Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Clin Cancer Res ; 30(1): 187-197, 2024 01 05.
Article in English | MEDLINE | ID: mdl-37819945

ABSTRACT

PURPOSE: Radiation and platinum-based chemotherapy form the backbone of therapy in human papillomavirus (HPV)-negative head and neck squamous cell carcinoma (HNSCC). We have correlated focal adhesion kinase (FAK/PTK2) expression with radioresistance and worse outcomes in these patients. However, the importance of FAK in driving radioresistance and its effects on chemoresistance in these patients remains unclear. EXPERIMENTAL DESIGN: We performed an in vivo shRNA screen using targetable libraries to identify novel therapeutic sensitizers for radiation and chemotherapy. RESULTS: We identified FAK as an excellent target for both radio- and chemosensitization. Because TP53 is mutated in over 80% of HPV-negative HNSCC, we hypothesized that mutant TP53 may facilitate FAK-mediated therapy resistance. FAK inhibitor increased sensitivity to radiation, increased DNA damage, and repressed homologous recombination and nonhomologous end joining repair in mutant, but not wild-type, TP53 HPV-negative HNSCC cell lines. The mutant TP53 cisplatin-resistant cell line had increased FAK phosphorylation compared with wild-type, and FAK inhibition partially reversed cisplatin resistance. To validate these findings, we utilized an HNSCC cohort to show that FAK copy number and gene expression were associated with worse disease-free survival in mutant TP53, but not wild-type TP53, HPV-negative HNSCC tumors. CONCLUSIONS: FAK may represent a targetable therapeutic sensitizer linked to a known genomic marker of resistance.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Papillomavirus Infections , Humans , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/genetics , Cisplatin/pharmacology , Cisplatin/therapeutic use , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Papillomavirus Infections/complications , Papillomavirus Infections/drug therapy , Papillomavirus Infections/genetics , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/genetics , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor
2.
J Immunother Cancer ; 11(8)2023 08.
Article in English | MEDLINE | ID: mdl-37604640

ABSTRACT

BACKGROUND: TP53, the most mutated gene in solid cancers, has a profound impact on most hallmarks of cancer. Somatic TP53 mutations occur in high frequencies in head and neck cancers, including oral squamous cell carcinoma (OSCC). Our study aims to understand the role of TP53 gain-of-function mutation in modulating the tumor immune microenvironment (TIME) in OSCC. METHODS: Short hairpin RNA knockdown of mutant p53R172H in syngeneic oral tumors demonstrated changes in tumor growth between immunocompetent and immunodeficient mice. HTG EdgeSeq targeted messenger RNA sequencing was used to analyze cytokine and immune cell markers in tumors with inactivated mutant p53R172H. Flow cytometry and multiplex immunofluorescence (mIF) confirmed the role of mutant p53R172H in the TIME. The gene expression of patients with OSCC was analyzed by CIBERSORT and mIF was used to validate the immune landscape at the protein level. RESULTS: Mutant p53R172H contributes to a cytokine transcriptome network that inhibits the infiltration of cytotoxic CD8+ T cells and promotes intratumoral recruitment of regulatory T cells and M2 macrophages. Moreover, p53R172H also regulates the spatial distribution of immunocyte populations, and their distribution between central and peripheral intratumoral locations. Interestingly, p53R172H-mutated tumors are infiltrated with CD8+ and CD4+ T cells expressing programmed cell death protein 1, and these tumors responded to immune checkpoint inhibitor and stimulator of interferon gene 1 agonist therapy. CIBERSORT analysis of human OSCC samples revealed associations between immune cell populations and the TP53R175H mutation, which paralleled the findings from our syngeneic mouse tumor model. CONCLUSIONS: These findings demonstrate that syngeneic tumors bearing the TP53R172H gain-of-function mutation modulate the TIME to evade tumor immunity, leading to tumor progression and decreased survival.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Mouth Neoplasms , Tumor Microenvironment , Tumor Suppressor Protein p53 , Animals , Humans , Mice , Carcinoma, Squamous Cell/genetics , CD8-Positive T-Lymphocytes , Cytokines , Disease Models, Animal , Gain of Function Mutation , Mouth Neoplasms/genetics , Mutation , Squamous Cell Carcinoma of Head and Neck/genetics , Tumor Suppressor Protein p53/genetics
3.
Clin Cancer Res ; 29(7): 1344-1359, 2023 04 03.
Article in English | MEDLINE | ID: mdl-36689560

