US20220226401A1 - Dengue vaccine unit dose and administration thereof - Google Patents

Dengue vaccine unit dose and administration thereof Download PDF

Info

Publication number
US20220226401A1
US20220226401A1 US17/274,016 US201917274016A US2022226401A1 US 20220226401 A1 US20220226401 A1 US 20220226401A1 US 201917274016 A US201917274016 A US 201917274016A US 2022226401 A1 US2022226401 A1 US 2022226401A1
Authority
US
United States
Prior art keywords
dengue
serotype
pfu
concentration
strain
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
US17/274,016
Other languages
English (en)
Inventor
Derek WALLACE
Inge LEFEVRE
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Takeda Pharmaceuticals International AG
Takeda Vaccines Inc
Original Assignee
Takeda Vaccines Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US16/295,611 external-priority patent/US11464815B2/en
Application filed by Takeda Vaccines Inc filed Critical Takeda Vaccines Inc
Priority to US17/274,016 priority Critical patent/US20220226401A1/en
Assigned to TAKEDA VACCINES, INC. reassignment TAKEDA VACCINES, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: TAKEDA PHARMACEUTICALS INTERNATIONAL AG
Assigned to TAKEDA PHARMACEUTICALS INTERNATIONAL AG reassignment TAKEDA PHARMACEUTICALS INTERNATIONAL AG ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: WALLACE, Derek, LEFEVRE, Inge
Publication of US20220226401A1 publication Critical patent/US20220226401A1/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0016Combination vaccines based on diphtheria-tetanus-pertussis
    • A61K39/0018Combination vaccines based on acellular diphtheria-tetanus-pertussis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/76Viruses; Subviral particles; Bacteriophages
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/295Polyvalent viral antigens; Mixtures of viral and bacterial antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/525Virus
    • A61K2039/5252Virus inactivated (killed)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/525Virus
    • A61K2039/5254Virus avirulent or attenuated
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/55Medicinal preparations containing antigens or antibodies characterised by the host/recipient, e.g. newborn with maternal antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/57Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
    • A61K2039/572Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2 cytotoxic response
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/57Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
    • A61K2039/575Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2 humoral response
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/70Multivalent vaccine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/20011Papillomaviridae
    • C12N2710/20034Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/24011Flaviviridae
    • C12N2770/24111Flavivirus, e.g. yellow fever virus, dengue, JEV
    • C12N2770/24122New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/24011Flaviviridae
    • C12N2770/24111Flavivirus, e.g. yellow fever virus, dengue, JEV
    • C12N2770/24134Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/24011Flaviviridae
    • C12N2770/24111Flavivirus, e.g. yellow fever virus, dengue, JEV
    • C12N2770/24171Demonstrated in vivo effect
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/32011Picornaviridae
    • C12N2770/32411Hepatovirus, i.e. hepatitis A virus
    • C12N2770/32434Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to a safe and effective method of inoculation against dengue disease and a corresponding safe and effective dengue vaccine.
  • the present invention relates to a safe and effective method of inoculation against dengue disease irrespective of serostatus and a corresponding safe and effective dengue vaccine.
  • the present invention relates also to unit doses of a dengue vaccine composition and methods for administering a unit dose of a dengue vaccine composition to a subject or a subject population in a broad age group.
  • the present invention is also related to particular concomitant administration regimes, wherein the unit does/vaccine composition is administered concomitantly with one or more of: a yellow fever (YF) vaccine, a hepatitis A vaccine, a human papillomavirus (HPV) vaccine, a combined measles, mumps and rubella (MMR) vaccine, a combined tetanus, diphtheria, and pertussis (whooping cough) (Tdap) vaccine, and/or a combined vaccine for diphtheria, tetanus, pertussis, poliomyelitis and Haemophilus influenzae type b (DTap/IPV/Hib), or any combination of the concomitant administration regimes mentioned above.
  • the unit dose according to this invention provides immune responses against all serotypes of dengue virus, i.e. DENV-1, DENV-2, DENV-3 and DENV-4.
  • Vaccines for protection against viral infections have been effectively used to reduce the incidence of human disease.
  • One of the most successful technologies for viral vaccines is to immunize animals or humans with a weakened or attenuated virus strain (a “live attenuated virus”). Due to limited replication after immunization, the attenuated virus strain does not cause disease. However, the limited viral replication is sufficient to express the full repertoire of viral antigens and can generate potent and long-lasting immune responses to the virus. Thus, upon subsequent exposure to a pathogenic virus strain, the immunized individual is protected from the disease.
  • live attenuated viral vaccines are among the most successful vaccines used in public health.
  • Dengue disease is a mosquito-borne disease caused by infection with a dengue virus. Dengue virus infections can lead to debilitating and painful symptoms, including a sudden high fever, headaches, joint and muscle pain, nausea, vomiting and skin rashes. To date, four serotypes of dengue virus have been identified: dengue-1 (DENV-1), dengue-2 (DENV-2), dengue-3 (DENV-3) and dengue-4 (DENV-4). Dengue virus serotypes 1 ⁇ 4 can also cause dengue hemorrhagic fever (DHF) and dengue shock syndrome (DSS). In the most severe cases, DHF and DSS can be life threatening.
  • DHF dengue hemorrhagic fever
  • DSS dengue shock syndrome
  • Dengue viruses cause 50-100 million cases of debilitating dengue fever, 500,000 cases of DHF/DSS, and more than 20,000 deaths each year, a large portion of which are children. All four dengue virus serotypes are endemic throughout the tropical regions of the world and constitute the most significant mosquito-borne viral threat to humans there. Dengue viruses are transmitted to humans primarily by Aedes aegypti mosquitoes, but also by Aedes albopictus mosquitoes. Infection with one dengue virus serotype results in life-long protection from re-infection by that serotype, but does not prevent secondary infection by one of the other three dengue virus serotypes. In fact, previous infection with one dengue virus serotype may lead to an increased risk of severe disease (DHF/DSS) upon secondary infection with a different serotype.
  • DHF/DSS severe disease
  • Dengvaxia® can enhance, rather than reduce, the risk of severe disease due to dengue infection in individuals who had not been previously infected by a dengue virus (seronegative populations). Therefore, Dengvaxia® is only recommended for use in individuals who had been previously infected with at least one dengue virus serotype (seropositive populations).
  • Dengvaxia® is only for use in people from 9 to 45 years of age who have been infected with dengue virus before and who live in areas where this infection is endemic.
  • Endemic areas are areas where the disease occurs regularly throughout the year.
  • Sridhar S et al Effect of Dengue Serostatus on Dengue Vaccine Safety and Efficacy. N Engl J Med 2018, 379:327-40; and World Health Organization. Dengue vaccine: WHO position paper—September 2018. Wkly. Epidemiol. Rec. 2018, 93:457-476.
  • Dengvaxia® One further disadvantage of the only currently approved dengue vaccine, Dengvaxia®, is that it must only be given to people who have had a positive test result showing a previous infection with dengue virus (EPAR), i.e. individuals with known serostatus for dengue. Thus, individuals with unknown serostatus for dengue cannot be vaccinated with Dengvaxia®.
  • EPAR dengue virus
  • Yellow fever is an acute viral hemorrhagic disease transmitted by infected mosquitoes of the Aedes aegypti specie. Symptoms of yellow fever take 3 to 6 days to develop and include fever, headache, jaundice, muscle pain, nausea, vomiting and fatigue. A small proportion of patients (about 15% of people) who contract the virus develop a severe disease that can lead to bleeding, shock, organ failure, and sometimes death. The virus is endemic in tropical areas of Africa and Central and South America.
  • Dengue fever and yellow fever (YF) viruses belong to the same family of flaviviridae and share antigenic determinants, which may result in cross-reacting antibodies. They are both transmitted between humans by mosquitoes (primarily Aedes aegypti ), and are both endemic in tropical areas of Africa and Latin America with a high public health impact.
  • YF-17D vaccine yellow fever vaccine
  • the YF-17D vaccine is highly effective (approaching 100%) and generally safe with the exception of very rare cases of vaccine-associated neurotropic and viscerotropic disease.
  • Vaccination against YF is also required for travelers to certain countries in accordance with the International Health Regulations, and is also recommended by the WHO for all subjects travelling to areas where there is evidence of persistent or periodic YF virus transmission.
  • a YF-17D vaccine is available under the product name YF-VAX® from Sanofi.
  • a yellow fever vaccine is recommended for people from nine months of age and older who are living in or traveling to endemic areas, persons travelling to or through countries requiring.
  • a single dose of yellow fever vaccine administered subcutaneously is usually sufficient to confer sustained lifelong protective immunity against yellow fever.
  • a booster dose is recommended every 10 years.
  • Hepatitis A is a liver disease caused by the hepatitis A virus (HAV).
  • HAV hepatitis A virus
  • the virus is primarily spread when an uninfected (and unvaccinated) person ingests food or water that is contaminated with the feces of an infected person.
  • the disease is closely associated with unsafe water or food, inadequate sanitation and poor personal hygiene.
  • the virus can also be transmitted through close physical contact with an infectious person.
  • hepatitis A infection does not cause chronic liver disease and is rarely fatal, but it can cause debilitating symptoms and fulminant hepatitis (acute liver failure), which is often fatal.
  • Hepatitis A occurs sporadically and in epidemics worldwide, with a tendency for cyclic recurrences.
  • the hepatitis A virus is one of the most frequent causes of foodborne infection. Epidemics related to contaminated food or water can erupt explosively, such as the epidemic in Shanghai in 1988 that affected about 300,000 people. Hepatitis A viruses persist in the environment and can withstand food-production processes routinely used to inactivate and/or control bacterial pathogens. The disease can lead to significant economic and social consequences in communities. It can take weeks or months for people recovering from the illness to return to work, school, or daily life. The impact on food establishments identified with the virus, and local productivity in general, can be substantial. In developing countries with poor sanitary conditions and hygienic practices, most children (90%) have been infected with the hepatitis A virus before the age of 10 years.
  • hepatitis A vaccination For routine hepatitis A vaccination, a two-dose schedule is recommended, particularly in travelers at substantial risk of contracting hepatitis A and in immunocompromised individuals. However, in healthy individuals, comparable effectiveness has been achieved with a single dose.
  • the vaccination schedule for children/adolescents (12 months through 18 years of age) as well as for adults ( ⁇ 19 years of age) consists of a primary dose administered intramuscularly, and a further booster dose administered intramuscularly 6 to 18 months later.
  • HAVRIX® HAVRIX®
  • VAQTA® Available hepatitis A vaccines
  • HPV Human papillomavirus
  • HPV vaccination prevents HPV-associated cervical cancers as well as HPV-associated cancers of the anus, vulva, vagina, and oropharynx. The vaccination can also prevent HPV-associated genital warts.
  • HPV vaccination is recommended in particular for 11 and 12 year-old girls. It is also recommended for girls and women age 13 through 26 years of age who have not yet been vaccinated or completed the vaccine series. HPV vaccine can also be given to girls beginning at age 9 years. The CDC recommends 11 to 12 year olds girls get two doses of HPV vaccine to protect against cancers caused by HPV. More recently, vaccination of boys in the same age ranges has also been recommended.
  • the routine HPV vaccination schedule for adolescents who start the vaccination series before the 15th birthday includes two doses of a HPV vaccine.
  • the two doses are usually separated by 6 to 12 months.
  • the minimum interval between doses is five calendar months.
  • a three dose schedule is recommended for subjects who start the series on or after the 15th birthday and for subjects with certain immunocompromising conditions (such as cancer, HIV infection, or taking immunosuppressive drugs).
  • the second dose is usually given 1 to 2 months after the first dose and the third dose 6 months after the first dose.
  • the minimum interval between the first and second doses of vaccine is usually 4 weeks.
  • the minimum interval between the second and third doses of vaccine is usually 12 weeks.
  • the minimum interval between the first and third doses is usually 5 calendar months. If the vaccination series is interrupted, the series does not need to be restarted.
  • HPV vaccines include Gardasil® 9, which is a recombinant 9-valent HPV (9vHPV) vaccine for preventing HPV serotypes 6, 11, 16, 18, 31, 33, 45, 52, and 58.
  • the HPV vaccine does not include any live or inactivated HPV, but the L1 proteins of the respective HPV serotypes.
  • Measles is a highly contagious infectious disease caused by the measles virus, a single-stranded, negative-sense, enveloped (non-segmented) RNA virus of the genus Morbillivirus within the family Paramyxoviridae. Complications occur in about 30% of cases and may include diarrhea, blindness, inflammation of the brain, and pneumonia, among others. Encephalitis occurs in approximately one of every 2000 reported cases; survivors often have permanent brain damage and mental retardation. Death, predominantly from respiratory and neurological causes, occurs in one of every 3000 reported measles cases. The risk of death is greater for infants and adults than for children and adolescents. Contracting measles during pregnancy increases fetal risk.
  • Subacute sclerosing panencephalitis a slow virus infection of the central nervous system, is associated with measles virus. Measles is an airborne disease which spreads easily through the coughs and sneezes of infected people and may also be spread through contact with saliva or nasal secretions.
  • Mumps is an acute disease of children and young adults, caused by the mumps virus, a single-stranded, negative-sense RNA virus of the genus Rubulavirus within the family Paramyxoviridae. Mumps virus produces no symptoms in about one-third of infected people. In those with a clinical response, glandular and nerve tissue are most often affected and the most common symptoms include fever and swelling of the parotid glands. Complications may include meningitis (15%), pancreatitis (4%), inflammation of the heart, or permanent deafness. Frequent viruria and abnormal renal function suggest that mumps virus may infect the kidneys. Mumps is highly contagious and spreads rapidly among people living closely together by respiratory droplets or direct contact with an infected person.
  • Rubella German measles
  • the virus usually results in a mild illness, accompanied by few constitutional symptoms, and occurs most commonly in childhood. If the infection occurs in a woman in early pregnancy however, the virus may cross the placenta to reach the fetus, in which the infection can induce birth defects. These defects may be serious and permanent and include congenital heart disease, cataract formation, deafness and mental retardation. Rubella is usually spread through the air via coughs of people who are infected.
  • Combined vaccine for measles, mumps and rubella is used widely for the immunization of children in certain regions of the world, because of its advantages over the individual vaccines. Combined vaccine provokes an adequate immune response in children simultaneously for the three infections.
  • MMR vaccines are indicated for simultaneous vaccination against measles, mumps, and rubella in individuals 12 months of age or older. Individuals first vaccinated at 12 months of age or older should be revaccinated prior to elementary school entry. Revaccination is intended to seroconvert those who do not respond to the first dose.
  • the Advisory Committee on Immunization Practices (ACIP) recommends administration of the first dose at 12 to 15 months of age and administration of the second dose at 4 to 6 years of age.
  • Tetanus is caused by an infection with the bacterium Clostridium tetani which is commonly found in soil, saliva, dust, and manure.
  • Clostridium tetani which is commonly found in soil, saliva, dust, and manure.
  • the bacteria generally enter the body through a break in the skin such as a cut or puncture wound by a contaminated object.
  • the bacteria produce toxins that interfere with muscle contractions, resulting in the typical signs of muscle spasms. It affects the brain and nervous system and causes extremely painful muscle spasms, usually all over the body. Spasms of the jaw can make it impossible to open the mouth, a condition called “lockjaw.” Tetanus kills one out of ten people infected with the disease.
  • Diphtheria is an infection caused by the bacterium Corynebacterium diphtheriae which primarily infects the throat and upper airways, and produces a toxin affecting other organs. Diphtheria has an acute onset and the main characteristics are sore throat, low fever and swollen glands in the neck. The toxin may, in severe cases, cause myocarditis or peripheral neuropathy. The diphtheria toxin causes a membrane of dead tissue to build up over the throat and tonsils, making breathing and swallowing difficult. Diphtheria is a very contagious infection and the bacteria usually spread between people by direct contact or through the air, but it may also be spread by contaminated objects.
  • Pertussis or whooping cough, caused by the bacterium Bordetella pertussis is an airborne disease that results in an extremely contagious respiratory infection that can lead to severe breathing problems, especially in infants.
  • Pertussis first appears like an ordinary cold, but then causes intense, uncontrollable coughing spells which can cause difficulty breathing, vomiting, and disturbed sleep.
  • a person may cough so hard that they vomit, break ribs, or become very tired from the effort. Children less than one year old may have little or no cough and instead have periods where they do not breathe.
  • a high-pitched “whoop” noise is heard when the person tries to take a breath after coughing.
  • Complications include pneumonia or death.
  • Pertussis can affect people of all ages, but can be very serious and even deadly, for babies less than a year old.
  • Tdap is a combination vaccine that protects against the three potentially life-threatening bacterial diseases tetanus, diphtheria, and pertussis (whooping cough).
  • Tdap stands for tetanus and diphtheria toxoids with acellular pertussis.
  • Tdap is an inactive vaccine produced by using dead bacteria. It is recommended that vaccination against tetanus, diphtheria and pertussis carried out in infancy (in children of less than 7 years of age) is done by using a particular diphtheria toxoid, tetanus toxoid and acellular pertussis absorbed vaccine, such as INFANRIX® from GlaxoSmithKline.
  • a further booster Tdap vaccine is recommended for children of greater than 10 years to ensure that immunity against tetanus, diphtheria and pertussis is maintained into adulthood.
  • a booster Tdap vaccine based on combined tetanus toxoid, reduced diphtheria toxoid and acellular pertussis (adsorbed) is available under the brand name BOOSTRIX® from GlaxoSmithKline. Tdap vaccination is administered intramuscularly as a single dose.
  • poliomyelitis In addition to diphtheria, tetanus and pertussis, poliomyelitis, often called polio or infantile paralysis, is a highly infectious viral disease caused by the poliovirus. It invades the nervous system, and can cause total paralysis in a matter of hours. The virus is transmitted by person-to-person spread mainly through the fecal-oral route or, less frequently, by a common vehicle (for example, contaminated water or food) and multiplies in the intestine. Initial symptoms are fever, fatigue, headache, vomiting, stiffness of the neck and pain in the limbs. 1 in 200 infections leads to irreversible paralysis (usually in the legs). Among those paralyzed, 5% to 10% die when their breathing muscles become immobilized. Polio mainly affects children under 5 years of age. There is no cure for polio, it can only be prevented.
  • Hib Haemophilus influenzae type b
  • Hib Haemophilus influenzae type b
  • Hib is a bacteria responsible for severe pneumonia, meningitis and other invasive diseases almost exclusively in children aged less than 5 years. It is transmitted through the respiratory tract from infected to susceptible individuals. Hib also causes potentially severe inflammatory infections of the face, mouth, blood, epiglottis, joints, heart, bones, peritoneum, and trachea. Although this problem occurs worldwide the burden of Hib disease was considerably higher in resource-poor countries, prior to the introduction of the vaccine into their national immunization programs. In 2000, Hib was estimated to have caused two to three million cases of serious disease, notably pneumonia and meningitis, and 386,000 deaths in young children. Hib disease is observed in all parts of the world but is difficult to confirm because it requires prompt laboratory investigation in patients that have not received prior antibiotic treatment.
  • Pentacel® which is based on combined diphtheria toxoid, tetanus toxoid, acellular pertussis (adsorbed), and inactivated poliovirus in combination with a Hib conjugate vaccine.
  • the present invention is therefore directed in part to a dengue vaccine for a method of mass vaccination without individual testing for serostatus or prior evaluation of seroprevalence rates.
  • This mass vaccination includes endemic regions including outbreak situations as well as in non-endemic regions for travelers.
  • the present invention is therefore directed to a tetravalent dengue virus composition including four live attenuated dengue virus strains representing serotype 1, serotype 2, serotype 3 and serotype 4, wherein preferably the tetravalent dengue virus composition includes a chimeric dengue serotype 2/1 strain and a dengue serotype 2 strain and a chimeric dengue serotype 2/3 strain and a chimeric dengue serotype 2/4 strain, wherein preferably the dengue serotype 2 strain is derived from the wild type virus strain DEN-2 16681 (represented by SEQ ID NO 11) and differs in at least three nucleotides from the wild type as follows:
  • the three chimeric dengue strains being derived from the serotype 2 strain by replacing the structural proteins prM and E from serotype 2 strain with the corresponding structural proteins from the other dengue serotypes, resulting in the following chimeric dengue strains:
  • composition or lyophilized unit dose which upon reconstitution with 0.5 mL of a pharmaceutically acceptable diluent comprises:
  • a dengue serotype 1 in a concentration of at least 3.3 log 10 pfu/0.5 ml
  • a dengue serotype 2 in a concentration of at least 2.7 log 10 pfu/0.5 ml
  • a dengue serotype 3 in a concentration of at least 4.0 log 10 pfu/0.5 ml
  • a dengue serotype 4 in a concentration of at least 4.5 log 10 pfu/0.5 ml.
  • the present invention is also in particular directed to such a composition or unit dose which upon reconstitution with a pharmaceutically acceptable diluent (i), (ii), (iii), and (iv) provide a total concentration of pfu/0.5 mL and based on said total concentration the concentration of (ii) in pfu/0.5 mL is less than 10%, and the concentration of (iv) in pfu/0.5 mL is at least 50%, and the concentration of (i) in pfu/0.5 mL is at least 1%, and the concentration of (iii) in pfu/0.5 mL is at least 8%, or preferably at least 10%, or at least 12%, or at least 14%, or at least 16%, or at least 18%.
  • the present invention is also in particular directed to such method and use wherein upon reconstitution with a pharmaceutically acceptable diluent (i), (ii), (iii), and (iv) provide a total concentration of pfu/0.5 mL and based on said total concentration the concentration of (ii) in pfu/0.5 mL is less than 2%, the concentration of (iv) in pfu/0.5 mL is at least 50%, the concentration of (i) in pfu/0.5 mL is at least 1%, and the concentration of (iii) in pfu/0.5 mL is at least 6%.
  • the present invention is therefore directed to a method including and corresponding use of a tetravalent dengue virus composition including four live attenuated dengue virus strains representing serotype 1, serotype 2, serotype 3 and serotype 4, the method being directed to inoculating a subject against virologically confirmable dengue disease, wherein in particular the tetravalent dengue virus composition includes a chimeric dengue serotype 2/1 strain and a dengue serotype 2 strain and a chimeric dengue serotype 2/3 strain and a chimeric dengue serotype 2/4 strain, wherein in particular the dengue serotype 2 strain is derived from the wild type virus strain DEN-2 16681 (represented by SEQ ID NO 11) and differs in at least three nucleotides from the wild type as follows:
  • the three chimeric dengue strains being derived from the serotype 2 strain by replacing the structural proteins prM and E from serotype 2 strain with the corresponding structural proteins from the other dengue serotypes, resulting in the following chimeric dengue strains:
  • the present invention is in particular directed to such method and use wherein the unit dose is lyophilized and upon reconstitution with 0.5 mL of a pharmaceutically acceptable diluent comprises:
  • a dengue serotype 1 in a concentration of at least 3.3 log 10 pfu/0.5 ml
  • a dengue serotype 2 in a concentration of at least 2.7 log 10 pfu/0.5 ml
  • a dengue serotype 3 in a concentration of at least 4.0 log 10 pfu/0.5 ml
  • a dengue serotype 4 in a concentration of at least 4.5 log 10 pfu/0.5 ml.
  • the present invention is also in particular directed to such method and use wherein upon reconstitution with a pharmaceutically acceptable diluent (i), (ii), (iii), and (iv) provide a total concentration of pfu/0.5 mL and based on said total concentration the concentration of (ii) in pfu/0.5 mL is less than 10%, and the concentration of (iv) in pfu/0.5 mL is at least 50%, and the concentration of (i) in pfu/0.5 mL is at least 1%, and the concentration of (iii) in pfu/0.5 mL is at least 8%, or at least 10%, or at least 12%, or at least 14%, or at least 16%, or at least 18%, and wherein the subject is preferably 2 to 17 years of age or 4 to 16 years of age.
  • the present invention is also in particular directed to such method and use wherein upon reconstitution with a pharmaceutically acceptable diluent (i), (ii), (iii), and (iv) provide a total concentration of pfu/0.5 mL and based on said total concentration the concentration of (ii) in pfu/0.5 mL is less than 2%, the concentration of (iv) in pfu/0.5 mL is at least 50%, the concentration of (i) in pfu/0.5 mL is at least 1%, and the concentration of (iii) in pfu/0.5 mL is at least 6%, wherein the subject is 18 to 60 years of age.
  • the present invention is therefore directed to a method and corresponding use, the method comprising a primary vaccination with only two administrations of the unit dose comprising the steps of:
  • such a method and use do not include the active surveillance with respect to febrile illness of the subject after the administration of the first- and second-unit dose.
  • active surveillance any subject with febrile illness (defined as fever ⁇ 38° C. on any 2 of 3 consecutive days) will be asked to return to the site for dengue fever evaluation by the Investigator.
  • Subjects/guardians will be contacted at least weekly to ensure robust identification of febrile illness by reminding subjects/guardians of their obligation to return to the site in case of febrile illness. This contact will be implemented through appropriate methods that may differ in each trial site (eg, phone calls, text messaging, home visits, school-based surveillance).
  • such a method and use do not include vaccine immunogenicity analysis including GMTs for dengue neutralizing antibodies.
  • Such a method and use do not include a reactogenicity analysis.
  • a reactogenicity analysis relates to solicited local AEs (injection site pain, injection site erythema, and injection site swelling) and solicited systemic AEs (child ⁇ 6 years: fever, irritability/fussiness, drowsiness and loss of appetite; child ⁇ 6 years: asthenia, fever, headache, malaise and myalgia) which will e.g. be assessed for 7 days and 14 days, respectively, following each vaccination (vaccination day included) via collection of diary cards.
  • the method according to the invention does not require the testing of the serostatus before vaccination and thus allows immediate treatment and outbreak control.
  • the invention is directed to a method and use wherein the subject is exposed to a dengue outbreak.
  • the outbreak is due to a dengue serotype 2, and/or due to a serotype 1.
  • the method of inoculation against the virologically confirmable dengue disease is due to a dengue serotype 2, and/or due to a dengue serotype 1.
  • A selecting a subject for administration of the unit doses of the tetravalent dengue virus composition in need for protection against dengue infection without determination of a previous dengue infection, and (B) administering a first unit dose of the tetravalent dengue virus composition to the subject, and (C) administering a second unit dose of the tetravalent dengue virus composition to the subject within 3 months of administration of the first unit dose. Therefore the method of inoculating is finalized without determination of a previous dengue infection.
  • the method further optionally comprises at least 1 years after the administration of the second unit dose a booster dose of the unit dose.
  • the method and use is directed to a method of inoculating a subject against virologically confirmable dengue disease with a tetravalent dengue virus composition including four live attenuated dengue virus strains representing serotype 1, serotype 2, serotype 3 and serotype 4, the dengue serotype 2 strain being derived from the wild type virus strain DEN-2 16681 (represented by SEQ ID NO 11) and differing in at least three nucleotides from the wild type as follows:
  • the method comprises a primary vaccination consisting of the steps of:
  • the method and use is applicable to subjects of all kinds of ages.
  • the present invention is in particular directed to such a method and use wherein the subject is under 9 years of age, or 4 to 5 years of age, or 6 to 11 years of age, or 12 to 16 years, or 6 to 16 years of age, or 4 to 16 years of age, or 2 to 17 years of age, or 9 years of age, or over 9 years of age, or 9 to 17 years of age, or 18 to 60 years of age, or 18 to 45 years of age, or 46 to 60 years of age, or over 60 years of age.
  • the present invention is directed to such use wherein the method is safe.
  • the present invention is directed to such use wherein the method provides a combined vaccine efficacy against virologically-confirmed dengue with hospitalization against all four serotypes with a 2-sided 95% confidence interval, wherein the lower bound is more than 65%, when measured against placebo in a subject population of at least 5,000 healthy 4 to 16 year old subjects irrespective of serostatus at baseline from first administration of the administration schedule until 12 to 18 months after the last administration of the administration schedule.
  • the present invention is directed to such use wherein the method is effective.
  • the present invention is directed to such use wherein the method provides a combined vaccine efficacy against all four serotypes, in preventing virologically confirmable dengue disease with a 2-sided 95% confidence interval, wherein the lower bound is more than 60%, when measured against placebo in a subject population of at least 5,000 healthy subjects irrespective of serostatus at baseline and 14 to 16 years of age, from the first administration of the administration schedule until 18 months after the last administration of the administration schedule.
  • the present invention is also directed in part to a reconstituted dengue vaccine composition for use in a method of preventing virologically confirmable dengue disease in a subject comprising consecutively administering at least a first and a second unit dose of the dengue vaccine composition to the subject, wherein said first and second unit dose are administered subcutaneously within 3 months and at least 4 weeks apart, optionally at about day 1 and at about day 90, wherein the dengue vaccine composition is a tetravalent dengue virus composition including four dengue virus strains representing dengue serotype 1, dengue serotype 2, dengue serotype 3 and dengue serotype 4, optionally wherein the dengue virus strains are live, attenuated, and wherein upon reconstitution with 0.5 mL of a pharmaceutically acceptable diluent
  • the present invention is therefore directed in part to a dengue vaccine composition for use in a method of preventing virologically confirmable dengue disease (VCD) in a subject comprising consecutively administering at least a first and a second unit dose of the dengue vaccine composition to the subject, wherein said first and second unit dose are administered subcutaneously within 3 months and at least 4 weeks apart, optionally at about day 1 and at about day 90, and wherein the dengue vaccine composition is a tetravalent dengue virus composition including four live, attenuated dengue virus strains representing dengue serotype 1, dengue serotype 2, dengue serotype 3 and dengue serotype 4, wherein the attenuated dengue virus strains comprise chimeric dengue viruses and preferably at least one non-chimeric dengue virus, and wherein the dengue serotype 1 and the dengue serotype 2 are present each in a concentration based on the total concentration in pfu/0.5 mL which is within 5%-points of each other and/or are together less than
  • the present invention is therefore directed in part to a unit dose of a dengue vaccine composition and use thereof, the unit dose comprising:
  • a tetravalent dengue virus composition including four live attenuated dengue serotypes (e.g. virus strains):
  • the present invention is further directed in part to a unit dose of a dengue vaccine composition and use thereof, the unit dose comprising:
  • a tetravalent virus composition including four live attenuated dengue virus strains:
  • the present invention is further directed in part to a unit dose of a dengue vaccine composition and use thereof, the unit dose comprising:
  • a tetravalent virus composition including four live attenuated dengue virus strains, wherein the unit dose is lyophilized and upon reconstitution with 0.5 mL of a pharmaceutically acceptable diluent comprises:
  • the present invention is further directed in part to a unit dose of a dengue vaccine composition and use thereof, wherein said unit dose is lyophilized and obtained by lyophilizing 0.5 mL of a solution comprising:
  • a tetravalent virus composition including four live attenuated dengue virus strains:
  • the present invention is in particular directed to such unit dose or composition wherein upon reconstitution with a pharmaceutically acceptable diluent (i), (ii), (iii), and (iv) provide a total concentration of pfu/0.5 mL and based on said total concentration the concentration of (ii) in pfu/0.5 mL is less than 10%, and the concentration of (iv) in pfu/0.5 mL is at least 50%, and the concentration of (i) in pfu/0.5 mL is at least 1%, and the concentration of (iii) in pfu/0.5 mL is at least 8%, or at least 10%, or at least 12%, or at least 14%, or at least 16%, or at least 18%, in particular wherein the concentration of (iii) in pfu/0.5 mL is at least 10%.
  • the present invention is in particular directed to such unit dose or composition, wherein upon reconstitution with a pharmaceutically acceptable diluent (i), (ii), (iii), and (iv) provide a total concentration of pfu/0.5 mL and based on said total concentration the concentration of (ii) in pfu/0.5 mL is less than 2%, the concentration of (iv) in pfu/0.5 mL is at least 50%, the concentration of (i) in pfu/0.5 mL is at least 1%, and the concentration of (iii) in pfu/0.5 mL is at least 6%.
  • the present invention is further directed in part to a kit for preparing a reconstituted unit dose and use thereof, the kit comprising the following components:
  • the present invention is further directed in part to a container, such as a vial, comprising one to ten unit doses of the present invention as described herein.
  • the present invention is further directed to a method of preventing dengue disease in a subject comprising administering to the subject a reconstituted unit dose of a dengue vaccine composition as described herein, for example by subcutaneous injection.
  • the present invention is further directed to the use of the reconstituted unit dose of a dengue vaccine composition as described herein for the manufacture of a medicament for preventing dengue disease in a subject, for example by subcutaneous injection.
  • the present invention is further directed to the reconstituted unit dose of a dengue vaccine composition as described herein for use in a method of preventing dengue disease in a subject, for example by subcutaneous injection.
  • the present invention is further directed to a method of preventing dengue disease in a subject population, comprising administering to the subject population a reconstituted unit dose of a vaccine composition as described herein, wherein the subject population is seronegative to all dengue serotypes.
  • the geometric mean neutralizing antibody titers (GMTs) when tested in at least 40, or at least 50, or at least 60 subjects at day 180 or day 365 after at least a first administration of said unit dose, and optionally a second administration of said unit dose 90 days after said first administration may provide a ratio of not more than 50, or not more than 40, or nor more than 30, or not more than 20 for the GMT of dengue serotype 2 to the GMT of dengue serotype 4 (GMT DENV-2:GMT DENV-4), and optionally a ratio of not more than 20 for the GMT of dengue serotype 2 to the GMT of dengue serotype 1 (GMT DENV-2:GMT DENV-1), and optionally a ratio of not more than 20 for the GMT of dengue serotype 2 to the GMT of dengue serotype 3 (GMT DENV-2:GMT DENV-3).
  • the present invention is further directed to a method of preventing dengue disease in a subject, comprising administering to the subject a reconstituted unit dose of a vaccine composition as described herein, wherein the subject is seronegative to all dengue serotypes.
  • the neutralizing antibody titers when tested in the subject at day 180 or day 365 after at least a first administration of said unit dose, and optionally a second administration of said unit dose 90 days after said first administration may provide a ratio of not more than 50, or not more than 40, or nor more than 30, or not more than 20 for the neutralizing antibody titer of dengue serotype 2 to the neutralizing antibody titer of dengue serotype 4, and optionally a ratio of not more than 20 for the neutralizing antibody titer of dengue serotype 2 to the neutralizing antibody titer of dengue serotype 1, and optionally a ratio of not more than 20 for the neutralizing antibody titer of dengue serotype 2 to the neutralizing antibody titer of den
  • the methods of preventing dengue disease of the present invention are not associated with an increased likelihood of solicited systemic adverse events, such as in children under 9 or seronegative individuals.
  • the present invention is therefore directed in part to a method of preventing dengue disease as well as yellow fever.
  • the present invention is therefore directed in part to a method of preventing dengue disease as well as hepatitis A.
  • the present invention is therefore directed in part to a method of preventing dengue disease as well as
  • HPV-associated cancers or genital warts HPV-associated cancers or genital warts.
  • the present invention is therefore directed in part to a method of preventing dengue disease as well as measles, mumps and rubella.
  • the present invention is therefore directed in part to a method of preventing dengue disease as well as tetanus, diphtheria, and pertussis.
  • the present invention is therefore directed in part to a method of preventing dengue disease as well as diphtheria, tetanus, pertussis, poliomyelitis and diseases caused by Haemophilus influenzae type b.
  • the present invention is therefore also directed in part to a method of prevention any combination of the above mentioned diseases.
  • the present invention is further directed in part to the use of the reconstituted unit dose of a dengue vaccine composition/tetravalent dengue virus composition as described herein for the manufacture of a medicament for preventing dengue disease in a subject, for example by subcutaneous injection.
  • the present invention is further directed in part to the reconstituted unit dose of a dengue vaccine composition/tetravalent dengue virus composition as described herein for use in a method of preventing dengue disease in a subject, as described herein.
  • unit dose of a dengue vaccine composition refers to the amount of a dengue vaccine which is administered to a subject in a single dose.
  • one unit dose is present in a vial and this unit dose is administered to a subject, e.g. optionally after reconstitution.
  • more than one unit dose of the dengue vaccine composition may be present in a vial so that with the content of one vial more than one subject can be vaccinated.
  • a “lyophilized unit dose” or “unit dose in lyophilized form” refers to the unit dose that is obtained by subjecting a given volume of the liquid dengue vaccine composition, such as 0.5 mL, to lyophilization.
  • the aqueous formulations of the dengue vaccine composition being produced by combining the pharmaceutically acceptable excipients and the dengue virus composition comprising the four dengue virus strains, preferably TDV-1 to TDV-4, is subjected to lyophilization to obtain the lyophilized unit dose.
  • a “reconstituted unit dose” or “unit dose in reconstituted form” is obtained from the lyophilized dose by reconstitution with a pharmaceutically acceptable diluent.
  • the diluent does not contain dengue virus.
  • the reconstituted unit dose is a liquid which can be administered to a subject, for example by injection, such as subcutaneous injection.
  • the term “upon reconstitution with 0.5 mL” is not limiting the reconstitution to be performed using 0.5 mL of the diluent, but refers to the concentration of the dengue viruses that will be present in the reconstituted unit dose when 0.5 mL diluent are used for reconstitution. While using a different volume for reconstitution (e.g. 0.8 mL) will result in a different concentration of dengue viruses in the reconstituted unit dose, the administration of the total volume of the unit dose (e.g. 0.8 mL) will result in the same total amount of dengue virus being administered.
  • a “concentration of at least X log 10 pfu/0.5 mL” refers to the concentration of a dengue serotype in 0.5 mL, but is not limiting the unit dose to be 0.5 mL. If the unit dose has a volume different than 0.5 mL, or is lyophilized from a volume different than 0.5 mL, or is reconstituted with a volume different than 0.5 mL, said concentration will differ from the “concentration of at least X log 10 pfu/0.5 mL”.
  • the unit dose has a volume of 0.5 mL, or is lyophilized from a volume of 0.5 mL, or is reconstituted with a volume of 0.5 mL, said concentration will be the “concentration of at least X log 10 pfu/0.5 mL”.
  • concentration may differ, the total amount of virus in the unit dose remains the same.
  • dengue serotype refers to a species of dengue virus which is defined by its cell surface antigens and therefore can be distinguished by serological methods known in the art. At present, four serotypes of dengue virus are known, i.e. dengue serotype 1 (DENV-1), dengue serotype 2 (DENV-2), dengue serotype 3 (DENV-3) and dengue serotype 4 (DENV-4).
  • tetravalent dengue virus composition refers to a dengue virus composition comprising four different immunogenic components from the four different dengue serotypes DENV-1, DENV-2, DENV-3 and DENV-4, preferably comprising four different live, attenuated dengue viruses, each representing one dengue serotype, and which aims to stimulate immune responses to all four dengue serotypes.
  • live attenuated dengue virus refers to a viable dengue virus which is mutated to provide reduced virulence.
  • the live attenuated dengue virus can be a dengue virus in which all components are derived from the same dengue serotype or it can be a chimeric dengue virus having parts from two or more dengue serotypes.
  • a “virus strain” and in particular a “dengue virus strain” is a genetic subtype of a virus, in particular of a dengue virus, which is characterized by a specific nucleic acid sequence.
  • a dengue serotype may comprise different strains with different nucleic acid sequences which have the same cell surface antigens.
  • a dengue virus strain can be a dengue virus in which all components are derived from the same dengue serotype or it can be a chimeric dengue virus having parts from two or more dengue serotypes.
  • TDV-2 refers to a molecularly characterized and cloned dengue serotype 2 strain derived from the live attenuated DEN-2 PDK-53 virus strain.
  • the PDK-53 strain is described for example in Bhamarapravati et al. (1987) Bulletin of the World Health Organization 65(2): 189-195.
  • the TDV-2 strain served as a backbone for the chimeric TDV-1, TDV-3 and TDV-4 strains into which parts from the TDV-1, TDV-3 and TDV-4 strains were introduced.
  • non-chimeric dengue virus or “non-chimeric dengue serotype strain” or “non-chimeric dengue strain” comprises only parts from one dengue serotype.
  • a non-chimeric dengue virus does not include parts from a different flavivirus such as yellow fever virus, Zika virus, West Nile virus, Japanese encephalitis virus, St. Louis encephalitis virus, tick-borne encephalitis virus.
  • TDV-2 is an example of a non-chimeric dengue virus.
  • a “chimeric dengue virus” or “chimeric dengue serotype strain” or “chimeric dengue strain” comprises parts from at least two different dengue serotypes.
  • the chimeric dengue virus does not include parts from a different flavivirus such as yellow fever virus, Zika virus, West Nile virus, Japanese encephalitis virus, St. Louis encephalitis virus, tick-borne encephalitis virus.
  • the chimeric dengue virus described herein does not include parts from the yellow fever virus.
  • a “chimeric dengue serotype 2/1 strain” or “DENV-2/1 chimera” or “TDV-1” refers to a dengue virus chimeric construct which comprises parts from both DENV-2 and DENV-1.
  • a “chimeric dengue serotype 2/3 strain” or “DENV-2/3 chimera” or “TDV-3” refers to a dengue virus chimeric construct which comprises parts from both DENV-2 and DENV-3.
  • the prM and E proteins from DENV-3 replace the prM and E proteins from DENV-2 as detailed below.
  • a “chimeric dengue serotype 2/4 strain” or “DENV-2/4 chimera” or “TDV-4” refers to a dengue virus chimeric construct which comprises parts from both DENV-2 and DENV-4.
  • the prM and E proteins from DENV-4 replace the prM and E proteins from DENV-2 as detailed below.
  • TDV refers to a tetravalent live attenuated dengue vaccine that comprises a mixture of the four live attenuated dengue virus strains TDV-1, TDV-2, TDV-3 and TDV-4 expressing surface antigens from the four dengue serotypes DENV-1, DENV-2, DENV-3 and DENV-4, respectively.
  • TDV-1 has the nucleotide sequence according to SEQ ID No. 1 and/or the amino acid sequence according to SEQ ID No. 2.
  • TDV-2 has the nucleotide sequence according to SEQ ID No. 3 and/or the amino acid sequence according to SEQ ID No. 4.
  • TDV-3 has the nucleotide sequence according to SEQ ID No. 5 and/or the amino acid sequence according to SEQ ID No. 6. In one embodiment, TDV-4 has the nucleotide sequence according to SEQ ID No. 7 and/or the amino acid sequence according to SEQ ID No. 8.
  • dengue disease refers to the disease which is caused by infection with dengue virus. Symptoms of dengue disease include sudden high fever, headaches, joint and muscle pain, nausea, vomiting and skin rashes. The term dengue disease also includes the more severe forms of dengue hemorrhagic fever (DHF) and dengue shock syndrome (DSS). Symptoms of DHF include increased vascular permeability, hypovolemia and abnormal blood clotting mechanisms. Subjects with DHF may present with severe manifestations of plasma leakage and hemorrhage. When a subject with DHF experiences shock he or she will be categorized as having DSS. Symptoms of DSS include bleeding that may appear as tiny spots of blood on the skin and larger patches of blood under the skin.
  • DHF dengue hemorrhagic fever
  • DSS dengue shock syndrome
  • DHF cases are defined as VCD cases meeting WHO 1997 DHF criteria.
  • preventing dengue disease preferably includes preventing DHF and/or DSS.
  • preventing dengue disease preferably includes preventing severe end-organ manifestations of dengue such as hepatomegaly and acute renal failure.
  • preventing dengue disease refers to preventing a subject from developing one or more symptoms of dengue disease because of an infection with a dengue virus.
  • preventing dengue disease is achieved by vaccinating or inoculating a subject with a dengue vaccine composition, such as the reconstituted unit dose described herein.
  • a dengue vaccine composition such as the reconstituted unit dose described herein.
  • prophylactically treating dengue disease is equivalent to “preventing dengue disease”.
  • preventing dengue disease includes preventing DHS and/or DSS.
  • VCD fever refers to febrile illness or illness clinically suspected to be dengue disease with a positive serotype-specific reverse transcriptase polymerase chain reaction (RT-PCR).
  • RT-PCR reverse transcriptase polymerase chain reaction
  • the term “virologically confirmable dengue” disease refers to a subject having febrile illness or illness clinically suspected to be dengue disease, wherein testing the subject, e.g. using RT-PCR, would confirm the presence of at least one dengue serotype. Severe forms of VCD fever will be identified as follows: Dengue Hemorrhagic Fever (DHF) was defined according to the WHO 1997 criteria.
  • DHF Dengue Hemorrhagic Fever
  • Severe dengue was defined through an assessment of an independent Dengue Case Adjudication Committee which will assess all hospitalized VCD cases (severe/non-severe) based on criteria redefined in a charter. All non-hospitalized cases are considered non-severe.
  • the term “febrile illness” is defined as temperature 38° C. on any 2 of 3 consecutive days.
  • the terms “virologically-confirmed dengue disease with hospitalization”, is considered to be a surrogate for severe dengue and the “incidence of virologically-confirmed dengue disease with hospitalization” is used as a safety parameter.
  • the “relative risk with respect to virologically-confirmed dengue disease with hospitalization” means the number of events of virologically confirmed dengue disease with hospitalization divided by the number of subjects treated with the unit dose as disclosed herein over the number of events of virologically confirmed dengue disease with hospitalization divided by the number of subjects treated with placebo.
  • the vaccine provides for the same or less risk for virologically-confirmed dengue disease with hospitalization as placebo and is considered “safe”.
  • the risk of virologically-confirmed dengue disease with hospitalization may be also 0.9 or less, 0.8 or less, 0.7 or less, 0.6 or less, 0.5 or less, 0.4 or less, 0.3 or less, 0.2 or less, or 0.1 or less, in particular when determined from 30 days after a second administration until 12 months after a second administration, in particular when determined in age groups selected from the age group of 4 to 16 year old subjects, the age group of 4 to under 9 year old subjects, the age group of 2 to under 9 year old subjects, the age group of 4 to 5 year old subjects, the age group of 6 to 11 year old subjects, and the age group of 12 to 16 year old subjects.
  • a vaccine is considered “safe” when the vaccine efficacy (VE) with respect to virologically-confirmed dengue disease with hospitalization is 0% or higher. This means that the vaccine provides for the same likelihood or less for virologically-confirmed dengue disease with hospitalization as placebo.
  • safety is the combined vaccine efficacy against virologically-confirmed dengue with hospitalization against all four serotypes with a 2-sided 95% confidence interval, wherein the lower bound is more than 25%, in particular when measured against placebo in a subject population of at least 1,500 or at least 2,000 healthy subjects (in particular when measured in age groups selected in particular from the age group of 4 to 16 year old subjects, the age group of 4 to under 9 year old subjects, the age group of 2 to under 9 year old subjects, the age group of 4 to 5 year old subjects, the age group of 6 to 11 year old subjects, and the age group of 12 to 16 year old subjects) being seronegative against all serotypes at baseline or being seropositive against at least one serotype at baseline, in particular when said unit dose or said placebo is administered at least twice within less than 6 months, such as within 3 months, about from first administration or from 30 days after the second or last administration of the administration schedule until at least 12 months, until 12 to 18 months, until 12 months, or until 18 months after the second or last
  • the lower bound may be more than 30%, more than 40%, more than 50%, more than 60%, more than 65%, more than 66%, more than 67%, more than 68% more than 70%, or more than 75%.
  • the 2-sided 95% confidence interval of the combined vaccine efficacy against virologically-confirmed dengue with hospitalization against all four serotypes when comparing seropositive and seronegative subjects provides for lower bounds of the 2-sided confidence interval which are within 10% points or within 15% points or within 20% points.
  • safety means providing a combined vaccine efficacy against virologically-confirmed dengue with hospitalization against all four serotypes with a 2-sided 95% confidence interval, wherein the lower bound is more than 65%, when measured against placebo in a subject population of at least 5,000 healthy 4 to 16 year old subjects irrespective of serostatus at baseline from first administration of the administration schedule until 12 to 18 months after the last administration of the administration schedule.
  • the vaccine is considered safe within the meaning of this invention.
  • safe in particular refers to a vaccine that is safe for all subjects irrespective of their serostatus at baseline. This means that the vaccine can be administered without the need to determine the occurrence of a previous dengue infection in the subject before administration.
  • the vaccine is safe as defined above with respect to all age groups starting from 4 years of age and preferably irrespective of the serostatus, in particular from 4 years of age to 60 years of age, or 4 years of age to 16 years of age.
  • VE against virologically-confirmed dengue disease with hospitalization is made to the disclosure below with respect to certain methods of treatment.
  • vaccine efficacy or “VE” measure the proportionate reduction in cases among vaccinated persons.
  • Vaccine efficacy is measured by calculating the risk of disease among vaccinated and unvaccinated persons and determining the percentage reduction in risk of disease among vaccinated persons relative to unvaccinated persons. The greater the percentage reduction of illness in the vaccinated group, the greater the vaccine efficacy. For example, a VE of 90% indicates a 90% reduction in disease occurrence among the vaccinated group, or a 90% reduction from the number of cases you would expect if they have not been vaccinated.
  • ⁇ v denote the hazard rate for the subjects vaccinated with a tetravalent dengue vaccine composition as disclosed herein and ⁇ c denote the hazard rate for unvaccinated subjects, i.e. subjects receiving placebo.
  • the hazard rate ratio HR is estimated from a Cox proportional hazard model with study vaccine as a factor, adjusted for age, and stratified by region.
  • the term“combined vaccine efficacy against all four serotypes” is defined as the vaccine efficacy in relation to the risk of dengue disease irrespective of the serotype being responsible for the virologically-confirmed dengue disease and the subject baseline serostatus.
  • a vaccine is considered “effective” in case the combined vaccine efficacy is above 30%.
  • the combined vaccine efficacy may be also 40% or more, 50% or more, 60% or more, 70% or more, 72% or more, or 80% or more, in particular when determined from 30 days after a second administration until 12 months after a second administration or 18 months after a second vaccination, in particular when determined in age groups selected from the age group of 4 to 16 year old subjects, the age group of 4 to under 9 year old subjects, the age group of 2 to under 9 year old subjects, the age group of 4 to 5 year old subjects, the age group of 6 to 11 year old subjects, and the age group of 12 to 16 year old subjects.
  • effective in particular refers to a vaccine that is effective for all subjects irrespective of their serostatus at baseline.
  • the vaccine is effective with respect to all age groups starting from 4 years of age and preferably irrespective of the serostatus, in particular from 4 years of age to 60 years of age or from 4 years of age to 16 years of age and irrespective of the serostatus.
  • Relevant subgroups in this context are under 9 years of age, from 2 years of age to under 9 years of age, from 4 years of age to under 9 years of age, 4 to 5 years of age, 6 to 11 years of age and 12 to 16 years of age or any age group within 4 to 16 years of age.
  • “effective” means providing a combined vaccine efficacy against all four serotypes, in preventing virologically confirmable dengue disease with a 2-sided 95% confidence interval, wherein the lower bound is more than 60%, when measured against placebo in a subject population of at least 5,000 healthy subjects irrespective of serostatus at baseline and 4 to 16 years of age, from the first administration of the administration schedule until 18 months after the last administration of the administration schedule. Further specific efficacies can be defined.
  • “combined vaccine efficacy against all four serotypes in seronegative subjects” refers to the efficacy measured in subjects which are seronegative at baseline.
  • “vaccine efficacy against a specific serotype e.g.
  • serotype 1 refers to the efficacy in relation to a specific serotype being responsible for the virologically-confirmed dengue disease.
  • combined vaccine efficacy against all four serotypes against virologically-confirmed dengue with hospitalization refers to the efficacy wherein only virologically-confirmed dengue cases with hospitalization are considered.
  • Such vaccine efficacies can be determined with respect to subjects being seronegative or seropositive at baseline and for different age groups.
  • the “relative risk” means the number of events of virologically confirmed dengue disease divided by the number of subjects treated with the unit dose as disclosed herein over the number of events of virologically confirmed dengue disease divided by the number of subjects treated with placebo.
  • the term“combined relative risk against all four serotypes” is defined as the relative risk in relation to the risk of dengue disease irrespective of the serotype being responsible for the virologically-confirmed dengue disease and the subject baseline serostatus.
  • vaccinating or “inoculating” refers to the administration of a vaccine to a subject, with the aim to prevent the subject, from developing one or more symptoms of a disease.
  • “vaccinating against dengue disease” or “inoculating against dengue disease” refers to the administration of a dengue vaccine composition to a subject, with the aim to prevent the subject, from developing one or more symptoms of dengue disease.
  • the method comprises a primary vaccination and optionally one or more booster vaccinations.
  • the primary vaccination is defined as the primary administration schedule for administering the composition or unit dose as disclosed herein to establish a protective immune response and e.g. consists of two administrations e.g. within three months.
  • booster vaccination refers to an administration or administration schedule which takes place after the primary vaccination e.g. at least 1 year or even 5 or 10 years after the last administration, e.g. the second administration, of the primary vaccination schedule.
  • the booster administration attempts at enhancing or reestablishing the immune response of the primary vaccination.
  • the terms “subject” or “subjects” are limited to human subjects (e.g. infants, children or adults).
  • the terms “elderly subject” or “elderly subjects” refer to subjects with an age of more than 60 years, such as 61 years to 100 years, 61 years to 90 years, 61 years to 80 years, 61 years to 75 years, or 61 years to 70 years.
  • subject population refers to a group of subjects.
  • the subject population may refer to least 40 subjects, at least 50 subjects, at least 60 subjects, at least 100 subjects or at least 1000 subjects and is defined by certain parameters.
  • the parameters that may be used to define a subject population include, but are not limited to, the age of the subjects, whether the subjects are from a dengue endemic region or from a dengue non-endemic region and the serostatus of the subjects.
  • endemic region refers to a region where a disease or infectious agent is constantly present and/or usually prevalent in a population within this region.
  • non-endemic region refers to a region from which the disease is absent or in which it is usually not prevalent.
  • a “dengue endemic region” refers to geographic areas in which an infection with dengue virus is constantly maintained at a baseline level.
  • a “dengue non-endemic region” is a geographic area in which an infection with dengue virus is not constantly maintained at a baseline level.
  • subject populations or subjects “from a dengue endemic region” or “from a dengue non-endemic region” refer to subject populations or subjects living in geographic areas as defined above.
  • Whether a geographic area or a subject population is dengue-endemic or not can be determined by different calculatory methods such as the ones described in Bhatt et al. (2013) Nature 496 (7446): 504-507 and supplementary material and in Stanaway et al. (2016) Lancet Infect Dis. 16(6): 712-723 and supplementary material.
  • Overviews of dengue endemic regions and dengue epidemiology are regularly published, for example, by the WHO or CDC. Typical dengue-endemic regions are in Latin America, Southeast Asia and the Pacific islands and dengue endemic countries include, but are not limited to, Australia, Brazil, Bangladesh, Colombia, China, Dominican Republic, Indonesia, India, Mexico, Malaysia, Portugal, Pakistan, Panama, Philippines, Puerto Rico, Singapore, Sri Lanka, Thailand and Vietnam.
