2018 NIAID
STRATEGIC PLAN
FOR
RESEARCH
ON
VACCINE
ADJUVANTS
NIAID
National Institute of Allergy and Infectious Diseases
2
TABLE OF CONTENTS
1. INTRODUCTION ................................................................................................................... 3
1.1. BRIEF OVERVIEW OF ADJUVANTS IN LICENSED VACCINES ........................... 3
1.2. THE NEED FOR NEW VACCINE ADJUVANTS
1.3. NIAID’S MISSION AND EXTRAMURAL FOCUS ON VACCINE ADJUVANT
........................................................ 4
RESEARCH ............................................................................................................................ 4
1.4. NIAID’S STRATEGIC PLAN FOR RESEARCH ON VACCINE ADJUVANTS ....... 5
2. FUNDAMENTAL IMMUNOLOGY AND ADJUVANT DISCOVERY ............................. 8
2.1. RECENT PROGRESS IN NIAID-SUPPORTED ACTIVITIES .................................... 8
2.2. PROGRAM GOALS AND RESEARCH CHALLENGES ........................................... 12
2.3. NIAID’S STRATEGY TO ADVANCE ADJUVANT DISCOVERY .......................... 13
3. ADJUVANT DEVELOPMENT AND PRECLINICAL EVALUATION ........................... 16
3.1. RECENT PROGRESS IN NIAID-SUPPORTED ACTIVITIES .................................. 16
3.2. PROGRAM GOALS AND RESEARCH CHALLENGES .. ......................................... 19
3.3. NIAID’S APPROACH TO ADVANCE ADJUVANT DEVELOPMENT AND
PRECLINICAL EVALUATION .......................................................................................... 21
4. CLINICAL EVALUATION OF ADJUVANTED VACCINES ........................................... 23
4.1. RECENT PROGRESS IN NIAID-SUPPORTED ACTIVITIES ............................ 23
4.2. PROGRAM GOALS AND RESEARCH CHALLENGES ........................................... 25
4.3. NIAID’S APPROACH TO ADVANCE THE CLINICAL EVALUATION OF
ADJUVANTED VACCINES ............................................................................................... 27
5. SUMMARY .......................................................................................................................... 28
6. APPENDICES ....................................................................................................................... 29
6.1. APPENDIX A: ABBREVIATIONS.............................................................................. 29
6.2. APPENDIX B: BLUE RIBBON PANEL MEMBERS ................................................. 31
6.3. APPENDIX C: NIAID-SPONSORED CLINICAL TRIALS FOR NON-HIV
ADJUVANTED VACCINES, RECENTLY COMPLETED OR ONGOING (AS OF APRIL
2018) ...................................................................................................................................... 33
6.4. APPENDIX D: NIAID-SPONSORED CLINICAL TRIALS FOR HIV ADJUVANTED
VACCINES, COMPLETED OR ONGOING (AS OF APRIL 2018) .................................. 34
3
1. INTRODUCTION
1.1. BRIEF OVERVIEW OF ADJUVANTS IN LICENSED VACCINES
The use of adjuvants began in 1920 when the French veterinarian Gaston Ramon
discovered that co-administration of inactivated diphtheria toxoid with starch,
breadcrumbs, or other substances led to an increase in antitoxin responses to diphtheria. In
1926, this finding was followed by Alexander Glenny’s discovery of the adjuvanticity of
aluminum salts and their clinical utility in diphtheria immunization.
Until 2009, aluminum-based adjuvants, collectively termed alum,were the only
adjuvants in vaccines licensed for use in the United States. That year, the U.S. Food and
Drug Administration (FDA) licensed the first of five vaccines containing novel adjuvants,
which are described below and summarized in Table 1:
Cervarix
®
, a human papillomavirus (HPV) vaccine containing AS04, a combination of
alum and 3-O-desacyl-4'-monophosphoryl lipid A (MPL), an immune-stimulating lipid
developed by GlaxoSmithKline (GSK). In the United States, Cervarix
®
is approved for
use in females 10 through 25 years of age to prevent infection from HPV types 16 and
18, which cause about 70% of cervical cancer cases.
Q-Pan H5N1, a pandemic H5N1 influenza vaccine containing AS03, an oil-in-water
emulsion combination adjuvant developed by GSK. This vaccine is part of the U.S.
vaccine stockpile but is not commercially available. It is intended for use in people 18
years of age and older.
FLUAD
®
, an influenza vaccine containing MF59
®
, a squalene-based oil-in-water
emulsion adjuvant developed by Novartis, for use in people 65 years of age and older.
FLUAD
®
was first licensed in Italy in 1997 and is now used as a pediatric and adult
influenza vaccine in more than 30 countries.
Shingrix
®
, a shingles (herpes zoster) vaccine containing AS01, a liposome-based
combination adjuvant containing MPL and the saponin QS-21, developed by GSK.
Shingrix
®
is for use in adults 50 years of age and older.
Heplisav-B
®
, a hepatitis B vaccine containing CpG-oligodeoxynucleotide (ODN), a
short, single-stranded DNA molecule that mimics unmethylated bacterial DNA and
triggers innate immune responses through activation of Toll-like receptor 9 (TLR9),
developed by Dynavax. This vaccine is for use in people 18 years of age and older.
Table 1. FDA-approved vaccines containing novel adjuvants (2009-2018)
Year Adjuvant Vaccine Name
2009 AS04 Cervarix
®
2013 AS03 Q-Pan H5N1
2015 MF59
®
FLUAD
®
2017 AS01B Shingrix
®
2017 CpG-oligodeoxynucleotide Heplisav-B
®
4
1.2. THE NEED FOR NEW VACCINE ADJUVANTS
Live-attenuated vaccines and some vaccines based on inactivated or killed agents do not
require exogenous adjuvants, because the vaccines contain their own natural/endogenous
adjuvants that can stimulate both the innate and adaptive arms of the immune system.
However, subunit vaccines, which contain specific purified molecular entities, generally
lack immune stimulators found in whole organism–based vaccines, leading to a need for
exogenous adjuvants to induce protective immunity and desirable vaccine efficacy.
Unfortunately, not all adjuvanted vaccines induce the types of immune responses required
for durable protective immunity. New or improved adjuvants may enhance the safety and
efficacy of current vaccines, and they also are needed to improve vaccine efficacy in at-risk
populations such as neonates, young children, pregnant women, the immunocompromised,
and the elderly. Finally, vaccines do not exist for many known and newly emerging
infectious agents, and the availability of new adjuvants, along with an understanding of
their mode of action, may facilitate development of such vaccines.
1.3. NIAID’S MISSION AND EXTRAMURAL FOCUS ON VACCINE ADJUVANT
RESEARCH
The overall mission of the National Institute of Allergy and Infectious Diseases (NIAID),
part of the National Institutes of Health (NIH), is to improve human health by supporting
research to understand, treat, and prevent infectious and immune-mediated diseases.
Critical components of NIAIDs research agenda in support of this mission are the
discovery and development of adjuvants, traditionally defined as vaccine components that
augment and target immune responses.
NIAIDs three extramural divisionsthe Division of Allergy, Immunology, and
Transplantation (DAIT); the Division of Microbiology and Infectious Diseases (DMID);
and the Division of AIDS (DAIDS)—all support aspects of adjuvant research that are
described in detail in Sections 2, 3, and 4. Briefly, the mission of each division and its
adjuvant research focus is described below.
DAIT supports basic and clinical research to understand immune system development;
define the mechanisms governing immune system function across the lifespan, providing
knowledge that can be applied to develop and improve treatment and prevention strategies
for inducing immunity against pathogenic infections; and develop better diagnostic, treatment,
and prevention strategies for immune-mediated diseases and the rejection of transplanted
organs. DAIT adjuvant research focuses on fundamental immunology, adjuvant discovery, and
early-stage development. The latter includes compound screening, determination of adjuvant
mechanisms of action, lead compound optimization and formulation, efficacy testing in
animals, and investigational new drug (IND)–enabling studies. Adjuvant science programs
supported by DAIT began in 2004 and include the Adjuvant Discovery Program, the
Adjuvant Development Program, and the Molecular Mechanisms of Combination
Adjuvants Program. Through these programs, more than 2 million compounds have been
screened, and numerous adjuvant candidates, encompassing eight different adjuvant types
5
or classes, are being further developed. Other programs invested in characterizing immune
responses to adjuvants include the NIAID Systems Approach to Immunity and
Inflammation Program and the Human Immunology Project Consortium (HIPC).
