Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 16 de 16
1.
Front Immunol ; 14: 1178741, 2023.
Article En | MEDLINE | ID: mdl-37287960

Background: Ocular infections with Chlamydia trachomatis serovars A-C cause the neglected tropical disease trachoma. As infection does not confer complete immunity, repeated infections are common, leading to long-term sequelae such as scarring and blindness. Here, we apply a systems serology approach to investigate whether systemic antibody features are associated with susceptibility to infection. Methods: Sera from children in five trachoma endemic villages in the Gambia were assayed for 23 antibody features: IgG responses towards two C. trachomatis antigens and three serovars [elementary bodies and major outer membrane protein (MOMP), serovars A-C], IgG responses towards five MOMP peptides (serovars A-C), neutralization, and antibody-dependent phagocytosis. Participants were considered resistant if they subsequently developed infection only when over 70% of other children in the same compound were infected. Results: The antibody features assayed were not associated with resistance to infection (false discovery rate < 0.05). Anti-MOMP SvA IgG and neutralization titer were higher in susceptible individuals (p < 0.05 before multiple testing adjustment). Classification using partial least squares performed only slightly better than chance in distinguishing between susceptible and resistant participants based on systemic antibody profile (specificity 71%, sensitivity 36%). Conclusions: Systemic infection-induced IgG and functional antibody responses do not appear to be protective against subsequent infection. Ocular responses, IgA, avidity, or cell-mediated responses may play a greater role in protective immunity than systemic IgG.


Trachoma , Child , Humans , Trachoma/diagnosis , Trachoma/epidemiology , Chlamydia trachomatis , Antibody Formation , Eye/metabolism , Immunoglobulin G
3.
Front Immunol ; 12: 790463, 2021.
Article En | MEDLINE | ID: mdl-34925371

The requirement for vaccine-induced tissue-resident immunity for protection against one or repeated infections with Chlamydia trachomatis (C.t.) is still not fully resolved. In this study, our aim was to investigate to which degree tissue-resident Th1/Th17 T cells in the genital tract (GT) could add to the protection mediated by circulating immunity. Out of several mucosal vaccine strategies, a strategy termed SIM (for simultaneous intrauterine and parenteral immunization with CAF01 adjuvanted CTH522), was superior in generating genital tract tissue-resident Th1/Th17 T cell immunity. This led to a faster and stronger local CD4 T cell response post infection, consisting of multifunctional IFNγ/TNFα-producing Th1 T cells and IFNγ/TNFα/IL-17-producing Th17 T cells, and a faster recruitment of innate immune cells. Post infection, SIM animals showed an additional significant reduction in bacterial levels compared to mice having received only a parenteral vaccine. Nevertheless, the parenteral strategy reduced bacterial levels by 75%, and interestingly, post infection, these mice generated their own vaccine-derived genital tract tissue-resident memory Th1/Th17 T cells, which upon a subsequent infection showed as fast an activation in the genital tract, as observed in SIM mice. Furthermore, in contrast to after the first infection, both groups of mice now showed a similar infection-induced boost in local vaginal IgA and IgG titers. Thus, vaccine-induced resident immunity, generated pre-infection, led to an advantage in the response against the first infection, but not the second infection, suggesting that a parenteral vaccine strategy is a suitable vaccine strategy against infections with Chlamydia trachomatis.


Bacterial Vaccines/administration & dosage , Chlamydia Infections/prevention & control , Chlamydia trachomatis/immunology , Immunity, Mucosal , Administration, Intravaginal , Animals , Antibodies, Bacterial , Antigens, Bacterial/administration & dosage , Antigens, Bacterial/immunology , Bacterial Vaccines/immunology , Chlamydia Infections/diagnosis , Chlamydia Infections/immunology , Chlamydia Infections/microbiology , Chlamydia trachomatis/isolation & purification , Disease Models, Animal , Female , Humans , Immunogenicity, Vaccine , Injections, Subcutaneous , Mice , Th1 Cells/immunology , Th17 Cells/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , Vagina/immunology , Vagina/microbiology
4.
NPJ Vaccines ; 6(1): 58, 2021 Apr 19.
Article En | MEDLINE | ID: mdl-33875654

Ocular and urogenital infections with Chlamydia trachomatis (C.t.) are caused by a range of different serovars. The first C.t. vaccine in clinical development (CTH522/CAF®01) induced neutralizing antibodies directed to the variable domain 4 (VD4) region of major outer membrane protein (MOMP), covering predominantly B and intermediate groups of serovars. The VD1 region of MOMP contains neutralizing B-cell epitopes targeting serovars of the C and C-related complex. Using an immuno-repeat strategy, we extended the VD1 region of SvA and SvJ to include surrounding conserved segments, extVD1A and extVD1J, and repeated this region four times. The extVD1A*4 was most immunogenic with broad cross-surface and neutralizing reactivity against representative members of the C and C-related complex serovars. Importantly, in vitro results for extVD1A*4 translated into in vivo biological effects, demonstrated by in vivo neutralization of SvA and protection/cross-protection against intravaginal challenge with both SvA and the heterologous SvIa strain.

5.
Front Microbiol ; 10: 197, 2019.
Article En | MEDLINE | ID: mdl-30800114

The development of a vaccine against genital chlamydia in women is advancing, and the evaluation of in situ immune responses following vaccination and challenge infections is crucial for development of a safe and protective vaccine. This study employs the sexually mature minipig model to characterize the genital in situ immune response to Chlamydia trachomatis infection in pigs previously immunized intramuscularly with UV-inactivated C. trachomatis serovar D (UV-SvD) adjuvanted/formulated with CAF01 adjuvant compared to a CAF01-alone control group. Pigs immunized with UV-SvD were significantly protected against vaginal challenge with C. trachomatis on day 3 post inoculation and showed significantly higher cervical infiltrations of approximately equal numbers of CD4+ and CD8+ T-cells, and IgG+ and IgA+ plasma cells compared to adjuvant-alone immunized controls. These immunological signatures correspond to findings in mice and are similar to those described in female chlamydia patients. This proves important potential for the pig model in elucidating immunological in situ signatures in future translational research in chlamydia vaccinology.

6.
Front Immunol ; 8: 569, 2017.
Article En | MEDLINE | ID: mdl-28567043

The selection of any specific immunization route is critical when defining future vaccine strategies against a genital infection like Chlamydia trachomatis (C.t.). An optimal Chlamydia vaccine needs to elicit mucosal immunity comprising both neutralizing IgA/IgG antibodies and strong Th1/Th17 responses. A strategic tool to modulate this immune profile and mucosal localization of vaccine responses is to combine parenteral and mucosal immunizations routes. In this study, we investigate whether this strategy can be adapted into a two-visit strategy by simultaneous subcutaneous (SC) and nasal immunization. Using a subunit vaccine composed of C.t. antigens (Ags) adjuvanted with CAF01, a Th1/Th17 promoting adjuvant, we comparatively evaluated Ag-specific B and T cell responses and efficacy in mice following SC and simultaneous SC and nasal immunization (SIM). We found similar peripheral responses with regard to interferon gamma and IL-17 producing Ag-specific splenocytes and IgG serum levels in both vaccine strategies but in addition, the SIM protocol also led to Ag-specific IgA responses and increased B and CD4+ T cells in the lung parenchyma, and in lower numbers also in the genital tract (GT). Following vaginal infection with C.t., we observed that SIM immunization gave rise to an early IgA response and IgA-secreting plasma cells in the GT in contrast to SC immunization, but we were not able to detect more rapid recruitment of mucosal T cells. Interestingly, although SIM vaccination in general improved mucosal immunity we observed no improved efficacy against genital infection compared to SC, a finding that warrants for further investigation. In conclusion, we demonstrate a novel vaccination strategy that combines systemic and mucosal immunity in a two-visit strategy.

7.
Front Immunol ; 8: 1652, 2017.
Article En | MEDLINE | ID: mdl-29312283

There is an unmet need for a vaccine to control Chlamydia trachomatis (C.t.) infections. We have recently designed a multivalent heterologous immuno-repeat 1 (Hirep1) vaccine construct based on major outer membrane protein variable domain (VD) 4 regions from C.t. serovars (Svs) D-F. Hirep1 administered in the Cationic Adjuvant Formulation no. 1 (CAF01) promoted neutralizing antibodies in concert with CD4+ T cells and protected against genital infection. In the current study, we examined the protective role of the antibody (Ab) response in detail. Mice were vaccinated with either Hirep1 or a vaccine construct based on a homologous multivalent construct of extended VD4's from SvF (extVD4F*4), adjuvanted in CAF01. Hirep1 and extVD4F*4 induced similar levels of Ab and cell-mediated immune responses but differed in the fine specificity of the B cell epitopes targeted in the VD4 region. Hirep1 induced a strong response toward a neutralizing epitope (LNPTIAG) and the importance of this epitope for neutralization was demonstrated by competitive inhibition with the corresponding peptide. Immunization with extVD4F*4 skewed the response to a non-neutralizing epitope slightly upstream in the sequence. Vaccination with Hirep1 as opposed to extVD4F*4 induced significant protection against infection in mice both in short- and long-term vaccination experiments, signifying a key role for Hirep1 neutralizing antibodies during protection against C.t. Finally, we show that passive immunization of Rag1 knockout mice with Hirep1 antibodies completely prevented the establishment of infection in 48% of the mice, demonstrating an isolated role for neutralizing antibodies in controlling infection. Our data emphasize the role of antibodies in early protection against C.t. and support the inclusion of neutralizing targets in chlamydia vaccines.

8.
BMC Vet Res ; 12(1): 200, 2016 Sep 10.
Article En | MEDLINE | ID: mdl-27614611

BACKGROUND: Chlamydia is one of the most common sexually transmitted diseases in humans worldwide, causing chronic lesions in the reproductive tract. Due to its often asymptomatic course, there is limited knowledge about the initial changes in the genital tract following infection. This study employs a novel sexually mature minipig model to investigate the initial histopathological changes following vaginal infection with Chlamydia trachomatis serovar D. RESULTS: A vaginal inoculation resulted in an infection primarily affecting the lower genital tract. The histopathological changes were characterized by a subepithelial inflammation consisting of neutrophils and mononuclear cells, followed by an increase in the number of plasma cells within the sub-epithelial stroma of the vagina. Detection of Chlamydia was associated with expression of cyclooxygenase-2 and interleukin-8 by superficial epithelial cells. The infection was self-limiting, with a duration of 7 days. CONCLUSION: Neutrophils, plasma cells and IL-8 have been linked with Chlamydia genital infection of unknown duration in human patients. In this study, we observe a similar pattern of local immune response/inflammation following experimental inoculation suggesting this porcine model shows promise as a model for translational chlamydia research.


Chlamydia Infections/veterinary , Swine Diseases/microbiology , Swine Diseases/pathology , Vagina/pathology , Animals , Chlamydia Infections/microbiology , Chlamydia Infections/pathology , Chlamydia trachomatis/classification , Chlamydia trachomatis/physiology , Cyclooxygenase 2/metabolism , Epithelial Cells/enzymology , Epithelial Cells/pathology , Female , Interleukin-8/metabolism , Serogroup , Swine , Swine, Miniature , Vagina/microbiology
9.
Immunol Cell Biol ; 94(2): 185-95, 2016 Feb.
Article En | MEDLINE | ID: mdl-26268662

Chlamydia is the most widespread sexually transmitted bacterial disease and a prophylactic vaccine is highly needed. Ideally, this vaccine is required to induce a combined response of Th1 cell-mediated immune (CMI) response in concert with neutralizing antibodies. Using a novel Göttingen minipig animal model, we evaluated the immunogenicity and efficacy of a multi-subunit vaccine formulated in the strong Th1-inducing adjuvant CAF01. We evaluated a mixture of two fusion proteins (Hirep1 and CTH93) designed to promote either neutralizing antibodies or cell-mediated immunity, respectively. Hirep1 is a novel immunogen based on the variant domain (VD) 4 region from major outer membrane protein (MOMP) serovar (Sv) D, SvE and SvF, and CTH93 is a fusion molecule of three antigens (CT043, CT414 and MOMP). Pigs were immunized twice intramuscularly with either Hirep1+CTH93/CAF01, UV-inactivated Chlamydia trachomatis SvD bacteria (UV-SvD/CAF01) or CAF01. The Hirep1+CTH93/CAF01 vaccine induced a strong CMI response against the vaccine antigens and high titers of antibodies, particularly against the VD4 region of MOMP. Sera from Hirep1+CTH93/CAF01 immunized pigs neutralized C. trachomatis SvD and SvF infectivity in vitro. Both Hirep1+CTH93/CAF01 and UV-SvD/CAF01 vaccination protected pigs against a vaginal C. trachomatis SvD infection. In conclusion, the Hirep1+CTH93/CAF01 vaccine proved highly immunogenic and equally protective as UV-SvD/CAF01 showing promise for the development of a subunit vaccine against Chlamydia.


Bacterial Outer Membrane Proteins/immunology , Bacterial Vaccines/immunology , Chlamydia Infections/prevention & control , Chlamydia/immunology , Th1 Cells/immunology , Animals , Antibodies, Neutralizing/blood , Bacterial Outer Membrane Proteins/administration & dosage , Bacterial Outer Membrane Proteins/genetics , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/genetics , Disease Models, Animal , Humans , Immunity, Cellular , Immunization , Interferon-gamma/immunology , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Swine , Swine, Miniature , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/genetics , Vaccines, Inactivated/immunology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology
10.
Front Immunol ; 6: 628, 2015.
Article En | MEDLINE | ID: mdl-26734002

International efforts in developing a vaccine against Chlamydia trachomatis have highlighted the need for novel immunization strategies for the induction of genital immunity. In this study, we evaluated an intramuscular (IM) prime/intranasal boost vaccination strategy in a Göttingen Minipig model with a reproductive system very similar to humans. The vaccine was composed of C. trachomatis subunit antigens formulated in the Th1/Th17 promoting CAF01 adjuvant. IM priming immunizations with CAF01 induced a significant cell-mediated interferon gamma and interleukin 17A response and a significant systemic high-titered neutralizing IgG response. Following genital challenge, intranasally boosted groups mounted an accelerated, highly significant genital IgA response that correlated with enhanced bacterial clearance on day 3 post infection. By detecting antigen-specific secretory component (SC), we showed that the genital IgA was locally produced in the genital mucosa. The highly significant inverse correlation between the vaginal IgA SC response and the chlamydial load suggests that IgA in the minipig model is involved in protection against C. trachomatis. This is important both for our understanding of protective immunity and future vaccination strategies against C. trachomatis and genital pathogens in general.

11.
Vaccine ; 32(6): 685-92, 2014 Feb 03.
Article En | MEDLINE | ID: mdl-24365515

A vaccine against genital tract infections caused by Chlamydia trachomatis is urgently needed. We have previously identified a number of immunodominant human T- and/or B-cell antigen targets in patients with a C. trachomatis infection. Herein we use a urogenital C. trachomatis mouse model to investigate the protective efficacy of these antigens. C3H/HeN mice were immunized with recombinant antigens formulated in the adjuvant CAF01. Immunity post vaccination was analyzed and the protective efficacy against vaginal challenge with C. trachomatis was monitored by vaginal swabbing. All antigens elicited a significant immune response when administered in CAF01 but the balance between CMI and humoral responses differed markedly for the different antigens. Six (CT443, CT043, CT858, CT610, CT004 and CT681) antigens were found to be protective. We demonstrated by T-cell depletion studies that the protection promoted by the two antigens CT043 and CT004 was mediated by CD4(+) T-cells. Both antigens are frequently recognized by T-cells during a natural Chlamydia infection in humans and if included in a future multi-component Chlamydia vaccine probably would operate mainly through the induction of a CMI response.


Antigens, Bacterial/immunology , CD4-Positive T-Lymphocytes/immunology , Chlamydia Infections/immunology , Chlamydia trachomatis , Immunity, Cellular , Animals , Antibodies, Bacterial/blood , Antibody Formation , Bacterial Vaccines/immunology , Disease Models, Animal , Female , Humans , Mice , Mice, Inbred C3H
12.
PLoS One ; 5(5): e10768, 2010 May 21.
Article En | MEDLINE | ID: mdl-20505822

BACKGROUND: The chlamydial proteins CT443 (OmcB) and CT521 (rl16) have previously been identified as human B and/or T cell targets during a chlamydial infection in humans. Here we compare the protective effector mechanism promoted by a fusion protein composed of CT521 and CT443 (CTH1) with a primary intranasal Chlamydia muridarum infection known to provide high levels of protection against a genital chlamydial challenge. METHODOLOGY/PRINCIPAL FINDINGS: The fusion protein CTH1, adjuvanted with a strong Th1 inducing cationic adjuvant (CAF01), significantly reduced the bacterial shedding compared to a control group in both a C. trachomatis Serovar D and C. muridarum challenge model. The CTH1/CAF01 vaccine was found to induce polyfunctional T cells consisting of TNFalpha/IL-2 and TNFalpha/IL-2/IFN-gamma positive cells and high titers of CTH1 specific IgG2a and IgG1. By depletion experiments the protection in the C. muridarum challenge model was demonstrated to be mediated solely by CD4(+) T cells. In comparison, an intranasal infection with C. muridarum induced a T cell response that consisted predominantly of TNFalpha/IFN-gamma co-expressing effector CD4(+) T cells and an antibody response consisting of C. muridarum specific IgG1, IgG2a but also IgA. This response was associated with a high level of protection against challenge-a protection that was only partially dependent on CD4(+) T cells. Furthermore, whereas the antibody response induced by intranasal infection was strongly reactive against the native antigens displayed in the chlamydial elementary body, only low levels of antibodies against this preparation were found after CTH1/CAF01 immunization. CONCLUSIONS/SIGNIFICANCE: Our data demonstrate that CTH1 vaccination promotes a CD4(+) T cell dependent protective response but compared with intranasal C. muridarum infection lacks a CD4 independent protective mechanism for complete protection.


Chlamydia Infections/immunology , Chlamydia Infections/prevention & control , Genitalia/immunology , Genitalia/microbiology , Vaccines, Subunit/immunology , Administration, Intranasal , Animals , Antibodies, Bacterial/immunology , Antibody Specificity/immunology , Bacterial Proteins/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/microbiology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/microbiology , Chlamydia Infections/microbiology , Chlamydia muridarum/immunology , Cytokines/biosynthesis , Disease Models, Animal , Electrophoresis, Polyacrylamide Gel , Flow Cytometry , Humans , Kinetics , Lymphocyte Depletion , Mice , Recombinant Fusion Proteins/immunology , Vaccination
13.
J Infect Dis ; 197(6): 897-905, 2008 Mar 15.
Article En | MEDLINE | ID: mdl-18288899

The obligate intracellular bacterium Chlamydia trachomatis is the causative agent of sexually transmitted chlamydia infections. A panel of 116 recombinant C. trachomatis proteins was evaluated comparatively to characterize both cell-mediated and humoral immune responses in patients with confirmed C. trachomatis genital infection. The antigens identified were categorized as being recognized exclusively by T cells (CT004, CT043, CT184, CT509, and CT611), B cells (CT082, CT089, CT322, CT396, and CT681), or both T cells and B cells (CT110 and CT443). This grouping of C. trachomatis antigens was correlated to their predicted cellular localization. The comparative evaluation presented here indicates that T cell antigens are located in all bacterial compartments, whereas antibody targets are mainly localized to the outer membrane (P = .0013). Overall, we have identified 5 T cell antigens, 5 B cell antigens, and 2 T/B cell antigens that are potential components for a future chlamydia vaccine.


Antigens, Bacterial/genetics , Chlamydia Infections/immunology , Chlamydia trachomatis/genetics , Chlamydia trachomatis/immunology , Adolescent , Adult , Aged , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Antigens, Bacterial/biosynthesis , Antigens, Bacterial/immunology , B-Lymphocytes/immunology , Chlamydia Infections/microbiology , Chlamydia trachomatis/isolation & purification , Female , Gene Expression , Gene Library , Humans , Male , Middle Aged , Subcellular Fractions/immunology , T-Lymphocytes/immunology
14.
J Infect Dis ; 196(10): 1546-52, 2007 Nov 15.
Article En | MEDLINE | ID: mdl-18008235

The specificity of the human T cell response to Chlamydia trachomatis was investigated by stimulating lymphocytes from 16 case patients with urogenital infection by use of a size-fractionated serovar D lysate. Considerable heterogeneity was found among case patients, and multiple protein fractions were recognized in each specimen. Mass spectrometry analysis of the 30-42-kDa T cell-stimulating region identified 10 C. trachomatis proteins. Of these, CT583, CT603, and CT610 were identified as strong antigens that induced significantly higher levels of IFN- gamma secretion in PBMCs from case patients, compared with PBMCs from control donors. All 3 proteins were recognized in specimens from case patients infected with serovars D-F, the most prevalent serovars. McDonald-Kreitman and Tajima's D tests involving clinical isolates from the same samples showed evidence for frequency-dependent selection on ct583. We predict that CT583 is a target of acquired protective immune responses in humans.


Chlamydia Infections/immunology , Chlamydia trachomatis/immunology , T-Lymphocytes/physiology , Case-Control Studies , Chlamydia Infections/blood , Chlamydia Infections/microbiology , Chlamydia trachomatis/genetics , DNA Primers , DNA, Bacterial/analysis , Humans , Polymerase Chain Reaction
15.
J Infect Dis ; 194(9): 1258-66, 2006 Nov 01.
Article En | MEDLINE | ID: mdl-17041852

BACKGROUND: The human immune response to a Chlamydia trachomatis serovar D lysate was investigated in patients with urogenital C. trachomatis infection, to identify novel T cell targets. METHODS: A C. trachomatis lysate was fractionated on the basis of molecular mass, and each fraction was used to stimulate peripheral-blood mononuclear cells from patients with C. trachomatis infection. In frequently recognized fractions, proteins were identified by mass spectrometry, recombinantly expressed, and tested for T cell recognition. RESULTS: T cell recognition of the fractions was highly heterogeneous in patients with C. trachomatis infection (n=16). Four patients exhibited responses that were strongly targeted to antigens of 16-20-kDa molecular mass. Three proteins were identified in this fraction: CT043, CT511, and CT521. The T cell response to the individual recombinant proteins were investigated, and CT521 was found to induce the highest level of interferon (IFN)- gamma. The recognition of CT521 was investigated in a larger study population (n=41), and a positive IFN-gamma response was measured in 83% of the patients. Several T cell epitopes were identified in CT521; in particular, peptide 5 in the central part of the protein was frequently recognized by T cells (63%). CONCLUSION: We have identified a novel C. trachomatis antigen, CT521, that is frequently recognized in patients with urogenital C. trachomatis infection.


Chlamydia Infections/immunology , Chlamydia trachomatis/immunology , Th1 Cells/physiology , Adult , Antigens, Bacterial/immunology , Epitopes, T-Lymphocyte/immunology , Female , Female Urogenital Diseases/microbiology , Humans , Male , Male Urogenital Diseases/microbiology
16.
Vaccine ; 24(26): 5452-60, 2006 Jun 29.
Article En | MEDLINE | ID: mdl-16675077

In this study, we evaluated the potential of a novel synthetic adjuvant designated IC31 for the ability to augment the immune response and protective efficacy of the well-known mycobacterial vaccine antigen, Ag85B-ESAT-6. The IC31 adjuvant, consisting of a vehicle based on the cationic peptide KLKL(5)KLK and the immunostimulatory oligodeoxynucleotide ODN1a signalling through the TLR9 receptor, was found to promote highly efficient Th1 responses. The combination of Ag85B-ESAT-6 and IC31 exhibited significant levels of protection in the mouse aerosol challenge model of tuberculosis and a detailed analysis of the immune response generated revealed the induction of CD4 T cells giving rise to high levels of IFN-gamma secretion. Furthermore, the combination of Ag85B-ESAT-6/IC31 was found to confer efficient protection in the guinea pig aerosol model of tuberculosis infection and is at present moving towards clinical testing.


Adjuvants, Immunologic/pharmacology , Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Tuberculosis Vaccines/immunology , Tuberculosis/prevention & control , Animals , Female , Guinea Pigs , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Recombinant Proteins
...