Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Vaccines (Basel) ; 12(4)2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38675812

ABSTRACT

Human papillomavirus (HPV) remains a global health concern because it contributes to the initiation of various HPV-associated cancers such as anal, cervical, oropharyngeal, penile, vaginal, and vulvar cancer. In HPV-associated cancers, oncogenesis begins with an HPV infection, which is linked to the activation of the Janus protein tyrosine kinase (JAK)/STAT signaling pathway. Various STAT signaling pathways, such as STAT3 activation, have been well documented for their tumorigenic role, yet the role of STAT1 in tumor formation remains unclear. In the current study, STAT1-/- mice were used to investigate the role of STAT1 in the tumorigenesis of a spontaneous HPV E6/E7-expressing oral tumor model. Subsequently, our candidate HPV DNA vaccine CRT/E7 was administered to determine whether the STAT1-/- host preserves a therapeutic-responsive tumor microenvironment. The results indicated that STAT1-/- induces robust tumorigenesis, yet a controlled tumor response was attained upon CRT/E7 vaccination. Characterizing this treatment effect, immunological analysis found a higher percentage of circulating CD4+ and CD8+ T cells and tumor-specific cytotoxic T cells. In addition, a reduction in exhaustive lymphocyte activity was observed. Further analysis of a whole-cell tumor challenge affirmed these findings, as spontaneous tumor growth was more rapid in STAT1-/- mice. In conclusion, STAT1 deletion accelerates tumorigenesis, but STAT1-/- mice maintains immunocompetency in CRT/E7 treatments.

2.
Sci Rep ; 13(1): 18476, 2023 10 28.
Article in English | MEDLINE | ID: mdl-37898690

ABSTRACT

The innate immune stimulator of interferon genes (STING) pathway is known to activate type I interferons (IFN-I) and participate in generating antitumor immunity. We previously produced hDT806, a recombinant diphtheria immunotoxin, and demonstrated its efficacy against head and neck squamous cell carcinoma (HNSCC). However, it's unknown whether the tumor-intrinsic STING plays a role in the anti-HNSCC effects of hDT806. In this study, we investigated the innate immune modulation of hDT806 on HNSCC. hDT806 significantly upregulated the level of STING and the ratio of p-TBK1/TBK1 in the HNSCC cells. Moreover, intratumoral hDT806 treatment increased the expression of STING-IFN-I signaling proteins including IFNA1, IFNB, CXCL10 and MX1, a marker of IFN-I receptor activity, in the HNSCC xenografts. Overexpression of STING mimicked the hDT806-induced upregulation of the STING-IFN-I signaling and induced apoptosis in the HNSCC cells. In the mouse xenograft models of HNSCC with STING overexpression, we observed a significant suppression of tumor growth and reduced tumor weight with increased apoptosis compared to their control xenograft counterparts without STING overexpression. Collectively, our data revealed that hDT806 may act as a stimulator of tumor-intrinsic STING-IFN-I signaling to inhibit tumor growth in HNSCC.


Subject(s)
Head and Neck Neoplasms , Immunotoxins , Interferon Type I , Humans , Animals , Mice , Squamous Cell Carcinoma of Head and Neck , Signal Transduction , Interferon Type I/genetics , Head and Neck Neoplasms/drug therapy
3.
Biology (Basel) ; 11(4)2022 Mar 22.
Article in English | MEDLINE | ID: mdl-35453686

ABSTRACT

Over 90% of head and neck squamous cell carcinoma (HNSCC) overexpresses the epidermal growth factor receptor (EGFR). However, the EGFR-targeted monotherapy response rate only achieves 10-30% in HNSCC. Recombinant immunotoxin (RIT) often consists of an antibody targeting a tumor antigen and a toxin (e.g., diphtheria toxin [DT]) that kills cancer cells. We produced a humanized RIT, designated as hDT806, targeting overexpressed EGFR and investigated its effects in HNSCC. Distinct from the EGFR-targeted tyrosine kinase inhibitor erlotinib or antibody cetuximab, hDT806 effectively suppressed cell proliferation in the four HNSCC lines tested (JHU-011, -013, -022, and -029). In JHU-029 mouse xenograft models, hDT806 substantially reduced tumor growth. hDT806 decreased EGFR protein levels and disrupted the EGFR signaling downstream effectors, including MAPK/ERK1/2 and AKT, while increased proapoptotic proteins, such as p53, caspase-9, caspase-3, and the cleaved PAPR. The hDT806-induced apoptosis of HNSCC cells was corroborated by flow cytometric analysis. Furthermore, hDT806 resulted in a drastic inhibition in RNA polymerase II carboxy-terminal domain phosphorylation critical for transcription and a significant increase in the γH2A.X level, a DNA damage marker. Thus, the direct disruption of EGFR signaling, transcription inhibition, DNA damage, as well as apoptosis induced by hDT806 may contribute to its antitumor efficacy in HNSCC.

