Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.303
Filter
1.
Anticancer Res ; 44(7): 2921-2931, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38925841

ABSTRACT

BACKGROUND/AIM: Human papillomavirus (HPV)-positive head and neck squamous cell carcinoma (HNSCC) is clinically and immunologically distinct from HPV-negative HNSCC. Herein, we investigated the presence of tumor antigens HPV E6/E7 and wild-type p53-specific T-cell responses, and the impact of immune checkpoint blockade in patients with HPV-positive HNSCC. MATERIALS AND METHODS: Peripheral blood mononuclear cells (PBMCs) from patients with HPV-positive HNSCC were stimulated with HPV E6/E7 or wild-type p53-derived peptide mixture and evaluated using the interferon-γ enzyme-linked immunosorbent spot assay. Flow cytometry was performed to analyze the proportion of T-cell subsets and T cells expressing immune checkpoint molecules. RESULTS: HPV E6/E7-specific T cells were detected in 22 (95.7%) of 23 patients, whereas wild-type p53-specific T cells were detected in 3 (15.0%) of 20 patients. Seven (43.8%) of 16 patients exhibited wild-type p53-specific T-cell responses, as determined using whole proteins instead of peptides. Immune checkpoint blockade enhanced wild-type p53-specific T-cell responses in 9 (45.0%) of 20 patients. Flow cytometric analysis of PBMCs revealed that responders exhibiting enhanced wild-type p53-specific T-cell responses following immune checkpoint blockade had a significantly higher proportion of Ki-67+CD4+ T cells, Ki-67+CD8+ T cells, regulatory T cells, PD-1+CD4+ T cells, and TIM-3+CD4+ T cells than non-responders. CONCLUSION: Our findings indicate that tumor antigen-specific T cells are present in the peripheral blood of patients with HPV-positive HNSCC. Blockade of checkpoint pathways can enhance T-cell responses in certain patients, probably via activated T cells, Tregs, and/or exhausted CD4+ T cells.


Subject(s)
Head and Neck Neoplasms , Immune Checkpoint Inhibitors , Papillomavirus Infections , Squamous Cell Carcinoma of Head and Neck , Humans , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Squamous Cell Carcinoma of Head and Neck/immunology , Squamous Cell Carcinoma of Head and Neck/virology , Squamous Cell Carcinoma of Head and Neck/drug therapy , Male , Female , Middle Aged , Aged , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/virology , Papillomavirus Infections/immunology , Papillomavirus Infections/virology , Antigens, Neoplasm/immunology , Oncogene Proteins, Viral/immunology , Tumor Suppressor Protein p53/immunology , Adult , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Papillomaviridae/immunology , T-Lymphocytes/immunology , Human Papillomavirus Viruses
2.
BMC Genomics ; 25(1): 507, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38778248

ABSTRACT

BACKGROUND: Alpha-papillomavirus 9 (α-9) is a member of the human papillomavirus (HPV) α genus, causing 75% invasive cervical cancers worldwide. The purpose of this study was to provide data for effective treatment of HPV-induced cervical lesions in Taizhou by analysing the genetic variation and antigenic epitopes of α-9 HPV E6 and E7. METHODS: Cervical exfoliated cells were collected for HPV genotyping. Positive samples of the α-9 HPV single type were selected for E6 and E7 gene sequencing. The obtained nucleotide sequences were translated into amino acid sequences (protein primary structure) using MEGA X, and positive selection sites of the amino acid sequences were evaluated using PAML. The secondary and tertiary structures of the E6 and E7 proteins were predicted using PSIPred, SWISS-MODEL, and PyMol. Potential T/B-cell epitopes were predicted by Industrial Engineering Database (IEDB). RESULTS: From 2012 to 2023, α-9 HPV accounted for 75.0% (7815/10423) of high-risk HPV-positive samples in Taizhou, both alone and in combination with other types. Among these, single-type-positive samples of α-9 HPV were selected, and the entire E6 and E7 genes were sequenced, including 298 HPV16, 149 HPV31, 185 HPV33, 123 HPV35, 325 HPV52, and 199 HPV58 samples. Compared with reference sequences, 34, 12, 10, 2, 17, and 17 nonsynonymous nucleotide mutations were detected in HPV16, 31, 33, 35, 52, and 58, respectively. Among all nonsynonymous nucleotide mutations, 19 positive selection sites were selected, which may have evolutionary significance in rendering α-9 HPV adaptive to its environment. Immunoinformatics predicted 57 potential linear and 59 conformational B-cell epitopes, many of which are also predicted as CTL epitopes. CONCLUSION: The present study provides almost comprehensive data on the genetic variations, phylogenetics, positive selection sites, and antigenic epitopes of α-9 HPV E6 and E7 in Taizhou, China, which will be helpful for local HPV therapeutic vaccine development.


Subject(s)
Oncogene Proteins, Viral , Phylogeny , China , Humans , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/immunology , Female , Papillomavirus E7 Proteins/genetics , Papillomavirus E7 Proteins/immunology , Alphapapillomavirus/genetics , Alphapapillomavirus/immunology , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/genetics , Epitopes/immunology , Epitopes/genetics , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/genetics , Papillomavirus Infections/virology , Amino Acid Sequence
3.
Viruses ; 16(5)2024 04 30.
Article in English | MEDLINE | ID: mdl-38793599

ABSTRACT

Breast cancer is the most common neoplasm worldwide. Viral infections are involved with carcinogenesis, especially those caused by oncogenic Human Papillomavirus (HPV) genotypes. Despite the detection of HPV in breast carcinomas, the virus's activity against this type of cancer remains controversial. HPV infection promotes remodeling of the host's immune response, resulting in an immunosuppressive profile. This study assessed the individual role of HPV oncogenes in the cell line MDA-MB-231 transfected with the E5, E6, and E7 oncogenes and co-cultured with peripheral blood mononuclear cells. Immunophenotyping was conducted to evaluate immune system modulation. There was an increase in CD4+ T cell numbers when compared with non-transfected and transfected MDA-MB-231, especially in the Treg profile. Pro-inflammatory intracellular cytokines, such as IFN-γ, TNF-α, and IL-17, were impaired by transfected cells, and a decrease in the cytolytic activity of the CD8+ and CD56+ lymphocytes was observed in the presence of HPV oncogenes, mainly with E6 and E7. The E6 and E7 oncogenes decrease monocyte expression, activating the expected M1 profile. In the monocytes found, a pro-inflammatory role was observed according to the cytokines released in the supernatant. In conclusion, the MDA-MB-231 cell lineage transfected with HPV oncogenes can downregulate the number and function of lymphocytes and monocytes.