  • the area's force of infection is measured by seroprevalence surveys provided as seroprevalence rate. Areas with very high force of infection are considered to have a seroprevalence rate of more than 80%.
  • region when it concerns seroprevalence rates refers to a geographic area where the seroprevalence rate could be determined or is known, e.g. a village, a town, a city, a region, a county, a state, a province or parts of the foregoing or a whole country.
  • serostatus refers to the amount of antibodies a subject has with respect to a certain infectious agent, in particular dengue virus.
  • seronegative or serona ⁇ ve means that the subject does not have neutralizing antibodies against any one of dengue serotypes DENV-1, DENV-2, DENV-3 and DENV-4 in the serum.
  • a seronegative or serona ⁇ ve subject or subject population is defined by a neutralizing antibody titer of less than 10 for each one of the four dengue serotypes.
  • a subject or subject population having a neutralizing antibody titer of equal to or more than 10 for at least one dengue serotype is defined as being “seropositive” with respect to said dengue serotype.
  • Serostatus at baseline refers to the serostatus before the administration of a dengue vaccine composition as described herein.
  • a “neutralizing antibody titer” refers to the amount of antibodies in the serum of a subject that neutralize the respective dengue serotype.
  • the neutralizing antibody titer against DENV-1, DENV-2, DENV-3 and DENV-4 is determined in a serum sample of the subject using known methods such as the plaque reduction neutralization test (PRNT) as described in the WHO Guidelines (World Health Organization Department of Immunization Vaccines Biologicals (2007) Guidelines for plaque reduction neutralization testing of human antibodies to dengue viruses, WHO/IVB/07.07) or a microneutralization (MNT50) assay as described herein.
  • PRNT plaque reduction neutralization test
  • MNT50 microneutralization
  • the “ratio of not more than 20 for the neutralizing antibody titer of dengue serotype 2 to the neutralizing antibody titer of dengue serotype 4” means that the neutralizing antibody titer of dengue serotype 2 is divided by the neutralizing antibody titer of dengue serotype 4 and that the ratio obtained hereby is no more than 20. In other words, the neutralizing antibody titer of dengue serotype 2 is not more than 20-times higher than the neutralizing antibody titer of dengue serotype 4 in the subject.
  • the terms “geometric mean neutralizing antibody titer” and “GMT” refer to the geometric mean value of the titer of neutralizing antibodies against the corresponding dengue serotype in the serum of subjects in a subject population.
  • the geometric mean value is calculated by a well-known formula.
  • the “ratio of not more than 20 for the GMT of dengue serotype 2 to the GMT of dengue serotype 4” means that the geometric mean neutralizing antibody titer of dengue serotype 2 (GMT DENV-2) is divided by the geometric mean neutralizing antibody titer of dengue serotype 4 (GMT DENV-4) and that the ratio obtained hereby is no more than 20.
  • the geometric mean neutralizing antibody titer of dengue serotype 2 is not more than 20-times higher than the geometric mean neutralizing antibody titer of dengue serotype 4 in the subject population.
  • an “immune response” refers to a subjects response to the administration of the dengue vaccine.
  • the immune response includes the formation of neutralizing antibodies to one or more dengue serotypes. It may also include the stimulation of a cell-mediated response or the formation of antibodies to non-structural proteins such as NS1.
  • An immune response is stimulated by the administration of a unit dose of the invention as described herein, if the titer of neutralizing antibodies against at least one dengue virus serotype and preferably against all four dengue virus serotypes is increased after said administration of said unit dose.
  • An immune response is stimulated by the administration of a unit dose of the invention as described herein, if the secretion of interferon gamma by peripheral blood mononuclear cells stimulated with peptides from dengue virus proteins is increased after said administration of said unit dose.
  • An immune response is stimulated by the administration of a unit dose of the invention as described herein, if the titer of antibodies to non-structural proteins such as NS1 is increased after said administration of said unit dose.
  • the administration of a reconstituted unit dose of the present invention as described herein stimulates the formation of neutralizing antibodies to one or more dengue serotypes, a cell-mediated response and the formation of antibodies to non-structural proteins such as NS1.
  • a “balanced immune response” means that the immune response to the four dengue serotypes is sufficient to provide protection against infection by all four dengue serotypes and preferably the immune response to the four dengue serotypes has a similar strength.
  • the neutralizing antibody titer against the four dengue serotypes at day 180 or day 365 after administration of a first reconstituted unit dose of the invention as described herein is similar, i.e. it differs by less than factor 30, by less than factor 25 or by less than factor 20.
  • total concentration in pfu/0.5 ml which serves as a base value for the calculation of the percentage concentration for each individual component of a tetravalent dengue vaccine is shown for one exemplary tetravalent vaccine composition comprising dengue serotype 1 in a concentration of 3.60 log 10 pfu/0.5 ml, a dengue serotype 2 concentration of 4.00 log 10 pfu/0.5 ml, a dengue serotype 3 concentration of 4.60 log 10 pfu/0.5 ml and a dengue serotype 4 concentration of 5.11 log 10 pfu/0.5 ml.
  • the logarithmic values of the concentrations are converted into numerical values.
  • the results of this conversion are 4 ⁇ 10 3 pfu/0.5 ml for serotype 1, 1 ⁇ 10 4 pfu/0.5 mll for serotype 2, 4 ⁇ 10 4 pfu/0.5 ml for serotype 3 and 1.3 ⁇ 10 5 pfu/0.5 ml for serotype 4.
  • the total concentration in pfu/0.5 ml is the sum of the preceding numerical values resulting in 1.84 ⁇ 10 5 pfu/0.5 ml.
  • the “percentage concentration” for each of the serotypes 1, 2, 3 and 4 is obtained by dividing the numerical concentration value (expressed as pfu/0.5 ml) of an individual serotype by the total concentration (expressed in pfu/0.5 ml) and multiplying the result by 100 i.e.:
  • the “concomitant” administration of vaccines refers to a combined administration of two or more vaccines.
  • the combined administration of two or more vaccines refers to combining the administration schedule of a dengue vaccine, such as the unit dose of the invention, with the administration schedule of a fellow fever vaccine, such as YF-17D, and/or of a hepatitis A vaccine, such as HAVRIX® or VAQTA®, and/or of a HPV vaccine, such as a 9vHPV vaccine, and/or of a combined measles, mumps and rubella (MMR) vaccine, and/or of a combined Tdap vaccine, such as a combined tetanus toxoid, reduced diphtheria toxoid and acellular pertussis (adsorbed) vaccine, such as BOOSTRIX®, and/or of a DTaP/IPV/HIB vaccine, in particular a combined DTaP/IP
  • “simultaneous” administration means an administration of at least two different vaccines, such as a dengue vaccine and a fellow fever vaccine, or a dengue vaccine and a hepatitis A vaccine, or a dengue vaccine and a HPV vaccine, or a dengue vaccine and a MMR vaccine, or a dengue vaccine and a Tdap vaccine, or a dengue vaccine and a DTaP/IPV/Hib vaccine on the same day.
  • the simultaneous administration may be administered by the same medical practitioner, such as during the same medical appointment.
  • sequential administration means an administration of at least two different vaccines, such as a dengue vaccine and a fellow fever vaccine, or a dengue vaccine and a hepatitis A vaccine, or a dengue vaccine and a HPV vaccine, or a dengue vaccine and a MMR vaccine, or a dengue vaccine and a Tdap vaccine, or a dengue vaccine and a DTaP/IPV/Hib vaccine on subsequent days, such as within 90 days, but in a combined administration schedule including the administration of the dengue vaccine and the fellow fever vaccine.
  • a dengue vaccine and a fellow fever vaccine such as a dengue vaccine and a fellow fever vaccine, or a dengue vaccine and a hepatitis A vaccine, or a dengue vaccine and a HPV vaccine, or a dengue vaccine and a MMR vaccine, or a dengue vaccine and a Tdap vaccine, or a dengue vaccine and a DTaP/IPV/Hib vaccine on subsequent days, such as within 90 days, but in
  • HPV-associated cancers or genital warts refers to cancers and genital wards caused by an HPV infection, respectively.
  • HPV serotypes 16, 18, 45, 31, 33, 52, 58 also referred to as “high-risk” HPV serotypes, are the types most common in cervical cancers, wherein the two HPV serotypes 16 and 18 cause about 70% of cervical cancers worldwide.
  • HPV serotypes 6 and 11, also referred to as “low-risk” HPV serotypes cause genital warts.
  • a “9vHPV vaccine” refers to a 9-valent HPV vaccine that provides protection against the HPV serotypes 6, 11, 16, 18, 31, 33, 45, 52, and 58.
  • the 9vHPV vaccine is used to prevent cervical, vulvar, vaginal, and anal cancers, precancerous or dysplastic lesions, and genital warts caused by one or more of these HPV serotypes.
  • MMR vaccine refers to a combined vaccine for measles, mumps and rubella.
  • MMR vaccine are known in the prior art and include M-M-R® II, Priorix®, Tresivac®, and Trimovax®.
  • Tdap vaccine refers to a combined vaccine for tetanus, diphtheria and pertussis.
  • Tdap vaccines known in the prior art include INFANRIX® (for vaccination of children from 6 weeks to 7 years of age), and BOOSTRIX® from GlaxoSmithKline and Adacel from Sanofi Pasteur (both for use in individuals of 10 years of age or older).
  • DTaP refers to diphtheria, tetanus and acellular pertussis.
  • IPV inactivated poliovirus
  • Hib refers to Haemophilus influenzae type b.
  • DTaP/IPV/Hib vaccine refers to a combined vaccine for diphtheria, tetanus, pertussis, poliomyelitis and Haemophilus influenzae type b.
  • a DTaP/IPV/Hib vaccine known in the prior art includes Pentacel® from Sanofi Pasteur.
  • chronic disease or condition includes those diseases and conditions which persist in an elderly subject for three months or more. In particular, it includes diabetes, hypertension, allergies, previous strokes, ischemic heart disease, chronic renal impairment and chronic obstructive pulmonary disease.
  • the term “impaired immune system” means that at least one function of at least one component of the immune system is weaker than in younger subjects, i.e. in subjects with an age of less than 60 years. These functions include a lower antioxidant response of monocytes against oxidative stress induced by dengue virus and lower T cell responses and cytokine production in response to dengue virus infection.
  • solicited systemic adverse events in children under 6 years are defined as fever, irritability/fussiness, drowsiness and loss of appetite that occurred within 14 days after each vaccination, and in children of 6 years or more are defined as fever, headache, asthenia, malaise and myalgia that occurred within 14 days after each vaccination.
  • solicited local adverse events are injection site pain, injection site erythema and injection site swelling that occurred within 7 days after each vaccination.
  • unsolicited adverse events are any adverse events (AEs) that are not solicited local or systemic AEs, as defined above.
  • a “serious adverse event” or “SAE” is any untoward medical occurrence or effect that at any dose results in death, is life-threatening, requires inpatient hospitalization or prolongation of existing hospitalization, results in persistent or significant disability/incapacity, is a congenital anomaly/birth defect or is medically important due to other reasons than the above mentioned criteria.
  • IP-Related AE or “vaccine related AE” means that there is suspicion that there is a relationship between the vaccine and the AE (without determining the extent of probability); there is a reasonable possibility that the vaccine contributed to the AE.
  • Non-IP Related or “non-vaccine related” means that there is no suspicion that there is a relationship between the vaccine and the AE; there are other more likely causes and administration of the vaccine is not suspected to have contributed to the AE.
  • a subject or subject population being “2 to 17 years of age” refers to a subject or subject population being 2 to 17 years of age on the first day of the administration of the dengue vaccine composition as described herein.
  • %-points refers to the difference of two %-values in a %-value. For example two values in % which are within 5%-points refers to e.g. one value at 1% and a second value at 6%.
  • the term “determination of the previous dengue infection in the subject before administration” means that a previous dengue infection has to be assessed before vaccination in that there is a laboratory confirmed history of dengue or through an appropriately validated serological test e.g. by the method as disclosed herein such as the MNT50 test described in Example 2 or any serotesting with adequate performance in terms of specificity and cross reactivity based on the locale disease epidemiology.
  • % w/v refers to % mg/ml wherein e.g. 150 mg/ml are 15% w/v.
  • FIG. 1 Genetic structure of the four dengue strains contained in TDV.
  • the solid red triangles indicate the three attenuating mutations present in the 5′NCR, NS1 and NS3 proteins.
  • the TDV-1, TDV-3 and TDV-4 strains are chimeric viruses where the prM and E genes from dengue serotype 1, 3 and 4, respectively, are inserted into the TDV-2 backbone.
  • FIG. 2 Schematic drawing illustrating the microneutralization test (MNT) used to determine the titer of neutralizing antibodies.
  • FIG. 3 Percentage of subjects ( ⁇ 95% confidence intervals) who were seropositive (reciprocal neutralizing titer 10) for each of the dengue serotypes at different time points of the trial in the HD-TDV group (dark colored, left bar) and TDV group (light colored, right bar) throughout the trial. Time points shown are baseline, day 15 (d15), day 30 (d30), day 90 (d90), day 180 (d180) and day 365 (d365).
  • Part A shows the results for participants seropositive (set of graphs on the left) and seronegative (set of graphs on the right) at baseline, per-protocol set.
  • Part B shows the results for the entire trial population (all) per-protocol set
  • FIG. 4 Geometric mean titers (GMTs) ( ⁇ 95% confidence intervals) of neutralizing antibodies against each of the four dengue serotypes during the course of the trial for HD-TDV (dark line with triangles) and TDV (light line with circles) recipients, for the entire trial population (part B) and for participants seropositive and seronegative at baseline (part A), per-protocol set.
  • GTTs Geometric mean titers
  • FIG. 5 IFN ⁇ ELISpot analysis of peripheral blood mononuclear cells before vaccination (baseline) and at different time points after administration of TDV. Shown are the response frequencies to the entire DENV-2 proteome. A subject was considered responsive if response to more than one peptide pool from DENV-2 was positive (i.e. ⁇ 4 ⁇ mean of negative control and ⁇ 50 SFC/10 6 PBMCs).
  • FIG. 6 IFN ⁇ ELISpot analysis of peripheral blood mononuclear cells before vaccination (baseline) and at different time points after administration of TDV. Shown are the response frequencies to peptide pools matching selected DENV-derived proteins as indicated. A subject was considered responsive if response to more than one peptide pool from DENV-2 was positive (i.e. ⁇ 4 ⁇ mean of negative control and ⁇ 50 SFC/10 6 PBMCs).
  • FIG. 7 Effect of sera from a seronegative subject (A) and a seropositive subject (A) to whom TDV was administered on DENV-2 NS1-induced hyperpermeability as determined by TEER.
  • HPMEC were grown on Transwell semi-permeable membranes (0.4 ⁇ m pore size), and serum samples (30 ⁇ l) were added to the apical chamber in the presence or absence of DENV2 NS1 (5 ⁇ g/ml).
  • DENV2 NS1 is depicted as squares; day 0 serum alone is depicted as diamonds; day 120 serum alone is depicted as triangles; day 0 serum+DENV2 NS1, is depicted as upside-down triangles; day 120 serum+DENV2 NS1 is depicted as X's.
  • ( ⁇ circumflex over ( ) ⁇ ) represents media change. Endothelial permeability was measured at indicated time-points over 48 hours. Relative TEER values from one independent experiment performed in duplicate are plotted. Error bars indicate standard error of the mean (SEM).
  • FIG. 8 Effect of sera from seronegative and seropositive subjects to which TDV was administered on NS1-induced sialic acid and heparan sulfate degradation. Shown is the quantification of mean fluorescence intensity (MFI) of (A) sialic acid and (B) heparan sulfate expression after staining with sialic acid- and heparan sulfate-specific fluorescent antibodies as visualized by confocal microscopy. Values are normalized to MFI from the NS1+positive control serum group (represented by dotted line at 100%) and expressed as percentage of control. Error bars indicate SEM. The left bar for each subject shows the results at day 0 (d0), the right car for each subject shows the results at day 120 (d120).
  • MFI mean fluorescence intensity
  • FIG. 9 Flow diagram of the clinical trial of Example 6.
  • FIG. 10 Cumulative incidence of A) virologically-confirmed dengue cases and B) hospitalized virologically-confirmed dengue cases over time during Part 1 study period by baseline serostatus (safety set data; data presented truncated at Month 18). Tables show numbers of participants under follow-up at various time points to end of Part 1 study period.
  • FIG. 11 Study design of phase III study described in example 6
  • the dengue virus is a single stranded, positive sense RNA virus of the family flaviviridae.
  • the taxonomy is outlined in Table 1.
  • the family flaviviridae includes three genera, flavivirus, hepacivirus and pestivirus.
  • the genus flavivirus contains highly pathogenic and potentially hemorrhagic fever viruses, such as yellow fever virus and dengue virus, encephalitic viruses, such as Japanese encephalitis virus, Murray Valley encephalitis virus and West Nile virus, and a number of less pathogenic viruses.
  • the flavivirus genome comprises in 5′ to 3′ direction (see FIG. 1 ):
  • C capsid protein
  • prM pre-membrane protein
  • NSI nonstructural proteins
  • the viral structural proteins are C, prM and E, and the nonstructural proteins are NSI to NS5.
  • the structural and nonstructural proteins are translated as a single polyprotein and processed by cellular and viral proteases.
  • the unit dose of the invention as described herein comprises a dengue virus composition that comprises four live attenuated dengue virus strains (tetravalent dengue virus composition) representing dengue serotype 1, dengue serotype 2, dengue serotype 3 and dengue serotype 4.
  • the composition comprises chimeric dengue viruses and optionally at least one non-chimeric dengue virus, in particular a molecularly characterized and cloned dengue serotype 2 strain derived from the live attenuated DEN-2 PDK-53 virus strain (TDV-2), and three chimeric dengue strains derived from the TDV-2 strain by replacing the structural proteins prM and E from TDV-2 with the corresponding structural proteins from the other dengue serotypes, resulting in the following chimeric dengue strains:
  • TDV-2 live attenuated DEN-2 PDK-53 virus strain
  • TDV-4 a DENV-2/4 chimera
  • the genetically modified tetravalent dengue vaccine TDV is based on a molecularly characterized and cloned dengue-2 virus strain (TDV-2).
  • TDV-2 strain was generated by cDNA cloning of the attenuated laboratory-derived DEN-2 PDK-53 virus strain that was originally isolated at Mahidol University, Bangkok, Thailand (Kinney et al. (1997) Virology 230(2): 300-308).
  • DEN-2 PDK-53 was generated by 53 serial passages in primary dog kidney (PDK) cells at 32° C. (Bhamarapravati et al. (1987) Bull. World Health Organ. 65(2): 189-195).
  • the attenuated DEN-2 PDK-53 strain (the precursor of TDV-2) was derived from the wild type virus strain DEN-2 16681 (SEQ ID NO 11) and differs in nine nucleotides from the wild type as follows (Kinney et al. (1997) Virology 230(2): 300-308):
  • the three nucleotide changes located in the 5′ noncoding region (NCR) (nucleotide 57) (mutation (i)), the NS-1 (amino acid 828 of SEQ ID NO. 4) (mutation (iv)) and NS-3 genes (amino acid 1725 of SEQ ID NO. 4) (mutation (vi)) form the basis for the attenuation phenotype of the DEN-2 PDK-53 strain (Butrapet et al. (2000) J. Virol. 74(7): 3111-3119) (Table 2). These three mutations are referred to herein as the “attenuating mutations” and are comprised in TDV-1, TDV-2, TDV-3 and TDV-4.
  • TDV-2 comprises in addition to the three attenuating mutations one or more mutations selected from:
  • the silent mutation in the NS5 gene at nucleotide 8571 from cytosine to thymidine of DEN-2 PDK-53 is not present in the TDV-2 strain.
  • TDV-2 comprises in addition to the three attenuating mutations one or more mutations selected from:
  • TDV-2 comprises in addition to the three attenuating mutations the mutations a) and g), preferably the mutations a), g), c), e) and h), more preferably the mutations a), g), c), e), h) and b), even more preferably the mutations a), g), c), e), h), b) and d), and most preferably the mutations a) to h).
  • the nucleotide positions and amino acids positions of TDV-2 refer to the nucleotide sequence as shown in SEQ ID NO. 3 and amino acid sequence as shown in SEQ ID NO. 4.
  • TDV-2 was modified by replacing the nucleic acid sequence encoding the DENV-2 prM and E glycoproteins with the nucleic acid sequence encoding the corresponding wild type prM and E glycoproteins from the DENV-1, DENV-3, and DENV-4 wild type strains DENV-1 16007, DENV-3 16562 or DENV-4 1036 virus, respectively, (see Table 3) using standard molecular genetic engineering methods (Huang et al. (2003) J. Virol. 77(21): 11436-11447).
  • FIG. 1 A diagram of the four TDV strains comprised in the dengue vaccine composition is shown in FIG. 1 .
  • the chimeric dengue strains TDV-1, TDV-3 and TDV-4 express the surface antigens prM and E of the DENV-1, DENV-3 or DENV-4 viruses, as depicted in Table 3 respectively, and retain the genetic alterations responsible for the attenuation of TDV-2.
  • each of the TDV-1, TDV-3 and TDV-4 strains comprises the attenuating mutations described in Table 2.
  • TDV-1 comprises in addition to the three attenuating mutations one or more mutations selected from:
  • k a mutation in the junction site between the prM-E gene and the DEN-2 PDK-53 backbone at nucleotides 2381/2382 from thymidine-guanine to cytosine-cytosine resulting in an amino acid change at position 762 from valine to alanine.
  • TDV-1 comprises in addition to the three attenuating mutations one or more mutations selected from:
  • n a silent mutation in the NS4B gene at nucleotide 7311 from adenine to guanine.
  • the TDV-1 strain comprises in addition to the three attenuating mutations the mutations l) and m), preferably the mutations l), m), c) and e), even more preferably the mutations l), m), c), e), d) and n), and most preferably the mutations l), m), c), e), d), n), i), j) and k).
  • the nucleotide positions and amino acids positions of TDV-1 refer to the nucleotide sequence as shown in SEQ ID NO. 1 and amino acid sequence as shown in SEQ ID NO. 2.
  • TDV-3 comprises in addition to the three attenuating mutations one or more mutations selected from:
  • k a mutation in the junction site between the prM-E gene and the DEN-2 PDK-53 backbone at nucleotides 2375/2376 from thymidine-guanine to cytosine-cytosine resulting in an amino acid change at position 760 from valine to alanine, and/or
  • TDV-3 comprises in addition to the three attenuating mutations one or more mutations selected from:
  • r a silent mutation in the NS5 gene at nucleotide 7620 from adenine to guanine.
  • TDV-3 comprises in addition to the three attenuating mutations the mutations p) and q), preferably the mutations p), q), c) and e), even more preferably the mutations p), q), c), e), d) and r), and most preferably the mutations p), q), c), e), d), r), j), k) and o).
  • the nucleotide positions and amino acids positions of TDV-3 refer to the nucleotide sequence as shown in SEQ ID NO. 5 and amino acid sequence as shown in SEQ ID NO. 6.
  • TDV-4 comprises in addition to the three attenuating mutations one or more mutations selected from:
  • k a mutation in the junction site between the prM-E gene and the DEN-2 PDK-53 backbone at nucleotides 2381/2382 from thymidine-guanine to cytosine-cytosine resulting in an amino acid change at position 762 from valine to alanine, and/or
  • v a mutation in the E gene at nucleotide 2275 from adenine to cytosine resulting in an amino acid change at position 727 from methionine to leucine.
  • TDV-4 comprises in addition to the three attenuating mutations one or more mutations selected from:
  • aa a mutation in the NS4A gene at nucleotide 6437 from cytosine to thymidine resulting in an amino acid change at position 2114 from alanine to valine, and/or
  • cc a silent mutation in the NS5 gene at nucleotide 9750 from adenine to cytosine.
  • TDV-4 comprises in addition to the three attenuating mutations the mutation s), u) and v), preferably the mutations s), u), v), c), e), x), y) and aa), even more preferably the mutations s), u), v), c), e), x), y), aa) and w), even more preferably the mutations s), u), v), c), e), x), y), aa), w), d), z), bb) and cc), and most preferably the mutations s), u), v), c), e), x), y), aa), w), d), z), bb), cc), j), k) and t).
  • the nucleotide positions and amino acids positions of TDV-4 refer to the nucleotide sequence as shown in SEQ ID NO. 7 and amino acid sequence as shown in SEQ ID NO. 8.
  • TDV-1 has the nucleotide sequence of SEQ ID NO. 1
  • TDV-2 has the nucleotide sequence of SEQ ID NO. 3
  • TDV-3 has the nucleotide sequence of SEQ ID NO. 5
  • TDV-4 has the nucleotide sequence of SEQ ID NO. 7.
  • TDV-1 has the amino acid sequence of SEQ ID NO. 2
  • TDV-2 has the amino acid sequence of SEQ ID NO. 4
  • TDV-3 has the amino acid sequence of SEQ ID NO. 6
  • TDV-4 has the amino acid sequence of SEQ ID NO. 8.
  • TDV-1 has a nucleotide sequence encoding the amino acid sequence of SEQ ID NO.
  • TDV-2 has a nucleotide sequence encoding the amino acid sequence of SEQ ID NO. 4
  • TDV-3 has a nucleotide sequence encoding the amino acid sequence of SEQ ID NO. 6
  • TDV-4 has a nucleotide sequence encoding the amino acid sequence of SEQ ID NO. 8.
  • TDV virus strains SEQ ID NO. dengue virus strain sequence type SEQ ID NO. 1 TDV-1 nucleotide sequence SEQ ID NO. 2 TDV-1 amino acid sequence SEQ ID NO. 3 TDV-2 nucleotide sequence SEQ ID NO. 4 TDV-2 amino acid sequence SEQ ID NO. 5 TDV-3 nucleotide sequence SEQ ID NO. 6 TDV-3 amino acid sequence SEQ ID NO. 7 TDV-4 nucleotide sequence SEQ ID NO. 8 TDV-4 amino acid sequence
  • the unit dose of the invention as described herein comprises the live attenuated dengue virus strains TDV-1, TDV-2, TDV-3 and TDV-4, wherein TDV-1, TDV-3 and TDV-4 are based on TDV-2 and comprise the prM and E regions of DENV-1, -3 and -4, respectively.
  • TDV-1 is characterized by the nucleotide sequence according to SEQ ID No. 1 and the amino acid sequence according to SEQ ID No. 2
  • TDV-2 is characterized by the nucleotide sequence according to SEQ ID No. 3 and the amino acid sequence according to SEQ ID No. 4
  • TDV-3 is characterized by the nucleotide sequence according to SEQ ID No. 5 and the amino acid sequence according to SEQ ID No. 6
  • TDV-4 is characterized by the nucleotide sequence according to SEQ ID No. 7 and the amino acid sequence according to SEQ ID No. 8.
  • the E protein of DENV-3 has two fewer amino acids than the E protein of DENV-2. Therefore, the nucleotides and encoded amino acid backbone of TDV-2 starting after the E region of DENV-3 at nucleotide 2374 of SEQ ID NO. 5 and amino acid 760 of SEQ ID NO. 6 are 6 nucleotides less and 2 amino acids less than the original TDV-2 nucleotide and amino acid positions, respectively.
  • the present invention is in part directed to a unit dose of a dengue vaccine composition as described.
  • the dengue vaccine composition comprises a tetravalent dengue virus composition, also referred to as dengue virus composition, and pharmaceutically acceptable excipients.
  • the present invention is in part directed to a unit dose of a dengue vaccine composition, wherein the dengue vaccine composition comprises a tetravalent dengue virus composition including four live attenuated dengue virus strains:
  • the dengue vaccine composition comprises a tetravalent dengue virus composition including four live attenuated dengue virus strains:
  • the present invention is further in part directed to a unit dose of a dengue vaccine composition, wherein the dengue vaccine composition comprises a tetravalent dengue virus composition including four live attenuated dengue virus strains:
  • the dengue vaccine composition comprises a tetravalent dengue virus composition including four live attenuated dengue virus strains:
  • the chimeric dengue serotype 2/1 strain is TDV-1
  • the dengue serotype 2 strain is TDV-2
  • the chimeric dengue serotype 2/3 strain is TDV-3
  • the chimeric dengue serotype 2/4 strain is TDV-4.
  • the dengue vaccine composition comprises a tetravalent dengue virus composition including four live attenuated dengue virus strains wherein:
  • the dengue vaccine composition comprises a tetravalent dengue virus composition including four live attenuated dengue virus strains wherein:
  • the dengue vaccine composition comprises a tetravalent dengue virus composition including four live attenuated dengue virus strains wherein:
  • the dengue vaccine composition comprises a tetravalent dengue virus composition including four live attenuated dengue virus strains wherein:
  • the dengue vaccine composition comprises a tetravalent dengue virus composition including four live attenuated dengue virus strains wherein:
  • the dengue vaccine composition comprises a tetravalent dengue virus composition including four live attenuated dengue virus strains wherein:
  • the dengue vaccine composition comprises a tetravalent dengue virus composition including four live attenuated dengue virus strains wherein:
  • the dengue vaccine composition comprises a tetravalent dengue virus composition including four live attenuated dengue virus strains wherein:
  • the dengue vaccine composition comprises a tetravalent dengue virus composition including four live attenuated dengue virus strains wherein the arithmetic sum of all four serotypes is less than 6.7 log 10 pfu/0.5 mL, preferably less than 5.5 log 10 pfu/0.5 mL. In certain such embodiments, the arithmetic sum of all four serotypes is at least 4.6 log 10 pfu/0.5 mL.
  • the dengue vaccine composition comprises a tetravalent dengue virus composition including four live attenuated dengue virus strains wherein the arithmetic sum of all four serotypes is in the range of 4.6 log 10 pfu/0.5 mL to 6.7 log 10 pfu/0.5 mL, preferably in the range of 4.6 log 10 pfu/0.5 mL to 5.5 log 10 pfu/0.5 mL.
  • the chimeric dengue serotype 2/1 strain is TDV-1
  • the dengue serotype 2 strain is TDV-2
  • the chimeric dengue serotype 2/3 strain is TDV-3
  • the chimeric dengue serotype 2/4 strain is TDV-4.
  • TDV-1 is characterized by the nucleotide sequence according to SEQ ID No. 1 and the amino acid sequence according to SEQ ID No. 2
  • TDV-2 is characterized by the nucleotide sequence according to SEQ ID No. 3 and the amino acid sequence according to SEQ ID No. 4
  • TDV-3 is characterized by the nucleotide sequence according to SEQ ID No. 5
  • TDV-4 is characterized by the nucleotide sequence according to SEQ ID No. 7 and the amino acid sequence according to SEQ ID No. 8.
  • the present invention is in part directed to a unit dose of a dengue vaccine composition, wherein the dengue vaccine composition comprises a tetravalent dengue virus composition including four live attenuated dengue virus strains:
  • the dengue vaccine composition comprises a tetravalent dengue virus composition including four live attenuated dengue virus strains wherein:
  • the dengue vaccine composition comprises a tetravalent dengue virus composition including four live attenuated dengue virus strains wherein:
  • the dengue vaccine composition comprises a tetravalent dengue virus composition including four live attenuated dengue virus strains wherein:
  • the dengue vaccine composition comprises a tetravalent dengue virus composition including four live attenuated dengue virus strains wherein:
  • the dengue vaccine composition comprises a tetravalent dengue virus composition including four live attenuated dengue virus strains wherein:
  • the dengue vaccine composition comprises a tetravalent dengue virus composition including four live attenuated dengue virus strains wherein:
  • the dengue vaccine composition comprises a tetravalent dengue virus composition including four live attenuated dengue virus strains wherein:
  • the dengue vaccine composition comprises a tetravalent dengue virus composition including four live attenuated dengue virus strains wherein:
  • the dengue vaccine composition comprises a tetravalent dengue virus composition including four live attenuated dengue virus strains wherein the arithmetic sum of all four serotypes is less than 6.7 log 10 pfu/dose, preferably less than 5.5 log 10 pfu/dose. In certain such embodiments, the arithmetic sum of all four serotypes is at least 4.6 log 10 pfu/dose.
  • the dengue vaccine composition comprises a tetravalent dengue virus composition including four live attenuated dengue virus strains wherein the arithmetic sum of all four serotypes is in the range of 4.6 log 10 pfu/dose to 6.7 log 10 pfu/dose, preferably in the range of 4.6 log 10 pfu/dose to 5.5 log 10 pfu/dose.
  • composition (i), (ii), (iii), and (iv) provide a total concentration of pfu/0.5 mL and based on said concentration, the concentration of (iii) at least 10% of the total concentration in pfu/0.5 mL.
  • composition (i), (ii), (iii), and (iv) provide a total concentration of pfu/0.5 mL and based on said total concentration the concentration of (ii) in pfu/0.5 mL is less than 10%, and the concentration of (iv) in pfu/0.5 mL is at least 50%, and the concentration of (i) in pfu/0.5 mL is at least 1%, and the concentration of (iii) in pfu/0.5 mL is at least 8%, or at least 10%, or at least 12%, or at least 14%, or at least 16%, or at least 18%.
  • concentration in the reconstituted unit dose of (iii) in pfu/0.5 mL is at least 10%.
  • composition (i), (ii), (iii), and (iv) provide a total concentration of pfu/0.5 mL and based on said total concentration the concentration of (ii) in pfu/0.5 mL is less than 2%, the concentration of (iv) in pfu/0.5 mL is at least 50%, the concentration of (i) in pfu/0.5 mL is at least 1%, and the concentration of (iii) in pfu/0.5 mL is at least 6%.
  • the chimeric dengue serotype 2/1 strain is TDV-1
  • the dengue serotype 2 strain is TDV-2
  • the chimeric dengue serotype 2/3 strain is TDV-3
  • the chimeric dengue serotype 2/4 strain is TDV-4.
  • TDV-1 is characterized by the nucleotide sequence according to SEQ ID No. 1 and the amino acid sequence according to SEQ ID No. 2
  • TDV-2 is characterized by the nucleotide sequence according to SEQ ID No. 3 and the amino acid sequence according to SEQ ID No. 4
  • TDV-3 is characterized by the nucleotide sequence according to SEQ ID No. 5
  • TDV-4 is characterized by the nucleotide sequence according to SEQ ID No. 7 and the amino acid sequence according to SEQ ID No. 8.
  • the concentration of the different dengue viruses is preferably determined by an immuno-focus assay known in the art.
  • the concentration may be determined by an immuno-focus assay wherein serial dilutions of dengue virus are applied to monolayers of adherent cells, such as Vero cells. After a period of time which allows infectious viruses to bind to the cells and to be taken up by the cells, an overlay containing thickening agents, such as agarose or carboxymethylcellulose, is added to prevent diffusion of viruses so that progeny viruses can only infect cells adjacent to the original infected cells.
  • an overlay containing thickening agents such as agarose or carboxymethylcellulose
  • cells are fixed and stained using serotype-specific anti-dengue monoclonal antibodies and a secondary antibody such as an antibody labeled with alkaline phosphatase.
  • the foci are stained by adding a suitable substrate for the enzyme attached to the secondary antibody, such as 5-bromo-4-chloro-3-indolyl-phosphate/nitro blue tetrazolium phosphatase substrate.
  • the number of plaques on the plate corresponds to the plaque forming units of the virus in the solutions applied to the cells. For example, a concentration of 1,000 pfu/ ⁇ l indicates that 1 ⁇ l of the solution applied to the cells contains enough viruses to produce 1,000 plaques in a cell monolayer.
  • the dengue vaccine composition comprises a tetravalent dengue virus composition including four live attenuated dengue virus strains, wherein a chimeric dengue serotype 2/1 strain, a dengue serotype 2 strain, a chimeric dengue serotype 2/3 strain, and a chimeric dengue serotype 2/4 strain provide a total concentration in pfu/0.5 mL.
  • total concentration in pfu/0.5 mL or “total concentration in pfu/dose” is the sum of the concentrations of the dengue serotype 1 (e.g. chimeric dengue serotype 2/1 strain), dengue serotype 2 (e.g.
  • the dengue serotype 2 strain preferably the sum of the concentrations of TDV-1, TDV-2, TDV-3 and TDV-4, and is defined as 100% of the dengue virus concentration as determined by pfu (plaque forming units) in 0.5 mL or in a dose.
  • the dengue vaccine composition comprises a tetravalent dengue virus composition including four live attenuated dengue virus strains, wherein a dengue serotype 1 (e.g. chimeric dengue serotype 2/1 strain), a dengue serotype 2 (e.g. dengue serotype 2 strain), a dengue serotype 3 (e.g. chimeric dengue serotype 2/3 strain), and a dengue serotype 4 (e.g. chimeric dengue serotype 2/4 strain) provide a total concentration in pfu/0.5 mL, wherein based on said total concentration the concentration of a dengue serotype 2 (e.g.
  • the concentration of a dengue serotype 2 (e.g. dengue serotype 2 strain) measured in pfu/0.5 mL is less than 10% of the total concentration, or less than 8%, or less than 6% of the total concentration, and wherein the concentration of a dengue serotype 4 (e.g. chimeric dengue serotype 2/4 strain) measured in pfu/0.5 mL is at least 50% or at least 60% or at least 65% of the total concentration.
  • the concentration of a dengue serotype 2 (e.g. dengue serotype 2 strain) measured in pfu/0.5 mL is 0.3 to 10% or 0.5 to 8% of the total concentration and the concentration of a dengue serotype 4 (e.g.
  • chimeric dengue serotype 2/4 strain measured in pfu/0.5 mL is 50% to 90% or 60% to 88% of the total concentration. This means that the concentration of the dengue serotype 2 (e.g. dengue serotype 2 strain) is lower than the concentration of the dengue serotype 4 (e.g. chimeric dengue serotype 2/4 strain).
  • the concentration of a dengue serotype 1 (e.g. chimeric dengue serotype 2/1 strain) measured in pfu/0.5 mL is at least 1% of the total concentration
  • the concentration of a dengue serotype 3 (e.g. chimeric dengue serotype 2/3 strain) measured in pfu/0.5 mL is at least 6% of the total concentration, or at least 7% or 8%, 10%, 12%, 14%, 16% or 18% of the total concentration.
  • the concentration of a dengue serotype 2 (e.g.
  • chimeric dengue serotype 2/1 strain measured in pfu/0.5 mL is 1% to 7% or 2% to 6% or 2.0% to 5.0% of the total concentration, and/or the concentration of a dengue serotype 3 (e.g. chimeric dengue serotype 2/3 strain) measured in pfu/0.5 mL is 6% to 25% or 7% to 25% or 10% to 25% or 18% to 25% of the total concentration.
  • concentration of the dengue serotype 1 e.g. chimeric dengue serotype 2/1 strain
  • the concentration of the dengue serotype 3 e.g. chimeric dengue serotype 2/3 strain.
  • the concentration of a dengue serotype 2 strain, such as TDV-2, measured in pfu/0.5 mL is less than 10% of the total concentration, preferably less than 6% or less than 2%
  • the concentration of a dengue serotype 4 (e.g. chimeric dengue serotype 2/4 strain), such as TDV-4, measured in pfu/0.5 mL is at least 50% of the total concentration, preferably at least 65%
  • the concentration of a dengue serotype 1 e.g.
  • chimeric dengue serotype 2/1 strain such as TDV-1
  • concentration of a dengue serotype 3 e.g. chimeric dengue serotype 2/3 strain
  • TDV-3 measured in pfu/0.5 mL is at least 6% of the total concentration, preferably between 6% and 25% or 10% to 25% or 18% to 25%.
  • a dengue virus composition comprising a dengue serotype 1 (e.g. chimeric dengue serotype 2/1 strain), a dengue serotype 2 (e.g. dengue serotype 2 strain), a dengue serotype 1 (e.g. chimeric dengue serotype 2/3 strain), and a dengue serotype 4 (e.g. chimeric dengue serotype 2/4 strain), such as TDV-1, TDV-2, TDV-3 and TDV-4, is provided, wherein the concentration of the dengue serotype 1 (e.g.
  • chimeric dengue serotype 2/1 strain measured in pfu/0.5 mL is at least 1% of the total concentration, preferably between 1% and 7% or 2.0% and 5.0%
  • the concentration of the dengue serotype 2 (e.g. dengue serotype 2 strain) measured in pfu/0.5 mL is less than 10% of the total concentration, preferably less than 6% or less than 2%
  • the concentration of the dengue serotype 3 (e.g. chimeric dengue serotype 2/3 strain) measured in pfu/0.5 mL is at least 6% of the total concentration, preferably between 6% and 25% or 10% to 25% or 18% to 25%.
  • the dengue serotype 4 e.g. chimeric dengue serotype 2/4 strain
  • the dengue vaccine composition comprises a tetravalent dengue virus composition including four live attenuated dengue virus strains, wherein the concentration of the dengue serotype 1 (e.g. chimeric dengue serotype 2/1 strain) measured in pfu/0.5 mL is 1% to 7% of the total concentration, the concentration of the dengue serotype 2 (e.g. dengue serotype 2 strain) measured in pfu/0.5 mL is less than 8% of the total concentration, such as in the range of 1% to 8% of the total concentration, the concentration of the dengue serotype 3 (e.g.
  • the arithmetic sum of all four serotypes is in the range of 4.6 log 10 pfu/0.5 mL to 6.7 log 10 pfu/0.5 mL, preferably in the range of 4.6 log 10 pfu/0.5 mL to 5.5 log 10 pfu/0.5 mL.
  • the dengue serotype 1 e.g. chimeric dengue serotype 2/1 strain
  • the dengue serotype 2 e.g. dengue serotype 2 strain
  • the dengue serotype 3 e.g. chimeric dengue serotype 2/3 strain
  • the dengue serotype 4 is preferably at least about 10% of the total concentration in pfu/0.5 mL and more preferably the dengue serotype 4 (e.g.
  • chimeric dengue serotype 2/4 strain such as TDV-4 is at least about 70% of the total concentration in pfu/0.5 mL.
  • the dengue serotype 4 e.g. chimeric dengue serotype 2/4 strain
  • TDV-4 represents the highest concentration in the composition of all four serotypes, preferably with at least about 70% of the total concentration in pfu/0.5 mL
  • dengue serotype 3 e.g. chimeric dengue serotype 2/3 strain
  • TDV-3 represents the second highest concentration in the composition of all four serotypes, preferably with at least about 10% of the total concentration in pfu/0.5 mL
  • dengue serotype 1 e.g.
  • chimeric dengue serotype 2/1 strain such as TDV-1 and dengue serotype 2 (e.g. dengue serotype 2 strain) such as TDV-2 each represent lower concentrations than the concentration of serotype 3 (e.g. chimeric dengue serotype 2/3 strain) such as TDV-3, and optionally together represent less than about 10% of the total concentration in pfu/0.5 mL.
  • the chimeric dengue serotype 2/1 strain is TDV-1
  • the dengue serotype 2 strain is TDV-2
  • the chimeric dengue serotype 2/3 strain is TDV-3
  • the chimeric dengue serotype 2/4 strain is TDV-4.
  • TDV-1 is characterized by the nucleotide sequence according to SEQ ID No. 1 and the amino acid sequence according to SEQ ID No. 2
  • TDV-2 is characterized by the nucleotide sequence according to SEQ ID No. 3 and the amino acid sequence according to SEQ ID No. 4
  • TDV-3 is characterized by the nucleotide sequence according to SEQ ID No. 5
  • TDV-4 is characterized by the nucleotide sequence according to SEQ ID No. 7 and the amino acid sequence according to SEQ ID No. 8.
  • the chimeric dengue serotype 2/4 strain preferably TDV-4
  • 0.5 ml is the reference volume for the determination of the concentrations of the virus strains in the composition in pfu/ml. The volume and/or amount per unit dose is described in the respective chapter.
  • the present invention is in part directed to a unit dose of a dengue vaccine composition, wherein the dengue vaccine composition comprises one or more pharmaceutically acceptable excipients.
  • the dengue vaccine composition comprises a non-reducing sugar, a surfactant, a protein and an inorganic salt.
  • the non-reducing sugar is trehalose
  • the surfactant is poloxamer 407
  • the protein is human serum albumin
  • the inorganic salt is sodium chloride.
  • the unit dose of a dengue vaccine composition comprises the following pharmaceutically acceptable excipients:
  • the unit dose of a dengue vaccine composition comprises the following pharmaceutically acceptable excipients when measured in 0.5 ml:
  • the unit dose of a dengue vaccine composition comprises the following pharmaceutically acceptable excipients when measured in 0.5 ml:
  • the lyophilized unit dose of the invention as described herein comprises the following pharmaceutically acceptable excipients:
  • the lyophilized unit dose of the invention as described herein comprises the following pharmaceutically acceptable excipients when measured in 0.5 ml:
  • the lyophilized unit dose of the invention as described herein comprises the following pharmaceutically acceptable excipients:
  • the reconstituted unit dose of the invention as described herein comprises the following pharmaceutically acceptable excipients:
  • the reconstituted unit dose of the invention as described herein comprises the following pharmaceutically acceptable excipients when measured in 0.5 ml:
  • the reconstituted unit dose of the invention as described herein comprises the following pharmaceutically acceptable excipients:
  • the human serum albumin may be a native or recombinant human serum albumin (rHSA).
  • the poloxamer 407 may be e.g. Pluronic F127.
  • the unit dose further comprises a buffer.
  • the buffer may be phosphate buffered saline (PBS).
  • the buffer may include at least one of sodium chloride (NaCl), monosodium dihydrogen phosphate (NaH 2 PO 4 ), disodium hydrogen phosphate (Na 2 HPO 4 ), potassium chloride (KCl), and potassium dihydrogen phosphate (KH 2 PO 4 ).
  • the buffer may include disodium hydrogen phosphate (Na 2 HPO 4 ), potassium chloride (KCl), and potassium dihydrogen phosphate (KH 2 PO 4 ).
  • the buffer may have a pH in the range of 7.0 to 8.5 at 25° C.
  • the present invention is directed in part to a unit dose of a dengue vaccine composition
  • a dengue vaccine composition comprising a tetravalent dengue virus composition as described herein and pharmaceutically acceptable excipients as described herein.
  • the present invention is directed in part to a unit dose of a dengue vaccine composition as described above e.g. of
  • the chimeric dengue serotype 2/1 strain is TDV-1
  • the dengue serotype 2 strain is TDV-2
  • the chimeric dengue serotype 2/3 strain is TDV-3
  • the chimeric dengue serotype 2/4 strain is TDV-4.
  • TDV-1 is characterized by the nucleotide sequence according to SEQ ID No. 1 and the amino acid sequence according to SEQ ID No. 2
  • TDV-2 is characterized by the nucleotide sequence according to SEQ ID No. 3 and the amino acid sequence according to SEQ ID No. 4
  • TDV-3 is characterized by the nucleotide sequence according to SEQ ID No. 5
  • TDV-4 is characterized by the nucleotide sequence according to SEQ ID No. 7 and the amino acid sequence according to SEQ ID No. 8.
  • the unit dose is lyophilized.
  • the lyophilized unit dose is obtained by subjecting a volume of 0.5 mL of the aqueous dengue vaccine composition produced by combining pharmaceutically acceptable excipients as described herein and the dengue vaccine composition as described herein comprising the four dengue virus strains, in particular TDV-1 to TDV-4, to lyophilization.
  • the residual moisture content as determined by Karl Fischer Determination is equal to or less than 5.0%, preferably equal to or less than 3%.
  • the unit dose is reconstituted.
  • the reconstituted unit dose is obtained by subjecting the lyophilized unit dose to reconstitution with a pharmaceutically acceptable diluent, preferably before administration of the dengue vaccine.
  • reconstitution will be accomplished by adding a pharmaceutically acceptable diluent, such as water for injection, phosphate buffered saline or an aqueous sodium chloride solution, to the lyophilized unit dose.
  • a pharmaceutically acceptable diluent such as water for injection, phosphate buffered saline or an aqueous sodium chloride solution
  • an aqueous sodium chloride solution such as a 37 mM aqueous sodium chloride solution, is added to the lyophilized unit dose for reconstitution.
  • the lyophilized unit dose will be reconstituted with 0.3 to 0.8 mL, or 0.4 to 0.7 mL, or 0.5 mL of diluent.
  • the lyophilized unit dose is reconstituted with 0.3 to 0.8 mL, 0.4 to 0.7 mL or 0.5 mL of 37 mM aqueous sodium chloride solution.
  • the lyophilized unit dose is reconstituted with 0.5 mL of 37 mM aqueous sodium chloride solution.
  • the reconstituted unit dose can subsequently be administered subcutaneously.
  • the unit dose in lyophilized form is the final product after manufacture of the unit dose and the storage form of the unit dose, wherein the unit dose in reconstituted form is prepared before administration of the unit dose to a subject.
  • the present invention is, moreover, directed in part to a unit dose of a dengue vaccine composition
  • a dengue vaccine composition comprising:
  • a tetravalent virus composition including four live attenuated dengue virus strains, wherein the unit dose is lyophilized and upon reconstitution with 0.5 mL of a pharmaceutically acceptable diluent comprises:
  • the reconstituted unit dose has a volume of e.g. 0.5 mL, wherein upon reconstitution with a pharmaceutically acceptable diluent (i), (ii), (iii), and (iv) provide a total concentration of pfu/0.5 mL and based on said concentration, the concentration of (iii) at least 10% of the total concentration in pfu/0.5 mL.
  • the reconstituted unit dose has a volume of e.g. 0.5 mL, wherein upon reconstitution with a pharmaceutically acceptable diluent (i), (ii), (iii), and (iv) provide a total concentration of pfu/0.5 mL and based on said total concentration the concentration of (ii) in pfu/0.5 mL is less than 10%, and the concentration of (iv) in pfu/0.5 mL is at least 50%, and the concentration of (i) in pfu/0.5 mL is at least 1%, and the concentration of (iii) in pfu/0.5 mL is at least 8%, or at least 10%, or at least 12%, or at least 14%, or at least 16%, or at least 18%.
  • concentration in the reconstituted unit dose of (iii) in pfu/0.5 mL is at least 10%.
  • the reconstituted unit dose has a volume of e.g. 0.5 mL, wherein upon reconstitution with a pharmaceutically acceptable diluent (i), (ii), (iii), and (iv) provide a total concentration of pfu/0.5 mL and based on said total concentration the concentration of (ii) in pfu/0.5 mL is less than 2%, the concentration of (iv) in pfu/0.5 mL is at least 50%, the concentration of (i) in pfu/0.5 mL is at least 1%, and the concentration of (iii) in pfu/0.5 mL is at least 6%.
  • the present invention is directed to a lyophilized unit dose of a dengue vaccine composition
  • a dengue serotype 1 e.g. chimeric dengue serotype 2/1 strain
  • a dengue serotype 2 e.g. dengue serotype 2 strain
  • a dengue serotype 3 e.g.
  • the chimeric dengue serotype 2/3 strain with a concentration of at least 4.0 log 10 pfu/0.5 mL
  • a dengue serotype 4 e.g. chimeric dengue serotype 2/4 strain
  • the unit dose is preferably formulated in 0.5 mL before lyophilization.
  • the chimeric dengue serotype 2/1 strain is TDV-1
  • the dengue serotype 2 strain is TDV-2
  • the chimeric dengue serotype 2/3 strain is TDV-3
  • the chimeric dengue serotype 2/4 strain is TDV-4.
  • TDV-1 is characterized by the nucleotide sequence according to SEQ ID No. 1 and the amino acid sequence according to SEQ ID No. 2
  • TDV-2 is characterized by the nucleotide sequence according to SEQ ID No. 3 and the amino acid sequence according to SEQ ID No. 4
  • TDV-3 is characterized by the nucleotide sequence according to SEQ ID No. 5 and the amino acid sequence according to SEQ ID No. 6
  • TDV-4 is characterized by the nucleotide sequence according to SEQ ID No. 7 and the amino acid sequence according to SEQ ID No. 8.
  • the lyophilized unit dose is obtained by lyophilizing 0.5 mL of a dengue vaccine composition comprising a dengue serotype 1 (e.g. chimeric dengue serotype 2/1 strain) in a concentration of 3.3 log 10 pfu/dose to 5.0 log 10 pfu/0.5 mL, a dengue serotype 2 (e.g. dengue serotype 2 strain) in a concentration of 2.7 log 10 pfu/dose to 4.9 log 10 pfu/0.5 mL, a dengue serotype 3 (e.g.
  • a dengue serotype 1 e.g. chimeric dengue serotype 2/1 strain
  • a dengue serotype 2 e.g. dengue serotype 2 strain
  • a dengue serotype 3 e.g.
  • chimeric dengue serotype 2/3 strain in a concentration of 4.0 log 10 pfu/dose to 5.7 log 10 pfu/0.5 mL
  • a dengue serotype 4 e.g. chimeric dengue serotype 2/4 strain
  • the chimeric dengue serotype 2/1 strain is TDV-1
  • the dengue serotype 2 strain is TDV-2
  • the chimeric dengue serotype 2/3 strain is TDV-3
  • the chimeric dengue serotype 2/4 strain is TDV-4.
  • the lyophilized unit dose is obtained by lyophilizing 0.5 mL of a dengue vaccine composition comprising a dengue serotype 1 (e.g. chimeric dengue serotype 2/1 strain) in a concentration of 3.3 log 10 pfu/0.5 mL to 3.6 log 10 pfu/0.5 mL, a dengue serotype 2 (e.g. dengue serotype 2 strain) in a concentration of 2.7 log 10 pfu/0.5 mL to 4.0 log 10 pfu/0.5 mL, a dengue serotype 3 (e.g.
  • a dengue serotype 1 e.g. chimeric dengue serotype 2/1 strain
  • a dengue serotype 2 e.g. dengue serotype 2 strain
  • a dengue serotype 3 e.g.
  • chimeric dengue serotype 2/3 strain in a concentration of 4.0 log 10 pfu/0.5 mL to 4.6 log 10 pfu/0.5 mL, and a dengue serotype 4 (e.g. chimeric dengue serotype 2/4 strain) in a concentration of 4.5 log 10 pfu/0.5 mL or 4.6 log 10 pfu/0.5 mL to 5.1 log 10 pfu/0.5 mL and pharmaceutically acceptable excipients as described herein.
  • a dengue serotype 4 e.g. chimeric dengue serotype 2/4 strain
  • the chimeric dengue serotype 2/1 strain is TDV-1
  • the dengue serotype 2 strain is TDV-2
  • the chimeric dengue serotype 2/3 strain is TDV-3
  • the chimeric dengue serotype 2/4 strain is TDV-4.
  • the lyophilized unit dose refers to 0.5 mL before lyophilization, wherein TDV-2 and TDV-4 are present in certain relative amounts, based on the total concentration of TDV-1, TDV-2, TDV-3 and TDV-4 in pfu/0.5 mL, and the concentration of TDV-2 measured in pfu/0.5 mL is less than 10% or less than 8% or less than 6%, and the concentration of TDV-4 measured in pfu/0.5 mL is at least 50% or at least 65%.
  • the concentration of TDV-1 measured in pfu/0.5 mL is at least 1% and/or the concentration of TDV-3 measured in pfu/0.5 mL is at least 6%, 7%, 8%, 10%, 12%, 14%, 16% k or at least 18%.
  • the reconstituted unit dose has a volume of 0.5 mL and TDV-2 and TDV-4 are present in certain relative amounts, based on the total concentration of TDV-1, TDV-2, TDV-3 and TDV-4 in pfu/0.5 mL, and the concentration of TDV-2 measured in pfu/0.5 mL is less than 10% or less than 8% or less than 6%, and the concentration of TDV-4 measured in pfu/0.5 mL is at least 50% or at least 65%.
  • the concentration of TDV-1 measured in pfu/0.5 mL is at least 1% and/or the concentration of TDV-3 measured in pfu/0.5 mL is at least 6%, 7%, 8%, 10%, 12%, 14%, 16% k or at least 18%.
  • the reconstituted unit dose has a volume of 0.5 mL and comprises a tetravalent dengue virus composition including four live attenuated dengue virus strains, wherein the concentration of the dengue serotype 1 (e.g. dengue serotype 2/1 strain) measured in pfu/0.5 mL is 1% to 7% of the total concentration, the concentration of the dengue serotype 2 (e.g dengue serotype 2 strain) measured in pfu/0.5 mL is less than 8% of the total concentration, such as in the range of 1% to 8% of the total concentration, the concentration of the dengue serotype 3 (e.g.
  • the dengue serotype 2/3 strain measured in pfu/0.5 mL is at least 10% of the total concentration
  • the concentration of the dengue serotype 4 (e.g. dengue serotype 2/4 strain) measured in pfu/0.5 mL is at least 65% of the total concentration, such as in the range of 65% to 80%.
  • the arithmetic sum of all four serotypes is in the range of 4.6 log 10 pfu/0.5 mL to 6.7 log 10 pfu/0.5 mL, preferably in the range of 4.6 log 10 pfu/0.5 mL to 5.5 log 10 pfu/0.5 mL.
  • the reconstituted unit dose has a volume of 0.5 mL and comprises a tetravalent dengue virus composition including four live attenuated dengue virus strains, wherein the dengue serotype 1 (e.g. chimeric dengue serotype 2/1 strain) such as TDV-1 and the dengue serotype 2 (e.g. dengue serotype 2 strain) such as TDV-2 are present each in a concentration based on the total concentration in pfu/0.5 mL which is within 5%-points of each other and/or are together less than about 10% of the total concentration in pfu/0.5 mL.
  • the dengue serotype 3 e.g.
  • the dengue serotype 4 e.g. chimeric dengue serotype 2/4 strain
  • the dengue serotype 4 represents the highest concentration in the composition of all four serotypes, preferably with at least about 70% of the total concentration in pfu/0.5 mL, dengue serotype 3 (e.g.
  • chimeric dengue serotype 2/3 strain such as TDV-3 represents the second highest concentration in the composition of all four serotypes, preferably with at least about 10% of the total concentration in pfu/0.5 mL
  • dengue serotype 1 e.g. chimeric dengue serotype 2/1 strain
  • dengue serotype 2 e.g. dengue serotype 2 strain
  • TDV-2 each represent lower concentrations than the concentration of serotype 3 (e.g. chimeric dengue serotype 2/3 strain) such as TDV-3, and optionally together represent less than about 10% of the total concentration in pfu/0.5 mL.
  • the lyophilized unit dose reconstituted in 0.5 mL will provide the above concentrations for the four dengue serotypes. While the unit dose of a dengue vaccine composition as described herein refers to the concentrations of the dengue serotypes in 0.5 mL, the lyophilized unit dose can be reconstituted with other volumes of a pharmaceutically acceptable diluent, such as an aqueous sodium chloride solution, without changing the absolute virus amount administered or the ratios of the viruses to one another.
  • a pharmaceutically acceptable diluent such as an aqueous sodium chloride solution
  • the lyophilized unit dose of the invention is prepared from a solution comprising a non-reducing sugar, a surfactant, a protein and an inorganic salt.
  • the lyophilized unit dose of the invention is prepared from a solution comprising trehalose, poloxamer 407, human serum albumin and sodium chloride.
  • the lyophilized unit dose of the invention is prepared from a solution comprising about 10% w/v to about 20% w/v ⁇ , ⁇ -trehalose dihydrate or an equimolar amount of other forms of ⁇ , ⁇ -trehalose, from about 0.5% w/v to about 1.5% w/v poloxamer 407, from about 0.05% w/v to about 2% w/v human serum albumin, and about 70 mM to about 120 mM sodium chloride.
  • the lyophilized unit dose of the invention as described herein is prepared from a solution comprising about 15% w/v ⁇ , ⁇ -trehalose dihydrate, about 1% w/v poloxamer 407, about 0.1% w/v human serum albumin and about 100 mM sodium chloride.
  • the solution from which the lyophilized unit dose is prepared further comprises a buffer.
  • the buffer may be phosphate buffered saline (PBS).
  • the buffer may include at least one of sodium chloride (NaCl), monosodium dihydrogen phosphate (NaH 2 PO 4 ), disodium hydrogen phosphate (Na 2 HPO 4 ), potassium chloride (KCl), and potassium dihydrogen phosphate (KH 2 PO 4 ).
  • the buffer may include disodium hydrogen phosphate (Na 2 HPO 4 ), potassium chloride (KCl), and potassium dihydrogen phosphate (KH 2 PO 4 ).
  • the buffer may have a pH in the range of about 7.0 to about 8.5 at 25° C. or a pH of about 6.8 to about 7.6 at 25° C., preferably a pH of about 7.2 at 25° C.
  • the reconstituted unit dose of the invention as described herein comprising about 15% w/v ⁇ , ⁇ -trehalose dihydrate, about 1% w/v poloxamer 407, about 0.1% w/v human serum albumin and about 137 mM sodium chloride.
  • the reconstituted unit dose may have a pH of about 7.0 to about 8.5 at 25° C., preferably a pH of about 7.2 at 25° C.
  • the unit dose of the invention as described herein activates multiple arms of the immune system—neutralizing antibodies, cellular immunity and anti-NS1 antibodies—in both seronegative and seropositive subject populations or in both seronegative and seropositive subjects.
  • the unit dose of the invention as described herein protects both dengue seronegative and dengue seropositive subject populations or subjects against dengue disease.
  • one unit dose is present in a container, preferably a vial, and said unit dose is administered to a subject after reconstitution.
  • more than one unit dose of the dengue vaccine composition may be present in a container, preferably a vial, so that with the content of one container, preferably a vial, more than one subject can be vaccinated.
  • the container comprising more than one unit doses of the invention as described herein is used for providing the reconstituted unit dose to be used in the methods of the invention as described herein.
  • the container comprising the unit dose of the invention is part of a kit.
  • the invention is directed in part to a kit for preparing a reconstituted unit dose comprising a lyophilized unit dose of the present invention as described herein, and a pharmaceutically acceptable diluent for reconstitution.
  • the diluent for reconstitution provided in a container, preferably a vial, or a pre-filled syringe.
  • the diluent for reconstitution is selected from water for injection, phosphate buffered saline or an aqueous sodium chloride solution.
  • the diluent for reconstitution is 30 to 40 mM sodium chloride, such as 37 mM sodium chloride.
  • the kit may further comprise a yellow fever vaccine, in particular YF-17D.
  • the yellow fever vaccine may be in a separate container, such as a vial.
  • the yellow fever vaccine and the unit dose of the invention may be in the same container.
  • the invention is directed in part to a combined dengue/yellow fever vaccine, wherein the unit dose of the invention as described herein is combined with a yellow fever vaccine.
  • a combined dengue/yellow fever vaccine comprises the unit dose of the invention as described herein and a yellow fever vaccine, in particular YF-17D, in the same formulation.
  • the invention is directed to a kit comprising such a combined dengue/yellow fever vaccine and a unit dose of the invention as described herein.
  • the kit may further comprise a hepatitis A vaccine, such as HAVRIX® or VAQTA®.
  • the hepatitis A vaccine may be in a separate container, such as a vial.
  • the hepatitis A vaccine and the unit dose of the invention may be in the same container.
  • the invention is directed in part to a combined dengue/hepatitis A vaccine, wherein the unit dose of the invention as described herein is combined with a hepatitis A vaccine.
  • a combined dengue/hepatitis A vaccine comprises the unit dose of the invention as described herein and a hepatitis A vaccine, such as HAVRIX® or VAQTA®, in the same formulation.
  • the invention is directed to a kit comprising such a combined dengue/hepatitis A vaccine and a unit dose of the invention as described herein.