DMID supports basic and applied research to control and prevent diseases caused by
virtually all human infectious agents except HIV. The DMID portfolio includes a broad
range of research from early discovery through product development for vaccines,
therapeutics, and diagnostics, including novel vaccines (adjuvanted and unadjuvanted) and
platform technologies. Currently, DMID is testing more than 100 different adjuvant
candidates/formulations encompassing all stages of vaccine development. DMID also
offers a comprehensive suite of preclinical services for the development and evaluation of
adjuvanted and unadjuvanted vaccines.
DAIDS supports a comprehensive research portfolio to advance biological knowledge of
HIV/AIDS and its related coinfections and comorbidities. The ultimate goal is to halt the
spread of HIV through the development and implementation of an effective vaccine and
biomedical prevention strategies that are safe and desirable. The adjuvants used in HIV
vaccine candidates, as of April 2018, are listed in Appendix D.
1.4. NIAIDS STRATEGIC PLAN FOR RESEARCH ON VACCINE ADJUVANTS
As technologic and scientific advances increased our understanding of the pathways
regulating innate and adaptive immune responses, NIAID recognized opportunities to
develop novel adjuvants or modify existing adjuvants to safely trigger long-term protective
immunity and durable vaccine efficacy. In 2010, NIAID prepared its first Strategic Plan for
Research on Vaccine Adjuvants and convened an Adjuvant Blue Ribbon Panel (BRP) to
review the plan.
The 2010 BRP made the following recommendations:
Support additional research to understand how new and existing adjuvants and
combinations of adjuvants enhance vaccine-specific immune protection
Foster discovery and advancement of promising adjuvant candidates through
optimization and preclinical testing stages
Develop/improve and standardize animal models to evaluate adjuvant safety and
efficacy, including development/use of animal models that more faithfully reflect
immune responses of neonates/young children and the elderly
Expand/standardize reagents for adjuvant discovery and development, including
pathogen-specific reagents, and immunologic reagents for animals beyond traditional
mouse models
Identify more accurate predictors of adjuvant efficacy and/or reactogenicity
Determine the effects of formulation on adjuvant mechanisms of action and safety and
effectiveness of adjuvanted vaccines
Between 2010 and 2018, NIAID’s extramural adjuvant discovery and development
research programs have implemented these recommendations. Because of significant
6
advances in adjuvant science, many of which were driven by NIAID-supported programs,
NIAID has updated its 2010 adjuvant research agenda, culminating in the 2018 Strategic
Plan, which focuses on three areas:
Fundamental immunology and adjuvant discovery
Adjuvant development and preclinical evaluation
Clinical evaluation of adjuvanted vaccines
NIAID convened a second BRP on April 23-24, 2018, to assess this Plan and provide
insights and recommendations, which are included in appropriate sections of the Plan and
summarized in Table 2.
Table 2. 2018 Blue Ribbon Panel Recommendations
Fundamental Immunology and Adjuvant Discovery
Fundamental Immunology
Expand fundamental mechanistic studies to:
o Identify innate correlates of adaptive immunity, including correlates of both protection
and persistent immunity, for healthy and at-risk populations
o Determine how antigen selection, including antigen processing/presentation
mechanisms, impacts adjuvant functionality
Adjuvant Discovery
Expand discovery efforts beyond TLRs, to include adjuvants that trigger
inflammasome components, target specific immune cells, or mimic immunologic
outcomes of natural infections that induce long-term protective immunity
Foster precision adjuvant discovery by first determining clinical/immunologic needs,
followed by designing a discovery process to identify compounds that meet those needs
Support development of functional screening methods that better mimic in vivo responses,
taking into account human variability and at-risk populations
Compare/optimize adjuvants for specific cell targeting and delivery routes
Encourage development of standard operating procedures and best practices that maximize
screening efforts and data sharing for cross-study analyses and benchmarking to licensed
products
Support central core services to further improve program integration: formulation
assistance, production of standardized reference antigens, and animal testing (multiple
models) for head-to-head adjuvant comparisons
Emphasize importance of considering formulation at the early stages of adjuvant discovery
7
Adjuvant Development and Preclinical Evaluation
Explore iterative testing of candidate adjuvants with a broad panel of antigens to identify
the most promising combinations for further development
Establish a public database/portal that describes the characteristics, functionality, and
safety profiles of novel compounds developed in NIAID’s adjuvant programs, including
links to publications and clinical trials information, and procedures for requesting access to
these adjuvants
Support studies that develop methods to extrapolate adjuvant outcomes seen with one
antigen to additional antigens
Expand support of current Good Manufacturing Practice (cGMP) capabilities to support
small-scale cGMP production of promising adjuvants for clinical trials
Clinical Evaluation of Adjuvanted Vaccines
Support clinical trials of novel adjuvants and expand clinical comparisons of different
adjuvants formulated with the same antigen
Support clinical trials of adjuvanted vaccines in at-risk populations (e.g., elderly, children)
Support comprehensive immunological analyses of samples from clinical trials to generate
detailed immune profiles of adjuvant functionality
Increase use of human challenge models, where applicable, to better assess the efficacy of
adjuvanted vaccines
Ensure at least a 12-month post-vaccination assessment for safety and immune durability
8
2. FUNDAMENTAL IMMUNOLOGY AND ADJUVANT DISCOVERY
2.1. RECENT PROGRESS IN NIAID-SUPPORTED ACTIVITIES
Fundamental Immunology
Fundamental immunology research supported by NIAID’s portfolio of investigator-
initiated grants and solicited research programs has identified many novel innate immune
molecules and increased our understanding of innate and adaptive immunity. For example,
investigator-initiated studies have led to the discovery of pathogen-associated molecular
patterns (PAMPs), which trigger innate immune responses and are the basis of many
adjuvants (e.g., they mimic PAMP-induced immune function). More recently, NIAID-
supported investigators have determined that live/viable bacteria contain a unique class of
PAMPs, termed vita-PAMPs, which are bacterial messenger RNAs (mRNAs) and cyclic
di-adenosine monophosphate (c-di-AMP) that serve as signatures of viability and trigger
specific innate immune responses. Similarly, investigators supported by one of NIAID’s
solicited research programs, Systems Approach to Immunity and Inflammation, have:
Identified novel functions for more than 260 genes, many of which provide insights
into signaling pathways triggered by innate immune receptors and will aid in
mechanism-of-action studies of novel candidate adjuvants
Developed a suite of ENU-mutant mice (https://mutagenetix.utsouthwestern.edu/) and
Collaborative Cross (CC) mice (http://csbio.unc.edu/CCstatus/index.py) that can be
used to confirm or determine an adjuvant’s mechanism of action. Mice from both
resources can be used in various ways to identify immune receptors or signaling
pathways responsible for adjuvant function.
Novel animal models also have been developed through investigator-initiated research
projects. For example, researchers at the University of Minnesota have developed the dirty
mouse” model by determining that the microbiome of pet store mice dramatically impacts
immune system function. Dirty mice exhibit immune functionality more reflective of adult
human responses, while immune function in laboratory mice is more similar to that of
human infants. Co-housing the pet store mice with laboratory mice increases the
microbiome diversity of the laboratory mice and changes their immune functionality to that
of the pet store mice (e.g., more similar to adult human responses).
Adjuvant Discovery
NIAID began its first solicited program in adjuvant science in 2003 with the Adjuvant
Discovery Program. This contract program supported the identification and
characterization of novel, effective, and safe vaccine adjuvants. The scientific scope of this
program included:
Use of high-throughput screening methods for adjuvant identification
9
Validation of adjuvant activity in human cells
Optimization of lead adjuvant candidates through formulation and medicinal chemistry
guided by structure–activity relationship (SAR) studies
Mechanism of action studies
Verification of adjuvanticity in animal models
The program was renewed in 2009. Building on knowledge gained from the 2003 and 2009
programs and to address recommendations made by the 2010 Adjuvant BRP, the 2014
program renewal emphasized discovery of adjuvants (1) for use in at-risk populations and
(2) that induced mucosal immune responses. Recognizing the limitations of a program that
renews only every five years, and following the recommendations of the 2010 Adjuvant
Blue Ribbon Panel to increase the investment in adjuvant research, in 2017, NIAID began
using the annual Small Business Innovation Research (SBIR) contract solicitation to
support additional adjuvant discovery activities. This program, while new, already has
expanded the investigator pool and increased the diversity of adjuvant candidates for the
development pipeline.