Subject(s)
Breast Neoplasms , Cytokines , Humans , Female , Cytokines/metabolism , Breast Neoplasms/immunology , Breast Neoplasms/virology , Breast Neoplasms/genetics , Cell Line, Tumor , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/virology , Papillomavirus Infections/immunology , Papillomavirus Infections/virology , Transfection , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/immunology , Oncogene Proteins, Viral/metabolism , Papillomaviridae/genetics , Papillomaviridae/immunology , Human Papillomavirus Viruses
4.
J Med Virol ; 96(6): e29685, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38783790

ABSTRACT

Human papillomaviruses (HPVs) are double-stranded DNA (dsDNA) tumor viruses causally associated with 5% of human cancers, comprising both anogenital and upper aerodigestive tract carcinomas. Despite the availability of prophylactic vaccines, HPVs continue to pose a significant global health challenge, primarily due to inadequate vaccine access and coverage. These viruses can establish persistent infections by evading both the intrinsic defenses of infected tissues and the extrinsic defenses provided by professional innate immune cells. Crucial for their evasion strategies is their unique intraepithelial life cycle, which effectively shields them from host detection. Thus, strategies aimed at reactivating the innate immune response within infected or transformed epithelial cells, particularly through the production of type I interferons (IFNs) and lymphocyte-recruiting chemokines, are considered viable solutions to counteract the adverse effects of persistent infections by these oncogenic viruses. This review focuses on the complex interplay between the high-risk HPV oncoproteins E6 and E7 and the innate immune response in epithelial cells and HPV-associated cancers. In particular, it details the molecular mechanisms by which E6 and E7 modulate the innate immune response, highlighting significant progress in our comprehension of these processes. It also examines forward-looking strategies that exploit the innate immune system to ameliorate existing anticancer therapies, thereby providing crucial insights into future therapeutic developments.


Subject(s)
Immune Evasion , Immunity, Innate , Oncogene Proteins, Viral , Papillomavirus Infections , Humans , Papillomavirus Infections/immunology , Papillomavirus Infections/virology , Oncogene Proteins, Viral/immunology , Papillomavirus E7 Proteins/immunology , Papillomaviridae/immunology , Papillomaviridae/pathogenicity , Host-Pathogen Interactions/immunology , Epithelial Cells/virology , Epithelial Cells/immunology
5.
Hum Vaccin Immunother ; 20(1): 2352908, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-38780076

ABSTRACT

Cervical cancer, among the deadliest cancers affecting women globally, primarily arises from persistent infection with high-risk human papillomavirus (HPV). To effectively combat persistent infection and prevent the progression of precancerous lesions into malignancy, a therapeutic HPV vaccine is under development. This study utilized an immunoinformatics approach to predict epitopes of cytotoxic T lymphocytes (CTLs) and helper T lymphocytes (HTLs) using the E6 and E7 oncoproteins of the HPV16 strain as target antigens. Subsequently, through meticulous selection of T-cell epitopes and other necessary elements, a multi-epitope vaccine was constructed, exhibiting good immunogenic, physicochemical, and structural characteristics. Furthermore, in silico simulations showed that the vaccine not only interacted well with toll-like receptors (TLR2/TLR3/TLR4), but also induced a strong innate and adaptive immune response characterized by elevated Th1-type cytokines, such as interferon-gamma (IFN-γ) and interleukin-2 (IL2). Additionally, our study investigated the effects of different immunization intervals on immune responses, aiming to optimize a time-efficient immunization program. In animal model experiments, the vaccine exhibited robust immunogenic, therapeutic, and prophylactic effects. Administered thrice, it consistently induced the expansion of specific CD4 and CD8 T cells, resulting in substantial cytokines release and increased proliferation of memory T cell subsets in splenic cells. Overall, our findings support the potential of this multi-epitope vaccine in combating HPV16 infection and signify its candidacy for future HPV vaccine development.


Through the stringent selection of T-cell epitopes and other necessary elements, a novel multi-epitope vaccine targeting HPV 16 E6 and E7 oncoproteins was constructed using an immunoinformatics approach.The vaccine designed can induce both cellular and humoral immune responses, encompassing all the required immunogenic, physicochemical, and structural characteristics for an ideal vaccine design. Moreover, it offers decent worldwide coverage.In animal studies, the vaccine demonstrated strong immune responses, including expansion of CD4 and CD8 T cells, cytokine release, and enhanced memory T cell proliferation, resulting in long-term anti-tumor effects, inhibition of tumor growth, and prolonged survival in tumor-bearing mice.The immunological evaluation of the designed vaccine suggests its potential as a novel vaccine candidate against HPV 16.