  • the kit may further comprise a HPV vaccine, in particular a 9vHPV vaccine, such as GARDASIL® 9.
  • the HPV vaccine may be in a separate container, such as a vial.
  • the HPV vaccine and the unit dose of the invention may be in the same container.
  • the invention is directed in part to a combined dengue/HPV vaccine, wherein the unit dose of the invention as described herein is combined with a HPV vaccine.
  • a combined dengue/HPV vaccine comprises the unit dose of the invention as described herein and a HPV vaccine, in particular a 9vHPV vaccine, such as GARDASIL® 9, in the same formulation.
  • the invention is directed to a kit comprising such a combined dengue/HPV vaccine and a unit dose of the invention as described herein.
  • the kit may further comprise a MMR vaccine, such as M-M-R® II.
  • MMR vaccine may be in a separate container, such as a vial.
  • the MMR vaccine and the unit dose of the invention may be in the same container.
  • the invention is directed in part to a combined dengue/MMR vaccine, wherein the unit dose of the invention as described herein is combined with a MMR vaccine.
  • a combined dengue/MMR vaccine comprises the unit dose of the invention as described herein and a MMR vaccine, such as M-M-R® II, in the same formulation.
  • the invention is directed to a kit comprising such a combined dengue/MMR vaccine and a unit dose of the invention as described herein.
  • the kit may further comprise a Tdap vaccine, in particular a combined tetanus toxoid, reduced diphtheria toxoid and acellular pertussis (adsorbed) vaccine, such as BOOSTRIX®.
  • a Tdap vaccine in particular a combined tetanus toxoid, reduced diphtheria toxoid and acellular pertussis (adsorbed) vaccine, such as BOOSTRIX®.
  • the Tdap vaccine may be in a separate container, such as a vial.
  • the Tdap vaccine and the unit dose of the invention may be in the same container.
  • the invention is directed in part to a combined dengue/Tdap vaccine, wherein the unit dose of the invention as described herein is combined with a Tdap vaccine.
  • Such a combined dengue/Tdap vaccine comprises the unit dose of the invention as described herein and a Tdap vaccine, in particular a combined tetanus toxoid, reduced diphtheria toxoid and acellular pertussis (adsorbed) vaccine, such as BOOSTRIX®, in the same formulation.
  • a Tdap vaccine in particular a combined tetanus toxoid, reduced diphtheria toxoid and acellular pertussis (adsorbed) vaccine, such as BOOSTRIX®, in the same formulation.
  • the invention is directed to a kit comprising such a combined dengue/Tdap vaccine and a unit dose of the invention as described herein.
  • the kit may further comprise a DTaP/IPV/Hib vaccine, in particular a combined DTaP/IPV/Hib vaccine, such as Pentacel®.
  • the DTaP/IPV/Hib vaccine may be in a separate container, such as a vial.
  • the DTaP/IPV/Hib vaccine and the unit dose of the invention may be in the same container.
  • the invention is directed in part to a combined dengue/DTaP/IPV/Hib vaccine, wherein the unit dose of the invention as described herein is combined with a DTaP/IPV/Hib vaccine.
  • Such a combined dengue/DTaP/IPV/Hib vaccine comprises the unit dose of the invention as described herein and a DTaP/IPV/Hib vaccine, in particular a combined DTaP/IPV/Hib vaccine, such as Pentacel®, in the same formulation.
  • the invention is directed to a kit comprising such a combined dengue/DTaP/IPV/Hib vaccine and a unit dose of the invention as described herein.
  • YF-VAX® a yellow fever vaccine from Sanofi, for subcutaneous use, is prepared by culturing the YF-17D strain of yellow fever virus in living avian leukosis virus-free (ALV-free) chicken embryos.
  • the vaccine contains sorbitol and gelatin as a stabilizer and is lyophilized. No preservative is added.
  • YF-VAX is formulated to contain not less than 4.74 log 10 pfu per 0.5 mL dose throughout the life of the product.
  • HAVRIX® a hepatitis A vaccine from GlaxoSmithKline
  • the virus (strain HM175) is propagated in MRC-5 human diploid cells. After removal of the cell culture medium, the cells are lysed to form a suspension. This suspension is purified through ultrafiltration and gel permeation chromatography procedures. Treatment of this lysate with formalin ensures viral inactivation.
  • Viral antigen activity is referenced to a standard using an enzyme linked immunosorbent assay (ELISA), and is therefore expressed in terms of ELISA Units (EL.U.). Each 1-mL dose for adults 18 years of age) of vaccine contains 1440 EL.U.
  • HAVRIX® contains the following excipients: Amino acid supplement (0.3% w/v) in a phosphate-buffered saline solution and polysorbate 20 (0.05 mg/mL).
  • HAVRIX® also contains residual MRC-5 cellular proteins (not more than 5 ⁇ g/mL), formalin (not more than 0.1 mg/mL), and neomycin sulfate (not more than 40 ng/mL), an aminoglycoside antibiotic included in the cell growth media. HAVRIX® is formulated without preservatives.
  • VAQTA® a hepatitis A vaccine from Merck Sharp & Dohme Corp.
  • VAQTA® is an inactivated whole virus vaccine derived from hepatitis A virus grown in cell culture in human MRC-5 diploid fibroblasts. It contains inactivated virus of a strain, which was originally derived by further serial passage of a proven attenuated strain. The virus is grown, harvested, purified by a combination of physical and high performance liquid chromatographic techniques developed at the Merck Research Laboratories, formalin inactivated, and then adsorbed onto amorphous aluminum hydroxyphosphate sulfate.
  • VAQTA® is a sterile suspension for intramuscular injection.
  • One milliliter of the vaccine contains approximately 50 U of hepatitis A virus antigen, which is purified and formulated without a preservative.
  • the 50 U dose of VAQTA® contains less than 0.1 ⁇ g of non-viral protein, less than 4 ⁇ 10 ⁇ 6 ⁇ g of DNA, less than 10 ⁇ 4 ⁇ g of bovine albumin, and less than 0.8 ⁇ g of formaldehyde.
  • Other process chemical residuals are less than 10 parts per billion (ppb), including neomycin.
  • Each 0.5-mL pediatric dose contains 25 U of hepatitis A virus antigen and adsorbed onto approximately 0.225 mg of aluminum provided as amorphous aluminum hydroxyphosphate sulfate, and 35 ⁇ g of sodium borate as a pH stabilizer, in 0.9% sodium chloride.
  • Each 1-mL adult dose contains 50 U of hepatitis A virus antigen and adsorbed onto approximately 0.45 mg of aluminum provided as amorphous aluminum hydroxyphosphate sulfate, and 70 ⁇ g of sodium borate as a pH stabilizer, in 0.9% sodium chloride.
  • GARDASIL® 9 a HPV vaccine from Merck, is a non-infectious recombinant 9-valent vaccine prepared from the purified virus-like particles (VLPs) of the major capsid (L1) protein of HPV serotypes 6, 11, 16, 18, 31, 33, 45, 52, and 58.
  • the L1 proteins are produced by separate fermentations using recombinant Saccharomyces cerevisiae and self-assembled into VLPs. The fermentation process involves growth of S. cerevisiae on chemically-defined fermentation media which include vitamins, amino acids, mineral salts, and carbohydrates.
  • the VLPs are released from the yeast cells by cell disruption and purified by a series of chemical and physical methods.
  • the purified VLPs are adsorbed on preformed aluminum-containing adjuvant (amorphous aluminum hydroxyphosphate sulfate or AAHS).
  • the 9-valent HPV VLP vaccine is a sterile liquid suspension that is prepared by combining the adsorbed VLPs of each HPV serotype and additional amounts of the aluminum-containing adjuvant and the final purification buffer.
  • GARDASIL 9 is a sterile suspension for intramuscular administration.
  • Each 0.5-mL dose contains approximately 30 ⁇ g of HPV serotype 6 L1 protein, 40 ⁇ g of HPV serotype 11 L1 protein, 60 ⁇ g of HPV serotype 16 L1 protein, 40 ⁇ g of HPV serotype 18 L1 protein, 20 ⁇ g of HPV serotype 31 L1 protein, 20 ⁇ g of HPV serotype 33 L1 protein, 20 ⁇ g of HPV serotype 45 L1 protein, 20 ⁇ g of HPV serotype 52 L1 protein, and 20 ⁇ g of HPV serotype 58 L1 protein.
  • Each 0.5-mL dose of the vaccine also contains approximately 500 ⁇ g of aluminum (provided as AAHS), 9.56 mg of sodium chloride, 0.78 mg of L-histidine, 50 ⁇ g of polysorbate 80, 35 ⁇ g of sodium borate.
  • MMR vaccines are known in the prior art and include M-M-R® II, Priorix®, Tresivac®, and Trimovax®.
  • M-M-R® II a MMR vaccine from Merck Sharp & Dohme Corp, is a live virus vaccine for vaccination against measles, mumps and rubella.
  • M-M-R® II is a sterile lyophilized preparation of (1) ATTENUVAX® (measles virus vaccine live), a more attenuated line of measles virus, derived from Enders' attenuated Edmonston strain and propagated in chick embryo cell culture, (2) MUMPSVAX® (mumps virus vaccine live), the Jeryl LynnTM (B level) strain of mumps virus propagated in chick embryo cell culture, and (3) MERUVAX® II (rubella virus vaccine live), the Wistar RA 27/3 strain of live attenuated rubella virus propagated in WI-38 human diploid lung fibroblasts.
  • the growth medium for measles and mumps is Medium 199 (a buffered salt solution containing vitamins and amino acids and supplemented with fetal bovine serum) containing SPGA (sucrose, phosphate, glutamate, and recombinant human albumin) as stabilizer and neomycin.
  • the growth medium for rubella is Minimum Essential Medium (MEM) (a buffered salt solution containing vitamins and amino acids and supplemented with fetal bovine serum) containing recombinant human albumin and neomycin. Sorbitol and hydrolyzed gelatin stabilizer are added to the individual virus harvests. The cells, virus pools, and fetal bovine serum are all screened for the absence of adventitious agents.
  • the reconstituted vaccine is for subcutaneous administration.
  • Each 0.5 mL dose contains not less than 1,000 TCID 50 (tissue culture infectious doses) of measles virus, 12,500 TCID 50 of mumps virus, and 1,000 TCID 50 of rubella virus.
  • Each dose of the vaccine is calculated to contain sorbitol (14.5 mg), sodium phosphate, sucrose (1.9 mg), sodium chloride, hydrolyzed gelatin (14.5 mg), recombinant human albumin ( ⁇ 0.3 mg), fetal bovine serum ( ⁇ 1 ppm), other buffer and media ingredients and approximately 25 ⁇ g of neomycin.
  • the product contains no preservative.
  • the lyophilized vaccine is reconstituted before administration.
  • BOOSTRIX® a combined tetanus toxoid, reduced diphtheria toxoid and acellular pertussis (adsorbed) (Tdap) vaccine, is a non-infectious, sterile vaccine for intramuscular administration. It contains tetanus toxoid, diphtheria toxoid, and pertussis antigens (inactivated pertussis toxin (iPT) and formaldehyde-treated filamentous hemagglutinin (FHA) and pertactin (PRN)). The antigens are the same as those in INFANRIX®, but BOOSTRIX® is formulated with reduced quantities of these antigens.
  • BOOSTRIX® is supplied as a 0.5 mL suspension for injection. Each 0.5 mL dose of BOOSTRIX® is formulated to contain 5 Limits of flocculation (Lf) of tetanus toxoid, 2.5 Lf of diphtheria toxoid, 8 ⁇ g of iPT, 8 ⁇ g of formaldehyde treated FHA, and 2.5 ⁇ g of PRN (69 kiloDalton outer membrane protein), aluminum hydroxide as adjuvant (not more than 0.39 mg aluminum by assay), 4.5 mg of sodium chloride, 1.00 ⁇ g of residual formaldehyde, and 1.00 ⁇ g of polysorbate 80 (Tween 80).
  • Lf Limits of flocculation
  • iPT diphtheria toxoid
  • PRN 69 kiloDalton outer membrane protein
  • PRN 69 kiloDalton outer membrane protein
  • aluminum hydroxide as adjuvant not more than 0.39 mg aluminum by as
  • Tetanus toxin is produced by growing Clostridium tetani in a modified Latham medium derived from bovine casein.
  • the diphtheria toxin is produced by growing Corynebacterium diphtheriae in Fenton medium containing a bovine extract. Both toxins are detoxified with formaldehyde, concentrated by ultrafiltration, and purified by precipitation, dialysis, and sterile filtration. Tetanus and diphtheria toxoid potency is determined by measuring the amount of neutralizing antitoxin in previously immunized guinea pigs.
  • the acellular pertussis antigens are isolated from Bordetella pertussis culture grown in modified Stainer-Scholte liquid medium.
  • iPT and FHA are isolated from the fermentation broth, PRN is extracted from the cells by heat treatment and flocculation.
  • the antigens are purified in successive chromatographic and precipitation steps.
  • iPT is detoxified using glutaraldehyde and formaldehyde.
  • FHA and PRN are treated with formaldehyde. Each antigen is individually adsorbed onto aluminum hydroxide.
  • the potency of the acellular pertussis components is determined by enzyme-linked immunosorbent assay (ELISA) on sera from previously immunized mice.
  • Pentacel® a combined DTaP/IPV/Hib vaccine, consists of a diphtheria and tetanus toxoids and acellular pertussis adsorbed and inactivated poliovirus (DTaP-IPV) component and an ActHIB® component combined through reconstitution for intramuscular injection.
  • ActHIB® Haemophilus b Conjugate Vaccine (Tetanus Toxoid Conjugate)
  • the DTaP-IPV component is supplied as a sterile liquid used to reconstitute the lyophilized ActHIB® component to form Pentacel®.
  • Each 0.5 mL dose contains 15 Limits of flocculation (Lf) diphtheria toxoid, 5 Lf tetanus toxoid, acellular pertussis antigens (20 ⁇ g detoxified pertussis toxin (PT), 20 ⁇ g filamentous hemagglutinin (FHA), 3 ⁇ g pertactin (PRN), 5 ⁇ g fimbriae types 2 and 3 (FIM)), inactivated polioviruses (40 D-antigen units (DU) Type 1 (Mahoney), 8 DU Type 2 (MEF-1), 32 DU Type 3 (Saukett)) and 10 ⁇ g PRP of Haemophilius influenzae type b covalently bound to 24 ⁇ g of tetanus toxoid (PRP-T).
  • Lf flocculation
  • PT detoxified pertussis toxin
  • FHA filamentous hemagglutinin
  • PRN 3 ⁇
  • ingredients per 0.5 mL dose include 1.5 mg aluminum phosphate (0.33 mg aluminum) as the adjuvant, polysorbate 80 (approximately 10 ppm by calculation), 42.5 mg sucrose, ⁇ 5 ⁇ g residual formaldehyde, ⁇ 50 ng residual glutaraldehyde, ⁇ 50 ng residual bovine serum albumin, 3.3 mg (0.6% v/v) 2-phenoxyethanol (not as a preservative), ⁇ 4 pg of neomycin and ⁇ 4 pg polymyxin B sulfate.
  • Corynebacterium diphtheriae is grown in modified Mueller's growth medium. After purification by ammonium sulfate fractionation, the diphtheria toxin is detoxified with formaldehyde and diafiltered.
  • Clostridium tetani is grown in modified Mueller-Miller casamino acid medium without beef heart infusion. Tetanus toxin is detoxified with formaldehyde and purified by ammonium sulfate fractionation and diafiltration.
  • Diphtheria and tetanus toxoids are individually adsorbed onto aluminum phosphate.
  • the acellular pertussis vaccine antigens are produced from Bordetella pertussis cultures grown in Stainer-Scholte medium modified by the addition of casamino acids and dimethyl-beta-cyclodextrin.
  • PT, FHA and PRN are isolated separately from the supernatant culture medium.
  • FIM are extracted and co-purified from the bacterial cells.
  • the pertussis antigens are purified by sequential filtration, salt-precipitation, ultrafiltration and chromatography.
  • PT is detoxified with glutaraldehyde.
  • FHA is treated with formaldehyde and the residual aldehydes are removed by ultrafiltration.
  • the individual antigens are adsorbed separately onto aluminum phosphate.
  • Poliovirus Type 1, Type 2 and Type 3 are each grown in separate cultures of MRC-5 cells, a line of normal human diploid cells, by the microcarrier method.
  • the cells are grown in CMRL (Connaught Medical Research Laboratories) 1969 medium, supplemented with calf serum.
  • the culture medium is replaced by Medium 199, without calf serum.
  • the viral suspensions are concentrated by ultrafiltration, and purified by liquid chromatography steps.
  • the monovalent viral suspensions are inactivated with formaldehyde.
  • Monovalent concentrates of each inactivated poliovirus are combined to produce a trivalent poliovirus concentrate.
  • the adsorbed diphtheria, tetanus and acellular pertussis antigens are combined with aluminum phosphate (as adjuvant), 2-phenoxyethanol (not as a preservative) and water for injection, into an intermediate concentrate.
  • aluminum phosphate as adjuvant
  • 2-phenoxyethanol not as a preservative
  • water for injection into an intermediate concentrate.
  • the trivalent poliovirus concentrate is added and the DTaP-IPV component is diluted to its final concentration.
  • the DTaP-IPV component does not contain a preservative.
  • Both diphtheria and tetanus toxoids induce at least 2 neutralizing units per mL in the guinea pig potency test.
  • the potency of the acellular pertussis antigens is evaluated by the antibody response of immunized mice to detoxified PT, FHA, PRN and FIM as measured by enzyme-linked immunosorbent assay (ELISA).
  • the potency of inactivated poliovirus antigens is determined by measuring antibody-mediated neutralization of poliovirus in sera from immunized rats.
  • PRP a high molecular weight polymer
  • the tetanus toxoid for conjugation to PRP is prepared by ammonium sulfate purification, and formalin inactivation of the toxin from cultures of Clostridium tetani (Harvard strain) grown in a modified Mueller and Miller medium.
  • the toxoid is filter sterilized prior to the conjugation process.
  • the ActHIB® component does not contain a preservative. Potency of the ActHIB® component is specified on each lot by limits on the content of PRP polysaccharide and protein per dose and the proportion of polysaccharide and protein that is characterized as high molecular weight conjugate.
  • the present invention is directed in part to a method of preventing dengue disease (in particular virologically confirmable dengue, VCD) in a subject.
  • the invention is directed to a method of preventing dengue disease in a subject, comprising administering to the subject, a unit dose/tetravalent dengue virus composition, in particular a reconstituted unit dose of the invention as described herein.
  • the present invention is directed in part to a method of preventing dengue hemorrhagic fever (DHF) and dengue shock syndrome (DSS).
  • DHF dengue hemorrhagic fever
  • DSS dengue shock syndrome
  • the invention is directed to a method of preventing dengue hemorrhagic fever (DHF) and dengue shock syndrome (DSS), comprising administering to the subject a reconstituted unit dose/tetravalent dengue virus composition of the invention as described herein.
  • the present invention is therefore directed to a method of inoculating a subject against virologically confirmable dengue disease with a tetravalent dengue virus composition including four live attenuated dengue virus strains representing serotype 1, serotype 2, serotype 3 and serotype 4, wherein in particular the tetravalent dengue virus composition includes a chimeric dengue serotype 2/1 strain and a dengue serotype 2 strain and a chimeric dengue serotype 2/3 strain and a chimeric dengue serotype 2/4 strain, wherein in particular the dengue serotype 2 strain is derived from the wild type virus strain DEN-2 16681 (SEQ ID NO 11) and differs in at least three nucleotides from the wild type as follows:
  • NCR 5′-noncoding region
  • nt-57 C-to-T major attenuation locus
  • chimeric dengue strains are derived from the serotype 2 strain by replacing the structural proteins prM and E from serotype 2 strain with the corresponding structural proteins from the other dengue serotypes, resulting in the following chimeric dengue strains:
  • the tetravalent dengue virus composition for such a method may be in the form of a unit dose comprising:
  • a dengue serotype 1 in a concentration of at least 3.3 log 10 pfu/0.5 ml
  • a dengue serotype 2 in a concentration of at least 2.7 log 10 pfu/0.5 ml
  • a dengue serotype 3 in a concentration of at least 4.0 log 10 pfu/0.5 ml
  • a dengue serotype 4 in a concentration of at least 4.5 log 10 pfu/0.5 ml.
  • the present invention is in particular directed to such a method wherein the unit dose is lyophilized and upon reconstitution with 0.5 mL of a pharmaceutically acceptable diluent comprises:
  • a dengue serotype 1 in a concentration of at least 3.3 log 10 pfu/0.5 ml
  • a dengue serotype 2 in a concentration of at least 2.7 log 10 pfu/0.5 ml
  • a dengue serotype 3 in a concentration of at least 4.0 log 10 pfu/0.5 ml
  • a dengue serotype 4 in a concentration of at least 4.5 log 10 pfu/0.5 ml.
  • the present invention is also in particular directed to such a method wherein upon reconstitution with a pharmaceutically acceptable diluent (i), (ii), (iii), and (iv) provide a total concentration of pfu/0.5 mL and based on said total concentration the concentration of (ii) in pfu/0.5 mL is less than 10%, and the concentration of (iv) in pfu/0.5 mL is at least 50%, and the concentration of (i) in pfu/0.5 mL is at least 1%, and the concentration of (iii) in pfu/0.5 mL is at least 8%, or at least 10%, or at least 12%, or at least 14%, or at least 16%, or at least 18%, and wherein preferably the subject is 2 to 17 years of age or 4 to 16 years of age.
  • the present invention is also in particular directed to such a method wherein upon reconstitution with a pharmaceutically acceptable diluent (i), (ii), (iii), and (iv) provide a total concentration of pfu/0.5 mL and based on said total concentration the concentration of (ii) in pfu/0.5 mL is less than 2%, the concentration of (iv) in pfu/0.5 mL is at least 50%, the concentration of (i) in pfu/0.5 mL is at least 1%, and the concentration of (iii) in pfu/0.5 mL is at least 6%, wherein preferably the subject is 18 to 60 years of age.
  • tetravalent composition or the unit dose can be derived from section “Dengue vaccine composition” and “Unit dose” above.
  • the present invention is therefore directed to a method and corresponding use, the method comprising a primary vaccination with only two administrations of the unit dose comprising the steps of:
  • Such method preferably provides a combined vaccine efficacy against all four serotypes in preventing virologically confirmable dengue disease with a 2-sided 95% confidence interval, wherein the lower bound is more than 60%, when measured against placebo in a subject population of at least 5,000 healthy subjects irrespective of serostatus at baseline and 14 to 16 years of age, from the first administration of the administration schedule until 18 months after the second administration of the administration schedule.
  • Such method also preferably provides a combined vaccine efficacy against all four serotypes, in preventing virologically confirmable dengue disease with a 2-sided 95% confidence interval, wherein the lower bound is more than 45%, when measured against placebo in a subject population of at least 1,500 or at least 2,000 healthy subjects seronegative against all serotypes at baseline and 14 to 16 years of age, from 30 days after the second administration of the administration schedule until 18 months after the second administration of the administration schedule.
  • the method of inoculation against the virologically confirmable dengue disease is due to a dengue serotype 2, and/or due to a dengue serotype 1.
  • the method has very high efficacy against dengue serotype 2 and dengue serotype 1 and the highest efficacy against dengue serotype 2.
  • the invention is directed to said methods having a vaccine efficacy against serotype 1, in preventing virologically confirmable dengue disease with a 2-sided 95% confidence interval, wherein the lower bound is more than 25%, when measured against placebo in a subject population of at least 1,500, or at least 2,000, or at least 5,000 healthy subjects (or at least 10,000, or at least 15,000 healthy subjects) irrespective of serostatus at baseline and 4 to 16 years of age from 30 days post second administration until 12 to 18 months (e.g. at 12 or at 18 months) after the second administration.
  • the lower bound is more than 30%, is more than 35% is more than 40%, is more than 45%, is more than 50%, or is more than 54%.
  • the subject population of at least 1,500 is seronegative against all serotypes at base line and the lower bound is more than 35%.
  • the seronegative and seropositive population each provide a vaccine efficacy against serotype 1 with a 2-sided 95% confidence interval, wherein the lower bounds are within 10%-points.
  • the invention is directed to said methods having a vaccine efficacy against serotype 1, in preventing virologically confirmable dengue disease, when measured against placebo in a subject population of at least 1,500, or at least 2,000, or at least 5,000 healthy subjects (or at least 10,000, or at least 15,000 healthy subjects) irrespective of serostatus at baseline and 4 to 16 years of age from 30 days post second administration until 12 to 18 months (e.g. at 12 or at 18 months) after the second administration.
  • the vaccine efficacy is more than 40%, is more than 50%, is more than 60%, or is more than 65%.
  • the subject population of at least 1,500 is seronegative against all serotypes at base line.
  • the seronegative and seropositive population each provide a vaccine efficacy against serotype 1 which are within 5%-points.
  • the invention is directed to said methods having a vaccine efficacy against serotype 2, in preventing virologically confirmable dengue disease with a 2-sided 95% confidence interval, wherein the lower bound is more than 25%, when measured against placebo in a subject population of at least 1,500, or at least 2,000, or at least 5,000 healthy subjects (or at least 10,000, or at least 15,000 healthy subjects) irrespective of serostatus at baseline and 4 to 16 years of age from 30 days post second administration until 12 to 18 months (e.g. at 12 or at 18 months) after the second administration.
  • the lower bound is more than 50%, is more than 60%, is more than 70%, is more than 80%, or is more than 85%.
  • the subject population of at least 1,500 is seronegative against all serotypes.
  • the seronegative and seropositive population each provide a vaccine efficacy against serotype 2 with a 2-sided 95% confidence interval, wherein the lower bounds are within 5%-points.
  • the invention is directed to said methods having a vaccine efficacy against serotype 2, in preventing virologically confirmable dengue disease, when measured against placebo in a subject population of at least 1,500, or at least 2,000, or at least 5,000 healthy subjects (or at least 10,000, or at least 15,000 healthy subjects) irrespective of serostatus at baseline and 4 to 16 years of age from 30 days post second administration until 12 to 18 months (e.g. at 12 or at 18 months) after the second administration.
  • the vaccine efficacy is more than 60%, is more than 70%, is more than 80%, or is more than 90%.
  • the subject population of at least 1,500 is seronegative against all serotypes at base line.
  • the seronegative and seropositive population each provide a vaccine efficacy against serotype 2 which are within 5%-points.
  • the unit dose is reconstituted and administered by subcutaneous injection.
  • the subcutaneous injection is administered to the arm, preferably to the deltoid region of the arm.
  • such a method does not include a step of determination whether there was a previous dengue infection in the subject before administration of the unit dose or wherein the serostatus of the subject is unknown before administration of the unit dose.
  • such a method does not include a step of determination of a previous dengue infection in the subjects preferably at any time before, during or after the steps of administration or wherein the serostatus of the subject is unknown preferably at any time before, during or after the steps of administration.
  • the method according to the invention does not require the testing of the serostatus before vaccination and thus allows immediate treatment and outbreak control.
  • the use is for a method wherein the subject is exposed to a dengue outbreak.
  • the outbreak is due to a dengue serotype 2, and/or due to a serotype 1.
  • the subject is from a region wherein the seroprevalence rate is unknown and/or wherein the seroprevalence rate is below 80%, or below 70%, or below 60%.
  • the subject is seronegative at baseline and is from a region or travels to a region wherein the seroprevalence rate is high with respect to serotype 1 and/or serotype 2 i.e. 80%, or 90% or above.
  • the vaccine and corresponding method is safe for seronegative and seropositive subjects and thus does not require an analysis of the serostatus or a determination of a previous dengue infection or a high seroprevalence rate in the region.
  • Such a method preferably provides a combined vaccine efficacy against virologically-confirmed dengue with hospitalization against all four serotypes with a 2-sided 95% confidence interval, wherein the lower bound is more than 65%, when measured against placebo in a subject population of at least 5,000 healthy 4 to 16 year old subjects irrespective of serostatus at baseline, preferably in at least 1,500 healthy 4 to 16 year old subjects seronegative at baseline, from first administration of the administration schedule until 12 to 18 months after the second administration of the administration schedule.
  • the 2-sided 95% confidence interval of the combined vaccine efficacy against virologically-confirmed dengue with hospitalization against all four serotypes when comparing seropositive and seronegative subjects provides for lower bounds of the 2-sided confidence interval which are within 10% points or within 15% points or within 20% points.
  • the method is preferably safe with respect to serotype 1 and serotype 2 which may therefore be used in outbreak situations due to serotype 1 and/or serotype 2 or even for seronegative subjects (e.g. travelers) or subjects with unknown serostatus in regions with very high seroprevalence rates (>80%) due to serotype 1 and/or serotype 2.
  • such a method does not include the active surveillance with respect to febrile illness of the subject after the administration of the first- and second-unit dose.
  • active surveillance any subject with febrile illness (defined as fever 38° C. on any 2 of 3 consecutive days) will be asked to return to the site for dengue fever evaluation by the Investigator.
  • Subjects/guardians will be contacted at least weekly to ensure robust identification of febrile illness by reminding subjects/guardians of their obligation to return to the site in case of febrile illness. This contact will be implemented through appropriate methods that may differ in each trial site (eg, phone calls, text messaging, home visits, school-based surveillance).
  • such a method does not include vaccine immunogenicity analysis including GMTs for dengue neutralizing antibodies.
  • Such a method does not include a reactogenicity analysis.
  • a reactogenicity analysis relates to solicited local AEs (injection site pain, injection site erythema, and injection site swelling) and solicited systemic AEs (child ⁇ 6 years: fever, irritability/fussiness, drowsiness and loss of appetite; child 6 years: asthenia, fever, headache, malaise and myalgia) which will e.g. be assessed for 7 days and 14 days, respectively, following each vaccination (vaccination day included) via collection of diary cards.
  • the method does not include an active surveillance, an immunogenicity analysis and a reactogenicity analysis.
  • the vaccine and the corresponding method of inoculation are safe and therefore do not require further steps of surveillance or analysis.
  • A selecting a subject for administration of the unit doses of the tetravalent dengue virus composition in need for protection against dengue infection without determination of a previous dengue infection, and (B) administering a first unit dose of the tetravalent dengue virus composition to the subject, and (C) administering a second unit dose of the tetravalent dengue virus composition to the subject within 3 months of administration of the first unit dose. Therefore the method of inoculating is finalized without determination of a previous dengue infection.
  • the method further optionally comprises at least 1 years after the administration of the second unit dose a booster dose of the unit dose. Selecting the subject may include all types of considerations but preferably not the determination of a previous dengue infection.
  • the selection may include consideration of the age, health conditions, and threat of infection.
  • the threat of infection includes consideration of the seroprevalence rate in the region in which the subject normally lives or intends to travel, the serotype specific seroprevalence rate and an outbreak situation or serotype specific outbreak situations.
  • the subject may be selected due to its exposure to serotype 1 and/or serotype 2 or due to the fact it requires protection against a specific dengue serotype, i.e. serotype 1 and/or serotype 2.
  • the method is applicable to subjects of all kinds of ages.
  • the subject is under 9 years of age, or 4 to 5 years of age, or 6 to 11 years of age or 12 to 16 years, or 6 to 16 years of age or 4 to 16 years of age, or 2 to 17 years of age, or 9 years of age, or over 9 years of age, or 9 to 17 years of age, or 18 to 45 years of age, or 46 to 60 years of age, or over 60 years of age.
  • the present invention is directed to such a method wherein the method which is safe.
  • the present invention is directed to such a method providing a combined vaccine efficacy against virologically-confirmed dengue with hospitalization against all four serotypes with a 2-sided 95% confidence interval, wherein the lower bound is more than 65%, when measured against placebo in a subject population of at least 5,000 healthy 4 to 16 year old subjects irrespective of serostatus at baseline from first administration of the administration schedule until 12 to 18 months after the last administration of the administration schedule.
  • the present invention is directed to such a method wherein the method which is effective.
  • the present invention is directed to such a method providing a combined vaccine efficacy against all four serotypes, in preventing virologically confirmable dengue disease with a 2-sided 95% confidence interval, wherein the lower bound is more than 60%, when measured against placebo in a subject population of at least 5,000 healthy subjects irrespective of serostatus at baseline and 14 to 16 years of age, from the first administration of the administration schedule until 18 months after the last administration of the administration schedule.
  • the invention is directed to said methods, wherein the subject is seronegative to all dengue serotypes.
  • the present invention is directed in part to a method of preventing dengue disease (in particular virologically confirmable dengue, VCD) in a subject population.
  • the invention is directed to a method of preventing dengue disease in a subject population, comprising administering to the subject population a unit dose, in particular a reconstituted unit dose of the invention as described herein.
  • the present invention is in part directed to said method for preventing dengue disease (in particular virologically confirmable dengue, VCD) in a subject population comprising administering to the subject population at least a first reconstituted unit dose of the invention as described herein, wherein certain ratios of geometric mean neutralizing antibody titers (GMTs) at day 180 or 365 after administration of said first unit dose to the subject population are achieved.
  • dengue disease in particular virologically confirmable dengue, VCD
  • VCD virologically confirmable dengue
  • the geometric mean neutralizing antibody titer for dengue serotype 2 (GMT DENV-2) and the geometric mean neutralizing antibody titer for dengue serotype 4 (GMT DENV-4) when tested in at least 40, or at least 50, or at least 60 subjects at day 180 or day 365 after at least a first administration of said reconstituted unit dose of the invention as described herein, and optionally a second administration of a reconstituted unit dose of the invention as described herein 90 days after said first administration, provide a ratio of GMT DENV-2:GMT DENV-4 of not more than 50, or not more than 40, or not more than 30, or not more than 20.
  • the ratio of GMT DENV-2:GMT DENV-1 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose
  • the ratio of GMT DENV-2:GMT DENV-3 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose.
  • the present invention is in part directed to said method for preventing dengue disease (in particular virologically confirmable dengue, VCD) in a subject comprising administering to the subject at least a first reconstituted unit dose of the invention as described herein, wherein certain ratios of neutralizing antibody titers at day 180 or 365 after administration of said first unit dose to the subject are achieved.
  • dengue disease in particular virologically confirmable dengue, VCD
  • the neutralizing antibody titer for dengue serotype 2 and the neutralizing antibody titer for dengue serotype 4 at day 180 or day 365 after at least a first administration of the reconstituted unit dose of the invention as described herein, and optionally a second administration of a reconstituted unit dose of the invention as described herein 90 days after said first administration provide a ratio of neutralizing antibody titer for DENV-2:neutralizing antibody titer for GMT DENV-4 of not more than 50, or not more than 40, or not more than 30, or not more than 20.
  • the ratio of the neutralizing antibody titers of DENV-2:DENV-1 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose, and/or the ratio of the neutralizing antibody titers of DENV-2:DENV-3 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose.
  • the geometric mean neutralizing antibody titers (GMTs) of a subject population or the neutralizing antibody titers of a subject are determined in accordance with the microneutralization test disclosed herein, for example according to the method described in Example 2.
  • a method inducing a more balanced immune response due to the administration of the reconstituted unit dose of the invention as described herein in terms of less differences between the geometric mean neutralizing antibody titers (GMTs) against the four dengue serotypes or the neutralizing antibody titers against the four dengue serotypes, is beneficial to the subject or subject population to be vaccinated.
  • a much greater response to any one of the four serotypes, such as to DENV-2 in comparison to the other serotypes is less beneficial.
  • the present invention is in part directed to said method for preventing dengue disease (in particular virologically confirmable dengue, VCD) in a subject or subject population wherein the method provides a seropositivity rate in a subject population of at least 50 subjects including the administration of two unit doses subcutaneously at day 1 and at day 90, wherein the subjects of the subject population are seronegative to all dengue serotypes at baseline.
  • dengue disease in particular virologically confirmable dengue, VCD
  • VCD virologically confirmable dengue
  • At least 80% of the subject population are seropositive for all four dengue serotypes at least one month after administration of the first unit dose, such as at day 30, and/or at least 80% of the subject population are seropositive for all four dengue serotypes before or at the time of the administration of the second unit dose, such as at day 90, and/or at least 80%, or at least 85%, or at least 90%, or at least 95% of the subject population are seropositive for all four dengue serotypes after the administration of the second unit dose, such as at day 120, and/or at least 80%, or at least 85%, or at least 90% of the subject population are seropositive for all four dengue serotypes after the administration of the second unit dose, such as at day 270.
  • the present invention is in part directed to said method for preventing dengue disease (in particular virologically confirmable dengue, VCD) in a subject or subject population wherein the method provides a seropositivity rate in a subject population of at least 100 subjects including administration of two unit doses subcutaneously at day 1 and at day 90, wherein the subjects of the subject population comprises from 20% to 40% subjects who are seronegative to all dengue serotypes and from 60% to 80% subjects who are seropositive to at least one dengue serotype at base line, wherein at day 120 and/or day 270 the seropositivity rate for all four dengue serotypes in the seronegative part of the subject population and the seropositivity rate for all four dengue serotypes in the seropositive part of the subject population do not deviate more than 10%-points and/or wherein at day 120 the seropositivity rate for all four dengue serotypes in the seronegative part of the subject population and the seropositivity rate for all four dengue ser
  • the present invention is in part directed to a method of preventing virologically confirmable dengue disease in a subject or subject population comprising administering to the subject or subject population a reconstituted unit dose of a tetravalent dengue virus composition including four live, attenuated dengue serotypes, in particular the virus strains as described herein.
  • the present invention is in part directed to a method of preventing virologically confirmable dengue disease with hospitalization in a subject or subject population comprising administering to the subject or subject population a reconstituted unit dose of a tetravalent dengue virus composition including four live, attenuated dengue serotypes, in particular the virus strains as described herein.
  • the method includes a reconstituted unit dose/tetravalent dengue virus composition of a dengue vaccine composition administered for preventing dengue disease in a subject or a subject population, the reconstituted unit dose comprising: a tetravalent virus composition including four live attenuated dengue virus strains, wherein a unit dose is lyophilized and upon reconstitution with 0.5 mL of a pharmaceutically acceptable diluent the reconstituted unit dose is obtained which comprises:
  • a dengue serotype 1 such as a chimeric dengue serotype 2/1 strain, in a concentration of at least 3.3 log 10 pfu/0.5 ml
  • a dengue serotype 2 such as a dengue serotype 2 strain, in a concentration of at least 2.7 log 10 pfu/0.5 ml
  • a dengue serotype 3 such as a chimeric dengue serotype 2/3 strain, in a concentration of at least 4.0 log 10 pfu/0.5 ml
  • a dengue serotype 4 such as a chimeric dengue serotype 2/4 strain, in a concentration of at least 4.5 log 10 pfu/0.5 ml.
  • the reconstituted unit dose/tetravalent dengue virus composition is used in the method of preventing dengue disease of the present invention, wherein upon reconstitution of the unit dose with a pharmaceutically acceptable diluent (i), (ii), (iii), and (iv) provide a total concentration of pfu/0.5 mL and based on said total concentration the concentration of (ii) in pfu/0.5 mL is less than 2%, the concentration of (iv) in pfu/0.5 mL is at least 50%, the concentration of (i) in pfu/0.5 mL is at least 1%, and the concentration of (iii) in pfu/0.5 mL is at least 6% and wherein the subject or subject population is of 18 to 60 years of age.
  • the reconstituted unit dose/tetravalent dengue virus composition is used in the method of preventing dengue disease of the present invention, wherein upon reconstitution with a pharmaceutically acceptable diluent (i), (ii), (iii), and (iv) provide a total concentration of pfu/0.5 mL and based on said total concentration the concentration of (ii) in pfu/0.5 mL is less than 10%, and the concentration of (iv) in pfu/0.5 mL is at least 50%, and the concentration of (i) in pfu/0.5 mL is at least 1%, and the concentration of (iii) in pfu/0.5 mL is at least 8% and wherein the subject or subject population is of 2 to 17 years of age.
  • the invention is directed to said methods, wherein said unit dose comprises a tetravalent dengue virus composition including four live attenuated dengue serotypes, in particular the virus strains described herein wherein the serotypes have certain concentrations as described herein with respect to the virus composition and unit dose such as:
  • the invention is directed to said methods, wherein said unit dose upon reconstitution with 0.5 mL of a pharmaceutically acceptable diluent has a concentration of 3.3 log 10 pfu/0.5 mL to 3.6 log 10 pfu/0.5 mL for dengue serotype 1 (e.g. chimeric dengue serotype 2/1 strain), has a concentration of 2.7 log 10 pfu/0.5 mL to 4.0 log 10 pfu/0.5 mL for dengue serotype 2 (e.g. dengue serotype 2 strain), has a concentration of 4.0 log 10 pfu/0.5 mL to 4.6 log 10 pfu/0.5 mL for dengue serotype 3 (e.g.
  • the subject or subject population is of 2 to 17 years of age, such as 4 to 16 years of age, and preferably less than 9 years of age. In other preferred embodiments, the subject or subject population is 4-5 years of age, 6-11 years of age or 12-16 years of age.
  • the invention is directed to said methods, wherein the concentration of the dengue serotype 1 (e.g. chimeric dengue serotype 2/1 strain) measured in pfu/0.5 mL is 1% to 7% of the total concentration, the concentration of the dengue serotype 2 (e.g. dengue serotype 2 strain) measured in pfu/0.5 mL is less than 8% of the total concentration, such as in the range of 1% to 8% of the total concentration, the concentration of the dengue serotype 3 (e.g. chimeric dengue serotype 2/3 strain) measured in pfu/0.5 mL is at least 10% of the total concentration, and the concentration of the dengue serotype 4 (e.g.
  • chimeric dengue serotype 2/4 strain measured in pfu/0.5 mL is at least 65% of the total concentration, such as in the range of 65% to 80%.
  • the arithmetic sum of all four serotypes is in the range of 4.6 log 10 pfu/0.5 mL to 6.7 log 10 pfu/0.5 mL, preferably in the range of 4.6 log 10 pfu/0.5 mL to 5.5 log 10 pfu/0.5 mL
  • the subject or subject population is of 2 to 17 years of age, such as 4 to 16 years of age, and even more preferably less than 9 years of age.
  • the subject or subject population is 4-5 years of age, 6-11 years of age or 12-16 years of age.
  • the invention is directed to said methods, wherein the dengue serotype 1 (e.g. chimeric dengue serotype 2/1 strain) such as TDV-1 and the dengue serotype 2 (e.g. dengue serotype 2 strain) such as TDV-2 are present each in a concentration based on the total concentration in pfu/0.5 mL which is within 5%-points of each other and/or are together less than about 10% of the total concentration in pfu/0.5 mL.
  • the dengue serotype 3 e.g.
  • the dengue serotype 4 e.g. chimeric dengue serotype 2/4 strain
  • the dengue serotype 4 represents the highest concentration in the composition of all four serotypes, preferably with at least about 70% of the total concentration in pfu/0.5 mL, dengue serotype 3 (e.g.
  • chimeric dengue serotype 2/3 strain such as TDV-3 represents the second highest concentration in the composition of all four serotypes, preferably with at least about 10% of the total concentration in pfu/0.5 mL
  • dengue serotype 1 e.g. chimeric dengue serotype 2/1 strain
  • dengue serotype 2 e.g. dengue serotype 2 strain
  • TDV-2 each represent lower concentrations than the concentration of serotype 3 (e.g. chimeric dengue serotype 2/3 strain) such as TDV-3, and optionally together represent less than about 10% of the total concentration in pfu/0.5 mL.
  • the chimeric dengue serotype 2/1 strain is TDV-1
  • the dengue serotype 2 strain is TDV-2
  • the chimeric dengue serotype 2/3 strain is TDV-3
  • the chimeric dengue serotype 2/4 strain is TDV-4.
  • TDV-1 is characterized by the nucleotide sequence according to SEQ ID No. 1 and the amino acid sequence according to SEQ ID No. 2
  • TDV-2 is characterized by the nucleotide sequence according to SEQ ID No. 3 and the amino acid sequence according to SEQ ID No. 4
  • TDV-3 is characterized by the nucleotide sequence according to SEQ ID No. 5
  • TDV-4 is characterized by the nucleotide sequence according to SEQ ID No. 7 and the amino acid sequence according to SEQ ID No. 8.
  • the invention is directed to said methods, wherein the reconstituted unit dose of the invention as described herein is administered by subcutaneous injection.
  • the subcutaneous injection is administered to the arm, preferably to the deltoid region of the arm.
  • the invention is directed to said methods, wherein the reconstituted unit dose is administered to a subject of unknown serostatus and/or wherein no test has been carried out to determine whether the subject is seropositive or seronegative (before) the unit dose as described herein is administered.
  • the invention is directed to said methods which do not include a step of determination of a previous dengue infection in the subject or subjects.
  • the invention is directed to said methods which do not include the analysis of the seroprevalence rate in the region or is conducted in a region with a seroprevalence of below 80%, below 70% or below 60%.
  • the invention is directed to a method wherein the serostatus of the subject is unknown. In such embodiments the serostatus is not determined at any time before and after administration in relation to this method.
  • the method is used in an outbreak situation.
  • the invention is directed to said methods being conducted outside a clinical trial
  • the invention is directed to said methods, wherein the subject, or subject population is seronegative to all dengue serotypes.
  • the invention is directed to said methods, wherein two unit doses of the invention as described herein are administered.
  • the two unit doses are administered within 12 months or more, or within six months, or within three months, and optionally at least 4 weeks apart such as at day 0 and day 90 or at day 1 and day 90.
  • a further third unit dose of the invention as described herein is administered after the second administration.
  • Such a third administration may act as a booster and may be administered between 6 to 12 months after the first administration, such as 12 months after the first administration, or later than 12 month after the first administration, such as 12 months (1 year) after the second administration or even 5 years or longer after the first or second administration.
  • the method of the invention comprises or consists of a single unit dose of the invention being administered.
  • the invention is directed to said methods, wherein the reconstituted unit dose of the invention as described herein is administered subcutaneously to a subject or subject population that is seronegative with respect to all dengue serotypes.
  • the subject or subject population is seropositive with respect to at least one dengue serotype.
  • the invention is directed to said methods, wherein the unit dose of the invention as described herein is administered to a subject or subject population from a dengue endemic region.
  • the subject or subject population is from Singapore, Dominican Republic, Panama, Philippines, Colombia, Puerto Rico or Thailand, in particular from Singapore, Dominican Republic, Panama, or Philippines.
  • the subject or subject population is from Asia Pacific or from Latin America.
  • the subject or subject population is from Thailand, Sri Lanka, Philippines, Panama, Portugal, Portugal, Portugal, Portugal, Portugal, Portugal, Portugal, Portugal, Portugal.
  • the subject, or subject population is from a dengue non-endemic region.
  • Such a subject population or such a subject may be vaccinated according to the present invention in the context of traveling to a dengue endemic region.
  • the reconstituted unit dose of the invention as described herein is administered subcutaneously to a subject, or subject population that is from a dengue endemic region or a dengue non-endemic region.
  • the invention is directed to said methods, wherein the reconstituted unit dose of the invention as described herein is administered subcutaneously to a subject or subject population of 2 to 60 years of age.
  • the subjects or subject population are adults of more than 17 years, or more than 18 years, or 18 to 60 years.
  • the subjects or subject population are adults of more than 21 years, or 21 to 60 years, or 21 to 45 years of age.
  • the invention is directed to said methods, wherein the reconstituted unit dose of the invention as described herein is administered subcutaneously to children and adolescents of 2 to 17 years of age.
  • the subjects or subject population are less than 9 years of age, or less than 4 years of age.
  • the subjects or subject population are from 2 to 9 years of age, or from 2 to 5 years of age, or from 4 to 9 years of age or from 6 to 9 years of age.
  • the subject or subject population is 4 to 16 years of age.
  • the subject or subject population is 4-5 years of age, 6-11 years of age or 12-16 years of age.
  • the subject or subject population is seronegative with respect to all dengue serotypes.
  • the invention is directed to said methods, wherein the unit dose of the invention as described herein is administered to a pediatric subject or pediatric subject population of less than 2 years of age, preferably of 2 months to 2 years or 2 months to 1.5 years or 2 months to 1 year.
  • the pediatric subject or pediatric subject population is seronegative and from a dengue endemic region.
  • the invention is directed to said methods, wherein the reconstituted unit dose of the invention as described herein is administered to a pediatric subject or pediatric subject population of less than 2 years of age, preferably of 2 months to 2 years or 2 months to 1.5 years or 2 months to 1 year, preferably by subcutaneous injection.
  • the pediatric subject or pediatric subject population is seronegative and from a dengue endemic region.
  • the invention is directed to said methods, wherein the subject or subject population is 4-5 years of age and from Asia Pacific, 6-11 years of age and from Asia Pacific, or 12-16 years of age and from Asia Pacific. In other embodiments, the subject or subject population is 4-5 years of age and from Latin America, 6-11 years of age and from Latin America, or 12-16 years of age and from Latin America.
  • the invention is directed to said methods, wherein the subject or subject population is 4-5 years of age and seropositive for at least 1 dengue serotype, 6-11 years of age and seropositive for at least 1 dengue serotype, or 12-16 years of age and seropositive for at least 1 dengue serotype.
  • the subject or subject population is 4-5 years of age and seronegative for all dengue serotypes, 6-11 years of age and seronegative for all dengue serotypes, or 12-16 years of age and seronegative for all dengue serotypes.
  • the invention is directed to said methods, wherein the subject or subject population is from Asia Pacific or Latin America and seropositive for at least one dengue serotype at baseline. In other embodiments, the subject or subject population is from Asia Pacific or Latin America and seronegative for at all dengue serotype at baseline.
  • the invention is directed to said methods, wherein the subject or subject population is from Asia Pacific, seropositive for at least one dengue serotype at baseline and 4-5 years of age, 6-11 years of age, or 12-16 years of age.
  • the subject or subject population is from Asia Pacific, seronegative for all dengue serotypes at baseline and 4-5 years of age, 6-11 years of age, or 12-16 years of age.
  • the subject or subject population is from Latin America, seropositive for at least one dengue serotype at baseline and 4-5 years of age, 6-11 years of age, or 12-16 years of age.
  • the subject or subject population is from America, seronegative for all dengue serotypes at baseline and 4-5 years of age, 6-11 years of age, or 12-16 years of age.
  • the invention is directed to said methods, wherein the subject or subject population had prior vaccination against Yellow Fever. In other embodiments, the subject or subject population had prior vaccination against Japanese Encephalitis. In yet other embodiments, the subject or subject population had no prior vaccination against Yellow Fever. In other embodiments, the subject or subject population had no prior vaccination against Japanese Encephalitis.
  • Prior vaccination indicates a vaccination prior to 30 days after a second administration, such as within 4 months after the first administration, with the reconstituted unit dose as described herein. For example for vaccine efficacy (VE) as determined in Example 6 from 30 days post-second vaccination, a prior vaccination of Yellow Fever is defined as a Yellow Fever vaccination occurring before 30 days post-second vaccination.
  • the subject or subject population received Dengvaxia® within the administration regimen as described herein or within 4.5 years after administration of the first dose.
  • the present invention is directed in part to a method of preventing virologically confirmable dengue disease in a subject comprising administering to the subject a tetravalent dengue virus composition including four dengue virus strains representing serotype 1, serotype 2, serotype 3 and serotype 4, wherein the virus strains are optionally live, attenuated dengue virus strains.
  • the present invention is directed in part to a method of preventing virologically confirmable dengue disease in a subject consisting of administering to the subject a tetravalent dengue virus composition including four dengue virus strains representing serotype 1, serotype 2, serotype 3 and serotype 4, wherein the virus strains are optionally live, attenuated dengue virus strains.
  • the invention is directed to said methods, wherein there is no step of determining the serostatus of the subject at baseline, in other words, said methods do not comprise a determination of a previous dengue infection of the subject at baseline before the administration of the tetravalent dengue virus composition.
  • said methods are safe and effective.
  • the subject has not been tested for the presence a previous dengue infection.
  • the invention is directed to said methods, wherein the vaccine administration is safe irrespective of whether there is a determination that the subject had a previous dengue infection before the administration of the tetravalent dengue virus composition.
  • the vaccine administration is safe irrespective of whether there is a determination that the subject had a previous dengue infection before the administration of the tetravalent dengue virus composition.
  • such methods are also effective.
  • the invention is directed to said methods, wherein the method is safe and/or effective.
  • the invention is directed to said methods, wherein the composition includes at least one chimeric dengue virus.
  • the invention is directed to said methods, wherein the composition includes at least one non-chimeric dengue virus and at least one chimeric dengue virus, in particular a chimeric dengue serotype 2/1 strain and a dengue serotype 2 strain and a chimeric dengue serotype 2/3 strain and a chimeric dengue serotype 2/4 strain.
  • the composition includes at least one non-chimeric dengue virus and at least one chimeric dengue virus, in particular a chimeric dengue serotype 2/1 strain and a dengue serotype 2 strain and a chimeric dengue serotype 2/3 strain and a chimeric dengue serotype 2/4 strain.
  • the invention is directed to said methods having a vaccine efficacy, preferably a combined vaccine efficacy against all four serotypes, in preventing virologically confirmable dengue disease with a 2-sided 95% confidence interval, wherein the lower bound is more than 25%, when measured against placebo in a subject population of at least 5,000 healthy subjects (or at least 10,000, or at least 15,000 healthy subjects) irrespective of serostatus at baseline and e.g. 14 to 16 years of age, wherein a reconstituted unit dose/tetravalent dengue virus composition as described herein or placebo is administered e.g.
  • the invention is directed to said methods having a vaccine efficacy, preferably a combined vaccine efficacy against all four serotypes, in preventing virologically confirmable dengue disease with a 2-sided 95% confidence interval, wherein the lower bound is more than 25%, when measured against placebo in a subject population of at least 5,000 healthy subjects (or at least 10,000, or at least 15,000 healthy subjects) irrespective of serostatus at baseline, wherein a reconstituted unit dose or tetravalent dengue virus composition as described herein or placebo is administered at least once, until 15 to 21 months (e.g.
  • the lower bound is more than 30%, more than 40%, more than 50%, more than 55%, more than 60%, more than 65%, more than 70% or more than 72%.
  • said reconstituted unit dose or placebo is administered subcutaneously within about 3 months, such as on days 0 and 90.
  • the invention is directed to said methods having a combined vaccine efficacy against all four serotypes, in preventing virologically confirmable dengue disease with a 2-sided 95% confidence interval, wherein the lower bound is more than 60%, when measured against placebo in a subject population of at least 5,000 healthy subjects (or at least 10,000, or at least 15,000 healthy subjects) irrespective of serostatus at baseline and 4 to 16 years of age, wherein a reconstituted unit dose/tetravalent dengue virus composition as described herein or placebo is administered e.g. at least twice within less than 6 months, such as within 3 months, after the first administration until 18 months after the last administration.
  • the lower bound is e.g. more than 62%, more than 64%, more than 66%, more than 68%, or more than 69%.
  • the invention is directed to said methods having a vaccine efficacy, preferably a combined vaccine efficacy against all four serotypes, in preventing virologically confirmable dengue disease of more than 30%, when measured against placebo in a subject population of at least 5,000 healthy subjects (or at least 10,000, or at least 15,000 healthy subjects) irrespective of serostatus at baseline and e.g. 14 to 16 years of age, wherein a reconstituted unit dose or tetravalent dengue virus composition as described herein or placebo is administered at least twice within less than 6 months, such as within 3 months, after first administration or 30 days after the second administration/last administration until at least 12 months or until 12 to 18 months (e.g. at 12 or at 18 months) after the second administration/last administration.
  • a vaccine efficacy preferably a combined vaccine efficacy against all four serotypes, in preventing virologically confirmable dengue disease of more than 30%, when measured against placebo in a subject population of at least 5,000 healthy subjects (or at least 10,000, or at least
  • the invention is directed to said methods having a vaccine efficacy, preferably a combined vaccine efficacy against all four serotypes, in preventing virologically confirmable dengue disease of more than 30%, when measured against placebo in a subject population of at least 5,000 healthy subjects (or at least 10,000, or at least 15,000 healthy subjects) irrespective of serostatus at baseline, wherein a reconstituted unit dose or tetravalent dengue virus composition as described herein or placebo is administered at least once, until 15 months after the first administration of the administration schedule.
  • the vaccine efficacy is more than 40%, more than 50%, more than 55%, more than 60%, more than 65%, more than 70%, more than 75%, more than 78%, more than 79% or about 80%.
  • said reconstituted unit dose or placebo is administered subcutaneously within about 3 month, such as on days 0 and 90.
  • the invention is directed to said methods having a combined vaccine efficacy against all four serotypes, in preventing virologically confirmable dengue disease of more than 66%, when measured against placebo in a subject population of at least 5,000 healthy subjects (or at least 10,000, or at least 15,000 healthy subjects) irrespective of serostatus at baseline and 14 to 16 years of age, wherein a reconstituted unit dose/tetravalent dengue virus composition as described herein or placebo is administered e.g. at least twice within less than 6 months, such as within 3 months, after the first administration until 18 months after the last administration.
  • the vaccine efficacy is e.g. more than 68%, more than 70%, more than 72%, or more than 74%.