ABSTRACT

PURPOSE: Cisplatin (CDDP)-based chemotherapy is a first-line treatment for patients with advanced head and neck squamous cell carcinomas (HNSCC), despite a high rate of treatment failures, acquired resistance, and subsequent aggressive behavior. The purpose of this study was to study the mechanism of CDDP resistance and metastasis in HNSCC. We investigated the role of NRF2 pathway activation as a driven event for tumor progression and metastasis of HNSCC. EXPERIMENTAL DESIGN: Human HNSCC cell lines that are highly resistant to CDDP were generated. Clonogenic survival assays and a mouse model of oral cancer were used to examine the impact of NRF2 activation in vitro and in vivo on CDDP sensitivity and development of metastasis. Western blotting, immunostaining, whole-exome sequencing, single-cell transcriptomic and epigenomic profiling platforms were performed to dissect clonal evolution and molecular mechanisms. RESULTS: Implantation of CDDP-resistant HNSCC cells into the tongues of nude mice resulted in a very high rate of distant metastases. The CDDP-resistant cells had significantly higher expression of NRF2 pathway genes in the presence of newly acquired KEAP1 mutations, or via epigenomic activation of target genes. Knockdown of NRF2 or restoration of the wild-type KEAP1 genes resensitized resistant cells to CDDP and decreased distant metastasis (DM). Finally, treatment with inhibitor of glutaminase-1, a NRF2 target gene, alleviated CDDP resistance. CONCLUSIONS: CDDP resistance and development of DM are associated with dysregulated and epigenetically reprogrammed KEAP1-NRF2 signaling pathway. A strategy targeting KEAP1/NRF2 pathway or glutamine metabolism deserves further clinical investigation in patients with CDDP-resistant head and neck tumors.


Subject(s)
Antineoplastic Agents , Head and Neck Neoplasms , NF-E2-Related Factor 2 , Squamous Cell Carcinoma of Head and Neck , Animals , Humans , Mice , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cisplatin/pharmacology , Cisplatin/therapeutic use , Drug Resistance, Neoplasm/genetics , Epigenesis, Genetic , Epigenomics , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/genetics , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , Mice, Nude , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Signal Transduction , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/genetics
4.
Adv Radiat Oncol ; 7(6): 100989, 2022.
Article in English | MEDLINE | ID: mdl-36420184

ABSTRACT

Purpose: An evolutionary action scoring algorithm (EAp53) based on phylogenetic sequence variations stratifies patients with head and neck squamous cell carcinoma (HNSCC) bearing TP53 missense mutations as high-risk, associated with poor outcomes, or low-risk, with similar outcomes as TP53 wild-type, and has been validated as a reliable prognostic marker. We performed this study to further validate prior findings demonstrating that EAp53 is a prognostic marker for patients with locally advanced HNSCC and explored its predictive value for treatment outcomes to adjuvant bio-chemoradiotherapy. Methods and Materials: Eighty-one resection samples from patients treated surgically for stage III or IV human papillomavirus-negative HNSCC with high-risk pathologic features, who received either radiation therapy + cetuximab + cisplatin (cisplatin) or radiation therapy + cetuximab + docetaxel (docetaxel) as adjuvant treatment in a phase 2 study were subjected to TP53 targeted sequencing and EAp53 scoring to correlate with clinical outcomes. Due to the limited sample size, patients were combined into 2 EAp53 groups: (1) wild-type or low-risk; and (2) high-risk or other. Results: At a median follow-up of 9.8 years, there was a significant interaction between EAp53 group and treatment for overall survival (P = .008), disease-free survival (P = .05), and distant metastasis (DM; P = .004). In wild-type or low-risk group, the docetaxel arm showed significantly better overall survival (hazard ratio [HR] 0.11, [0.03-0.36]), disease-free survival (HR 0.24, [0.09-0.61]), and less DM (HR 0.04, [0.01-0.31]) than the cisplatin arm. In high-risk or other group, differences between treatments were not statistically significant. Conclusions: The docetaxel arm was associated with better survival than the cisplatin arm for patients with wild-type or low-risk EAp53. These benefits appear to be largely driven by a reduction in DM.

5.
Commun Biol ; 5(1): 757, 2022 07 28.
Article in English | MEDLINE | ID: mdl-35902768

ABSTRACT

The critical role of the tumor immune microenvironment (TIME) in determining response to immune checkpoint inhibitor (ICI) therapy underscores the importance of understanding cancer cell-intrinsic mechanisms driving immune-excluded ("cold") TIMEs. One such cold tumor is oral cavity squamous cell carcinoma (OSCC), a tobacco-associated cancer with mutations in the TP53 gene which responds poorly to ICI therapy. Because altered TP53 function promotes tumor progression and plays a potential role in TIME modulation, here we developed a syngeneic OSCC models with defined Trp53 (p53) mutations and characterized their TIMEs and degree of ICI responsiveness. We observed that a carcinogen-induced p53 mutation promoted a cold TIME enriched with immunosuppressive M2 macrophages highly resistant to ICI therapy. p53-mutated cold tumors failed to respond to combination ICI treatment; however, the combination of a programmed cell death protein 1 (PD-1) inhibitor and stimulator of interferon genes (STING) agonist restored responsiveness. These syngeneic OSCC models can be used to gain insights into tumor cell-intrinsic drivers of immune resistance and to develop effective immunotherapeutic approaches for OSCC and other ICI-resistant solid tumors.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Mouth Neoplasms , Carcinoma, Squamous Cell/genetics , Genes, p53 , Head and Neck Neoplasms/genetics , Humans , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Mouth Neoplasms/therapy , Squamous Cell Carcinoma of Head and Neck/genetics , Tumor Microenvironment/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
6.
Semin Radiat Oncol ; 31(4): 274-285, 2021 10.
Article in English | MEDLINE | ID: mdl-34455983