Since its inception, researchers funded by NIAID’s Adjuvant Discovery Program have
screened more than 2 million compounds in vitro and more than 10
24
in silico and
identified a wide variety of promising adjuvant candidates, including:
Synthetic TLR2, 4, 5, 7, 8, 7/8, and 9 agonists; nanoemulsions (for mucosal
delivery)
C-type lectin receptor (CLR) agonists
CD1d ligands (natural killer T [NKT] cell inducers)
Synthetic RIG-I agonists
Carbohydrate-based adjuvants
Figure 1 shows representative immune receptors and pathways targeted by compounds and
formulations that have resulted from the NIAID Adjuvant Discovery Program.
10
Fig. 1
Figure 1: Vaccine adjuvants from NIAID’s adjuvant discovery and adjuvant
development programs target a variety of cellular receptors and signaling pathways.
Some of these adjuvants (solid arrows) engage cell-surface and intracellular RIG-I-like
receptors and C-type lectin receptors. In addition, adjuvants have been developed that
directly act on signaling molecules of these receptors, act by rearranging lipid rafts and
associated receptors, or provide adjuvanticity through yet undefined mechanisms
(dashed-line arrows).
Specific accomplishments of these programs include identification of the following
molecules with adjuvant activity, including:
Novel CpG-ODNs that exhibit higher adjuvanticity than previously published CpG-
ODN and are not species specific (e.g., able to trigger both mouse and human TLR9).
One of these novel CpG-ODNs, originally identified through in silico screening, is now
part of an inulin-CpG combination adjuvant that has been used in two clinical trials of
influenza vaccines.
Co-adjuvants in the form of small molecules that enhance the activity of an adjuvant
but do not exhibit intrinsic adjuvant activity themselves. Such co-adjuvants have been
shown to extend NF-KB signaling in antigen-presenting cells (APCs) and thus enhance
lymphocyte activation.
Novel oxoadenine-based TLR7/8 agonists with high potency and low reactogenicity
11
Amphotericin B as a novel TLR2/TLR4 agonist with adjuvant activity and low
reactogenicity
Adjuvant formulations that target adjuvant activity to specific APCs or lymph nodes,
reducing reactogenicity of potent imidazoquinoline (IMDQ)-based TLR7 agonists, such
as:
o Lysophospholipid conjugates of IMDQ
o IMDQ-ligated nanogel
o Hyaluronic-acid conjugated IMDQ
First small-molecule TLR4 agonist, which is currently being developed as a
combination adjuvant with TLR7 agonists for influenza vaccines
Novel RIG-I agonists, as follows:
o A small molecule compound called KIN, currently being developed for vaccines
against emerging RNA viruses, including pandemic influenza and Zika
o Synthetic, Sendai-virus-derived double-strand RNA (dsRNA) adjuvant for
intranasal immunization against influenza
Nanoemulsions (NEs) for intranasal or intramuscular injection, which act as delivery
platform and adjuvant. NEs inactivate viruses and bacteria and can be used to produce
killed, whole organism adjuvanted vaccines. Intranasal formulations can elicit strong
Th1/Th17 responses upon boost with NE-adjuvanted vaccine, overcoming preexisting
antigen-specific Th2 dominance. This approach is being developed for a recombinant
influenza vaccine and a B. pertussis vaccine.
Selective TLR8-agonist (methylpyrimidine-diamine) without TLR7 activity and ultra-
low inflammatory profile
Dendrimeric IMDQ construct with pure TLR7 activity, which has low reactogenicity
and enhanced adjuvanticity (compared to monomeric IMDQ) and promotes antibody
affinity maturation and epitope spreading
First chemokine receptor CCR1 agonist adjuvant (bis-quinoline)
NOD-1 agonist adjuvant (glutamyldiaminopimelic acid-derivative)
Novel water-soluble TLR2 agonist
Synthetic CRX compounds (MPL-derivatives), which are TLR4 agonists with high
safety profiles. Several of these compounds were developed as adjuvants for
intramuscular delivery and sublingual formulation (combination adjuvant with
methylglycol chitosan) for influenza vaccines. They also are being developed as
combination adjuvants with novel TLR7/8 agonists.
Protein cage nanoparticles (PCN) for mucosal delivery to induce tertiary lymphatic
tissue
The Adjuvant Discovery Program has also identified molecules with valuable, non-
adjuvant properties that include:
Novel antimicrobials
o Antivirals against Zika and dengue viruses
o Antibiotics such as a modified TLR7/8 agonist without TLR activity, which acts as
a potent inhibitor of Gram-positive bacteria, including methicillin-resistant
Staphylococcus aureus (MRSA)
o Pure TLR7 agonists currently being evaluated as HIV latency-reversal agents
12
Large libraries of TLR7, TLR8, and TLR7/8 agonists that modulate immune responses
and can be used as tools to study TLR7 and TLR8 signaling pathways
2.2. PROGRAM GOALS AND RESEARCH CHALLENGES
Goal 1: Strengthen Immunology Research
The most significant advances in NIAIDs Adjuvant Discovery Programs have come from
increased knowledge of innate immune receptors and their mechanisms of action (Figure
1).
The first innate immune receptor agonists developed as adjuvants targeted TLR4 and
TLR9. NIAID-supported researchers have expanded screening to include molecules that
bind to NOD-like receptors and C-type lectin receptors, as well as molecules involved in
signal transduction from innate immune receptors, such as various kinases and interferon
regulatory factors. Furthermore, an increased understanding of how innate and adaptive
immune cell subsets contribute to vaccine-induced immunity is leading to the discovery of
adjuvants that target cells other than macrophages and dendritic cells. These cells include B
cells, mast cells, and NKT cells.
While considerable progress has been made in discovery of innate immune receptors and
their mechanisms of action, several challenges remain. These include:
Limitations of existing methods to evaluate potential adjuvant function in vivo.
Most screening approaches are based on a limited number of parameters, such as NF-
KB activation or the induction of select cytokines from either cell lines or primary cells.
In vitro induction of those factors may not translate into adjuvanticity in vivo. Better
methods to evaluate adjuvant function, based on increased knowledge of immunologic
mechanisms, will ensure that promising candidates are not being overlooked.
Selection of appropriate animal models for adjuvant screening. Host species
differences in immune receptor specificity and cellular distribution, immunogenetics,
and pathogen susceptibility can complicate the discovery of adjuvants for human use.
One example is TLR8, which was thought to be nonfunctional in mice until the recent
discovery of the mouse TLR8 ligand and the realization that it differs significantly from
the human TLR8 ligand. The selection of appropriate animal models for adjuvant
screening is critically important.
Adjuvants for at-risk populations, especially the very young and frail elderly.
The recent successes of the AS01-adjuvanted Shingrix
®
vaccine and MF59
®
-
adjuvanted influenza vaccine in the elderly and the AS03- and MF59
®
-adjuvanted
influenza vaccines licensed outside the U.S. for young children clearly demonstrate that
adjuvants can overcome age-associated immune deficits. However, additional adjuvants
are still needed for use in the very young and the frail elderly.
13
Goal 2: Support Technology Development for Adjuvant Discovery
Adjuvant discovery methods have improved in recent years for two main reasons:
Technologic advances have allowed for more efficient in vitro screening of compounds
with progressively higher throughput rates.
Advances in medicinal chemistry have enabled rapid and systematic modifications of
molecules to alter their immune stimulatory properties.
In addition, the focus of adjuvant discovery has shifted from natural compounds and/or
derivatives of known innate immune receptor agonists to small molecules that bear no
structural similarities to natural compounds. These molecules may bind to sites on an
immune molecule that may or may not overlap with the binding site for the natural ligand.
These types of adjuvants are identified by high-throughput screening of chemical libraries.
Recently, in silico approaches have been employed (1) to pre-screen compound libraries to
reduce the number of compounds that need to be screened in vitro or (2) to explore the
potential utility of computer-generated structures before synthesis and in vitro testing.