  • the invention is directed to said methods having a vaccine efficacy, preferably a combined vaccine efficacy against all four serotypes, in preventing virologically confirmable dengue disease with hospitalization with a 2-sided 95% confidence interval, wherein the lower bound is more than 0%, when measured against placebo in a subject population of at least 5,000 healthy subjects (or at least 10,000, or at least 15,000 healthy subjects) irrespective of serostatus at baseline, wherein a reconstituted unit dose or tetravalent dengue virus composition as described herein or placebo is administered at least twice within less than 6 months, such as within 3 months, 30 days after the second administration until at least 18 months after the second administration.
  • the lower bound is more than 10%, is more than 20%, is more than 30%, is more than 40%, is more than 50%, is more than 55%, is more than 60%, is more than 65%, is more than 70% or is more than 80%, or more than 90%.
  • the invention is directed to said methods having a combined vaccine efficacy against all four dengue serotypes in seronegative subjects with a 2-sided 95% confidence interval, wherein the lower bound is more than 25%, when measured against placebo in a subject population of at least 1,500 or at least 2,000 healthy subjects being seronegative against all serotypes at baseline, wherein said unit dose/tetravalent dengue virus composition or said placebo is administered at least twice within less than 6 months, such as within 3 months, about 30 days after the second administration of the administration schedule until at least 12 months or until 12 to 18 months (e.g. at 12 or at 18 months) after the second administration of the administration schedule.
  • the lower bound is more than 30%, is more than 40%, is more than 50%, or is more than 55%.
  • the invention is directed to said methods having a combined vaccine efficacy against all four serotypes, in preventing virologically confirmable dengue disease with a 2-sided 95% confidence interval, wherein the lower bound is more than 25%, when measured against placebo in a subject population of at least 1,500 or at least 2,000 or at least 5,000 healthy subjects (or at least 10,000, or at least 15,000 healthy subjects) being seronegative against all serotypes at baseline and 4 to 16 years of age, wherein a reconstituted unit dose/tetravalent dengue virus composition as described herein or placebo is e.g. administered at least twice within less than 6 months, such as within 3 months, from 30 days post last administration until 12 to 18 months (e.g. at 12 months or at 18 months) after the last administration.
  • the lower bound is more than 30%, is more than 35%, is more than 40%, or is more than 45%.
  • the invention is directed to said methods having a combined vaccine efficacy against all four serotypes, in preventing virologically confirmable dengue disease with a 2-sided 95% confidence interval, wherein the lower bound is more than 25%, when measured against placebo in a subject population of at least 5,000 healthy subjects (or at least 10,000, or at least 15,000 healthy subjects) being seropositive at baseline and 4 to 16 years of age, wherein a reconstituted unit dose/tetravalent dengue virus composition as described herein or placebo is e.g. administered at least twice within less than 6 months, such as within 3 months, from 30 days post last administration until 12 to 18 months (e.g. at 12 months or at 18 months) after the last administration.
  • the lower bound is more than 40%, is more than 45%, is more than 50%, is more than 60%, or is more than 65%.
  • the invention is directed to said methods having a combined vaccine efficacy against all four serotypes, in preventing virologically confirmable dengue disease with a 2-sided 95% confidence interval, wherein the lower bound is more than 25%, when measured against placebo in a subject population of at least 5,000 healthy subjects (or at least 10,000, or at least 15,000 healthy subjects) being seropositive at baseline being or seronegative against all serotypes at baseline and 4 to 16 years of age, wherein a reconstituted unit dose/tetravalent dengue virus composition as described herein or placebo is e.g. administered at least twice within less than 6 months, such as within 3 months, from 30 days post last administration until 12 to 18 months (e.g. at 12 months or at 18 months) after the last administration.
  • the difference between the lower bound provided by the seropositive subjects at baseline and the subjects seronegative against all serotypes at baseline is no more than 15%-points.
  • the invention is directed to said methods having a combined vaccine efficacy against all four dengue serotypes in seronegative subjects of more than 30%, when measured against placebo in a subject population of at least 1,500 or at least 2,000 healthy subjects being seronegative against all serotypes at baseline, wherein said unit dose/tetravalent dengue virus composition or said placebo is administered at least twice within less than 6 months, such as within 3 months, 30 days after the second administration until at least 12 months or until 12 to 18 months (e.g. at 12 or at 18 months) after the second administration.
  • the combined vaccine efficacy against all four dengue serotypes in seronegative subjects is more than 40%, is more than 50%, is more than 60%, is more than 65%, or is more than 70%.
  • the invention is directed to said methods having a combined vaccine efficacy against all four serotypes, in preventing virologically confirmable dengue disease, when measured against placebo in a subject population of at least 1,500 or at least 2,000 or at least 5,000 healthy subjects (or at least 10,000, or at least 15,000 healthy subjects) being seronegative against all serotypes at baseline and 4 to 16 years of age, wherein a reconstituted unit dose/tetravalent dengue virus composition as described herein or placebo is e.g. administered at least twice within less than 6 months, such as within 3 months, from 30 days post last administration until 12 to 18 months (e.g. at 12 months or at 18 months) after the last administration.
  • the said vaccine efficacy is more than 30%, is more than 40%, is more than 50%, is more than 55%, is more than 60%, or is more than 65%.
  • the invention is directed to said methods having a combined vaccine efficacy against all four serotypes, in preventing virologically confirmable dengue disease, when measured against placebo in a subject population of at least 5,000 healthy subjects (or at least 10,000, or at least 15,000 healthy subjects) being seropositive at baseline and 4 to 16 years of age, wherein a reconstituted unit dose/tetravalent dengue virus composition as described herein or placebo is e.g. administered at least twice within less than 6 months, such as within 3 months, from 30 days post last administration until 12 to 18 months (e.g. at 12 months or at 18 months) after the last administration.
  • the said vaccine efficacy is more than 40%, is more than 50%, is more than 60%, is more than 65%, is more than 70%, or is more than 75%.
  • the invention is directed to said methods having a combined vaccine efficacy against all four serotypes, in preventing virologically confirmable dengue disease with a 2-sided 95% confidence interval, wherein the lower bound is more than 25%, when measured against placebo in a subject population of at least 5,000 healthy subjects (or at least 10,000, or at least 15,000 healthy subjects) being seropositive at baseline being or seronegative against all serotypes at baseline and 4 to 16 years of age, wherein a reconstituted unit dose/tetravalent dengue virus composition as described herein or placebo is e.g. administered at least twice within less than 6 months, such as within 3 months, from 30 days post last administration until 12 to 18 months (e.g. at 12 months or at 18 months) after the last administration.
  • the difference between the lower bound provided by the seropositive subjects at baseline and the subjects seronegative against all serotypes at baseline is no more than 15%-points, or is no more than 10%-points.
  • the invention is directed to said methods having a combined vaccine efficacy against all four dengue serotypes with a 2-sided 95% confidence interval, wherein the lower bound is more than 25%, when measured against placebo in a subject population of at least 1,000 healthy subjects 4 to 5 years of age at the time of randomization and irrespective of serostatus at baseline, wherein said unit dose/tetravalent dengue virus composition or said placebo is administered at least twice within less than 6 months, such as within 3 months, about 30 days after the second administration of the administration schedule until at least 12 months or until 12 to 18 months (e.g. at 12 or at 18 months) after the second administration of the administration schedule.
  • the lower bound is more than 30%, is more than 40%, is more than 45%.
  • the invention is directed to said methods having a combined vaccine efficacy against all four dengue serotypes of more than 30%, when measured against placebo in a subject population of at least 1,000 healthy subjects 4 to 5 years of age at the time of randomization and irrespective of serostatus at baseline, wherein said unit dose/tetravalent dengue virus composition or said placebo is administered at least twice within less than 6 months, such as within 3 months, 30 days after the second administration until at least 12 months or until 12 to 18 months (e.g. at 12 or at 18 months) after the second administration.
  • the combined vaccine efficacy against all four dengue serotypes is more than 40%, is more than 50%, is more than 60%, is more than 65%, or is more than 70%.
  • the invention is directed to said methods having a combined vaccine efficacy against all four dengue serotypes with a 2-sided 95% confidence interval, wherein the lower bound is more than 25%, when measured against placebo in a subject population of at least 1,000 healthy subjects 6 to 11 years of age at the time of randomization and irrespective of serostatus at baseline, wherein said unit dose/tetravalent dengue virus composition or said placebo is administered at least twice within less than 6 months, such as within 3 months, about 30 days after the second administration of the administration schedule until at least 12 months or until 12 to 18 months (e.g. at 12 or at 18 months) after the second administration of the administration schedule.
  • the lower bound is more than 30%, is more than 40%, is more than 50%, is more than 60%, or is more than 70%.
  • the invention is directed to said methods having a combined vaccine efficacy against all four dengue serotypes of more than 30%, when measured against placebo in a subject population of at least 1,000 healthy subjects 6 to 11 years of age at the time of randomization and irrespective of serostatus at baseline, wherein said unit dose/tetravalent dengue virus composition or said placebo is administered at least twice within less than 6 months, such as within 3 months, 30 days after the second administration until at least 12 months or until 12 to 18 months (e.g. at 12 or at 18 months) after the second administration.
  • the combined vaccine efficacy against all four dengue serotypes is more than 40%, is more than 50%, is more than 60%, is more than 70%, is more than 75%, or is more than 80%.
  • the invention is directed to said methods having a combined vaccine efficacy against all four dengue serotypes with a 2-sided 95% confidence interval, wherein the lower bound is more than 25%, when measured against placebo in a subject population of at least 1,000 healthy subjects 12 to 16 years of age at the time of randomization and irrespective of serostatus at baseline, wherein said unit dose/tetravalent dengue virus composition or said placebo is administered at least twice within less than 6 months, such as within 3 months, about 30 days after the second administration of the administration schedule until at least 12 months or until 12 to 18 months (e.g. at 12 or at 18 months) after the second administration of the administration schedule.
  • the lower bound is more than 30%, is more than 40%, is more than 50%, is more than 60%, is more than 65%, or is more than 68%.
  • the invention is directed to said methods having a combined vaccine efficacy against all four dengue serotypes of more than 30%, when measured against placebo in a subject population of at least 1,000 healthy subjects 12 to 16 years of age at the time of randomization and irrespective of serostatus at baseline, wherein said unit dose/tetravalent dengue virus composition or said placebo is administered at least twice within less than 6 months, such as within 3 months, 30 days after the second administration until at least 12 months or until 12 to 18 months (e.g. at 12 or at 18 months) after the second administration.
  • the combined vaccine efficacy against all four dengue serotypes is more than 40%, is more than 50%, is more than 60%, is more than 70%, is more than 75%, or is more than 80%.
  • the invention is directed to said methods having a vaccine efficacy against dengue serotype 1 with a 2-sided 95% confidence interval, wherein the lower bound is more than 25%, when measured against placebo in a subject population of at least 5,000 healthy subjects, or at least 10,000 healthy subjects, or at least 15,000 healthy subjects irrespective of serostatus at baseline, wherein said unit dose/tetravalent dengue virus composition or said placebo is administered at least twice within less than 6 months, such as within 3 months, about 30 days after the second administration of the administration schedule until at least 12 months or until 12 to 18 months (e.g. at 12 or at 18 months) after the second administration of the administration schedule.
  • the lower bound is more than 30%, is more than 40%, or is more than 50%.
  • the invention is directed to said methods having a vaccine efficacy against serotype 1, in preventing virologically confirmable dengue disease with a 2-sided 95% confidence interval, wherein the lower bound is more than 25%, when measured against placebo in a subject population of at least 1,500, or at least 2,000, or at least 5,000 healthy subjects (or at least 10,000, or at least 15,000 healthy subjects) irrespective of serostatus at baseline and 4 to 16 years of age, wherein a reconstituted unit dose/tetravalent dengue virus composition as described herein or placebo is e.g. administered at least twice within less than 6 months, such as within 3 months, from 30 days post last administration until 12 to 18 months (e.g. at 12 or at 18 months) after the last administration.
  • the lower bound is more than 30%, is more than 35% is more than 40%, is more than 45%, is more than 50%, or is more than 54%.
  • the subject population of at least 1,500 is seronegative against all serotypes at base line and the lower bound is more than 35%.
  • the seronegative and seropositive population each provide a vaccine efficacy against serotype 1 with a 2-sided 95% confidence interval, wherein the lower bounds are within 10%-points.
  • the invention is directed to said methods having a vaccine efficacy against dengue serotype 1 of more than 30%, when measured against placebo in a subject population of at least 5,000 healthy subjects, or at least 10,000 healthy subjects, or at least 15,000 healthy subjects irrespective of serostatus at baseline, wherein said unit dose/tetravalent dengue virus composition or said placebo is administered at least twice within less than 6 months, such as within 3 months, 30 days after the second administration until at least 12 months or until 12 to 18 months (e.g. at 12 or at 18 months) after the second administration.
  • the vaccine efficacy against dengue serotype 1 is more than 40%, is more than 50%, is more than 60%, is more than 65%, or is more than 70%.
  • the invention is directed to said methods having a vaccine efficacy against serotype 1, in preventing virologically confirmable dengue disease, when measured against placebo in a subject population of at least 1,500, or at least 2,000, or at least 5,000 healthy subjects (or at least 10,000, or at least 15,000 healthy subjects) irrespective of serostatus at baseline and 4 to 16 years of age, wherein a reconstituted unit dose/tetravalent dengue virus composition as described herein or placebo is e.g. administered at least twice within less than 6 months, such as within 3 months, from 30 days post last administration until 12 to 18 months (e.g. at 12 or at 18 months) after the last administration.
  • the vaccine efficacy is more than 40%, is more than 50%, is more than 60%, or is more than 65%.
  • the subject population of at least 1,500 is seronegative against all serotypes at base line.
  • the seronegative and seropositive population each provide a vaccine efficacy against serotype 1 which are within 5%-points.
  • the invention is directed to said methods having a vaccine efficacy against dengue serotype 2 with a 2-sided 95% confidence interval, wherein the lower bound is more than 25%, when measured against placebo in a subject population of at least 5,000 healthy subjects, or at least 10,000 healthy subjects, or at least 15,000 healthy subjects irrespective of serostatus at baseline, wherein said unit dose/tetravalent dengue virus composition or said placebo is administered at least twice within less than 6 months, such as within 3 months, about 30 days after the second administration of the administration schedule until at least 12 months or until 12 to 18 months (e.g. at 12 or at 18 months) after the second administration of the administration schedule.
  • the lower bound is more than 30%, is more than 40%, is more than 50, is more than 60, is more than 70, is more than 80, or is more than 90%.
  • the invention is directed to said methods having a vaccine efficacy against serotype 2, in preventing virologically confirmable dengue disease with a 2-sided 95% confidence interval, wherein the lower bound is more than 25%, when measured against placebo in a subject population of at least 1,500, or at least 2,000, or at least 5,000 healthy subjects (or at least 10,000, or at least 15,000 healthy subjects) irrespective of serostatus at baseline and 4 to 16 years of age, wherein a reconstituted unit dose/tetravalent dengue virus composition as described herein or placebo is e.g. administered at least twice within less than 6 months, such as within 3 months, from 30 days post last administration until 12 to 18 months (e.g. at 12 or at 18 months) after the last administration.
  • the lower bound is more than 50%, is more than 60%, is more than 70%, is more than 80%, or is more than 85%.
  • the subject population of at least 1,500 is seronegative against all serotypes.
  • the seronegative and seropositive population each provide a vaccine efficacy against serotype 2 with a 2-sided 95% confidence interval, wherein the lower bounds are within 5%-points.
  • the invention is directed to said methods having a vaccine efficacy against dengue serotype 2 of more than 30%, when measured against placebo in a subject population of at least 5,000 healthy subjects, or at least 10,000 healthy subjects, or at least 15,000 healthy subjects irrespective of serostatus at baseline, wherein said unit dose/tetravalent dengue virus composition or said placebo is administered at least twice within less than 6 months, such as within 3 months, 30 days after the second administration until at least 12 months or until 12 to 18 months (e.g. at 12 or at 18 months) after the second administration.
  • the vaccine efficacy against dengue serotype 2 is more than 40%, is more than 50%, is more than 60%, is more than 70%, is more than 80, or is more than 90%.
  • the invention is directed to said methods having a vaccine efficacy against serotype 2, in preventing virologically confirmable dengue disease, when measured against placebo in a subject population of at least 1,500, or at least 2,000, or at least 5,000 healthy subjects (or at least 10,000, or at least 15,000 healthy subjects) irrespective of serostatus at baseline and 4 to 16 years of age, wherein a reconstituted unit dose/tetravalent dengue virus composition as described herein or placebo is e.g. administered at least twice within less than 6 months, such as within 3 months, from 30 days post last administration until 12 to 18 months (e.g. at 12 or at 18 months) after the last administration.
  • the vaccine efficacy is more than 60%, is more than 70%, is more than 80%, or is more than 90%.
  • the subject population of at least 1,500 is seronegative against all serotypes at base line.
  • the seronegative and seropositive population each provide a vaccine efficacy against serotype 2 which are within 5%-points.
  • the invention is directed to said methods having a vaccine efficacy against dengue serotype 3 with a 2-sided 95% confidence interval, wherein the lower bound is more than 25%, when measured against placebo in a subject population of at least 5,000 healthy subjects, or at least 10,000 healthy subjects, or at least 15,000 healthy subjects irrespective of serostatus at baseline, wherein said unit dose/tetravalent dengue virus composition or said placebo is administered at least twice within less than 6 months, such as within 3 months, about 30 days after the second administration of the administration schedule until at least 12 months after the second administration of the administration schedule.
  • the lower bound is more than 30%, is more than 40%.
  • the invention is directed to said methods having a vaccine efficacy against dengue serotype 3 of more than 30%, when measured against placebo in a subject population of at least 5,000 healthy subjects, or at least 10,000 healthy subjects, or at least 15,000 healthy subjects irrespective of serostatus at baseline, wherein said unit dose/tetravalent dengue virus composition or said placebo is administered at least twice within less than 6 months, such as within 3 months, 30 days after the second administration until at least 12 months after the second administration.
  • the vaccine efficacy against dengue serotype 3 is more than 40%, is more than 50%, is more than 55%, or is more than 60%.
  • the invention is directed to said methods having a combined vaccine efficacy against all four serotypes, in preventing virologically confirmable dengue disease with hospitalization with a 2-sided 95% confidence interval, wherein the lower bound is more than 25%, when measured against placebo in a subject population of at least 5,000 healthy subjects (or at least 10,000, or at least 15,000 healthy subjects) irrespective of serostatus at baseline and 4 to 16 years of age, wherein a reconstituted unit dose/tetravalent dengue virus composition as described herein or placebo is e.g. administered at least twice within less than 6 months, such as within 3 months, from first administration until 12 to 18 months (e.g.
  • the lower bound is more than 10%, is more than 20%, is more than 30%, is more than 40%, is more than 50%, is more than 55%, is more than 60%, is more than 65%, is more than 66%, is more than 67%, is more than 70%, is more than 75%, is more than 77%, or is more than 80%.
  • the invention is directed to said methods having a combined vaccine efficacy against all four serotypes, in preventing virologically confirmable dengue disease with hospitalization, when measured against placebo in a subject population of at least 5,000 healthy subjects (or at least 10,000, or at least 15,000 healthy subjects) irrespective of serostatus at baseline and 4 to 16 years of age, wherein a reconstituted unit dose/tetravalent dengue virus composition as described herein or placebo is e.g. administered at least twice within less than 6 months, such as within 3 months, from first administration until 12 to 18 months (e.g. at 12 months or at 18 months) after the last administration, or from 30 days post last administration until 12 to 18 months (e.g. at 12 or at 18 months) after the last administration.
  • the vaccine efficacy is more than is more than 70%, is more than 75%, is more than 80%, or is more than 82%, or is more than 85%, more than 88%.
  • the invention is directed to said methods having a combined vaccine efficacy against virologically-confirmed dengue with hospitalization against all four serotypes with a 2-sided 95% confidence interval, wherein the lower bound is more than 25%, when measured against placebo in a subject population of at least 1,500 or at least 2,000 healthy subjects being seronegative against all serotypes at baseline, wherein said unit dose/tetravalent dengue virus composition or said placebo is administered at least twice within less than 6 months, such as within 3 months, about 30 days after the second administration of the administration schedule until at least 12 months or until 12 to 18 months (e.g. at 12 or at 18 months) after the second administration of the administration schedule.
  • the lower bound is more than 30%, is more than 40%, is more than 50%, is more than 60%, is more than 70%, or is more than 75%.
  • the invention is directed to said methods having a combined vaccine efficacy against all four serotypes, in preventing virologically confirmable dengue disease with hospitalization with a 2-sided 95% confidence interval, wherein the lower bound is more than 25%, when measured against placebo in a subject population of at least 5,000 healthy subjects (or at least 10,000, or at least 15,000 healthy subjects) being seronegative against all serotypes at baseline and 4 to 16 years of age, wherein a reconstituted unit dose/tetravalent dengue virus composition as described herein or placebo is e.g. administered at least twice within less than 6 months, such as within 3 months, from 30 days post last administration until 12 to 18 months (e.g. at 12 months or at 18 months) after the last administration.
  • the lower bound is more than 60%, is more than 65%, is more than 66%, is more than 67%, is more than 70%, is more than 75%, is more than 77% or is more than 80%.
  • the invention is directed to said methods having a combined vaccine efficacy against virologically-confirmed dengue with hospitalization against all four serotypes of more than 30%, when measured against placebo in a subject population of at least 1,500 or at least 2,000 healthy subjects, healthy subjects being seronegative against all serotypes at baseline, wherein said unit dose/tetravalent dengue virus composition or said placebo is administered at least twice within less than 6 months, such as within 3 months, 30 days after the second administration until at least 12 months or until 12 to 18 months (e.g. at 12 or at 18 months) after the second administration.
  • the combined vaccine efficacy against virologically-confirmed dengue with hospitalization against all four serotypes is more than 40%, is more than 50%, is more than 60%, is more than 70%, is more than 80%, or is more than 90%.
  • the invention is directed to said methods having a combined vaccine efficacy against all four serotypes, in preventing virologically confirmable dengue disease with hospitalization, when measured against placebo in a subject population of at least 5,000 healthy subjects (or at least 10,000, or at least 15,000 healthy subjects) being seronegative against all serotypes at baseline and 4 to 16 years of age, wherein a reconstituted unit dose/tetravalent dengue virus composition as described herein or placebo is e.g. administered at least twice within less than 6 months, such as within 3 months, from 30 days post last administration until 12 to 18 months (e.g. at 12 months or at 18 months) after the last administration.
  • the said vaccine efficacy is more than 60%, is more than 65%, is more than 66%, is more than 67%, is more than 70%, is more than 75%, is more than 77%, is more than 80, or is more than 85%.
  • the invention is directed to said methods having a combined vaccine efficacy against virologically-confirmed dengue with hospitalization against all four serotypes with a 2-sided 95% confidence interval, wherein the lower bound is more than 25%, when measured against placebo in a subject population of at least 1,500 or at least 2,000 healthy subjects being seropositive at baseline, wherein said unit dose/tetravalent dengue virus composition or said placebo is administered at least twice within less than 6 months, such as within 3 months, about 30 days after the second administration of the administration schedule until at least 12 months or until 12 to 18 months (e.g. at 12 or at 18 months) after the second administration of the administration schedule.
  • the lower bound is more than 30%, is more than 40%, is more than 50%, is more than 60%, is more than 70%, or is more than 80%.
  • the invention is directed to said methods having a combined vaccine efficacy against all four serotypes, in preventing virologically confirmable dengue disease with hospitalization with a 2-sided 95% confidence interval, wherein the lower bound is more than 25%, when measured against placebo in a subject population of at least 5,000 healthy subjects (or at least 10,000, or at least 15,000 healthy subjects) being seropositive at baseline and 4 to 16 years of age, wherein a reconstituted unit dose/tetravalent dengue virus composition as described herein or placebo is e.g. administered at least twice within less than 6 months, such as within 3 months, from 30 days post last administration until 12 to 18 months (e.g. at 12 months or at 18 months) after the last administration.
  • the lower bound is more than 60%, is more than 65%, is more than 70%, is more than 75%, or is more than 80%.
  • the invention is directed to said methods having a combined vaccine efficacy against virologically-confirmed dengue with hospitalization against all four serotypes of more than 30%, when measured against placebo in a subject population at least 1,500 or of at least 2,000 healthy subjects, healthy subjects being seropositive at baseline, wherein said unit dose/tetravalent dengue virus composition or said placebo is administered at least twice within less than 6 months, such as within 3 months, 30 days after the second administration until at least 12 months or until 12 to 18 months (e.g. at 12 or at 18 months) after the second administration.
  • the combined vaccine efficacy against virologically-confirmed dengue with hospitalization against all four serotypes is more than 40%, is more than 50%, is more than 60%, is more than 70%, is more than 80%, or is more than 90%.
  • the invention is directed to said methods having a combined vaccine efficacy against all four serotypes, in preventing virologically confirmable dengue disease with hospitalization, when measured against placebo in a subject population of at least 5,000 healthy subjects (or at least 10,000, or at least 15,000 healthy subjects) being seropositive at baseline and 4 to 16 years of age, wherein a reconstituted unit dose/tetravalent dengue virus composition as described herein or placebo is e.g. administered at least twice within less than 6 months, such as within 3 months, from 30 days post last administration until 12 to 18 months (e.g. at 12 months or at 18 months) after the last administration.
  • the vaccine efficacy is more than 75%, is more than 70%, is more than 80%, is more than 85%, or is more than 90%.
  • the invention is directed to said methods having a combined vaccine efficacy against all four serotypes, in preventing virologically confirmable dengue disease with hospitalization with a 2-sided 95% confidence interval, wherein the lower bound is more than 25%, when measured against placebo in a subject population of at least 5,000 healthy subjects (or at least 10,000, or at least 15,000 healthy subjects) being seropositive at baseline being or seronegative against all serotypes at baseline and 4 to 16 years of age, wherein a reconstituted unit dose/tetravalent dengue virus composition as described herein or placebo is e.g. administered at least twice within less than 6 months, such as within 3 months, from 30 days post last administration until 12 to 18 months (e.g. at 12 months or at 18 months) after the last administration.
  • the difference between the lower bound provided by the seropositive subjects at baseline and the subjects seronegative against all serotypes at baseline is no more than 15%-points.
  • the invention is directed to said methods having a combined vaccine efficacy against all four serotypes, in preventing virologically confirmable dengue disease with hospitalization, when measured against placebo in a subject population of at least 5,000 healthy subjects (or at least 10,000, or at least 15,000 healthy subjects) being seropositive at baseline being or seronegative against all serotypes at baseline and 4 to 16 years of age, wherein a reconstituted unit dose/tetravalent dengue virus composition as described herein or placebo is e.g. administered at least twice within less than 6 months, such as within 3 months, from 30 days post last administration until 12 to 18 months (e.g. at 12 months or at 18 months) after the last administration.
  • the difference between the vaccine efficacy provided by the seropositive subjects at baseline and the subjects seronegative against all serotypes at baseline is no more than 10%-points or no more than 5%-points.
  • the invention is directed to said methods having a relative risk, preferably a combined relative risk against all four serotypes, with a 2-sided 95% confidence interval, wherein the upper bound is less than 0.75, when measured against placebo in a subject population of at least 5,000 healthy subjects (or at least 10,000, or at least 15,000 healthy subjects) irrespective of serostatus at baseline, wherein a reconstituted unit dose/tetravalent dengue virus composition as described herein or placebo is administered at least twice within less than 6 months, such as within 3 months, 30 days after the second administration until at least 12 months after the second administration.
  • the upper bound is less than 0.70, less than 0.65, less than 0.60, less than 0.55, less than 0.50, less than 0.45, less than 0.40, less than 0.35, less than 0.30 or less than 0.28.
  • said reconstituted unit dose or placebo is administered subcutaneously within about 3 month, such as on days 0 and 90.
  • the invention is directed to said methods having a relative risk, preferably a combined relative risk against all four serotypes, of less than 0.70, when measured against placebo in a subject population of at least 5,000 healthy subjects (or at least 10,000, or at least 15,000 healthy subjects) irrespective of serostatus at baseline, wherein a reconstituted unit dose/tetravalent dengue virus composition as described herein or placebo is administered at least twice within less than 6 months, such as within 3 months, 30 days after the second administration until at least 12 months after the second administration.
  • the relative risk is less than 0.65, less than 0.60, less than 0.55, less than 0.50, less than 0.45, less than 0.40, less than 0.35, less than 0.30, less than 0.25 or less than 0.23.
  • said reconstituted unit dose or placebo is administered subcutaneously within about 3 month, such as on days 0 and 90.
  • the invention is directed to said methods, wherein virologically confirmable dengue disease occurs in less than 2.5% of the subjects, when measured against placebo in a subject population of at least 5,000 healthy subjects (or at least 10,000, or at least 15,000 healthy subjects) irrespective of serostatus at baseline, wherein a reconstituted unit dose/tetravalent dengue virus composition as described herein or placebo is administered at least twice within less than 6 months, such as within 3 months, 30 days after the second administration until at least 12 months or at least 18 months after the second administration.
  • virologically confirmable dengue disease occurs in less than 2.0% of the subjects, less than 1.5% of the subjects, less than 1.0% of the subjects, less than 0.8% of the subjects, or less than 0.6% of the subjects.
  • said reconstituted unit dose or placebo is administered subcutaneously within about 3 month, such as on days 0 and 90.
  • the invention is directed to said methods having a combined vaccine efficacy against all four serotypes with a 2-sided 95% confidence interval, wherein the lower bound is more than 61.0%, or more than 65.0 or more than 70.0% or more than 72.0% when measured against placebo in a subject population of at least 5,000 healthy subjects (or at least 10,000, or at least 15,000 healthy subjects) from endemic irrespective of serostatus at baseline and being selected from the group consisting of 4 to 16 year old subjects at the time of randomization, wherein said unit dose/tetravalent dengue virus composition or said placebo is administered at least twice within 6 months or less, about 30 days after the last administration of the administration schedule until at least 12 or 13 months after the last administration of the administration schedule.
  • the invention is directed to said methods having a combined vaccine efficacy against all four serotypes of more than 66%, or of more than 70%, or of more than 75%, or of more than 77%, or of more than 80.0%, when measured against placebo in a subject population of at least 5,000 healthy subjects (or at least 10,000, or at least 15,000 healthy subjects) from endemic areas irrespective of serostatus at baseline and being selected from the group consisting of 4 to 16 year old subjects at the time of randomization, wherein said unit dose/tetravalent dengue virus composition or said placebo is administered at least twice within 6 months or less, about 30 days after the last administration of the administration schedule until at least 12 months or 13 month after the last administration of the administration schedule.
  • the invention is directed to said methods, wherein the combined vaccine efficacy against all four serotypes is measured about 30 days after the last administration of the administration schedule until 12 or 13 months after the last administration of the administration schedule.
  • the invention is directed to said methods, wherein said unit dose or said placebo is administered at least twice within three months, in particular at about day 1 and about day 90, and wherein the combined vaccine efficacy against all four serotypes is measured 30 days after the second administration until 12 or 13 months after the second administration of the administration schedule.
  • the invention is directed to said methods, wherein said methods are effective and safe.
  • the subject or subject population is under 9 years of age, under 4 years of age, or under 2 years of age or from 2 to 9 years of age, or from 2 to 5 years of age, or from 4 to 9 years of age or from 6 to 9 years of age.
  • the subject is seronegative with respect to all dengue serotypes.
  • the invention is directed to said methods, wherein said methods having a relative risk for virologically confirmed dengue with hospitalization of 1 or less, or 0.8 or less, or 0.6 or less, when measured against placebo in a subject population of at least 5,000 healthy subjects (or at least 10,000, or at least 15,000 healthy subjects).
  • the subject or subject population is under 9 years of age, under 4 years of age, or under 2 years of age or from 2 to 9 years of age, or from 2 to 5 years of age, or from 4 to 9 years of age or from 6 to 9 years of age.
  • the subject is seronegative with respect to all dengue serotypes.
  • the invention is directed to said methods, wherein the healthy subjects of the subject population are 4 to 16 years of age. In some of such embodiments, the healthy subjects of the subject population are 4 to 5 years of age, 6 to 11 years of age, or 12 to 16 years of age.
  • the invention is directed to said methods, wherein the healthy subjects of the subject population are defined as being healthy in view of the exclusion criteria specified in Example 6.
  • the invention is directed to said methods, wherein the healthy subjects of the subject population are from Asia Pacific or Latin America.
  • the invention is directed to said methods, wherein the healthy subjects of the subject population are seropositive with respect to at least one serotype. In other embodiments, the healthy subjects of the subject population are seronegative with respect to all serotypes.
  • the invention is directed to said methods, wherein the healthy subjects of the subject population are 4-5 years of age and from Asia Pacific, 6-11 years of age and from Asia Pacific, or 12-16 years of age and from Asia Pacific. In other embodiments, the healthy subjects of the subject population are 4-5 years of age and from Latin America, 6-11 years of age and from Latin America, or 12-16 years of age and from Latin America.
  • the invention is directed to said methods, wherein the healthy subjects of the subject population are 4-5 years of age and seropositive for at least 1 dengue serotype, 6-11 years of age and seropositive for at least 1 dengue serotype, or 12-16 years of age and seropositive for at least 1 dengue serotype.
  • the healthy subjects of the subject population are 4-5 years of age and seronegative for all dengue serotypes, 6-11 years of age and seronegative for all dengue serotypes, or 12-16 years of age and seronegative for all dengue serotypes.
  • the invention is directed to said methods, wherein the healthy subjects of the subject population are from Asia Pacific or Latin America and seropositive for at least one dengue serotype at baseline. In other embodiments, the healthy subjects of the subject population are from Asia Pacific or Latin America and seronegative for at all dengue serotype at baseline.
  • the invention is directed to said methods, wherein the healthy subjects of the subject population are from Asia Pacific, seropositive for at least one dengue serotype at baseline and 4-5 years of age, 6-11 years of age, or 12-16 years of age.
  • the healthy subjects of the subject population are from Asia Pacific, seronegative for all dengue serotypes at baseline and 4-5 years of age, 6-11 years of age, or 12-16 years of age.
  • the healthy subjects of the subject population are from Latin America, seropositive for at least one dengue serotype at baseline and 4-5 years of age, 6-11 years of age, or 12-16 years of age.
  • the healthy subjects of the subject population are from America, seronegative for all dengue serotypes at baseline and 4-5 years of age, 6-11 years of age, or 12-16 years of age.
  • the invention is directed to said methods, wherein the healthy subjects of the subject population had prior vaccination against Yellow Fever. In other embodiments, the healthy subjects of the subject population had no prior vaccination against Yellow Fever.
  • Prior vaccination indicates a vaccination prior to the first vaccination with the reconstituted unit dose as described herein. For example for vaccine efficacy (VE) as determined in Example 6 from 30 days post-second vaccination, a prior vaccination of Yellow Fever is defined as a Yellow Fever vaccination occurring before 30 days post-second vaccination.
  • VE vaccine efficacy
  • the invention is directed to said methods, wherein the healthy subjects of the subject population had prior vaccination against Japanese Encephalitis. In other embodiments, the healthy subjects of the subject population had no prior vaccination against Japanese Encephalitis.
  • the invention is directed to said methods, wherein the healthy subjects of the subject population received Dengvaxia® within the administration regimen as described herein or within 4.5 years after administration of the first dose. In certain embodiments, the invention is directed to said methods, wherein the occurrence of vaccine related serious adverse events is less than 0.1%.
  • the invention is directed to said methods, wherein the occurrence of vaccine related unsolicited adverse events occurring within 4 weeks of administration is less than 2%.
  • the invention is directed to said methods, wherein the occurrence of vaccine related solicited adverse events occurring within 2 weeks of administration is less than 35%.
  • the invention is directed to said methods, wherein the occurrence of vaccine related solicited local reactions occurring within 1 weeks of administration is less than 40%.
  • the invention is directed to said methods, wherein the method does not increase the risk of virologically-confirmed dengue with hospitalization in the individual, such as in a seronegative individual.
  • the unit dose is co-administered with a vaccine selected from the group of a
  • DTaP/IPV/Hib vaccine in particular a combined DTaP/IPV/Hib vaccine, an MMR vaccine, a yellow fever vaccine, in particular YF-17D, an HPV vaccine, in particular a 9vHPV vaccine, a tetanus, diphtheria, and pertussis (Tdap) vaccine, in particular a combined tetanus toxoid, reduced diphtheria toxoid and acellular pertussis (adsorbed) vaccine, and a hepatitis A vaccine as described in more detail below.
  • the present invention is directed in part to the composition or unit dose of the invention as described herein for use in a method of preventing dengue disease (in particular virologically-confirmable dengue, VCD) in a subject.
  • dengue disease in particular virologically-confirmable dengue, VCD
  • the present invention is directed in part to the composition or unit dose of the invention as described herein for use in a method of preventing dengue disease (in particular virologically-confirmable dengue, VCD) in a subject population.
  • dengue disease in particular virologically-confirmable dengue, VCD
  • the present invention is directed in part to a method of preventing dengue hemorrhagic fever (DHF) and dengue shock syndrome (DSS).
  • DHF dengue hemorrhagic fever
  • DSS dengue shock syndrome
  • Method of preventing, method of inoculating is to be understood to be also disclosed as unit dose for use in such a method of preventing dengue disease in a subject or subject population irrespective of being expressly stated below.
  • the composition or unit dose of the invention is a tetravalent dengue virus composition including a chimeric dengue serotype 2/1 strain and a dengue serotype 2 strain and a chimeric dengue serotype 2/3 strain and a chimeric dengue serotype 2/4 strain.
  • the dengue serotype 2 strain is derived from the wild type virus strain DEN-2 16681 (represented by SEQ ID NO 11) and differs in at least three nucleotides from the wild type as follows:
  • the tetravalent dengue virus composition for such use may be in the form of a unit dose comprising:
  • a dengue serotype 1 in a concentration of at least 3.3 log 10 pfu/0.5 ml
  • a dengue serotype 2 in a concentration of at least 2.7 log 10 pfu/0.5 ml
  • a dengue serotype 3 in a concentration of at least 4.0 log 10 pfu/0.5 ml
  • a dengue serotype 4 in a concentration of at least 4.5 log 10 pfu/0.5 ml.
  • the tetravalent dengue virus composition for such use may also be in the form of a lyophilized unit dose which upon reconstitution with 0.5 mL of a pharmaceutically acceptable diluent comprises:
  • a dengue serotype 1 in a concentration of at least 3.3 log 10 pfu/0.5 ml
  • a dengue serotype 2 in a concentration of at least 2.7 log 10 pfu/0.5 ml
  • a dengue serotype 3 in a concentration of at least 4.0 log 10 pfu/0.5 ml
  • a dengue serotype 4 in a concentration of at least 4.5 log 10 pfu/0.5 ml.
  • the concentration of (ii) in pfu/0.5 mL is preferably less than 10%, and the concentration of (iv) in pfu/0.5 mL is preferably at least 50%, and the concentration of (i) in pfu/0.5 mL is preferably at least 1%, and the concentration of (iii) in pfu/0.5 mL is preferably at least 8%, or more preferred at least 10%, or at least 12%, or at least 14%, or at least 16%, or at least 18% and wherein the subject is preferably 2 to 17 years of age or 4 to 16 years of age.
  • the concentration of (ii) in pfu/0.5 mL is preferably less than 2%
  • the concentration of (iv) in pfu/0.5 mL is preferably at least 50%
  • the concentration of (i) in pfu/0.5 mL is at preferably least 1%
  • the concentration of (iii) in pfu/0.5 mL is preferably at least 6% and wherein the subject preferably is 18 to 60 years of age.
  • tetravalent composition or the unit dose can be derived from section “Dengue vaccine composition” and “Unit dose” above.
  • Such method preferably provides a combined vaccine efficacy against all four serotypes in preventing virologically confirmable dengue disease with a 2-sided 95% confidence interval, wherein the lower bound is more than 60%, when measured against placebo in a subject population of at least 5,000 healthy subjects irrespective of serostatus at baseline and 14 to 16 years of age, from the first administration of the administration schedule until 18 months after the second administration of the administration schedule.
  • Such method also preferably provides a combined vaccine efficacy against all four serotypes, in preventing virologically confirmable dengue disease with a 2-sided 95% confidence interval, wherein the lower bound is more than 45%, when measured against placebo in a subject population of at least 1,500 or at least 2,000 healthy subjects seronegative against all serotypes at baseline and 14 to 16 years of age, from 30 days after the second administration of the administration schedule until 18 months after the second administration of the administration schedule.
  • the use is for a method of inoculation against the virologically confirmable dengue disease is due to a dengue serotype 2, and/or due to a dengue serotype 1.
  • the method has very high efficacy against dengue serotype 2 and dengue serotype 1 and the highest efficacy against dengue serotype 2.
  • the invention is directed to said methods having a vaccine efficacy against serotype 1, in preventing virologically confirmable dengue disease with a 2-sided 95% confidence interval, wherein the lower bound is more than 25%, when measured against placebo in a subject population of at least 1,500, or at least 2,000, or at least 5,000 healthy subjects (or at least 10,000, or at least 15,000 healthy subjects) irrespective of serostatus at baseline and 4 to 16 years of age from 30 days post second administration until 12 to 18 months (e.g. at 12 or at 18 months) after the second administration.
  • the lower bound is more than 30%, is more than 35% is more than 40%, is more than 45%, is more than 50%, or is more than 54%.
  • the subject population of at least 1,500 is seronegative against all serotypes at base line and the lower bound is more than 35%.
  • the seronegative and seropositive population each provide a vaccine efficacy against serotype 1 with a 2-sided 95% confidence interval, wherein the lower bounds are within 10%-points.
  • the invention is directed to said methods having a vaccine efficacy against serotype 1, in preventing virologically confirmable dengue disease, when measured against placebo in a subject population of at least 1,500, or at least 2,000, or at least 5,000 healthy subjects (or at least 10,000, or at least 15,000 healthy subjects) irrespective of serostatus at baseline and 4 to 16 years of age from 30 days post second administration until 12 to 18 months (e.g. at 12 or at 18 months) after the second administration.
  • the vaccine efficacy is more than 40%, is more than 50%, is more than 60%, or is more than 65%.
  • the subject population of at least 1,500 is seronegative against all serotypes at base line.
  • the seronegative and seropositive population each provide a vaccine efficacy against serotype 1 which are within 5%-points.
  • the invention is directed to said methods having a vaccine efficacy against serotype 2, in preventing virologically confirmable dengue disease with a 2-sided 95% confidence interval, wherein the lower bound is more than 25%, when measured against placebo in a subject population of at least 1,500, or at least 2,000, or at least 5,000 healthy subjects (or at least 10,000, or at least 15,000 healthy subjects) irrespective of serostatus at baseline and 4 to 16 years of age from 30 days post second administration until 12 to 18 months (e.g. at 12 or at 18 months) after the second administration.
  • the lower bound is more than 50%, is more than 60%, is more than 70%, is more than 80%, or is more than 85%.
  • the subject population of at least 1,500 is seronegative against all serotypes.
  • the seronegative and seropositive population each provide a vaccine efficacy against serotype 2 with a 2-sided 95% confidence interval, wherein the lower bounds are within 5%-points.
  • the invention is directed to said methods having a vaccine efficacy against serotype 2, in preventing virologically confirmable dengue disease, when measured against placebo in a subject population of at least 1,500, or at least 2,000, or at least 5,000 healthy subjects (or at least 10,000, or at least 15,000 healthy subjects) irrespective of serostatus at baseline and 4 to 16 years of age from 30 days post second administration until 12 to 18 months (e.g. at 12 or at 18 months) after the second administration.
  • the vaccine efficacy is more than 60%, is more than 70%, is more than 80%, or is more than 90%.
  • the subject population of at least 1,500 is seronegative against all serotypes at base line.
  • the seronegative and seropositive population each provide a vaccine efficacy against serotype 2 which are within 5%-points.
  • the unit dose is reconstituted and administered by subcutaneous injection.
  • the subcutaneous injection is administered to the arm, preferably to the deltoid region of the arm.
  • such a method does not include a step of determination of a previous dengue infection in the subjects preferably at any time before, during or after the steps of administration or wherein the serostatus of the subject is unknown preferably at any time before, during or after the steps of administration.
  • the method according to the invention does not require the testing of the serostatus before vaccination and thus allows immediate treatment and outbreak control.
  • the use is for a method wherein the subject is exposed to a dengue outbreak.
  • the outbreak is due to a dengue serotype 2, and/or due to a serotype 1.
  • the subject is from a region wherein the seroprevalence rate is unknown and/or wherein the seroprevalence rate is below 80%, or below 70%, or below 60%.
  • the subject is seronegative at baseline and is from a region or travels to a region wherein the seroprevalence rate is high with respect to serotype 1 and/or serotype 2 i.e. 80%, or 90% or above.
  • the vaccine and corresponding method is safe for seronegative and seropositive subjects and thus does not require an analysis of the serostatus or a determination of a previous dengue infection or a high seroprevalence rate in the region.
  • Such a method preferably provides a combined vaccine efficacy against virologically-confirmed dengue with hospitalization against all four serotypes with a 2-sided 95% confidence interval, wherein the lower bound is more than 65%, when measured against placebo in a subject population of at least 5,000 healthy 4 to 16 year old subjects irrespective of serostatus at baseline, preferably in at least 1,500 healthy 4 to 16 year old subjects seronegative at baseline, from first administration of the administration schedule until 12 to 18 months after the second administration of the administration schedule.
  • the 2-sided 95% confidence interval of the combined vaccine efficacy against virologically-confirmed dengue with hospitalization against all four serotypes when comparing seropositive and seronegative subjects provides for lower bounds of the 2-sided confidence interval which are within 10% points or within 15% points or within 20% points.
  • the method is preferably safe with respect to serotype 1 and serotype 2 which may therefore be used in outbreak situations due to serotype 1 and/or serotype 2 or even for seronegative subjects (e.g. travelers) or subjects with unknown serostatus in regions with very high seroprevalence rates (>80%) due to serotype 1 and/or serotype 2.
  • such a method does not include the active surveillance with respect to febrile illness of the subject after the administration of the first- and second-unit dose.
  • active surveillance any subject with febrile illness (defined as fever 38° C. on any 2 of 3 consecutive days) will be asked to return to the site for dengue fever evaluation by the Investigator.
  • Subjects/guardians will be contacted at least weekly to ensure robust identification of febrile illness by reminding subjects/guardians of their obligation to return to the site in case of febrile illness. This contact will be implemented through appropriate methods that may differ in each trial site (eg, phone calls, text messaging, home visits, school-based surveillance).
  • such a method does not include vaccine immunogenicity analysis including GMTs for dengue neutralizing antibodies.
  • Such a method does not include a reactogenicity analysis.
  • a reactogenicity analysis relates to solicited local AEs (injection site pain, injection site erythema, and injection site swelling) and solicited systemic AEs (child ⁇ 6 years: fever, irritability/fussiness, drowsiness and loss of appetite; child 6 years: asthenia, fever, headache, malaise and myalgia) which will e.g. be assessed for 7 days and 14 days, respectively, following each vaccination (vaccination day included) via collection of diary cards.
  • the method does not include an active surveillance, an immunogenicity analysis and a reactogenicity analysis.
  • the vaccine and the corresponding method of inoculation are safe and therefore do not require further steps of surveillance or analysis.
  • A selecting a subject for administration of the unit doses of the tetravalent dengue virus composition in need for protection against dengue infection without determination of a previous dengue infection, and (B) administering a first unit dose of the tetravalent dengue virus composition to the subject, and (C) administering a second unit dose of the tetravalent dengue virus composition to the subject within 3 months of administration of the first unit dose. Therefore the method of inoculating is finalized without determination of a previous dengue infection.
  • the method however further optionally comprises at least 1 years after the administration of the second unit dose a booster dose of the unit dose.
  • Selecting the subject may include all types of considerations but preferably not the determination of a previous dengue infection.
  • the selection may include consideration of the age, health conditions, and threat of infection.
  • the threat of infection includes consideration of the seroprevalence rate in the region in which the subject normally lives or intends to travel, the serotype specific seroprevalence rate and an outbreak situation or serotype specific outbreak situations.
  • the subject may be selected due to its exposure to serotype 1 and/or serotype 2 or due to the fact it requires protection against a specific dengue serotype, i.e. serotype 1 and/or serotype 2.
  • the method is applicable to subjects of all kinds of ages.
  • the subject is under 9 years of age, or 4 to 5 years of age, or 6 to 11 years of age or 12 to 16 years, or 6 to 16 years of age or 4 to 16 years of age, or 2 to 17 years of age, or 9 years of age, or over 9 years of age, or 9 to 17 years of age, or 18 to 45 years of age, or 46 to 60 years of age, or over 60 years of age.
  • the unit dose of the tetravalent dengue virus composition comprising (i), (ii), (iii), and (iv) upon reconstitution with a pharmaceutically acceptable diluent provides a total concentration of pfu/0.5 mL of (i), (ii), (iii), and (iv) and based on said total concentration the concentration of (ii) in pfu/0.5 mL is preferably less than 10%, and the concentration of (iv) in pfu/0.5 mL is preferably at least 50%, and the concentration of (i) in pfu/0.5 mL is preferably at least 1%, and the concentration of (iii) in pfu/0.5 mL is preferably at least 8%, or more preferred at least 10%, or at least 12%, or at least 14%, or at least 16%, or at least 18% and the subject is 2 to 17 years of age or 4 to 16 years of age.
  • the unit dose of the tetravalent dengue virus composition comprising (i), (ii), (iii), and (iv) upon reconstitution with a pharmaceutically acceptable diluent provides a total concentration of pfu/0.5 mL of (i), (ii), (iii), and (iv) and based on said total concentration the concentration of (ii) in pfu/0.5 mL is preferably less than 2%, the concentration of (iv) in pfu/0.5 mL is preferably at least 50%, the concentration of (i) in pfu/0.5 mL is at preferably least 1%, and the concentration of (iii) in pfu/0.5 mL is preferably at least 6% and the subject is 18 to 60 years of age.
  • the present invention is directed in part to a reconstituted unit dose of a dengue vaccine composition as described herein for use in a method of preventing virologically confirmable dengue disease in a subject comprising administering at least a first unit dose of the dengue vaccine composition to the subject, wherein the dengue vaccine composition is a tetravalent dengue virus composition including four dengue virus strains representing dengue serotype 1, dengue serotype 2, dengue serotype 3 and dengue serotype 4, optionally wherein the dengue virus strains are live, attenuated dengue virus strains and/or comprise chimeric dengue viruses and/or at least one non-chimeric dengue virus, and wherein upon reconstitution with 0.5 mL of a pharmaceutically acceptable diluent
  • the present invention is directed in part to a reconstituted unit dose of a dengue vaccine composition as described herein for use in a method of preventing virologically confirmable dengue disease in a subject comprising consecutively administering at least a first and a second unit dose of the dengue vaccine composition to the subject, wherein said first and second unit dose are administered subcutaneously within 3 months and at least 4 weeks apart, optionally at about day 1 and at about day 90, wherein the dengue vaccine composition is a tetravalent dengue virus composition including four dengue virus strains representing dengue serotype 1, dengue serotype 2, dengue serotype 3 and dengue serotype 4, optionally wherein the dengue virus strains are live, attenuated, and wherein upon reconstitution with 0.5 mL of a pharmaceutically acceptable diluent
  • the invention is directed to a reconstituted unit dose of a dengue vaccine composition for use in a method of preventing virologically confirmable dengue disease in a subject comprising consecutively administering at least a first and a second unit dose of the dengue vaccine composition to the subject, wherein said first and second unit dose are administered subcutaneously within 3 months and at least 4 weeks apart, optionally at about day 1 and at about day 90, wherein the dengue vaccine composition is a tetravalent dengue virus composition including four dengue virus strains representing dengue serotype 1, dengue serotype 2, dengue serotype 3 and dengue serotype 4, optionally wherein the dengue virus strains are live, attenuated, wherein the subject is under 9 years of age and/or when the serostatus of the subject is unknown or seronegative and wherein upon reconstitution with 0.5 mL of a pharmaceutically acceptable diluent
  • the reconstituted unit dose is administered to a subject of unknown serostatus and/or wherein no test has been carried out to determine whether the subject is seropositive or seronegative before the unit dose as described herein is administered.
  • the subject is under 9 years of age and/or the serostatus of the subject is unknown or seronegative. In certain such embodiments, the subject is under 9 years of age and the serostatus of the subject is unknown or seronegative, preferably seronegative.
  • the method is safe.
  • the subject is under 9 years of age or from 4 years of age and/or the serostatus of the subject is unknown or seronegative.
  • the subject is from 4 years of age and the serostatus of the subject is unknown or seronegative, preferably seronegative.
  • the method is effective.
  • the subject is under 9 years of age and/or the serostatus of the subject is unknown or seronegative.
  • the subject is under 9 years of age and the serostatus of the subject is unknown or seronegative, preferably seronegative.
  • the dengue serotype 1 and the dengue serotype 2 are present each in a concentration based on the total concentration in pfu/0.5 mL which is within 5%-points of each other and/or are together less than about 10% of the total concentration in pfu/0.5 mL.
  • the dengue serotype 3 is at least about 10% of the total concentration in pfu/0.5 mL and/or the dengue serotype 4 is at least about 70% of the total concentration in pfu/0.5 mL.