4.
Cell Rep ; 37(3): 109838, 2021 10 19.
Article in English | MEDLINE | ID: mdl-34648735

ABSTRACT

As severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spreads, variants with enhanced virulence and transmissibility have emerged. Although in vitro systems allow rapid characterization, they do not fully recapitulate the dynamic interaction of virions and neutralizing antibodies in the airway. Here, we demonstrate that the N501Y variant permits respiratory infection in unmodified mice. We utilize N501Y to survey in vivo pseudovirus infection dynamics and susceptibility to reinfection with the L452R (Los Angeles), K417N + E484K (South Africa), and L452R + K417N + E484Q (India) variants. Human coronavirus disease 2019 (COVID-19)+ or vaccinated antibody isotypes, titers, variant receptor binding domain (RBD) binding, and neutralization potential are studied, revealing numerous significant correlations. Immune escape of the K417N + E484K variant is observed because infection can be appreciated in the nasopharynx, but not lungs, of mice transferred with low-antibody-tier plasma. Conversely, near-complete protection is observed in animals receiving high-antibody-tier plasma, a phenomenon that can only be appreciated in vivo.


Subject(s)
Antibodies, Viral/immunology , COVID-19/immunology , COVID-19/therapy , SARS-CoV-2/genetics , SARS-CoV-2/immunology , Animals , Antibodies, Neutralizing/immunology , Cell Line , Cricetinae , Genetic Variation , HEK293 Cells , Humans , Immune System , Immunization, Passive/methods , In Vitro Techniques , Mice , Mutation , Nasopharynx/virology , Protein Binding , Recombinant Proteins/metabolism , Spike Glycoprotein, Coronavirus/genetics , COVID-19 Serotherapy
5.
Vaccines (Basel) ; 9(4)2021 Apr 01.
Article in English | MEDLINE | ID: mdl-33916109

ABSTRACT

The flavivirus Zika (ZIKV) has emerged as a global threat, making the development of a ZIKV vaccine a priority. While live-attenuated vaccines are known to induce long-term immunity but reduced safety, inactivated vaccines exhibit a weaker immune response as a trade-off for increased safety margins. To overcome the trade-off between immunogenicity and safety, the concept of a third-generation flavivirus vaccine based on single-cycle flaviviruses has been developed. These third-generation flavivirus vaccines have demonstrated extreme potency with a high level of safety in animal models. However, the production of these single-cycle, encapsidation-defective flaviviruses requires a complicated virion packaging system. Here, we investigated a new single-cycle flavivirus vaccine, a vertebrate-specific replication-defective ZIKV (VSRD-ZIKV), in a mouse model. VSRD-ZIKV replicates to high titers in insect cells but can only initiate a single-round infection in vertebrate cells. During a single round of infection, VSRD-ZIKV can express all the authentic viral antigens in vertebrate hosts. VSRD-ZIKV immunization elicited a robust cellular and humoral immune response that protected against a lethal ZIKV challenge in AG129 mice. Additionally, VSRD-ZIKV-immunized pregnant mice were protected against vertically transferring a lethal ZIKV infection to their offspring. Immunized male mice were protected and prevented viral accumulation in the testes after being challenged with lethal ZIKV. Overall, our results indicate that VSRD-ZIKV induces a potent protective immunity against ZIKV in a mouse model and represents a promising approach to develop novel single-cycle arbovirus vaccines.

6.
Virology ; 552: 73-82, 2021 01 02.
Article in English | MEDLINE | ID: mdl-33075709

ABSTRACT

Zika virus (ZIKV) is a mosquito-borne flavivirus that replicates in both vertebrate and insect cells, whereas insect-specific flaviviruses (ISF) replicate only in insect cells. We sought to convert ZIKV, from a dual-tropic flavivirus, into an insect-specific virus for the eventual development of a safe ZIKV vaccine. Reverse genetics was used to introduce specific mutations into the furin cleavage motif within the ZIKV pre-membrane protein (prM). Mutant clones were selected, which replicated well in C6/36 insect cells but exhibited reduced replication in non-human primate (Vero) cells. Further characterization of the furin cleavage site mutants indicated they replicated poorly in both human (HeLa, U251), and baby hamster kidney (BHK-21) cells. One clone with the induced mutation in the prM protein and at positions 291and 452 within the NS3 protein was totally and stably replication-defective in vertebrate cells (VSRD-ZIKV). Preliminary studies in ZIKV sensitive, immunodeficient mice demonstrated that VSRD-ZIKV-infected mice survived and were virus-negative. Our study indicates that a reverse genetic approach targeting the furin cleavage site in prM can be used to select an insect-specific ZIKV with the potential utility as a vaccine strain.