ABSTRACT

In multiple anatomic sites, patients with cancers associated with the Human Papillomavirus (HPV) experience better locoregional control and overall survival after radiotherapy and/or chemoradiotherapy than patients with HPV-negative cancers. These improved outcomes suggest that relatively unique biological features in HPV-positive cancers may increase sensitivity to DNA damaging agents as well as an impaired DNA damage response. This review will address potential biological mechanisms driving this increased sensitivity of HPV-positive cancer to radiation and/or chemotherapy. This review will discuss the clinical and preclinical observations that support the intrinsic radiosensitivity and/or chemosensitivity of HPV-positive cancers. Furthermore, this review will highlight the molecular mechanisms for increased radiation sensitivity using the classical "4 Rs" of radiobiology: repair, reassortment, repopulation, and reoxygenation. First, HPV-positive cancers have increased DNA damage due to increased oxidative stress and impaired DNA damage repair due to the altered activity TP53, p16, TIP60, and other repair proteins. Second, irradiated HPV-positive cancer cells display increased G2/M arrest leading to reassortment of cancer cells in more radiosensitive phases of the cell cycle. In addition, HPV-positive cancers have less radioresistant cancer stem cell subpopulations that may limit their repopulation during radiotherapy. Finally, HPV-positive cancers may also have less hypoxic tumor microenvironments that make these cancers more sensitive to radiation than HPV-negative cells. We will also discuss extrinsic immune and microenvironmental factors enriched in HPV-positive cancers that facilities responses to radiation. Therefore, these potential biological mechanisms may underpin the improved clinical outcomes often observed in these virally induced cancers.


Subject(s)
Alphapapillomavirus , Head and Neck Neoplasms , Papillomavirus Infections , Apoptosis , Cell Line, Tumor , G2 Phase Cell Cycle Checkpoints , Humans , Papillomaviridae , Papillomavirus Infections/complications , Radiobiology , Tumor Microenvironment
7.
Cancer Prev Res (Phila) ; 14(8): 767-778, 2021 08.
Article in English | MEDLINE | ID: mdl-34021022

ABSTRACT

Although the principle of systemic treatment to prevent the progression of oral premalignant lesions (OPL) has been demonstrated, there remains a lack of consensus about an optimal approach that balances clinical efficacy with toxicity concerns. Recent advances in cancer therapy using approaches targeting the tumor immune microenvironment (TIME) including immune-checkpoint inhibitors indicate that these agents have significant clinically activity against different types of cancers, including oral cancer, and therefore they may provide an effective oral cancer prevention strategy for patients with OPLs. Our past work showed that systemic delivery of a monoclonal antibody to the programmed death receptor 1 (PD-1) immune checkpoint can inhibit the progression of OPLs to oral cancer in a syngeneic murine oral carcinogenesis model. Here we report a novel approach of local delivery of a PD-1 immune-checkpoint inhibitor loaded using a hydrogel, which significantly reduces the progression of OPLs to carcinomas. In addition, we detected a significant infiltration of regulatory T cells associated with oral lesions with p53 mutation, and a severe loss of expression of STING, which correlated with a decreased infiltration of dendritic cells in the oral lesions. However, a single local dose of PD-1 inhibitor was found to restore stimulator of interferon response cGAMP interactor 1 (STING) and CD11c expression and increase the infiltration of CD8+ T cells into the TIME irrespective of the p53 mutational status. Overall, we provide evidence for the potential clinical value of local delivery of biomaterials loaded with anti-PD-1 antibodies to prevent malignant progression of OPLs. PREVENTION RELEVANCE: Oral cancer is an aggressive disease, with an overall survival rate of 50%. Preinvasive histologic abnormalities such as tongue dysplasia represent an early stage of oral cancer; however, there are no treatments to prevent oral carcinoma progression. Here, we combined biomaterials loaded with an immunotherapeutic agent preventing oral cancer progression.


Subject(s)
Immune Checkpoint Inhibitors/administration & dosage , Mouth Neoplasms/prevention & control , Precancerous Conditions/drug therapy , Programmed Cell Death 1 Receptor/immunology , 4-Nitroquinoline-1-oxide , Animals , Antibodies, Monoclonal/administration & dosage , Carcinogenesis/chemically induced , Carcinogenesis/drug effects , Disease Models, Animal , Drug Delivery Systems , Female , Genes, p53/genetics , Head and Neck Neoplasms/chemically induced , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/prevention & control , Humans , Injections, Intralesional , Male , Mice , Mice, Transgenic , Mouth Neoplasms/pathology , Precancerous Conditions/chemically induced , Precancerous Conditions/genetics , Precancerous Conditions/pathology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Quinolones , Squamous Cell Carcinoma of Head and Neck/chemically induced , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/pathology , Squamous Cell Carcinoma of Head and Neck/prevention & control
8.
Mol Cancer Ther ; 20(7): 1257-1269, 2021 07.
Article in English | MEDLINE | ID: mdl-33947685