Subject(s)
Epitopes, T-Lymphocyte , Human papillomavirus 16 , Oncogene Proteins, Viral , Papillomavirus Infections , Papillomavirus Vaccines , Uterine Cervical Neoplasms , Vaccines, DNA , Female , Papillomavirus Vaccines/immunology , Papillomavirus Vaccines/administration & dosage , Human papillomavirus 16/immunology , Vaccines, DNA/immunology , Vaccines, DNA/administration & dosage , Papillomavirus Infections/prevention & control , Papillomavirus Infections/immunology , Epitopes, T-Lymphocyte/immunology , Animals , Oncogene Proteins, Viral/immunology , Oncogene Proteins, Viral/genetics , Uterine Cervical Neoplasms/prevention & control , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/virology , Papillomavirus E7 Proteins/immunology , Mice , Humans , T-Lymphocytes, Cytotoxic/immunology , Repressor Proteins/immunology , T-Lymphocytes, Helper-Inducer/immunology , Mice, Inbred C57BL , Interferon-gamma/metabolism , Interferon-gamma/immunology
6.
Virol J ; 21(1): 124, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38822328

ABSTRACT

Cervical cancer (CC) and other malignant malignancies are acknowledged to be primarily caused by persistent human papillomavirus (HPV) infection. Historically, vaccinations against viruses that produce neutralizing antibodies unique to the virus have been an affordable way to manage viral diseases. CC risk is decreased, but not eliminated, by HPV vaccinations. Since vaccinations have been made available globally, almost 90% of HPV infections have been successfully avoided. On the lesions and diseases that are already present, however, no discernible treatment benefit has been shown. As a result, therapeutic vaccines that elicit immune responses mediated by cells are necessary for the treatment of established infections and cancers. mRNA vaccines possess remarkable potential in combating viral diseases and malignancy as a result of their superior industrial production, safety, and efficacy. Furthermore, considering the expeditiousness of production, the mRNA vaccine exhibits promise as a therapeutic approach targeting HPV. Given that the HPV-encoded early proteins, including oncoproteins E6 and E7, are consistently present in HPV-related cancers and pre-cancerous lesions and have crucial functions in the progression and persistence of HPV-related diseases, they serve as ideal targets for therapeutic HPV vaccines. The action mechanism of HPV and HPV-related cancer mRNA vaccines, their recent advancements in clinical trials, and the potential for their therapeutic applications are highlighted in this study, which also offers a quick summary of the present state of mRNA vaccines. Lastly, we highlight a few difficulties with mRNA HPV vaccination clinical practice and provide our thoughts on further advancements in this quickly changing sector. It is expected that mRNA vaccines will soon be produced quickly for clinical HPV prevention and treatment.


Subject(s)
Papillomavirus Infections , Papillomavirus Vaccines , Uterine Cervical Neoplasms , mRNA Vaccines , Humans , Papillomavirus Infections/prevention & control , Papillomavirus Infections/virology , Papillomavirus Infections/immunology , Papillomavirus Vaccines/immunology , Papillomavirus Vaccines/administration & dosage , Uterine Cervical Neoplasms/prevention & control , Uterine Cervical Neoplasms/virology , Uterine Cervical Neoplasms/therapy , Female , Papillomaviridae/immunology , Papillomaviridae/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage , Oncogene Proteins, Viral/immunology , Oncogene Proteins, Viral/genetics , Human Papillomavirus Viruses
7.
Mol Ther ; 32(7): 2340-2356, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38715363

ABSTRACT

Human papillomavirus (HPV) 16 and 18 infections are related to many human cancers. Despite several preventive vaccines for high-risk (hr) HPVs, there is still an urgent need to develop therapeutic HPV vaccines for targeting pre-existing hrHPV infections and lesions. In this study, we developed a lipid nanoparticle (LNP)-formulated mRNA-based HPV therapeutic vaccine (mHTV)-03E2, simultaneously targeting the E2/E6/E7 of both HPV16 and HPV18. mHTV-03E2 dramatically induced antigen-specific cellular immune responses, leading to significant CD8+ T cell infiltration and cytotoxicity in TC-1 tumors derived from primary lung epithelial cells of C57BL/6 mice expressing HPV E6/E7 antigens, mediated significant tumor regression, and prolonged animal survival, in a dose-dependent manner. We further demonstrated significant T cell immunity against HPV16/18 E6/E7 antigens for up to 4 months post-vaccination in immunological and distant tumor rechallenging experiments, suggesting robust memory T cell immunity against relapse. Finally, mHTV-03E2 synergized with immune checkpoint blockade to inhibit tumor growth and extend animal survival, indicating the potential in combination therapy. We conclude that mHTV-03E2 is an excellent candidate therapeutic mRNA vaccine for treating malignancies caused by HPV16 or HPV18 infections.


Subject(s)
Oncogene Proteins, Viral , Papillomavirus Infections , Papillomavirus Vaccines , RNA, Messenger , Animals , Mice , Papillomavirus Vaccines/immunology , Humans , Papillomavirus Infections/immunology , Papillomavirus Infections/virology , Papillomavirus Infections/therapy , Papillomavirus Infections/prevention & control , Female , Oncogene Proteins, Viral/immunology , Oncogene Proteins, Viral/genetics , RNA, Messenger/genetics , RNA, Messenger/immunology , Nanoparticles/chemistry , Human papillomavirus 16/immunology , Human papillomavirus 16/genetics , Mice, Inbred C57BL , Human papillomavirus 18/immunology , Human papillomavirus 18/genetics , Papillomavirus E7 Proteins/immunology , Papillomavirus E7 Proteins/genetics , Cancer Vaccines/immunology , Cancer Vaccines/administration & dosage , Cell Line, Tumor , Disease Models, Animal , CD8-Positive T-Lymphocytes/immunology , Repressor Proteins/immunology , Repressor Proteins/genetics , DNA-Binding Proteins , Liposomes
8.
J Med Virol ; 96(5): e29630, 2024 May.
Article in English | MEDLINE | ID: mdl-38659368

ABSTRACT

The human papillomavirus type 16 (HPV16) causes a large fraction of genital and oropharyngeal carcinomas. To maintain the transformed state, the tumor cells must continuously synthesize the E6 and E7 viral oncoproteins, which makes them tumor-specific antigens. Indeed, specific T cell responses against them have been well documented and CD8+ T cells engineered to express T cell receptors (TCRs) that recognize epitopes of E6 or E7 have been tested in clinical studies with promising results, yet with limited clinical success. Using CD8+ T cells from peripheral blood of healthy donors, we have identified two novel TCRs reactive to an unexplored E618-26 epitope. These TCRs showed limited standalone cytotoxicity against E618-26-HLA-A*02:01-presenting tumor cells. However, a single-signaling domain chimeric antigen receptor (ssdCAR) targeting L1CAM, a cell adhesion protein frequently overexpressed in HPV16-induced cancer, prompted a synergistic effect that significantly enhanced the cytotoxic capacity of NK-92/CD3/CD8 cells armored with both TCR and ssdCAR when both receptors simultaneously engaged their respective targets, as shown by live microscopy of 2-D and 3-D co-cultures. Thus, virus-specific TCRs from the CD8+ T cell repertoire of healthy donors can be combined with a suitable ssdCAR to enhance the cytotoxic capacity of the effector cells and, indirectly, their specificity.