Subject(s)
Insecta/virology , Membrane Proteins/metabolism , Vertebrates/virology , Viral Nonstructural Proteins/metabolism , Virus Replication , Zika Virus Infection/virology , Zika Virus/physiology , Animals , Cell Line , Chlorocebus aethiops , Cricetinae , Furin/metabolism , HeLa Cells , Host Specificity , Humans , Isoquinolines , Mice , Mutation , Reverse Genetics/methods , Vero Cells , Vertebrates/immunology , Viral Proteins/metabolism , Zika Virus Infection/immunology
7.
Int J Gynecol Pathol ; 40(2): 134-140, 2021 Mar 01.
Article in English | MEDLINE | ID: mdl-32265359

ABSTRACT

A 44-yr-old woman with menorrhagia and uterine fibroids underwent total laparoscopic hysterectomy, revealing several submucosal, intramural, and subserosal tan-white nodules in the uterus. Microscopic examination revealed tumors displaying 3 distinct morphologies: 1 tumor with features of conventional leiomyoma; 1 tumor with increased cellularity, staghorn/hemangiopericytoma-like vasculature, and occasional atypical cells with prominent red nucleoli and some perinucleolar halos suggesting a fumarate hydratase (FH)-deficient atypical leiomyoma; and 1 tumor with an admixture of epithelioid and spindled cells with the former arranged around blood vessels suggesting a perivascular epithelioid cell tumor (PEComa). Immunohistochemical studies confirmed these diagnoses by demonstrating loss of FH expression in the atypical leiomyoma and diffuse expression of HMB45 and cathepsin K in the tumor with epithelioid features. Sanger sequencing analysis revealed that the FH-deficient atypical leiomyoma harbored a c.181A>G (p.Lys61Glu) mutation in exon 2 of the FH gene. As this mutation was not present in either the other tumors or peripheral blood, the mutation is somatic and hereditary leiomyomatosis and renal cell cancer syndrome is excluded. This case highlights the importance of thorough examination of uterine mesenchymal tumors with atypical and epithelioid features so that tumors with some potential for recurrence (PEComas) and those that might indicate a hereditary cancer syndrome (FH-deficient atypical leiomyoma) are identified and can trigger appropriate clinical investigation and follow-up.


Subject(s)
Fumarate Hydratase/genetics , Leiomyoma/diagnosis , Neoplastic Syndromes, Hereditary/diagnosis , Perivascular Epithelioid Cell Neoplasms/diagnosis , Uterine Neoplasms/diagnosis , Adult , Amino Acid Substitution , Diagnosis, Differential , Exons/genetics , Female , Fumarate Hydratase/metabolism , Humans , Hysterectomy , Immunohistochemistry , Leiomyoma/complications , Leiomyoma/genetics , Leiomyoma/pathology , Neoplastic Syndromes, Hereditary/complications , Neoplastic Syndromes, Hereditary/genetics , Neoplastic Syndromes, Hereditary/pathology , Perivascular Epithelioid Cell Neoplasms/complications , Perivascular Epithelioid Cell Neoplasms/genetics , Perivascular Epithelioid Cell Neoplasms/pathology , Uterine Neoplasms/complications , Uterine Neoplasms/genetics , Uterine Neoplasms/pathology , Uterus/pathology , Uterus/surgery
8.
Histopathology ; 76(3): 354-365, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31564060

ABSTRACT

AIMS: Hereditary leiomyomatosis and renal cell cancer (HLRCC) syndrome is caused by germline mutations in the Fumarate hydratase (FH) gene. In young women, the syndrome often presents with symptomatic uterine leiomyomas, leading to myomectomy or hysterectomy. In this study, we aimed to investigate the incidence and mutational profiles of FH-negative leiomyomas from young patients, thus allowing for early identification and triage of syndromic patients for surveillance. METHODS AND RESULTS: We evaluated 153 cases of uterine leiomyomas from women aged up to 30 years for loss of FH expression by tissue microarray (TMA)-based immunohistochemical staining. Mutational analysis of tumours with loss of FH was carried out by polymerase chain reaction (PCR) amplification of 10 exons within the FH gene and subsequent Sanger sequencing. The status of promoter methylation was assessed by bisulphite sequencing. Loss of FH protein expression was detected in seven (4.6%) of 153 tested uterine leiomyomas from young patients. All FH-negative leiomyomas displayed staghorn vasculature and fibrillary/neurophil-like cytoplasm. We found that six (86%) of seven FH-negative tumours detected by immunohistochemistry harboured FH mutations, 50% of which contained germline mutations. In particular, the germline mutational rate in FH gene was 2.0% (three of 153 cases). Bisulphite sequencing analysis failed to detect promoter methylation in any of the seven tumours. CONCLUSION: Our study showed a relatively high rate of FH germline mutation in FH-negative uterine leiomyomas from patients aged up to 30 years. While genetic mutations confer protein expression loss, epigenetic regulation of the FH gene appears to be unrelated to this phenotype.


