Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
BMC Genomics ; 25(1): 409, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38664626

ABSTRACT

OBJECTIVE: To evaluate the contribution of germline genetics to regulating the briskness and diversity of T cell responses in CRC, we conducted a genome-wide association study to examine the associations between germline genetic variation and quantitative measures of T cell landscapes in 2,876 colorectal tumors from participants in the Molecular Epidemiology of Colorectal Cancer Study (MECC). METHODS: Germline DNA samples were genotyped and imputed using genome-wide arrays. Tumor DNA samples were extracted from paraffin blocks, and T cell receptor clonality and abundance were quantified by immunoSEQ (Adaptive Biotechnologies, Seattle, WA). Tumor infiltrating lymphocytes per high powered field (TILs/hpf) were scored by a gastrointestinal pathologist. Regression models were used to evaluate the associations between each variant and the three T-cell features, adjusting for sex, age, genotyping platform, and global ancestry. Three independent datasets were used for replication. RESULTS: We identified a SNP (rs4918567) near RBM20 associated with clonality at a genome-wide significant threshold of 5 × 10- 8, with a consistent direction of association in both discovery and replication datasets. Expression quantitative trait (eQTL) analyses and in silico functional annotation for these loci provided insights into potential functional roles, including a statistically significant eQTL between the T allele at rs4918567 and higher expression of ADRA2A (P = 0.012) in healthy colon mucosa. CONCLUSIONS: Our study suggests that germline genetic variation is associated with the quantity and diversity of adaptive immune responses in CRC. Further studies are warranted to replicate these findings in additional samples and to investigate functional genomic mechanisms.


Subject(s)
Colorectal Neoplasms , Genome-Wide Association Study , Polymorphism, Single Nucleotide , Tumor Microenvironment , Humans , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Male , Female , Middle Aged , Quantitative Trait Loci , Aged , Lymphocytes, Tumor-Infiltrating/immunology , Germ-Line Mutation , RNA-Binding Proteins/genetics , Genotype , Germ Cells/metabolism
2.
Front Immunol ; 14: 1268117, 2023.
Article in English | MEDLINE | ID: mdl-37942321

ABSTRACT

Objective: Reduced diversity at Human Leukocyte Antigen (HLA) loci may adversely affect the host's ability to recognize tumor neoantigens and subsequently increase disease burden. We hypothesized that increased heterozygosity at HLA loci is associated with a reduced risk of developing colorectal cancer (CRC). Methods: We imputed HLA class I and II four-digit alleles using genotype data from a population-based study of 5,406 cases and 4,635 controls from the Molecular Epidemiology of Colorectal Cancer Study (MECC). Heterozygosity at each HLA locus and the number of heterozygous genotypes at HLA class -I (A, B, and C) and HLA class -II loci (DQB1, DRB1, and DPB1) were quantified. Logistic regression analysis was used to estimate the risk of CRC associated with HLA heterozygosity. Individuals with homozygous genotypes for all loci served as the reference category, and the analyses were adjusted for sex, age, genotyping platform, and ancestry. Further, we investigated associations between HLA diversity and tumor-associated T cell repertoire features, as measured by tumor infiltrating lymphocytes (TILs; N=2,839) and immunosequencing (N=2,357). Results: Individuals with all heterozygous genotypes at all three class I genes had a reduced odds of CRC (OR: 0.74; 95% CI: 0.56-0.97, p= 0.031). A similar association was observed for class II loci, with an OR of 0.75 (95% CI: 0.60-0.95, p= 0.016). For class-I and class-II combined, individuals with all heterozygous genotypes had significantly lower odds of developing CRC (OR: 0.66, 95% CI: 0.49-0.87, p= 0.004) than those with 0 or one heterozygous genotype. HLA class I and/or II diversity was associated with higher T cell receptor (TCR) abundance and lower TCR clonality, but results were not statistically significant. Conclusion: Our findings support a heterozygote advantage for the HLA class-I and -II loci, indicating an important role for HLA genetic variability in the etiology of CRC.


Subject(s)
Colorectal Neoplasms , Histocompatibility Antigens Class I , Humans , Heterozygote , Gene Frequency , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class II/genetics , HLA Antigens , Colorectal Neoplasms/genetics , Receptors, Antigen, T-Cell/genetics
3.
Viruses ; 15(6)2023 06 06.
Article in English | MEDLINE | ID: mdl-37376627

ABSTRACT

The aim of this study was to classify the diversity of anal HPV and non-HPV sexually transmitted infections (STIs) and compare the concordance between anal and genital infections in HIV-infected and uninfected women living in the Tapajós region, Amazon, Brazil. A cross-sectional study was performed with 112 HIV-uninfected and 41 HIV-infected nonindigenous women. Anal and cervical scrapings were collected and analyzed for HPV, Chlamydia trachomatis (CT), Neisseria gonorrheae (NG), Trichomonas vaginalis (TV), Mycoplasma genitalium (MG), and Human alphaherpesvirus 2 (HSV-2). The Kappa test evaluated the concordance between anal and genital infections. The overall prevalence of anal HPV infection was 31.3% in HIV-uninfected and 97.6% in HIV-infected women. The most frequent anal high-risk HPV (hrHPV) types were HPV18 and HPV16 in HIV-uninfected women and HPV51, HPV59, HPV31, and HPV58 in HIV-infected women. Anal HPV75 Betapapillomavirus was also identified. Anal non-HPV STIs were identified in 13.0% of all participants. The concordance analysis was fair for CT, MG, and HSV-2, almost perfect agreement for NG, moderate for HPV, and variable for the most frequent anal hrHPV types. Thus, a high prevalence of anal HPV infection with moderate and fair concordance between anal and genital HPV and non-HPV STIs was observed in our study.


Subject(s)
Chlamydia Infections , HIV Infections , Papillomavirus Infections , Sexually Transmitted Diseases , Humans , Female , Papillomavirus Infections/complications , Papillomavirus Infections/epidemiology , Brazil/epidemiology , Cross-Sectional Studies , Sexually Transmitted Diseases/complications , Sexually Transmitted Diseases/epidemiology , Chlamydia trachomatis , Cervix Uteri , Neisseria gonorrhoeae , HIV Infections/complications , HIV Infections/epidemiology , Prevalence , Chlamydia Infections/complications , Chlamydia Infections/epidemiology
4.
Cancers (Basel) ; 14(17)2022 Sep 05.
Article in English | MEDLINE | ID: mdl-36077873

ABSTRACT

Therapeutic vaccine studies should be designed to elicit durable, high magnitude, and efficacious T cell responses, all of which can be impacted by the choice of the vaccination schedule. Here, we compare different prime-boost intervals (PBI) in a human papillomavirus (HPV) model using a HPV16E7E6 Venezuelan equine encephalitis virus replicon particle (VRP) vaccination to address the optimal boosting schedule, quality of immune response, and overall in vivo efficacy. Six different vaccine regimens were tested with each group receiving booster vaccinations at different time intervals. Analysis of T-cell responses demonstrated a significant HPV16 E7 specific CD8+ T cell response with at minimum a one-week PBI between antigen re-exposure. Significant E7-specific in vivo cytotoxicity was also observed with longer PBIs. Additionally, longer PBIs led to an enhanced memory recall response to tumor challenge, which correlated with differential expansion of T cell memory subsets. Our findings imply that when using alphavirus vector platforms as a vaccination strategy, a one-week PBI is sufficient to induce high magnitude effector T cells with potent anti-tumor activity. However, longer PBIs lead to enhanced long-term protective anti-tumor immunity. These findings have implications for therapeutic vaccine clinical trials in which shorter intervals of prime-boost regimens may lead to suboptimal durable immune responses.

5.
Arch Gynecol Obstet ; 305(2): 439-447, 2022 02.
Article in English | MEDLINE | ID: mdl-34392396

ABSTRACT

OBJECTIVE: To validate our previous findings of high-level EGFR expression in GCCC using an expanded cohort of specimens and to further examine the molecular and cellular features of this aggressive malignancy to identify potentially actionable therapeutic targets. METHODS: The SEER database was queried to obtain the epidemiological data regarding the current national survival trends for GCCC. Immunohistochemistry (IHC) was used to examine the expression of EGFR, PD-1, and PD-L1. CiberSort analysis was used to analyze a previously published RNA-sequencing dataset obtained from a single patient diagnosed with GCCC. RESULTS: In comparison to squamous cell carcinomas and adenocarcinoma/adenosquamous carcinomas, GCCC was observed in younger patients (p < 0.001) and demonstrated inferior survival (p < 0.001). All (100%) of the specimens (8/8) exhibited immunoreactivity when stained for CD3ε (T-cell marker), EGFR, PD-1, and PD-L1 whereas CTLA4 expression was not detected. Analysis of RNA-sequencing data revealed that cetuximab and erlotinib altered the chemokine profile, lymphocyte abundance, and expression of inhibitory immune checkpoints in a single patient when combined with cytotoxic chemotherapy in a single patient. CONCLUSIONS: The data from this descriptive study suggests that immune checkpoint blockade, whether single agent or in combination, may be a suitable therapeutic option for a disease for which targeted approaches do not currently exist.


Subject(s)
Carcinoma, Adenosquamous , Carcinoma, Squamous Cell , Uterine Cervical Neoplasms , B7-H1 Antigen/metabolism , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/pathology , Cervix Uteri/pathology , Female , Humans , Uterine Cervical Neoplasms/pathology
6.
Am J Obstet Gynecol ; 225(6): 656.e1-656.e11, 2021 12.
Article in English | MEDLINE | ID: mdl-34273278

ABSTRACT

BACKGROUND: The risk of a high-grade lesion in women undergoing colposcopy following an abnormal screening result may be different by human papillomavirus vaccination status, because women who are vaccinated are presumably less likely to harbor human papillomavirus types 16 and 18. OBJECTIVE: This study aimed to evaluate whether the risk of high-grade cervical lesion diagnosed through colposcopy is lower in women with human papillomavirus vaccination than in women without vaccination referred to colposcopy based on equal abnormal screening findings. STUDY DESIGN: Kaiser Permanente Orange County female patients between ages 21 and 38 years were included following an abnormal screening if they had ≥1 colposcopies between July 2017 and August 2018 and had at least 1 pathology diagnosis from the colposcopy visits. Data on demographic characteristics, clinical and sexual histories, and human papillomavirus vaccination were collected using a colposcopy registry smart form and from electronic medical records. Human papillomavirus genotyping was performed for tissues from confirmed cervical intraepithelial neoplasm grade 2+ diagnoses. A multilevel generalized linear model with a logic function was used to evaluate the association between human papillomavirus vaccination history and the outcome of a cervical intraepithelial neoplasm grade 2+ diagnosis and for human papillomavirus type 16- or 18-positive cervical intraepithelial neoplasm grade 2+ as an alternative outcome, adjusting for screening results and potential confounders. RESULTS: Of 730 women included in the study, 170 had a histologic diagnosis of cervical intraepithelial neoplasm grade 2+ (23.2%). Moreover, 68 cases (40.0%) were histologically human papillomavirus type 16 and/or 18 positive. Of the 730 women, 311 (43%) were vaccinated for the human papillomavirus before colposcopy. Most women (206 [66.2%]) with human papillomavirus vaccination received the vaccine between the ages 18 and 26 years. A history of human papillomavirus vaccination overall, before sexual debut, before the age of 18 years, or with complete dosing was not associated with lower odds of a cervical intraepithelial neoplasm grade 2+ diagnosis (odds ratio, 1.07 [95% confidence interval, 0.70-1.64]; odds ratio, 1.11 [95% confidence interval, 0.55-2.24]; odds ratio, 0.96 [95% confidence interval, 0.49-1.91]; and odds ratio, 0.84 [95% confidence interval, 0.53-1.35], respectively, in reference to no vaccination). Human papillomavirus vaccination history was not significantly associated with the odds of a human papillomavirus type 16- or 18-positive cervical intraepithelial neoplasm grade 2+ diagnosis (P=.45). Notably, 8 cases (4.8% of all cervical intraepithelial neoplasm grade 2+ cases) showed a human papillomavirus type 16 on a cervical intraepithelial neoplasm grade 2+ histologic polymerase chain reaction analysis despite reported or documented human papillomavirus vaccination before sexual debut, including 2 cases who started vaccination before the age of 13 years. CONCLUSION: Our study did not support modifying the colposcopy management guidelines for abnormal screening results for women with human papillomavirus vaccination, especially those vaccinated in the catch-up age range. Our findings on the 8 cases of human papillomavirus 16-positive cervical intraepithelial neoplasm grade 2+ vaccination before sexual debut suggested that lowering the recommended age for human papillomavirus vaccination may have additional benefits for preventing human papillomavirus infection that could occur early in life in some women.


Subject(s)
Papillomavirus Infections/epidemiology , Papillomavirus Vaccines , Uterine Cervical Dysplasia/epidemiology , Uterine Cervical Neoplasms/epidemiology , Vaccination , Adult , California/epidemiology , Cohort Studies , Colposcopy , Female , Humans , Neoplasm Staging , Papillomavirus Infections/pathology , Papillomavirus Infections/surgery , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/surgery , Young Adult , Uterine Cervical Dysplasia/pathology , Uterine Cervical Dysplasia/surgery
7.
Front Immunol ; 11: 561843, 2020.
Article in English | MEDLINE | ID: mdl-33154746

ABSTRACT

Persistent infection with high-risk human papillomavirus (hrHPV) genotypes results in a large number of anogenital and head and neck cancers worldwide. Although prophylactic vaccination coverage has improved, there remains a need to develop methods that inhibit viral transmission toward preventing the spread of HPV-driven disease. Defensins are a class of innate immune effector peptides that function to protect hosts from infection by pathogens such as viruses and bacteria. Previous work utilizing α and ß defensins from humans has demonstrated that the α-defensin HD5 is effective at inhibiting the most common high-risk genotype, HPV16. A third class of defensin that has yet to be explored are θ-defensins: small, 18-amino acid cyclic peptides found in old-world monkeys whose unique structure makes them both highly cationic and resistant to degradation. Here we show that the prototype θ-defensin, rhesus theta defensin 1, inhibits hrHPV infection through a mechanism involving capsid clustering that inhibits virions from binding to cell surface receptor complexes.


Subject(s)
Alphapapillomavirus/physiology , Capsid/metabolism , Defensins/metabolism , Host-Pathogen Interactions , Papillomavirus Infections/metabolism , Papillomavirus Infections/virology , Alphapapillomavirus/drug effects , Alphapapillomavirus/ultrastructure , Capsid Proteins/metabolism , Cell Line , Cell Membrane/metabolism , Cell Membrane/virology , Defensins/pharmacology , Dose-Response Relationship, Drug , Genome, Viral , Genotype , Humans , Immunity, Innate , Papillomavirus Infections/immunology , Peptides, Cyclic/metabolism , Protein Binding , Virion/ultrastructure , alpha-Defensins/metabolism
8.
Clin Cancer Res ; 26(21): 5621-5630, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32816895

ABSTRACT

PURPOSE: A phase I clinical trial (GOG-9929) examined the safety and efficacy of adjuvant immune-modulation therapy with the checkpoint inhibitor ipilimumab [anti-CTL antigen-4 (anti-CTLA-4)] following chemoradiation therapy (CRT) for newly diagnosed node-positive human papillomavirus (HPV)-related cervical cancer. To better understand the mechanism of action and to identify predictive biomarkers, immunologic and viral correlates were assessed before, during, and after treatment. PATIENTS AND METHODS: Twenty-one patients who received CRT and ≥2 doses of ipilimumab and 5 patients who received CRT only were evaluable for translational endpoints. Circulating T-cell subsets were evaluated by multiparameter flow cytometry. Cytokines were evaluated by multiplex ELISA. HPV-specific T cells were evaluated in a subset of patients by IFNγ ELISpot. RESULTS: Expression of the activation markers ICOS and PD-1 significantly increased on T-cell subsets following CRT and were sustained or increased following ipilimumab treatment. Combined CRT/ipilimumab treatment resulted in a significant expansion of both central and effector memory T-cell populations. Genotype-specific E6/E7-specific T-cell responses increased post-CRT in 1 of 8 HPV16+ patients and in 2 of 3 HPV18+ patients. Elevation in levels of tumor-promoting circulating cytokines (TNFα, IL6, IL8) post-CRT was significantly associated with worse progression-free survival. CONCLUSIONS: Our data indicate that CRT alone and combined with ipilimumab immunotherapy show immune-modulating activity in women with locally advanced cervical cancer and may be a promising therapeutic option for the enhancement of antitumor immune cell function after primary CRT for this population at high risk for recurrence and metastasis. Several key immune biomarkers were identified that were associated with clinical response.


Subject(s)
CTLA-4 Antigen/genetics , Ipilimumab/administration & dosage , Neoplasm Recurrence, Local/drug therapy , Uterine Cervical Neoplasms/drug therapy , Adult , Biomarkers, Tumor/genetics , CTLA-4 Antigen/antagonists & inhibitors , Chemoradiotherapy/adverse effects , Chemoradiotherapy/methods , Dose-Response Relationship, Drug , Female , Human papillomavirus 16/genetics , Human papillomavirus 16/pathogenicity , Human papillomavirus 18/genetics , Human papillomavirus 18/pathogenicity , Humans , Immune Checkpoint Inhibitors/administration & dosage , Immune Checkpoint Inhibitors/adverse effects , Interferon-gamma/genetics , Ipilimumab/adverse effects , Middle Aged , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/radiotherapy , Progression-Free Survival , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/genetics , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/radiotherapy
9.
Front Immunol ; 11: 922, 2020.
Article in English | MEDLINE | ID: mdl-32499782

ABSTRACT

Tumor necrosis factor superfamily member 14 (LIGHT) has been in pre-clinical development for over a decade and shows promise as a modality of enhancing treatment approaches in the field of cancer immunotherapy. To date, LIGHT has been used to combat cancer in multiple tumor models where it can be combined with other immunotherapy modalities to clear established solid tumors as well as treat metastatic events. When LIGHT molecules are delivered to or expressed within tumors they cause significant changes in the tumor microenvironment that are primarily driven through vascular normalization and generation of tertiary lymphoid structures. These changes can synergize with methods that induce or support anti-tumor immune responses, such as checkpoint inhibitors and/or tumor vaccines, to greatly improve immunotherapeutic strategies against cancer. While investigators have utilized multiple vectors to LIGHT-up tumor tissues, there are still improvements needed and components to be found within a human tumor microenvironment that may impede translational efforts. This review addresses the current state of this field.


Subject(s)
Immunotherapy/methods , Neoplasms/therapy , Tumor Microenvironment/drug effects , Tumor Necrosis Factor Ligand Superfamily Member 14/therapeutic use , Animals , Clinical Trials as Topic , Combined Modality Therapy , Humans , Immunity , Mice , Neoplasms/immunology , Tumor Microenvironment/immunology , Tumor Necrosis Factor Ligand Superfamily Member 14/immunology
10.
Cells ; 9(4)2020 04 15.
Article in English | MEDLINE | ID: mdl-32326440

ABSTRACT

Langerhans cells (LC) are the resident antigen presenting cells of the mucosal epithelium and play an essential role in initiating immune responses. LC are the only cells in the body to contain Birbeck granules (BG), which are unique cytoplasmic organelles comprised of c-type lectin langerin. Studies of BG have historically focused on morphological characterizations, but BG have also been implicated in viral antigen processing which suggests that they can serve a function in antiviral immunity. This study focused on investigating proteins that could be involved in BG formation to further characterize their structure using transmission electron microscopy (TEM). Here, we report a critical role for the protein annexin A2 (anxA2) in the proper formation of BG structures. When anxA2 expression is downregulated, langerin expression decreases, cytoplasmic BG are nearly ablated, and the presence of malformed BG-like structures increases. Furthermore, in the absence of anxA2, we found langerin was no longer localized to BG or BG-like structures. Taken together, these results indicate an essential role for anxA2 in facilitating the proper formation of BG.


Subject(s)
Annexin A2/metabolism , Cytoplasmic Granules/metabolism , Langerhans Cells/metabolism , Antigens, CD , Cell Line , Cytoplasmic Granules/ultrastructure , Humans , Langerhans Cells/ultrastructure , Lectins, C-Type , Mannose-Binding Lectins , Protein Subunits/metabolism , Protein Transport
11.
Cancer Immunol Res ; 8(7): 856-868, 2020 07.
Article in English | MEDLINE | ID: mdl-32295785

ABSTRACT

Vaccination of patients against neoantigens expressed in concurrent tumors, recurrent tumors, or tumors developing in individuals at risk of cancer is posing major challenges in terms of which antigens to target and is limited to patients expressing neoantigens in their tumors. Here, we describe a vaccination strategy against antigens that were induced in tumor cells by downregulation of the peptide transporter associated with antigen processing (TAP). Vaccination against TAP downregulation-induced antigens was more effective than vaccination against mutation-derived neoantigens, was devoid of measurable toxicity, and inhibited the growth of concurrent and future tumors in models of recurrence and premalignant disease. Human CD8+ T cells stimulated with TAPlow dendritic cells elicited a polyclonal T-cell response that recognized tumor cells with experimentally reduced TAP expression. Vaccination against TAP downregulation-induced antigens overcomes the main limitations of vaccinating against mostly unique tumor-resident neoantigens and could represent a simpler vaccination strategy that will be applicable to most patients with cancer.


Subject(s)
Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/administration & dosage , Neoplasm Recurrence, Local/therapy , Neoplasms/therapy , ATP-Binding Cassette Transporters/antagonists & inhibitors , ATP-Binding Cassette Transporters/immunology , Animals , Antigen Presentation/immunology , Cancer Vaccines/immunology , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasm Recurrence, Local/immunology , Neoplasms/immunology , RNA, Small Interfering/genetics
12.
JAMA Oncol ; 6(1): 92-99, 2020 Jan 01.
Article in English | MEDLINE | ID: mdl-31774464

ABSTRACT

IMPORTANCE: Despite standard chemoradiotherapy (CRT), most women with lymph node (LN)-positive cervical cancer experience disease recurrence. Immunotherapy is being investigated in the up-front treatment setting. OBJECTIVES: To assess the safety of sequential immunotherapy after CRT and to investigate human papillomavirus (HPV) genotype and HLA allele status on survival and programmed cell death 1 (PD-1) expression before and after CRT and sequential immunotherapy. DESIGN, SETTING, AND PARTICIPANTS: This prospective phase 1 trial conducted in 29 Gynecology Oncology Cooperative Group member institutions enrolled participants from December 18, 2012, to August 31, 2016, with a 14.8-month median follow-up and translational end points. Thirty-four women with International Federation of Gynecology and Obstetrics stage IB2 to IVA cervical cancer with positive pelvic LNs, para-aortic LNs, or both were enrolled; 13 did not receive ipilimumab and were excluded from the analysis. Data were analyzed from January 21 to April 4, 2018. INTERVENTIONS: Treatment consisted of 6 weekly doses of cisplatin, 40 mg/m2, concurrent with radiotherapy. After completion of chemotherapy, sequential ipilimumab was given every 21 days for 4 doses. Two dosage levels of ipilimumab, 3 mg/kg and 10 mg/kg, were studied to identify the maximum tolerated dose. MAIN OUTCOMES AND MEASURES: The primary end point was safety, and the secondary end points were overall survival and progression-free survival. Exploratory end points included HPV genotype, HLA allele status, and PD-1 expression measured in peripheral blood. RESULTS: The median age of the 32 participants included in the intent-to-treat analysis was 50 (range, 26-61) years, and 22 patients (69%) were white. Of the 21 patients who received ipilimumab, all had positive pelvic LN, and 6 (29%) had positive para-aortic LNs. All patients completed CRT, and of the 21 patients who received at least 2 cycles of ipilimumab, 18 (86%) completed 4 cycles of ipilimumab, and 3 (14%) completed 2 cycles. The maximum tolerated dose was 10 mg/kg. Two of the 21 patients (9.5%) who received ipilimumab had self-limiting grade 3 toxic effects (lipase increase; dermatitis). The 12-month overall survival was 90%, and progression-free survival was 81%. Human papillomavirus genotype and HLA subtype were not associated with progression-free survival or overall survival. T cells expressing PD-1 increased after CRT, and levels were sustained with ipilimumab. CONCLUSIONS AND RELEVANCE: This study's findings suggest that the use of immunotherapy after CRT for curative-intent treatment of patients with cervical cancer is tolerable and effective. The results indicated that PD-1 was upregulated after CRT and sustained with sequential ipilimumab therapy. These immune findings may help guide future therapies to harness the activated T-cell phenotype in patients with node-positive cervical cancer.


Subject(s)
Uterine Cervical Neoplasms , Adult , Chemoradiotherapy , Female , Humans , Ipilimumab/adverse effects , Middle Aged , Neoplasm Recurrence, Local/pathology , Prospective Studies , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/genetics
13.
Vaccine ; 37(22): 2915-2924, 2019 05 16.
Article in English | MEDLINE | ID: mdl-31010714

ABSTRACT

Persistent human papillomavirus (HPV) infection is causally linked to the development of several human cancers, including cervical, vulvar, vaginal, anal, penile, and oropharyngeal cancers. To address the need for a therapeutic vaccine against HPV-associated diseases, here we test and compare the immunogenicity and therapeutic efficacy of a bacterial exotoxin fusion protein covalently linked to the HPV16 E7 oncoprotein adjuvanted with CpG or GPI-0100 in the C3.43 preclinical HPV16-transformed tumor model. We show that TVGV-1 protein vaccine adjuvanted with either CpG or GPI-0100 adjuvant induces a high frequency of E7-specific CD8+ T cells, and both adjuvants are able to assist the immune response in inducing polyfunctional cytokine-secreting lytic T cells that show therapeutic efficacy against well-established C3.43 tumors. CpG-adjuvanted TVGV-1 resulted in higher frequencies of IFNγ secreting and degranulating E7-specific T cells compared to GPI-0100-adjuvanted TVGV-1, resulting in marginally increased in vivo efficacy. Despite minor differences in immune response outcomes, we consider both CpG ODN and GPI-0100 to be promising vaccine adjuvants to increase the immunogenicity and therapeutic efficacy of the TVGV-1 protein for HPV16-driven cancers.


Subject(s)
Human papillomavirus 16/pathogenicity , Papillomavirus E7 Proteins/metabolism , Papillomavirus Infections/metabolism , Papillomavirus Vaccines/therapeutic use , Saponins/metabolism , Animals , Female , Humans , Immunophenotyping , Mice , Mice, Inbred C57BL , Papillomavirus E7 Proteins/genetics , Papillomavirus E7 Proteins/immunology , Papillomavirus Infections/genetics , Papillomavirus Vaccines/immunology
14.
BMC Cancer ; 18(1): 970, 2018 Oct 11.
Article in English | MEDLINE | ID: mdl-30309325

ABSTRACT

BACKGROUND: Prior small studies have shown increased expression of sperm protein 17 (Sp17) in epithelial ovarian cancer (EOC) tissue and suggest Sp17 as a potential biomarker for EOC. However, how Sp17 expression varies with histology, grade, and stage of EOC and its expression in other ovarian neoplasms has not been defined. It is unknown whether patients with EOC have elevated serum Sp17 levels or if Sp17 expression is associated with survival outcomes. METHODS: The study included 982 patients with benign, borderline, and malignant ovarian neoplasms and normal ovary. There were 878 patients with tissue only, 39 with serum only, and 65 with matching serum and tissue. Immunohistochemical (IHC) staining with anti-Sp17 antibody was performed on tissue specimens and the intensity scored as weak, moderate, or strong. A sandwich enzyme-linked immunosorbent assay (ELISA) was performed to measure Sp17 sera concentrations. RESULTS: Sp17 expression was most commonly seen in serous cystadenomas (83%) and serous borderline tumors (100%). Of the 773 EOC specimens, 223 (30%) expressed Sp17. Grade and histology were significantly associated with Sp17 expression among EOC specimens (p < 0.001) on both univariate and multivariable analysis, with grade 1 serous adenocarcinomas showing the highest expression (51%). Sp17 expression was limited in other benign and non-epithelial malignant neoplasms. Neither Sp17 tissue expression nor serum concentration correlated with survival outcomes. Serum concentrations were higher in patients with Sp17 tissue expression, and the highest concentrations were noted among patients with serous and clear cell adenocarcinomas. CONCLUSIONS: Sp17 is highly expressed in benign, borderline, and low grade malignant serous ovarian neoplasms and can be quantified in serum. Sp17 expression may have diagnostic significance in this subset of patients.


Subject(s)
Antigens, Surface/metabolism , Biomarkers, Tumor/metabolism , Carcinoma, Ovarian Epithelial/metabolism , Carrier Proteins/metabolism , Cystadenoma, Serous/metabolism , Ovarian Neoplasms/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Antigens, Surface/blood , Biomarkers, Tumor/blood , Calmodulin-Binding Proteins , Carcinoma, Ovarian Epithelial/pathology , Carrier Proteins/blood , Cell Line, Tumor , Child , Cystadenoma, Serous/pathology , Female , Humans , Membrane Proteins , Middle Aged , Neoplasm Grading , Ovarian Neoplasms/pathology , Prognosis , Retrospective Studies , Survival Analysis , Up-Regulation , Young Adult
15.
Gynecol Oncol ; 151(1): 102-110, 2018 10.
Article in English | MEDLINE | ID: mdl-30087059

ABSTRACT

OBJECTIVE: We evaluated acceptability of cervico-vaginal self-collection (CVSC) and prevalence of human papillomavirus (HPV) in Human immunodeficiency virus (HIV)-infected and HIV-uninfected women living in the Tapajós region, Amazon, Brazil. METHODS: Cross-sectional study recruited 153 non-indigenous women (HIV-uninfected, n = 112 and HIV-infected, n = 41) who voluntarily sought assistance in health services. Peripheral blood for HIV screening and cervical scraping (CS) for HPV detection were collected. Women who accepted to perform CVSC received instructions and individual collection kits. Risk factors for high-risk HPV genotypes (hrHPV) were identified by uni- and multivariate analyses. RESULTS: The overall acceptability of CVSC was 87%. Only HIV-infected women had cytological abnormalities (12.2%). Prevalence of any HPV and hrHPV infection was 42.9% and 47.9% for HIV-uninfected and 97.6% and 77.5% for HIV-infected women, respectively. There was significant agreement in the detection of HPV (88%, 0.76, 95% confidence interval [CI], 0.65-0.87) and hrHPV (79.7%, 0.56, 95% CI, 0.41-0.71) between self-collected and clinician-collected samples. The most prevalent hrHPV types were HPV16 and HPV18 in HIV-uninfected and HPV16, HPV51 and HPV59 in HIV-infected women. HIV-infected women with hrHPV infection had multiple hrHPV infections (p = 0.005) and lower CD4 count (p = 0.018). Risk factors for hrHPV infection included being HIV-infected and having five or more sexual partners. CONCLUSIONS: CVSC had high acceptability and high prevalence of hrHPV types in women living in the Tapajós region, Amazon, Brazil.


Subject(s)
Early Detection of Cancer/methods , Mass Screening/methods , Papillomaviridae/isolation & purification , Papillomavirus Infections/epidemiology , Patient Acceptance of Health Care/statistics & numerical data , Uterine Cervical Neoplasms/prevention & control , Adolescent , Adult , Aged , Brazil/epidemiology , CD4 Lymphocyte Count , Cervix Uteri/pathology , Cervix Uteri/virology , Cross-Sectional Studies , DNA, Viral/isolation & purification , Early Detection of Cancer/statistics & numerical data , Female , Genotype , HIV Infections/blood , HIV Infections/epidemiology , HIV Infections/pathology , HIV Infections/virology , Humans , Mass Screening/statistics & numerical data , Middle Aged , Papillomaviridae/genetics , Papillomavirus Infections/blood , Papillomavirus Infections/diagnosis , Papillomavirus Infections/pathology , Prevalence , Risk Factors , Specimen Handling/methods , Specimen Handling/statistics & numerical data , Uterine Cervical Neoplasms/diagnosis , Uterine Cervical Neoplasms/virology , Vagina/pathology , Vagina/virology , Young Adult
16.
Sci Rep ; 8(1): 11642, 2018 08 03.
Article in English | MEDLINE | ID: mdl-30076379

ABSTRACT

Human papillomavirus (HPV) entry into epithelial cells is independent of canonical endocytic pathways. Upon interaction with host cells, HPV establishes infection by traversing through an endocytic pathway that is clathrin- and caveolin-independent, but dependent on the annexin A2/S100A10 heterotetramer (A2t). We examined the contribution of monomeric annexin A2 (AnxA2) vs. A2t in HPV infection and endocytosis, and further characterized the role of these molecules in protein trafficking. We specifically show that cell surface A2t is not required for HPV attachment, and in the absence of A2t virion internalization remains clathrin-independent. Without A2t, viral progression from early endosomes to multivesicular endosomes is significantly inhibited, capsid uncoating is dramatically reduced, and lysosomal degradation of HPV is accelerated. Furthermore, we present evidence that AnxA2 forms a complex with CD63, a known mediator of HPV trafficking. Overall, the observed reduction in infection is less significant in the absence of S100A10 alone compared to full A2t, supporting an independent role for monomeric AnxA2. More broadly, we show that successful infection by multiple oncogenic HPV types is dependent on A2t. These findings suggest that A2t is a central mediator of high-risk HPV intracellular trafficking post-entry and pre-viral uncoating.


Subject(s)
Annexin A2/genetics , Papillomaviridae/genetics , Papillomavirus Infections/genetics , S100 Proteins/genetics , Annexin A2/chemistry , Capsid Proteins/chemistry , Capsid Proteins/genetics , Endocytosis/genetics , Epithelial Cells/virology , HeLa Cells , Human papillomavirus 16/genetics , Human papillomavirus 16/pathogenicity , Humans , Lysosomes/genetics , Papillomaviridae/pathogenicity , Papillomavirus Infections/pathology , Papillomavirus Infections/virology , Protein Multimerization/genetics , Protein Transport/genetics , Proteolysis , S100 Proteins/chemistry , Virion/genetics , Virion/pathogenicity
17.
Cancer Immunol Res ; 6(7): 870-880, 2018 07.
Article in English | MEDLINE | ID: mdl-29792298

ABSTRACT

High-risk human papillomavirus-associated cancers express viral oncoproteins (e.g., E6 and E7) that induce and maintain the malignant phenotype. The viral origin of these proteins makes them attractive targets for development of a therapeutic vaccine. Camelid-derived single-domain antibody fragments (nanobodies or VHHs) that recognize cell surface proteins on antigen-presenting cells (APC) can serve as targeted delivery vehicles for antigens attached to them. Such VHHs were shown to induce CD4+ and CD8+ T-cell responses against model antigens conjugated to them via sortase, but antitumor responses had not yet been investigated. Here, we tested the ability of an anti-CD11b VHH (VHHCD11b) to target APCs and serve as the basis for a therapeutic vaccine to induce CD8+ T-cell responses against HPV+ tumors. Mice immunized with VHHCD11b conjugated to an H-2Db-restricted immunodominant E7 epitope (E749-57) had more E7-specific CD8+ T cells compared with those immunized with E749-57 peptide alone. These CD8+ T cells acted prophylactically and conferred protection against a subsequent challenge with HPV E7-expressing tumor cells. In a therapeutic setting, VHHCD11b-E749-57 vaccination resulted in greater numbers of CD8+ tumor-infiltrating lymphocytes compared with mice receiving E749-57 peptide alone in HPV+ tumor-bearing mice, as measured by in vivo noninvasive VHH-based immune-positron emission tomography (immunoPET), which correlated with tumor regression and survival outcome. Together, these results demonstrate that VHHs can serve as a therapeutic cancer vaccine platform for HPV-induced cancers. Cancer Immunol Res; 6(7); 870-80. ©2018 AACR.


Subject(s)
Antigens/immunology , Neoplasms/etiology , Papillomaviridae/immunology , Papillomavirus Infections/complications , Papillomavirus Infections/immunology , Single-Domain Antibodies/immunology , Animals , Biomarkers , Cancer Vaccines/immunology , Disease Models, Animal , Female , Immunization , Immunotherapy , Mice , Mice, Knockout , Models, Biological , Neoplasms/diagnosis , Neoplasms/metabolism , Neoplasms/therapy , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/immunology , Papillomavirus Infections/virology , Positron-Emission Tomography , Protein Binding
18.
PLoS One ; 13(1): e0191311, 2018.
Article in English | MEDLINE | ID: mdl-29324830

ABSTRACT

Nano-Pulse Stimulation (NPS) is a non-thermal pulsed electric field modality that has been shown to have cancer therapeutic effects. Here we applied NPS treatment to the human papillomavirus type 16 (HPV 16)-transformed C3.43 mouse tumor cell model and showed that it is effective at eliminating primary tumors through the induction of immunogenic cell death while subsequently increasing the number of tumor-infiltrating lymphocytes within the tumor microenvironment. In vitro NPS treatment of C3.43 cells resulted in a doubling of activated caspase 3/7 along with the translocation of phosphatidylserine (PS) to the outer leaflet of the plasma membrane, indicating programmed cell death activity. Tumor-bearing mice receiving standard NPS treatment showed an initial decrease in tumor volume followed by clearing of tumors in most mice, and a significant increase in overall survival. Intra-tumor analysis of mice that were unable to clear tumors showed an inverse correlation between the number of tumor infiltrating lymphocytes and the size of the tumor. Approximately half of the mice that cleared established tumors were protected against tumor re-challenge on the opposite flank. Selective depletion of CD8+ T cells eliminated this protection, suggesting that NPS treatment induces an adaptive immune response generating CD8+ T cells that recognize tumor antigen(s) associated with the C3.43 tumor model. This method may be utilized in the future to not only ablate primary tumors, but also to induce an anti-tumor response driven by effector CD8+ T cells capable of protecting individuals from disease recurrence.


Subject(s)
Adaptive Immunity , Cell Death/immunology , Cell Transformation, Viral , Electric Stimulation , Human papillomavirus 16/physiology , Animals , CD8 Antigens/metabolism , Caspase 3/metabolism , Caspase 7/metabolism , Cell Line, Tumor , Enzyme Activation , Humans , Mice , Nanotechnology , Tumor Burden
19.
Virol J ; 13(1): 187, 2016 11 18.
Article in English | MEDLINE | ID: mdl-27863502

ABSTRACT

During sexual transmission of human immunodeficiency virus (HIV), macrophages are initial targets for HIV infection. Secretory leukocyte protease inhibitor (SLPI) has been shown to protect against HIV infection of macrophages through interactions with annexin A2 (A2), which is found on the macrophage cell surface as a heterotetramer (A2t) consisting of A2 and S100A10. Therefore, we investigated potential protein-protein interactions between A2 and HIV-1 gp120 through a series of co-immunoprecipitation assays and a single molecule pulldown (SiMPull) technique. Additionally, inhibitors of A2t (A2ti) that target the interaction between A2 and S100A10 were tested for their ability to impair productive HIV-1 infection of macrophages. Our data suggest that interactions between HIV-1 gp120 and A2 exist, though this interaction may be indirect. Furthermore, an anti-A2 antibody impaired HIV-1 particle production in macrophages in vitro, whereas A2ti did not indicating that annexin A2 may promote HIV-1 infection of macrophages in its monomeric rather than tetrameric form.


Subject(s)
Annexin A2/antagonists & inhibitors , HIV-1/immunology , HIV-1/physiology , Host-Pathogen Interactions , Macrophages/virology , Virus Replication , Annexin A2/metabolism , Antibodies/metabolism , Centrifugation , HIV Envelope Protein gp120/metabolism , Humans , Immunoprecipitation , Protein Binding , Protein Interaction Mapping
20.
AIDS Patient Care STDS ; 30(7): 291-306, 2016 07.
Article in English | MEDLINE | ID: mdl-27410493

ABSTRACT

In the last three decades, extensive research on human immunodeficiency virus (HIV) has highlighted its capability to exploit a variety of strategies to enter and infect immune cells. Although CD4(+) T cells are well known as the major HIV target, with infection occurring through the canonical combination of the cluster of differentiation 4 (CD4) receptor and either the C-C chemokine receptor type 5 (CCR5) or C-X-C chemokine receptor type 4 (CXCR4) coreceptors, HIV has also been found to enter other important immune cell types such as macrophages, dendritic cells, Langerhans cells, B cells, and granulocytes. Interestingly, the expression of distinct cellular cofactors partially regulates the rate in which HIV infects each distinct cell type. Furthermore, HIV can benefit from the acquisition of new proteins incorporated into its envelope during budding events. While several publications have investigated details of how HIV manipulates particular cell types or subtypes, an up-to-date comprehensive review on HIV tropism for different immune cells is lacking. Therefore, this review is meant to focus on the different receptors, coreceptors, and cofactors that HIV exploits to enter particular immune cells. Additionally, prophylactic approaches that have targeted particular molecules associated with HIV entry and infection of different immune cells will be discussed. Unveiling the underlying cellular receptors and cofactors that lead to HIV preference for specific immune cell populations is crucial in identifying novel preventative/therapeutic targets for comprehensive strategies to eliminate viral infection.


Subject(s)
CD4 Antigens/metabolism , HIV Infections/virology , HIV-1/metabolism , Receptors, CCR5/metabolism , Receptors, CXCR4/metabolism , HIV-1/physiology , Humans , Receptors, HIV/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...