ABSTRACT

Despite advances in surgery, chemotherapy, and radiation, there are limited treatment options for advanced head and neck squamous cell carcinoma (HNSCC) and survival remains very poor. Therefore, effective therapies are desperately needed. Recently, selective exploitation of DNA damage and replication stress responses has become a novel approach for cancer treatment. Wee1 kinase and Rad51 recombinase are two proteins involved in regulating replication stress and homologous recombination repair in cancer cells. In this study, we investigated the combined effect of Rad51 inhibitor (B02) and Wee1 inhibitor (AZD1775) in vitro and in vivo in various HNSCC cell lines. Clonogenic survival assays demonstrated that B02 synergized with AZD1775 in vitro in all HNSCC cell lines tested. The synergy between these drugs was associated with forced CDK1 activation and reduced Chk1 phosphorylation leading to induction of excessive DNA damage and replication stress, culminating in aberrant mitosis and apoptosis. Our results showed that elevated Rad51 mRNA expression correlated with worse survival in HNSCC patients with HPV-positive tumors. The combination of B02 and AZD1775 significantly inhibited tumor growth in vivo in mice bearing HPV-positive HNSCC tumors as compared to HPV-negative HNSCC. This differential sensitivity appears to be linked to HPV-positive tumors having more in vivo endogenous replication stress owing to transformation by E6 and E7 oncogenes. Furthermore, addition of B02 radiosensitized the HPV-negative HNSCC tumors in vitro and in vivo In conclusion, our data implicate that a novel rational combination with Rad51 and Wee1 inhibitors holds promise as synthetic lethal therapy, particularly in high-risk HPV-positive HNSCC.


Subject(s)
Apoptosis/drug effects , Cell Cycle Proteins/antagonists & inhibitors , DNA Damage/drug effects , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Rad51 Recombinase/antagonists & inhibitors , Animals , Cell Cycle/drug effects , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cells, Cultured , Computational Biology/methods , DNA Repair/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Synergism , Gene Expression Profiling , Homologous Recombination , Humans , Mice , Pyrazoles/pharmacology , Pyrimidinones/pharmacology , Radiation-Sensitizing Agents/pharmacology , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/etiology , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology , Xenograft Model Antitumor Assays
9.
Nature ; 578(7795): 449-454, 2020 02.
Article in English | MEDLINE | ID: mdl-32051587

ABSTRACT

The solid tumour microenvironment includes nerve fibres that arise from the peripheral nervous system1,2. Recent work indicates that newly formed adrenergic nerve fibres promote tumour growth, but the origin of these nerves and the mechanism of their inception are unknown1,3. Here, by comparing the transcriptomes of cancer-associated trigeminal sensory neurons with those of endogenous neurons in mouse models of oral cancer, we identified an adrenergic differentiation signature. We show that loss of TP53 leads to adrenergic transdifferentiation of tumour-associated sensory nerves through loss of the microRNA miR-34a. Tumour growth was inhibited by sensory denervation or pharmacological blockade of adrenergic receptors, but not by chemical sympathectomy of pre-existing adrenergic nerves. A retrospective analysis of samples from oral cancer revealed that p53 status was associated with nerve density, which was in turn associated with poor clinical outcomes. This crosstalk between cancer cells and neurons represents mechanism by which tumour-associated neurons are reprogrammed towards an adrenergic phenotype that can stimulate tumour progression, and is a potential target for anticancer therapy.


Subject(s)
Adrenergic Neurons/pathology , Cell Transdifferentiation , Cellular Reprogramming , Mouth Neoplasms/pathology , Sensory Receptor Cells/pathology , Tumor Suppressor Protein p53/deficiency , Adrenergic Antagonists/pharmacology , Adrenergic Antagonists/therapeutic use , Animals , Cell Division , Disease Models, Animal , Disease Progression , Female , Humans , Male , Mice , Mice, Inbred BALB C , MicroRNAs/genetics , Mouth Neoplasms/drug therapy , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , Nerve Fibers/pathology , Neurites/pathology , Receptors, Adrenergic/metabolism , Retrospective Studies , Tumor Microenvironment , Tumor Suppressor Protein p53/genetics , Xenograft Model Antitumor Assays
10.
J Natl Cancer Inst ; 112(3): 266-277, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31124563

ABSTRACT

BACKGROUND: Alterations in the epidermal growth factor receptor and PI3K pathways in head and neck squamous cell carcinomas (HNSCCs) are frequent events that promote tumor progression. Ectopic expression of the epidermal growth factor receptor-targeting microRNA (miR), miR-27a* (miR-27a-5p), inhibits tumor growth. We sought to identify mechanisms mediating repression of miR-27a* in HNSCC, which have not been previously identified. METHODS: We quantified miR-27a* in 47 oral cavity squamous cell carcinoma patient samples along with analysis of miR-27a* in 73 oropharyngeal and 66 human papillomavirus-positive (HPV+) samples from The Cancer Genome Atlas. In vivo and in vitro TP53 models engineered to express mutant TP53, along with promoter analysis using chromatin immunoprecipitation and luciferase assays, were used to identify the role of TP53 and TP63 in miR-27a* transcription. An HNSCC cell line engineered to conditionally express miR-27a* was used in vitro to determine effects of miR-27a* on target genes and tumor cells. RESULTS: miR-27a* expression was repressed in 47 oral cavity tumor samples vs matched normal tissue (mean log2 difference = -0.023, 95% confidence interval = -0.044 to -0.002; two-sided paired t test, P = .03), and low miR-27a* levels were associated with poor survival in HPV+ and oropharyngeal HNSCC samples. Binding of ΔNp63α to the promoter led to an upregulation of miR-27a*. In vitro and in vivo findings showed that mutant TP53 represses the miR-27a* promoter, downregulating miR-27a* levels. ΔNp63α and nucleoporin 62, a protein involved in ΔNP63α transport, were validated as novel targets of miR-27a*. CONCLUSION: Our results characterize a negative feedback loop between TP63 and miR-27a*. Genetic alterations in TP53, a frequent event in HNSCC, disrupt this regulatory loop by repressing miR-27a* expression, promoting tumor survival.


