Nasdaq: VBIV Corporate Presentation December 2014 Forward-Looking Statement Disclaimer This presentation contains forward-looking statements within the meaning of the provisions of Section 27A of the Securities Act of 1933, as amended, and Section 21E of the Securities Exchange Act of 1934, as amended. Forward-looking statements are generally identifiable by the use of words like "may," "will," "should," "could," "expect," "anticipate," "estimate," "believe," "intend," or "project" or the negative of these words or other variations on these words or comparable terminology. The reader is cautioned not to put undue reliance on these forward-looking statements, as these statements are subject to numerous factors and uncertainties outside of our control that can make such statements untrue, including, but not limited to, inadequate capital, adverse economic conditions, intense competition, lack of meaningful research results, entry of new competitors and products, adverse federal, state and local government regulation, termination of contracts or agreements, technological obsolescence of our products, technical problems with our research and products, price increases for supplies and components, inability to carry out research, development and commercialization plans, loss or retirement of key executives and research scientists and other specific risks. We currently have no commercial products intended to diagnose, treat, prevent, or cure any disease. The statements contained in this presentation regarding our ongoing research and development and the results attained by us to-date have not been evaluated by the Food and Drug Administration. There can be no assurance that further research and development, and/or whether clinical trial results, if any, will validate and support the results of our preliminary research and studies. Further, there can be no assurance that the necessary regulatory approvals will be obtained or that we will be able to develop new products on the basis of our technologies. In addition, other factors that could cause actual results to differ materially are discussed in a Proxy Statement filed with the SEC on June 30th, 2014. Investors and security holders are urged to read these documents free of charge on the SEC's web site at www.sec.gov. We undertake no obligation to publicly update or revise our forward-looking statements as a result of new information, future events, or otherwise. 2 VBI – A Novel Vaccine Biotech Co. VBI is developing novel technologies that seek to expand vaccine protection in significant markets of unmet medical need. VBI IN BRIEF Public market debut in late July 2014 with a $16.25M private placement and a $6M secured debt facility. NasdaqCM: VBIV Share Price: $2.68 (12/8/14 – market close) Common Stock Currently Outstanding: 20M, 50-Day Avg Vol: 39,000 (12/8/14) Market Cap: $54M (12/8/14); Cash $14.8M (9/30/14) Headquartered in Cambridge, MA with main research site in Ottawa, Canada. U.S. incorporation in 2006. 3 Opportunity Highlights T W O C O M P L E M E N TA R Y T E C H N O L O G Y P L AT F O R M S Enveloped Virus-Like Particle (eVLP) Technology Platform for the development of thirdgeneration synthetic vaccines that closely resemble the structure of the virus they mimic. Thermostable LPV™ Platform enables the development of vaccines that can withstand storage or shipment at elevated or constantly fluctuating temperatures. Prophylactic cytomegalovirus (CMV) vaccine candidate with strong proof of concept in preclinical studies.1 Large and well-defined market opportunities: CMV market could exceed $1B annually; CMV vaccine doses could exceed 7.6M by 20302 with pricing similar to HPV at $140 per dose.3 Thermostable LPV™ Platform – currently, more than 90% of all vaccines require “cold chain” shipment at 4°C.4 IP strategy protects core platforms; CMV IP extends to 2032 and LPV™ IP extends to 2030. World-class leadership team includes Steve Gillis and Jeff Baxter. 