  • the dengue serotype 4 represents the highest concentration in the composition of all four serotypes, preferably with at least about 70% of the total concentration in pfu/0.5 mL
  • dengue serotype 3 represents the second highest concentration in the composition of all four serotypes, preferably with at least about 10% of the total concentration in pfu/0.5 mL
  • dengue serotype 1 and dengue serotype 2 each represent lower concentrations than the concentration of serotype 3, and optionally together represent less than about 10% of the total concentration in pfu/0.5 mL.
  • the composition includes at least one chimeric dengue virus. In certain such embodiments, the composition includes at least one non-chimeric dengue virus and at least one chimeric dengue virus.
  • the subject is seronegative to all dengue serotypes at baseline and/or is from 4 years of age, optionally to 60 years of age.
  • the subject is 4 to 16 years of age, under 9 years of age, from 2 years of age to under 9 years of age, from 4 years of age to under 9 years of age, 4 to 5 years of age, 6 to 11 years of age, or 12 to 16 years of age.
  • the subject is seropositive to at least one dengue serotypes at baseline and/or is from 4 years of age, optionally to 60 years of age.
  • the subject is 4 to 16 years of age, under 9 years of age, from 2 years of age to under 9 years of age, from 4 years of age to under 9 years of age, 4 to 5 years of age, 6 to 11 years of age, or 12 to 16 years of age.
  • the method does not comprise a determination of a previous dengue infection in the subject before the administration of the first unit dose of the tetravalent dengue virus composition.
  • the subject has not been tested for the presence a previous dengue infection.
  • the dengue serotype 1 is a chimeric dengue serotype 2/1 strain
  • the dengue serotype 2 is a non-chimeric dengue serotype 2 strain
  • the dengue serotype 3 is a chimeric dengue serotype 2/3 strain
  • the dengue serotype 4 is a chimeric dengue serotype 2/4 strain and the dengue serotype 1 has the amino acid sequence of SEQ ID NO. 2
  • the dengue serotype 2 has the amino acid sequence of SEQ ID NO. 4
  • the dengue serotype 3 has the amino acid sequence of SEQ ID NO. 6
  • the dengue serotype 4 has the amino acid sequence of SEQ ID NO. 8.
  • the unit dose further comprises from about 10% w/v to about 20% w/v 0,0-trehalose dihydrate or an equimolar amount of other forms of ⁇ , ⁇ -trehalose, from about 0.5% w/v to about 1.5% w/v poloxamer 407, from about 0.05% w/v to about 2% w/v human serum albumin, and from about 70 mM to 140 mM sodium chloride when measured in 0.5 mL.
  • the unit dose comprises about 15% (w/v) ⁇ , ⁇ -trehalose dihydrate, about 1% (w/v) poloxamer 407, about 0.1% (w/v) human serum albumin, and about 100 mM sodium chloride when measured in 0.5 mL.
  • the method is for preventing dengue hemorrhagic fever (DHF) or dengue shock syndrome (DSS).
  • DHF dengue hemorrhagic fever
  • DSS dengue shock syndrome
  • the subject is from a dengue endemic region. In other embodiments, the subject is from a dengue non-endemic region.
  • the subject is from Asia Pacific or Latin America.
  • the reconstituted unit dose provides a seropositivity rate when it is administered to a subject population of at least 50 subjects in two unit doses subcutaneously at day 1 and at day 90, wherein the subjects of the subject population are seronegative to all dengue serotypes at baseline.
  • At least 80% of the subject population are seropositive for all four dengue serotypes at least one month after administration of the first unit dose, such as at day 30, and/or at least 80% of the subject population are seropositive for all four dengue serotypes before or at the time of the administration of the second unit dose, such as at day 90, and/or at least 80%, or at least 85% or at least 90%, or at least 95% of the subject population are seropositive for all four dengue serotypes after the administration of the second unit dose, such as at day 120, and/or at least 80%, or at least 85%, or at least 90% of the subject population are seropositive for all four dengue serotypes after the administration of the second unit dose, such as at day 270.
  • At least 80% of the subject population are seropositive for all four dengue serotypes at least one month after administration of the first unit dose, such as at day 30, and before or at the time of the administration of the second unit dose, such as at day 90, and after the administration of the second unit dose, such as at day 120 and at day 270.
  • the reconstituted unit dose provides a seropositivity rate, when it is administered to a subject population of at least 100 subjects in two unit doses subcutaneously at day 1 and at day 90, wherein the subjects of the subject population comprises from 20% to 40% subjects who are seronegative to all dengue serotypes and from 60% to 80% subjects who are seropositive to at least one dengue serotype at base line, wherein at day 120 and/or day 270 the seropositivity rate for all four dengue serotypes in the seronegative part of the subject population and the seropositivity rate for all four dengue serotypes in the seropositive part of the subject population do not deviate more than 10%-points and/or wherein at day 120 the seropositivity rate for all four dengue serotypes in the seronegative part of the subject population and the seropositivity rate for all four dengue serotypes in the seropositive part of the subject population do not deviate more than 5%-points.
  • the invention is directed to a dengue vaccine composition as described herein for use in a method of preventing virologically confirmable dengue disease in a subject comprising consecutively administering at least a first and a second unit dose of the dengue vaccine composition to the subject, wherein said first and second unit dose are administered subcutaneously within 3 months and at least 4 weeks apart, optionally at about day 1 and at about day 90, and wherein the dengue vaccine composition is a tetravalent dengue virus composition including four live, attenuated dengue virus strains representing dengue serotype 1, dengue serotype 2, dengue serotype 3 and dengue serotype 4, wherein the attenuated dengue virus strains comprise chimeric dengue viruses and at least one non-chimeric dengue virus, and wherein the dengue serotype 1 and the dengue serotype 2 are present each in a concentration based on the total concentration in pfu/0.5 mL which is within 5%-points of each other and/or are together less than about
  • the method does not comprise a determination of a previous dengue infection of the subject before the administration of the first unit dose of the tetravalent dengue virus composition and wherein the method is safe and effective.
  • the subject has not been tested for the presence a previous dengue infection.
  • the dengue serotype 3 is at least about 10% of the total concentration in pfu/0.5 mL and/or the dengue serotype 4 is at least about 70% of the total concentration in pfu/0.5 mL.
  • the dengue serotype 4 represents the highest concentration in the composition of all four serotypes, preferably with at least about 70% of the total concentration in pfu/0.5 mL
  • dengue serotype 3 represents the second highest concentration in the composition of all four serotypes, preferably with at least about 10% of the total concentration in pfu/0.5 mL
  • dengue serotype 1 and dengue serotype 2 each represent lower concentrations than the concentration of serotype 3, and optionally together represent less than about 10% of the total concentration in pfu/0.5 mL.
  • the dengue serotype 1 is a chimeric dengue serotype 2/1 strain
  • the dengue serotype 2 is a non-chimeric dengue serotype 2 strain
  • the dengue serotype 3 is a chimeric dengue serotype 2/3 strain
  • the dengue serotype 4 is a chimeric dengue serotype 2/4 strain and the dengue serotype 1 has the amino acid sequence of SEQ ID NO. 2
  • the dengue serotype 2 has the amino acid sequence of SEQ ID NO. 4
  • the dengue serotype 3 has the amino acid sequence of SEQ ID NO. 6
  • the dengue serotype 4 has the amino acid sequence of SEQ ID NO. 8.
  • the unit doses are administered to the deltoid region of the arm.
  • the composition is administered without determining the serostatus of the subject at baseline and wherein the administration is safe and effective regardless of the serostatus at base line.
  • the subject is seronegative to all dengue serotypes at baseline and/or is from 4 years of age, optionally to 60 years of age.
  • the subject is 4 to 16 years of age, under 9 years of age, from 2 years of age to under 9 years of age, from 4 years of age to under 9 years of age, 4 to 5 years of age, 6 to 11 years of age, or 12 to 16 years of age.
  • the subject may be under 9 years of age and seronegative to all four dengue serotypes at baseline.
  • the subject is seropositive to at least one dengue serotypes at baseline and/or is from 4 years of age, optionally to 60 years of age.
  • the subject is 4 to 16 years of age, under 9 years of age, from 2 years of age to under 9 years of age, from 4 years of age to under 9 years of age, 4 to 5 years of age, 6 to 11 years of age, or 12 to 16 years of age.
  • the subject may be under 9 years of age and seropositive to at least one dengue serotypes at baseline.
  • the subject is 4 to 5 years of age, 6 to 11 years of age or 12 to 16 years of age.
  • the method is for preventing dengue hemorrhagic fever (DHF) or dengue shock syndrome (DSS).
  • DHF dengue hemorrhagic fever
  • DSS dengue shock syndrome
  • the subject is from a dengue endemic region or from a dengue non-endemic region.
  • the subject is from Asia Pacific or Latin America.
  • the composition provides a seropositivity rate when it is administered to a subject population of at least 50 subjects in two unit doses subcutaneously at day 1 and at day 90, wherein the subjects of the subject population are seronegative to all dengue serotypes at baseline, in particular wherein at least one month after administration of the first unit dose, such as at day 30, at least 80% of the subject population are seropositive for all four dengue serotypes, and/or at least 80% of the subject population are seropositive for all four dengue serotypes before or at the time of the administration of the second unit dose, such as at day 90, and/or at least 80%, or at least 85% or at least 90%, or at least 95% of the subject population are seropositive for all four dengue serotypes after the administration of the second unit dose, such as at day 120, and/or at least 80%, or at least 85%, or at least 90% of the subject population are seropositive for all four dengue serotypes after the administration of the second unit dose,
  • the composition provides a seropositivity rate, when it is administered to a subject population of at least 100 subjects in two unit doses subcutaneously at day 1 and at day 90, wherein the subjects of the subject population comprises from 20% to 40% subjects who are seronegative to all dengue serotypes and from 60% to 80% subjects who are seropositive to at least one dengue serotype at base line, wherein at day 120 and/or day 270 the seropositivity rate for all four dengue serotypes in the seronegative part of the subject population and the seropositivity rate for all four dengue serotypes in the seropositive part of the subject population do not deviate more than 10%-points and/or wherein at day 120 the seropositivity rate for all four dengue serotypes in the seronegative part of the subject population and the seropositivity rate for all four dengue serotypes in the seropositive part of the subject population do not deviate more than 5%-points.
  • the present invention is in part directed to the unit dose of the invention as described herein for use in a method of preventing dengue disease (in particular virologically confirmable dengue, VCD) in a subject population comprising administering to the subject population at least a first reconstituted unit dose of the invention as described herein, wherein certain ratios of geometric mean neutralizing antibody titers (GMTs) at day 180 or 365 after administration of said first unit dose to the subject population are achieved.
  • GCTs geometric mean neutralizing antibody titers
  • the geometric mean neutralizing antibody titer for dengue serotype 2 (GMT DENV-2) and the geometric mean neutralizing antibody titer for dengue serotype 4 (GMT DENV-4) when tested in at least 40, or at least 50, or at least 60 subjects at day 180 or day 365 after at least a first administration of said reconstituted unit dose of the invention as described herein, and optionally a second administration of a reconstituted unit dose of the invention as described herein 90 days after said first administration, provide a ratio of GMT DENV-2:GMT DENV-4 of not more than 50, or not more than 40, or not more than 30, or not more than 20.
  • the ratio of GMT DENV-2:GMT DENV-1 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose
  • the ratio of GMT DENV-2:GMT DENV-3 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose.
  • the present invention is in part directed to the unit dose of the invention as described herein for use in a method of preventing dengue disease (in particular virologically confirmable dengue, VCD) in a subject comprising administering to the subject at least a first reconstituted unit dose of the invention as described herein, wherein certain ratios of neutralizing antibody titers at day 180 or 365 after administration of said first unit dose to the subject are achieved.
  • dengue disease in particular virologically confirmable dengue, VCD
  • the neutralizing antibody titer for dengue serotype 2 and the neutralizing antibody titer for dengue serotype 4 at day 180 or day 365 after at least a first administration of the reconstituted unit dose of the invention as described herein, and optionally a second administration of a reconstituted unit dose of the invention as described herein 90 days after said first administration provide a ratio of neutralizing antibody titer for DENV-2:neutralizing antibody titer for GMT DENV-4 of not more than 50, or not more than 40, or not more than 30, or not more than 20.
  • the ratio of the neutralizing antibody titers of DENV-2:DENV-1 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose, and/or the ratio of the neutralizing antibody titers of DENV-2:DENV-3 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose.
  • the geometric mean neutralizing antibody titers (GMTs) of a subject population or the neutralizing antibody titers of a subject are determined in accordance with the microneutralization test disclosed herein, for example according to the method described in Example 2.
  • the invention is directed to the reconstituted unit dose of the invention as described herein for said uses, wherein said unit dose is administered by subcutaneous injection.
  • the subcutaneous injection is administered to the arm, preferably to the deltoid region of the arm.
  • the invention is directed to a reconstituted unit dose of the invention as described herein for said uses, wherein the subject or subject population is seronegative to all dengue serotypes.
  • the invention is directed to a reconstituted unit dose of the invention as described herein for said uses, wherein a single unit dose of the invention as described herein is administered.
  • the invention is directed to a reconstituted unit dose of the invention as described herein for said uses, wherein two reconstituted unit doses of the invention as described herein are administered.
  • the two reconstituted unit doses are administered within 12 months or more, or within six months, or within three months, such as at day 0 and day 90 or at day 1 and day 90.
  • a third reconstituted unit dose of the invention as described herein may be administered after the second administration.
  • Such a third administration may act as a booster and may be administered between 6 to 12 months after the first administration, such as 12 months after the first administration, or later than 12 month after the first administration, such as 12 months (1 year) after the second administration or even 5 years or longer after the first or second administration.
  • the invention is directed to a reconstituted unit dose of the invention as described herein for said uses, wherein the reconstituted unit dose of the invention as described herein is administered at most in two doses or in one dose.
  • the subject is seronegative with respect to all dengue serotypes.
  • the subject is seronegative with respect to all dengue serotypes and the reconstituted unit dose is administered to the seronegative subject by subcutaneous injection.
  • the subject is seropositive with respect to at least one dengue serotype.
  • the invention is directed to the reconstituted unit dose of the invention as described herein for said uses, wherein the reconstituted unit dose of the invention as described herein is administered to a subject or subject population from a dengue endemic region.
  • the subject or subject population is from Singapore, Dominican Republic, Panama, Philippines, Colombia, Puerto Rico or Thailand, in particular from Singapore, Dominican Republic, Panama, or Philippines.
  • the subject or subject population is from a dengue non-endemic region.
  • Such a subject population or such a subject may be vaccinated according to the invention in the context of traveling to a dengue-endemic region.
  • the reconstituted unit dose of the invention as described herein is administered subcutaneously to a subject or subject population from a dengue endemic region or from a dengue non-endemic region.
  • the invention is directed to the unit dose of the invention as described herein for said uses, wherein the subject or subject population is of 2 to 60 years of age, or more than 17 years, or more than 18 years, or 18 to 60 years of age. In certain embodiments, the subject or subject population is of 1 to 17 years of age, or less than 9 years of age, or less than 4 years of age or less than 2 years of age. In further specific embodiments, the subjects or subject population are adults of more than 21 years, or 21 to 60 years, or 21 to 45 years of age. According to some of these embodiments the subject or subject population is seronegative and from a dengue-endemic region.
  • the invention is directed to the reconstituted unit dose of the invention as described herein for said uses, wherein the unit dose of the invention as described herein is administered to a pediatric subject or pediatric subject population of less than 2 years of age, preferably of 2 months to 2 years of age or 2 months to 1.5 years of age or 2 months to 1 year of age.
  • the pediatric subject or pediatric subject population is seronegative and from a dengue endemic region.
  • the invention is directed to the reconstituted unit dose of the invention as described herein for said uses, wherein the reconstituted unit dose is administered subcutaneously to a pediatric subject or pediatric subject population of less than 2 years of age, preferably of 2 months to 2 years of age or 2 months to 1.5 years of age or 2 months to 1 year of age.
  • the pediatric subject or pediatric subject population is seronegative and from a dengue endemic region.
  • the unit dose for use in the methods described above may be any unit dose of a dengue vaccine composition as described above under the headings “Unit dose” or “Dengue vaccine composition” and comprise any dengue virus strain as described above under the heading “Dengue virus strain”.
  • the present invention is directed in part to the unit dose of the invention as described herein for use in a method of preventing dengue hemorrhagic fever (DHF) and dengue shock syndrome (DSS) in an elderly subject.
  • DHF dengue hemorrhagic fever
  • DSS dengue shock syndrome
  • the invention is directed to a reconstituted unit dose of the invention as described herein for said uses, wherein the elderly subject is seronegative to all dengue serotypes.
  • the invention is directed to a reconstituted unit dose of the invention as described herein for said uses, wherein the elderly subject is seronegative to all dengue serotypes.
  • the invention is directed to a reconstituted unit dose of the invention as described herein for said uses, wherein two reconstituted unit doses of the invention as described herein are administered.
  • the two reconstituted unit doses are administered within 12 months or more, or within six months, or within three months, such as at day 0/1 and day 90.
  • a third reconstituted unit dose of the invention as described herein may be administered after the second administration.
  • Such a third administration may act as a booster and may be administered between 6 to 12 months after the first administration, such as 12 months after the first administration, or later than 12 month after the first administration, such as 12 months (1 year) after the second administration or even 5 years or longer after the first or second administration.
  • the elderly subject is seronegative with respect to all dengue serotypes. In certain embodiments of the invention the elderly subject is seronegative with respect to all dengue serotypes and the reconstituted unit dose is administered to the seronegative subject by subcutaneous injection.
  • the elderly subject is seropositive with respect to at least one dengue serotype.
  • the invention is directed to the reconstituted unit dose of the invention as described herein for said uses, wherein the reconstituted unit dose of the invention as described herein is administered to an elderly subject from a dengue endemic region.
  • the elderly subject is from Singapore, Dominican Republic, Panama, Philippines, Colombia, Puerto Rico or Thailand, in particular from Singapore, Dominican Republic, Panama, or Philippines.
  • the elderly subject is from a dengue non-endemic region.
  • Such an elderly subject may be vaccinated according to the invention in the context of traveling to a dengue-endemic region.
  • the reconstituted unit dose of the invention as described herein is administered subcutaneously to an elderly subject from a dengue endemic region or from a dengue non-endemic region.
  • the invention is directed to the reconstituted unit dose of the invention as described herein for said uses, wherein the elderly subject has at least one chronic condition or disease.
  • the at least one chronic condition or disease may be selected from diabetes, hypertension, allergies, previous strokes, ischemic heart disease, chronic renal impairment and chronic obstructive pulmonary disease.
  • the invention is directed to the reconstituted unit dose of the invention as described herein for said uses, wherein the elderly subject has an impaired immune system.
  • the present invention is directed in part to the use of a unit dose of the invention as described herein for the manufacture of a medicament for preventing dengue disease (in particular virologically confirmable dengue, VCD) in a subject.
  • dengue disease in particular virologically confirmable dengue, VCD
  • the present invention is directed in part to the use of a unit dose of the invention as described herein for the manufacture of a medicament for preventing dengue disease (in particular virologically confirmable dengue, VCD) in a subject population.
  • dengue disease in particular virologically confirmable dengue, VCD
  • the present invention is therefore directed to the use of a tetravalent dengue virus composition including four live attenuated dengue virus strains representing serotype 1, serotype 2, serotype 3 and serotype 4 in the manufacture of a medicament for the method of inoculating a subject against virologically confirmable dengue disease, wherein in particular the tetravalent dengue virus composition includes a chimeric dengue serotype 2/1 strain and a dengue serotype 2 strain and a chimeric dengue serotype 2/3 strain and a chimeric dengue serotype 2/4 strain, wherein in particular the dengue serotype 2 strain is derived from the wild type virus strain DEN-2 16681 (SEQ ID NO 11) and differs in at least three nucleotides from the wild type as follows:
  • chimeric dengue strains are derived from the serotype 2 strain by replacing the structural proteins prM and E from serotype 2 strain with the corresponding structural proteins from the other dengue serotypes, resulting in the following chimeric dengue strains:
  • the present invention is in particular directed to such use wherein the tetravalent dengue virus composition is in the form of a unit dose comprising:
  • a dengue serotype 1 in a concentration of at least 3.3 log 10 pfu/0.5 ml
  • a dengue serotype 2 in a concentration of at least 2.7 log 10 pfu/0.5 ml
  • a dengue serotype 3 in a concentration of at least 4.0 log 10 pfu/0.5 ml
  • a dengue serotype 4 in a concentration of at least 4.5 log 10 pfu/0.5 ml.
  • the present invention is in particular directed to such use wherein the unit dose is lyophilized and upon reconstitution with 0.5 mL of a pharmaceutically acceptable diluent comprises:
  • a dengue serotype 1 in a concentration of at least 3.3 log 10 pfu/0.5 ml
  • a dengue serotype 2 in a concentration of at least 2.7 log 10 pfu/0.5 ml
  • a dengue serotype 3 in a concentration of at least 4.0 log 10 pfu/0.5 ml
  • a dengue serotype 4 in a concentration of at least 4.5 log 10 pfu/0.5 ml.
  • the present invention is also in particular directed to such use wherein upon reconstitution with a pharmaceutically acceptable diluent (i), (ii), (iii), and (iv) provide a total concentration of pfu/0.5 mL and based on said total concentration the concentration of (ii) in pfu/0.5 mL is less than 10%, and the concentration of (iv) in pfu/0.5 mL is at least 50%, and the concentration of (i) in pfu/0.5 mL is at least 1%, and the concentration of (iii) in pfu/0.5 mL is at least 8%, or at least 10%, or at least 12%, or at least 14%, or at least 16%, or at least 18%, and wherein the subject is preferably 2 to 17 years of age or 4 to 16 years of age.
  • the present invention is also in particular directed to such use wherein upon reconstitution with a pharmaceutically acceptable diluent (i), (ii), (iii), and (iv) provide a total concentration of pfu/0.5 mL and based on said total concentration the concentration of (ii) in pfu/0.5 mL is less than 2%, the concentration of (iv) in pfu/0.5 mL is at least 50%, the concentration of (i) in pfu/0.5 mL is at least 1%, and the concentration of (iii) in pfu/0.5 mL is at least 6% and wherein the subject preferably is 18 to 60 years of age.
  • Such method preferably provides a combined vaccine efficacy against all four serotypes in preventing virologically confirmable dengue disease with a 2-sided 95% confidence interval, wherein the lower bound is more than 60%, when measured against placebo in a subject population of at least 5,000 healthy subjects irrespective of serostatus at baseline and 14 to 16 years of age, from the first administration of the administration schedule until 18 months after the second administration of the administration schedule.
  • Such method also preferably provides a combined vaccine efficacy against all four serotypes, in preventing virologically confirmable dengue disease with a 2-sided 95% confidence interval, wherein the lower bound is more than 45%, when measured against placebo in a subject population of at least 1,500 or at least 2,000 healthy subjects seronegative against all serotypes at baseline and 14 to 16 years of age, from 30 days after the second administration of the administration schedule until 18 months after the second administration of the administration schedule.
  • the use is for a method of inoculation against the virologically confirmable dengue disease is due to a dengue serotype 2, and/or due to a dengue serotype 1.
  • the method has very high efficacy against dengue serotype 2 and dengue serotype 1 and the highest efficacy against dengue serotype 2.
  • the invention is directed to the use wherein said methods have a vaccine efficacy against serotype 1, in preventing virologically confirmable dengue disease with a 2-sided 95% confidence interval, wherein the lower bound is more than 25%, when measured against placebo in a subject population of at least 1,500, or at least 2,000, or at least 5,000 healthy subjects (or at least 10,000, or at least 15,000 healthy subjects) irrespective of serostatus at baseline and 4 to 16 years of age from 30 days post second administration until 12 to 18 months (e.g. at 12 or at 18 months) after the second administration.
  • the lower bound is more than 30%, is more than 35% is more than 40%, is more than 45%, is more than 50%, or is more than 54%.
  • the subject population of at least 1,500 is seronegative against all serotypes at base line and the lower bound is more than 35%.
  • the seronegative and seropositive population each provide a vaccine efficacy against serotype 1 with a 2-sided 95% confidence interval, wherein the lower bounds are within 10%-points.
  • the invention is directed to said use wherein said methods have a vaccine efficacy against serotype 1, in preventing virologically confirmable dengue disease, when measured against placebo in a subject population of at least 1,500, or at least 2,000, or at least 5,000 healthy subjects (or at least 10,000, or at least 15,000 healthy subjects) irrespective of serostatus at baseline and 4 to 16 years of age from 30 days post second administration until 12 to 18 months (e.g. at 12 or at 18 months) after the second administration.
  • the vaccine efficacy is more than 40%, is more than 50%, is more than 60%, or is more than 65%.
  • the subject population of at least 1,500 is seronegative against all serotypes at base line.
  • the seronegative and seropositive population each provide a vaccine efficacy against serotype 1 which are within 5%-points.
  • the invention is directed to said use wherein said methods have a vaccine efficacy against serotype 2, in preventing virologically confirmable dengue disease with a 2-sided 95% confidence interval, wherein the lower bound is more than 25%, when measured against placebo in a subject population of at least 1,500, or at least 2,000, or at least 5,000 healthy subjects (or at least 10,000, or at least 15,000 healthy subjects) irrespective of serostatus at baseline and 4 to 16 years of age from 30 days post second administration until 12 to 18 months (e.g. at 12 or at 18 months) after the second administration.
  • the lower bound is more than 50%, is more than 60%, is more than 70%, is more than 80%, or is more than 85%.
  • the subject population of at least 1,500 is seronegative against all serotypes.
  • the seronegative and seropositive population each provide a vaccine efficacy against serotype 2 with a 2-sided 95% confidence interval, wherein the lower bounds are within 5%-points.
  • the invention is directed to said use wherein said methods have a vaccine efficacy against serotype 2, in preventing virologically confirmable dengue disease, when measured against placebo in a subject population of at least 1,500, or at least 2,000, or at least 5,000 healthy subjects (or at least 10,000, or at least 15,000 healthy subjects) irrespective of serostatus at baseline and 4 to 16 years of age from 30 days post second administration until 12 to 18 months (e.g. at 12 or at 18 months) after the second administration.
  • the vaccine efficacy is more than 60%, is more than 70%, is more than 80%, or is more than 90%.
  • the subject population of at least 1,500 is seronegative against all serotypes at base line.
  • the seronegative and seropositive population each provide a vaccine efficacy against serotype 2 which are within 5%-points.
  • the unit dose is reconstituted and administered by subcutaneous injection.
  • the subcutaneous injection is administered to the arm, preferably to the deltoid region of the arm.
  • such use is directed to a method which does not include a step of determination of a previous dengue infection in the subjects preferably at any time before, during or after the steps of administration or wherein the serostatus of the subject is unknown preferably at any time before, during or after the steps of administration.
  • the method according to the invention does not require the testing of the serostatus before vaccination and thus allows immediate treatment and outbreak control.
  • the use is for a method wherein the subject is exposed to a dengue outbreak.
  • the outbreak is due to a dengue serotype 2, and/or due to a serotype 1.
  • the subject is from a region wherein the seroprevalence rate is unknown and/or wherein the seroprevalence rate is below 80%, or below 70%, or below 60%.
  • the subject is seronegative at baseline and is from a region or travels to a region wherein the seroprevalence rate is high with respect to serotype 1 and/or serotype 2 i.e. 80%, or 90% or above.
  • the vaccine and corresponding method is safe for seronegative and seropositive subjects and thus does not require an analysis of the serostatus or a determination of a previous dengue infection or a high seroprevalence rate in the region.
  • Such a method preferably provides a combined vaccine efficacy against virologically-confirmed dengue with hospitalization against all four serotypes with a 2-sided 95% confidence interval, wherein the lower bound is more than 65%, when measured against placebo in a subject population of at least 5,000 healthy 4 to 16 year old subjects irrespective of serostatus at baseline, preferably in at least 1,500 healthy 4 to 16 year old subjects seronegative at baseline, from first administration of the administration schedule until 12 to 18 months after the second administration of the administration schedule.
  • the 2-sided 95% confidence interval of the combined vaccine efficacy against virologically-confirmed dengue with hospitalization against all four serotypes when comparing seropositive and seronegative subjects provides for lower bounds of the 2-sided confidence interval which are within 10% points or within 15% points or within 20% points.
  • the method is preferably safe with respect to serotype 1 and serotype 2 which may therefore be used in outbreak situations due to serotype 1 and/or serotype 2 or even for seronegative subjects (e.g. travelers) or subjects with unknown serostatus in regions with very high seroprevalence rates (>80%) due to serotype 1 and/or serotype 2.
  • such a method does not include the active surveillance with respect to febrile illness of the subject after the administration of the first- and second-unit dose.
  • active surveillance any subject with febrile illness (defined as fever 38° C. on any 2 of 3 consecutive days) will be asked to return to the site for dengue fever evaluation by the Investigator.
  • Subjects/guardians will be contacted at least weekly to ensure robust identification of febrile illness by reminding subjects/guardians of their obligation to return to the site in case of febrile illness. This contact will be implemented through appropriate methods that may differ in each trial site (eg, phone calls, text messaging, home visits, school-based surveillance).
  • such a method does not include vaccine immunogenicity analysis including GMTs for dengue neutralizing antibodies.
  • Such a method does not include a reactogenicity analysis.
  • a reactogenicity analysis relates to solicited local AEs (injection site pain, injection site erythema, and injection site swelling) and solicited systemic AEs (child ⁇ 6 years: fever, irritability/fussiness, drowsiness and loss of appetite; child 6 years: asthenia, fever, headache, malaise and myalgia) which will e.g. be assessed for 7 days and 14 days, respectively, following each vaccination (vaccination day included) via collection of diary cards.
  • the method does not include an active surveillance, an immunogenicity analysis and a reactogenicity analysis.
  • the vaccine and the corresponding method of inoculation are safe and therefore do not require further steps of surveillance or analysis.
  • A selecting a subject for administration of the unit doses of the tetravalent dengue virus composition in need for protection against dengue infection without determination of a previous dengue infection, and (B) administering a first unit dose of the tetravalent dengue virus composition to the subject, and (C) administering a second unit dose of the tetravalent dengue virus composition to the subject within 3 months of administration of the first unit dose. Therefore the method of inoculating is finalized without determination of a previous dengue infection.
  • the method however further optionally comprises at least 1 years after the administration of the second unit dose a booster dose of the unit dose.
  • Selecting the subject may include all types of considerations but preferably not the determination of a previous dengue infection.
  • the selection may include consideration of the age, health conditions, and threat of infection.
  • the threat of infection includes consideration of the seroprevalence rate in the region in which the subject normally lives or intends to travel, the serotype specific seroprevalence rate and an outbreak situation or serotype specific outbreak situations.
  • the subject may be selected due to its exposure to serotype 1 and/or serotype 2 or due to the fact it requires protection against a specific dengue serotype, i.e. serotype 1 and/or serotype 2.
  • the method is applicable to subjects of all kinds of ages.
  • the subject is under 9 years of age, or 4 to 5 years of age, or 6 to 11 years of age or 12 to 16 years, or 6 to 16 years of age or 4 to 16 years of age, or 2 to 17 years of age, or 9 years of age, or over 9 years of age, or 9 to 17 years of age, or 18 to 45 years of age, or 46 to 60 years of age, or over 60 years of age.
  • the unit dose of the tetravalent dengue virus composition comprising (i), (ii), (iii), and (iv) upon reconstitution with a pharmaceutically acceptable diluent provides a total concentration of pfu/0.5 mL of (i), (ii), (iii), and (iv) and based on said total concentration the concentration of (ii) in pfu/0.5 mL is preferably less than 10%, and the concentration of (iv) in pfu/0.5 mL is preferably at least 50%, and the concentration of (i) in pfu/0.5 mL is preferably at least 1%, and the concentration of (iii) in pfu/0.5 mL is preferably at least 8%, or more preferred at least 10%, or at least 12%, or at least 14%, or at least 16%, or at least 18% and the subject is 2 to 17 years of age or 4 to 16 years of age.
  • the unit dose of the tetravalent dengue virus composition comprising (i), (ii), (iii), and (iv) upon reconstitution with a pharmaceutically acceptable diluent provides a total concentration of pfu/0.5 mL of (i), (ii), (iii), and (iv) and based on said total concentration the concentration of (ii) in pfu/0.5 mL is preferably less than 2%, the concentration of (iv) in pfu/0.5 mL is preferably at least 50%, the concentration of (i) in pfu/0.5 mL is at preferably least 1%, and the concentration of (iii) in pfu/0.5 mL is preferably at least 6% and the subject is 18 to 60 years of age.
  • Method of preventing Any method described herein above under the heading “Method of preventing” is to be understood to be also disclosed as the use of a unit dose for the manufacture of a medicament for preventing dengue disease in a subject or subject population with such a method irrespective of whether it is expressly stated below.
  • the present invention is in part directed to the use of a unit dose of the invention as described herein for the manufacture of a medicament for preventing dengue disease in a subject population, comprising administering to the subject population at least a first reconstituted unit dose of the invention as described herein, wherein certain ratios of geometric mean neutralizing antibody titers (GMTs) at day 180 or 365 after administration of said first unit dose to the subject population are achieved.
  • GCTs geometric mean neutralizing antibody titers
  • the geometric mean neutralizing antibody titer for dengue serotype 2 (GMT DENV-2) and the geometric mean neutralizing antibody titer for dengue serotype 4 (GMT DENV-4) when tested in at least 40, or at least 50, or at least 60 subjects at day 180 or day 365 after at least a first administration of said reconstituted unit dose of the invention as described herein, and optionally a second administration of a reconstituted unit dose of the invention as described herein 90 days after said first administration, provide a ratio of GMT DENV-2:GMT DENV-4 of not more than 50, or not more than 40, or not more than 30, or not more than 20.
  • the ratio of GMT DENV-2:GMT DENV-1 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose
  • the ratio of GMT DENV-2:GMT DENV-3 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose.
  • the present invention is in part directed to the use of a unit dose of the invention as described herein for the manufacture of a medicament for preventing dengue disease in a subject, comprising administering to the subject at least a first reconstituted unit dose of the invention as described herein, wherein certain ratios of neutralizing antibody titers at day 180 or 365 after administration of said first unit dose to the subject are achieved.
  • the neutralizing antibody titer for dengue serotype 2 and the neutralizing antibody titer for dengue serotype 4 at day 180 or day 365 after at least a first administration of the reconstituted unit dose of the invention as described herein, and optionally a second administration of a reconstituted unit dose of the invention as described herein 90 days after said first administration provide a ratio of neutralizing antibody titer for DENV-2:neutralizing antibody titer for GMT DENV-4 of not more than 50, or not more than 40, or not more than 30, or not more than 20.
  • the ratio of the neutralizing antibody titers of DENV-2:DENV-1 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose, and/or the ratio of the neutralizing antibody titers of DENV-2:DENV-3 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose.
  • the geometric mean neutralizing antibody titers (GMTs) of a subject population or the neutralizing antibody titers of a subject are determined in accordance with the microneutralization test disclosed herein, for example according to the method described in Example 2.
  • the invention is directed to said uses, wherein the reconstituted unit dose of the invention as described herein is administered by subcutaneous injection.
  • the subcutaneous injection is administered to the arm, preferably to the deltoid region of the arm.
  • the invention is directed to said uses, wherein one reconstituted unit dose of the invention as described is administered by subcutaneous injection.
  • the subcutaneous injection is administered to the arm, preferably to the deltoid region of the arm.
  • the invention is directed to said uses, wherein two reconstituted unit doses of the invention as described herein are administered.
  • the two unit doses are administered within 12 months or more, or within six months, or within three months, such as at day 0 and day 90 or at day 1/0 and day 90.
  • a third unit dose of the invention as described herein may be administered after the second administration.
  • Such a third administration may act as a booster and may be administered between 6 to 12 months after the first administration, such as 12 months after the first administration, or later than 12 month after the first administration, such as 12 months (1 year) after the second administration or even 5 years or longer after the first or second administration.
  • the subject is seronegative with respect to all dengue serotypes.
  • the subject is seropositive with respect to at least one dengue serotype.
  • the invention is directed to said uses, wherein the reconstituted unit dose is administered to the seronegative subject by subcutaneous injection.
  • the invention is directed to said uses, wherein the reconstituted unit dose is administered to a subject of unknown serostatus and/or wherein no test has been carried out to determine whether the subject is seropositive or seronegative before the unit dose is administered.
  • the invention is directed to said uses, wherein the reconstituted unit dose of the invention as described herein is administered to a subject or subject population from a dengue endemic region.
  • the subject or subject population is from Singapore, Dominican Republic, Panama, Philippines, Colombia, Puerto Rico or Thailand, in particular from Singapore, Dominican Republic, Panama, or Philippines.
  • the subject or subject population is from a dengue non-endemic region.
  • Such a subject population or subject may be vaccinated according to the invention in the context of traveling to a dengue endemic region.
  • the reconstituted unit dose of the invention as described herein is administered subcutaneously to a subject or subject population from a dengue endemic region or from a dengue non-endemic region.
  • the invention is directed to said uses, wherein the subject is of 2 to 60 years of age or more than 17 years, or more than 18 years, or 18 to 60 years of age. In certain embodiments the subject is 1 to 17 years of age, or less than 9 years of age, or less than 4 years of age or less than 2 years of age. In further embodiments, the subjects or subject population are adults of more than 21 years, or 21 to 60 years, or 21 to 45 years of age. According to some of these embodiments the subject is seronegative and from a dengue-endemic region.
  • the invention is directed to said uses, wherein the unit dose of the invention as described herein is administered to a pediatric subject or pediatric subject population of less than 2 years of age, preferably of 2 months to 2 years of age or 2 months to 1.5 years of age or 2 months to 1 year of age.
  • the pediatric subject or pediatric subject population is seronegative and from a dengue endemic region.
  • the invention is directed to said uses, wherein the reconstituted unit dose of the invention as described herein is administered subcutaneously to a pediatric subject or pediatric subject population of less than 2 years of age, preferably of 2 months to 2 years of age or 2 months to 1.5 years of age or 2 months to 1 year of age.
  • the pediatric subject or pediatric subject population is seronegative and from a dengue endemic region.
  • the present invention is directed in part to the use of a unit dose of the invention as described herein for the manufacture of a medicament for preventing dengue disease in an elderly subject.
  • the present invention is directed in part to the use of a unit dose of the invention as described herein for the manufacture of a medicament for preventing dengue hemorrhagic fever (DHF) and dengue shock syndrome (DSS) in an elderly subject.
  • DHF dengue hemorrhagic fever
  • DSS dengue shock syndrome
  • the invention is directed to said uses, wherein the reconstituted unit dose of the invention as described herein is administered by subcutaneous injection.
  • the subcutaneous injection is administered to the arm, preferably to the deltoid region of the arm.
  • the invention is directed to said uses, wherein two reconstituted unit doses of the invention as described herein are administered.
  • the two unit doses are administered within 12 months or more, or within six months, or within three months, such as at day 0/1 and day 90.
  • a third unit dose of the invention as described herein may be administered after the second administration.
  • Such a third administration may act as a booster and may be administered between 6 to 12 months after the first administration, such as 12 months after the first administration, or later than 12 month after the first administration, such as 12 months (1 year) after the second administration or even 5 years or longer after the first or second administration.
  • the elderly subject is seronegative with respect to all dengue serotypes.
  • the elderly subject is seropositive with respect to at least one dengue serotype.
  • the invention is directed to said uses, wherein the reconstituted unit dose is administered to the seronegative elderly subject by subcutaneous injection.
  • the invention is directed to said uses, wherein the reconstituted unit dose of the invention as described herein is administered to an elderly subject from a dengue endemic region.
  • the elderly subject is from Singapore, Dominican Republic, Panama, Philippines, Colombia, Puerto Rico or Thailand, in particular from Singapore, Dominican Republic, Panama, or Philippines.
  • the elderly subject is from a dengue non-endemic region.
  • Such an elderly subject may be vaccinated according to the invention in the context of traveling to a dengue endemic region.
  • the reconstituted unit dose of the invention as described herein is administered subcutaneously to an elderly subject from a dengue endemic region or from a dengue non-endemic region.
  • the invention is directed to said uses, wherein the elderly subject has at least one chronic condition or disease.
  • the at least one chronic condition or disease may be selected from diabetes, hypertension, allergies, previous strokes, ischemic heart disease, chronic renal impairment and chronic obstructive pulmonary disease.
  • the invention is directed to said uses, wherein the elderly subject has an impaired immune system.
  • the present invention is directed in part to a method of preventing dengue disease as well as yellow fever in a subject.
  • the invention is directed to a method of preventing dengue disease in a subject, comprising administering to the subject a reconstituted unit dose of the invention as described herein, wherein the method further comprises preventing yellow fever in the subject by concomitant administration of a yellow fever vaccine, in particular YF-17D, to the subject.
  • a yellow fever vaccine in particular YF-17D
  • the present invention is directed in part to a method of preventing dengue disease as well as yellow fever in a subject population.
  • the invention is directed to a method of preventing dengue disease in a subject population, comprising administering to the subject population a reconstituted unit dose of the invention as described herein, wherein the method further comprises preventing yellow fever in the subject population by concomitant administration of a yellow fever vaccine, in particular YF-17D, to the subject population.
  • the present invention is in part directed to said method for preventing dengue disease and yellow fever in a subject population comprising administering to the subject population at least a first reconstituted unit dose of the invention as described herein, wherein certain ratios of geometric mean neutralizing antibody titers (GMTs) at day 180 or 365 after administration of said first unit dose to the subject population are achieved, and concomitantly administering a yellow fever vaccine, in particular YF-17D, to the subject population.
  • GTTs geometric mean neutralizing antibody titers
  • the geometric mean neutralizing antibody titer for dengue serotype 2 (GMT DENV-2) and the geometric mean neutralizing antibody titer for dengue serotype 4 (GMT DENV-4) when tested in at least 40, or at least 50, or at least 60 subjects at day 180 or day 365 after at least a first administration of said reconstituted unit dose of the invention as described herein, and optionally a second administration of a reconstituted unit dose of the invention as described herein 90 days after said first administration, provide a ratio of GMT DENV-2:GMT DENV-4 of not more than 50, or not more than 40, or not more than 30, or not more than 20.
  • the ratio of GMT DENV-2:GMT DENV-1 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose
  • the ratio of GMT DENV-2:GMT DENV-3 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose.
  • the present invention is in part directed to said method for preventing dengue disease and yellow fever in a subject comprising administering to the subject at least a first reconstituted unit dose of the invention as described herein, wherein certain ratios of neutralizing antibody titers at day 180 or 365 after administration of said first unit dose to the subject are achieved, and concomitantly administering a yellow fever vaccine, in particular YF-17D, to the subject.
  • the neutralizing antibody titer for dengue serotype 2 and the neutralizing antibody titer for dengue serotype 4 at day 180 or day 365 after at least a first administration of the reconstituted unit dose of the invention as described herein, and optionally a second administration of a reconstituted unit dose of the invention as described herein 90 days after said first administration provide a ratio of neutralizing antibody titer for DENV-2:neutralizing antibody titer for DENV-4 of not more than 50, or not more than 40, or not more than 30, or not more than 20.
  • the ratio of the neutralizing antibody titers of DENV-2:DENV-1 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose, and/or the ratio of the neutralizing antibody titers of DENV-2:DENV-3 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose.
  • the geometric mean neutralizing antibody titers (GMTs) of a subject population or the neutralizing antibody titers of a subject are determined in accordance with a microneutralization test, for example according to the method described in Example 2.
  • the invention is directed to said methods, wherein the unit dose of the invention as described herein and the yellow fever vaccine, in particular YF-17D, are administered simultaneously.
  • the simultaneous administration is on day 0 or day 90, preferably on day 0.
  • the administration of the unit dose of the invention as described herein and the yellow fever vaccine, in particular YF-17D are done sequentially.
  • the invention is directed to said methods, wherein the reconstituted unit dose of the invention as described herein is administered and the yellow fever vaccine, in particular YF-17D, are administered by subcutaneous injection.
  • the subcutaneous injections are administered to the arm, preferably to the deltoid region of the arm.
  • the subcutaneous injections of the unit dose of the invention as described herein and yellow fever vaccine, in particular YF-17D are administered to different anatomical sites, such as to opposite arms, in particular when the vaccines are administered simultaneously.
  • the invention is directed to said methods, wherein two unit doses of the invention as described herein are administered.
  • the two unit doses of the invention as described herein are administered within 12 month or more, or within 6 month, or within three months, such as at day 0/1 and day 90.
  • a further third unit dose of the invention as described herein is administered after the second.
  • Such a third administration may act as a booster and may be administered between 6 to 12 months after the first administration, such as 12 months after the first administration, or later than 12 month after the first administration, such as 12 months (1 year) after the second administration or even 5 years or longer after the first or second administration.
  • the invention is directed to said methods, wherein two reconstituted unit doses of the invention as described herein and one dose of a yellow fever vaccine, in particular YF-17D, are administered, in particular according to the following schedule
  • the invention is directed to said methods, wherein two reconstituted unit doses of the invention as described herein and one dose of a yellow fever vaccine, in particular YF-17D, are administered, in particular according to the following schedule
  • the invention is directed to said methods, wherein two reconstituted unit doses of the invention as described herein and one dose of a yellow fever vaccine, in particular YF-17D, are administered, in particular according to the following schedule
  • the yellow fever vaccine and unit dose of the invention as described herein are administered simultaneously on day 0 or simultaneously on day 90.
  • the invention is directed to said methods, wherein the subject or subject population is seronegative to all dengue serotypes.
  • the invention is directed to said methods, wherein the reconstituted unit dose of the invention as described herein is administered subcutaneously to a subject or subject population and the yellow fever vaccine, in particular YF-17D vaccine, is administered subcutaneously to a subject or subject population, and wherein the subject or the subject population is seronegative with respect to all dengue serotypes.
  • the subject or subject population is seropositive with respect to at least one dengue serotype.
  • the invention is directed to said methods, wherein the unit dose of the invention as described herein and the yellow fever vaccine, in particular YF-17D, are administered to a subject or subject population from a dengue endemic region.
  • the reconstituted unit dose of the invention as described herein and the yellow fever vaccine, in particular YF-17D are administered subcutaneously to a subject or subject population from a dengue endemic region.
  • the subject or subject population is from a dengue non-endemic region.
  • Such a subject population or such a subject may be vaccinated according to the present invention in the context of traveling to a dengue endemic region and yellow fever endemic region.
  • the invention is directed to said methods, wherein the reconstituted unit dose of the invention as described herein and of the yellow fever vaccine, in particular YF-17D, are administered subcutaneously to a subject or subject population of more than 17 years, or more than 18 years, or 18 to 60 years of age.
  • the subjects or subject population are adults of more than 21 years, or 21 to 60 years, or 21 to 45 years of age.
  • the subject or subject population is from a dengue endemic region.
  • the subject or subject population is from a dengue non-endemic region, preferably from a dengue non-endemic and yellow fever non-endemic region.
  • the subject or subject population are seronegative for all four dengue serotypes.
  • the present invention is directed in part to the unit dose of the invention as described herein for use in a method of preventing dengue disease in a subject, wherein the method also comprises the prevention of yellow fever in the subject with a yellow fever vaccine, in particular YF-17D.
  • the present invention is directed in part to a unit dose of a dengue vaccine composition as described herein and a yellow fever vaccine, in particular YF-17D, for use in a method of preventing dengue disease and yellow fever in a subject, respectively.
  • the present invention is directed in part to the unit dose of the invention as described herein for use in a method of preventing dengue disease in a subject population, wherein the method also comprises the prevention of yellow fever in the subject population with a yellow fever vaccine, in particular YF-17D.
  • the present invention is directed in part to a unit dose of a dengue vaccine composition as described herein and a yellow fever vaccine, in particular YF-17D, for use in a method of preventing dengue disease and yellow fever in a subject population, respectively.
  • the present invention is in part directed to the unit dose of a dengue vaccine composition as described herein and the yellow fever vaccine, in particular YF-17D, for use in a method of preventing dengue disease and yellow fever in a subject population, comprising administering to the subject population at least a first reconstituted unit dose of the invention as described herein, wherein certain ratios of geometric mean neutralizing antibody titers (GMTs) at day 180 or 365 after administration of said first unit dose to the subject population are achieved, and concomitantly administering a yellow fever vaccine, in particular YF-17D, to the subject population.
  • GTTs geometric mean neutralizing antibody titers
  • the geometric mean neutralizing antibody titer for dengue serotype 2 (GMT DENV-2) and the geometric mean neutralizing antibody titer for dengue serotype 4 (GMT DENV-4) when tested in at least 40, or at least 50, or at least 60 subjects at day 180 or day 365 after at least a first administration of said reconstituted unit dose of the invention as described herein, and optionally a second administration of a reconstituted unit dose of the invention as described herein 90 days after said first administration, provide a ratio of GMT DENV-2:GMT DENV-4 of not more than 50, or not more than 40, or not more than 30, or not more than 20.
  • the ratio of GMT DENV-2:GMT DENV-1 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose
  • the ratio of GMT DENV-2:GMT DENV-3 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose.
  • the present invention is in part directed to the unit dose of a dengue vaccine composition as described herein and the yellow fever vaccine, in particular YF-17D, for use in a method of preventing dengue disease and yellow fever in a subject comprising administering to the subject at least a first reconstituted unit dose of the invention as described herein, wherein certain ratios of neutralizing antibody titers at day 180 or 365 after administration of said first unit dose to the subject are achieved, and concomitantly administering a yellow fever vaccine, in particular YF-17D, to the subject.
  • the neutralizing antibody titer for dengue serotype 2 and the neutralizing antibody titer for dengue serotype 4 at day 180 or day 365 after at least a first administration of the reconstituted unit dose of the invention as described herein, and optionally a second administration of a reconstituted unit dose of the invention as described herein 90 days after said first administration provide a ratio of neutralizing antibody titer for DENV-2:neutralizing antibody titer for DENV-4 of not more than 50, or not more than 40, or not more than 30, or not more than 20.
  • the ratio of the neutralizing antibody titers of DENV-2:DENV-1 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose, and/or the ratio of the neutralizing antibody titers of DENV-2:DENV-3 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose.
  • the geometric mean neutralizing antibody titers (GMTs) of a subject population or the neutralizing antibody titers of a subject are determined in accordance with a microneutralization test, for example according to the method described in Example 2.
  • the invention is directed to the unit dose of the invention as described herein and the yellow fever vaccine, in particular YF-17D, for said uses, wherein said unit dose and said yellow fever vaccine are administered simultaneously.
  • the simultaneous administration is on day 0 or day 90, preferably on day 0.
  • the administration of said unit dose and said yellow fever vaccine, in particular YF-17D are done sequentially.
  • the invention is directed to the unit dose of the invention as described herein and the yellow fever vaccine, in particular YF-17D, for said uses, wherein said unit dose is administered by subcutaneous injection and wherein said yellow fever vaccine is administered by subcutaneous injection.
  • the subcutaneous injections are administered to the arm, preferably to the deltoid region of the arm.
  • the subcutaneous injection of said unit dose and the subcutaneous injection of said yellow fever vaccine are administered to different anatomical sites, such as to opposite arms, in particular when the vaccines are administered simultaneously.
  • the invention is directed to the unit dose of the invention as described herein and the yellow fever vaccine, in particular YF-17D, for said uses, wherein two reconstituted unit doses of the invention as described herein are administered.
  • the two unit doses of the invention as described herein are administered within 12 months or more, or within six months, or within three months, such as at day 0/1 and day 90.
  • a further third reconstituted unit dose of the invention as described herein is administered after the second administration.
  • Such a third administration may act as a booster and may be administered between 6 to 12 months after the first administration, such as 12 months after the first administration, or later than 12 month after the first administration, such as 12 months (1 year) after the second administration or even 5 years or longer after the first or second administration.