Subject(s)
Fumarate Hydratase/genetics , Leiomyoma/genetics , Leiomyomatosis/genetics , Neoplastic Syndromes, Hereditary/genetics , Skin Neoplasms/genetics , Uterine Neoplasms/genetics , Adolescent , Adult , DNA Mutational Analysis , Female , Fumarate Hydratase/metabolism , Germ-Line Mutation , Humans , Immunohistochemistry , Leiomyoma/enzymology , Leiomyoma/pathology , Leiomyomatosis/enzymology , Leiomyomatosis/pathology , Mutation , Neoplastic Syndromes, Hereditary/enzymology , Neoplastic Syndromes, Hereditary/pathology , Prevalence , Retrospective Studies , Skin Neoplasms/enzymology , Skin Neoplasms/pathology , Tissue Array Analysis , Uterine Neoplasms/enzymology , Uterine Neoplasms/pathology , Young Adult
9.
Hum Pathol ; 92: 67-80, 2019 10.
Article in English | MEDLINE | ID: mdl-31437519

ABSTRACT

Using a comprehensive next-generation sequencing pipeline (143 genes), Oncomine Comprehensive v.2, we analyzed genetic alterations on a set of vulvar squamous cell carcinomas (SCCs) with emphasis on the primary and metastatic samples from the same patient, to identify amenable therapeutic targets. Clinicopathologic features were reported and genomic DNA was extracted from 42 paraffin-embedded tumor tissues of 32 cases. PD-L1 expression was evaluated in 20 tumor tissues (10 cases with paired primary and metastatic tumors). Fifteen (88%) of 17 successfully analyzed HPV-unrelated SCCs harbored TP53 mutations. 2 different TP53 mutations had been detected in the same tumor in 4 of 15 cases. Other recurrent genetic alterations in this group of tumors included CDKN2a mutations (41%), HRAS mutations (12%), NOTCH1 mutations (12%) and BIRC3 (11q22.1-22.2) amplification (12%). Six HPV-related tumors harbored PIK3CA, BAP1, PTEN, KDR, CTNNB1, and BRCA2 mutations, of which, one case also contained TP53 mutation. Six cases showed identical mutations in paired primary site and distant metastatic location and four cases displayed different mutational profiles. PD-L1 expression was seen in 6 of 10 primary tumors and all 6 paired cases showed discordant PD-L1 expression in the primary and metastatic sites. Our results further confirmed the genetic alterations that are amenable to targeted therapy, offering the potential for individualized management strategies for the treatment of these aggressive tumors with different etiology. Discordant PD-L1 expression in the primary and metastatic vulvar SCCs highlights the importance of evaluation of PD-L1 expression in different locations to avoid false negative information provided for immunotherapy.


Subject(s)
B7-H1 Antigen/genetics , Carcinoma, Squamous Cell/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Tumor Suppressor Protein p53/genetics , Vulvar Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Baculoviral IAP Repeat-Containing 3 Protein/genetics , Biomarkers, Tumor/genetics , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/secondary , Female , High-Throughput Nucleotide Sequencing , Humans , Middle Aged , Mutation , Proto-Oncogene Proteins p21(ras)/genetics , Receptor, Notch1/genetics , Vulvar Neoplasms/pathology , Vulvar Neoplasms/secondary
10.
Am J Surg Pathol ; 42(6): 750-760, 2018 06.
Article in English | MEDLINE | ID: mdl-29505425