4 Experienced Leadership VBI’s management and board brings vaccine discovery, development, financing, and commercialization expertise. DR. STEVE GILLIS, CHAIRMAN OF THE BOARD Dr. Gillis is a Ph.D. immunologist by training with expertise in molecular and tumor immunology. Previously, he served as the Founder and CEO of Corixa, which was acquired in 2005 by GSK for $300M. Prior to Corixa, Dr. Gillis was Founder and Director of Immunex, acquired in 2002 by Amgen for $16B. Dr. Gillis is a managing Director of ARCH Venture Partners. JEFF BAXTER, PRESIDENT & CEO Mr. Baxter joined VBI in September of 2009. Previously, he was a managing partner of The Column Group, a venture capital firm. Until July 2006, Mr. Baxter was SVP, R&D Finance and Operations, of GlaxoSmithKline (GSK). His 19 year pharma career has spanned finance, commercial operations, manufacturing, R&D operations, and SROne, GSK’s in-house venture fund. 5 Experienced Leadership Cont. Dr. Michel De Wilde, Ph.D, Director was Senior Vice President, R&D, at Sanofi Pasteur, the human vaccines division of Sanofi from 2001 until June 2013. In this position, he was responsible for managing approximately 1,500 employees and a broad portfolio of development projects. Dr. Michael Steinmetz, Ph.D., Director, has been Managing Director of Clarus Ventures since the firm’s inception in 2005. Prior to Clarus, Dr. Steinmetz was a General Partner at MPM Capital, a healthcare venture capital firm. He has over 25 years of healthcare industry investment experience. Sam Chawla, Director, is a Portfolio Manager of Perceptive Advisors LLC, an investment fund focused on the healthcare sector. Prior to joining Perceptive Advisors in 2013, Mr. Chawla was a Managing Director at UBS Investment Bank in the Global Healthcare Group. Dr. David E. Anderson, Ph.D., Senior VP Research, is a dynamic and well-published immunologist with broad expertise in the areas of vaccine development, autoimmunity, and tumor immunology. Dr. Anderson joined VBI full time in 2009 from Harvard Medical School. Egidio Nascimento, CA , Chief Financial Officer, joined VBI in 2005, bringing his experience in finance and accounting at early-stage ventures. Mr. Nascimento previously served as VP of Finance at Genome Canada and as the CFO of two start-up companies. 6 Platform Technologies Two complementary vaccine platform technologies in development. e V L P P L AT F O R M T H E R M O S TA B L E P L AT F O R M Enveloped (e) virus-like particle (VLP) vaccines resemble viruses, but are noninfectious and do not contain viral genetic material. Lipid Particle Vaccines (LPV™) platform enables the development of vaccines that can withstand months of storage or shipment at constantly fluctuating or elevated temperatures. eVLPs are capable of generating strong immune response due to similarity to viruses found in nature. eVLP platform is applicable to diseases; established proof of concept in Flu, HCV, West Nile and CMV.1 Lead candidate is a prophylactic CMV vaccine that, if approved for marketing, is expected to addresses a large unmet market need.2 Completed proof of concept studies on vaccine and biologic targets including Influenza, Rabies, MMR, and Herceptin®like monoclonal antibody.3 LPV™ allows for rapid reformulation of existing vaccines; pursuing self-funding collaborations with undisclosed prospects. 7 eVLP Platform Virus-like Particle Vaccine Evolution Virus-like particle vaccines have evolved considerably since their introduction in early 1990s. 1 S T G E N E R AT I O N Design: Antigens are produced and self-assemble. Key Advantage: Simple structures and repetitive pattern of antigenic epitopes. Key Limitation: Only a very limited number of antigens spontaneously form orderly VLP structures; cannot be applied to all enveloped viruses. Examples: Gardasil®, Cervarix®, Engerix-B®, Recombivax HB®. 