Subject(s)
CD8-Positive T-Lymphocytes , Oncogene Proteins, Viral , Receptors, Antigen, T-Cell , Receptors, Chimeric Antigen , Repressor Proteins , Humans , Oncogene Proteins, Viral/immunology , Oncogene Proteins, Viral/genetics , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/genetics , Repressor Proteins/immunology , Repressor Proteins/genetics , CD8-Positive T-Lymphocytes/immunology , Killer Cells, Natural/immunology , Human papillomavirus 16/immunology , Human papillomavirus 16/genetics , Cytotoxicity, Immunologic , Cell Line, Tumor
9.
Int J Cancer ; 155(2): 251-260, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38577820

ABSTRACT

Human papillomavirus (HPV) proteins may elicit antibody responses in the process toward HPV-related malignancy. However, HPV seroepidemiology in noncervical HPV-related cancers remains poorly understood, particularly in populations with a high prevalence of human immunodeficiency virus (HIV). Using a glutathione S-transferase-based multiplex serology assay, antibodies against E6, E7 and L1 proteins of HPV16 and HPV18 were measured in sera of 535 cases of noncervical HPV-related cancers (anal (n = 104), vulval (n = 211), vaginal (n = 49), penile (n = 37) and oropharyngeal (n = 134)) and 6651 non-infection-related cancer controls, from the Johannesburg Cancer Study that recruited Black South African with newly diagnosed cancer between 1995 and 2016. Logistic and Poisson regression models were used to calculate adjusted odds ratios (aOR) and prevalence ratios (aPR) and 95% confidence intervals (CI) in cases versus controls. HPV16 E6 was more strongly associated with noncervical HPV-related cancers than HPV16 L1 or E7, or HPV18 proteins: anal (females (HPV16 E6 aOR = 11.50;95%CI:6.0-22.2), males (aOR = 10.12;95%CI:4.9-20.8), vulval (aOR = 11.69;95%CI:7.9-17.2), vaginal (aOR = 10.26;95%CI:5.0-21), penile (aOR = 18.95;95%CI:8.9-40), and oropharyngeal (females (aOR = 8.95;95%CI:2.9-27.5), males (aOR = 3.49;95%CI:1.8-7.0)) cancers. HPV16-E6 seropositivity ranged from 24.0% to 35.1% in anal, vulval, vaginal and penile cancer but was significantly lower (11.2%) in oropharyngeal cancer. After adjustment for HIV, prevalence of which increased from 22.2% in 1995-2005 to 54.1% in 2010-2016, HPV16 E6 seropositivity increased by period of diagnosis (aPR for 2010-2016 vs. 1995-2006 = 1.84;95%CI:1.1-3.0). Assuming HPV16 E6 seroprevalence reflects HPV attributable fraction, the proportion of certain noncervical-HPV-related cancers caused by HPV is increasing over time in South Africa. This is expected to be driven by the increasing influence of HIV.


Subject(s)
Antibodies, Viral , HIV Infections , Oncogene Proteins, Viral , Papillomavirus Infections , Humans , Male , Female , South Africa/epidemiology , Papillomavirus Infections/virology , Papillomavirus Infections/epidemiology , Papillomavirus Infections/immunology , Middle Aged , Adult , Antibodies, Viral/blood , Antibodies, Viral/immunology , Oncogene Proteins, Viral/immunology , HIV Infections/epidemiology , HIV Infections/virology , Human papillomavirus 16/immunology , Aged , Oropharyngeal Neoplasms/virology , Oropharyngeal Neoplasms/epidemiology , Seroepidemiologic Studies , Case-Control Studies , Human papillomavirus 18/immunology , Vulvar Neoplasms/virology , Vulvar Neoplasms/epidemiology , Vulvar Neoplasms/blood , Penile Neoplasms/virology , Penile Neoplasms/epidemiology , Penile Neoplasms/blood , Anus Neoplasms/virology , Anus Neoplasms/epidemiology , Anus Neoplasms/blood , Vaginal Neoplasms/virology , Vaginal Neoplasms/epidemiology , Black People , Repressor Proteins/immunology , Neoplasms/epidemiology , Neoplasms/virology , Neoplasms/blood , Neoplasms/immunology , Human Papillomavirus Viruses
10.
Immunology ; 172(3): 375-391, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38471664

ABSTRACT

Persistent human papillomavirus (HPV) infection is associated with multiple malignancies. Developing therapeutic vaccines to eliminate HPV-infected and malignant cells holds significant value. In this study, we introduced a lipid nanoparticle encapsulated mRNA vaccine expressing tHA-mE7-mE6. Mutations were introduced into E6 and E7 of HPV to eliminate their tumourigenicity. A truncated influenza haemagglutinin protein (tHA), which binds to the CD209 receptor on the surface of dendritic cells (DCs), was fused with mE7-mE6 in order to allow efficient uptake of antigen by antigen presenting cells. The tHA-mE7-mE6 (mRNA) showed higher therapeutic efficacy than mE7-mE6 (mRNA) in an E6 and E7+ tumour model. The treatment resulted in complete tumour regression and prevented tumour formation. Strong CD8+ T-cell immune response was induced, contributing to preventing and curing of E6 and E7+ tumour. Antigen-specific CD8+ T were found in spleens, peripheral blood and in tumours. In addition, the tumour infiltration of DC and NK cells were increased post therapy. In conclusion, this study described a therapeutic mRNA vaccine inducing strong anti-tumour immunity in peripheral and in tumour microenvironment, holding promising potential to treat HPV-induced cancer and to prevent cancer recurrence.


Subject(s)
Cancer Vaccines , Dendritic Cells , Oncogene Proteins, Viral , Papillomavirus E7 Proteins , Papillomavirus Infections , Papillomavirus Vaccines , mRNA Vaccines , Animals , Papillomavirus Infections/immunology , Papillomavirus Infections/prevention & control , Papillomavirus E7 Proteins/immunology , Cancer Vaccines/immunology , Oncogene Proteins, Viral/immunology , Oncogene Proteins, Viral/genetics , Papillomavirus Vaccines/immunology , Dendritic Cells/immunology , Humans , Mice , Female , CD8-Positive T-Lymphocytes/immunology , Mice, Inbred C57BL , Nanoparticles , Antigen-Presenting Cells/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Killer Cells, Natural/immunology , Repressor Proteins/immunology , Repressor Proteins/genetics , Neoplasms/therapy , Neoplasms/immunology , RNA, Messenger/genetics , Cell Line, Tumor , Liposomes
11.
Virology ; 567: 15-25, 2022 02.
Article in English | MEDLINE | ID: mdl-34942562

ABSTRACT

HPV68 is a common HR-HPV, its persistent infection is closely related with the occurrence of cervical cancer. In this study, 2939 (27.60%, 2939/10650) positive samples were detected, and 174 (5.92%, 174/2939) were HPV68. 150 HPV68 E6-E7 were successful sequenced, 4 non-synonymous mutations were detected in E6, and E7 were 12. N133S non-synonymous mutations of HPV 68 E6 and C67G, T68 A/M of HPV68 E7 are E6, E7 positive selection sites, they all located in the key domains and major motifs of E6/E7 protein, the above amino-acid substitutions changed the protein structure, disturbed the interaction with other protein or cellular factors and make a difference in epitopes affinity, may affect the pathogenicity and adaptability of HPV68 to the environment. The enrichment of HPV68 data is of great significance for understanding the inherent geographical and biological differences of HPV68 in China.


Subject(s)
Alphapapillomavirus/genetics , Mutation , Oncogene Proteins, Viral/genetics , Papillomavirus E7 Proteins/genetics , Papillomavirus Infections/epidemiology , Alphapapillomavirus/chemistry , Alphapapillomavirus/classification , Alphapapillomavirus/immunology , Amino Acid Sequence , Amino Acid Substitution , B-Lymphocytes/immunology , B-Lymphocytes/virology , Binding Sites , Cervix Uteri/immunology , Cervix Uteri/virology , China/epidemiology , Epitopes/chemistry , Epitopes/genetics , Epitopes/immunology , Female , Genotype , Histocompatibility Antigens Class I/chemistry , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Humans , Models, Molecular , Molecular Typing , Oncogene Proteins, Viral/chemistry , Oncogene Proteins, Viral/immunology , Papillomavirus E7 Proteins/chemistry , Papillomavirus E7 Proteins/immunology , Papillomavirus Infections/immunology , Papillomavirus Infections/virology , Phylogeny , Prevalence , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Structure, Tertiary , Sequence Alignment , Sequence Homology, Amino Acid , T-Lymphocytes/immunology , T-Lymphocytes/virology
12.
Front Immunol ; 12: 742861, 2021.
Article in English | MEDLINE | ID: mdl-34759925

ABSTRACT

Background: Cervical cancer - caused by persistent High Risk Human Papilloma Virus (HR HPV) infections - is the second most common cancer affecting women globally. HIV infection increases the risk for HPV persistence, associated disease progression and malignant cell transformation. We therefore hypothesized that this risk increase is directly linked to HIV infection associated dysfunction or depletion of HPV-oncoprotein-specific T-cell responses. Methods: The 2H study specifically included HIV+ and HIV- women with and without cervical lesions and cancer to analyze HPV oncogene-specific T cell responses in relation to HPV infection, cervical lesion status and HIV status. Oncoprotein E6 and E7 specific T-cell responses were quantified for the most relevant types HPV16, 18 and 45 and control antigens (CMV-pp65) and M.tb-PPD in 373 women, using fresh peripheral blood mononuclear cells in an IFN-γ release ELISpot assay. Results: Overall, systemic E6- and E7-oncoprotein-specific T-cell responses were infrequent and of low magnitude, when compared to CMV-pp65 and M.tb-PPD (p < 0.001 for all HR HPV types). Within HIV negative women infected with either HPV16, 18 or 45, HPV16 infected women had lowest frequency of autologous-type-E6/E7-specific T-cell responses (33%, 16/49), as compared to HPV18 (46% (6/13), p = 0.516) and HPV45 (69% (9/13), p = 0.026) infected women. Prevalent HPV18 and 45, but not HPV16 infections were linked to detectable oncoprotein-specific T-cell responses, and for these infections, HIV infection significantly diminished T-cell responses targeting the autologous infecting genotype. Within women living with HIV, low CD4 T-cell counts, detectable HIV viremia as well as cancerous and precancerous lesions were significantly associated with depletion of HPV oncoprotein-specific T-cell responses. Discussion: Depletion of HPV-oncoprotein-specific T-cell responses likely contributes to the increased risk for HR HPV persistence and associated cancerogenesis in women living with HIV. The low inherent immunogenicity of HPV16 oncoproteins may contribute to the exceptional potential for cancerogenesis associated with HPV16 infections.


Subject(s)
Alphapapillomavirus/immunology , Coinfection/immunology , HIV Infections/immunology , Oncogene Proteins, Viral/immunology , Papillomavirus Infections/immunology , T-Lymphocytes/immunology , Adult , Female , Humans , Middle Aged , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/virology
13.
Nat Commun ; 12(1): 5945, 2021 10 12.
Article in English | MEDLINE | ID: mdl-34642315

ABSTRACT

Although several oropharyngeal cancer (OPC) susceptibility loci have been identified, most previous studies lacked detailed information on human papillomavirus (HPV) status. We conduct a genome-wide analysis by HPV16 serology status in 4,002 oral cancer cases (OPC and oral cavity cancer (OCC)) and 5,256 controls. We detect four susceptibility loci pointing to a distinct genetic predisposition by HPV status. Our most notable finding in the HLA region, that is now confirmed to be specific of HPV(+)OPC risk, reveal two independent loci with strong protective effects, one refining the previously reported HLA class II haplotype association. Antibody levels against HPV16 viral proteins strongly implicate the protective HLA variants as major determinants of humoral response against L1 capsid protein or E6 oncoprotein suggesting a natural immune response against HPV(+)OPC promoted by HLA variants. This indicates that therapeutic vaccines that target E6 and attenuate viral response after established HPV infections might protect against HPV(+)OPC.


Subject(s)
HLA Antigens/immunology , Human papillomavirus 16/immunology , Immunity, Humoral , Mouth Neoplasms/immunology , Oropharyngeal Neoplasms/immunology , Papillomavirus Infections/immunology , Aged , Antibodies, Viral/biosynthesis , Capsid Proteins/genetics , Capsid Proteins/immunology , Case-Control Studies , Female , Gene Expression , Genetic Predisposition to Disease , Genome-Wide Association Study , HLA Antigens/classification , HLA Antigens/genetics , Haplotypes , Human papillomavirus 16/pathogenicity , Humans , Male , Meta-Analysis as Topic , Middle Aged , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Mouth Neoplasms/virology , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/immunology , Oropharyngeal Neoplasms/genetics , Oropharyngeal Neoplasms/pathology , Oropharyngeal Neoplasms/virology , Papillomavirus Infections/genetics , Papillomavirus Infections/pathology , Papillomavirus Infections/virology , Quantitative Trait Loci , Repressor Proteins/genetics , Repressor Proteins/immunology , Risk Factors , Smoking/physiopathology
14.
Int J Mol Sci ; 22(17)2021 Aug 24.
Article in English | MEDLINE | ID: mdl-34502053

ABSTRACT

In recent decades, recombinant antibodies against specific antigens have shown great promise for the therapy of infectious diseases and cancer. Human papillomaviruses (HPVs) are involved in the development of around 5% of all human cancers and HPV16 is the high-risk genotype with the highest prevalence worldwide, playing a dominant role in all HPV-associated cancers. Here, we describe the main biological activities of the HPV16 E6, E7, and E5 oncoproteins, which are involved in the subversion of important regulatory pathways directly associated with all known hallmarks of cancer. We then review the state of art of the recombinant antibodies targeted to HPV oncoproteins developed so far in different formats, and outline their mechanisms of action. We describe the advantages of a possible antibody-based therapy against the HPV-associated lesions and discuss the critical issue of delivery to tumour cells, which must be addressed in order to achieve the desired translation of the antibodies from the laboratory to the clinic.


Subject(s)
Antibodies, Viral/therapeutic use , Neoplasms/drug therapy , Single-Domain Antibodies/therapeutic use , Animals , Antibodies, Viral/immunology , Humans , Neoplasms/virology , Oncogene Proteins, Viral/immunology , Papillomavirus E7 Proteins/immunology , Recombinant Proteins/immunology , Recombinant Proteins/therapeutic use , Repressor Proteins/immunology , Single-Domain Antibodies/immunology
15.
Nature ; 597(7875): 279-284, 2021 09.
Article in English | MEDLINE | ID: mdl-34471285

ABSTRACT

T cells are important in tumour immunity but a better understanding is needed of the differentiation of antigen-specific T cells in human cancer1,2. Here we studied CD8 T cells in patients with human papillomavirus (HPV)-positive head and neck cancer and identified several epitopes derived from HPV E2, E5 and E6 proteins that allowed us to analyse virus-specific CD8 T cells using major histocompatibility complex (MHC) class I tetramers. HPV-specific CD8 T cells expressed PD-1 and were detectable in the tumour at levels that ranged from 0.1% to 10% of tumour-infiltrating CD8 T lymphocytes (TILs) for a given epitope. Single-cell RNA-sequencing analyses of tetramer-sorted HPV-specific PD-1+ CD8 TILs revealed three transcriptionally distinct subsets. One subset expressed TCF7 and other genes associated with PD-1+ stem-like CD8 T cells that are critical for maintaining T cell responses in conditions of antigen persistence. The second subset expressed more effector molecules, representing a transitory cell population, and the third subset was characterized by a terminally differentiated gene signature. T cell receptor clonotypes were shared between the three subsets and pseudotime analysis suggested a hypothetical differentiation trajectory from stem-like to transitory to terminally differentiated cells. More notably, HPV-specific PD-1+TCF-1+ stem-like TILs proliferated and differentiated into more effector-like cells after in vitro stimulation with the cognate HPV peptide, whereas the more terminally differentiated cells did not proliferate. The presence of functional HPV-specific PD-1+TCF-1+CD45RO+ stem-like CD8 T cells with proliferative capacity shows that the cellular machinery to respond to PD-1 blockade exists in HPV-positive head and neck cancer, supporting the further investigation of PD-1 targeted therapies in this malignancy. Furthermore, HPV therapeutic vaccination efforts have focused on E6 and E7 proteins; our results suggest that E2 and E5 should also be considered for inclusion as vaccine antigens to elicit tumour-reactive CD8 T cell responses of maximal breadth.


Subject(s)
Alphapapillomavirus/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/virology , Lymphocytes, Tumor-Infiltrating/immunology , Programmed Cell Death 1 Receptor/metabolism , Stem Cells/cytology , Alphapapillomavirus/isolation & purification , CD8-Positive T-Lymphocytes/classification , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines/immunology , Cell Differentiation , Cell Proliferation , DNA-Binding Proteins/immunology , Humans , Lymphocytes, Tumor-Infiltrating/classification , Lymphocytes, Tumor-Infiltrating/cytology , Lymphocytes, Tumor-Infiltrating/metabolism , Oncogene Proteins, Viral/immunology , Papillomavirus Infections/immunology , Papillomavirus Infections/virology , Papillomavirus Vaccines/immunology , RNA-Seq , Receptors, Antigen, T-Cell/immunology , Single-Cell Analysis , Stem Cells/immunology , T Cell Transcription Factor 1/metabolism , T-Lymphocytes/immunology , Transcription, Genetic
16.
Life Sci ; 285: 119945, 2021 Nov 15.
Article in English | MEDLINE | ID: mdl-34516991

ABSTRACT

AIMS: Human papillomavirus (HPV) L1, L2 and E7 proteins were used as target antigens for development of preventive and therapeutic vaccines. Moreover, linkage of antigens to heat shock proteins (HSPs) could enhance the potency of vaccines. Curcumin and nanocurcumin compounds were suggested as the chemopreventive and chemotherapeutic agents against cancer. In this study, two multiepitope DNA and peptide-based vaccine constructs (L1-L2-E7 and HSP70-L1-L2-E7) were used along with curcumin and nanocurcumin to evaluate immune responses, and protective/therapeutic effects in tumor mouse model. MAIN METHODS: At first, the multiepitope L1-L2-E7 and HSP70-L1-L2-E7 fusion genes were subcloned in eukaryotic and prokaryotic expression vectors. The recombinant multiepitope peptides were generated in E. coli strain. Then, the cytotoxic effects of curcumin and nanocurcumin were evaluated on HEK-293 T non-cancerous and C3 cancerous cells. Finally, mice vaccination was performed using different regimens. Curcumin and nanocurcumin compounds were administered alone or along with different vaccine constructs. KEY FINDINGS: Our data indicated that the use of nanocurcumin along with the multiepitope HSP70-L1-L2-E7 vaccine construct could completely protect mice against HPV-related C3 tumor cells, and eradicate tumors in a therapeutic test. Furthermore, nanocurcumin showed higher protection than curcumin alone. Generally, curcumin and nanocurcumin compounds could reduce tumor growth synergistically with the multiepitope vaccine constructs, but they did not influence the immune responses in different regimens. SIGNIFICANCE: These data demonstrated that the designed multiepitope vaccine constructs along with curcumin and nanocurcumin can be used as a promising method for HPV vaccine development.


Subject(s)
Antineoplastic Agents/pharmacology , Cancer Vaccines/immunology , Capsid Proteins/immunology , Curcumin/pharmacology , Oncogene Proteins, Viral/immunology , Papillomavirus E7 Proteins/immunology , Papillomavirus Vaccines/immunology , Uterine Cervical Neoplasms/immunology , Animals , Antineoplastic Agents/administration & dosage , Cancer Vaccines/genetics , Capsid Proteins/administration & dosage , Capsid Proteins/genetics , Cloning, Molecular , Curcumin/administration & dosage , Cytokines/metabolism , Epitopes, T-Lymphocyte/administration & dosage , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Escherichia coli , Female , Genetic Vectors , HEK293 Cells , HSP70 Heat-Shock Proteins/administration & dosage , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/immunology , Humans , Mice, Inbred C57BL , Neoplasms, Experimental/immunology , Neoplasms, Experimental/therapy , Oncogene Proteins, Viral/administration & dosage , Oncogene Proteins, Viral/genetics , Papillomavirus E7 Proteins/administration & dosage , Papillomavirus E7 Proteins/genetics , Papillomavirus Vaccines/administration & dosage , Papillomavirus Vaccines/genetics , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Uterine Cervical Neoplasms/therapy , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology
17.
J Immunother ; 44(8): 292-306, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34432728

ABSTRACT

Next-generation T-cell therapies will likely continue to utilize T-cell receptors (TCRs) and chimeric antigen receptors (CARs) because each receptor type has advantages. TCRs often possess exceptional properties even when tested unmodified from patients' T cells. CARs are generally less sensitive, possibly because their ligand-binding domains are grafted from antibodies selected for binding affinity or avidity and not broadly optimized for a functional response. Because of the disconnect between binding and function among these receptor types, the ultimate potential of CARs optimized for sensitivity and selectivity is not clear. Here, we focus on a thoroughly studied immuno-oncology target, the HLA-A*02/HPV-E629-38 complex, and show that CARs can be optimized by a combination of high-throughput binding screens and low-throughput functional assays to have comparable activity to clinical TCRs in acute assays in vitro. These results provide a case study for the challenges and opportunities of optimizing high-performing CARs, especially in the context of targets utilized naturally by TCRs.


Subject(s)
Immunotherapy, Adoptive , Neoplasms/therapy , Papillomavirus Infections/therapy , Receptors, Chimeric Antigen/immunology , Cell Line , Green Fluorescent Proteins , HLA-A2 Antigen/immunology , Humans , Interferon-gamma/immunology , Luciferases, Firefly , Neoplasms/immunology , Oncogene Proteins, Viral/immunology , Papillomavirus E7 Proteins/immunology , Papillomavirus Infections/immunology , Peptides/immunology , Repressor Proteins/immunology , Single-Chain Antibodies/immunology
18.
Sci Rep ; 11(1): 13404, 2021 06 28.
Article in English | MEDLINE | ID: mdl-34183739

ABSTRACT

Cervical cancer continues to impose a heavy burden worldwide, and human papilloma virus (HPV) infection, especially persistent infection with type 16 (HPV-16), is known to be the primary etiological factor. Therapeutic vaccines are urgently needed because prophylactic vaccines are ineffective at clearing pre-existing HPV infection. Here, two recombinant Listeria strains (LMΔ-E6E7 & LIΔ-E6E7) with deletions of the actA and plcB genes, expressing the shuffled HPV-16 E6E7 protein were constructed. The strains were delivered into the spleen and liver by intravenous inoculation, induced antigen-specific cellular immunity and were eliminated completely from the internal organs several days later. Intravenously treating with single strain for three times, or with both strains alternately for three times significantly reduced the tumor size and prolonged the survival time of model mice. Combination immunotherapy with two strains seemed more effective than immunotherapy with single strain in that it enhanced the survival of the mice, and the LMΔ-E6E7-prime-LIΔ-E6E7-boost strategy showed significant stronger efficacy than single treatment with the LIΔ-E6E7 strain. The antitumor effect of this treatment might due to its ability to increase the proportion of CD8+ T cells and reduce the proportion of T regulatory cells (Tregs) in the intratumoral milieu. This is the first report regarding Listeria ivanovii-based therapeutic vaccine candidate against cervical cancer. Most importantly we are the first to confirm that combination therapy with two different recombinant Listeria strains has a more satisfactory antitumor effect than administration of a single strain. Thus, we propose a novel prime-boost treatment strategy.


Subject(s)
Human papillomavirus 16/immunology , Listeria/immunology , Oncogene Proteins, Viral/immunology , Papillomavirus E7 Proteins/immunology , Papillomavirus Infections/immunology , Repressor Proteins/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Cancer Vaccines/immunology , Disease Models, Animal , Female , Humans , Immunity, Cellular/immunology , Immunotherapy/methods , Mice , Mice, Inbred C57BL , Papillomavirus Infections/virology , Papillomavirus Vaccines/immunology , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/therapy , Uterine Cervical Neoplasms/virology , Vaccination/methods
19.
Sci Rep ; 11(1): 12397, 2021 06 11.
Article in English | MEDLINE | ID: mdl-34117331

ABSTRACT

Cervical cancer, caused by human papillomavirus (HPV), is the fourth most common type of cancer among women worldwide. While HPV prophylactic vaccines are available, they have no therapeutic effects and do not clear up existing infections. This study aims to design a therapeutic vaccine against cervical cancer using reverse vaccinology. In this study, the E6 and E7 oncoproteins from HPV16 were chosen as the target antigens for epitope prediction. Cytotoxic T lymphocytes (CTL) and helper T lymphocytes (HTL) epitopes were predicted, and the best epitopes were selected based on antigenicity, allergenicity, and toxicity. The final vaccine construct was composed of the selected epitopes, along with the appropriate adjuvant and linkers. The multi-epitope vaccine was evaluated in terms of physicochemical properties, antigenicity, and allergenicity. The tertiary structure of the vaccine construct was predicted. Furthermore, several analyses were also carried out, including molecular docking, molecular dynamics (MD) simulation, and in silico cloning of the vaccine construct. The results showed that the final proposed vaccine could be considered an effective therapeutic vaccine for HPV; however, in vitro and in vivo experiments are required to validate the efficacy of this vaccine candidate.


Subject(s)
Cancer Vaccines/immunology , Epitopes/immunology , Papillomavirus Vaccines/immunology , Uterine Cervical Neoplasms/prevention & control , Cancer Vaccines/chemistry , Computational Biology , Epitopes/chemistry , Female , Humans , Immunogenicity, Vaccine , Molecular Docking Simulation , Oncogene Proteins, Viral/chemistry , Oncogene Proteins, Viral/immunology , Papillomavirus E7 Proteins/chemistry , Papillomavirus E7 Proteins/immunology , Papillomavirus Vaccines/chemistry , Repressor Proteins/chemistry , Repressor Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Helper-Inducer/immunology , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/virology
20.
Front Immunol ; 12: 593161, 2021.
Article in English | MEDLINE | ID: mdl-33717073

ABSTRACT

HPV E5 is an oncoprotein mainly expressed in premalignant lesions, which makes it an important target for a vaccine to prevent or cure cervical cancer (CC). In this study, we evaluated whether E5 targeted to DEC-205, present in dendritic cells (DCs), could induce a therapeutic protection against HPV16-induced tumor cells in a mouse model. The HPV-16 E5 (16E5) protein was cross-linked to a monoclonal antibody (mAb) specific to mouse DEC-205 (anti-DEC-205:16E5) or to an isotype control mAb (isotype:16E5). Rotavirus VP6 was cross-linked to the mouse anti-DEC-205 mAb (anti-DEC-205:VP6) as a non-specific antigen control. BALB/c mice were inoculated subcutaneously (s.c.) with the 16E5-expressing BMK-16/myc tumor cells, and 7 and 14 days later the mice were immunized s.c. with the conjugates, free 16E5 or PBS in the presence of adjuvant. Tumor growth was monitored to evaluate protection. A strong protective immune response against the tumor cells was induced when the mice were inoculated with the anti-DEC-205:16E5 conjugate, since 70% of the mice controlled the tumor growth and survived, whereas the remaining 30% developed tumors and died by day 72. In contrast, 100% of the mice in the control groups died by day 30. The anti-DEC-205:16E5 conjugate was found to induce 16E5-specific memory T cells, with a Th1/Th17 profile. Both CD4+ and CD8+ T cells contributed to the observed protection. Finally, treating mice that had developed tumors with an anti-PD-1 mAb, delayed the tumor growth for more than 20 days. These results show that targeting 16E5 to DEC-205, alone or combined with an immune checkpoint blockade, could be a promising protocol for the treatment of the early stages of HPV-associated cancer.


Subject(s)
Dendritic Cells/immunology , Human papillomavirus 16/immunology , Neoplasms/etiology , Neoplasms/therapy , Oncogene Proteins, Viral/immunology , Papillomavirus Infections/complications , Animals , Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Biomarkers, Tumor , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Cell Line, Tumor , Dendritic Cells/metabolism , Disease Models, Animal , Female , Humans , Immunization , Immunologic Memory , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Mice , Neoplasms/diagnosis , Papillomavirus Infections/virology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...