ABSTRACT

Small cell neuroendocrine carcinoma (SCNEC) of the uterine cervix is a rare but extremely aggressive tumor. While high-risk human papillomavirus (HPV) is involved at an early stage of oncogenesis in many tumors, additional driving events have been postulated to facilitate the progression of SCNECs. Identification of oncogenic drivers could guide targeted therapy of this neoplasm. Clinicopathologic features of 10 cervical SCNECs are reported. Analyses included immunohistochemical evaluation of p16, p53, synaptophysin, and chromogranin expression; in situ hybridizations and polymerase chain reaction for high-risk HPV and/or HPV 18; and next-generation sequencing based on a 637-gene panel. The patients ranged in age from 28 to 68 years (mean, 45.6 y; median, 40.5 y). All tumors had diffuse p16 and synaptophysin expression. All but 1 tumor was positive for chromogranin (extent of staining ranged from focal to diffuse). HPV 18 was detected in 6 tumors and HPV 35 in 1 tumor. At least 1 driver mutation was detected in 8 tumors. Four cases harbored TP53 somatic mutations, 3 of which correlated with an aberrant p53 staining pattern. Four PIK3CA mutations (p.G106A, p.N345T, p.E545K, and p.E545D) were detected in 3 tumors, 2 of which also harbored TP53 mutations. Oncogenic driver mutations involving KRAS, Erbb2, c-Myc, NOTCH1, BCL6, or NCOA3 were detected in 4 tumors. Mutations in caretaker tumor suppressors PTEN, RB1, BRCA1, BRCA2, and ARID1B were also identified in 4 tumors that commonly coharbored activating oncogenic mutations. Targeted next-generation gene sequencing identified genetic alterations involving the MAPK, PI3K/AKT/mTOR, and TP53/BRCA pathways in SCNECs. The presence of genetic alterations that are amenable to targeted therapy in SCNECs offers the potential for individualized management strategies for treatment of this aggressive tumor.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Neuroendocrine/genetics , Carcinoma, Small Cell/genetics , DNA Mutational Analysis/methods , High-Throughput Nucleotide Sequencing , Mutation , Uterine Cervical Neoplasms/genetics , Adult , Aged , Biomarkers, Tumor/analysis , Carcinoma, Neuroendocrine/chemistry , Carcinoma, Neuroendocrine/pathology , Carcinoma, Neuroendocrine/virology , Carcinoma, Small Cell/chemistry , Carcinoma, Small Cell/pathology , Carcinoma, Small Cell/virology , Female , Genetic Predisposition to Disease , Human Papillomavirus DNA Tests , Humans , Immunohistochemistry , Middle Aged , Phenotype , Predictive Value of Tests , United States , Uterine Cervical Neoplasms/chemistry , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/virology
11.
Laryngoscope ; 127(12): 2713-2720, 2017 12.
Article in English | MEDLINE | ID: mdl-28714529

ABSTRACT

OBJECTIVES/HYPOTHESIS: Recurrent respiratory papillomatosis (RRP) is a benign disease caused by human papillomavirus (HPV) types 6 and 11. Although a prophylactic vaccine against RRP is available, a therapeutic vaccine is needed to treat those already infected. The objective of our study was to design and test a DNA vaccine targeting HPV11 proteins. STUDY DESIGN: Preclinical scientific investigation. METHODS: A DNA vaccine encoding the HPV11 E6 and E7 genes linked to calreticulin (CRT) was generated. Immunologic response to the HPV11 CRT/E6E7 vaccine was measured by vaccinating C57BL/6 mice via electroporation and measuring CD8 + T cell responses from harvested splenocytes. A tumor cell line containing HPV11-E6E7 was created, and the ability of novel DNA vaccine to control tumor growth was measured in vivo. RESULTS: Our vaccine generated a significant and specific CD8 + T-cell response against the HPV11-E6aa41-70 peptide. The CD8 + T-cell responses did not recognize E7 epitopes, indicating E6 immunodominance. CD8 + responses were augmented in the CRT-linked vaccine compared to a control non-CRT vaccine. The HPV11 CRT/E6E7 vaccine was used to treat mice inoculated with a HPV11 E6E7 expressing tumor cell line after temporary CD3 depletion to facilitate tumor growth. Vaccinated mice had a significantly lower tumor growth rate (P = .029) and smaller tumor volumes compared to control mice, indicating an augmented immunologic response in vaccinated mice. CONCLUSIONS: A DNA vaccine targeting HPV11 E6E7 generates a specific HPV11 CD-8 + T-cell response capable of reducing the growth of HPV11-expressing tumors. DNA vaccines are a promising immunologic strategy for treating RRP. LEVEL OF EVIDENCE: NA. Laryngoscope, 127:2713-2720, 2017.


Subject(s)
Human papillomavirus 11/immunology , Oncogene Proteins, Viral/immunology , Papillomavirus Infections/prevention & control , Papillomavirus Vaccines/immunology , Respiratory Tract Infections/prevention & control , Vaccines, DNA/immunology , Animals , Cell Line, Tumor , Female , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Papillomavirus Infections/virology , Respiratory Tract Infections/virology
12.
Cancer Immunol Immunother ; 65(3): 261-71, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26759151

ABSTRACT

Recurrent respiratory papillomatosis is caused by human papillomavirus (HPV) infection, most commonly types 6 (HPV-6) and 11 (HPV-11). Due to failed host immune responses, HPV is unable to be cleared from the host, resulting in recurrent growth of HPV-related lesions that can obstruct the lumen of the airway within the upper aerodigestive tract. In our murine model, the HPV-6b and HPV-11 E7 antigens are not innately immunogenic. In order to enhance the host immune responses against the HPV E7 antigen, we linked calreticulin (CRT) to HPV-6b E7 and found that vaccinating C57BL/6 mice with the HPV-6b CRT/E7 DNA vaccine is able to induce a CD8+ T cell response that recognizes an H-2D(b)-restricted E7aa21-29 epitope. Additionally, vaccination of HLA-A*0201 transgenic mice with HPV-6b CRT/E7 DNA generated a CD8+ T cell response against the E7aa82-90 epitope that was not observed in the wild-type C57BL/6 mice, indicating this T cell response is restricted to HLA-A*0201. In vivo cytotoxic T cell killing assays demonstrated that the vaccine-induced CD8+ T cells are able to efficiently kill target cells. Interestingly, the H-2D(b)-restricted E7aa21-29 sequence and the HLA-A*0201-restricted E7aa82-90 sequence are conserved between HPV-6b and HPV-11 and may represent shared immunogenic epitopes. The identification of the HPV-6b/HPV-11 CD8+ T cell epitopes facilitates the evaluation of various immunomodulatory strategies in preclinical models. More importantly, the identified HLA-A*0201-restricted T cell epitope may serve as a peptide vaccination strategy, as well as facilitate the monitoring of vaccine-induced HPV-specific immunologic responses in future human clinical trials.


Subject(s)
Epitopes, T-Lymphocyte/immunology , HLA-A2 Antigen/immunology , Histocompatibility Antigen H-2D/immunology , Oncogene Proteins, Viral/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Calreticulin/pharmacology , Cell Line, Tumor , Female , Humans , Mice , Mice, Inbred C57BL , Papillomavirus Vaccines/immunology , Vaccination , Vaccines, DNA/immunology
13.
J Biomed Sci ; 22: 7, 2015 Jan 16.
Article in English | MEDLINE | ID: mdl-25591912

ABSTRACT

BACKGROUND: Bortezomib, a proteasome inhibitor and suberoylanilide hydroxamic acid (SAHA, also known as Vorinostat), a histone deacetylase inhibitor, have been recognized as potent chemotherapeutic drugs. Bortezomib and SAHA are FDA-approved for the treatment of cutaneous T cell lymphoma and multiple myeloma/mantle cell lymphoma, respectively. Furthermore, the combination of the bortezomib and SAHA has been tested in a variety of preclinical models and in clinical trials and may be ideal for the treatment of cancer. However, it remains unclear how this treatment strategy affects the host immune response against tumors. RESULTS: Here, we used a well-defined E6/E7-expressing tumor model to examine how the immune system can be motivated to act against tumor cells expressing tumor antigens. We demonstrate that the combination of bortezomib and SAHA elicits potent antitumor effects in TC-1 tumor-bearing mice. Additionally, we are the first to show that treatment with bortezomib and SAHA leads to tumor-specific immunity by rendering tumor cells more susceptible to killing by antigen-specific CD8+ T cells than treatment with either drug alone. CONCLUSIONS: The current study serves an important foundation for the future clinical application of both drugs for the treatment of cervical cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Bortezomib/pharmacology , CD8-Positive T-Lymphocytes/drug effects , Hydroxamic Acids/pharmacology , Immunity, Innate/drug effects , Uterine Cervical Neoplasms/drug therapy , Animals , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Female , Histone Deacetylase Inhibitors/pharmacology , Humans , Mice , Mice, Inbred C57BL , Proteasome Inhibitors/pharmacology , Uterine Cervical Neoplasms/immunology , Vorinostat
14.
J Biomed Opt ; 20(5): 051008, 2015 May.
Article in English | MEDLINE | ID: mdl-25423046

ABSTRACT

There is an urgent need for rapid methods to develop vaccines in response to emerging viral pathogens. Whole inactivated virus (WIV) vaccines represent an ideal strategy for this purpose; however, a universal method for producing safe and immunogenic inactivated vaccines is lacking. Conventional pathogen inactivation methods such as formalin, heat, ultraviolet light, and gamma rays cause structural alterations in vaccines that lead to reduced neutralizing antibody specificity, and in some cases, disastrous T helper type 2-mediated immune pathology. We have evaluated the potential of a visible ultrashort pulsed (USP) laser method to generate safe and immunogenic WIV vaccines without adjuvants. Specifically, we demonstrate that vaccination of mice with laser-inactivated H1N1 influenza virus at about a 10-fold lower dose than that required using conventional formalin-inactivated influenza vaccines results in protection against lethal H1N1 challenge in mice. The virus, inactivated by the USP laser irradiation, has been shown to retain its surface protein structure through hemagglutination assay. Unlike conventional inactivation methods, laser treatment did not generate carbonyl groups in protein, thereby reducing the risk of adverse vaccine-elicited T helper type 2 responses. Therefore, USP laser treatment is an attractive potential strategy to generate WIV vaccines with greater potency and safety than vaccines produced by current inactivation techniques.


Subject(s)
Influenza Vaccines/chemistry , Orthomyxoviridae Infections/prevention & control , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , CD8-Positive T-Lymphocytes/cytology , Cell Line , Dogs , Female , Hemagglutination Tests , Humans , Influenza A Virus, H1N1 Subtype , Influenza, Human/prevention & control , Lasers , Mice , Mice, Inbred BALB C , Microscopy, Electron, Transmission , Neutralization Tests , Vaccination , Vaccines, Inactivated/chemistry
15.
PLoS One ; 9(7): e103562, 2014.
Article in English | MEDLINE | ID: mdl-25072795

ABSTRACT

Vitamin E has been shown to have strong anticarcinogenic properties, including antioxidant characteristics, making it an ideal candidate for use in combination with immunotherapies that modify the tumor microenvironment. The tumor microenvironment contains immunosuppressive components, which can be diminished, and immunogenic components, which can be augmented by immunotherapies in order to generate a productive immune response. In the current study, we employ the α-tocopherol succinate isomer of vitamin E to reduce immunosuppression by myeloid derived suppressor cells (MDSCs) as well as adoptive transfer of antigen-specific CD8+ T cells to generate potent antitumor effects against the HPV16 E7-expressing TC-1 tumor model. We show that vitamin E alone induces necrosis of TC-1 cells and elicits antitumor effects in TC-1 tumor-bearing mice. We further demonstrate that vitamin E reverses the suppression of T cell activation by MDSCs and that this effect is mediated in part by a nitric oxide-dependent mechanism. Additionally, treatment with vitamin E reduces the percentage of MDSCs in tumor loci, and induces a higher percentage of T cells, following T cell adoptive transfer. Finally, we demonstrate that treatment with vitamin E followed by E7-specific T cell adoptive transfer experience elicits potent antitumor effects in tumor-bearing mice. Our data provide additional evidence that vitamin E has anticancer properties and that it has promise for use as an adjuvant in combination with a variety of cancer therapies.


Subject(s)
CD8-Positive T-Lymphocytes/drug effects , Myeloid Cells/drug effects , alpha-Tocopherol/pharmacology , Animals , CD11b Antigen/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Female , Immunotherapy, Adoptive , Mice , Mice, Inbred C57BL , Myeloid Cells/cytology , Myeloid Cells/metabolism , Necrosis , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , Nitric Oxide/chemistry , Nitric Oxide/toxicity , Papillomavirus E7 Proteins/immunology , Papillomavirus E7 Proteins/metabolism , alpha-Tocopherol/therapeutic use
16.
Sci Transl Med ; 5(172): 172ra20, 2013 Feb 13.
Article in English | MEDLINE | ID: mdl-23408053

ABSTRACT

Although many human cancers are located in mucosal sites, most cancer vaccines are tested against subcutaneous tumors in preclinical models. We therefore wondered whether mucosa-specific homing instructions to the immune system might influence mucosal tumor outgrowth. We showed that the growth of orthotopic head and neck or lung cancers was inhibited when a cancer vaccine was delivered by the intranasal mucosal route but not the intramuscular route. This antitumor effect was dependent on CD8⁺ T cells. Indeed, only intranasal vaccination elicited mucosal-specific CD8⁺ T cells expressing the mucosal integrin CD49a. Blockade of CD49a decreased intratumoral CD8⁺ T cell infiltration and the efficacy of cancer vaccine on mucosal tumor. We then showed that after intranasal vaccination, dendritic cells from lung parenchyma, but not those from spleen, induced the expression of CD49a on cocultured specific CD8⁺ T cells. Tumor-infiltrating lymphocytes from human mucosal lung cancer also expressed CD49a, which supports the relevance and possible extrapolation of these results in humans. We thus identified a link between the route of vaccination and the induction of a mucosal homing program on induced CD8⁺ T cells that controlled their trafficking. Immunization route directly affected the efficacy of the cancer vaccine to control mucosal tumors.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/administration & dosage , Chemotaxis, Leukocyte , Head and Neck Neoplasms/therapy , Immunity, Mucosal , Lung Neoplasms/therapy , Nasal Mucosa/immunology , Papillomavirus Vaccines/administration & dosage , Adjuvants, Immunologic/administration & dosage , Administration, Intranasal , Animals , Antigens, CD/metabolism , Cancer Vaccines/immunology , Cell Proliferation , Cells, Cultured , Dendritic Cells/immunology , Female , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/pathology , Humans , Injections, Intramuscular , Integrin alpha Chains/metabolism , Integrin alpha1/metabolism , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lymph Nodes/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mice, Inbred C57BL , Papillomavirus Vaccines/immunology , Shiga Toxins/administration & dosage , Spleen/immunology , Tumor Burden
17.
J Biomed Sci ; 19: 62, 2012 Jul 06.
Article in English | MEDLINE | ID: mdl-22768792

ABSTRACT

The threat of emerging pathogens and microbial drug resistance has spurred tremendous efforts to develop new and more effective antimicrobial strategies. Recently, a novel ultrashort pulsed (USP) laser technology has been developed that enables efficient and chemical-free inactivation of a wide spectrum of viral and bacterial pathogens. Such a technology circumvents the need to introduce potentially toxic chemicals and could permit safe and environmentally friendly pathogen reduction, with a multitude of possible applications including the sterilization of pharmaceuticals and blood products, and the generation of attenuated or inactivated vaccines.