2 N D G E N E R AT I O N 3 R D GENERATION – VBI Design: Antigens of interest are covalently attached to the surface of a backbone protein. Design: Common protein backbone and lipid membrane in which the antigen of interest can be expressed. Key Advantage: Can be applied to multiple different target antigens; VLP structure is not limited to the properties of the antigen. Key Limitation: Antigen of interest is artificially bound to the structural protein and not represented in a natural configuration. Example: Qb VLP Platform. Key Advantage: Enables more natural presentation of target antigen within a membrane that more closely mimics a virus; can express multiple target antigens in a single VLP. Limitation: More effort to meet FDA/EMA purification standards. Ideal Candidates: CMV, HCV, Dengue, RSV, West Nile. 9 eVLP Platform Overview eVLPs are a third-generation class of synthetic vaccines that closely resemble the structure of the virus they mimic. e V L P P L AT F O R M H I G H L I G H T S Same size and structure as enveloped viruses. Present antigens in their natural state (lipid bilayer) to provoke an optimal immune response. Demonstrated ability to trigger strong, broadly neutralizing antibodies in multiple preclinical models (CMV, HCV, and Flu).1 Antigenic Protein Internal Protein Provides Viral Structure Natural Lipid Bilayer Antigenic protein retains natural conformation in the Lipid Bilayer Suitable to a wide array of viruses including CMV, HCV, Dengue, RSV, and West Nile. Strong intellectual property estate. Top: eVLP Diagram – the foundation of the eVLP technology is a stable, protein-based core on which additional vaccine antigens of interest can be added; Bottom: Electron Microscopy image of VBI’s CMV eVLP captured at Sripps Institute. 10 eVLP Platform Characteristics eVLP Platform provides VBI with a novel technology designed to address unmet vaccine needs. KEY CHARACTERISTICS Highly Immunogenic: eVLP immune responses comparable to or better than natural infection by closely mimicking structure of target virus.1 Customizable: eVLPs provide ability to rationally design a vaccine by including different antigens and controlling their relative expression. Safe: Unlike live-attenuated vaccines, eVLPs cannot revert back to an infectious state. Commercially Viable: Manufactured and purified using scalable methods (unlike some vectored delivery approaches); demonstrated commercially suitable yields.2 VBI’s Ottawa, Canada-based research facility. 11 eVLP Vaccine Candidates Novel eVLP vaccines with the goal of creating first-in-class or best-inclass preventives or therapeutics. C U R R E N T VA C C I N E C A N D I D AT E S Prophylactic Cytomegalovirus (CMV) Vaccine: CMV is a common virus that can cause serious, life-threatening complications in persons with weakened immune systems. Unborn babies whose mothers become infected with CMV during pregnancy are at particularly high risk of suffering from developmental delays. There is currently no commercially available preventative vaccine for CMV.1 Therapeutic Hepatitis C (HCV) Vaccine: Hepatitis C is an infectious disease affecting primarily the liver, caused by the hepatitis C virus. Chronic infection can lead to scarring of the liver and ultimately to cirrhosis. Approximately 80% of those exposed to the virus develop a chronic infection.2 VBI is seeking to partner or co-develop its HCV therapeutic vaccine candidate. 12 CMV Vaccine Candidate CMV Opportunity Cytomegalovirus (CMV) is a common virus that can cause serious, lifethreatening complications in persons with weakened immune systems; developing a CMV vaccine is a leading public health priority.1 C O N G E N I TA L I N F E C T I O N Unborn babies whose mothers become infected with CMV during pregnancy are at high risk.2 In the U.S., congenital CMV causes one child to become disabled every hour.3 Congenital CMV infection causes more long-term problems and childhood deaths than Down Syndrome or Fetal Alcohol Syndrome.4 IMMUNOCOMPROMISED A primary CMV infection can cause serious disease persons with weakened immune systems5, including: Organ and bone marrow transplant recipients. Cancer patients. Persons receiving immunosuppressive drugs. 