While in silico screening has the potential to revolutionize adjuvant science, this approach
still faces challenges, which include the needs to:
Determine structure–activity relationship (SAR) to improve the accuracy of in
silico screening methods. SAR analysis defines the relationship between a
compound’s chemical or three-dimensional structure and its biological activity. Insights
gained through SAR analysis can be used to rationally alter structure and hence the
potency or functionality of a compound. Application of SAR to in silico screening of
adjuvants requires knowledge of the structure of the immune receptor being targeted.
Define binding site requirements. Recent identification of small-molecule compounds
that can bind to innate immune receptors but lack structural similarity to the natural
receptor ligands highlights gaps in our understanding of the requirements for receptor
activation. In addition, ligand binding is not necessarily associated with receptor
activation. Thus, in silico modeling alone can be misleading and needs to be coupled
with laboratory validation to improve overall accuracy.
Provide access to supercomputing facilities. The most frequently used algorithms for
in silico screening simulate static docking of putative ligands to their receptors, rather
than the dynamic interactions that occur naturally between receptor and ligands.
Modeling such dynamic interactions requires supercomputing capacity that is not
readily available to many researchers.
2.3. NIAIDS STRATEGY TO ADVANCE ADJUVANT DISCOVERY
Based on NIAID’s goals and recommendations from the BRP, NIAIDs 2018 Strategic
Plan for Research on Vaccine Adjuvants focuses on two major areas: strengthen
14
immunology research and support technology development.
Strengthen immunology research, to specifically improve evaluation of adjuvant
function, provide new animal models for adjuvant screening, and facilitate the
discovery of adjuvants for use in at-risk populations. To meet this goal, NIAID will
continue to support and expand current programs, and develop new programs, as
follows:
o Current programs
The Systems Approach to Immunity and Inflammation Program, which is
identifying novel immune response genes through development and analysis of
novel mouse models. The data and novel mouse models generated through this
program will enhance adjuvant discovery efforts.
The Immunity in Neonates and Infants program and the Immunity in the Elderly
program, which are characterizing immune functionality and facilitating
identification of possible adjuvant targets in these at-risk populations.
The Adjuvant Discovery Program, which continues screening, preclinical
testing, and early-stage development of new adjuvant candidates and will pursue
discovery of immune modulating or tolerogenic adjuvants.
The Molecular Mechanisms of Combination Adjuvants (MMCA) program,
which is providing novel insights into the mechanisms of combination adjuvant
activity and the basis for synergistic adjuvanticity.
o FY 2019 new NIAID initiatives
Maintaining Immunity After Immunization, which will increase understanding
of how durable immunity is induced and maintained in response to infection or
vaccination and will identify novel targets for adjuvant discovery
Collaborative Cross (CC) Mouse Model Generation and Discovery of
Immunoregulatory Mechanisms, which will support the use of CC mouse lines
to increase understanding of the host genetics involved in immune regulation
and function
o NIAID will continue to use the SBIR contract mechanism to fund additional
adjuvant discovery by small business entrepreneurs and the development of
immunologic reagents for underrepresented animal models that may be better
models of human immune responses (e.g., ferrets for influenza research).
o In addition to its solicited adjuvant portfolio, NIAID will maintain and expand its
portfolio of unsolicited adjuvant grants using a variety of mechanisms, such as
SBIR, R01, R03, R21, and T grants.
Support technology development for adjuvant discovery. For example:
o NIAID will expand support of structural immunology studies to enhance SAR
analyses and improve the accuracy of in silico adjuvant discovery methods.
Currently, most of the structural immunology research supported by NIAID is
through investigator-initiated grants. While these studies have provided valuable
insights into the structure–function relationships of many important immune
molecules, technologic advances in this area would increase the number of
15
available structures for use by the adjuvant discovery community. NIAID will
consider using the SBIR contract funding mechanism and other approaches to grow
this important research area.
o NIAID’s Office of Cyber Infrastructure and Computational Biology (OCICB)
provides sophisticated biocomputing resources to NIAID’s intramural researchers.
NIAID extramural staff will work with OCICB staff to make these supercomputing
capabilities available to adjuvant researchers in the extramural community.
16
3. ADJUVANT DEVELOPMENT AND PRECLINICAL EVALUATION
3.1. RECENT PROGRESS IN NIAID-SUPPORTED ACTIVITIES
Adjuvant Development Program
NIAID expanded its solicited program in adjuvant science in 2008 with the Adjuvant
Development Program. This contract program supports:
Preclinical development of novel adjuvants coupled with specific vaccines, including
mechanistic analyses of the novel adjuvant/antigen combinations
Formulation and dosing studies
Optimization of immunization regimens and delivery routes
cGMP production
This program was recompeted in 2013 and 2018 with the goal of ensuring further
development of promising adjuvant candidates through preclinical testing and IND-
enabling studies, thus expanding the number of adjuvants available for clinical evaluation.
Several of the compounds evaluated through the Adjuvant Development Program
originated from NIAID’s Adjuvant Discovery Program, and some have been or are
currently being tested in more than 20 clinical trials. In 2016, NIAID began using the
annual SBIR contract solicitation to support additional adjuvant development activities.
The adjuvant/antigen or adjuvant/vaccine combinations assessed through the Adjuvant
Development program and SBIR contract program include:
Microcrystalline inulin with seasonal, pandemic, and universal influenza vaccine
candidates
Nanoemulsions with a plant-derived, recombinant H5 influenza vaccine or HIV
envelope virus-like particle (VLP) vaccine
Fully synthetic TLR4 agonists plus plant-derived saponin (QS-21), coupled with a
recombinant E protein from West Nile virus
A small-molecule RIG-I agonist combined with split H5N1 influenza virus, UV-
inactivated West Nile virus, or a VLP-based vaccine for Zika virus
The protein-based mucosal double-mutant heat-labile toxin (dmLT) adjuvant, with
BCG and the recombinant fusion protein, 5fu, from Mycobacterium tuberculosis and a
recombinant IpaB-IpaD fusion protein from Shigella sonnei
A novel semi-synthetic analog of QS-21 (TiterQuil-1055™) with Fluzone
®
, Fluarix
®
,
and Flublok
®
influenza vaccines
Adjuvants in HIV Preclinical Vaccine Development
NIAID also supports adjuvant development within vaccine-focused research programs.
NIAIDs HIV preclinical vaccine development activities focus predominately on variations
of HIV Env immunogens in combination with an array of adjuvants and/or delivery
17
systems to enhance potency and durability. These studies are conducted in both small
animals and nonhuman primates. An extensive variety of adjuvants and adjuvant
combinations have been tested, with the most common adjuvants being Alhydrogel
and
other aluminum salts formulations, MF59
®
, AS0 series, Adjuplex, ISCOMATRIX
®
, pIL-
12, and multiple TLR agonists alone or in combination (3M-052, CpG, poly ICLC, R848,
GLA-SE).
Adjuvants in Non-HIV Preclinical Vaccine Development
NIAID’s vaccine research and development activities for non-HIV infectious diseases
include three different types of vaccines: live attenuated vaccines; inactivated vaccines; and
subunit vaccines composed of purified protein, recombinant protein, or carbohydrate. These
preclinical research and development efforts focus on early discovery of adjuvanted vaccine
targets and IND-enabling activities, including providing biological resources, animal
testing, process development, Good Laboratory Practice (GLP) toxicity assessment, and
cGMP production of vaccines or vaccine components. Early-stage research on adjuvanted
vaccines includes pairing of antigens with adjuvants, formulation studies, and the design
and construction of novel vaccine candidates in which an adjuvant is built into the vaccine
construct. Examples of this latter approach include co-display of the adjuvant and antigen
on the surface of nonpathogenic engineered microbes and nanoparticle molding
technologies for co-delivery of adjuvant and antigen. NIAID also supports development of
animal models that are relevant for or mimic natural infection or diseases of public health
importance to evaluate vaccine formulation. These models are especially important for
vaccine research for biodefense or emerging infectious diseases, since the data generated
may be utilized for vaccine emergency use authorization, accelerated approval, or pivotal
efficacy studies under the Animal Rule.
NIAID also supports the development of adjuvanted vaccines in some high-priority
research areas through investigator-initiated research grants and various program
initiatives, such as SBIR contracts, Centers of Excellence for Translational Research,
Advanced Development of Vaccine Candidates for Biodefense and Emerging Infectious
Diseases, and Research to Advance Vaccine Safety. Selected examples of current activities
include evaluation of:
Candidate tuberculosis (TB) vaccines containing traditional and novel adjuvants or
adjuvant systems, as follows:
o The cyclic dinucleotide (CDN) class of molecules (a vita-PAMP), which includes c-
di-AMP, as regulators of M. tuberculosis physiology and the host immune response
to infection and vaccination. Research is underway to determine the adjuvant
potential of c-di-AMP in a preventative tuberculosis vaccine.
o The cationic liposomal adjuvant CAF09, which induces both CD4+ and CD8+ T
cells
o An adjuvant platform to produce micelles and vesicles incorporating mycolic acid,
CpG, and MPL for intranasal immunization
o A wide range of adjuvants, including alum, GLA-SE, GLA-AF, QS-21, CpG-SE,
and liposomes, which are being evaluated independently and in combination with
18
either anionic, neutral, or PEG liposomes to identify the optimal adjuvant pairing
with the M. tuberculosis antigen ID93
Candidate malaria vaccines containing:
o The novel glycolipid adjuvant 7DW8-5 combined with a live attenuated sporozoite
vaccine
o Traditional adjuvants such as Alhydrogel
, MPL/Alhydrogel
, ISA 51 VG, ISA
720 VG, GLA-SE, GLA-LSQ, AddaVax
, and CpG 7909, in malaria subunit-based
vaccines
o Nano/microparticle technology platforms for a malaria transmission-blocking
vaccine
Programs in Support of Adjuvanted Vaccine Development
Several of NIAID’s basic immunology programs facilitate selection of appropriate antigens
and evaluation of antigen-specific immunity generated by candidate adjuvanted vaccines,
including:
The Immune Epitope Database and Analysis Resource (IEDB, www.iedb.org), which
provides public access to B and T cell immune epitope information curated from the
scientific literature or through direct submission by the research community, and
sophisticated epitope prediction and analysis bioinformatics tools
The B-Cell Epitope Discovery and Mechanisms of Antibody Protection program and
the Large-Scale T-Cell Epitope Discovery program
The NIH Tetramer Core Facility (http://tetramer.yerkes.emory.edu/)
The Human Immunology Project Consortium (HIPC) program, which uses novel
approaches, including metabolomics, transcriptomics, and proteomics, to conduct
comprehensive systems immunologic analyses of human immune responses to
infections, adjuvants, and/or vaccines
The Cooperative Centers on Human Immunology (CCHI) program, which is
developing novel technologies and bioinformatics tools to conduct mechanistic
analyses of human immune responses to infections and vaccines (adjuvanted and
unadjuvanted)
The Modeling Immunity for Biodefense program, which includes the development of
computational tools that analyze antibody-antigen interactions and facilitate design of
more immunogenic or cross-protective antigens that can be paired with adjuvants
Preclinical Development Services and Support
NIAID also provides a comprehensive suite of preclinical development services that fill
particular knowledge and experimental gaps critical to move adjuvanted vaccine candidates
along the product development pathway. These resources include:
BEI Resources, a central repository that supplies organisms and reagents, including
those relevant for vaccine or adjuvant research to the research community
The Preclinical Models of Infectious Diseases program, which provides services to
researchers to study the full range of pathogens, including bacteria, viruses, parasites,
19
fungi, and other agents such as toxins and prion proteins. The program’s main focus is
to help researchers who have developed a product such as a vaccine that needs to be
tested in an animal model, but who lack either the resources or expertise needed to
perform that testing, and to support the development and refinement of animal models
for infectious diseases of importance to humans.
NIAIDs Vaccine Development Services, which are intended for use in the
investigation, control, prevention, and treatment of a wide range of infectious agents
and to support the development of vaccines; vaccine components, including adjuvants;
vaccine delivery systems; and challenge material. Vaccine testing services include
assay development, immunogenicity and efficacy studies, clinical and nonclinical
sample testing, and safety and toxicity testing. Vaccine manufacturing services include
writing product development plans, product optimization, non-cGMP and cGMP
manufacturing of antigens and adjuvants, assay development, and regulatory activities,
including audits.
NIAID will continue to support new program initiatives to address the development and
evaluation of important vaccines and vaccine technologies, including adjuvants. Examples
of some existing programs are:
Centers of Excellence for Translational Research (for emerging/reemerging vaccine
technologies)
Novel Vaccine Technologies and Strategies to Promote Sustained Vaccine Efficacy (for
malaria and pertussis)
Advanced Development of Vaccine Candidates for Biodefense and Emerging
Infectious Disease (for vaccines, including influenza and TB; and technologies,
including adjuvants)
Research to Advance Vaccine Safety
These services and programs support a wide range of preclinical development needs for
vaccines, including adjuvanted vaccine construction; in vitro and in vivo assessment of
candidate products; generation of data regarding optimization, synthesis, and formulation
of candidates; manufacturing using cGMP standards; and safety assessments such as GLP
toxicity studies.
3.2. PROGRAM GOALS AND RESEARCH CHALLENGES
Goal 1: Expand Availability of Novel Adjuvants for Preclinical Vaccine Development
and Clinical Evaluation
Vaccine developers tend to look for adjuvants with well-established safety records,
preferably in humans, when selecting an adjuvant for their candidate vaccines. NIAID’s
adjuvant development activities include mechanism-of-action studies to gain insights into
immune pathways triggered by adjuvants and ensure that off-target effects are identified
and mitigated early in the development process. These efforts will be crucial in assembling
a pool of well-characterized, potent adjuvants with proven safety records that can be used
to accelerate development of effective vaccines.
20
The major challenge associated with early stage development and preclinical assessment of
novel adjuvants and adjuvant vaccines is:
The ability to correlate safety and efficacy observed in animal models to human
responses. Animal models, while valuable, are not able to emulate many of the
factors affecting responses in humans. These factors include:
o Pre-existing conditions and medications; recent infection history and chronic
infection status (e.g., cytomegalovirus [CMV], Epstein-Barr virus [EBV])
o Genetics/epigenetics (e.g., species-specific TLR expression patterns and ligand
binding)
o Microbiome composition
o Behavior, including exercise and recreational drug use
o Nutrition and diet
o Environmental exposures
In addition, current animal models may not identify the most effective vaccine regimens or
predict adjuvant-vaccine reactogenicity in humans. Although certain cytokines, other
soluble molecules, and transcriptional networks have been associated with reactogenicity,
well-defined signatures that reliably predict reactogenicity have not been identified.
Concerns about adverse events have slowed the development and licensure of adjuvanted
vaccines, especially for at-risk populations such as the very young, pregnant women, and
the elderly.
Goal 2: Optimize Formulation, Antigen/Adjuvant Pairing, and Support cGMP
Production
Development of novel adjuvanted vaccines involves activities that require specialized,
multidisciplinary teams with strong formulation expertise. NIAID’s adjuvant and vaccine
development programs support formulation studies, including the iterative process required
to optimize antigen/adjuvant pairing, and cGMP production of lead adjuvant and adjuvant-
antigen formulations for further downstream preclinical development and clinical
evaluation. Formulation studies are crucial during adjuvant development and vaccine
construction. However, they also must be considered during the early stages of adjuvant
discovery and adjuvant-antigen pairing, since inadequate or suboptimal formulation may
prevent promising novel adjuvants or adjuvanted vaccines from moving forward in
development.
The major challenges associated with these activities include:
Formulation considerations. Subtle differences in formulations can significantly
impact immunogenicity, stability, and safety of adjuvanted vaccines. In addition, due to
significant advances in antigen discovery and development of protein expression
platforms, most vaccines currently in development are subunit vaccines. Pairing the
correct protein antigens with the appropriate adjuvants during the early development
stage is critical for future success of a safe and efficacious vaccine. Currently, given the
21
lack of correlates or surrogates of protection for most pathogens, head-to-head
comparisons of multiple adjuvants with specific vaccine antigens may be the most
efficient process for identifying the optimal adjuvants for a given antigen.