  • the invention is directed to the unit dose of the invention as described herein and the yellow fever vaccine, in particular YF-17D, for said uses, wherein two reconstituted unit doses of the invention as described herein and one dose of the yellow fever vaccine are administered, in particular according to the following schedule
  • the invention is directed to the unit dose of the invention as described herein and the yellow fever vaccine, in particular YF-17D, for said uses, wherein two unit doses of the invention as described herein and one dose of the yellow fever vaccine are administered, in particular according to the following schedule
  • the invention is directed to the unit dose of the invention as described herein and the yellow fever vaccine, in particular YF-17D, for said uses, wherein two unit doses of the invention as described herein and one dose of the yellow fever vaccine are administered, in particular according to the following schedule
  • the yellow fever vaccine and unit dose of the invention as described herein are administered simultaneously on day 0 or simultaneously on day 90.
  • the invention is directed to the unit dose of the invention as described herein and the yellow fever vaccine, in particular YF-17D, for said uses, wherein the subject or subject population is seronegative to all dengue serotypes.
  • the invention is directed to the unit dose of the invention as described herein and the yellow fever vaccine, in particular YF-17D, for said uses, wherein said unit dose is reconstituted and administered subcutaneously to a subject or subject population and said yellow fever vaccine is administered subcutaneously to a subject or subject population, and wherein the subject or the subject population is seronegative with respect to all dengue serotypes.
  • the subject or subject population is seropositive with respect to at least one dengue serotype.
  • the invention is directed to the unit dose of the invention as described herein and the yellow fever vaccine, in particular YF-17D, for said uses, wherein said unit dose and said yellow fever vaccine are administered to a subject or subject population from a dengue endemic region.
  • the reconstituted unit dose of the invention as described herein and the yellow fever vaccine, in particular YF-17D are administered subcutaneously to a subject or subject population from a dengue endemic region.
  • the subject or subject population is from a dengue non-endemic region.
  • Such a subject population or such a subject may be vaccinated according to the present invention in the context of traveling to a dengue endemic region and yellow fever endemic region.
  • the invention is directed to the unit dose of the invention as described herein and the yellow fever vaccine, in particular YF-17D, for said uses, wherein the reconstituted unit dose of the invention as described herein and the yellow fever vaccine, in particular YF-17D, are administered subcutaneously to a subject or a subject population of more than 17 years, or more than 18 years, or 18 to 60 years of age.
  • the subject or subject population is from a dengue endemic region.
  • the subject or subject population is from a dengue non-endemic region, preferably from a dengue non-endemic and yellow fever non-endemic region.
  • the subject or subject population is seronegative for all four dengue serotypes.
  • the present invention is directed in part to the use of a unit dose of the invention as described herein for the manufacture of a medicament for preventing dengue disease in a subject, further comprising the use of a yellow fever vaccine, in particular YF-17D, for the manufacture of a medicament for preventing yellow fever in the subject.
  • the present invention is directed in part to a use of a unit dose of a dengue vaccine composition as described herein and a yellow fever vaccine, in particular YF-17D, for the manufacture of a medicament for preventing dengue disease and yellow fever in a subject, respectively.
  • the present invention is directed in part to the use of a unit dose of the invention as described herein for the manufacture of a medicament for preventing dengue disease in a subject population, further comprising the use of a yellow fever vaccine, in particular YF-17D, for the manufacture of a medicament for preventing yellow fever in the subject population.
  • the present invention is directed in part to a use of a unit dose of a dengue vaccine composition as described herein and a yellow fever vaccine, in particular YF-17D, for the manufacture of a medicament for preventing dengue disease and yellow fever in a subject population, respectively.
  • the present invention is in part directed to the use of a unit dose of the invention as described herein and a yellow fever vaccine, in particular YF-17D, for the manufacture of a medicament for preventing dengue disease and yellow fever in a subject population, comprising administering to the subject population at least a first reconstituted unit dose of the invention as described herein, wherein certain ratios of geometric mean neutralizing antibody titers (GMTs) at day 180 or 365 after administration of said first unit dose to the subject population are achieved, and concomitantly administering a yellow fever vaccine, in particular YF-17D, to the subject population.
  • GTTs geometric mean neutralizing antibody titers
  • the geometric mean neutralizing antibody titer for dengue serotype 2 (GMT DENV-2) and the geometric mean neutralizing antibody titer for dengue serotype 4 (GMT DENV-4) when tested in at least 40, or at least 50, or at least 60 subjects at day 180 or day 365 after at least a first administration of said reconstituted unit dose of the invention as described herein, and optionally a second administration of a reconstituted unit dose of the invention as described herein 90 days after said first administration, provide a ratio of GMT DENV-2:GMT DENV-4 of not more than 50, or not more than 40, or not more than 30, or not more than 20.
  • the ratio of GMT DENV-2:GMT DENV-1 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose
  • the ratio of GMT DENV-2:GMT DENV-3 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose.
  • the present invention is in part directed to the use of a unit dose of the invention as described herein and a yellow fever vaccine, in particular YF-17D, for the manufacture of a medicament for preventing dengue disease and yellow fever in a subject, comprising administering to the subject at least a first reconstituted unit dose of the invention as described herein, wherein certain ratios of neutralizing antibody titers at day 180 or 365 after administration of said first unit dose to the subject are achieved, and concomitantly administering a yellow fever vaccine, in particular YF-17D, to the subject.
  • the neutralizing antibody titer for dengue serotype 2 and the neutralizing antibody titer for dengue serotype 4 at day 180 or day 365 after at least a first administration of the reconstituted unit dose of the invention as described herein, and optionally a second administration of a reconstituted unit dose of the invention as described herein 90 days after said first administration provide a ratio of neutralizing antibody titer for DENV-2:neutralizing antibody titer for DENV-4 of not more than 50, or not more than 40, or not more than 30, or not more than 20.
  • the ratio of the neutralizing antibody titers of DENV-2:DENV-1 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose, and/or the ratio of the neutralizing antibody titers of DENV-2:DENV-3 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose.
  • the geometric mean neutralizing antibody titers (GMTs) of a subject population or the neutralizing antibody titers of a subject are determined in accordance with a microneutralization test, for example according to the method described in Example 2.
  • the invention is directed to said uses, wherein the unit dose of the invention as described herein and the yellow fever vaccine, in particular YF-17D, are administered simultaneously.
  • the simultaneous administration is on day 0 or day 90, preferably on day 0.
  • the administration of the unit dose of the invention as described herein and the yellow fever vaccine, in particular YF-17D are done sequentially.
  • the invention is directed to said uses, wherein the unit dose of the invention as described herein is reconstituted and administered by subcutaneous injection and wherein the yellow fever vaccine, in particular YF-17D, is administered by subcutaneous injection.
  • the injections are administered to the arm, preferably to the deltoid region of the arm.
  • the subcutaneous injection of the unit dose of the invention as described herein and of the yellow fever vaccine, in particular YF-17D are administered to different anatomical sites such as to opposite arms, in particular when the vaccines are administered simultaneously.
  • the invention is directed to said uses, wherein two reconstituted unit doses of the invention as described herein are administered.
  • the two unit doses of the invention as described herein are administered within 12 months or more, or within six months, or within three months, such as at day 0/1 and day 90.
  • a further third unit dose of the invention as described herein is administered after the second administration.
  • Such a third administration may act as a booster and may be administered between 6 to 12 months after the first administration, such as 12 months after the first administration, or later than 12 month after the first administration, such as 12 months (1 year) after the second administration or even 5 years or longer after the first or second administration.
  • the invention is directed to said uses, wherein two unit doses of the invention as described herein and one dose of a yellow fever vaccine, in particular YF-17D, are administered, in particular according to the following schedule
  • the invention is directed to said uses, wherein two unit doses of the invention as described herein and one dose of a yellow fever vaccine, in particular YF-17D, are administered, in particular according to the following schedule
  • the invention is directed to said uses, wherein two unit doses of the invention as described herein and one dose of a yellow fever vaccine, in particular YF-17D, are administered, in particular according to the following schedule
  • the invention is directed to said uses, wherein the subject or subject population is seronegative to all dengue serotypes.
  • the invention is directed to said uses, wherein the unit dose of the invention as described herein is reconstituted and administered subcutaneously to a subject or subject population and the yellow fever vaccine, in particular YF-17D, is administered subcutaneously to a subject or subject population, and wherein the subject or the subject population is seronegative with respect to all dengue serotypes.
  • the subject or subject population is seropositive with respect to at least one dengue serotype.
  • the invention is directed to said uses, wherein the unit dose of the invention as described herein and the yellow fever vaccine, in particular YF-17D, are administered to a subject or subject population from a dengue endemic region.
  • the reconstituted unit dose of the invention as described herein and the yellow fever vaccine, in particular YF-17D are administered subcutaneously to a subject or subject population from a dengue endemic region.
  • the subject or subject population is from a dengue non-endemic region.
  • Such a subject population or such a subject may be vaccinated according to the present invention in the context of traveling to a dengue endemic region and yellow fever endemic region.
  • the invention is directed to said uses, wherein the reconstituted unit dose of the invention as described herein and the yellow fever vaccine, in particular YF-17D, are administered subcutaneously to a subject or subject population of more than 17 years, or more than 18 years, or 18 to 60 years of age.
  • the subjects or subject population are adults of more than 21 years, or 21 to 60 years, or 21 to 45 years of age.
  • the subject or subject population is from a dengue endemic region.
  • the subject or subject population is from a dengue non-endemic region, preferably from a dengue non-endemic and yellow fever non-endemic region.
  • the subject or subject population is seronegative for all four dengue serotypes.
  • the present invention is directed in part to a method of preventing dengue disease as well as hepatitis A in a subject.
  • the invention is directed to a method of preventing dengue disease in a subject, comprising administering to the subject a reconstituted unit dose of the invention as described herein, wherein the method further comprises preventing hepatitis A in the subject by concomitant administration of a hepatitis A vaccine, such as HAVRIX® or VAQTA®, to the subject.
  • a hepatitis A vaccine such as HAVRIX® or VAQTA®
  • the present invention is directed in part to a method of preventing dengue disease as well as hepatitis A in a subject population.
  • the invention is directed to a method of preventing dengue disease in a subject population, comprising administering to the subject population a reconstituted unit dose of the invention as described herein, wherein the method further comprises preventing hepatitis A in the subject population by concomitant administration of a hepatitis A vaccine, such as HAVRIX® or VAQTA®, to the subject population.
  • a hepatitis A vaccine such as HAVRIX® or VAQTA®
  • the present invention is in part directed to said method for preventing dengue disease and hepatitis A in a subject population comprising administering to the subject population at least a first reconstituted unit dose of the invention as described herein, wherein certain ratios of geometric mean neutralizing antibody titers (GMTs) at day 180 or 365 after administration of said first unit dose to the subject population are achieved, and concomitantly administering a hepatitis A vaccine, such as HAVRIX® or VAQTA®, to the subject population.
  • GTTs geometric mean neutralizing antibody titers
  • the geometric mean neutralizing antibody titer for dengue serotype 2 (GMT DENV-2) and the geometric mean neutralizing antibody titer for dengue serotype 4 (GMT DENV-4) when tested in at least 40, or at least 50, or at least 60 subjects at day 180 or day 365 after at least a first administration of said reconstituted unit dose of the invention as described herein, and optionally a second administration of a reconstituted unit dose of the invention as described herein 90 days after said first administration, provide a ratio of GMT DENV-2:GMT DENV-4 of not more than 50, or not more than 40, or nor more than 30, or not more than 20.
  • the ratio of GMT DENV-2:GMT DENV-1 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose
  • the ratio of GMT DENV-2:GMT DENV-3 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose.
  • the present invention is in part directed to said method for preventing dengue disease and hepatitis A in a subject comprising administering to the subject at least a first reconstituted unit dose of the invention as described herein, wherein certain ratios of neutralizing antibody titers at day 180 or 365 after administration of said first unit dose to the subject are achieved, and concomitantly administering a hepatitis A vaccine, such as HAVRIX® or VAQTA®, to the subject.
  • a hepatitis A vaccine such as HAVRIX® or VAQTA®
  • the neutralizing antibody titer for dengue serotype 2 and the neutralizing antibody titer for dengue serotype 4 at day 180 or day 365 after at least a first administration of the reconstituted unit dose of the invention as described herein, and optionally a second administration of a reconstituted unit dose of the invention as described herein 90 days after said first administration provide a ratio of neutralizing antibody titer for DENV-2:neutralizing antibody titer for DENV-4 of not more than 50, or not more than 40, or not more than 30, or not more than 20.
  • the ratio of the neutralizing antibody titers of DENV-2:DENV-1 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose, and/or the ratio of the neutralizing antibody titers of DENV-2:DENV-3 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose.
  • the geometric mean neutralizing antibody titers (GMTs) of a subject population or the neutralizing antibody titers of a subject are determined in accordance with a microneutralization test, for example according to the method described in Example 2.
  • the invention is directed to said methods, wherein the unit dose of the invention as described herein and the hepatitis A vaccine, such as HAVRIX® or VAQTA®, are administered simultaneously.
  • the simultaneous administration is on day 0 or day 90, preferably on day 0.
  • the administration of the unit dose of the invention as described herein and the hepatitis A vaccine, such as HAVRIX® or VAQTA® are done sequentially.
  • the invention is directed to said methods, wherein the reconstituted unit dose of the invention as described herein is administered by subcutaneous injection and wherein the hepatitis A vaccine, such as HAVRIX® or VAQTA®, is administered by intramuscular injection.
  • the injections are administered to the arm, preferably to the deltoid region of the arm.
  • the subcutaneous injection of the reconstituted unit dose of the invention as described herein and the intramuscular injection of the hepatitis A vaccine, such as HAVRIX® or VAQTA® are administered to different anatomical sites, such as to opposite arms, in particular when the vaccines are administered simultaneously.
  • the invention is directed to said methods, wherein two unit doses of the invention as described herein are administered.
  • the two unit doses of the invention as described herein are administered within 12 month or more, or within 6 month, or within three months, such as at day 0/1 and day 90.
  • a further third unit dose of the invention as described herein is administered after the second administration.
  • Such a third administration may act as a booster and may be administered between 6 to 12 months after the first administration, such as 12 months after the first administration, or later than 12 month after the first administration, such as 12 months (1 year) after the second administration or even 5 years or longer after the first or second administration.
  • the invention is directed to said methods, wherein two reconstituted unit doses of the invention as described herein and one dose of a hepatitis A vaccine, such as HAVRIX® or VAQTA®, are administered, in particular according to the following schedule
  • the invention is directed to said methods, wherein the subject or subject population is seronegative to all dengue serotypes.
  • the invention is directed to said methods, wherein the reconstituted unit dose of the invention as described herein is administered subcutaneously to a subject or subject population and the hepatitis A vaccine, such as HAVRIX® or VAQTA®, is administered intramuscularly to a subject or subject population, and wherein the subject or the subject population is seronegative with respect to all dengue serotypes.
  • the subject or subject population is seropositive with respect to at least one dengue serotype.
  • the invention is directed to said methods, wherein the unit dose of the invention as described herein and the hepatitis A vaccine, such as HAVRIX® or VAQTA®, are administered to a subject or subject population from a dengue endemic region.
  • the reconstituted unit dose of the invention as described herein is administered subcutaneously and the hepatitis A vaccine, such as HAVRIX® or VAQTA®, is administered intramuscularly to a subject or subject population from a dengue endemic region.
  • a second dose of a hepatitis A vaccine such as HAVRIX® or VAQTA®
  • the second dose of the hepatitis A vaccine may be administered after the first administration of the hepatitis A vaccine.
  • Such a second administration may act as a booster and may be administered 9 months after the first administration of the hepatitis A vaccine, such as on day 270.
  • the invention is directed to said methods, wherein the reconstituted unit dose of the invention as described herein is administered subcutaneously and wherein the hepatitis A vaccine, such as HAVRIX® or VAQTA®, is administered intramuscularly to a subject or subject population of more than 17 years, or more than 18 years, or 18 to 60 years of age.
  • the subjects or subject population are adults of more than 21 years, or 21 to 60 years, or 21 to 45 years of age.
  • the subject or subject population is from a dengue endemic region.
  • the subject or subject population is from a dengue non-endemic region, preferably from a dengue non-endemic and a hepatitis A non-endemic region.
  • the subject or subject population is seronegative for all four dengue serotypes.
  • the present invention is directed in part to the unit dose of the invention as described herein for use in a method of preventing dengue disease in a subject, wherein the method also comprises the prevention of hepatitis A in the subject with a hepatitis A vaccine, such as HAVRIX® or VAQTA®.
  • the present invention is directed in part to a unit dose of a dengue vaccine composition as described herein and a hepatitis A vaccine, such as HAVRIX® or VAQTA®, for use in a method of preventing dengue disease and hepatitis A in a subject, respectively.
  • the present invention is directed in part to the unit dose of the invention as described herein for use in a method of preventing dengue disease in a subject population, wherein the method also comprises the prevention of hepatitis A in the subject population with a hepatitis A vaccine, such as HAVRIX® or VAQTA®.
  • the present invention is directed in part to a unit dose of a dengue vaccine composition as described herein and a hepatitis A vaccine, such as HAVRIX® or VAQTA®, for use in a method of preventing dengue disease and hepatitis A in a subject population, respectively.
  • the present invention is in part directed to the unit dose of a dengue vaccine composition as described herein and the hepatitis A vaccine, such as HAVRIX® or VAQTA®, for use in a method of preventing dengue disease and hepatitis A in a subject population, comprising administering to the subject population at least a first reconstituted unit dose of the invention as described herein, wherein certain ratios of geometric mean neutralizing antibody titers (GMTs) at day 180 or 365 after administration of said first unit dose to the subject population are achieved, and concomitantly administering a hepatitis A vaccine, such as HAVRIX® or VAQTA®, to the subject population.
  • GTTs geometric mean neutralizing antibody titers
  • the geometric mean neutralizing antibody titer for dengue serotype 2 (GMT DENV-2) and the geometric mean neutralizing antibody titer for dengue serotype 4 (GMT DENV-4) when tested in at least 40, or at least 50, or at least 60 subjects at day 180 or day 365 after at least a first administration of said reconstituted unit dose of the invention as described herein, and optionally a second administration of a reconstituted unit dose of the invention as described herein 90 days after said first administration, provide a ratio of GMT DENV-2:GMT DENV-4 of not more than 50, or not more than 40, or not more than 30, or not more than 20.
  • the ratio of GMT DENV-2:GMT DENV-1 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose
  • the ratio of GMT DENV-2:GMT DENV-3 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose.
  • the present invention is in part directed to the unit dose of a dengue vaccine composition as described herein and the hepatitis A vaccine, such as HAVRIX® or VAQTA®, for use in a method of preventing dengue disease and hepatitis A in a subject comprising administering to the subject a hepatitis A vaccine, such as HAVRIX® or VAQTA®, and at least a first reconstituted unit dose of the invention as described herein, wherein certain ratios of neutralizing antibody titers at day 180 or 365 after administration of said first unit dose to the subject are achieved, and concomitantly administering a hepatitis A vaccine, such as HAVRIX® or VAQTA®, to the subject.
  • a hepatitis A vaccine such as HAVRIX® or VAQTA®
  • the neutralizing antibody titer for dengue serotype 2 and the neutralizing antibody titer for dengue serotype 4 at day 180 or day 365 after at least a first administration of the reconstituted unit dose of the invention as described herein, and optionally a second administration of a reconstituted unit dose of the invention as described herein 90 days after said first administration provide a ratio of neutralizing antibody titer for DENV-2:neutralizing antibody titer for DENV-4 of not more than 50, or not more than 40, or not more than 30, or not more than 20, wherein the method further comprises preventing hepatitis A in the subject by concomitant administration of a hepatitis A vaccine, such as HAVRIX® or VAQTA®, to the subject.
  • a hepatitis A vaccine such as HAVRIX® or VAQTA®
  • the ratio of the neutralizing antibody titers of DENV-2:DENV-1 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose, and/or the ratio of the neutralizing antibody titers of DENV-2:DENV-3 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose.
  • the geometric mean neutralizing antibody titers (GMTs) of a subject population or the neutralizing antibody titers of a subject are determined in accordance with a microneutralization test, for example according to the method described in Example 2.
  • the invention is directed to the unit dose of the invention as described herein and the hepatitis A vaccine, such as HAVRIX® or VAQTA®, for said uses, wherein said unit dose and said hepatitis A vaccine are administered simultaneously.
  • the simultaneous administration is on day 0 or day 90, preferably on day 0.
  • the administration of said unit dose and said hepatitis A vaccine are done sequentially.
  • the invention is directed to the unit dose of the invention as described herein and the hepatitis A vaccine, such as HAVRIX® or VAQTA®, for said uses, wherein said unit dose is administered by subcutaneous injection and wherein said hepatitis A vaccine is administered by intramuscular injection.
  • the injections are administered to the arm, preferably to the deltoid region of the arm.
  • the subcutaneous injection of said unit dose and the intramuscular injection of said hepatitis A vaccine are administered to different anatomical sites, such as to opposite arms, in particular when the vaccines are administered simultaneously.
  • the invention is directed to the unit dose of the invention as described herein and the hepatitis A vaccine, such as HAVRIX® or VAQTA®, for said uses, wherein two reconstituted unit doses of the invention as described herein are administered.
  • the two unit doses of the invention as described herein are administered within 12 months or more, or within six months, or within three months, such as at day 0/1 and day 90.
  • a further third reconstituted unit dose of the invention as described herein is administered after the second administration.
  • Such a third administration may act as a booster and may be administered between 6 to 12 months after the first administration, such as 12 months after the first administration, or later than 12 month after the first administration, such as 12 months (1 year) after the second administration or even 5 years or longer after the first or second administration.
  • the invention is directed to the unit dose of the invention as described herein and the hepatitis A vaccine, such as HAVRIX® or VAQTA®, for said uses, wherein two reconstituted unit doses of the invention as described herein and one dose of the hepatitis A vaccine are administered, in particular according to the following schedule
  • the invention is directed to the unit dose of the invention as described herein and the hepatitis A vaccine, such as HAVRIX® or VAQTA®, for said uses, wherein the subject or subject population is seronegative to all dengue serotypes.
  • the invention is directed to the unit dose of the invention as described herein and the hepatitis A vaccine, such as HAVRIX® or VAQTA®, for said uses, wherein said unit dose is reconstituted and administered subcutaneously to a subject or subject population and said hepatitis A vaccine is administered intramuscularly to a subject or subject population, and wherein the subject or the subject population is seronegative with respect to all dengue serotypes.
  • the subject or subject population is seropositive with respect to at least one dengue serotype.
  • the invention is directed to the unit dose of the invention as described herein and the hepatitis A vaccine, such as HAVRIX® or VAQTA®, for said uses, wherein said unit dose and said hepatitis A vaccine are administered to a subject or subject population from a dengue endemic region.
  • the reconstituted unit dose of the invention as described herein is administered subcutaneously and the hepatitis A vaccine, such as HAVRIX® or VAQTA®, is administered intramuscularly to a subject or subject population from a dengue endemic region.
  • the invention is directed to the unit dose of the invention as described herein and the hepatitis A vaccine, such as HAVRIX® or VAQTA®, for said uses, wherein a further second dose of a hepatitis A vaccine, such as HAVRIX® or VAQTA®, is administered.
  • the second dose of the hepatitis A vaccine may be administered after the first administration of the hepatitis A vaccine.
  • Such a second administration may act as a booster and may be administered 9 months after the first administration of the hepatitis A vaccine, such as on day 270.
  • the invention is directed to the unit dose of the invention as described herein and the hepatitis A vaccine, such as HAVRIX® or VAQTA®, for said uses, wherein the reconstituted unit dose of the invention as described herein is administered subcutaneously and the hepatitis A vaccine, such as HAVRIX® or VAQTA®, is administered intramuscularly to a subject or subject population of more than 17 years, or more than 18 years, or 18 to 60 years of age.
  • the subjects or subject population are adults of more than 21 years, or 21 to 60 years, or 21 to 45 years of age.
  • the subject or subject population is from a dengue endemic region.
  • the subject or subject population is from a dengue non-endemic region, preferably from a dengue non-endemic and a hepatitis A non-endemic region. According to certain embodiments, the subject or subject population is seronegative for all four dengue serotypes.
  • the present invention is directed in part to the use of a unit dose of the invention as described herein for the manufacture of a medicament for preventing dengue disease in a subject, further comprising the use of a hepatitis A vaccine, such as HAVRIX® or VAQTA®, for the manufacture of a medicament for preventing hepatitis A in the subject.
  • a hepatitis A vaccine such as HAVRIX® or VAQTA®
  • the present invention is directed in part to a use of a unit dose of a dengue vaccine composition as described herein and a hepatitis A vaccine, such as HAVRIX® or VAQTA®, for the manufacture of a medicament for preventing dengue disease and hepatitis A in a subject, respectively.
  • the present invention is directed in part to the use of a unit dose of the invention as described herein for the manufacture of a medicament for preventing dengue disease in a subject population, further comprising the use of a hepatitis A vaccine, such as HAVRIX® or VAQTA®, for the manufacture of a medicament for preventing hepatitis A in the subject population.
  • a hepatitis A vaccine such as HAVRIX® or VAQTA®
  • the present invention is directed in part to a use of a unit dose of a dengue vaccine composition as described herein and a hepatitis A vaccine, such as HAVRIX® or VAQTA®, for the manufacture of a medicament for preventing dengue disease and hepatitis A in a subject population, respectively.
  • the present invention is in part directed to the use of a unit dose of the invention as described herein and a hepatitis A vaccine, such as HAVRIX® or VAQTA®, for the manufacture of a medicament for preventing dengue disease and hepatitis A in a subject population, comprising administering to the subject population at least a first reconstituted unit dose of the invention as described herein, wherein certain ratios of geometric mean neutralizing antibody titers (GMTs) at day 180 or 365 after administration of said first unit dose to the subject population are achieved and concomitantly administering a hepatitis A vaccine, such as HAVRIX® or VAQTA®, to the subject population.
  • GTTs geometric mean neutralizing antibody titers
  • the geometric mean neutralizing antibody titer for dengue serotype 2 (GMT DENV-2) and the geometric mean neutralizing antibody titer for dengue serotype 4 (GMT DENV-4) when tested in at least 40, or at least 50, or at least 60 subjects at day 180 or day 365 after at least a first administration of said reconstituted unit dose of the invention as described herein, and optionally a second administration of a reconstituted unit dose of the invention as described herein 90 days after said first administration, provide a ratio of GMT DENV-2:GMT DENV-4 of not more than 50, or not more than 40, or not more than 30, or not more than 20.
  • the ratio of GMT DENV-2:GMT DENV-1 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose
  • the ratio of GMT DENV-2:GMT DENV-3 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose.
  • the present invention is in part directed to the use of a unit dose of the invention as described herein and a hepatitis A vaccine, such as HAVRIX® or VAQTA®, for the manufacture of a medicament for preventing dengue disease and hepatitis A in a subject, comprising administering to the subject at least a first reconstituted unit dose of the invention as described herein, wherein certain ratios of neutralizing antibody titers at day 180 or 365 after administration of said first unit dose to the subject are achieved and concomitantly administering a hepatitis A vaccine, such as HAVRIX® or VAQTA®, to the subject.
  • a hepatitis A vaccine such as HAVRIX® or VAQTA®
  • the neutralizing antibody titer for dengue serotype 2 and the neutralizing antibody titer for dengue serotype 4 at day 180 or day 365 after at least a first administration of the reconstituted unit dose of the invention as described herein, and optionally a second administration of a reconstituted unit dose of the invention as described herein 90 days after said first administration provide a ratio of neutralizing antibody titer for DENV-2:neutralizing antibody titer for DENV-4 of not more than 50, or not more than 40, or not more than 30, or not more than 20.
  • the ratio of the neutralizing antibody titers of DENV-2:DENV-1 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose, and/or the ratio of the neutralizing antibody titers of DENV-2:DENV-3 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose.
  • the geometric mean neutralizing antibody titers (GMTs) of a subject population or the neutralizing antibody titers of a subject are determined in accordance with a microneutralization test, for example according to the method described in Example 2.
  • the invention is directed to said uses, wherein the unit dose of the invention as described herein and the hepatitis A vaccine, such as HAVRIX® or VAQTA®, are administered simultaneously.
  • the simultaneous administration is on day 0 or day 90, preferably on day 0.
  • the administration of the unit dose of the invention as described herein and the hepatitis A vaccine, such as HAVRIX® or VAQTA® are done sequentially.
  • the invention is directed to said uses, wherein the unit dose of the invention as described herein is reconstituted and administered by subcutaneous injection and wherein the hepatitis A vaccine, such as HAVRIX® or VAQTA®, is administered by intramuscular injection.
  • the injections are administered to the arm, preferably to the deltoid region of the arm.
  • the subcutaneous injection of the unit dose of the invention as described herein and the intramuscular injection of the hepatitis A vaccine, such as HAVRIX® or VAQTA® are administered to different anatomical sites, such as to opposite arms, in particular when the vaccines are administered simultaneously.
  • the invention is directed to said uses, wherein two reconstituted unit doses of the invention as described herein are administered.
  • the two unit doses of the invention as described herein are administered within 12 months or more, or within six months, or within three months, such as at day 0/1 and day 90.
  • a further third unit dose of the invention as described herein is administered after the second administration.
  • Such a third administration may act as a booster and may be administered between 6 to 12 months after the first administration, such as 12 months after the first administration, or later than 12 month after the first administration, such as 12 months (1 year) after the second administration or even 5 years or longer after the first or second administration.
  • the invention is directed to said uses, wherein two unit doses of the invention as described herein and one dose of a hepatitis A vaccine, such as HAVRIX® or VAQTA®, are administered, in particular according to the following schedule
  • the invention is directed to said uses, wherein the subject or subject population is seronegative to all dengue serotypes.
  • the invention is directed to said uses, wherein the unit dose of the invention as described herein is reconstituted and administered subcutaneously to a subject or subject population and the hepatitis A vaccine, such as HAVRIX® or VAQTA®, is administered intramuscularly to a subject or subject population, and wherein the subject or the subject population is seronegative with respect to all dengue serotypes.
  • the subject or subject population is seropositive with respect to at least one dengue serotype.
  • the invention is directed to said uses, wherein the unit dose of the invention as described herein and the hepatitis A vaccine, such as HAVRIX® or VAQTA®, are administered to a subject or subject population from a dengue endemic region.
  • the reconstituted unit dose of the invention as described herein is administered subcutaneously and the hepatitis A vaccine, such as HAVRIX® or VAQTA®, is administered intramuscularly to a subject or subject population from a dengue endemic region.
  • the invention is directed to said uses, wherein a further second dose of a hepatitis A vaccine, such as HAVRIX® or VAQTA®, is administered.
  • the second dose of the hepatitis A vaccine may be administered after the first administration of the hepatitis A vaccine.
  • Such a second administration may act as a booster and may be administered 9 months after the first administration of the hepatitis A vaccine such as on day 270.
  • the invention is directed to said uses, wherein the reconstituted unit dose of the invention as described herein is administered subcutaneously and the hepatitis A vaccine, such as HAVRIX® or VAQTA®, is administered intramuscularly to a subject or subject population of more than 17 years, or more than 18 years, or 18 to 60 years of age.
  • the subjects or subject population are adults of more than 21 years, or 21 to 60 years, or 21 to 45 years of age.
  • the subject or subject population is from a dengue endemic region.
  • the subject or subject population is from a dengue non-endemic region, preferably from a dengue non-endemic and hepatitis A non-endemic region.
  • the subject or subject population is seronegative for all four dengue serotypes.
  • the present invention is directed in part to a method of preventing dengue disease as well as HPV-associated cancers or genital warts in a subject.
  • the invention is directed to a method of preventing dengue disease in a subject, comprising administering to the subject a reconstituted unit dose of the invention as described herein, wherein the method further comprises preventing HPV-associated cancers or genital warts in the subject by concomitant administration of a HPV vaccine, in particular a 9vHPV vaccine, such as GARDASIL® 9, to the subject.
  • the present invention is directed in part to a method of preventing dengue disease as well as HPV-associated cancers or genital warts in a subject population.
  • the invention is directed to a method of preventing dengue disease in a subject population, comprising administering to the subject population a reconstituted unit dose of the invention as described herein, wherein the method further comprises preventing HPV-associated cancers or genital warts in the subject population by concomitant administration of a HPV vaccine, in particular a 9vHPV vaccine, such as GARDASIL® 9, to the subject population.
  • the present invention is in part directed to said method for preventing dengue disease and HPV-associated cancers or genital warts in a subject population comprising administering to the subject population at least a first reconstituted unit dose of the invention as described herein, wherein certain ratios of geometric mean neutralizing antibody titers (GMTs) at day 180 or 365 after administration of said first unit dose to the subject population are achieved, and concomitantly administering a HPV vaccine, in particular a 9vHPV vaccine, such as GARDASIL® 9, to the subject population.
  • GTTs geometric mean neutralizing antibody titers
  • the geometric mean neutralizing antibody titer for dengue serotype 2 (GMT DENV-2) and the geometric mean neutralizing antibody titer for dengue serotype 4 (GMT DENV-4) when tested in at least 40, or at least 50, or at least 60 subjects at day 180 or day 365 after at least a first administration of said reconstituted unit dose of the invention as described herein, and optionally a second administration of a reconstituted unit dose of the invention as described herein 90 days after said first administration, provide a ratio of GMT DENV-2:GMT DENV-4 of not more than 50, or not more than 40, or not more than 30, or not more than 20.
  • the ratio of GMT DENV-2:GMT DENV-1 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose
  • the ratio of GMT DENV-2:GMT DENV-3 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose.
  • the present invention is in part directed to said method for preventing dengue disease and HPV-associated cancers or genital warts in a subject comprising administering to the subject at least a first reconstituted unit dose of the invention as described herein, wherein certain ratios of neutralizing antibody titers at day 180 or 365 after administration of said first unit dose to the subject are achieved, and concomitantly administering a HPV vaccine, in particular a 9vHPV vaccine, such as GARDASIL® 9, to the subject.
  • a HPV vaccine in particular a 9vHPV vaccine, such as GARDASIL® 9
  • the neutralizing antibody titer for dengue serotype 2 and the neutralizing antibody titer for dengue serotype 4 at day 180 or day 365 after at least a first administration of the reconstituted unit dose of the invention as described herein, and optionally a second administration of a reconstituted unit dose of the invention as described herein 90 days after said first administration provide a ratio of neutralizing antibody titer for DENV-2:neutralizing antibody titer for DENV-4 of not more than 50, or not more than 40, or not more than 30, or not more than 20.
  • the ratio of the neutralizing antibody titers of DENV-2:DENV-1 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose, and/or the ratio of the neutralizing antibody titers of DENV-2:DENV-3 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose.
  • the geometric mean neutralizing antibody titers (GMTs) of a subject population or the neutralizing antibody titers of a subject are determined in accordance with a microneutralization test, for example according to the method described in Example 2.
  • the invention is directed to said methods, wherein the unit dose of the invention as described herein and the HPV vaccine, in particular a 9vHPV vaccine, such as GARDASIL® 9, are administered simultaneously.
  • the simultaneous administration is on day 0 or day 90, preferably on day 0.
  • the administration of the unit dose of the invention as described herein and the HPV vaccine, in particular a 9vHPV vaccine, such as GARDASIL® 9, are done sequentially.
  • the invention is directed to said methods, wherein the reconstituted unit dose of the invention as described herein is administered by subcutaneous injection and wherein the HPV vaccine, in particular a 9vHPV vaccine, such as GARDASIL® 9, is administered by intramuscular injection.
  • the injections are administered to the arm, preferably to the deltoid region of the arm.
  • the subcutaneous injection of the unit dose of the invention as described herein and the intramuscular injection of the HPV vaccine, in particular a 9vHPV vaccine, such as GARDASIL® 9 are administered to different anatomical sites, such as to opposite arms, in particular when the vaccines are administered simultaneously.
  • the invention is directed to said methods, wherein two unit doses of the invention as described herein are administered.
  • the two unit doses of the invention as described herein are administered within 12 month or more, or within 6 month, or within three months, such as at day 0/1 and day 90.
  • a further third unit dose of the invention as described herein is administered after the second.
  • Such a third administration may act as a booster and may be administered between 6 to 12 months after the first administration, such as 12 months after the first administration, or later than 12 month after the first administration, such as 12 months (1 year) after the second administration or even 5 years or longer after the first or second administration.
  • the invention is directed to said methods, wherein two reconstituted unit doses of the invention as described herein and two doses of a HPV vaccine, in particular a 9vHPV vaccine, such as GARDASIL® 9, are administered, in particular according to the following schedule
  • the invention is directed to said methods, wherein the subject or subject population is seronegative to all dengue serotypes.
  • the invention is directed to said methods, wherein the reconstituted unit dose of the invention as described herein is administered subcutaneously to a subject or subject population and the HPV vaccine, in particular a 9vHPV vaccine, such as GARDASIL® 9, is administered intramuscularly to a subject or subject population, and wherein the subject or the subject population is seronegative with respect to all dengue serotypes.
  • the subject or subject population is seropositive with respect to at least one dengue serotype.
  • the invention is directed to said methods, wherein the unit dose of the invention as described herein and the HPV vaccine, in particular a 9vHPV vaccine, such as GARDASIL® 9, are administered to a subject or subject population from a dengue endemic region.
  • the reconstituted unit dose of the invention as described herein is administered subcutaneously and the HPV vaccine, in particular a 9vHPV vaccine, such as GARDASIL® 9, is administered intramuscularly to a subject or subject population from a dengue endemic region.
  • the invention is directed to said methods, wherein the reconstituted unit dose of the invention as described herein is administered subcutaneously and wherein the HPV vaccine, in particular a 9vHPV vaccine, such as GARDASIL® 9, is administered intramuscularly to a subject or subject population of 9 to 26 years of age, preferably 9 to 15 years of age.
  • the subject or subject population is from a dengue endemic region.
  • the subject or subject population is female.
  • the subject or subject population is seronegative for all four dengue serotypes.
  • the invention is directed to a method of preventing dengue disease and HPV-associated cancers or genital warts in a subject comprising administering to the subject a subcutaneous injection of the reconstituted unit dose of the invention as described herein and an intramuscular injection of the HPV vaccine, in particular a 9vHPV vaccine, such as GARDASIL® 9, according to the above described administration schedule, wherein the subject is a female subject of 9 to 15 years of age from a dengue endemic region.
  • a 9vHPV vaccine such as GARDASIL® 9
  • the present invention is directed in part to the unit dose of the invention as described herein for use in a method of preventing dengue disease in a subject, wherein the method also comprises the prevention of HPV-associated cancers or genital warts in the subject with a HPV vaccine, in particular a 9vHPV vaccine, such as GARDASIL® 9.
  • a HPV vaccine in particular a 9vHPV vaccine, such as GARDASIL® 9
  • the present invention is directed in part to a unit dose of a dengue vaccine composition as described herein and a HPV vaccine, in particular a 9vHPV vaccine, such as GARDASIL® 9, for use in a method of preventing dengue disease and HPV-associated cancers or genital warts in a subject, respectively.
  • the present invention is directed in part to the unit dose of the invention as described herein for use in a method of preventing dengue disease in a subject population, wherein the method also comprises the prevention of HPV-associated cancers or genital warts in the subject population with a HPV vaccine, in particular a 9vHPV vaccine, such as GARDASIL® 9.
  • a HPV vaccine in particular a 9vHPV vaccine, such as GARDASIL® 9.
  • the present invention is directed in part to a unit dose of a dengue vaccine composition as described herein and a HPV vaccine, in particular a 9vHPV vaccine, such as GARDASIL® 9, for use in a method of preventing dengue disease and HPV-associated cancers or genital warts in a subject population, respectively.
  • the present invention is in part directed to the unit dose of a dengue vaccine composition as described herein and the HPV vaccine, in particular a 9vHPV vaccine, such as GARDASIL® 9, for use in a method of preventing dengue disease and HPV-associated cancers or genital warts in a subject population, comprising administering to the subject population at least a first reconstituted unit dose of the invention as described herein, wherein certain ratios of geometric mean neutralizing antibody titers (GMTs) at day 180 or 365 after administration of said first unit dose to the subject population are achieved, and concomitantly administering a HPV vaccine, in particular a 9vHPV vaccine, such as GARDASIL® 9, to the subject population.
  • GTTs geometric mean neutralizing antibody titers
  • the geometric mean neutralizing antibody titer for dengue serotype 2 (GMT DENV-2) and the geometric mean neutralizing antibody titer for dengue serotype 4 (GMT DENV-4) when tested in at least 40, or at least 50, or at least 60 subjects at day 180 or day 365 after at least a first administration of said reconstituted unit dose of the invention as described herein, and optionally a second administration of a reconstituted unit dose of the invention as described herein 90 days after said first administration, provide a ratio of GMT DENV-2:GMT DENV-4 of not more than 50, or not more than 40, or not more than 30, or not more than 20.
  • the ratio of GMT DENV-2:GMT DENV-1 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose
  • the ratio of GMT DENV-2:GMT DENV-3 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose.
  • the present invention is in part directed to the unit dose of a dengue vaccine composition as described herein and the HPV vaccine, in particular a 9vHPV vaccine, such as GARDASIL® 9, for use in a method of preventing dengue disease and HPV-associated cancers or genital warts in a subject comprising administering to the subject at least a first reconstituted unit dose of the invention as described herein, wherein certain ratios of neutralizing antibody titers at day 180 or 365 after administration of said first unit dose to the subject are achieved, and concomitantly administering a HPV vaccine, in particular a 9vHPV vaccine, such as GARDASIL® 9, to the subject.
  • a 9vHPV vaccine such as GARDASIL® 9
  • the neutralizing antibody titer for dengue serotype 2 and the neutralizing antibody titer for dengue serotype 4 at day 180 or day 365 after at least a first administration of the reconstituted unit dose of the invention as described herein, and optionally a second administration of a reconstituted unit dose of the invention as described herein 90 days after said first administration provide a ratio of neutralizing antibody titer for DENV-2:neutralizing antibody titer for DENV-4 of not more than 50, or not more than 40, or not more than 30, or not more than 20.
  • the ratio of the neutralizing antibody titers of DENV-2:DENV-1 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose, and/or the ratio of the neutralizing antibody titers of DENV-2:DENV-3 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose.
  • the geometric mean neutralizing antibody titers (GMTs) of a subject population or the neutralizing antibody titers of a subject are determined in accordance with a microneutralization test, for example according to the method described in Example 2.
  • the invention is directed to the unit dose of the invention as described herein and the HPV vaccine, in particular a 9vHPV vaccine, such as GARDASIL® 9, for said uses, wherein said unit dose and said HPV vaccine are administered simultaneously.
  • the simultaneous administration is on day 0 or day 90, preferably on day 0.
  • the administration of said unit dose and said HPV vaccine are done sequentially.
  • the invention is directed to the unit dose of the invention as described herein and the HPV vaccine, in particular a 9vHPV vaccine, such as GARDASIL® 9, for said uses, wherein said unit dose is administered by subcutaneous injection and wherein said HPV vaccine is administered by intramuscular injection.
  • the injections are administered to the arm, preferably to the deltoid region of the arm.
  • the subcutaneous injection of said unit dose and the intramuscular injection of said HPV vaccine are administered to different anatomical sites, such as to opposite arms, in particular when the vaccines are administered simultaneously.
  • the invention is directed to the unit dose of the invention as described herein and the HPV vaccine, in particular a 9vHPV vaccine, such as GARDASIL® 9, for said uses, wherein two reconstituted unit doses of the invention as described herein are administered.
  • the two unit doses of the invention as described herein are administered within 12 months or more, or within six months, or within three months, such as at day 0/1 and day 90.
  • a further third reconstituted unit dose of the invention as described herein is administered after the second administration.
  • Such a third administration may act as a booster and may be administered between 6 to 12 months after the first administration, such as 12 months after the first administration, or later than 12 month after the first administration, such as 12 months (1 year) after the second administration or even 5 years or longer after the first or second administration.
  • the invention is directed to the unit dose of the invention as described herein and the HPV vaccine, in particular a 9vHPV vaccine, such as GARDASIL® 9, for said uses, wherein two reconstituted unit doses of the invention as described herein and two doses of the HPV vaccine are administered, in particular according to the following schedule
  • the invention is directed to the unit dose of the invention as described herein and the HPV vaccine, in particular a 9vHPV vaccine, such as GARDASIL® 9, for said uses, wherein the subject or subject population is seronegative to all dengue serotypes.
  • the invention is directed to the unit dose of the invention as described herein and the HPV vaccine, in particular a 9vHPV vaccine, such as GARDASIL® 9, for said uses, wherein said unit dose is reconstituted and administered subcutaneously to a subject or subject population and said HPV vaccine is administered intramuscularly to a subject or subject population, and wherein the subject or the subject population is seronegative with respect to all dengue serotypes.
  • the subject or subject population is seropositive with respect to at least one dengue serotype.
  • the invention is directed to the unit dose of the invention as described herein and the HPV vaccine, in particular a 9vHPV vaccine, such as GARDASIL® 9, for said uses, wherein said unit dose and said HPV vaccine are administered to a subject or subject population from a dengue endemic region.
  • the reconstituted unit dose of the invention as described herein is administered subcutaneously and the HPV vaccine, in particular a 9vHPV vaccine, such as GARDASIL® 9, is administered intramuscularly to a subject or subject population from a dengue endemic region.
  • the invention is directed to the unit dose of the invention as described herein and the HPV vaccine, in particular a 9vHPV vaccine, such as GARDASIL® 9, for said uses, wherein the reconstituted unit dose of the invention as described herein is administered subcutaneously and wherein the HPV vaccine, in particular a 9vHPV vaccine, such as GARDASIL® 9, is administered intramuscular to a subject or subject population of 9 to 25 years of age, preferably of 9 to 15 years of age.
  • the subject or subject population is from a dengue endemic region.
  • the subject or subject population is female.
  • the subject or subject population is seronegative for all four dengue serotypes.
  • the invention is directed to the unit dose of the invention as described herein and the HPV vaccine, in particular a 9vHPV vaccine, such as GARDASIL® 9, for use in a method of preventing dengue disease and HPV-associated cancers or genital warts in a subject comprising administering to the subject a subcutaneous injection of the reconstituted unit dose of the invention as described herein and an intramuscular injection of the HPV vaccine, in particular a 9vHPV vaccine, such as GARDASIL® 9, according to the above described administration schedule, wherein the subject is a female subject of 9 to 15 years of age from a dengue endemic region.
  • a 9vHPV vaccine such as GARDASIL® 9
  • the present invention is directed in part to the use of a unit dose of the invention as described herein for the manufacture of a medicament for preventing dengue disease in a subject, further comprising the use of a HPV vaccine, in particular a 9vHPV vaccine, such as GARDASIL® 9, for the manufacture of a medicament for preventing HPV-associated cancers or genital warts in the subject.
  • a HPV vaccine in particular a 9vHPV vaccine, such as GARDASIL® 9
  • GARDASIL® 9vHPV vaccine such as GARDASIL® 9
  • the present invention is directed in part to the use of a unit dose of the invention as described herein for the manufacture of a medicament for preventing dengue disease in a subject population, further comprising the use of a HPV vaccine, in particular a 9vHPV vaccine, such as GARDASIL® 9, for the manufacture of a medicament for preventing HPV-associated cancers or genital warts in the subject population.
  • a HPV vaccine in particular a 9vHPV vaccine, such as GARDASIL® 9
  • GARDASIL® 9vHPV vaccine such as GARDASIL® 9vHPV vaccine
  • the present invention is in part directed to the use of a unit dose of the invention as described herein and a HPV vaccine, in particular a 9vHPV vaccine, such as GARDASIL® 9, for the manufacture of a medicament for preventing dengue disease and HPV-associated cancers or genital warts in a subject population, comprising administering to the subject population at least a first reconstituted unit dose of the invention as described herein, wherein certain ratios of geometric mean neutralizing antibody titers (GMTs) at day 180 or 365 after administration of said first unit dose to the subject population are achieved, and concomitantly administering a HPV vaccine, in particular a 9vHPV vaccine, such as GARDASIL® 9, to the subject population.
  • a HPV vaccine in particular a 9vHPV vaccine, such as GARDASIL® 9
  • the geometric mean neutralizing antibody titer for dengue serotype 2 (GMT DENV-2) and the geometric mean neutralizing antibody titer for dengue serotype 4 (GMT DENV-4) when tested in at least 40, or at least 50, or at least 60 subjects at day 180 or day 365 after at least a first administration of said reconstituted unit dose of the invention as described herein, and optionally a second administration of a reconstituted unit dose of the invention as described herein 90 days after said first administration, provide a ratio of GMT DENV-2:GMT DENV-4 of not more than 50, or not more than 40, or not more than 30, or not more than 20.
  • the ratio of GMT DENV-2:GMT DENV-1 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose
  • the ratio of GMT DENV-2:GMT DENV-3 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose.
  • the present invention is in part directed to the use of a unit dose of the invention as described herein and a HPV vaccine, in particular a 9vHPV vaccine, such as GARDASIL® 9, for the manufacture of a medicament for preventing dengue disease and HPV-associated cancers or genital warts in a subject, comprising administering to the subject at least a first reconstituted unit dose of the invention as described herein, wherein certain ratios of neutralizing antibody titers at day 180 or 365 after administration of said first unit dose to the subject are achieved, and concomitantly administering a HPV vaccine, in particular a 9vHPV vaccine, such as GARDASIL® 9, to the subject.
  • a HPV vaccine in particular a 9vHPV vaccine, such as GARDASIL® 9
  • the neutralizing antibody titer for dengue serotype 2 and the neutralizing antibody titer for dengue serotype 4 at day 180 or day 365 after at least a first administration of the reconstituted unit dose of the invention as described herein, and optionally a second administration of a reconstituted unit dose of the invention as described herein 90 days after said first administration provide a ratio of neutralizing antibody titer for DENV-2:neutralizing antibody titer for DENV-4 of not more than 50, or not more than 40, or not more than 30, or not more than 20.
  • the ratio of the neutralizing antibody titers of DENV-2:DENV-1 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose, and/or the ratio of the neutralizing antibody titers of DENV-2:DENV-3 is not more than 20, or not more than 18, or not more than 15 at day 180 or 365 after administration of said first reconstituted unit dose.
  • the geometric mean neutralizing antibody titers (GMTs) of a subject population or the neutralizing antibody titers of a subject are determined in accordance with a microneutralization test, for example according to the method described in Example 2.
  • the invention is directed to said uses, wherein the unit dose of the invention as described herein and the HPV vaccine, in particular a 9vHPV vaccine, such as GARDASIL® 9, are administered simultaneously.
  • the simultaneous administration is on day 0 or day 90, preferably on day 0.
  • the administration of the unit dose of the invention as described herein and the HPV vaccine, in particular a 9vHPV vaccine, such as GARDASIL® 9, are done sequentially.
  • the invention is directed to said uses, wherein the unit dose of the invention as described herein is reconstituted and administered by subcutaneous injection and wherein the HPV vaccine, in particular a 9vHPV vaccine, such as GARDASIL® 9, is administered by intramuscular injection.
  • the injections are administered to the arm, preferably to the deltoid region of the arm.
  • the subcutaneous injection of the unit dose of the invention as described herein and the intramuscular injection of the HPV vaccine, in particular a 9vHPV vaccine, such as GARDASIL® 9 are administered to different anatomical sites such as to opposite arms, in particular when the vaccines are administered simultaneously.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Virology (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Microbiology (AREA)
  • Immunology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Molecular Biology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
US17/274,016 2018-09-05 2019-09-05 Dengue vaccine unit dose and administration thereof Pending US20220226401A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/274,016 US20220226401A1 (en) 2018-09-05 2019-09-05 Dengue vaccine unit dose and administration thereof