14 CMV Opportunity Cont. In the U.S., approximately 40,000 cases of congenital CMV infection occur each year. As a result, approximately 5,000 infants are born with or develop permanent complications, some of them severe.1 C O M P L I C AT I O N S Children Born With or Developing Long-Term Medical Conditions Each Year in the U.S. Hearing Loss Vision Loss Mental Disability 6,000 5,000 5,000 4,000 4,000 Lack of Coordination 3,000 Small Head 2,000 Seizures 1,000 Death 5,500 3,000 200 60 0 Congenital Fetal Alcohol Down Spina Bifida / Pediatric CMV Disease Syndrome Syndrome Anencephaly HIV/AIDS Invasive Hib Cannon, M. J., and K. F. Davis. 2005. Washing our hands of the congenital cytomegalovirus disease epidemic. BMC Public Health 5:70 15 CMV Vaccine Landscape VBI’s CMV vaccine candidate (VBI-1501A) addresses limitations (potency, durability) within the field of past prospects. N O TA B L E C M V VA C C I N E C A N D I D AT E S Competing vaccine candidates tested in earlier Phase I trials have stalled or reverted to preclinical development. A 2009 Phase II Study of a prophylactic CMV vaccine had projected 50% efficacy.1 Natural immunity imparts ~90% protection,2 but vaccine-induced nAb titers against epithelial cell infection were 10X lower.3 Efficacy appeared to wane quickly after the first year.4 16 CMV Vaccine Candidate: VBI-1501A Strong proof of concept demonstrated in animal studies. PRECLINICAL HIGHLIGHTS Neutralization of CMV up to 32X greater than natural immunity in multiple preclinical animal models.1 Structure of the eVLP platform generates stronger neutralizing antibodies than does immunization with the same (gB) target protein alone.2 gB antigen is a major antigen for the induction of neutralizing antibodies against CMV.3 Potent neutralization in fibroblasts and epithelial cells, two clinically-relevant cell types that are susceptible to CMV infection.4 Durable and high titer antibodies anticipated to impart long-lasting immunity in vaccine recipients.5 Potent immunity alone or in combination with alum, a safe, FDA-approved adjuvant.6 17 CMV Vaccine Candidate: Proof of Concept eVLP presentation of the gB antigen improves responses relative to recombinant gB protein; modified form of gB (gB-G) further improves responses beyond that conferred by the eVLPs alone.1 In comparison to standard gB recombinant protein, gB eVLPs demonstrate enhanced potency and improved consistency of responses in epithelial cells. Modified gB-G antigen further enhances the consistency of responses in epithelial cells. Neutralizing antibody titers for individual mice immunized with comparable doses of Recombinant gB, gB eVLPs, or optimized gB-G eVLPs (VBI-1501). 1,000 50% Epithelial cell nAb Titer (1/x) P R E C L I N I C A L R E S U LT S 100 10 1 Recombinant gB gB eVLPs gB-G eVLPs (VBI-1501) 18 CMV Vaccine Candidate: Proof of Concept Cont. After two vaccinations, VBI-1501A induces immunity that greatly exceeds a benchmark for naturally acquired CMV immunity (Cytogam®).1 Pooled sera from vaccinated mice (n=8) tested for the ability to neutralize CMV infection in both Fibroblast and Epithelial cells, two clinically relevant cell types susceptible to CMV infection. Durable responses to the vaccine observed more than six months after the second immunization. 10,000 10,000 Cytogam® VBI-1501A [alum] VBI-1501 Epithelial cell nAb Titre (1/x) gB-G eVLPs produced using a GMP compliant HEK 293 cell line and purified to meet FDA standards. VBI-1501A elicits rapid, potent, and durable neutralizing antibody titers, which exceed naturally acquired levels of immunity (Cytogam®) after two immunizations (weeks 0 and 8) in mice. Fibroblast cell nAb Titre (1/x) P R E C L I N I C A L R E S U LT S 1,000 100 10 1 0 4 8 12 16 20 24 28 32 0 4 8 12 16 20 24 28 32 1,000 100 10 1 Time (weeks) 19 CMV Vaccine Candidate: Phase I Trial Design VBI will follow a well-defined clinical development path; initial human proof of concept is anticipated during Phase I clinical trials. A N T I C I PAT E D T R I A L D E S I G N Target Population: 125 CMV-Negative Healthy Adults Duration: 12 Months with Interim Data at Six Months Q4 2015: Expected IND Approval and Phase I Start Q1 2016: Interim Phase I Data Expected Q3 2016: Final Phase I Data Expected Primary Endpoint: Safety and Tolerability Secondary Endpoint: Anti-CMV nAb Effective in Fibroblasts / Epithelial Cells nAb are neutralizing antibodies that indicate acquired CMV immunity.1 Estimated Cost: $7M 20 CMV Vaccine Candidate: Development Plan VBI’s CMV Vaccine Candidate may demonstrate immunologic proof of concept in humans at an early stage; VBI may have multiple pathways to further product development. C L I N I C A L D E V E L O P M E N T A N D PA R T N E R S H I P O P T I O N S Interim Phase I data is expected in Q1 2016, at which time VBI expects to demonstrate safety, tolerability, and also immunological proof of concept in humans by measuring CMV neutralizing antibodies in fibroblasts and epithelial cells. Upon the receipt of Phase I data, VBI may explore partnering with a major vaccine franchise on a Phase II trial. Phase II human proof of concept can also be achieved with shorter duration trials in the transplant setting. 21 Thermostable Platform Thermostable LPV™ Technology Enables thermostable vaccine delivery, which is expected to increase vaccine access, safety, and efficacy. IMPROVED ACCESS D O S E S PA R I N G U N LO C K S VA LU E Enables safe storage and transfer outside of the refrigerator / freezer “cold chain”.1 Improved stability and immunogenicity makes it possible to deliver a lower effective dose.2 Adds convenience and portability to improve vaccine developer value proposition. 23 LPV™ Market Opportunity Without a constant temperature in a very narrow range above freezing, many vaccines lose their potency, become ineffective, or hazardous. MARKET HIGHLIGHTS TECHNOLOGY BENEFITS Currently, more than 90% of all vaccines require “cold chain” shipment at 4°C.1 Reduced moisture content during lyophilization cycles to prevent moisture ingress – critical to providing stability and thermostability; can reduce protein aggregation.5 Reliance on a cold chain can increase vaccine costs by up to 20%.2 The World Health Organization (WHO) estimates that the global vaccine market was $24B in 2013.3 Proprietary combination of lipids ratios (MPG, DCP and Cholesterol) and methods can be tailored for desired formulation properties. Nearly 50% of lyophilized and 25% of liquid vaccines are wasted each year; one of the biggest contributors to this wastage is disruption of the cold chain. Patent-pending production method – simple and scalable; production consistency indicates commerciallyviable process and technology.6 24 LPV™ Development Plan LPV™ technology can confer thermostability to commercially-available vaccines and to vaccines under development.1 C O M M E R C I A L I Z AT I O N VBI’s formulation team has expertise working with multiple vaccine types. Demonstrated pilot scale manufacture at a GMP compliant contract facility. Currently pursuing LPV™ collaborations with undisclosed potential prospects. All partnerships are expected to be self funding, with upfront and royalty payments anticipated. VBI’s Ottawa, Canada-based research facility. 25 LPV™ Technology Proof of Concept Preclinical proof of concept data has been collected using commercial products acquired through retail channels and through undisclosed collaborations with prospects. PROOF OF CONCEPT Protein subunit vaccine (Influenza)1 12 Months at 40° C Live Biologic Vaccine (MMR)2 8 Weeks at 37° C Monoclonal Antibody Therapeutics3 8 Weeks at 40° C Complex Protein Vaccine (Rabies)4 18 Months at 40° C VBI’s Ottawa, Canada-based research facility. 26 LPV™ Technology Proof of Concept Cont. C O M M E R C I A L F L U VA C C I N E Six month of stability at 40°C was achieved for an LPV ™ formulation of H3N2 seasonal flu vaccine formulated at pilot scale by a contract manufacturing organization.1 1024 HAI Titer (1/x) 512 256 128 64 32 16 8 Storage Condition Formulation Antigen 1 2 3 FLU LPV™ Reference Vaccine 6 Months @ 4°C Reference Vaccine 6 Months @ 40°C 4 FLU LPV™ H3N2 Component of Seasonal Flu Vaccine 27 VBI Opportunity VBI Opportunity Summary Emerging company with experienced leadership, innovative vaccine assets, and runway to meet anticipated near-term milestones. HIGHLIGHTS CMV vaccine candidate with $1B annual market potential.1,2 Additional ‘catch-up’ cohorts could increase initial market size significantly. CMV Phase I clinical development program has the potential to provide human proof of concept by benchmarking against naturally acquired immunity (Cytogam™).3 VBI’s LPV™ technology can confer thermostability to vaccines and biologics; currently more than 90% of all vaccines require “cold chain” shipment at 4°C.4 Strong intellectual property portfolio. World-class leadership team. 29 Anticipated CMV Program Milestones Forecast Timing Milestone Q4 2014 Complete GMP Tech Transfer of VBI-1501A; Initiate GMP Production for Toxicology Q1 2015 Complete GMP Manufacturing for VBI-1501A Q2 2015 Initiate Formal Toxicology Q3 2015 Complete Formal Toxicology Q4 2015 VBI-1501A - IND Approval and Phase I Start Q1 2016 Interim Phase I Data Q3 2016 Final Phase I Data 30 VBI Vaccines, Inc. 222 Third Street, Suite 2241 Cambridge, MA 02142 (617) 830-3031 [email protected] Appendix Intellectual Property IP strategy protects core platforms; CMV IP extends to 2032 and LPV™ IP extends to 2030. IN T ELLEC T UAL PROPERT Y PORT FOLIO Platform IP Licensed eVLP Patents Internally developed LPV Patents Product Specific Embodiments eVLP: HCV, CMV Compositions LPV: Flu, Live Attenuated, Tx Proteins, Etc. Coverage of Manufacturing Processes eVLP: Purification Process LPV: Inverse Melt Process, Lipid Ratios Extension IP Analytical Tools Manufacturing Product IP Coverage of Proprietary Analytical Tools Proprietary Potency Assay Proprietary Reagents Platform IP Second Generation Platform Extension IP Undisclosed Future Efforts 33 LPV™ Technology Proof of Concept C O M M E R C I A L M M R VA C C I N E Eight weeks of stability at 37°C was achieved with an LPV™ formulation of a commercial MMR vaccine. Potency was measured in quadruplicate in VERO cells in vitro after eight weeks of storage. VBI LPV™ formulations (orange) provided greater stability than standard sucrose lyophilized control (blue) or off-the-shelf reference vaccine (green).1 100,000 TCID50/ml 10,000 1,000 100 10 Storage Temperature Formulation Lyophilization Cycle 1 2 4°C 37°C 3 4 4°C 37°C 5 6 4°C 37°C VBI LPV™ Formulation NO LPV™ Formulation Control Reference (Commercial Vaccine) 2 LYO Cycle 2 LYO Cycle 1 LYO Cycle 34 LPV™ Technology Proof of Concept Cont. LPV™ formulation of a HER-2 mAb reduces moisture, increases purity, and maintains potency under stress conditions.1 THERAPEUTIC mAb Liquid bulk drug substance (Her-2-mAb-liquid) was compared to lyophilization in sucrose in the absence (Her-2-mAb-lyo) or presence of the LPV™ lipids (Her-2 mAb-LPV™). Moisture Content Purity (SEC Test) 0.5 100 100 99 90 98 80 Potency (%) 1 Purity (%) Moisture Content (%) 1.5 97 96 0 70 60 95 Her-2mAb Her-2mAb Her-2mAb (liquid) (lyo) (LPV) Potency (After 8 weeks at 40° C) 50 Her-2mAb Her-2mAb Her-2mAb (liquid) (lyo) (LPV) Her-2mAb Her-2mAb Her-2mAb (liquid) (lyo) (LPV) 35 Sources PAGE 4: OPPORTUNITY HIGHLIGHTS 1. VBI Studies: 15BC04, 15BC19; Garrone, P (2011) Sci Transl Med 3, 94ra71; Bellier, B (2009) Vaccine 25, 5772; Szecsi, J (2006) Virol J 3, 70. 2. Estimates adapted from Vakzine Analytics, CMV Marketview, August 2012 3. Vaccine prices adapted from CDC published pricing: http://www.cdc.gov/vaccines/programs/vfc/awardees/vaccinemanagement/price-list/archive.html 4. Biopharma Cold Chain Sourcebook 2010 PAGE 7: PLATFORM TECHNOLOGIES PAGE 12: eVLP VACCINE CANDIDATES 1. http://www.cdc.gov/cmv/trends-stats.html 2. Nelson, PK; Mathers BM; Cowie B; Hagan H; Des Jarlais D; Horyniak D; Degenhardt L (2011-08-13). "Global epidemiology of hepatitis B and hepatitis C in people who inject drugs: results of systematic reviews". Lancet 378 (9791): 571–83. PAGE 14: CMV OPPORTUNITY 1. Stratton KR et al, Committee to Study Priorities for Vaccine Development, Inst. of Med.; Washington, DC 2. http://www.cdc.gov/cmv/trends-stats.html 3. http://www.cdc.gov/CMV/index.html 1. VBI Studies: 15BC04, 15BC19; Garrone, P (2011) Sci Transl Med 3, 94ra71; Bellier, B (2009) Vaccine 25, 5772; Szecsi, J (2006) Virol J 3, 70. 2. Stratton KR et al, Committee to Study Priorities for Vaccine Development, Inst. of Med.; Washington, DC 4. Cannon, M. J., and K. F. Davis. 2005. Washing our hands of the congenital cytomegalovirus disease epidemic. BMC Public Health 5:70 3. VBI Studies: 12BC04 (Flu), 07CH12 (MMR), 07CH17 (Rabies), undisclosed partner feasibility study (biologic) 5. http://www.cdc.gov/CMV/risk/weak-immune.html PAGE 15: CMV OPPORTUNITY CONT. PAGE 10: eVLP PLATFORM OVERVIEW 1. 1. VBI studies 15BC04, 15BC19 (CMV); Garrone, P (2011) Sci Transl Med 3, 94ra71 (HCV); Szecsi, J (2006) Virol J 3, 70 (Flu) Demmler GJ. Congenital cytomegaloviral infection and disease. Semin Pediatr Infect Dis 1999;10:195-200. PAGE 16: CMV VACCINE LANDSCAPE PAGE 12: eVLP PLATFORM CHARACTERISTICS 1. VBI Studies: 15BC04, 15BC19 2. VBI Studies: 15CH19, 15CH38 1. Pass RF (2009) N Eng J Med 360, 1191-1199 2. Adler SP (1995) J Infect Dis 171, 26-32 3. Cui X (2008) Vaccine 26, 5760-5766 4. Lilja AE (2013) Vaccine 30, 6980-6990 36 Sources Cont. PAGE 17: CMV VACCINE CANDIDATE: VBI-1501A PAGE 24: LPV™ MARKET OPPORTUNITY 1. VBI Studies: 15BC19 1. Biopharma Cold Chain Sourcebook 2010 2. VBI Studies: 15BC04 2. World Health Organization (WHO) 3. http://www.ncbi.nlm.nih.gov/pubmed/10466818 3. VBI Studies: 12BC04 4. VBI Studies: 15BC04, 15BC19, 15BC39 4. Thermostability of vaccines, A. Galazka, J. Milstien, M. Zaffran, World Health Organization 1998. 5. VBI Studies: 15BC19 5. VBI Studies: 12BC01, undisclosed partner feasibility study 6. VBI Studies: 15BC19, 15BC3 6. VBI Studies: 12BC04 PAGE 18: CMV VACCINE CANDIDATE: PROOF OF CONCEPT 1. VBI Studies: 15BC04 PAGE 19: CMV VACCINE CANDIDATE: PROOF OF CONCEPT CONT. 1. 1. VBI Studies: VBI Studies 12BC04 (Flu), 07CH12 (MMR), 07CH17 (Rabies) PAGE 26: LPV™ TECHNOLOGY PROOF OF CONCEPT 1. VBI Studies: 12BC04 2. VBI Studies: 07CH12 3. Undisclosed partner feasibility study 4. VBI Studies: 07CH17 VBI Studies: 15BC19 PAGE 20: CMV VACCINE CANDIDATE: PHASE I TRIAL DESIGN 1. PAGE 25: LPV™ DEVELOPMENT PLAN Nozawa, N (2009) J Clin Virol 46S4, S58-63. PAGE 23: THERMOSTABLE LPV™ TECHNOLOGY 1. VBI Studies: 12BC04 (Flu), 07CH12 (MMR), 07CH17 (rabies), undisclosed partner feasibility study (biologic) 2. VBI Studies: 12BC07, 12MK01 37 Sources Cont. PAGE 27: LPV™ TECHNOLOGY PROOF OF CONCEPT CONT. 1. VBI Studies: 12BC04 PAGE 29: VBI OPPORTUNITY SUMMARY 1. Estimates adapted from Vakzine Analytics, CMV Marketview, August 2012 2. Vaccine prices adapted from CDC published pricing. http://www.cdc.gov/vaccines/programs/vfc/awardees/vaccinemanagement/price-list/archive.html 3. Potency of Cytogam™, live-attenuated Towne vaccine and adjuvanted gB subunit vaccine adapted from Cui, et al., 2008 4. Biopharma Cold Chain Sourcebook 2010 PAGE 34: LPV™ TECHNOLOGY PROOF OF CONCEPT CONT. 1. VBI Studies: 07CH12 PAGE 35: LPV™ TECHNOLOGY PROOF OF CONCEPT CONT. 1. Undisclosed partner feasibility study 38
© Copyright 2024 ExpyDoc