Subject(s)
Head and Neck Neoplasms/genetics , MicroRNAs/genetics , Squamous Cell Carcinoma of Head and Neck/genetics , Transcription Factors/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/genetics , Case-Control Studies , Chromatin Immunoprecipitation , Feedback, Physiological , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , MicroRNAs/metabolism , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Mutation , Neoplasm Staging , Promoter Regions, Genetic , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology , Survival Rate , Transcription Factors/metabolism , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism
11.
Clin Cancer Res ; 25(18): 5650-5662, 2019 09 15.
Article in English | MEDLINE | ID: mdl-31308060

ABSTRACT

PURPOSE: TP53 mutations are highly prevalent in head and neck squamous cell carcinoma (HNSCC) and associated with increased resistance to conventional treatment primarily consisting of chemotherapy and radiation. Restoration of wild-type p53 function in TP53-mutant cancer cells represents an attractive therapeutic approach and has been explored in recent years. In this study, the efficacy of a putative p53 reactivator called COTI-2 was evaluated in HNSCC cell lines with different TP53 status.Experimental Design: Clonogenic survival assays and an orthotopic mouse model of oral cancer were used to examine in vitro and in vivo sensitivity of HNSCC cell lines with either wild-type, null, or mutant TP53 to COTI-2 alone, and in combination with cisplatin and/or radiation. Western blotting, cell cycle, live-cell imaging, RNA sequencing, reverse-phase protein array, chromatin immunoprecipitation, and apoptosis analyses were performed to dissect molecular mechanisms. RESULTS: COTI-2 decreased clonogenic survival of HNSCC cells and potentiated response to cisplatin and/or radiation in vitro and in vivo irrespective of TP53 status. Mechanistically, COTI-2 normalized wild-type p53 target gene expression and restored DNA-binding properties to the p53-mutant protein in HNSCC. In addition, COTI-2 induced DNA damage and replication stress responses leading to apoptosis and/or senescence. Furthermore, COTI-2 lead to activation of AMPK and inhibition of the mTOR pathways in vitro in HNSCC cells. CONCLUSIONS: COTI-2 inhibits tumor growth in vitro and in vivo in HNSCC likely through p53-dependent and p53-independent mechanisms. Combination of COTI-2 with cisplatin or radiation may be highly relevant in treating patients with HNSCC harboring TP53 mutations.


Subject(s)
Antineoplastic Agents/pharmacology , Signal Transduction/drug effects , Squamous Cell Carcinoma of Head and Neck/metabolism , Thiosemicarbazones/pharmacology , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cellular Senescence/drug effects , Cellular Senescence/genetics , Cisplatin/pharmacology , DNA Damage , DNA Replication , Disease Models, Animal , Gene Expression Profiling , Humans , Mice , Protein Binding , Signal Transduction/radiation effects , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/pathology , Stress, Physiological , Transcription, Genetic , Tumor Suppressor Protein p53/genetics , Xenograft Model Antitumor Assays
12.
Oral Oncol ; 87: 49-57, 2018 12.
Article in English | MEDLINE | ID: mdl-30527243

ABSTRACT

OBJECTIVES: Currently there are no standard biomarkers of head and neck squamous cell carcinoma (HNSCC) response to therapy. This is, due to a lack of adequate predictive tumor models. To this end, we established cancer organoid lines from individual patient's tumors, and characterized their growth characteristics and response to different drug treatments with the objective of using these models for prediction of treatment response. MATERIALS AND METHODS: Forty-three patients' samples were processed to establish organoids. To analyze the character of these organoids, immunohistochemistry, Western blotting, drug sensitivity assays, clonogenic survival assays, and animal experiments were performed. The HPV status and TP53 mutational status were also confirmed in these lines. RESULTS: HNSCC organoids were successfully established with success rate of 30.2%. Corresponding two-dimensional cell lines were established from HNSCC organoids at higher success rate (53.8%). These organoids showed similar histological features and stem cell, epithelial and mesenchymal marker expression to the original tumors, thus recapitulating many of the characteristics of the original tumor cells. The cisplatin and docetaxel IC50 were determined for HNSCC organoids and the corresponding 2D cell lines using drug sensitivity and clonogenic survival assays. Responses to drug treatment in vivo were found to be similar to the IC50 calculated from organoids by drug sensitivity assays in vitro. CONCLUSION: We established novel in vitro HNSCC cancer organoid lines retaining many properties of the original tumors from they were derived. These organoids can predict in vivo drug sensitivity and may represent useful tools to develop precision treatments for HNSCC.


Subject(s)
Antineoplastic Agents/pharmacology , Head and Neck Neoplasms/drug therapy , Organoids/drug effects , Primary Cell Culture/methods , Squamous Cell Carcinoma of Head and Neck/drug therapy , Animals , Antineoplastic Agents/therapeutic use , Cisplatin/pharmacology , Cisplatin/therapeutic use , Docetaxel/pharmacology , Docetaxel/therapeutic use , Drug Screening Assays, Antitumor/methods , Female , Head and Neck Neoplasms/pathology , Humans , Inhibitory Concentration 50 , Male , Mice , Organoids/pathology , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology , Squamous Cell Carcinoma of Head and Neck/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
13.
Oncogene ; 37(10): 1279-1292, 2018 03.
Article in English | MEDLINE | ID: mdl-29269868

ABSTRACT

Many mutant p53 proteins exert oncogenic gain-of-function (GOF) properties that promote cancer cell invasive growth and metastasis, yet the mechanisms mediating these functions still largely remain elusive. We show here that overexpression of the GOF mutant p53 G245D and other GOF p53 mutants enhances the invasive cell growth of p53-deficient head and neck squamous cell carcinoma (HNSCC) UM-SCC-1 cells both in in vitro three-dimensional culture and in an in vivo orthotopic nude mouse model of HNSCC through a novel transcription-independent mechanism. We demonstrate that the expression of the oncogenic forkhead transcription factor FOXM1 is upregulated by GOF mutant p53s. Moreover, we show that overexpression of GOF mutant p53 G245D decreases the AMP-activated protein kinase (AMPK)-mediated phosphorylation of FOXO3a, a tumor suppressive forkhead transcription factor, leading to its cytoplasmic accumulation. This downregulation of FOXO3a's activity, in turn, leads to de-repression of FOXM1 expression. Importantly, we show that either overexpression of FOXO3a or downregulation of FOXM1 impairs both GOF mutant p53-mediated cell invasion in vitro and pulmonary metastases of UM-SCC-1 cells in vivo. Finally, not only do oral cancer patients with p53 mutations exhibit higher levels of FOXM1 expression than patients with wild-type p53, but also HNSCC patients with TP53 mutations and high levels of FOXM1 expression have the poorest survival outcomes. Given our prior demonstration that GOF mutant p53s inhibit AMPK, our current study, establishes and demonstrates a novel transcription-independent GOF mutant p53-AMPK-FOXO3a-FOXM1 signaling cascade that plays an important role in mediating mutant p53s' gain-of-function activities in HNSCCs.


Subject(s)
Carcinogenesis/genetics , Forkhead Box Protein M1/genetics , Forkhead Box Protein O3/genetics , Gain of Function Mutation , Squamous Cell Carcinoma of Head and Neck/genetics , Tumor Suppressor Protein p53/genetics , Amino Acid Substitution/genetics , Animals , Cell Line, Tumor , Gain of Function Mutation/physiology , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mice, Nude , Signal Transduction/genetics , Squamous Cell Carcinoma of Head and Neck/pathology , Transcriptional Activation/genetics
14.
Clin Cancer Res ; 23(21): 6541-6554, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28790110

ABSTRACT

Purpose: The cure rate for patients with advanced head and neck squamous cell carcinoma (HNSCC) remains poor due to resistance to standard therapy primarily consisting of chemoradiation. As mutation of TP53 in HNSCC occurs in 60% to 80% of non-HPV-associated cases and is in turn associated with resistance to these treatments, more effective therapies are needed. In this study, we evaluated the efficacy of a regimen combining vorinostat and AZD1775 in HNSCC cells with a variety of p53 mutations.Experimental Design: Clonogenic survival assays and an orthotopic mouse model of oral cancer were used to examine the in vitro and in vivo sensitivity of high-risk mutant p53 HNSCC cell lines to vorinostat in combination with AZD1775. Cell cycle, replication stress, homologous recombination (HR), live cell imaging, RNA sequencing, and apoptosis analyses were performed to dissect molecular mechanisms.Results: We found that vorinostat synergizes with AZD1775 in vitro to inhibit growth of HNSCC cells harboring high-risk mutp53. These drugs interact synergistically to induce DNA damage, replication stress associated with impaired Rad51-mediated HR through activation of CDK1, and inhibition of Chk1 phosphorylation, culminating in an early apoptotic cell death during the S-phase of the cell cycle. The combination of vorinostat and AZD1775 inhibits tumor growth and angiogenesis in vivo in an orthotopic mouse model of oral cancer and prolongs animal survival.Conclusions: Vorinostat synergizes with AZD1775 in HNSCC cells with mutant p53 in vitro and in vivo A strategy combining HDAC and WEE1 inhibition deserves further clinical investigation in patients with advanced HNSCC. Clin Cancer Res; 23(21); 6541-54. ©2017 AACR.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Head and Neck Neoplasms/drug therapy , Hydroxamic Acids/administration & dosage , Pyrazoles/administration & dosage , Pyrimidines/administration & dosage , Tumor Suppressor Protein p53/genetics , Animals , Apoptosis/drug effects , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Cycle Proteins/antagonists & inhibitors , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Damage/drug effects , DNA Replication/drug effects , Drug Synergism , Female , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Histone Deacetylase Inhibitors/administration & dosage , Humans , Hydroxamic Acids/adverse effects , Mice , Mutation , Nuclear Proteins/antagonists & inhibitors , Phosphorylation/drug effects , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrazoles/adverse effects , Pyrimidines/adverse effects , Pyrimidinones , Risk Factors , S Phase/drug effects , Squamous Cell Carcinoma of Head and Neck , Vorinostat
15.
Clin Cancer Res ; 21(21): 4831-44, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26124202

