![]() Multivalent dengue virus vaccine
专利摘要:
The present invention provides a vaccine composition of attenuated dengue virus. More specifically, the attenuated virus is produced by serial passage in PDK cells. In addition, the present invention stimulates an individual's immune system by administration of attenuated dengue-1, dengue-2, dengue-3 and dengue-4 viruses to induce protection against all four dengue virus serotypes. Provide a method. 公开号:KR20020008136A 申请号:KR1020017012302 申请日:2000-03-24 公开日:2002-01-29 发明作者:엑켈스케네쓰에이치;풋낙조셉알;더보이스도리아알;인니스브루스엘;호크찰스에이치;웰링톤썬;니란잔카네사-타산 申请人:왈터 리드 아미 인스티튜트 오브 리써치; IPC主号:
专利说明:
MULTIVALENT DENGUE VIRUS VACCINE} [2] Dengue fever is caused by one of four serotypes: dengue virus, dengue-1, dengue-2, dengue-3 and dengue-4, which are transferred to humans by mosquitoes. In adults, dengue infections usually cause self-limiting but inoperable acute diseases involving fever, muscle pain, headaches and intermittent rashes. This disease can be exacerbated by hemorrhagic fever, which can be evidenced by a positive tourniquet test, natural pointed bleeding, clear bleeding and / or shock. Hemorrhagic dengue has a mortality rate of about 0.5% of the cases. It has been found that infections infected by another dengue strain after antibodies have been formed by early dengue infection may be at higher risk for hemorrhagic dengue. [3] Mosquito mediators of the dengue virus are found in all tropical and subtropical regions of the world and in some temperate regions of the United States, Europe, Africa and Central Asia. Recently, endemic and epidemic dengue infections have occurred in Central and South America, Southeast Asia, India, Africa, the Caribbean, and the Pacific. Vector control is impractical. [4] There is a need for an effective vaccine that can protect against all four serotypes of dengue. [5] Summary of the Invention [6] The present invention satisfies the aforementioned needs. The present invention relates to vaccine compositions comprising the attenuated dengue virus from all four serotypes. The attenuated virus is provided with a physiologically acceptable excipient in an amount sufficient to induce an immune response in a human host, and may optionally comprise an adjuvant that enhances the host's immune response. [7] Accordingly, one of the objects of the present invention is an attenuated dengue virus comprising more than one attenuated dengue virus selected from the group consisting of dengue-1, dengue-2, dengue-3 and dengue-4 in any combination It is to provide a composition. [8] It is another object of the present invention to provide a method of stimulating an individual's immune system to induce protection against a dengue virus. The method involves administering to the subject an immunologically sufficient amount of a dengue virus derived from all four serotypes attenuated by serial passage. [1] This application is incorporated by reference in U.S.C. Claim the contents of priority under § 119 (e). [9] 1: Incidence of> Grade 1 symptoms as a result of vaccine administration. [10] Figure 2: Frequency of distribution of Reactogenicity Index by serotype. [11] Figure 3: Table showing the results of a dose-ranging 4-valent dengue vaccine study. [12] 4: Table showing immunogenicity of a dengue vaccine with full-dose tetravalent out of 10 subjects. [13] Figure 5: Table showing details of selected formulations of the 4-valent vaccine study. [14] Figure 6, A-H: Interferon γ production by PBMCs collected from vaccine volunteers and stimulated with virus-specific serotypes. All volunteers receive only one serotype of vaccine. Graphs (A-D) show the results of volunteers given a second dose around day 32. The graph (E-H) shows the results of volunteers given a second dose around 92 days. Responses above 1000 pg / ml appeared immediately before the second dose in most volunteers. Only four volunteers responded at least 1000 pg / ml within the first 15 days given the primary vaccine dose. [15] 7, A-D: Interferon γ production of PBMCs collected from vaccine volunteers vaccinated with the tetravalent vaccine. PBMCs were individually stimulated with each serotype of virus. Individual lines in each graph represent the response of PBMC of volunteer 1 to each serotype of virus. Slow response was observed in the case of monovalent vaccination. [16] 8, A and B: Granzyme B mRNA preparation of PBMCs in which 1 and 4 were collected from vaccine volunteers. Cells were collected from all individuals whose PBMCs were always secreted at> 1000 pg IFNγ / ml. This is a semiquantitative indication of the amount of mRNA detected by RTPCR. Top chart (A) shows the intensity of the bands seen for all samples. Lower gel (B) is from selected volunteers showing examples of positive and negative RTPCR assays. [17] The present invention provides attenuated dengue virus of all four serotypes suitable for use as a vaccine in humans. The dengue virus described herein is prepared by serial passage of infectious dengue virus isolates in a suitable host cell line, such as primary kidney cells, to allow for the accumulation of mutations that attenuate the isolates. Continuous passage refers to infection of a cell line with viral isolates, recovery of viral offspring from the host cell and subsequent infection of the host cell with the virus to generate the next passage. [18] Preferably, regardless of whether attenuated or inactivated, other viral compositions of any serotype of serotypes may be used with the attenuated strains described herein, the compositions of the invention Use a virus. The attenuated dengue-1 virus derived from the 45AZ5 isolate was deposited with the accession number VR-2648 in the American Treaty Culture Collection (ATCC) (10801 University Boulevard, Manassas, Virginia 20110-2209, U.S.A) under the Budapest Treaty. [19] The attenuated dengue-2 virus derived from the S16803 isolate was deposited with the accession number of VR-2653 in the Budapest Treaty American Type Culture Collection (ATCC) (10801 University Boulevard, Manassas, Virginia 20110-2209, U.S.A). [20] The attenuated dengue-3 virus derived from the CH53489 isolate was deposited with the accession number VR-2647 in the Budapest Treaty American Type Culture Collection (ATCC) (10801 University Boulevard, Manassas, Virginia 20110-2209, U.S.A). [21] The attenuated dengue-4 virus derived from the 341750 isolate was deposited with the accession number VR-2652 to the American Type Culture Collection (ATCC) (10801 University Boulevard, Manassas, Virginia 20110-2209, U.S.A) under the Budapest Treaty. [22] Modified viruses have been isolated that can be attenuated by serial passage of virulent (disease-causing) strains of dengue, ie not infectious or cause disease. This modified virus was tested for reduced infectivity in monkeys. Viruses with reduced infectivity were then tested in humans. Humans are the only primates that show signs of clinical disease. Attenuated viruses that showed at least to no clinical reactivity but still elicit an immune response. [23] In one embodiment of the present invention, attenuated strains were induced by serial passage in primary dog kidney (PDK) cells of toxic dengue isolates derived from all four dengue serotypes. Serial subculture is performed by infecting PDK cells with toxic strains, incubating the infected cells for several days, and harvesting the supernatant containing the virus. The harvested virus is then applied to fresh PDK cells to generate the next passage. [24] A series of various passages were tested for clinical effects after final passage in fetal Rhesus monkey lung cells (FRhL). Using FRhL cells, typically, passage 1 is considered a master seed, passage 2 is considered a production seed, and passage 3 is optimized for virus titers considered as a lot of vaccine. I was. Vaccines were prepared at various PDK passage levels and vaccine products were tested for attenuation in monkeys and humans. The toxicity of the passaged virus, ie its ability to cause disease, was named in part by daily monitoring and evaluation of symptoms such as temperature (fever), headache and rash. Judging from the inability of the virus to induce clinical symptoms of dengue fever in vaccinates, the passage is considered attenuated. [25] Propagation of the attenuated virus of the present invention is possible in many cell lines capable of dengue virus growth. Dengue virus grows in a variety of human and animal cells. Preferred cell lines for propagation of attenuated dengue virus for vaccine use include DBS-FRhL-2, Vero cells and other monkey cells. The highest viral yield is usually obtained by heteroploid cell lines such as Vero cells. Cells are typically inoculated in a number of infections ranging from about 0.01 to 0.005, and under conditions permitting the replication of the virus, such as about 30-37 ° C. and about 3-5 days, or the time required for the virus to reach an appropriate titer Incubate as much as possible. The virus can be removed from the cell culture, separated from the cellular components by conventionally known purification methods such as centrifugation, and further purified as desired using methods known to those skilled in the art. [26] After isolation of the attenuated virus, sequencing of its genome can be performed to obtain the basis for the attenuated phenotype. This is accomplished by sequencing viral DNA to identify changes in nucleotides in the attenuated isolates compared to the genomic sequence of the control virus. Thus, molecular changes conferring attenuation to toxic strains can be characterized. [27] One embodiment of the invention provided herein includes introducing a sequence change alone or in combination at any position listed in the table above to generate attenuated virus progeny. Viral genomes with these changes can be prepared by any standard recombinant DNA technique known to those skilled in the art to introduce nucleotide changes into cloned DNA (Ausubel et al., Current Protocols in Molecular Biology, Greene Publishing Associates & Wiley Interscience, New York). , 1989). The genome can then be ligated to a suitable vector for transfection of host cells for the production of viral offspring. [28] The ability to generate viral offspring through plasmid-mediated introduction of the viral genome can also be used to prepare viruses with limited molecular changes. In this embodiment of the invention, stable viral stock can be prepared comprising altered sequences that confer desired properties on the virus, such as reduced toxicity. This approach can also be used to introduce the effects of molecular changes on the various properties of the virus, ie antigenic form, toxicity, or desired sequence change, into the viral genome, produce viral offspring from the genome, and then characterize it for characterization. Attenuation to recover viral offspring can be assessed. This approach can also be used to construct viruses with heterologous sequences inserted into the viral genome that are simultaneously delivered by viruses that generate an immune response against other diseases. [29] Construction of viral genomes with limited molecular changes can be achieved using standard techniques such as oligonucleotide-directed, linker-scanning or mutagenesis techniques based on polymerase chain reactions known to those skilled in the art. (Zoller and Smith, 1984, DNA 3, 479-488; Botstein and Shortle, 1985, Science 229, 1193). Ligation of the genome into a vector suitable for delivery can be accomplished through standard techniques known to those skilled in the art. Transfection of the vector into host cells for the production of viral progeny is carried out using any standard technique such as calcium-phosphate or DEAE-dextran mediated transfection, electroporation, protoplast fusion and other techniques known to those skilled in the art. Can be done. (Sambrook et al., Molecular Cloning: A laboratory Manual , Cold Spring Harbor Laboratory Press, 1989). [30] For vaccine use, the attenuated virus of the present invention may be used directly in vaccine formulations or, if necessary, by lyophilization in a stabilizer, using lyophilization protocols known to those skilled in the art (Hoke, 1990, Am J Trop Med Hyg 43, 219-226). Lyophilized virus is typically maintained at about 4 ° C. In use, the lyophilized virus may be reconstituted in water, as described further below, or, if necessary, in a stabilizing solution, such as saline or stabilizing solution comprising Mg ++ and HEPES, with or without adjuvant. Can be. All references cited herein are hereby incorporated by reference in their entirety. [31] Accordingly, the dengue virus vaccine of the present invention is immunologically effective for treating one or more attenuated dengue viruses selected from the group consisting of dengue-1, dengue-2, dengue-3 and dengue-4 as described herein as active ingredients. Included in quantity. The attenuated viral composition can be introduced into a subject, in particular a human, with pharmacologically acceptable excipients and / or adjuvant. Useful excipients are well known in the art and include, for example, water, buffered water, 0.4% saline, 0.3% glycine, hyaluronic acid and the like. The resulting aqueous solution is packaged or lyophilized, depending on the application, and the lyophilized formulation is rehydrated prior to administration as described above. The composition may be formulated with pharmaceutically acceptable auxiliary substances such as pH adjusting and buffering agents, tonicity adjusting agents, wetting agents, etc., such as sodium acetate, sodium lactate, sodium chloride, potassium chloride, calcium chloride, sorbitan, to optimize physiological conditions as needed. Monolaurate, triethanolamine oleate, and the like. [32] Administration of the live attenuated virus disclosed herein may be parenteral injection (eg, intraperitoneal, subcutaneous or intramuscular injection), ovo injection in birds, oral injection and virus on the airway surface (usually with medication) Can be performed by any suitable means including locally applying. Local application of the virus to the airway surface may be performed intranasally (eg, by using a dropper, swab or inhaler to deposit the medicament intranasally). Local application of the virus to the airway surface may also produce particles suitable for respiration of the drug containing the virus (including solid particles and liquid particles) by inhalation administration, such as as an aerosol suspension, and then subject the subject to inhalation of particles suitable for respiration. This can be done by. Methods and devices for the administration of particles suitable for the respiration of a medicament are known, and any conventional technique can be used. As a result of vaccination, the host is at least partially or fully immune to dengue virus infection of the serotype administered, or is resistant to ongoing moderate or severe dengue virus infection. [33] Vaccine compositions comprising the attenuated dengue virus of the present invention are administered to a person who is vulnerable to or otherwise at risk of dengue virus infection to enhance an individual's autoimmune response. This amount is defined as "immunologically effective amount." In this use, the precise amount again depends on the subject's state of health and body weight, mode of administration, the nature of the formulation, etc., but typically ranges from about 10 2 to 10 6 pfu of each serotype of dengue virus per subject. The amount of viral vaccine of each serotype is adjusted, i.e. increased or decreased, so as to obtain an agent that provides sufficient protection from infection with the desired dengue virus. When combining four serotypes, the preferred composition comprises the same amount of each dengue serotype. In either case, the vaccine formulation should be provided with an amount of attenuated dengue virus of each serotype sufficient to effectively protect the patient against a serious or life-threatening dengue virus infection of one serotype in the vaccine formulation, and crossover Other serotypes are possible if protection is desired. [34] The attenuated dengue virus of the present invention of one particular serotype can be used in combination with the attenuated virus of other serotypes of dengue virus to protect against multiple dengue viruses. Typically different modified viruses are administered in combination and simultaneously, but can also be administered separately. [35] In some cases, it may be desirable to combine attenuated dengue virus vaccines of the invention with vaccines that induce a protective response to other pathogens. [36] The vaccine composition of the present invention can be administered in a single or multiple doses. Multiple administrations may be necessary to elicit sufficient levels of immunity. The level of immunity induced can be monitored by measuring the amount of neutralizing secretory serum antibodies, and by repeated vaccinations or adjusted doses as necessary to maintain the desired level of protection. [37] The following examples are provided for illustration and not limitation. [38] The following materials and methods are used in the examples that follow. [39] Materials and Methods for Vaccine Preparation [40] Virus strains. [41] DEN virus was isolated from human and mosquito sources after passage in primary kidney (PDK) cell culture. Table 1 lists strains adopted and passaged in PDK cells. After subculture in PDK cells, virus strains were further acclimated to FRhL cells for seed and vaccine preparation. This consists of an additional 3-4 passages for the final vaccine lot formulation. In addition, the parent virus strains listed in Table 1 were derived from low cell culture passages in cells capable of DEN virus replication. [42] Vaccine manufacturing [43] DEN vaccines for all four serotypes were prepared in FRhL cell culture using similar procedures. Collected, pre-tested (see Table 2 for test results) FRhL cells were removed from the liquid nitrogen reservoir, Eagle Minimum Essential Medium (EMEM) (Biowhittaker, Waldersville, MD) in a 150 cm 2 flask, non-essential amino acids, fetal bovine serum, Plated in cell media supplemented with FBS (2%) (Biowhittaker, Waldersville, MD) and antibiotics. After the flask reached confluency, the medium was removed, the flask was inoculated with DEN producing seeds diluted with 0.01 MOI and adsorbed at 32 ° C. for 1 hour. After adsorption and feeding into fresh EMEM medium, the flask was switched to 32 ° C. and maintained for 4 days. Four days after inoculation, the medium was discarded from all flasks and the cell monolayers were washed three times with 100 ml of Hanks BSS (Biowhittaker, Waldersville, MD). After washing, the flask was fed with EMEM medium containing 0.25% human serum albumin (HSA, Alpha Therapeutic Corp, Los Angeles, CA) instead of FBS. After an additional two days of incubation at 32 ° C., the supernatant was removed from all flasks and combined. After sampling for safety testing, the remaining cultures were combined and purified by filtration with a 0.45 micron nonprotein binding membrane filter. The filtered liquids were combined and mixed with an equal amount of stabilizer comprising 15% lactose and 5% HSA. The complete liquid was stored at −70 ° C. and lyophilized. For final vialing, the stabilized whole liquid was thawed rapidly at 41 ° C. and dispensed 3 ml into serum vials. The tray of the vial was frozen at a temperature of −40 ° C. in a Hull freeze dryer and then dried for 1 day. After capping, the vial was stored at −20 ° C. in the monitored freezer. [44] Vaccine testing [45] All cell banks used for seed production as well as seed and vaccine lots were tested for the presence of contaminants. Test articles and results are listed in Table 2. No contaminants were detected in any of the products. [46] Rhesus monkey inoculation [47] Aged female and male rhesus macaques (6-15 kg) were immunized with 0.5 ml subcutaneous inlay on the upper arm with a DEN vaccine lot or parental virus. Blood for virus isolation and antibody testing was taken daily in the femoral vein before and 14 days after inoculation. Blood was also collected at 30 and 60 days post immunization. Viral challenge was performed similarly. [48] Virus Isolation by Amplification in C6 / 36 Cells [49] Virus isolation by C6 / 36 cell culture amplification is described in Putnak et al., 1996 (J. Infect Dis 174, 1176-1184). Briefly, after inoculation of monkeys, blood samples were obtained daily for 1-14 days. Serum was separated and frozen at -80 ° C. To recover the virus from the serum, thawed serum was diluted 1: 3 in cell culture medium and then used to inoculate a 25 cm 2 flask containing a monolayer of C6 / 36 mosquito cells. After adsorption of the virus, EMEM holding medium was added to the flask and maintained at 28 ° C. After 7 days, the medium was changed and the flasks were incubated for an additional 7 days. At 14 days after inoculation, the supernatant was discarded and frozen at −80 ° C. and mixed with the same amount of heat inactivated fetal bovine serum (FBS). Frozen samples were later analyzed for infectious virus by plaque assay. [50] Dengue virus strain used for the development of attenuated vaccines Serotype Original isolate Vaccine strain: Passage from human isolate PDK passage selected for vaccine production FRhL Passage for Seed and Vaccine Manufacturing Parent strain: Passage from human isolate DEN-1 (West Pac 74; 45AZ5) Human isolate, Nauru, 1974 20xFRhL (plaque sorting and mutation by 5AZ); vaccine prepared at p-20 causing dengue (2 vols) 10,20,27 1: master seed 2: production seed 3: vaccine lot 9 x FRhL DEN-2 (S16803) Human Isolation, Thailand, 1974 1x Mosquito; 4x PGMK 10,20,30,40,50 1: master seed 2: production seed 3: vaccine lot 4 x PGMK; 2xC6 / 36 DEN-3 (CH53489) Human Isolation, Thailand, 1973 4xPGMK; 5xC6 / 36 10,20,30 1: master seed 2: production seed 3: vaccine lot 4xPGMK DEN-4 (341750) Human Isolate, Colombia, 1982 1x Mosquito 6,10,15,20 1: pre-master seed (PDK-20 only) 2: master seed 3: production seed 4: vaccine lot 1x Mosquito; 5xPGMK; 4xFRhL [51] Preclinical Trials of FRhl Cell Bank and DEN LAV Seed and Vaccine Lot exam FRhl Cell Bank Master seed Production seed Vaccine (bulk) Vaccine (Final Container) Aseptic x x x x x Mycoplasma x x x RT x x Blood cell adsorption x x x Cell culture safety (4 cell lines) xx Fertilized egg safety x Animal Safety: Growth Mouse xx Animal Safety: Suckling Mouse xx Animal Safety: Guinea Pigs xx Animal safety: Rabbit x Carcinogenic x NA NA NA NA Nuclear cell x NA NA NA NA Monkey Safety: Neurotoxicity NA x (DEN-4) Monkey Infectious / Immunogenic NA x Monkey efficacy NA x (DEN-2, DEN-4) Infectious (Plaque Assay) NA x x x x Total safety NAx x Residual moisture NA x Reconstitution pH NA x Reconstitution osmotic NA x Endotoxin NA x Equality (DEN) NA x x x [52] DEN virus strain set used for inoculation of rhesus monkeys and adapted to PDK cells DEN virus strain virus Inoculation: PFU / 0.5 ml Mks Viremia / Total (Average Days of Viremia) Mk seroconversion / total (GMT PRNT 50 at 1-2 mo after inoculation) DEN-1, 45AZ5 PDK-0 (mo) 3.3 x 10 4 4/4 (6.8) 4/4 (760) PDK-10 (Generated Seed) * 7.0 x 10 4 4/4 (4.75) 4/4 (1030) PDK-20 (Generated Seeds) 1.7 x 10 4 4/4 (4.5) 4/4 (640) PDK-27 (Generated seed 1.8 x 10 4 0/4 (0) 4/4 (50) DEN-2, S16803 PDK-0 (Mo) 5.0 x 10 6 4/4 (5) 4/4 (600) PDK-10 (Generated Seeds) 3.8 x 10 5 4/4 (4.75) 4/4 (570) PDK-20 (Generated Seeds) 2.2 x 10 5 4/4 (6.5) 4/4 (920) PDK-30 (Generated Seed 1 ) 4.4 x 10 5 2/3 (3.3) 4/4 (640) PDK-30 (Generated Seed 2 ) 2.1 x 10 5 3/3 (6.0) 3/3 (640) PDK-40 (Generated Seeds) 1.0 x 10 4 2/4 (1) 3/4 (90) PDK-50 (Generated Seed 1 ) 2.6 x 10 6 2/4 (1) 4/4 (310) [53] Table 3 continued [54] PDK-50 (Generated Seed 2 ) 5.9 x 10 5 3/4 (3.25) 4/4 (280) PDK-50 (vaccine) 1 x 10 6 ND 4/4 (270) DEN-3, CH53489 PDK-0 (parent) 8.0 x 10 3 3/3 (3) 3/3 (660) PDK-10 (Generated Seeds) 2.5 x 10 6 @ 2/3 (1.3) 3/3 (150) PDK-20 (Generated Seeds) 1.0 x 10 6 @ 0/3 3/3 (130) PDK-30 (Generated Seed) 9.3 x 10 5 @ 0/3 0/3 (<10) DEN-4, 341750 PDK-0 (parent) 1.0 x 10 3 3/3 (4.7) 3/3 (420) PDK-6 (Generated Seeds) 1.7 x 10 5 1/4 (0.5) 4/4 (250) PDK-10 (Generated Seeds) 2.9 x 10 5 1/4 (1.3) 2/4 (90) PDK-15 (Generated Seeds) 5.5 x 10 4 1/4 (0.25) 2/4 (40) PDK-20 (Generated Seeds) 5.5 x 10 4 1/4 (0.25) 2/4 (70) PDK-20 (vaccine) 1.2 x 10 5 1/3 (0.3) 3/3 (50) 1,2: 2 separate monkey experiment groups Plaque assay performed on @ C6 / 36 cells [55] Plaque analysis . In Rhesus monkey kidney (LLC-Mk 2 , ATCC CCL7) cells according to the method of Sukhavachana et al. (1966) (Bull WHO 35, 65-66), directly from monkey serum by plaque analysis, or Infectious virus was titrated from amplified viremia isolates. Assays in C6 / 36 cells were performed as described in Putnak et al. (1996) supra. [56] Neutralization test . DEN neutralizing antibodies were measured from monkey serum using a plaque reduction neutralization test similar to that used by Russell et al. (1967) (J Immunol 99, 285-290). Plaque reduction 50% endpoint (PRNT50) was measured in serum samples using the parental viruses listed in Table 1. [57] Example 1 [58] DEN virus modification and vaccine lot generation in PDK cells. [59] DEN virus strains selected for vaccine development have a variety of passage history prior to PDK passage. For DEN-4 341750, there was only one mosquito passage before inoculation of PDK cell culture, whereas DEN-1 West Pac 74 strain had a history of 20 FRhL cell passages prior to PDK passage (Table 1). Except for DEN-3, all strains were adapted after a small number of PDK passages. For DEN-3, additional efforts were needed to increase viral feed in the initial passage to adapt the strain to PDK cells. As a general case after adaptation to PDK cells, the DEN virus titers were found to be in the range of 10 4 -10 5 PFU / ml. Attempts to increase titers have not been successful, and alternative cell substrates have been found for vaccine production. DBS-FRhL-2 (FRhL) cells were selected for this purpose for a variety of reasons: 1) In these cells, the DEN virus replicated at a titer of about 10 6 PFU / ml to enable the preparation of the DEN vaccine. and; 2) the cells have been used for the preparation of several DEN vaccines tested in Phase I clinical trials without side effects that may be associated with vaccine cell substrates; 3) FRhL cells are rhesus monkey lung diploid normal cells that have no oncogenic potential and are free of reverse transcriptase activity and contaminants; 4) There are no regulatory or other requirements for purification of the vaccine because the cells are "normal" diploid cells; 5) FRhL cell banks can be established in the cell generation available for vaccine manufacture starting with the low passage number of cells available. Thus, PDK passages provide an excellent model for those who want to study the empirical process of selective attenuation. However, since PDK serial passage represents a cumulative selection process, additional passages on other cell substrates provide their own selection pressure. Whether FRhL passage increases or decreases viral toxicity in humans is not known. The use of stable cells that must be fully characterized is only attractive once. However, published experience with FRhL cells has shown that the cells can completely alter or destabilize the biological properties obtained during serial passage in PDK (Halstead et al., 1984, Am J Trop Med Hyg 33, 654). -665; Halstead et al., 1984, Am J Trop Med Hyg 33, 666-671; Halstead et al., 1984, Am Trop Med Hyg 33, 672-678; Halstead et al., 1984, Am Trop Med Hyg 33, 679-683; Eckels Et al., 1984, Am J Trop Med Hyg 33, 679-683). [60] Adaptation of PDK-passage virus to FRhL was uniformly successful in all strains of DEN virus and was not dependent on PDK passage. Virus titers from harvests of FRhL passages 1-4 ranged from 10 5 -10 6 PFU / ml. By the third to fourth FRhL passages, vaccine lots of all DEN strain set viruses were prepared and tested as shown in Table 2. Data for the FRhL cell bank test, and master and production seed tests are also provided in Table 2. The test results required to ensure safety and contamination free were either negative or within acceptable limits. For DEN-4341750 PDK-20 producing seeds, monkey neurotoxicity tests were performed. The results of this study can be found in Hoke (1990) Am J Trop Med Hyg 43, 219-226. The DEN-4 producing seeds used for comparison, and the DEN-4 parental virus, were not neuropathogenic. For the remaining candidate DEN vaccines, whether there is a need to assess for neurotoxicity remains questionable based on this experience and data from other DEN monkey neurotoxicity tests (personal communication). [61] Example 2 [62] Rhesus monkeys inoculated with PDK-passage DEN virus. [63] The infectivity of the DEN virus passaged in PDK cells and represented as the "main set" was compared with the unmodified parental virus in each serotype. Table 3 lists the results of this study, which measures the degree of infection in monkeys by the number of days of viremia that can be found in serum taken sequentially two weeks after inoculation. In groups of 3-4 monkeys inoculated with DEN-1, DEN-2, DEN-3, and DEN-4, the average number of days of viremia when vaccinated with parental virus was 6.8 days, 5 days, and 3 days, respectively. , And 4.7. For DEN-2 mothers, additional data (not shown) was specified that infections including measurable viral viremia were highly reproducible over time using similar monkey and isolation techniques. Unfortunately, there is only partial data on viral titers in monkey serum. Most of the data present comes from experience with monkey viremia blood using titrated DEN-2 parent virus in mosquito cell culture. Peak virus titers at 4-8 days post inoculation reached 10 5 PFU per ml of serum (Putnak et al., 1996, supra). [64] For each strain set, PDK passages modify the DEN virus as indicated by the reduced ability of the virus to infect monkeys. For several strain sets, this is clearly demonstrated by the complete lack of viremia at the highest PDK passages. Inoculation of monkeys with DEN-1 in PDK passage 27 resulted in viremia on day 0 in four monkeys. This translates to zero isolation from all 56 bleeding bodies tested. Similar results were seen with DEN-3 PDK-20 and PDK-30. In PDK-30 for this virus, all evidence of monkey infectivity was lost (ie, no viremia in monkeys vaccinated with 10 6 PFU of virus, no evidence of seroconversion). The DEN-2 strain required the largest number of PDK passages to obtain strains of monkey infectivity. For the virus, more than 40 passages in PDK cell culture were required to reduce viremia. In contrast to the above experience, DEN-4 strain 341750 only required 6 passages in PDK cells for monkey infection modification. In another DEN-1 strain, 1009, even after 50 PDK passages, there was no evidence of monkey infection modifications compared to the parental virus (data not shown). In conclusion, PDK cell passage has been shown to be an effective empirical method for the modification and attenuation of various DEN isolates. This is an unnatural host for DENs that places selection pressure on a population of viruses that are probably suitable for PDK replication but not necessarily for replication in target cells in monkeys and humans. [65] Materials and Methods of Candidate Vaccine Research in Humans [66] Applicants . Healthy male and female volunteers aged 18-45 years old were treated for hematology, hematology, prothrombin time, partial thromboplastin time, urination, rapid plasma lysine antibodies, and antibodies against hepatitis B virus surface antigen and HIV. Irradiation and screening was carried out by a test panel containing serology. Candidates were excluded on the basis of persistent and significant abnormality or positive testing. Female volunteers were eligible to participate if the pregnancy test was negative within 48 hours of vaccination and willingly signed an agreement to avoid pregnancy using conventional contraceptive methods for 3 months after vaccination. In addition, if the volunteer already has flavivirus immunity that may affect the response to the dengue vaccine (Scott, 1983, J Infect Dis 148, 1055-1060), or neomycin, streptomycin, or gentamicin If you already have a history of allergies, they are excluded. Prior flavivirus immunity does not have any detectable hemagglutination inhibitory antibody (serum dilution of 1:10) against dengue type 1-4, Japanese encephalitis, or yellow fever, or yellow fever vaccine or flavivirus infection It was defined as having no history at all. [67] Applicants scored ≥70% on written tests designed to test knowledge of all aspects of clinical trials. Thereafter, an informed agreement was obtained from each of the applicants according to [US 21 CFR Part 50-Protection of Human Subjects]. Clinical protocols include all relevant regulatory requirements, including Declaration of Helsinki (Protocol), and Army Regulations 70-25-Use of Volunteeers as Subjects of Research, and 40-7-Use of Investigational Drugs in Humans and the Use of Schedule I Controlled Substances. Followed. The study was conducted in Human Subject Research Review Board, Office of the Surgeon General, U.S. Approved by the Army, the WRAIR Human Use Research Committee, and the Institutional Review Board, University of Maryland (Baltimore). [68] Study vaccine . Study vaccines are listed in Table 4. Vaccine virus was repeatedly passaged in primary dog kidney cells followed by fetal rhesus monkey lung (FRhL) continuous diploid cell cultures as three final passages to prepare seeds and vaccines. Each candidate identified prior to attempts at the volunteers a substantial reduction in viremia in vaccinated rhesus monkeys compared to their wild type parental virus. Appropriate attenuation, measured by infection of rhesus monkeys, has shown that the dengue vaccine strain is a suitable vaccine for human testing. [69] Just before immunization, lyophilized vaccine vials were reconstituted with sterile water for injection (USP). After immunization, the unused portion of the rehydration vaccine was kept on ice and titrated within 4 hours in LLC-MK 2 cell monolayers (Sukhavachana et al., 1966, Bull WHO 35, 65-66). Each volunteer was awarded 1.0 × 10 5 to 4.5 × 10 6 pfu of virus depending on the candidate vaccine to be injected (Table 4). The passage history of individual study vaccines is summarized below. [70] WRAIR live attenuated dengue vaccine vaccinePDK Passage *yearStudy placeNumber of applicantsDose (x 10 5 pfu) Dengue 1 (45AZ5)271991CVD104.4-45 2019911992CVD #107.7-38 1019911992CVD92.8-3.501984USAMRIID ##2 Dengue 2 (S16803)501991CVD36.8 401996USAMRIID35 3019911992CVD105.6-10 Dengue 3 (CH53489)201992CVD61.0-1.4 101992CVD33.801986USAMRIID2 Dengue 4 (341750)201989USAMRIID81.0 151991CVD34.8 all10--69- * Primary dog kidney passage level # Center for Vaccine Development, University of Maryland, Baltimore ## United States Army Medical Research Institute of Infectious Diseases, Frederick, MD [71] Dengue 1 45AZ5 Vaccine : DEN-1 strain West Pac 74 was isolated in 1974 from a human case of DEN fever on Nairu Island (West Pacific). The isolate was passaged 20 times in FrhL cell culture and a vaccine lot was prepared. Passages included mutagenesis and plaque selection to recover attenuated viruses suitable for human vaccination. [72] After vaccination of two human volunteers, it was decided to stop using the vaccine due to DEN disease in one of the volunteers. The vaccine was further attenuated by passage in PDK and FrhL cell culture. The current candidate vaccine is DEN-1 45AZ5 PDK-20. [73] Dengue 2 S16803 Vaccine : Dengue 2 strain S16803 virus was derived from Thai virus isolate from a patient with dengue fever. For seed and vaccine preparation, the virus was passaged PDK a total of 50 times and finally passaged in fetal rhesus monkey lung diploid cells (DBS-FRhL-2). Initially, two vaccine candidates were prepared at the 30th and 50th PDK passage levels and selected for testing. Another vaccine candidate was developed in WRAIR from the same Dengue 2 parental strain S16803 virus and was made at the 40th passage level by the Salk Institute (Swiftwater, PA). [74] Type 3 Dengue CH53489 Vaccine : Type 3 Dengue strain CH53489 virus is derived from a Thai strain and is 30 times in primary dog kidney (PDK) cells after initial passage in primary green monkey kidney (PGMK) and C6 / 36 insect cells. Passed. Viruses from 10, 20 and 30 PDK passages were used to inoculate fetal rhesus monkey lung diploid cell cultures. [75] Dengue 4 341750 Carib Vaccine : The dengue 4 vaccine candidate was derived from a Caribbean strain of dengue 4 (Columbia, 1982), passaged at the University of Hawaii, and manufactured at WRAIR [Marchette, 1990, Am J Trop Med Hyg 43 , 212-218. Antibodies to the parental virus neutralize other dengue 4 virus strains, including H-241, the prototype strain. Attenuation of human isolates was achieved by 20 passages in primary dog kidney (PDK) cell culture. [76] Research design . A standard randomized single-blind inpatient clinical protocol was used for all pilot studies. Most of the research was conducted at the Center for Vaccine Development, University of Maryland, Baltimore, Maryland. Pilot studies of Dengue 2 S16803 PDK 40 vaccine and Dengue 4 CH341750 PDK 20 vaccine were performed at the Medical Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Ft Dentrick, Maryland, USA. [77] In the initial clinical study of the vaccine, three volunteers were first tested with the largest passage available for a particular strain. Symptoms were closely monitored for 3 weeks, and if the volunteers were still good, the next lower passage was tested. If one or more volunteers suffered, lower passage testing of the vaccine strain was not performed because the lower passage would have been less attenuated. After all acceptable passage levels were tested in three volunteers, the lowest level that did not cause illness was chosen to further test up to seven additional volunteers. [78] To enable careful observation, to prevent exposure to foreign infectious diseases, and to prevent possible vector mosquito infections, volunteers were limited to the study ward from 3 days before inoculation to 20 days after immunization. Regardless of severity or whether all adverse experiences were associated with immunization, all adverse experiences that occurred during this period after administration of each vaccine were recorded. Acceptable safety of the vaccine was first defined as the absence of the following serious side effects: any serious clinical illness not explained by a diagnosis not associated with vaccination; Continuous heat (oral temperature ≧ 38.5 ° C. in four measurements over 24 hours, maximum daily oral temperature ≧ 38.5 ° C. in three consecutive days, or temperature above 40 ° C. in any individual measurement); Thrombocytopenia (<100,000 platelets / mm 3 ), leukopenia (absolute neutrophil count <1000) in two consecutive measurements; Or serum amino alanine transferase (ALT) levels greater than four times normal on three or more consecutive days (which are not otherwise described). In addition, experiences that suggest significant side effects that could be associated with the use of the vaccine were recorded as serious events. [79] On day 0, 0.5 ml of undiluted vaccine was subcutaneously inoculated to the volunteers. After immunization, vital signs were recorded every 6 hours. The injection site was examined and measured daily for maximum diameter and sclerosis of erythema. First 20 years after immunization of clinical signs (fever [> 37.8 ° C.], rash, vomiting, bleeding, and liver and spleen dilation) and symptoms (boredom, headache, myalgia, arthralgia, nausea, and ocular pain or photophobia) Evaluated daily for days. Symptoms were graded as mild (noticing symptoms but continuing surveillance) or as severe (forcing beds by symptoms). When volunteers requested, painful symptoms were treated with propoxyphen hydrochloride; No antipyretic was used. The observations were recorded on a normal checklist of symptoms and physical findings. On day 21, volunteers were released from study surveillance and asked to return for serological studies 1, 6, 12 and 24 months after inoculation. [80] Two healthy flavivirus-immune volunteers were immunized with the parent strain of Dengue 1 45AZ5 vaccine in USAMRIID and two years later with the parent strain of Dengue 3 CH53489 vaccine. Medical records from the study were reviewed for the presence or absence of the following signs and symptoms: fever, rash, boredom, headache, myalgia, arthralgia, and ocular pain or photophobia. Viremia was measured daily. In contrast to the attempts of the present invention, symptoms were not systematically recorded and the intensity of symptoms was not graded. In addition, the clinical experience with Dengue 4 341750 Carib PDK 20, given to eight volunteers in USAMRIID during the later study, was summarized and compared with that of those who received the current immunization [Hoke, 1990, supra]. [81] Laboratory evaluation . Hemoglobin and hematocrit, leukocyte cell counts using differential counts, platelet counts, and every other day for routinely available medical tests at aspartate aminotransferase (AST) and alanine aminotransferase (ALT) levels. Blood was collected from volunteers on day. In addition, blood was collected every other day for 20 days for virus isolation and antibody studies. Blood (20 ml) was allowed to coagulate for 2 hours at 4 ° C. and serum was decanted into 1-ml aliquots and stored frozen at −70 ° C. until study. [82] Virus isolation . For determination of dengue virusemia, serum was thawed and seeded on C6 / 36 mosquito cell monolayers and incubated at 28 ° C. for 14 days. Viruses were analyzed for supernatant culture harvests (Sukhavachana et al., 1966, Bull WHO 35, 65-66) by plaque analysis on LLC-MK 2 cells. To quantify the amount of virus in serum, plaque assays were performed on C6 / 36 clones of Aedes albopictus mosquito cells (Hoke, 1990, supra). Plasma dilutions were inoculated into the cell culture flasks and adsorbed at 35 ° C. for 1-2 hours. Hank's Balanced Salt Solution and overlay medium consisting of 0.75% agarose, 5% lactalbumin hydrolyzate, 0.12 M NaHCO 3 , and antibiotics were added and all flasks were incubated at 35 ° C. After 7 days, the flask was stained with 5% liquid neutral red for 3-5 hours. After 18 hours extra pigmentation was removed and plaques were read. [83] Serology. Antibody tests include ELISA, HAI, and plaque reduction neutralization test (PRNT) performed using the same serotype dengue virus as strain in the vaccine being tested. Detection of anti-dengue IgM antibodies is carried out by modification of ELISA, where values above 0.10 OD units are considered positive [Innis, 1989]. HAI testing is performed by standard techniques modified with microvolume using 4-8 units of independent antigen, using serum extracted with acetone to remove inhibitors [Clarke and Casals, 1958, Am J Trop Med]. Hyg 7, 561-573]. PRNT analysis was performed by the method described by Russell et al. [Russell, 1967, supra]. [84] Statistical analysis. Cochran-Armitage test for trends and Spearman's correlation, respectively, for the dengue 2 vaccine S16803 (PDK 30, 40 and 50) and dengue 3 vaccine CH53489 (PDK 10 and 20), frequency and extent and passage of response The relationship between them was analyzed. Independently analyzed symptoms and signs included eye symptoms, headache, boredom, myalgia, arthralgia, rash and the number of days of fever (temperature> 37.8 ° C.) and their presence or absence. The null hypothesis that higher PDK levels are not related to lower response is assessed with a 5% probability. Examining the data, the optimal passage for each virus was determined based on the clinical and immunological response of each volunteer. According to the US Army Medical Research and Development Command's Flavivirus Vaccine Steering Committee, passages that immunized about 80% of volunteers without causing unacceptable side effects were selected for further development. [85] Definition of infection by vaccine [86] Infection with the vaccine is defined as replication of the dengue virus in the volunteer, which is detected by the presence of post-immune serum form-specific neutralizing antibodies or IgM anti-dengue antibodies. Viremia is not included as essential for infection diagnosis because it is never detected in the absence of an antibody response. Vaccine failure is defined as an unacceptable reverse clinical response or failure to develop IgM or PRNT antibodies in the recovery phase. [87] Example 3 [88] Clinical response to attenuated dengue vaccine [89] Dengue 2 S16803 Vaccine [90] Dengue 2 strain S16803 virus generated from passage 50 of PDK cells was tested in three volunteers. The volunteers were healthy with oral temperature not exceeding 38.0 ° C. Two of the three volunteers had transient mild symptoms of boredom, headache, and eye symptoms (eye pain or photophobia). Experimental findings included mild ALT elevation (<2 × normal) in two of the three volunteers and mild leukopenia in one of the three volunteers. Due to the acceptable safety profile of the PDK 50 vaccine, the next lower acceptable passage, PDK 30, was selected for clinical evaluation. [91] PDK 30 vaccine tested in 10 patients was underattenuated and developed symptoms that were mild to some degree compatible with dengue. Four volunteers (40%) had a low grade fever over 9-14 days post vaccination (average 12 days) to a maximum temperature of 38.5 ° C. 80% caused a rash. While most volunteers experienced eye symptoms (10/10), headaches (9/10), and malaise, 70 percent had one or more headaches, severe eye and photophobia, severe symptoms of boredom or muscle pain. . Three volunteers slightly elevated their alanine aminotransferase (ALT), a measure of liver pathology. [92] The PDK 30 vaccine was generated from the master seed because the PDK 30 vaccine was so reactive that it could not be further tested on volunteers. Two of the three volunteers vaccinated with PDK 40 had mild dengue syndrome for 9-10 days with low grade body temperature (<38.1 ° C.), rash, myalgia, and headache after vaccination. Symptoms resolved spontaneously over several days without drug demand or disability. The accompanying symptom was an unexpected elevation of serum liver enzymes, one at a maximum ALT level of 199 IU / ml (4 times normal) and the other at 77 IU / ml maximum ALT (1.5 times higher than normal). The third volunteer was asymptomatic, but the ALT also doubled (up to 10 2 ). All experimental abnormalities resolved within days without intervention, and all volunteers returned to health within 21 days of receiving the vaccine. Due to the abnormal frequency of hepatitis development associated with the PDK 40 vaccine, no further development of this product is planned. [93] Table 5 summarizes the initial clinical experience with the WRAIR Dengue 2 vaccine. Reduced frequency of signs of fever and rash are evident between passage 30 and 50 vaccines. In addition, although the oral temperature falls from the maximum of 38.5 ° C to normal as the passage is increased, there is no change in the exothermic period exceeding one day. For the Dengue 2 vaccine, the frequency and duration of eye symptoms, rash, headache, boredom and myalgia were significantly associated with passage. [94] Clinical Response in Dengue 2 S16803 Virus Vaccine Recipients PassageennuiheadacheMuscle painArthralgiaInner sxrashExothermic T> 37.8 ℃Fever days (average)Fever 2-S16803-309/109/107/104/1010/108/104/109-14 (12)38.5 2-S16803-402/32/32/31/31/32/31/38,938.0 2-S16803-502/32/30/31/32/30/30/3-- [95] Symptoms-days [96] PassageennuiheadacheMuscle painArthralgiaInner sxrashFever T> 37.8 ℃ 2-S16803-302.23.62.41.73.35.40.5 2-S16803-402.01.72.01.05.71.70.7 2-S16803-500.60.70.00.31.00.00.0 [97] Clinical Response to Dengue 3 CH53489 Virus Vaccine Recipients A: number of patients responding vaccineennuiheadacheMuscle painArthralgiaInner sxrashT> 37.8 ℃ (days)Fever 3-CH53489-02/22/22/21/22/22/22/2 (5-9)40.6 3-CH53489-101/32/32/31/31/32/31/3 (10,11)38.2 3-CH53489-203/65/63/64/64/61/61/6 (3)38.7 [98] B: days of symptoms vaccineennuiheadacheMuscle painArthralgiaInner sxrashT> 37.8 ℃ (days) 3-CH53489-03.54.04.52.03.57.55.0 3-CH53489-100.33.32.31.01.36.30.7 3-CH53489-201.72.81.02.01.30.80.2 [99] [100] [101] a: primary dog kidney passage [102] b: defined as anti-dengue IgM positive or PRNT50 seroconversion rate [103] c: defined as neutralizing antibody titer> 1:10 (PRNT50) [104] Suggested strains for expanded clinical studies [105] Dengue 3 CH53489 vaccine. The Dengue 3 vaccine (CH53489, PDK 0) developed at WRAIR was administered to two healthy yellow fever-immune male volunteers as a 0.5 ml subcutaneous inoculation of virus 2 × 10 4 pfu. There was no immediate post-immune process. By day 6, both volunteers had some severe dengue fever characterized by high fever, chills, myalgia, headache, boredom, and diffuse erythematous rash. Both volunteers had thrombocytopenia and leukopenia, but there were no signs of hemorrhagic fever. After a five-day fever period, both men recovered rapidly and were complete around 21 days. Due to the serious illness experienced by both patients, no further testing of the passage was made. Subsequently, PDK 10 and PDK 20 passages were prepared as vaccine candidates. [106] PDK 20 vaccine was given to 6 volunteers, resulting in mild response. One patient experienced premature fever on day 3 with transient fever (up to 38.2 ° C.), pharyngitis, and cervical lymphadenopathy. No dengue virus was isolated from the sera of volunteers. The patient was thought to have a concomitant illness with a fever that was not directly related to vaccination. 4 of 6 volunteers had short-lived mild dengue symptoms without rash; Arthralgia, ocular pain, and headache were the most frequent complaints. However, one volunteer suffered from more severe symptoms of headache, boredom, and ocular pain for three days. In addition, he suffered from leukopenia and maintained a rise in ALT levels; The laboratory abnormalities were resolved on day 31. Another volunteer had a small, reversible, ALT rise below 2 × normal. Since PDK 20 vaccines are safe with marginally acceptable response, the next lowest available passage vaccine virus (PDK 10) was tested. [107] PDK 10 virus has proven to be too reactive for the recipient. One of the three volunteers had a low grade fever on days 10 and 11 (up to 38.3 ° C.) and a flowering rash on day 13. Another volunteer suffered from persistent itching associated with waxing and Waning rashes and weak cervical and axillary lymph nodules 6 to 9 days after vaccination. He then suffered from a speculative rash with 10 to 12 days of boredom, headache and muscle pain. The volunteer developed a typical dengue disease after showing a specific constitutional allergic reaction to the vaccine. The two volunteers also exhibited laboratory abnormalities of <2 × normal elevated ALT levels and leukopenia, which were resolved on day 31. [108] Table 6 summarizes the response to the Dengue 3 CH53489 vaccine. Despite the tendency of less frequent and shorter duration of signs and symptoms with passage, passage did not reach statistical significance in either analysis. [109] Dengue 4 341750 vaccine. Eight volunteers received PDK 20 vaccine [Hoke, 1990, supra] 10 5 PFU. Five volunteers suffered from little-identified spots, bleaching rashes and slight temperature rises (up to 38.1 ° C). Viremia and antibody responses were also seen in these 5 volunteers (63%). [110] In anticipation that lower passages may be more infectious, a new DEN-4 341750 candidate vaccine was prepared from PDK passage 15. Three volunteers received the vaccine and two experienced mild symptoms. The third volunteer suffered suddenly with fever, swelling of the face and limbs, severe fatigue, rash, ocular pain, photophobia, and arthralgia on day 8. Over the next three days, fever continued up to 39.6 ° C., but signs and symptoms resolved spontaneously. Because of the serious side effects of vaccination, the further use of the PDK-15 vaccine was terminated and PDK-20 was selected for further evaluation. [111] Example 4 [112] Immune Response to Viremia and Attenuated Dengue Vaccines [113] Table 7 describes the viremia and immune response with the WRAIR Dengue vaccine. The infectivity of each vaccine is summarized below. [114] Dengue 2 S16803 vaccine. Although all recipients of the PDK 50 vaccine had no viremia, two out of three developed low-titration neutralizing antibodies by 60 days. The finding suggests that the infectivity of vaccine viruses to humans has been reduced. In contrast, two of the three Dengue 2 PDK 40 vaccinates suffered from apparent viremia, and all of the high titer antibodies developed after vaccination. As desired, the dengue 2 PDK 30 vaccine had the highest infectivity: viremia was detected in all 10 volunteers and all volunteers were transfected by neutralizing antibody titers> 1:60 by 60 days. [115] Dengue 3 CH53489 vaccine. Dengue-3 virus, which retains the small plaque phenotype and temperature sensitivity of the vaccine virus, was recovered from two yellow fever immune recipients of the Dengue 3 PDK 0 vaccine for 6 days and 7 days. High titer PRNT50 and hemagglutination inhibitory (HAI) antibodies with secondary-infectious cross reactivity were then measured in serum collected on days 30 and 60 in both volunteers. Infectiousness was similar in patients receiving the Dengue 3 PDK 10 attenuated vaccine: two of three had viremia and all developed vaccination-inducing neutralizing antibodies. In contrast, two out of six dengue 3 PDK 20 vaccinates suffered from detectable viremia, then three volunteers seroconverted to reflect reduced infectivity. [116] Dengue 4 341750 vaccine. Eight volunteers received PDK 20 vaccine 10 5 PFU, and five had viremia and antibody responses (63%). The vaccine, PDK 15, prepared from the low passage of the candidate was more infectious. Viruses were isolated from single volunteers at 8 and 10 days post vaccination with a maximum titration of 15 pfu / ml. Subsequently, the volunteers neutralized antibody titration 450 with the secondary HAI reaction, Preexposure to Louis encephalitis virus was found (PRNT titration 1:20 before vaccination). Two volunteers without detectable viremia were neutralized titrated at 1:10 and 1:40 by 30 days post vaccination. [117] Example 5 [118] Selection of candidate vaccine [119] An extension program for testing the safety of WRAIR PDK-attenuated vaccines is shown in Table 8 listing the salient features of the vaccine for each serotype. As a result of increasing PDK passages, the mean disease score decreases, which assesses the number and duration of symptoms per volunteer. In addition, elevated PDK passage is also associated with reduced mean days of viremia, except for the Dengue 4 vaccine. Of the dengue 2, 3, and 4 vaccines tested, only one passage was considered safe and tolerably reactive and suitable for extended clinical studies: dengue 2 PDK 50, dengue 3 PDK 20, and dengue 4 PDK 20. However, the percentage of infected recipients declined with increasing PDK passages. Serum conversion, defined as a percentage with neutralizing antibody titration ≧ 1: 10, declined similarly within a wide confidence interval. [120] Argument [121] WRAIR has long been involved in the development of live, attenuated dengue vaccines. Both the WRAIR and Mahidol dengue vaccine programs have developed several live vaccines by attenuation through several passages (PDKs) of the dog's kidney cells (repeated growth in tissue culture). Preliminary test results from a small number of volunteers established the safety of the WRAIR candidate vaccine. None of the 65 recipients needed emergency care and did not sustain severe damage. Three volunteers experienced transient specific reactions related to dengue vaccination and the vaccine received from further clinical development was recovered. Despite the inconvenience, experimental infection with attenuated vaccines was acceptable. [122] Clinical trials showed that increasing PDK passage of the vaccine virus increased attenuation for volunteers. The effect is best shown with dengue 1 and dengue 3 viruses, where the parental non-passive viaus results in unmodified dengue fever and subsequent 20 PDK passages acceptable reactiveity. However, increasing PDK passage reduced the infectivity of the vaccine virus, which reduced immunogenicity. In addition, reduced viremia with vaccine virus in humans appears to correlate with that in rhesus monkeys (except Dengue 4 PDK 15). The findings suggest that the infectivity of the attenuated dengue virus vaccine in volunteers has proven to be equivalent to immunogenicity. The relationship between passage and reactogenicity should be carefully understood because patients experiencing one symptom will experience several symptoms. Since our assay assumes the independence of these symptoms, the understanding described in the independent p-values may be poor. In addition, we believe that the rash was strongly associated with the passage (p = 0.009 independent of existence and p = 0.01 for duration). This is supported by the lack of a significant correlation between rash and other symptoms for dengue 2 or 3 vaccines (Spearman's test). [123] Only vaccines with an acceptable safety profile are selected for extended clinical testing: Dengue 1 45AZ5 PDK 20, Dengue 2 S16803 PDK 50, Dengue 3 CH53489 PDK 20, and Dengue 4 341750 PDK 20. Serum conversion due to few volunteers Due to the wide confidence interval in, the subsequent study increased the number of recipients of each of the four selected vaccines. In addition, another test will determine whether the immunogenicity of the attenuated vaccine can be elevated through the administration of two doses instead of the single dose used in the study. [124] Example 6 [125] Extended studies of monovalent vaccines; Monovalent vaccines provided as two doses; And monovalent vaccines mixed as tetravalent agents provided in one and two doses. [126] Study Design: The objective was to evaluate the safety and immunogenicity of four monovalent vaccines given on a single dose followed by a two-dose vaccination schedule. Combination 4 then assessed the safety and immunogenicity of the vaccine. Patients were recruited from two locations, the University of Maryland in Baltimore and WRAIR in Washington, DC. The first group of 22 patients was divided into four or four groups of four who received single dose of monovalent dengue or yellow fever 17D virus (Connaught), respectively. The 17 D yellow fever vaccine served as a control and reference point for reactogenicity. Another 31 patients were divided into four groups of seven to eight who provided one monovalent vaccine in two doses, one half a month and the other half a month. Finally, 10 volunteers were given a tetravalent vaccine in two or three doses. The first four tetravalent recipients were vaccinated at 0 and 1 month. The latter six tetravalent recipients were vaccinated at 0, 1 and 4 months. All patients except ten tetravalent vaccine recipients received vaccine serotypes randomly and in a double-blind manner. [127] Patients: Patients were normal healthy adults aged 18 to 50 years. All patients were seronegative for hepatitis B, C and HIV. All patients were seronegative for dengue 1-4, JE, SLE, and YF by the hemagglutination inhibition assay prior to the start of the study. [128] Vaccines: Four serotype vaccine candidates were isolated from scratch from people with clinical disease. Each was then modified by serial passage in primary dog kidney (PDK) followed by fetal rhesus monkey lung cells as described above. The volunteers were selected based on preliminary experiments in human volunteers. Each lyophilized monovalent vaccine was reconstituted with sterile water and provided in a volume of 0.5 cc. Doses of serotypes 1-4 were 10 6 , 10 6 , 10 5 and 10 5 pfu of dengue 1, 2, 3, and 4, respectively. The tetravalent vaccine dose was prepared by mixing each reconstituted monovalent 0.25 cc and provided a final volume of 1 cc. The dose of the tetravalent vaccine was 1.1-2.8 × 10 6 pfu. All vaccinations were given subcutaneously in the upper part of the arm. [129] Clinical safety: Response to vaccination was assessed by a combination of daily symptom recording and periodic physician evaluation for 3 weeks after each vaccination. Patients were stayed in the study period for detailed observation during the period of 1 week post-vaccination, which was the closest period of response and viremia in that period. The patient was examined and specifically asked about fever, chills, headache, orbital pain, myalgia, arthralgia, rash and other symptoms. Each symptom can be at the stage of 0 (none), 1 (does not affect normal activity; does not require medication), 2 (requires medication or changes in activity), or 3 (pathologically stable or alleviated by medication) Was rated as. The most common symptoms were classified into four categories. These categories are as follows: gastrointestinal disorders including 1) subjective fever and chills, 2) headache and orbital pain, 3) myalgia and joint pain, and 4) nausea, vomiting and abdominal pain. The symptom index of each category was calculated by multiplying the duration of symptoms manifested at the highest symptom grade of each day and after several days. If symptoms have occurred for 24 hours, they correspond to a period of 1 day. The response factor index (RI) is simply the sum of the symptom indices for each category. RI summarizes the vaccine response for each patient. The symptom category index and RI allow for semi-quantitative comparisons of vaccine response and vaccine serotypes between patients. [130] The blood and liver toxicity of patients was investigated during the study with a series of CBCs, platelet counts, AST and ALT. [131] Severe adverse events are defined as serious illnesses that lack other possible causes, such as fever exceeding 38.5 ° C continuously for more than 24 hours or Tmax above 38.5 ° C or more than 104 ° C daily oral temperature above 104 ° C, neutropenia less than 1,000 / ml Or thrombocytopenia less than 90,000 / ml, or when serum ALT or AST is more than five times normal. [132] Immunogenicity: Hemagglutination inhibition assay was performed using Clarke and Cassals, 1958 (Am J Trop Hyg 7, 561-573) method, and dengue IgM and IgG were measured by capturing ELISA in almost the last six tetravalent patients. Dengue and yellow fever neutralizing antibodies were determined by plaque reduction neutralization tests on days 0 and 30 after each vaccination. Endpoint quantitation studies measure 30 days of neutralizing antibodies after the final vaccination. Neutralizing antibody seroconversion is defined as a 50% reduction in plaque at a serum dilution ratio of at least 1: 5. Viremia is measured with serum at 7-14 days after the initial and secondary vaccinations. The method used for virus isolation uses LLC-MK 2 or C6 / 36 cells for amplification and Vero for plaque formation and delayed plaques adopted in Yuill, 1968 (Am J Trop Med Hyg 17, 441-448). It was how. [133] One and two dose studies were combined for this report. Patient characteristics are shown in Table 9. A total of 59 normal patients received the dengue virus vaccine; 49 received a monovalent test substance and 10 received a tetravalent vaccine. Four received a certified 17D yellow fever vaccination (Connaught). [134] Patient features vaccineThe number of patients (the number of two doses)castleraceAverage age Den-112 (8)7M / 5F6W / 6B32 Den-212 (8)7M / 5F7W / 5B36 Den-313 (8)9M / 4F8W / 5B36 Den-412 (7)6M / 6F4W / 6B / 1H / 1AmI33 4th4 (4)3M / 1F4 W26 YF 17D4 (0)3M / 1F3W / 1B30 [135] Example 7 [136] Reactivity [137] Local reaction. Of the 59 dengue vaccine recipients, 19 (32%) had mild arm pain at the injection site. Seven of them received DEN-1, four DEN-2, one DEN-3, one DEN-4, and five four. Only 5 patients experienced pain at the injection site after 24 hours. None affected the use of the arm. [138] Systemic reaction. Of the 59 dengue recipients, 20% had no symptoms at the first vaccination but 70% of the patients were asymptomatic at the second vaccination. Four patients who received the third dose had no symptoms associated with it. The most commonly reported reactions in dengue vaccination are headache and muscle pain. They vary in severity. 1 is the incidence of grade 1 or higher symptoms in the first vaccination, which causes changes in daily activity or undergoes medication for stability. After the first dose of the vaccine, five patients (8%), one serotype 1, one serotype 4, and three quadrants had severe grade 3 symptoms of colds, muscle pain, headache or nausea lasting less than one day. . None of the patients had grade 3 symptoms of re-vaccination. [139] RI is in the range of 0 to 35. Table 10 compares the reported reactivity of each vaccine. DEN-1 monovalent and tetravalent vaccines are associated with more reactive. Secondary or tertiary administration of all dengue vaccines resulted in almost constant response, even in those patients with moderate to severe symptoms from initial vaccination. [140] Average reactive index vaccineTotal number of patientsFirst dose RI (n)Second dose RI (n)3rd dose RI (n) Den-1127.4 (12)0.5 (8)- Den-2123.8 (12)0.3 (8)- Den-3132.9 (13)0.8 (8)- Den-4123.7 (12)0.5 (6)- 4th109.3 (10)1.9 (10)0.0 (4) YF 17D43.8 (4)-- -= Not implemented [141] 2 shows the frequency distribution of RI by serum. Eight patients (14%) developed fever (> 100.4 ° F). Of the eight, four received DEN-1, one DEN-2, one DEN-3, and two four. The highest and longest fever was seen in TDEN 103.3 F and 3 days in DEN-1 recipients. Only one other patient who received DEN-1 had more than one day of fever. Seven of eight fever attacks followed the first vaccination. [142] Sixteen patients (27%) had a generalized rash including torso and limbs at the first vaccination. The rash is usually erythematous, punctate papules and oily benign. Of the 16 people with generalized rashes, only 7 had fever. Of the 16 patients with rash, five received DEN-1, two DEN-2, one DEN-3, three DEN-4 and five quartet. The rash is typically pronounced 8-10 days after vaccination and resolves 3-4 days later. None of the patients developed spotting, purpura or scarring. None of the patients developed rashes from re-vaccination. [143] Gastrointestinal symptoms are relatively common, occurring in one third of the patients, but soft and simple, lasting no more than 24 hours. One DEN-4 recipient developed severe nausea associated with convulsive abdominal pain for one day. [144] Six patients (10%), five dengue and one yellow fever 17D recipient, developed transient neutropenia with an absolute neutrophil count below 1000 / ml. The minimum was 288 in DEN-1 patients. Neutropenia usually resolves after 2-3 days. None of the patients developed thrombocytopenia. There was no clinically significant increase in AST or ALT. [145] None of the patients had clinical evidence of dengue hemorrhagic fever, as expected in a group of non-immune adults who received primary dengue virus exposure. [146] Example 8 [147] Immunogenicity [148] Viremia was detected in 10 patients (17%), one with DEN-2, four with DEN-3, one with DEN-4 and four with tetravalent. No DEN-1 viremia was detected. The serotypes of virus isolated in tetravalent patients have not yet been identified. All detected viremia appeared after the first dose of virus. Strangely, fever was present with only viremia in three tetravalent recipients. All viremia patients had neutralizing antibodies. One did not develop an IgM or IgG response even in viremia. [149] Table 11 summarizes the antibody response to monovalent vaccination. Neutralizing antibodies were detected more frequently than IgM and IgG. No seroconversion was detected by IgM or IgG, which was also not found by PRNT 50 at serum dilution ratio 1:10. When present, IgM was positive at 41% by day 14, 17% by day 21, and 42% by day 30 after vaccination. IgM typically peaked up to 30 days after the first vaccination. One exception occurred in tetravalent recipients, whose IgM peaked three days after the second vaccination. IgM can last more than three months. The seroconversion rates with neutralizing antibodies were 100%, 92%, 54% and 58% for monovalent serotypes 1, 2, 3 and 4 respectively. When present, neutralizing antibodies were typically detected up to 30 days after the first vaccination. No time zone between 0 and 30 was evaluated for neutralizing antibodies. Second doses of the vaccine resulted in more than four-fold increase in DEN-2 GMT, which was not observed in other serotypes. Two DEN-3 patients converted sera after the second dose of the vaccine, one at 1 month and the other at 3 months. They did not develop neutralizing antibodies after a single dose. Interestingly, the IgM / IgG pattern of these two patients presents a secondary response after the second dose, suggesting that the first dose has become immunologically sensitive. [150] Despite preliminary negative hemagglutination inhibition analysis of Dengue, SLE, JE and YF, 5 out of 53 (9%) tested had secondary antibody response patterns with an IgM / IgG ratio <1.8. All five were negative for the same dengue neutralizing antibody prior to vaccination. This suggests a previous potential exposure to flaviviruses. We did not find any significant differences between the mean RIs of the secondary and primary antibody responders (9.6 vs 5.8, p = 0.19). [151] There were 12 monovalent patients without IgM / IgG or neutralizing antibodies. One received DEN-2, six received DEN-3 and five received DEN-4. The mean reactivity of the non-responders group was less than 1, which was significantly different from the mean RI (0.9 vs 4.9, p <0.003) of form 2, 3 and 4 neutralizing antibody responders. [152] Our study consisted of 25 black and 31 white patients. There was no significant difference between the average RIs of these two ethnic groups. This is interesting because there is some epidemiological evidence that suggests the milder dengue disease severity among blacks. [153] Monovalent Vaccine Serum Conversion Rate by IgM and PRNT 50 vaccineSerum conversion after first doseFirst dose GMT -1 * Serum Conversion After Second AdministrationSecond dose GMT -1 Cumulative seroconversionIgM (+) PRNT 50 IgM (+) PRNT 50 IgMPRNT 50DEN-110/1212/12 (100%)6680/2-51310/1212/12 (100%) DEN-29/1211/12 (92%)1120/30/15599/1211/12 (92%) DEN-34/136/13 (53%)152/91/7166/137/13 (54%) DEN-45/127/12 (58%)170/70/595/127/12 (58%) YF 17D0/44/4 (100%)2935---0/44/4 (100%)* Use potency of 30 days after vaccination; Calculation uses 1 for voice titer-= not performed [154] Reactivity and immunogenicity of tetravalent vaccine recipients applicantVaccine plan (months)Reactive indexSerotype neutralizing antibodies measured 30 days after Dosing 1Dosing 2Dosing 3Dosing 1Dosing 2Dosing 3 330,1160-1,2,3,41,2,3,4- 340,100-21,2- 350,140-1,2,3,41,2,3,4- 360,1153-One1,3- 370,1,4200OneOne1,2,3 380,43514-1,21,2,3- 390,1,418001,3,41,31,2,3,4 400,1,4200OneOne1,3 410,1,4One20221,2,3,4 420,100-21,2- [155] Monovalent seroconversion rate and tetravalent multidose vaccineDEN-1 AbDEN-2 AbDEN-3 AbDEN-4 Ab Single dose once12/1211/126/137/12 One dose of tetravalent7/10 p <.056/10 p> .073/10 p> .43/10 p> .18 4-valent administration9/106/105/102/10 Four doses three times4/43/44/42/4 [156] Age, gender [157] Table 12 shows the results of PRNT seroconversion in ten tetravalent vaccine subjects. The first four subjects received two vaccinations at 0 and 1 month. One subject missed the second vaccination at 30 days and was vaccinated at 60 days. The other six subjects received vaccinations at 0 and 1 month and tertiary vaccination at 4 months if the response was incomplete. Both subjects produced neutralizing antibodies to serotypes after all four doses. The other two tetravalent receptors were serotyped into all four serotypes after vaccination at 4 months. The other two responded trivalently. A given tetravalent second dose at 1 or 2 months did not significantly increase seroconversion. The total seroconversion rates among these 10 tetravalent subjects were 100%, 80%, 80% and 40% for DEN-1, 2, 3 and 4, respectively. [158] Example 9 [159] One study, a two-level 2 4 factorial design, was designed to measure the interaction of each serotype composition in a tetravalent vaccine. [160] 54 subjects were given 15 permutations of 2 dose levels of each serotype. The results are shown in FIG. High dose H indicates undiluted vaccine in the range of 10 5 to 10 6 pfu / ml; Low dosing L represents a 1:30 dilution of the undiluted vaccine at about 10 3.5 to 10 4.5 pfu / ml. [161] Six subjects were pre-administered four vaccinated at 0 and 1 month. If the subject does not have a tetravalent neutralizing antibody response, the third dose is given at 4 months. The results are shown in FIG. [162] At 0 and 1 month 4 human subjects were injected with a mixed full-dose tetravalent vaccine. The endpoint was clinical stability and neutralizing antibodies at 1 month after the second vaccination. T-cell responses were measured in the first 4 subjects. The results are shown in FIG. [163] The results show that the tetravalent vaccine (16 formulations) was safe among 64 non-immune US volunteers. Reactivity was varied. Four formulations elicited trivalent or tetravalent neutralizing antibody responses from all volunteers. According to the monovalent experiment, the second dose of the tetravalent vaccine at 1 month did not induce noticeable reactogenicity but also increased the neutralizing antibody response. Terminal titration of neutralizing antibody reactions is ongoing. Memory interferon-gamma responses in T-cells can be measured in the absence of neutralizing antibodies. An interval of four months or more can result in an improved tetravalent seroconversion. [164] discussion [165] The vaccines appear to be weakened in humans compared to the traditional description of wild type dengue infection experiments (Simmons et al., 1931, Manila Bureau of Printing). To quantify the reactivity, a fractional scale based on the duration and severity of self-reported symptoms was used. The method tends to overestimate vaccine-related responses. Ideally it should be identified as a case of natural dengue infection. However, inaccurate RI also makes it possible to reasonably compare symptoms between individuals and groups. The results of the monovalent vaccine test showed that the degree of weakness may vary among four dengue vaccine candidates. 45AZ5 PDK20 was the weakest, causing the highest titers and uniform seroconversion. The DEN-2 candidate, S16803 PDK50, similarly resulted in nearly 100% seroconversion with a positive reactive profile. Den-3 and Den-4 had a low reactive profile, but seroconversion was only 50-60%. It should be noted that administration of types 3 and 4, lower immunogenic species is 10 times less than types 1 and 2. [166] Secondary administration of the virus is significantly less involved in the response. However, the benefits of secondary administration of monovalent vaccines at 1 or 3 months are small. Den-1 and 2 are already homogeneously immunogens and may not require further administration. Nevertheless, the GMT of Den-2 was more than quadrupled. This may be evidence of administration containing a sufficient antigen mass that elicits low levels of virus replication or amplification response after the second dose. The pattern of this neutralizing antibody response can also be seen as a second vaccination with 17D YF (Wisseman, 1962, Am J Trop Med Hyg 11, 570-575). The first dose of Den-3 is a secondary antibody response pattern, which can make two monovalent subjects seroconverted after the second dose. This suggests that neutralizing antibody assessments may not be sensitive enough to detect the proper immune response of type 3 vaccine candidates. Secondary administration did not add any new seroconverters to type 4. There was no obvious additional effect on the schedules administered and tested in conferring a second dose of monovalent DEN-1 or DEN-4. [167] Twelve monovalent subjects who did not have a neutralizing antibody response to the monovalent vaccine also did not react with measurable dengue IgM or IgG. All these non-responders apparently obtained viable virus in the same vial that had been replicated in another subject. They did not produce any response to vaccination. Thus, by all signs, there was no evidence of viral replication among these subjects. The mechanism for this non-reactivity is unknown. It may be the result of a lack of host substrate required for infection or an effective innate immunity. [168] The value of multiple administrations may be clearer in bio-attenuated vaccine combinations as a strategy for bypass viral interference. The administration of each component may be important here as well as the interval of administration. Interference and enhancement can potentially occur when dengue viruses are given in combination. Four subjects developed neutralizing antibodies in all four serotypes, two after the first dose and two after the third dose at 4 months. Four months out of five volunteers who were re-vaccinated, three or more serotypes were converted. The explanation for this difference is that there is sufficient cross-reactive neutralizing antibody that inhibits replication of the heterotype virus in the vaccine one month after each vaccination. Sabine has found this temporary cross protection lasting up to 3 months when given a human serotype virus (Sabin, 1959, Vira and Rickettsial Infections of Man. Philadelphia: JB Lippioncott Company). Our forthcoming tetravalent study will use the 0.6 month vaccination schedule. [169] The low immunogenicity of DEN-3 and 4 is that 10 5 pfu / ml Den-3 and 4 administrations have a replication disadvantage compared to DEN-1 and 2 (both are 10 6 pfu / ml as tetravalent agents). Can be. Alternative manufacturing strategies are under way to increase the drop in DEN-3 and DEN-4. [170] Without confirming viral infection of all four viruses among the tetravalent responders, it cannot be convinced that the presence of neutralizing antibodies necessarily implies replication of all four serotypes. The neutralizing antibodies measured can be cross reactive and have low binding activity. This problem should be addressed by examining the long-term persistence of the antibody for each serotype. Sensitivity and serotype-specific RT-PCR investigations may be useful for measuring multiple viral infections as evidence of viral replication. [171] Only two of the four vaccinates developed neutralizing antibodies to all four serotypes after the first vaccination. This incomplete response to tetravalent antibodies raises the question of the risk of dengue hemorrhagic fever in the setting of exposure to toxic heterologous serotypes. If antibody dependent proliferation is the pathophysiological mechanism of DHF, there may be a risk even when all four serotype antibodies are induced by the vaccine (but one or more serotype antibodies differentially decrease below the neutralization point). In the following, it is reported that the TH1 T-cell response can be measured among the four vaccinates even in the absence of neutralizing antibodies. Is it enough to protect These questions can only be answered by careful long-term field testing of tetravalent vaccines in the community. [172] In conclusion, our results show that the four serotypes are monovalent vaccines, which are more reactive for type 1 than serotypes 2, 3 and 4. Serotypes 1 and 2 lead to greater than 90% neutralizing antibodies, while serotypes 3 and 4 are less immunogenic. The tetravalent combination is safe, reasonably well tolerated, and induced neutralizing antibodies to all 4 serotypes in 4 of 10 subjects. Two doses of the tetravalent vaccine did not improve seroconversion rates during the one or two month dosing period tested. Longer dosing periods of four months may improve serum conversion. [173] Example 10 [174] Materials and Methods for T-Cell Responses to Dengue Vaccines [175] Subject. 35 healthy adult volunteers (21 males, 14 females) aged 18-50 participated in phase I clinical trials conducted by Walter Reed Army Institute of Research, including the candidate Dengue virus vaccine. Participants were selected from a group of volunteers without circulating anti-flavivirus antibodies. Additional selection criteria were good health based on HIV negative status and response to physical experiments and questionnaires. [176] Vaccine group. 30 individuals received two doses of the individually randomized live-attenuated monovalent vaccine; Four received two doses of the live-attenuated tetravalent vaccine. One monovalent recipient (Applicant ID 1) withdrew only after the first dose. Prior to vaccination, Dengue virus types 1 to 4, Japanese Encephalitis Virus, St. Louis encephalitis virus, or yellow fever virus, was devoid of detectable hemagglutination-inhibiting serum antibodies. Each dose was given by subcutaneous injection of 0.5 ml undiluted virus (s). [177] PBMC harvest. Vacutainer Cell Preparation by intravenous puncture from each volunteer with peripheral blood (8 ml) on day 0 and 5 times after the first dose (3, 7, 9, 14, 28/30/31/60 or 91 days) Tube (CPT) [Becton-Dickinson, Franklin Lakes, NJ]. Blood is also collected on the 2nd day of administration and 4 times thereafter (3, 7, 9 and 14 days after the second dose). The duration of the second dose was approximately 1 to 3 months after the first dose, depending on the volunteer. In about a month's pooling time, the variation occurred due to the variety of applicants' schedules. Cells were separated from whole blood by centrifugation at 1000 × g for 30 minutes. PBMCs were harvested (cell layer on gels in CPT tubes) and washed twice with Hank's balanced salt solution with 500 xg centrifugation (Life Technologies, Rockville, MD). The isolated PBMCs are resuspended in 4 ml (per CPT tube) of cell coagulation medium / DMSO (Sigma, St. Louis, MO) and coagulate overnight at −70 ° C. in an aliquot of 1 ml. The PBMCs are then transported in vapor phase liquid nitrogen for long term storage. [178] Vaccine virus. The following live-attenuated dengue virus species described above are used for monovalent vaccines: 45AZ5PDK20 (DEN 1), S16803PDK50 (DEN 2), CH53489 (DEN 3), 341750PDK20 (DEN 4). The tetravalent vaccine was an equivalent mixture of all four of these species. [179] Cell culture viruses. The following dengue viruses, propagated in Vero cells, were used for PBMC stimulation in culture: Westpac 74 (DEN 1), S16803 (DEN 2), CH53489 (DEN 3), and TVP360 (DEN 4). All four serotypes By Robert Putnak, it was stored at −70 ° C. until use and provided in 1 ml aliquots. The dropping rate of the virus ranges from .30-2.4 x 10 6 pfu / ml. [180] Bulk culture of PBMCs and stimulation with live-virus. Condensed vials of PBMC are removed from the liquid nitrogen reservoir and slowly dissolved at 37 ° C. PBMC was washed twice with RPMI medium 1640 (Life Technologies, Rockville, MD) and supplemented with 10% human male AB serum (Sigma) [enicillin (100 U / ml) -streptomycin (0.1 mg / ml)- It is suspended in complete medium containing fungisone (0.25 mg / ml) [Sigma], 2 mM L-glutamine (Life Technologies), and 0.5 mM 2-mercaptoethanol (Sigma). The cells are suspended at a concentration of 2.5 million cells / ml. Some probes require 3.2 million cells / ml. The PBMC (100 ml) is added to individual wells of 96-well V-bottom plates (Costar, Action, Mass.). An equal volume of Dengue virus 1, 2, 3 or 4 diluted in 10% complete medium at a concentration of 3000 to 24000 pfu / 100ml is added to each well. Control wells received the same volume of medium without virus. The cells are then incubated at 37 ° C. in 5% CO 2 for 4 days. [181] Immunoassay. [182] Chemiluminescent immunoassay was performed to determine the amount of lymphokine secreted from the tissue culture supernatant at the end of 4 days of culture. A 96 well immunoassay plate, Microlite 2 (Dynatech Laboratories, Inc., Chantilly, Virginia) was given an unlabeled anti-lymphokine (IL-4, IL-10 or interferon γ) antibody in 0.1 M potassium bicarbonate buffer overnight. Pharmingin San Diego, Calif.) Was coated with 50 ul / well of 10 mg / ml. The plate was washed and 100 ul I-block buffer (Tropix, Bedford, MA) was added for one hour. The standard group (recombinant IL-4, IL-10 and interferon γ, Pharmingen, San Didego, Calif.) Was previously diluted in I-blocks starting at a concentration of 10 ng / ml. The standard group was diluted 8-3 times. Samples, controls and standard groups were diluted in the same volume of I-block buffer. An aliquot of 50 ul was added to each assay plate. Samples were incubated at room temperature for 1 hour. The plate was washed. Secondary biotinlyated antibody was diluted 1: 1000 with I-block and 50 ul / well was added to the irradiation plate. The plate was washed and 50 ul / well of avidin-alkaline phosphatase (Avidix AP, Tropix, Bedford, Mass.) Was added to the irradiation plate. The plates were incubated for one hour at room temperature. The washed plates were incubated twice for 1 minute in irradiation buffer (Tropix). The CDP-Star substrate (Tropix) was added to each well (100 ul / well). After 10 minutes, the plates are read on an MD2250 luminometer (Dyatech, Chantilly, VA). The first sample is examined using a modified protocol. Avidin-equarin (Sealite Sciences, Atlanta, GA) was used instead of the detector step with avidin-alkaline phosphatase. The material became unavailable during the study and the protocol changed. The results using the standard and control specimens were identical for the two survey formats. [183] Serotype cross-reactivity. To test serotype uniqueness, PBMCs collected at 42, 45, or 105 days at selected receptors of monovalent attenuated vaccine (see results) were harvested at 250,000 cells / well for 4 days with virus of each serotype in independent culture. Stimulated. Culture supernatants were then analyzed using chemiluminescent lymphokine ELISA. [184] T-cell partial deficiency. To test the intrinsic cell source of lymphokine production, PBMCs lacked CD 3+ or CD 8+ T lymphocytes prior to stimulation. Selected PBMCs were washed twice with RPMI medium 1640 and suspended at 3.52 million cells / ml in 5% complete medium (more than 30% PBMC was used as a feed to compensate for cell loss during the deficiency process). For negative deficiency, cells (650,000 PBMCs) were incubated with washed antibodies coated with magnetic beads. Two kinds of beads were used: M-450 anti-CD3 and anti-CD8 beads (Dynal, Oslo, Norway). Anti-CD3 beads were used at a concentration of 5.2 million particles / tube giving approximately 20: 1 beads to the desired cell ratio. Anti-CD8 beads were used at a concentration of 4 million particles per tube, giving approximately 31: 1 beads (DYNABEADS ™ (Dynal)) to the desired cell ratio in 1.5 ml microcentrifuge tubes. Cells were incubated at 4 ° C. for 30 minutes with moderate agitation. Non-deficient PBMCs were used as controls. Labeled cells were removed from the cell mixture using an MPC-2 magnetic particle concentrator (Dynal). CD3 + and CD8 + negatively selected PBMCs were transferred to fresh microcentrifuge tubes. For lack of all residual unbound cells, concentrated Dynabead was washed once with 200 ul complete medium. After transfer, the final volume (400 ul) was equally divided into two wells of 96-well V-bottom culture plates. Deficient and control PBMC culture supernatants were analyzed after 4 days using chemiluminescent lymphokine ELISA. In addition, cultured PBMCs were examined for intracellular Granzyme B mRNA (see below). CD4 + deficiency is similarly done except that separation is done after stimulation with M-450 CD4 + (28.6 ml / 4.004 million particles, approximately 31: 1 beads at the desired cell ratio) dynabead. CD4 + negative selected PBMCs are only investigated for intracellular granzyme B mRNA. [185] Flow cytometry. Randomly selected, unstimulated PBMC populations (both non-deficient controls and CD3 + or CD8 + deficient sets) were stained in duplicate, and then the deficiency efficiency (measured as% deficiency) was measured using FACS analysis. Cells were incubated with PE labeled anti-CD4 + or anti-CD8 + antibody and FITC labeled anti-CD3 + antibody (Becton-Dickinson) at 4 ° C. for 30 minutes. The labeled PBMCs were then washed three times with fluorescent buffer [PBS (Sigma), 0.05% Na Azide, 1% Fetal Bovine Serum (Summit Biotechnology, Boulder, CO)] and fluorescent fixative [PBS, 1% formalin before analysis]. , 0.05% Na azide]. Deficiency efficiency with CD4 + Dynabeads was not measured. [186] Granzyme B Analysis. Non-deficient control PBMCs and PBMCs deficient in T cell subsets were stimulated with wild-type virus for 4 days and then analyzed for intracellular granzyme B mRNA. Reverse transcriptase chain polymerization (RT-PCR) assay format on a 96-well plate was used. [187] MRNA purification was performed using the "Straight A's" mRNA Isolation System (Novagen, Madison, Wis.). After centrifugation and PBMC culture supernatant were removed for lymphokine ELISA analysis, the pelleted PBMCs were lysed with 200 ul / well of lysis buffer containing 10 mM dithiothritol, and washed oligo dT magnetic beads. Incubate with 200 mg / well for 30 minutes at room temperature. To remove DNA, protein and cell debris, the beads were thoroughly washed with an 8-fold volume of wash buffer using MPC-96 (Dynal) magnetic particle concentrator, and then mRNA was washed with H 2 O 200 ul / well for 20 minutes. Elute at 70 ° C. Transfer the eluent to a 1.5 ml microcentrifuge tube and perform a second elution with additional H 2 O 200 ml / well. Then, 400 ul of eluate was precipitated using 50 ul of 3M sodium acetate (pH 5.2), 20 mg of glycogen (Novagen), and 300 ul of isopropanol. After a final wash with 70% cold ethanol, mRNA pellets were suspended in 30 ul of H 2 O. [188] RT-PCR steps were performed in 96-well plates. Oligonucleotide primers (22 bp), corresponding to exons of human granzyme B (CTLA-1) and amplifying the 120 bp region, were synthesized by Dr Stuart Cohen of Walter Read Army Institute of Research. This primer has the following sequence: grb2a (sense) 5'AGC CGA CCC AGC AGT TTA TCC C (SEQ ID NO: 1), grb2b (antisense) 5'C TCT GGT CCG CTT GGC CTT TCT (SEQ ID NO: 2). [189] For each reverse transcriptase reaction, the total reactant volume was 40 ul and contained: MgCl 2 (5 mM), 10X Buffer II (10 mM Tris-HCL, 50 mM KCL, pH 8.3), dNTPs (1 mM each), And RNase inhibitor (40 units) [Perkin Elmer, Norwalk, CT] AMV reverse transcriptase (10 units) [Siekagaku], grb2b primer (3 pmoles), sH 2 O and 4 ml of mRNA template. The RT reaction step was performed on a 9600 thermocycler (Perkin Elmer) with parameters set at 42 ° C. (90 minutes), 99 ° C. (5 minutes), 4 ° C. (indefinite). For each PCR, the total reaction volume was 50 ul and contained: MgCl 2 (2 mM), 10X Buffer II (same as above), dNTPs (.4 mM each), amplitaq gold (1.25 units), grb2a And grb2b primers (1 pmoles each), sH 2 O and cDNA template 5 ul. In addition, the PCR incubation step includes 95 ° C. initial denaturation / enzyme activation (10 min), 30 cycles: [95 ° C. denaturation (30 sec with 10 sec lamp) / 60 ° C. annealing (30 sec with 30 sec lamp) / 72 ° C. Extension (30 sec with 30 sec ramp), 72 ° C. final extension (7 min), 4 ° C. (indefinite) with a 9600 thermocycler (Perkin Elmer). [190] Using electrophoresis, the final amplified PCR product (10 ul) was separated on 2% agarose (SeaKem) / 1X TAE (tris-acetate-EDTA) gel stained with ethidium bromide, and digital camera (Scientific Imagine Systems). , New Haven, CT). [191] If a booster response to booster administration of live vaccines could occur, it was estimated that more attenuated live vaccines could be used. Booster responses to antibody and T cell responses were found. [192] T cell responses to dengue vaccines were measured, but less T cell responses were measured compared to antibody responses. Thus, the T cell response to administration of live dengue vaccines was not well characterized. One of the aims of this study is to determine the nature of the T cell response, namely T helper response, serotype specificity and cytotoxic potential. [193] The predominant T cell response to these vaccines was the Th1 response. This was determined by the secretion of interferon γ by peripheral blood monocytes (PBMC) stimulated by live dengue virus during 4 days of culture. Interferon γ was secreted from CD3 + CD8 − T cells. T cell responses were slightly cross-reactive and specific for dengue virus serotypes. Reactions occurred in some of the subjects and not in others. [194] Lymphokine secretion by dengue stimulating cells. [195] Live dengue virus was used to stimulate PBMC cultures. The serotype of the stimulating virus used in the culture was the same as the serotype of the vaccine virus. After 4 days, tissue culture supernatants were analyzed for the presence of interferon γ, IL-4 and IL-10. In all cultures, IL-4 and IL-10 were consistently negative. Two analytical controls were used to confirm that the analysis was successful. First, recombinant lymphokines were used in the standard curve, and second, control samples were used to confirm that lymphokines could be measured even in the presence of tissue culture supernatants. [196] Despite the negative expression of IL-4 and IL-10, high levels of interferon γ were measured in some culture supernatants. 6 shows the interferon γ expression kinetics of cells harvested from volunteers receiving monovalent vaccines. Overall, the highest interferon γ responses were seen in PBMCs collected from recipients of Dengue 1 and Dengue 2 candidate vaccines, but some high responses were also seen in Dengue 3 and 4 recipients. Occasionally, interferon γ was sometimes detected up to 14 days of the first inoculation, but expression was often detected immediately before or immediately after the second dose. Thus, the kinetics of secretion was much slower than expected. There was no consistent pattern of booster response in the monovalent recipients of this study. Depending on the individual, after the second dose, interferon γ levels increased or decreased. [197] Interferon γ levels were unmeasurable in unstimulated PBMCs of all volunteers at all pools. Mean expression at day 0 of stimulation was 127 pg / ml with a standard deviation of 230 pg / ml. [198] In monovalent vaccine recipients, 16 positive and 14 negative interferon γ responders appeared (mean ± 3 standard deviations). Sixteen of 30 monovalent vaccine recipients had interferon g results of PBMC culture> 1000 pg / ml at one or more time points. Twelve patients sustained> 1000 pg / ml of interferon g secretion above two time points. In addition, 12 of 30 had a secretion> 1000 pg / ml at the time of final analysis. [199] Four volunteers received the tetravalent vaccine (same mixture of all four monovalent strains). 7 shows the interferon γ production following PBMCs where these 4 were collected in the recipients. PBMCs were stimulated in isolated cultures using one of each of the four serotypes of dengue virus. PBMCs of volunteers # 33 and # 36 secreted significant amounts of interferon γ,> 1000 pg / ml, at one or more time points after stimulation with each of the four serotypes. PBMC of volunteer # 35 secreted a significant amount of interferon γ for three of the four serotypes (except Dengue 3). PBMC of volunteer # 34 secreted significant amounts of interferon γ only for dengue 2 virus. The highest reaction occurred predominantly for DEN 1 and 2. As in the case of monovalent volunteers, the interferon γ production kinetics were delayed in the tetravalent vaccine volunteers. High levels of interferon γ were detected immediately before and after the second inoculation. As with the monovalent recipient, the booster response was inconsistent with the pattern of ylteron γ secretion after the second dose. [200] In aggregates, the above results indicate that the predominant T lymphocyte response in both monovalent and tetravalent vaccine recipients is an antigen specific Th1 response. [201] Example 11 [202] Serotype cross-reactivity [203] PBMCs from 12 monovalent vaccine recipients are tested for dengue serotype-specific response and cross reactivity. Based on kinetics, individuals are selected that secrete at least 1000 pg / ml of interferon γ in PBMC culture supernatant at the final time point (day 2 of the last harvest). From the date of final harvest, PBMCs are stimulated with each dengue serotype for 4 days in independent culture and analyzed for secreted interferon γ in the culture supernatant. Although there were some serotype cross-reactivity, there was always the highest response in PBMCs stimulated with the same serotype virus as the original vaccination (Table 14). Therefore, the interferon γ response seen in PBMCs from selected monovalent vaccine recipients is dengue serotype-specific. [204] Crossreactivity is less than half of serotype-specific responses. The dengue 2 vaccine recipients were most cross-reactive with dengue 4 virus. The dengue 4 vaccine recipients were most cross-reactive with dengue 2 virus. For dengue 1 vaccine recipients, cross-reactivity varied. There was only one dengue 3 vaccine recipient in this group and the response was serotype specific. [205] [206] Example 12 [207] T cell subset deficiency [208] Identification of cells secreting interferon γ to demonstrate that this is a Th1 response. This is done by lacking T cells or T cell subsets prior to culture. The cells used in this study are mixed with PBMC isolated from whole blood using density gradient centrifugation. The main cells of the PBMC population are T cells, B cells, monocytes and NK cells. For this evaluation, the time points with the highest interferon γ were selected based on the kinetics in 13 monovalent and three tetravalent volunteers. [209] Immune magnetic cell isolation is used to remove cells from PBMCs. Deficiency efficacy is assessed using flow cytometry in test deficiency. Analysis of cultured PBMCs is not performed because only a few are accessible. In test deprivation, removal of CD3 + cells using CD3 monovalent antibodies results in a 92% reduction in CD3 + cells compared to non-deficient PBMC controls. CD3 deficiency is monitored using a double label for CD3 and CD4, a double label for CD3 and CD8 and a single label for CD3. CD3 deficiency is more complete through CD4 + cells than CD8 + cells, with 98% of CD3 / CD4 T cells deficient and 90% of CD3 / CD8 T cells deficient in the CD3 deficient group. Removal of CD8 + cells using CD8 monovalent antibodies results in a 99.9% decrease in CD8 + cells. [210] Selected PBMCs lack CD3 + or CD8 + T lymphocytes and are stimulated in culture with dengue virus for 4 days and then tested for secreted interferon γ. The results are compared to those obtained from non-deficient PBMC controls cultured simultaneously. CD4 + T lymphocytes are not deficient before stimulation because other cell populations require CD4 + T helper cells for the production of interferon γ. [211] Removing CD3 + cells prior to culture substantially reduces the production of interferon γ as shown in Table 15. The range of reduction of interferon γ after CD3 + deficiency is 59-100%. Although reduced in CD3 + deficient cultures, a significant amount of interferon γ is produced. This residual production indicates that a small amount of residual CD3 + cells remaining after immunomagnetic cell isolation secretes interferon γ and / or another cell population also secretes interferon γ. [212] [213] With the exception of only one individual, elimination of pre-culture CD8 + cells does not reduce the production of interferon γ. In 9 of 16 cultures, the removal of CD8 + cells actually increases their production either by the removal of inhibition by these cells or by the reduction of the death of infected antigen presenting cells by CD8 + cytotoxic lymphocytes. [214] Together, the results indicate that interferon γ seen in PBMC cultures secrete cells affected by CD4 + T lymphocytes and / or CD4 + T lymphocytes. This fact is also supported by elucidating the Th1 response. [215] Example 13 [216] Granzyme B. Th1 responses are associated with cytotoxic lymphocyte responses, among others. In an effort to determine whether cells capable of cell mediated killing exist in these vaccine volunteers, granzyme B mRNA was measured in PBMCs cultured for deficiency experiments. After removal of the culture supernatant for lymphokine analysis, cells were pelleted and lysed for extraction of mRNA. Granzyme B specific primers were used for RT-PCR. PCR products were analyzed by agarose gel electrophoresis. Gel band intensities were converted to +, − scale using a comparative reference photograph (FIG. 8). Extra cells from 7 of the volunteers were incubated without viruses. From the seven volunteers, unstimulated PBMC showed little (-or +) granzyme B mRNA expression. With antigen specific stimulation, expression was substantially upregulated in all 16 of the selected vaccine recipients (FIG. 8). T cell subset deficiency using CD8 monoclonal antibodies did not significantly reduce granzyme B expression compared to control PBMCs. There were 3 individuals (ID 16, 22 and 33) in which granzyme B expression was reduced in the CD8 deficient group. In one (ID 33), the reduction was substantial. In contrast, T cell subset deficiency using CD3 monoclonal antibodies reduced expression in 14 of volunteers. The decline was substantial in eight of the single volunteers and in all three of the four volunteers. Four of the interferon γ free responders were also tested for granzyme B mRNA. All showed low levels of expression (data not shown). [217] In cells from 7 of the volunteers, T cell subset deficiency using CD4 monoclonal antibodies was performed 4 days after culture. In order to provide T helper activity to all cells in need of help during the cultivation, deficiency was performed after culturing. Removal of CD4 + cells after stimulation did not affect granzyme B expression compared to the nondeficient control of the seven volunteers analyzed. Thus, although there is an antigen dependent production of granzyme B mediated by CD4 + Th1 cells, the actual cells producing granzyme B have been shown to be cells other than T cells. It is not known whether it is produced by NK cells or macrophages. [218] discussion [219] The two objectives of this study were to determine if there was a measurable T cell response in the vaccine recipient and if a cell mediated response to the second dose of the vaccine was observed. For these purposes, T cell response kinetics were measured by restimulation of cells collected at two dose intervals. Restimulation was performed with live virus in bulk cultures of PBMCs collected during the study. [220] The third objective of this study was to: 1. T cell in view of measurement of cell types limited by the lymphokine list, 2. dengue serotype specificity and cross-reactivity, and 3. cytotoxic potential, granzyme B production It was to measure the nature of the reaction. This response was measured in PBMCs from both monovalent and tetravalent vaccine recipients. For tetravalent vaccine recipients, it was important to determine if the response could be measured for all four serotypes of dengue virus. [221] Human and mouse T helper responses can be classified into two groups based on their pattern of lymphokine expression 5. T helper 1 (Th1) cells are characterized by the secretion of IL-2 and interferon γ. Of the two lymphokines, interferon γ is the most important for identifying Th1 cells. T helper 2 (Th2) cells are characterized by the secretion of IL-4, IL-5, IL-6 and IL-10. Of the mixed population of cells or PBMC bulk culture, one of the two secretion patterns is generally dominant. [222] One factor influencing the Th1 to Th2 response is the nature of aggressive interferon. Viral infections, and some bacterial infections such as Listeria and Mycobacterium (Peters, 1996, Hepatology 23, 909-916) often induce Th1 responses, while some parasitic infections will induce Th2 responses (Conrad et al., 1990, J Exp Med 171, 1497-1508). The rate of the two reactions can change during the course of the infection. For example, although viral infections generally begin with Th1 responses, Th2 responses can be generated after infection. Early Th1 responses can augment CTL responses and direct immunoglobulin isotype switching, while subsequent Th2 responses can augment antibody production by B cells. [223] During natural dengue infection, one study showed a Th1 response in most individuals. The Th1 response was associated with an effective immune response without associated severe pathogenicity. In contrast, some individuals developed a Th2 response associated with more pathogenicity. [224] Despite its relevance to the Th1 response with an effective anti-dengue immune response, interferon γ, the key lymphokine of the Th1 response, has both positive and negative effects on the immune response. In Thailand, Kurane found high levels of interferon γ in the serum of DHF patients compared to low levels in the serum of DF patients (Kurane et al., 1991, J Clin Invest 88, 1473-1480). Increased interferon γ may be a measure of immune activity. Interferon γ is required to activate and maintain activity of cytotoxic cells (CD4 + T cells, CD8 + cells and NK cells). While this mechanism may contribute to pathogenicity in severe infections, the same response may be beneficial in weaker infections by reducing the number of virus infected cells through antigen specific cytolysis. The positive role of interferon γ in controlling dengue virus infection is demonstrated in recent mouse knockout models that lack interferon α, β and γ. In contrast to normal adult controls that are resistant to infection, knockout mice are susceptible to lethal infection by dengue virus (Johnson and Roehrig, 1999, J Virol 73, 783-786). [225] Alternatively, interferon γ may contribute to the pathogenicity of dengue virus infection. One mechanism for pathogenicity may be by immune boosting by increasing infection of macrophage, one major target cell. In culture, interferon γ increased antibody mediated infection of macrophage line U937 by increasing the number of Fc receptors on the surface of cells (Kontny et al., 1988, J Virol 62, 3928-3933). However, other studies using normal cultured macrophages have had the opposite effect of reducing infection (Sittisombut et al., 1995, J Med Virol 45, 43-49). Through these conflicting results, it is not clear whether interferon γ contributes to an increase in macrophage infection. [226] In this study, the Th1 response was dominant. Assays for IL-4 and IL-10 were consistently negative, indicating a lack of TH2 response. High levels of interferon γ were detected in the supernatants of many cultures, indicating the presence of a Th1 response in the culture. [227] Since the stimulated cells were total PBMCs, it was necessary to measure the cells that caused the secretion of interferon γ. This is accomplished by deficiency of T cell subsets using immunomagnetic procedures. Negative deficiency was performed before incubation with an antibody that recognizes either CD3 or CD8. Since CD3 deficiency resulted in interruption of interferon γ secretion and CD8 deficiency was not, it was concluded that CD3 + CD8- lymphocytes secrete interferon γ or at least are a cell population that controls the secretion of interferon γ. This demonstrated that interferon γ is the result of the Th1 response. Residual interferon γ in some cultures after deficiency may be due to some remaining CD4 + T lymphocytes or other cells, possibly NK cells or macrophages, after deficiency in culture. [228] Peak interferon γ response was serotype specific. When cells from monovalent vaccine recipients were separately stimulated by each of the four serotypes of dengue virus, peak interferon γ production was in response to stimulation by the vaccine virus and homogeneous dengue virus. Less cross-reactive responses to other dengue viruses have been noted in various cultures. This is similar to lymphocyte proliferation, results obtained by others using different measurements. In one study, cells from individuals receiving the Dengue 2 vaccine showed the greatest response to the Dengue 2 virus, but a cross reactive response was noted (Dharakul, J Infect Dis 170, 27-33). Although many clones from dengue 3 vaccine recipients responded best to dengue 3 antigens, it was demonstrated at the clone level that they had cross-reactive responses to the other three dengue antigens (Kurane et al., 1989, J Exp Med 170, 763-775). The conclusion of the latter study was that primary dengue virus infection mainly produces cross-reactive CD4 + lymphocyte responses (proliferation and interferon γ production). [229] In this study, the cross-reactivity of PBMCs of monovalent vaccine recipients was generally less than half of serotype specific responses. In tetravalent vaccine recipients, interferon γ secretion in response to each serotype of dengue virus was significant in three of four tetravalent vaccine recipients. The response varied sufficiently within the individual vaccine recipients, making it impossible to determine whether the lower response was serotype specific or cross reactive. [230] The kinetics of T cell activity exhibited by interferon γ secretion was slower than expected. In some instances, the response could be detected up to 14 days. In most cases, however, no response was detected until immediately before administration of the second vaccine dose. It is not clear what is the reason for the delayed dynamics. One explanation is that antigen production by vaccine virus infected cells can be slow and persistent. Equally, however, the method may optionally detect a memory response rather than an acute response. For example, if active CD8 + cells inhibit the CD4 + response in PBMCs collected during early infection, the measurable response can be attenuated. In cultures deficient in CD8 + lymphocytes, interferon γ secretion by remaining lymphocytes increased in more than half of the culture. This inhibition can be greater during the initial infection. [231] Others observed more acute lymphokine production kinetics. For 17 days after inoculation of the attenuated dengue vaccine, serum lymphokines including serum interferon γ were measured. In this study, acute reaction peaked in viremia was noted (Kurane et al., 1995, J Clin Lab Immunol 46, 35-40). [232] The response to the second dose was mixed. Some individuals showed an increase in interferon γ production, while others showed a decrease. Interferon γ production by cells collected from vaccine recipients immediately prior to the second dose is sufficiently large to mask any memory response to the second dose. In addition, late interferon γ responses can make the measurement of memory T cell responses more difficult. It is clear that some individuals responded to the second dose. This may indicate that there is some localized viral growth in the presence of an active immune response. [233] In summary, the predominant T cell response to administration of the live attenuated dengue virus was the Th1 response. This was evidenced by the secretion of interferon γ by restimulated PBMCs collected from vaccine recipients. None of the PBMC cultures from the cells of the vaccine recipients showed significant IL-4 or IL-10 secretion into the culture supernatant after restimulation. The Th1 response was demonstrated by indicating that CD3 + CD8 − lymphocytes secrete interferon γ. The Th1 response was mainly dengue serotype specificity, but less cross-reactive response was noted. [234] Example 14 [235] Clinical and immunological evaluation of four dengue viruses as challenge strains in immune and susceptible volunteers [236] The main purpose of this study is to characterize the clinical response to each of the four candidate dengue challenge viruses in susceptible and immune volunteers to assess their suitability as challenge strains for human vaccine efficacy studies. The second purpose of the study is to hypothesize the immune correlation of protection against dengue fever. [237] Dose, Schedule and Route: All volunteers will receive one of four dengue challenge viruses or placebos in a single dose of 0.5 ml subcutaneously in the deltoid zone on day 0 of study day. [238] Study group: [239] Volunteer Set # 1 (Sensitive): One of DEN-1, DEN-2, DEN-3, DEN-4 or placebo. [240] Volunteer Set # 2 (Immune): Administered either the DEN virus (serotype corresponding to a previously administered vaccine) or placebo. [241] General Eligibility Criteria: Age 18-35 years, good health without any chronic medical condition, score greater than 75% for study comprehension written test, informed consent, likelihood of study duration, approval for participation from the command line Letter (for soldiers only), serological transition to previous dengue vaccinations (only in volunteer set # 2). [242] Statistics: Data analysis will mainly be described in this pilot trial with a small number of volunteers in each test sample group. The main concern will be to document the frequency of (prespecified and unexpected) clinical events within four study groups compared to placebo. [243] A hypothesis was developed for the immune correlation of protection, comparing pre-challenge immune measurements and post-challenge immune responses of all challenged volunteers with dengue fever with those of all challenged volunteers that were well maintained. [244] Application of the human dengue challenge model: In contrast to most histological human dengue challenge experiments designed to assess attenuation of live vaccine candidates or to characterize dengue disease, these challenge studies are: 1) flavivirus-non-immune (flavivirus) -naive) to validate the correlation of immunity in recipients of a single dengue vaccine when validating four dengue viruses as challenge strains of volunteers (Sponsor Set # 1), and subsequent challenge with homologous dengue virus. (Volunteer set # 2) will be. [245] If the clinical response in volunteer set # 1 suggests that the strain is appropriate for the challenge in subsequent control experiments, the challenge strain may be selected as a dengue vaccine candidate or placebo to select the most promising vaccine candidate for further development. Will be administered to the recipient. [246] If the immunological response in volunteer set # 2 suggests that some aspect of pre-challenge immunity (antibody and / or T cell memory) correlates with protection, this correlation of protection may simplify dengue vaccine development. [247] Category Restriction of Dengue Viruses Tested as Challenge Strains: Appropriate dengue challenge viruses 1) regenerate non-complex dengue fever lasting 3-7 days in volunteer set # 1, and 2) are prepared according to appropriate manufacturing criteria (GMP) 3) free of adjuvant or reactive non-viral components and 3) lyophilized virus in sufficient amounts (> 100 doses). Challenge viruses include inactivated DEN-1 45AZ5 (PDK-0), DEN-2 S16803 (PDK-10), DEN-3 cl 24/28 (PDK-0), DEN-4 341750 (PDK-6) ( PDK = primary dog kidney cells. [248] The dose of each challenge virus in the plaque forming unit (pfu.) Will be 0.5 × titer. [249] Study challenge viruses fall into the latter two categories. This study is to demonstrate that the study candidate challenge strains fall into the first category. We have some evidence that the four challenge viruses tested in this study are moderately pathogenic. DEN-1 and DEN-3 challenge viruses have already been shown to cause noncomplex recessive disease among volunteers. Although the DEN-2 and DEN-4 challenge viruses to be administered in this study have not been tested in volunteers, they are considered pathogenic as they are the precursors of dengue virus vaccine candidates rejected because they developed recessive disease among volunteers. The only reason for rejecting any of the four study candidate dengue challenge viruses is that they do not cause any disease in flavivirus-uninfected volunteers (volunteer set # 1) or are excessive in any volunteers (volunteer set # 1 or # 2). It causes disease. [250] Volunteer Set # 1: Ten healthy flavivirus-uninfected volunteers will be randomized to receive a dengue virus challenge with one of four serotypes (two volunteers per serotype) or placebo. Volunteers and researchers will be blinded to vaccination. Applicants anticipate that eight volunteers who received the Dengue Challenge Virus will be moderately ill with 3-7 days of fever, severe headache and myalgia. A sufficient recovery can take as long as 14 days after the onset of the disease. Each challenge virus will be considered appropriate based on the clinical response of two recipients, which must satisfy the study definition of dengue fever. Dengue fever is defined as: A disease with two or more of the following: headache, myalgia, erythematous rash, post-orbital pain, arthralgia, and persistent for more than 48 hours, allowing for decreased body temperature due to acetaminophen use Fever, and periods of fever following and subsequent tissue reactions presented by neutropenia or thrombocytopenia or liver damage, and evidence of dengue virusemia during the period of fever. [251] Volunteer Set # 2: Young and healthy volunteers under the age of 12 will receive homologous dengue challenge virus (N = 10, independent of serotype) or placebo (N = 2). Immune volunteers are previous recipients of a monovalent live attenuated dengue vaccine with a primary neutralizing antibody response. Immune volunteers are expected to remain well. Correlation of protection can be confirmed by measurements between pre-challenge immune status or immune activation challenge.
权利要求:
Claims (16) [1" claim-type="Currently amended] An immunogenic composition containing more than one attenuated dengue virus selected from the group consisting of dengue-1, dengue-2, dengue-3, dengue-4, among physiologically acceptable excipients. [2" claim-type="Currently amended] 2. The immunogenic composition of claim 1, further comprising an adjuvant that enhances the immune response. [3" claim-type="Currently amended] The immunogenic composition of claim 1 formulated with 10 2 to 10 6 PFU doses of attenuated virus. [4" claim-type="Currently amended] Administering to the subject an immunologically sufficient amount of more than one attenuated virus selected from the group consisting of dengue-1, dengue-2, dengue-3 and dengue-4 in a physiologically acceptable carrier. How to stimulate dengue virus specific immune response. [5" claim-type="Currently amended] The method of claim 4, wherein the attenuated virus is administered parenterally. [6" claim-type="Currently amended] The method of claim 4, wherein the attenuated virus is administered intranasally. [7" claim-type="Currently amended] A multivalent live attenuated dengue virus vaccine containing any combination of dengue virus serotypes selected from the group consisting of dengue 1, dengue 2, dengue 3 and dengue 4. [8" claim-type="Currently amended] The vaccine of claim 7, wherein said dengue 1 is 45AZ5 PDK20 (ATCC No. VR-2648). [9" claim-type="Currently amended] The vaccine of claim 7, wherein said dengue 2 is S16803 PDK50 (ATCC No. VR-2653). [10" claim-type="Currently amended] 8. The vaccine of claim 7 wherein said dengue 3 is CH5389 PDK20 (ATCC No. VR-2647). [11" claim-type="Currently amended] The vaccine of claim 7, wherein said dengue 4 is 341750 PDK20 (ATCC No. VR-2652). [12" claim-type="Currently amended] The method according to claim 7, wherein Dengue 1 is 45AZ5 PDK20 (ATCC No. VR-2648), Dengue 2 is S16803 PDK50 (ATCC No. VR-2653), Dengue 3 is CH5389 PDK20 (ATCC No. VR-2647) And dengue 4 is 341750 PDK20 (ATCC No. VR-2652). [13" claim-type="Currently amended] 8. The dengue virus vaccine of claim 7, wherein said dengue virus is produced in vertebrate cells. [14" claim-type="Currently amended] The dengue virus vaccine of claim 13, wherein said cells are Vero cells. [15" claim-type="Currently amended] 8. The method according to claim 7, wherein the dengue-1 virus is in an amount of 10 2 to 10 7 pfu / ml, the dengue-2 virus is in an amount of 10 2 to 10 7 pfu, and the dengue-3 virus is 10 2 to 10 A dengue virus vaccine in an amount of 7 pfu and wherein the dengue-4 virus is in an amount of 10 2 to 10 7 pfu / ml. [16" claim-type="Currently amended] The dengue virus vaccine of claim 15, wherein said vaccine is administered subcutaneously.
类似技术:
公开号 | 公开日 | 专利标题 US20190192649A1|2019-06-27|Flavivirus vaccines Powers2018|Vaccine and therapeutic options to control chikungunya virus Guy et al.2010|Preclinical and clinical development of YFV 17D-based chimeric vaccines against dengue, West Nile and Japanese encephalitis viruses USRE45123E1|2014-09-09|Recombinant attenuated dengue viruses comprising a deletion in the 3′ untranslated region and additional attenuating mutations induced by chemical mutagenesis US20180010099A1|2018-01-11|Dengue tetravalent vaccine containing a common 30 nucleotide deletion in the 3'-utr of dengue types 1, 2, 3, and 4, or antigenic chimeric dengue viruses 1, 2, 3, and 4 Scherret et al.2001|The relationships between West Nile and Kunjin viruses. Durbin et al.2011|Development and clinical evaluation of multiple investigational monovalent DENV vaccines to identify components for inclusion in a live attenuated tetravalent DENV vaccine Bhamarapravati et al.1987|Immunization with a live attenuated dengue-2-virus candidate vaccine |: clinical, immunological and biological responses in adult volunteers AU2009212794B2|2012-02-09|Avirulent, immunogenic flavivirus chimeras AU740961B2|2001-11-15|Chimeric flavivirus vaccines Hollidge et al.2010|Arboviral encephalitides: transmission, emergence, and pathogenesis Kurane2007|Dengue hemorrhagic fever with special emphasis on immunopathogenesis Monath2005|Yellow fever vaccine US8029802B2|2011-10-04|Vaccines against Japanese encephalitis virus and West Nile virus Ikegami et al.2009|Rift valley fever vaccines Pletnev et al.2006|Chimeric West Nile/dengue virus vaccine candidate: preclinical evaluation in mice, geese and monkeys for safety and immunogenicity Guirakhoo et al.2001|Construction, safety, and immunogenicity in nonhuman primates of a chimeric yellow fever-dengue virus tetravalent vaccine Durbin et al.2006|rDEN2/4Δ30 |, a live attenuated chimeric dengue serotype 2 vaccine, is safe and highly immunogenic in healthy dengue-naïve adults AU2003290985B2|2010-04-01|West nile virus vaccine Guirakhoo et al.1999|Immunogenicity, genetic stability, and protective efficacy of a recombinant, chimeric yellow fever-Japanese encephalitis virus | as a live, attenuated vaccine candidate against Japanese encephalitis US20190119654A1|2019-04-25|Development of dengue virus vaccine components Durbin et al.2005|rDEN4Δ30, a live attenuated dengue virus type 4 vaccine candidate, is safe, immunogenic, and highly infectious in healthy adult volunteers Pugachev et al.2003|Traditional and novel approaches to flavivirus vaccines US6962708B1|2005-11-08|Chimeric flavivirus vaccines JP3681358B2|2005-08-10|Chimeric and / or growth-restricted flavivirus
同族专利:
公开号 | 公开日 ES2322327T3|2009-06-19| EP1165127B1|2008-12-31| CN1351502A|2002-05-29| AT419006T|2009-01-15| WO2000057907A2|2000-10-05| WO2000057907A9|2002-04-11| CA2368674A1|2000-10-05| AU779280B2|2005-01-13| JP2002540168A|2002-11-26| US7217418B2|2007-05-15| KR100721745B1|2007-05-25| US6638514B1|2003-10-28| MXPA01009683A|2003-06-24| US20070087015A1|2007-04-19| DE60041250D1|2009-02-12| EP1165127A2|2002-01-02| WO2000057907A3|2001-04-12| AU4038200A|2000-10-16| CN1191092C|2005-03-02|
引用文献:
公开号 | 申请日 | 公开日 | 申请人 | 专利标题
法律状态:
1999-03-26|Priority to US12631399P 1999-03-26|Priority to US60/126,313 2000-02-11|Priority to US18172400P 2000-02-11|Priority to US60/181,724 2000-03-24|Application filed by 왈터 리드 아미 인스티튜트 오브 리써치 2002-01-29|Publication of KR20020008136A 2007-05-25|Application granted 2007-05-25|Publication of KR100721745B1
优先权:
[返回顶部]
申请号 | 申请日 | 专利标题 US12631399P| true| 1999-03-26|1999-03-26| US60/126,313|1999-03-26| US18172400P| true| 2000-02-11|2000-02-11| US60/181,724|2000-02-11| 相关专利
Sulfonates, polymers, resist compositions and patterning process
Washing machine
Washing machine
Device for fixture finishing and tension adjusting of membrane
Structure for Equipping Band in a Plane Cathode Ray Tube
Process for preparation of 7 alpha-carboxyl 9, 11-epoxy steroids and intermediates useful therein an
国家/地区
|