Applications Claiming Priority (23)

Application Number Priority Date Filing Date Title
EP18192800.3 2018-09-05
EP18192701.3 2018-09-05
EP18192793 2018-09-05
EP18192701 2018-09-05
EP18192800 2018-09-05
EP18192776 2018-09-05
EP18192776.5 2018-09-05
EP18192787 2018-09-05
EP18192711.2 2018-09-05
EP18192793.0 2018-09-05
EP18192814.4 2018-09-05
EP18192711 2018-09-05
EP18192717 2018-09-05
EP18192814 2018-09-05
EP18192787.2 2018-09-05
EP18192717.9 2018-09-05
EP19154334.7 2019-01-29
EP19154334 2019-01-29
US16/295,611 US11464815B2 (en) 2018-09-05 2019-03-07 Dengue vaccine unit dose and administration thereof
EP19161184 2019-03-07
EP19161184.7 2019-03-07
PCT/US2019/049749 WO2020051334A1 (en) 2018-09-05 2019-09-05 Dengue vaccine unit dose and administration thereof
US17/274,016 US20220226401A1 (en) 2018-09-05 2019-09-05 Dengue vaccine unit dose and administration thereof

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US16/295,611 Continuation US11464815B2 (en) 2018-09-05 2019-03-07 Dengue vaccine unit dose and administration thereof