ABSTRACT

PURPOSE: Although the majority of patients with HPV(+) oropharyngeal cancers have a favorable prognosis, there are some patients with tumors that are resistant to aggressive chemoradiotherapy with unusual patterns of locoregional and systemic recurrences. Therefore, more effective therapies are needed. In this study, we investigated the chemosensitizing efficacy of the selective Wee-1 kinase inhibitor, AZD-1775, in HPV(+) head and neck squamous cell carcinoma (HNSCC). EXPERIMENTAL DESIGN: Clonogenic survival assays and an orthotopic mouse model of HPV(+) oral cancer were used to examine the in vitro and in vivo sensitivity of HPV(+) HNSCC cell lines to AZD-1775 in combination with cisplatin, respectively. Cell-cycle analysis, DNA damage (γH2AX), homologous recombination (HR), and apoptosis were examined to dissect molecular mechanisms. RESULTS: We found that AZD-1775 displays single-agent activity and enhances the response of HPV(+) HNSCC cells to cisplatin both in vitro and in vivo. The sensitivity of the HPV(+) HNSCC cells to AZD-1775 alone or in combination with cisplatin was associated with G2 checkpoint abrogation, persistent DNA damage, and apoptosis induction. This finding of AZD-1775 increasing the sensitivity of HPV(+) HNSCC cells to cisplatin through apoptosis was not seen previously in the HPV(-) HNSCC cancer cells and is accompanied by a decreased expression of the antiapoptotic proteins, MCl-1and XIAP, which appear to be cleaved following AZD-1775 treatment. CONCLUSIONS: AZD-1775 selectively sensitizes HPV(+) HNSCC cells and orthotopic oral xenografts to cisplatin through apoptosis and support the clinical investigation of AZD-1775 in combination with cisplatin particularly in patients with advanced and recurrent metastatic HPV(+) HNSCC tumors.


Subject(s)
Apoptosis/drug effects , Carcinoma, Squamous Cell/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Head and Neck Neoplasms/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Nuclear Proteins/antagonists & inhibitors , Papillomavirus Infections/complications , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , X-Linked Inhibitor of Apoptosis Protein/metabolism , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/etiology , Caspases/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cisplatin/pharmacology , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Drug Synergism , G2 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Genes, p53 , Head and Neck Neoplasms/etiology , Humans , Inhibitory Concentration 50 , Male , Mice , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Papillomavirus Infections/virology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Pyrimidinones , Squamous Cell Carcinoma of Head and Neck , Tumor Burden/drug effects , X-Linked Inhibitor of Apoptosis Protein/genetics , Xenograft Model Antitumor Assays
16.
Cancer Res ; 75(7): 1205-15, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25691460

ABSTRACT

TP53 is the most frequently altered gene in head and neck squamous cell carcinoma (HNSCC), with mutations occurring in over two thirds of cases; however, the predictive response of these mutations to cisplatin-based therapy remains elusive. In the current study, we evaluate the ability of the Evolutionary Action score of TP53-coding variants (EAp53) to predict the impact of TP53 mutations on response to chemotherapy. The EAp53 approach clearly identifies a subset of high-risk TP53 mutations associated with decreased sensitivity to cisplatin both in vitro and in vivo in preclinical models of HNSCC. Furthermore, EAp53 can predict response to treatment and, more importantly, a survival benefit for a subset of head and neck cancer patients treated with platinum-based therapy. Prospective evaluation of this novel scoring system should enable more precise treatment selection for patients with HNSCC.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/genetics , Cisplatin/pharmacology , Tongue Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Animals , Carcinoma, Squamous Cell/drug therapy , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , Gene Expression , Humans , Male , Mice, Nude , Middle Aged , Mutation , Tongue Neoplasms/drug therapy , Xenograft Model Antitumor Assays
17.
Cancer Res ; 75(7): 1527-36, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25634208

ABSTRACT

TP53 is the most frequently altered gene in head and neck squamous cell carcinoma, with mutations occurring in over two-thirds of cases, but the prognostic significance of these mutations remains elusive. In the current study, we evaluated a novel computational approach termed evolutionary action (EAp53) to stratify patients with tumors harboring TP53 mutations as high or low risk, and validated this system in both in vivo and in vitro models. Patients with high-risk TP53 mutations had the poorest survival outcomes and the shortest time to the development of distant metastases. Tumor cells expressing high-risk TP53 mutations were more invasive and tumorigenic and they exhibited a higher incidence of lung metastases. We also documented an association between the presence of high-risk mutations and decreased expression of TP53 target genes, highlighting key cellular pathways that are likely to be dysregulated by this subset of p53 mutations that confer particularly aggressive tumor behavior. Overall, our work validated EAp53 as a novel computational tool that may be useful in clinical prognosis of tumors harboring p53 mutations.