Despite the vital importance of formulation, including co-formulation of
adjuvant/antigen pairing, adjuvant development efforts frequently lack adequate
consideration of formulation, which can impact:
o Proper adjuvant selection and retention of antigenicity/immunogenicity
o Targeting of the adjuvant to the correct immune cells and lymphatic tissues
o Co-localization of antigen and adjuvant, or proper antigen conformation
o Timed release of the adjuvant to prevent induction of excessive innate immune
responses and the potential for reactogenicity
o Selection of the optimal delivery system and vaccination strategy
cGMP manufacturing. Bottlenecks for vaccine developers in academia and small
businesses are (1) limited access to proprietary adjuvants; (2) development of scalable,
robust manufacturing processes for promising novel adjuvants; and (3) lack of
sufficient quantities of cGMP material for well-characterized adjuvants/adjuvanted
vaccines required for early phase clinical trials.
3.3. NIAIDS APPROACH TO ADVANCE ADJUVANT DEVELOPMENT AND
PRECLINICAL EVALUATION
To address the issues raised above and to continue the development of promising
adjuvanted vaccine candidates, NIAID will support the following activities:
Expand availability of novel adjuvants for preclinical vaccine development and clinical
evaluation by improving the ability to correlate safety and efficacy profiles generated in
animal models to human responses.
To accomplish these efforts, NIAID will:
o Continue NIAIDs Preclinical Models of Infectious Disease program, which
provides a central resource for development and refinement of animal models,
including traditional small laboratory animals, nonhuman primates, and
nontraditional animals such as swine. This program also offers in vivo screening
and efficacy testing in appropriate animal models.
o Continue NIAID’s SBIR contract program for the development of immunologic
reagents for underrepresented mammalian models, including ferrets, guinea pigs,
and swine, to expand the types of animal models that can be used for detailed
immunologic analyses of adjuvant function
o Maintain and expand development of novel animal models that more accurately
reflect human immune responses to adjuvanted vaccines across all age
groups/populations through NIAID’s Systems Approach to Immunity and
Inflammation program and through a new funding opportunity, starting in 2019, to
22
support the use of CC and CC-RIX mouse lines that more faithfully reproduce
human immune responses
o Increase support to HIPC and CCHI investigators to:
Accelerate further development and validation of fully human artificial
immune systems” and immune tissue explants
Conduct additional systems immunology analyses of head-to-head comparisons
of novel adjuvant/antigen combinations
Optimize formulation and antigen/adjuvant pairing activities.
To accomplish these efforts, NIAID will:
o Renew the Adjuvant Development Program in 2018, which includes a stronger
emphasis on formulation and head-to-head comparisons of adjuvant/antigen
combinations. NIAID also will continue to award SBIR contracts to increase
adjuvant development. Community access to adjuvants developed under these
programs will be expanded through targeted NIAID supplements.
o Expand development of novel adjuvanting technologies, including conjugation
technologies or self-adjuvanting vaccine platforms, through the SBIR adjuvant
development contracts and other funding mechanisms
o Expand development of co-adjuvants to include checkpoint inhibitors that enhance
APC or T-cell responses by extending immune activation or by blocking negative
regulators
o Continue to support adjuvant process development to enable cGMP production
capabilities for adjuvant production through NIAID’s Adjuvant Development
program
o Encourage and continue to support preclinical development of adjuvanted vaccines
in the areas of product optimization, process development, formulation studies,
cGMP production, and GLP safety assessment through NIAID’s Vaccine
Development Services, Broad Agency Announcements (BAAs), partnership
programs, and SBIR contract solicitations to accelerate the transition of adjuvanted
vaccine candidates into clinical evaluation
23
4. CLINICAL EVALUATION OF ADJUVANTED VACCINES
4.1. RECENT PROGRESS IN NIAID-SUPPORTED ACTIVITIES
Adjuvanted Vaccines for Non-HIV Infectious Diseases
Since 1998, NIAID has supported 133 vaccine clinical trials for non-HIV infectious
diseases, primarily through the Vaccine and Treatment Evaluation Units (VTEUs). Forty-
seven of these clinical trials have included adjuvanted vaccines, with adjuvants such as
Alum (used in 13 of these trials), MF59
®
, AS03, glucopyranosyl lipid adjuvant (GLA, a
synthetic TLR4 agonist), and GLA with QS-21 in liposomal formulation. Recently
completed and ongoing trials of adjuvanted vaccines for non-HIV infectious diseases are
summarized in Appendix C and described below:
Phase I and II clinical trials of inactivated influenza vaccine candidates with established
adjuvants in multiple combinations, as part of its influenza pandemic preparedness
program
Exploration of novel vaccination strategies, including heterologous prime-boost interval
studies using H7 influenza vaccines
In collaboration with GSK, the direct comparison of two adjuvants, AS03 and MF59
®
,
with seasonal influenza vaccines. The trial has been completed and results will be
released to the public once data analyses are completed.
A first-in-human Phase I clinical trial with a purified formalin-inactivated Zika virus
vaccine candidates with alum adjuvant, in a rapid response to this emerging viral
disease threat
Two clinical trials exploring vaccines with novel adjuvants
o TLR7/8 agonist, Imiquimod, to enhance the efficacy of a pre-pandemic H5
influenza vaccine
o Recombinant double-mutant heat-labile toxin (dmLT), which acts as both antigen
and adjuvant in a vaccine against enterotoxigenic E. coli (ETEC). The dmLT
adjuvant can induce strong mucosal immune responses after parenteral delivery.
Evaluation of tuberculosis (TB) vaccine candidates in Phase I clinical trials using new
combination adjuvants, GLA-LSQ and GLA-SE
Evaluation of new combination adjuvants such as GLA-LSQ for a malaria vaccine or
Alhydrogel
plus GLA in Aqueous Formulation (GLA-AF) for schistosomiasis
vaccines
Use of the human challenge models to evaluate, compare, or narrow the field of choices
of different vaccine formulations for hookworm vaccines
Adjuvanted Vaccines for HIV
NIAIDs HIV vaccine program has been exploring multiple vaccine platforms to achieve
protective immunity against HIV that include HIV DNA vaccines or viral vectors boosted
or combined with adjuvanted recombinant Env protein(s). Two complementary strategies
are being used: an empirical approach that builds on partial clinical success to
expeditiously move vaccine candidates into human testing and a theoretical approach that
24
selects vaccine candidates for further development based on an understanding of the
immune response to HIV infection. Completed and ongoing trials of HIV adjuvanted
vaccines are described below and summarized in Appendix D.
The Empirical Approach
This approach relies mostly on the landmark RV144 study in Thailand, the first and
only (as of April 2018) large clinical trial to demonstrate modest efficacy for an
investigational HIV vaccine. RV144 evaluated the safety and estimated the efficacy of
a prime-boost combination of two vaccine components given sequentially: ALVAC-
HIV, which uses a canarypox virus as a vectoror carrier—to deliver HIV genes; and
the recombinant AIDSVAX B/E, HIV Env protein formulated with alum hydroxide gel
(Rehydragel). At the end of the 3.5-year study period, investigators observed a 31%
reduction in risk of HIV infection among vaccine recipients compared to those who
received a placebo.
Analysis of the immune responses induced by the vaccine suggests that a lower risk of
infection is associated with:
o Plasma immunoglobulin G (IgG) antibodies targeting the V1V2 region of HIV Env
o Low plasma levels of HIV-Env-specific IgA (which compete with protective IgG)
o Polyfunctional CD4+ T cells (CD154, interleukin-2 [IL-2], IL-4, interferon-gamma
[IFN-γ], and tumor necrosis factor alpha [TNF-α])
o IgG antibodies harboring Fc effector functions needed for antibody-dependent cell-
mediated cytotoxicity (ADCC), but only in vaccinees with low-plasma, HIV-Env-
specific IgA
To follow up on this encouraging outcome, the P5 partnership (Pox-Protein-Public-
Private-Partnership, http://www.vaccineenterprise.org/content/P5Partnership) was
established in 2010 to substantiate and improve upon RV144 using a modified pox-
protein HIV vaccine. Phase I studies include:
o Side-by-side comparison of adjuvant formulations
Boost of HIV Env formulated with Rehydragel vs. MF59
®
after ALVAC
prime (HVTN 107)
Boost of HIV Env formulated with MF59
®
vs. AS01B after DNA prime (HVTN
108)
Boost of HIV Env formulated with MF59
®
vs. AS01B after ALVAC prime
(HVTN 120)
Prior to completion of the RV144 trial, many adjuvants were tested in Phase I clinical
trials in combination with HIV immunogens: Rehydragel, Alhydrogel
, Adju-Phos
,
MF59
®
, AS01B, and GLA-SE. Two Phase IIb studies used similar recombinant Env
subunit proteins only (without the ALVAC prime) as part of the regimen (VAX003,
VAX004) that were formulated with Rehydragel, but these studies did not show
efficacy.
25
The Theoretical Approach
This approach targets the induction of broadly neutralizing antibodies (bNAbs) against
HIV. Based on HIV natural infection studies and an exquisite characterization of the
structure of the HIV Env spike, there is a better understanding of the requirements to
induce bNAbs. The immunogens must first engage the B cell harboring the germline
lineage of desired antibodies, and then, through multiple cycles of affinity maturation,
the gene encoding immunoglobulins may acquire the mutations needed to provide
broad and potent HIV neutralization activity. T follicular helper (Tfh) cells, the drivers
of affinity maturation in the lymph node, also are sought to generate better bNAbs. In
addition to high levels of somatic hypermutation, other unusual structural features of
bNAbs include a long heavy chain complementarity-determining region 3 (HCDR3),
capable of reaching through the glycan shield of the virus, and autoreactivity.
Two additional clinical trials involving direct comparisons of adjuvants are under
development. The first is supported by the HIV Vaccine Trials Network (HVTN) and
will use a stabilized trimer BG505 SOSIP.664 immunogen, adjuvanted with
3M052+Alum, CpG 1018, or GLA-LSQ. The second trial, a partnership between
NIAID and the U.S. Military HIV Research Program, will test a DNA prime, gp145
HIV Env protein boost, adjuvanted with several Army Liposome Formulations (ALFs),
Alhydrogel
®
, and dmLT.
4.2. PROGRAM GOALS AND RESEARCH CHALLENGES
Goal 1: Increase the Availability of Well-Characterized Novel Adjuvants for Use in
Vaccine Clinical Trials
One of NIAID’s major goals is to develop vaccines against pathogens and their toxins. This
goal includes clinical evaluation in both domestic and international populations, which
often have distinct pathogen exposure histories that can impact host immunity. Therefore,
adjuvants capable of inducing Th1/Th17, Tfh, CD8+ T cell, or mucosal immune responses
or that are effective in at-risk populations are especially needed.
Challenges associated with these activities include:
Difficulty in reconciling human clinical data and animal preclinical data to
improve the design and further evaluation of adjuvanted vaccine candidates. The
primary endpoints of early-phase clinical trials of adjuvanted vaccines are safety and
immunogenicity. However, sample sizes are generally small and lack sufficient
statistical power to dissect safety or identify correlates of protection, both of which are
critical parameters in determining whether a vaccine candidate moves into later-phase
clinical trials or needs further refinement. Preclinical animal efficacy studies may
provide some insights regarding safety and efficacy, but species differences in
immunity, pharmacogenetics, and reactogenicity need to be considered.
Limited number of adjuvants available from commercial sources. Although NIAID
has supported many clinical trials of vaccine candidates against infectious diseases,
26
those requiring adjuvants continue to rely on a limited number of sources. Access to
novel adjuvants already in clinical development is hampered by intellectual property
(IP) restrictions. In addition, adjuvant IP holders may either oppose or not be interested
in direct comparisons of their formulation with those they do not own. Bringing more
potent adjuvants into the clinic represents a significant financial and administrative
burden for any adjuvant developer. Many of these developers reside at academic
institutions or own small businesses and have neither experience with the process nor
the funding needed to bring products into the clinic.
Goal 2: Maximize Evaluation of Data from Clinical Trials
Although many well-designed clinical trials generate valuable datasets and contain a wealth
of information, the lack of correlates of protection for most pathogens complicates the
rational selection of an adjuvant for the clinical evaluation of a novel vaccine. Other issues
related to evaluation of data from clinical trials of adjuvanted vaccines include:
Formulations identified in Phase I clinical trials as safe and immunogenic that may fail
in advanced clinical development or efficacy field trials
Immunological analyses of clinical trial samples commonly focus on certain parameters
known or assumed to be associated with protection against a specific disease. However,
such assumptions limit the breadth of immunologic information produced and our
ability to identify and validate immune correlates of protection associated with adjuvant
functionality.
The lack of standardized assays to measure the immunogenicity of an adjuvant
combined with different antigens. This data would establish immunological profiles of
adjuvants and determine how much those profiles are affected by the antigen
component of the vaccine.
Sample-sparing techniques, such as those supported by NIAID’s Sample Sparing Program,
have improved significantly in recent years allowing more parameters to be determined
with small numbers of cells or volumes of whole blood or serum. Simultaneously, the cost
of multiplex assays has decreased making the routine generation of high-density datasets
from clinical trials samples highly feasible.
The major challenge associated with this goal is:
Access to data from adjuvant studies and adjuvanted vaccine and human
challenge clinical trials. Many investigators conducting adjuvant research or clinical
trials do not deposit their datasets into publicly accessible databases such as ImmPort,
because they either are unaware of relevant databases or lack incentives or the expertise
to comply with data submission requirements. These requirements may include the
collection of specific meta-data parameters, data types, and use of controlled
vocabularies.
27
4.3. NIAIDS APPROACH TO ADVANCE THE CLINICAL EVALUATION OF
ADJUVANTED VACCINES
To address the challenges raised above and to continue the evaluation of novel adjuvants in
clinical trials, NIAID plans to foster/support the following activities:
Generation, collection, curation, and public availability of clinical data on the
immunological activity of vaccine adjuvants to inform their use in subsequent trials
Expansion of NIAID-sponsored research using human challenge models to evaluate the
efficacy of candidate adjuvanted vaccine formulations
Support of clinical trials to compare and down-select novel adjuvanted vaccine
candidates
Continued/expanded investment in the development of computational models that can
reconcile data from human clinical trials and preclinical animal studies to improve the
design and efficacy of adjuvanted vaccines
Continued investment in the development of sample sparing assays to enable evaluation
of more immunological parameters in small clinical samples
Use of NIAID-sponsored programs such as HIPC, CCHI, and Systems Biology
programs to:
o Conduct research to identify biomarkers and correlates/surrogates of immune
protection or safety/reactogenicity
o Assist in the design of immunological analyses of clinical trials samples
o Enable collaborations between existing NIAID networks and clinical trials sites
through targeted administrative supplements
Expansion of NIAID’s Bioinformatics resources, including ImmPort
(www.immport.org), to:
o Develop a public database of reference adjuvants and their immunologic activity
o Provide access to immunologic and other datasets generated by NIAID-supported
clinical trials
28
5. SUMMARY
The goal of NIAID’s adjuvant discovery and development programs is to generate a diverse
panel of well-defined, safe, and effective adjuvants that can be paired with candidate vaccines
to induce the desired protective immune profiles in all relevant populations.
To achieve this goal, NIAID will continue to:
Support and expand its fundamental immunology research portfolio to identify the
requirements for inducing effective immune responses and long-lasting protection in all
individuals
Advance adjuvant discovery and early stage development, specifically targeting the critical
immunologic parameters described in the relevant sections above
Support the discovery of adjuvanted vaccines; animal testing and screening, including GLP
safety assessment of adjuvanted vaccine formulations; process development; and
production of clinical grade materials of vaccines and vaccine adjuvants
Explore the feasibility of a NIAID-sponsored cGMP vaccine/adjuvant manufacturing
facility for small-scale clinical trials
Explore creative approaches to promote and accelerate clinical testing of promising vaccine
candidates formulated with novel adjuvants
29
6. APPENDICES
6.1. APPENDIX A: ABBREVIATIONS
ADCC Antibody-Dependent Cell-Mediated Cytotoxicity
ALF Army Liposome Formulation
APC Antigen-Presenting Cell
BAA Broad Agency Announcement
bNAb Broadly Neutralizing Antibody
BRP Blue Ribbon Panel
CC Collaborative Cross
CCHI Cooperative Centers for Human Immunology
c-di-AMP Cyclic Di-Adenosine Monophosphate
CDN Cyclic Dinucleotide
cGMP Current Good Manufacturing Practice
CLR C-Type Lectin Receptor
CMV Cytomegalovirus
DAIDS Division of AIDS
DAIT Division of Allergy, Immunology, and Transplantation
DMID Division of Microbiology and Infectious Diseases
dmLT Double-Mutant Heat-Labile E. coli Toxin
dsRNA double-strand RNA
EBV Epstein-Barr virus
ETEC Enterotoxigenic E. coli
FDA Food and Drug Administration
GLA Glucopyranosyl Lipid Adjuvant
GLP Good Laboratory Practice
GSK GlaxoSmithKline
HCDR3 Heavy Chain Complementarity-Determining Region 3
HIPC Human Immunology Project Consortium
HPV Human Papillomavirus
HVTN HIV Vaccine Trials Network
IEDB Immune Epitope Database and Analysis Resource
IgG immunoglobulin G
IL-2 interleukin-2
IFN-γ interferon-gamma
MMCA Molecular Mechanisms of Combination Adjuvants
MPL Monophosphoryl Lipid
MRSA Methicillin-Resistant Staphylococcus aureus
NE Nanoemulsion
NIAID National Institute of Allergy and Infectious Diseases
NIH National Institutes of Health
NKT Natural Killer T
OCICB Office of Cyber Infrastructure and Computational Biology
ODN Oligodeoxynucleotide
PAMP Pathogen-Associated Molecular Patterns
30
PCN Protein Cage Nanoparticles
SAR Structure–Activity Relationship
SBIR Small Business Innovation Research
TB Tuberculosis
Tfh T Follicular Helper
TLR Toll-Like Receptor
TNF-α tumor necrosis factor alpha
VLP Virus-Like Particle
VTEU Vaccine and Treatment Evaluation Units
31
6.2. APPENDIX B: BLUE RIBBON PANEL MEMBERS
Rafi Ahmed, Ph.D.
Professor
Emory University School of Medicine
James Baker, M.D.
Director, Michigan Nanotechnology Institute for Medicine and Biological Sciences
University of Michigan
Mario Barro, Ph.D.
Senior Director
Head of Technology and External Networks
Sanofi Pasteur
John Clements, Ph.D.
Professor
Tulane University
Robert Coffman, Ph.D.
Senior Vice President and Chief Scientific Officer
Dynavax Technologies
Victor DeFilippis, Ph.D.
Assistant Professor
Oregon Health and Sciences University/Vaccine and Gene Therapy Institute
Jay Evans, Ph.D.
Professor
Director, Center for Translational Medicine
University of Montana
Michael Gale, Ph.D.
Professor
University of Washington
Hana Golding, Ph.D.
Chief, Laboratory of Retroviruses
Center for Biologics Evaluation and Research
FDA
32
Nir Hacohen, Ph.D.
Professor
Broad Institute and Massachusetts General Hospital
Ross Kedl, Ph.D.
Professor
University of Colorado
Marian Kohut, Ph.D.
Professor
Iowa State University
Ofer Levy, M.D., Ph.D.
Director
Precision Vaccines Program
Boston Children’s Hospital
Julie McElrath, M.D., Ph.D.
Senior Vice President
Director, Vaccine and Infectious Disease Division
Fred Hutchinson Cancer Research Center
Nikolai Petrovsky, M.B.B.S., Ph.D.
Professor
Vaxine/Flinders University
Rino Rappuoli, Ph.D.
Chief Scientist
GSK Vaccines
Steven Reed, Ph.D.
President and CEO
Infectious Disease Research Institute
Amy Weiner, Ph.D.
Senior Program Officer, Discovery & Translational Sciences
Bill & Melinda Gates Foundation
33
6.3. APPENDIX C: NIAID-SPONSORED CLINICAL TRIALS FOR NON-HIV
ADJUVANTED VACCINES, RECENTLY COMPLETED OR ONGOING (AS
OF APRIL 2018)
Adjuvant Pathogen
Pathogen
Vaccine Antigen/Approach
Phase
ClinicalTrials.gov
Identifier
dmLT
Enterotoxigenic
E. coli
dmLT antigen
Phase I
NCT02531685
AS03 vs.
MF59
®
Influenza
A/H5N8
A/gyrfalcon/Washington/41088-
6/2014
Phase I
NCT02624219
AS03 vs.
MF59
®
Influenza
A/H5N8
A/gyrfalcon/Washington/41088-
6/2014
Phase I
NCT03014310
AS03
Influenza
A/H7N9
Monovalent A/Hong
Kong/125/2017
Phase II
NCT03312231
AS03
Influenza
A/H7N9
Monovalent A/Hong
Kong/125/2017, with seasonal
IIV4
Phase II
NCT03318315
AS03
Influenza
A/H7N9
Monovalent A/H7N9
A/Shanghai/2/2013
Phase II
NCT02921997
MF59
®
Influenza
A/H7N9
Monovalent A/H7N9
A/Shanghai/2/2013, with live
attenuated H7N9
Phase I
NCT02251288
GLA-SE vs.
AP10-602
M. tuberculosis
ID93 antigen
Phase I
NCT02508376
Alhydrogel
+/ GLA-AF
N. americanus
Co-administered Na-GST-1 and
Na-APR-1 antigens
Phase Ib
NCT02476773
Alhydrogel
+/ GLA-AF or
CpG 10104
N. americanus
Na-GST-1 antigen
Phase II
NCT03172975
Alhydrogel
+/ GLA-AF
(AP 10-701)
Schistosoma
spp.
Sm-TSP-2 antigen
Phase Ia
NCT02337855
Alhydrogel
®
+/ GLA-AF
(AP 10-701)
Schistosoma
spp.
Sm-TSP-2 antigen
Phase Ib
NCT03110757
Alum Zika
Zika virus purified inactivated
vaccine
Phase I
NCT02963909
34
6.4. APPENDIX D: NIAID-SPONSORED CLINICAL TRIALS FOR HIV
ADJUVANTED VACCINES, COMPLETED OR ONGOING (AS OF APRIL
2018)
Adjuvant HIV Vaccine Antigen/Approach
Phase
ClinicalTrials.gov Identifier
Adju-Phos
Ad26.Mos.HIV or Ad26.Mos4.HIV
boosted by gp140 Clade C protein
Phase I
NCT02788045
Adju-Phos
Ad26.Mos4.HIV boosted by gp140
Clade C protein
Phase I
NCT03060629
Alhydrogel
gp145 Clade C Env protein
Phase I
NCT03382418
GLA-SE EnvSeq1: CH505 TF gp120 series
Phase I
NCT03220724
GLA-SE
Polyvalent DNA boosted by gp120 (A,
B, C, AE)
Phase I
NCT03409276
pIL-12 PENNVAX
®
-GP DNA
Phase I
NCT02431767
MF59
®
ALVAC (vCP2438) boosted by
bivalent subtype C gp120
Phase I/II
NCT02404311
MF59
®
ALVAC (vCP2438) boosted by
bivalent subtype C gp120
Phase IIb/III
NCT02968849
MF59
®
DNA-HIV-PT123 boosted by bivalent
subtype C gp120
Phase I
NCT02997969
MF59
®
gp140 Clade C boost to HVTN073
Phase I
NCT01423825
MF59
®
gp140 Clade C
Phase I
NCT01376726
MF59
®
MVA-C boosted by gp140, DNA-C2
boosted by MVA-C, or DNA-C2
boosted by MVA-C and gp140
Phase I
NCT01418235
Rehydragel
DNA-HIV-PT123 or NYVAC-HIV-
PT1/PT4 prime with AIDSVAX B/E
boost
Phase I
NCT01799954
Rehydragel
ALVAC (vCP1521) boosted by
AIDSVAX gp120 B/E
Phase I
NCT02109354
Rehydragel
DNA-HIV-PT123 boosted by
AIDSVAX gp120 B/E
Phase I
NCT02207920
35
Adjuvant HIV Vaccine Antigen/Approach
Phase
ClinicalTrials.gov Identifier
Rehydragel
ALVAC (vCP1521) boosted by
AIDSVAX gp120 B/E
Phase III
NCT00223080
pIL-12 and pIL-15 PENNVAX-B DNA
Phase I
NCT00528489
MF59
®
vs. AS01B
DNA-HIV-PT123 boosted by bivalent
subtype C gp120
Phase I
NCT02915016
MF59
®
vs. AS01B
ALVAC (vCP2438) boosted by
bivalent subtype C gp120
Phase I/IIa
NCT03122223
MF59
®
vs.
Rehydragel
ALVAC (vCP2438) boosted by
bivalent subtype C gp120
Phase I
NCT03284710