Publications (1)

Publication Number Publication Date
US20220226401A1 true US20220226401A1 (en) 2022-07-21

Family

ID=69722836

Family Applications (3)

Application Number Title Priority Date Filing Date
US16/561,953 Active US11590221B2 (en) 2018-09-05 2019-09-05 Dengue vaccine unit dose and administration thereof
US17/274,016 Pending US20220226401A1 (en) 2018-09-05 2019-09-05 Dengue vaccine unit dose and administration thereof
US18/149,742 Pending US20230355748A1 (en) 2018-09-05 2023-01-04 Dengue vaccine unit dose and administration thereof

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US16/561,953 Active US11590221B2 (en) 2018-09-05 2019-09-05 Dengue vaccine unit dose and administration thereof

Family Applications After (1)

Application Number Title Priority Date Filing Date
US18/149,742 Pending US20230355748A1 (en) 2018-09-05 2023-01-04 Dengue vaccine unit dose and administration thereof

Country Status (18)

Country Link
US (3) US11590221B2 (ko)
EP (3) EP3620174B1 (ko)
JP (1) JP2021536463A (ko)
KR (1) KR20210071979A (ko)
CN (1) CN112912106A (ko)
AU (1) AU2019335006A1 (ko)
BR (1) BR112021003962A2 (ko)
CA (1) CA3111332A1 (ko)
CO (1) CO2021003931A2 (ko)
DK (1) DK3620174T3 (ko)
EC (1) ECSP21023398A (ko)
ES (1) ES2908304T3 (ko)
IL (2) IL298307A (ko)
MX (1) MX2021002586A (ko)
PE (1) PE20211545A1 (ko)
PH (1) PH12021550479A1 (ko)
SG (1) SG11202102104SA (ko)
WO (1) WO2020051334A1 (ko)

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MX2018012459A (es) * 2016-04-13 2019-06-06 Takeda Vaccines Inc Composiciones y metodos de vacunacion contra virus de dengue en niños y adultos jovenes.
CA3111332A1 (en) * 2018-09-05 2020-03-12 Takeda Vaccines, Inc. Dengue vaccine unit dose and administration thereof
WO2021174059A1 (en) * 2020-02-27 2021-09-02 Takeda Vaccines, Inc. Method for removing host cell dna from virus preparation
CN112813089A (zh) * 2021-03-22 2021-05-18 苏州诺威百奥生物科技有限公司 黄病毒科检测用多位点重组抗原的制备方法
CN113913551B (zh) * 2021-06-22 2022-05-13 中国检验检疫科学研究院 一种登革病毒分型的测序引物、检测方法及试剂盒
WO2024086605A1 (en) 2022-10-18 2024-04-25 Takeda Vaccines, Inc. Dengue vaccine formulation
EP4357464A1 (en) * 2022-10-20 2024-04-24 Takeda Vaccines, Inc. A method for detecting the presence of a dengue virus serotype in a sample containing at least one dengue virus serotype

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11464815B2 (en) * 2018-09-05 2022-10-11 Takeda Vaccines, Inc. Dengue vaccine unit dose and administration thereof
US11590221B2 (en) * 2018-09-05 2023-02-28 Takeda Vaccines, Inc. Dengue vaccine unit dose and administration thereof

Family Cites Families (42)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4810092A (en) 1986-02-21 1989-03-07 Midac Corporation Economical spectrometer unit having simplified structure
JPH084508B2 (ja) 1987-09-16 1996-01-24 国立予防衛生研究所長 組み換えワクチニアウイルス
JP2511494B2 (ja) 1988-05-12 1996-06-26 善治 松浦 日本脳炎ウイルス表面抗原蛋白質の製造法
US6184024B1 (en) 1988-07-14 2001-02-06 The United States Of America As Represented By The Department Of Health And Human Services Chimeric and/or growth-restricted flaviviruses
IL91304A0 (en) 1988-08-20 1990-03-19 Us Health Recombinant vaccinia virus for prevention of disease caused by flavivirus
US5514375A (en) 1990-08-15 1996-05-07 Virogenetics Corporation Flavivirus recombinant poxvirus vaccine
MY109299A (en) 1990-08-15 1996-12-31 Virogenetics Corp Recombinant pox virus encoding flaviviral structural proteins
US5494671A (en) 1990-08-27 1996-02-27 The United States Of America As Represented By The Department Of Health And Human Services C-terminally truncated dengue and Japanese encephalitis virus envelope proteins
DE69231570T2 (de) 1991-09-19 2001-06-21 Us Health Chimäre und/oder wachstumgehemmte Flaviviren
JPH05276941A (ja) 1992-02-28 1993-10-26 Nippon Zeon Co Ltd フラビウイルス科に属するウイルスの表面抗原タンパク質を含む非感染性の構造物粒子の製造方法
CA2224724C (en) 1995-05-24 2007-12-04 Hawaii Biotechnology Group, Inc. Subunit vaccine against flavivirus infection
WO1996040933A1 (en) 1995-06-07 1996-12-19 The Government Of The United States Of America, Represented By The Secretary Department Of Health And Human Services Infectious dengue 2 virus pdk-53 as quadravalent vaccine
IL131600A0 (en) 1997-02-28 2001-01-28 Ora Vax Inc Chimeric flavivirus vaccines
AU9794198A (en) 1997-10-08 1999-04-27 Government Of The United States Of America, As Represented By The Secretary Of The Department Of Health And Human Services, The Chimeric vaccine against tick-borne encephalitis virus
CA2331368C (en) 1998-06-04 2009-02-10 The Government Of The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Nucleic acid vaccines for prevention of flavivirus infection
PT2278012E (pt) 2000-02-16 2015-11-30 Univ Mahidol Quimeras imunogénicas do vírus do dengue 2
CA2440593A1 (en) 2001-03-12 2002-09-19 Yale University Compositions and methods comprising west nile virus polypeptides
US20040259224A1 (en) 2002-05-31 2004-12-23 Farshad Guirakhoo Tetravalent Dengue vaccines
WO2006134443A1 (en) 2005-06-17 2006-12-21 Sanofi Pasteur Dengue serotype 2 attenuated strain
WO2007021672A2 (en) 2005-08-10 2007-02-22 Acambis Inc. Vaccination against dengue virus infection
FR2903605A1 (fr) 2006-07-12 2008-01-18 Sanofi Pasteur Sa Methode d'immunisation contre les quatres serotypes de la dengue
FR2906724B1 (fr) 2006-10-04 2009-03-20 Sanofi Pasteur Sa Methode d'immunisation contre les 4 serotypes de la dengue.
FR2909286B1 (fr) 2006-12-01 2012-06-08 Sanofi Pasteur Methode d'immunisation contre les 4 serotypes de la dengue
AP3172A (en) 2007-07-13 2015-03-31 Florida Gulf Coast University Optimized dengue virus entry inhibitory peptide (DN81)
SG157244A1 (en) 2008-05-15 2009-12-29 Mp Biomedicals Asia Pacific Pte Ltd Point of care test for the detection of exposure or immunity to dengue virus
WO2010085358A2 (en) 2009-01-23 2010-07-29 The Board Of Regents Of The University Of Texas System Flaviviruses expressing the prm, e, and ns1 proteins of other flaviviruses and uses thereof
EP2438198A4 (en) 2009-06-01 2014-05-14 Inviragen Inc COMPOSITIONS AND METHODS FOR THE DELIVERY OF VACCINES AGAINST DENGUE VIRUS
US8968996B2 (en) 2009-06-01 2015-03-03 Takeda Vaccines, Inc. Compositions and methods for rapid immunization against dengue virus
BRPI0904020B8 (pt) 2009-10-01 2021-05-25 Fundacao Oswaldo Cruz composição vacinal contra o vírus da dengue, e, kit
EP2353609A1 (en) 2010-02-04 2011-08-10 Sanofi Pasteur Immunization compositions and methods
TW201402143A (zh) 2012-06-10 2014-01-16 Inviragen Inc 抗登革熱病毒疫苗之組成物及投藥方法
MY168959A (en) 2012-07-24 2019-01-28 Sanofi Pasteur Vaccine compositions for the prevention of dengue virus infection
CN104812408A (zh) 2012-07-24 2015-07-29 赛诺菲巴斯德有限公司 用于防止登革热病毒感染的疫苗组合物
TW201428101A (zh) 2012-11-08 2014-07-16 Inviragen Inc 登革熱病毒血清型4型之建構物的組成物、方法及用途
CA2933472A1 (en) 2012-12-14 2014-06-19 Takeda Vaccines, Inc. Compositions, methods of administration and uses for trivalent dengue virus formulations
ES2933970T3 (es) 2013-03-15 2023-02-15 Takeda Vaccines Inc Composiciones y métodos para construcciones quiméricas de virus del dengue en vacunas
TW201620546A (zh) 2014-09-02 2016-06-16 賽諾菲巴斯德公司 疫苗組合物
WO2017005654A1 (en) 2015-07-03 2017-01-12 Sanofi Pasteur Concomitant dengue, diphtheria, tetanus, whooping cough (pertussis), polio, and haemophilus influenzae type b vaccination.
BR112017028212A2 (pt) 2015-07-03 2018-09-11 Sanofi Pasteur vacinação concomitante contra dengue e febre amarela
US10004795B2 (en) 2015-09-08 2018-06-26 Fundacao Butantan Process for preparing an attenuated tetravalent dengue vaccine
MX2018012459A (es) * 2016-04-13 2019-06-06 Takeda Vaccines Inc Composiciones y metodos de vacunacion contra virus de dengue en niños y adultos jovenes.
SG10201607778XA (en) 2016-09-16 2018-04-27 Chugai Pharmaceutical Co Ltd Anti-Dengue Virus Antibodies, Polypeptides Containing Variant Fc Regions, And Methods Of Use

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11464815B2 (en) * 2018-09-05 2022-10-11 Takeda Vaccines, Inc. Dengue vaccine unit dose and administration thereof
US11590221B2 (en) * 2018-09-05 2023-02-28 Takeda Vaccines, Inc. Dengue vaccine unit dose and administration thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Osorio JE, Wallace D, Stinchcomb DT. A recombinant, chimeric tetravalent dengue vaccine candidate based on a dengue virus serotype 2 backbone. Expert Rev Vaccines. 2016;15(4):497-508. doi: 10.1586/14760584.2016.1128328. Epub 2016 Feb 22. PMID: 26635182. *

Also Published As

Publication number Publication date
SG11202102104SA (en) 2021-04-29
US20230355748A1 (en) 2023-11-09
EP3620174B1 (en) 2022-01-19
PH12021550479A1 (en) 2021-10-25
EP3620174A1 (en) 2020-03-11
CO2021003931A2 (es) 2021-04-19
IL281177B2 (en) 2024-03-01
EP4082568A1 (en) 2022-11-02
IL298307A (en) 2023-01-01
AU2019335006A1 (en) 2021-05-06
ES2908304T3 (es) 2022-04-28
JP2021536463A (ja) 2021-12-27
DK3620174T3 (da) 2022-03-14
US11590221B2 (en) 2023-02-28
ECSP21023398A (es) 2021-06-30
CA3111332A1 (en) 2020-03-12
WO2020051334A1 (en) 2020-03-12
KR20210071979A (ko) 2021-06-16
MX2021002586A (es) 2021-06-08
EP3846849A1 (en) 2021-07-14
CN112912106A (zh) 2021-06-04
IL281177B1 (en) 2023-11-01
US20200230230A1 (en) 2020-07-23
BR112021003962A2 (pt) 2021-05-25
PE20211545A1 (es) 2021-08-16
IL281177A (en) 2021-04-29

Similar Documents

Publication Publication Date Title
US11590221B2 (en) Dengue vaccine unit dose and administration thereof
TWI406673B (zh) 對抗4種登革熱血清型之免疫方法
US11464815B2 (en) Dengue vaccine unit dose and administration thereof
AU2011212647B2 (en) Immunization compositions and methods
US10946087B2 (en) Vaccine compositions against dengue virus diseases
US10857222B2 (en) Concomitant dengue and yellow fever vaccination
WO2017005654A1 (en) Concomitant dengue, diphtheria, tetanus, whooping cough (pertussis), polio, and haemophilus influenzae type b vaccination.
Fiore et al. Hepatitis A vaccines
JP2022544613A (ja) デング熱及びa型肝炎を予防するための方法
WO2014083194A1 (en) Methods for inducing antibodies
US11426461B2 (en) Methods for preventing dengue and hepatitis A
Noor et al. Comparative Analysis of Emerging Viruses to Inform Development of Safe and Effective Vaccines for COVID-19

Legal Events

Date Code Title Description
AS Assignment

Owner name: TAKEDA VACCINES, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:TAKEDA PHARMACEUTICALS INTERNATIONAL AG;REEL/FRAME:056927/0192

Effective date: 20200212

Owner name: TAKEDA PHARMACEUTICALS INTERNATIONAL AG, SWITZERLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:WALLACE, DEREK;LEFEVRE, INGE;SIGNING DATES FROM 20200211 TO 20200212;REEL/FRAME:056923/0238

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED