studies were placebo-controlled, proceeded in
parallel with almost similar sample sizes, and achieved
good seroconversion rates, but the Latin America trial
also demonstrated protective efficacy against two
serotypes of rotavirus.
 As described for Phase I studies, the humoral and CMI




response to the immunogen(s) in the vaccine candidates
should be evaluated.
The mode of protection of the vaccine guides the
measurement of immune response.
With RotarixTM (oral), serum IgA antibody concentration was
monitored, while in the case of GardasilTM (parenteral), serum
IgG was measured to determine the seroconversion rates
Rarely, Phase II studies can be the definitive study for
licensure with immune markers as outcomes
For example, the meningococcal C conjugate (MCC) vaccine
was licensed on the basis of serological correlates of
protection without efficacy data in United Kingdom
 The type of adverse events (solicited, unsolicited, laboratory) collected
during Phase II trials and the mode of collection (through visits to clinics
and subject diary cards/questionnaires) are similar to Phase I trials.
 However, as Phase II studies are statistically powered and better
designed, they are able to provide meaningful differentiation in terms of
distribution and differences in adverse events between groups.
 In some cases, Phase II data may also provide information regarding
specific adverse events that should be evaluated more carefully in larger
Phase III trials.
 Phase II trials can also provide preliminary information on protective





efficacy through human challenge studies, wherein healthy participants
are deliberately infected with the pathogen.
Such studies are commonly referred to as Phase IIa studies and are
appropriate only for selected diseases wherever it is scientifically and
ethically justified, where the pathogen does not cause lethal infection and is
not resistant to available treatment, and a complete and successful cure
can be obtained.
Human challenge studies have been conducted to test the preliminary
efficacy of vaccine candidates against malaria, typhoid, and cholera.
In case of RTS, S, investigators conducted multiple combined Phase I/IIa
studies to assess preliminary efficacy.
Such studies offer rapid assessment of the usefulness of a vaccine
candidate in a limited number of subjects, thereby preventing the
unnecessary exposure of thousands of individuals, mostly children and
infants, in large Phase II/III trials to a potentially ineffective vaccine.
It thus allows quicker vaccine development and serves as a go/no-go step
for advancing development.
 Pivotal Phase III trials, essential for registration and approval to market of
a vaccine, assess the effect of the final formulation.
 These trials are designed to evaluate efficacy and safety.
 Vaccine Efficacy (VE) is defined as the percent reduction in
incidence (of disease or infection) among the vaccinated.
 If incidence of disease in unvaccinated subjects is “Iu” and in vaccinated
subjects is “Iv”, then the VE is calculated as:
(Iu-Iv/Iu) × 100% = (1- Iv/Iu) × 100% = (1-RR) × 100%
 Iu = incidence in unvaccinated population
Iv = incidence in vaccinated population
 RR = relative risk

 Occurrence of disease is the most common endpoint; however, the
trial may be based on other clinical end points, such as
immunological correlates of protection.
Clinical Trials: Phase Ill
Pivotal Clinical Trials
• Large clinical trials which could involve thousands of individuals
• Demonstrate in target population of Immunogenicity /Efficacy:
 Demonstrate efficacy: vaccine prevents infection /disease
 Demonstrate immunogenicity (e.g. correlate of protection)
• Further evaluate the safety profile large safety database (Health Authorities)
• Co-administration with licensed vaccines /standard of care vaccines
• Manufacturing process evaluation :
 Immunological bridge from lots used in Phase Il (-III) trials to final
commercial scale lots
 Demonstrate consistency of 3 successive lots produced at industrial scale
 Phase III trials are large-scale clinical trials enrolling thousands of subjects
from the target population.
 They are conducted in “field” conditions that are similar to future routine
use.
 Incidence of disease in the study population impacts the sample size:

a low incidence means that large numbers of subjects are required to estimate vaccine
efficacy in comparison to the numbers needed if disease incidence is greater.
 In diseases where an immunological end point correlates with clinical
protection, it can be used as a primary efficacy end point, and smaller
sample sizes often suffice

RCTs are considered the “gold standard,” where participants are randomly
allocated to receive either the investigational or the control vaccine (placebo, different
vaccine, or nothing).

A prospective RCT controls variables, prevents bias, and maximizes the chances of
detecting a difference between the investigational vaccine and control.

Superiority trial designs are employed if there is currently no effective vaccine for
the disease

For comparisons against existing vaccines, a non inferiority trial is planned to
demonstrate that the relative risk of disease or infection with the new vaccine is not
greater than the available vaccine
 Vaccine efficacy can also be studied through group randomized trials.
 Such group/cluster randomized trials study the indirect protection
(herd immunity) offered by the vaccine in a community
 However, for licensure studies, individual randomization studies are
preferred because if the product is not giving any direct protection, it is
unlikely to have any indirect effect.
 Further, safety evaluations are difficult to conduct in cluster
randomized trials
 Secondary attack rate study or household contact
study (can be randomized):

These are pre-exposure cohort trials for infections with a high secondary attack rate

The unit of intervention may be an individual, family, or community

The indirect effect is the difference in the outcome in an unvaccinated individual when
living in a vaccinated community or living in a comparable unvaccinated community

Such clinical trials are not done for vaccine licensure as yet but may become
common in the near future
 Observational cohort studies
 May be considered where:
 a RCT is not ethically justified
 the clinical end point requires long- term follow-up
(e.g., hepatitis B vaccination in neonates)
 the number of individuals is too large to follow up
 It should be of importance to public health and should also be
clinically important.
 Often the evaluation of protective efficacy focuses on the
ability of the vaccine to prevent clinical disease!
 However, if an organism causes a range of infections (e.g.,
from mild infection to severe disease), then the primary end
point can be carefully selected in accordance with the
proposed indication.
Ways to demonstrate
efficacy
 Clinical endpoint
 Immune response
endpoint (ICP)
 Animal models

Vaccine efficacy is normally assessed on the basis of a single end point (e.g.,
severe gastroenteritis in Phase III study of RotarixTM)

However, a composite efficacy end point can also be studied : e.g., incidence of
genital warts, vulvar or vaginal cervical intraepithelial neoplasia (CIN), or cancer, or
cancer associated with HPV type 6, 11, 16, or 18 were composite end points for
evaluating GardasilTM.

While selecting an end point, it is important to note that the more serious the end
point, the lower is its incidence, which in turn lowers the probability of studying the
impact of a vaccine

The primary end point should be carefully chosen because that will generally
determine the size of the trial

The remaining outcomes can be studied as secondary endpoints.

To accurately identify the end points in clinical protection studies, confirmation of
cases through laboratory methods, antigenic detection, and the clinical picture
is necessary to support a clinical case definition

Case definitions for the trial endpoints should be clearly defined in the protocol e.g., during Phase III study of GardasilTM, investigators were given standard
protocols for anogenital examination, conducting colposcopy, and classifying
cervical samples on a standard scale

Similarly, for the RotarixTM trial, standard case definitions were used for defining an
episode of severe gastroenteritis

The validity of the diagnosis is most important for evaluation of efficacy or safety

If no well-validated methods for establishing infection and/or progression of infection exist
during the period of prelicensure clinical development, then experimental laboratory
methods can be used

The sensitivity, specificity, and reproducibility of the methods used to ascertain a case are
included in the study reports

In clinical trials where prevention of disease is used as an endpoint, considerable efforts
should be made to establish the immunological correlate of protection (e.g. HVB)

To determine immunogenicity, serological data are usually collected from a subset of the
immunized population at predefined intervals and from persons classifiable as vaccine
failures
 In the prelicensure studies, it is recommended to enroll approximately
3000 subjects.
 However, where Phase III studies are designed with safety as the
primary end point, huge sample sizes are seen, e.g., >37000 subjects
were enrolled in a 7-valent pneumococcal conjugate vaccine study, while
for Rotarix™ and RotaTeq™ ~63,000 and ~70,000 infant subjects,
respectively, were assessed to detect the risk of intussusception.

“Phase IV trial” or “post-marketing surveillance study (PMS)”

Phase IV trials involve the safety surveillance (pharmacovigilance) over a much larger patient
population and a longer time period.

Assesses effectiveness of vaccination programs

Assessment whether the vaccine works under normal/field use.

Phase IV studies may also be REQUIRED by regulatory authorities,


For example, on certain population groups such as pregnant women, HIV (drug interactions), ...or

May be undertaken by the sponsoring company for finding a new market for the drug/vaccine
Harmful effects discovered by Phase IV trials may result in a drug or vaccine being no
longer sold, or restricted to certain uses (E.g. Rotashield® and the recent
Dengvaxia®)
Clinical Trials: Phase IV
Post-Authorization Clinical Trials
• Trials performed post-licensure could be:
 Requested by Regulatory Authorities requests as
conditional
for vaccine's approval -Y Regulatory Commitments
 Performed for label updates (e.g. age extension, coadministrations, long-term antibodies persistence, new
target populations ...)
 Unlike drugs, which are given to patients, vaccines are received by




healthy individuals, thus the safety margin should be very high
As vaccines have to be stored under refrigeration, there are always
logistical challenges during clinical trials considering that Phase II and
Phase III are “field studies”
As healthy children also receive immunization under the national
program, the trial design gets complicated due to the possibility of
interference during co-immunization
The compatibility of the adjuvant with the vaccine antigen and the
quality and stability evaluation of antigen/adjuvant formulation are
important aspects of clinical development.
The immune response primarily measured during early stages of
vaccine development (Phase I/II) should evaluate: amount, class,
subclass, and function of each specific antibody
 Relationship between functional and nonfunctional antibody assays
 Kinetics of immune response such as lag time for onset, antibody





persistence, seroconversion rate, and induction of immune memory
Components of the immune response according to mode of delivery
[whether immunoglobulin A (IgA) or immunoglobulin G (IgG)]
Quality of the antibody response: specificity and/or epitope recognition and
avidity
Potential for formation of cross-reactive antibodies or immune complexes
Immunological factors that might affect the humoral immune response as
preexisting antibodies (including maternal antibodies).
Cell-mediated immune (CMI) response and the possibility of immune
interference and/or cross-reacting immune responses when vaccines
containing more than one antigen or two or more vaccines are coadministered, especially to children and young infants with immature immune
systems
 These studies are conducted in healthy individuals, thus,





SAFETY remains to be the utmost priority through out the
development
The disease endpoints (preventable infectious disease for
e.g.) could be so rare, for this reason, pivotal studies are
very large (involving thousands of subjects)
Complexity of enrolling subjects (why?)
Requirement of cold chain maintenance
The studies are community-based rather than hospitalbased
Long manufacturing lead times for clinical trial lots
Vaccines HAVE to be safe!
• The most effective tool for improving public health
• Given to healthy individuals (from babies to elderly)
• No immediate obvious health benefit for vaccinated individual
• A high standard of safety is expected
• Acceptable risk is minimal
 Approval stage
 Pre-clinical and clinical data to support approval
 Post-clinical phase: Phase 4: Effectiveness of the vaccination
program
 Post marketing surveillance and program evaluation
 Assessment whether the vaccine works under field condition while
phase 3 involves clinical trial to measure efficacy under controlled
setting
Vaccine Development Process: summary
Early Stages
SAFETY
SAFETY
Efficacy
Effectiveness
Development,
Evaluation, & Validation
Post-licensure
PROGRAM
IMPLEMENTATION
 Antigen
identification &
discovery
 Adjuvant discovery
& selection
 Ag/adj. Physical &
Chemical
characterization
 Formulation
PRECLINICAL
STUDIES
CLINICAL
STUDIES
 Pharmacokinetic
s
 Safety/Toxicity
Studies
 Phase-I to III
 Mechanisms of
action
 Immunogenicit
y studies
 Efficacy
studies
 Discussion
with regulatory
bodies
 Filing
application
and
Registration
 Post-marketing
surveillance
(Phase IV)
 Impact studies
Thanks!
Questions??
Download