Subject(s)
Bacteria/radiation effects , Lasers , Viruses/radiation effects , Bacteria/pathogenicity , Humans , Sterilization/methods , Vaccines, Inactivated/radiation effects , Viruses/pathogenicity
18.
Clin Cancer Res ; 11(13): 4717-23, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-16000566

ABSTRACT

PURPOSE: Persistent infection with oncogenic human papillomaviruses (HPV) plays a central etiologic role in the development of squamous carcinomas of the cervix and their precursor lesions, cervical intraepithelial neoplasias (CIN). We carried out a prospective observational cohort study evaluating known, quantifiable prognostic variables of clinical behavior in women with high-grade cervical lesions. EXPERIMENTAL DESIGN: Our study cohort included healthy women with high-grade cervical lesions (CIN2/3) with residual visible lesions after colposcopically directed biopsy. We prospectively followed 100 women over 15 weeks before standard resection. HPV typing was done using PCR and a reverse line blot detection method. RESULTS: The rate of spontaneous histologic regression, defined as (CIN1 or less at resection) was 28%. The overall rate of HPV infection was 100%. HPV16 was identified in 68% of the lesions. Women with HPV16 only were significantly less likely to regress, compared with women with HPV types other than HPV16 (odds ratio, 0.342; 95% confidence interval, 0.117-0.997; P = 0.049). In the cohort with HPV16 only, patients who had an HLA*A201 allele had similar outcomes to those who did not carry A201. However, among patients with HPV types other than HPV16, the HLA*A201 allele interaction was significant; patients with HLA*A201 were the least likely to resolve. CONCLUSIONS: CIN2/3 lesions associated with HPV16 alone are significantly less likely to resolve spontaneously than those caused by other types. Interactions among HPV type, HLA type, and regression rate support a role for HLA-restricted HPV-specific immune responses in determining disease outcome.


Subject(s)
Papillomavirus Infections/pathology , Uterine Cervical Dysplasia/pathology , Adolescent , Adult , Aged , Alleles , Cell Line, Tumor , Cohort Studies , DNA, Viral/analysis , DNA, Viral/genetics , Female , Genotype , HLA Antigens/genetics , HLA-A2 Antigen/genetics , Humans , K562 Cells , Middle Aged , Papillomaviridae/genetics , Papillomaviridae/growth & development , Papillomavirus Infections/genetics , Papillomavirus Infections/virology , Prospective Studies , Remission, Spontaneous , Uterine Cervical Dysplasia/genetics , Uterine Cervical Dysplasia/virology
19.
Cancer ; 98(9 Suppl): 2064-9, 2003 Nov 01.
Article in English | MEDLINE | ID: mdl-14603543

ABSTRACT

Revelation of the connection between the human papillomavirus (HPV) and cervical neoplasia and invasive cervical cancer is prompting new investigations to expand that understanding and promote vaccines, gene therapy, and other interventions. At the Second International Conference on Cervical Cancer (Houston, TX, April 11-14, 2002), laboratory and clinical researchers reported advances in new studies meant to increase understanding of the natural history of HPV and cervical intraepithelial neoplasia, to evaluate new cervical cancer screening techniques, and to promote new therapies. Using K14-HPV type 16 transgenic mice, researchers are investigating the effects of estrogen on cervical cancer carcinogenesis, and results are lending support to epidemiological theories showing a difference in HPV infection rates and the development of cervical lesions in women using oral contraceptives. Other work involves investigating genes that are up-regulated by HPV infection and the role of the p53 homologue, p63, in cervical neoplasia evolution. Telomerase also is under investigation as a biomarker in high-risk populations. Gene therapy that replaced p53 in cervical cancer cell lines in vitro and a nude mouse model inhibited cell and tumor growth, confirming previous findings in squamous epithelial carcinomas of the head and neck. Furthermore, research in intracellular targeting of antigens to subcellular locations shows promise for treating cervical cancer preclinically. Identification of molecular changes in cervical cancer and knowledge about the importance of HPV infection in cervical cancer can lead to new therapies to treat existing cervical cancer and, in the long term, prevent the disease.


Subject(s)
Uterine Cervical Neoplasms , Animals , Contraceptives, Oral/adverse effects , Female , Humans , Mice , Mice, Nude , Mice, Transgenic , Papillomaviridae , Papillomavirus Infections/genetics , Tumor Suppressor Protein p53 , Uterine Cervical Neoplasms/therapy
SELECTION OF CITATIONS
SEARCH DETAIL