Subject(s)
Carcinoma, Squamous Cell/genetics , Head and Neck Neoplasms/genetics , Lung Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/secondary , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Genetic Association Studies , Genomics , Head and Neck Neoplasms/mortality , Head and Neck Neoplasms/pathology , Humans , Kaplan-Meier Estimate , Lung Neoplasms/mortality , Lung Neoplasms/secondary , Mutation , Neoplasm Invasiveness , Proportional Hazards Models , Transcriptome
18.
Mol Cancer Ther ; 14(2): 608-19, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25504633

ABSTRACT

Although cisplatin has played a role in "standard-of-care" multimodality therapy for patients with advanced squamous cell carcinoma of the head and neck (HNSCC), the rate of treatment failure remains particularly high for patients receiving cisplatin whose tumors have mutations in the TP53 gene. We found that cisplatin treatment of HNSCC cells with mutant TP53 leads to arrest of cells in the G2 phase of the cell cycle, leading us to hypothesize that the wee-1 kinase inhibitor MK-1775 would abrogate the cisplatin-induced G2 block and thereby sensitize isogenic HNSCC cells with mutant TP53 or lacking p53 expression to cisplatin. We tested this hypothesis using clonogenic survival assays, flow cytometry, and in vivo tumor growth delay experiments with an orthotopic nude mouse model of oral tongue cancer. We also used a novel TP53 mutation classification scheme to identify which TP53 mutations are associated with limited tumor responses to cisplatin treatment. Clonogenic survival analyses indicate that nanomolar concentration of MK-1775 sensitizes HNSCC cells with high-risk mutant p53 to cisplatin. Consistent with its ability to chemosensitize, MK-1775 abrogated the cisplatin-induced G2 block in p53-defective cells leading to mitotic arrest associated with a senescence-like phenotype. Furthermore, MK-1775 enhanced the efficacy of cisplatin in vivo in tumors harboring TP53 mutations. These results indicate that HNSCC cells expressing high-risk p53 mutations are significantly sensitized to cisplatin therapy by the selective wee-1 kinase inhibitor, supporting the clinical evaluation of MK-1775 in combination with cisplatin for the treatment of patients with TP53 mutant HNSCC.


Subject(s)
Cell Cycle Checkpoints/drug effects , Cell Cycle Proteins/antagonists & inhibitors , Cisplatin/pharmacology , Drug Resistance, Neoplasm/drug effects , Head and Neck Neoplasms/genetics , Mutation/genetics , Nuclear Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics , Animals , CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cellular Senescence/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Damage , Drug Synergism , Humans , Mice, Nude , Mitosis/drug effects , Nuclear Proteins/metabolism , Phenotype , Phosphorylation/drug effects , Protein-Tyrosine Kinases/metabolism , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Pyrimidinones , Reactive Oxygen Species/metabolism
19.
Mol Cell ; 54(6): 960-974, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24857548

ABSTRACT

Many mutant p53 proteins (mutp53s) exert oncogenic gain-of-function (GOF) properties, but the mechanisms mediating these functions remain poorly defined. We show here that GOF mutp53s inhibit AMP-activated protein kinase (AMPK) signaling in head and neck cancer cells. Conversely, downregulation of GOF mutp53s enhances AMPK activation under energy stress, decreasing the activity of the anabolic factors acetyl-CoA carboxylase and ribosomal protein S6 and inhibiting aerobic glycolytic potential and invasive cell growth. Under conditions of energy stress, GOF mutp53s, but not wild-type p53, preferentially bind to the AMPKα subunit and inhibit AMPK activation. Given the importance of AMPK as an energy sensor and tumor suppressor that inhibits anabolic metabolism, our findings reveal that direct inhibition of AMPK activation is an important mechanism through which mutp53s can gain oncogenic function.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Carcinoma, Squamous Cell/genetics , Energy Metabolism/genetics , Head and Neck Neoplasms/genetics , Tumor Suppressor Protein p53/metabolism , AMP-Activated Protein Kinases/antagonists & inhibitors , Acetyl-CoA Carboxylase/metabolism , Animals , Antimetabolites, Antineoplastic/pharmacology , Cell Movement/genetics , Cell Proliferation , Enzyme Activation/genetics , Fluorouracil/pharmacology , Humans , Mice , Mice, Nude , Neoplasm Invasiveness/genetics , Neoplasm Transplantation , Protein Binding/genetics , RNA Interference , RNA, Small Interfering , Ribosomal Protein S6/metabolism , Signal Transduction/genetics , Spheroids, Cellular/cytology , Squamous Cell Carcinoma of Head and Neck , Transplantation, Heterologous , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
20.
Mol Cancer Ther ; 12(9): 1860-73, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23839309

ABSTRACT

Despite the use of multimodality therapy using cisplatin to treat patients with advanced stage squamous cell carcinoma of the head and neck (HNSCC), there is an unacceptably high rate of treatment failure. TP53 is the most commonly mutated gene in HNSCC, and the impact of p53 mutation on response to cisplatin treatment is poorly understood. Here, we show unambiguously that wild-type TP53 (wtp53) is associated with sensitivity of HNSCC cells to cisplatin treatment, whereas mutation or loss of TP53 is associated with cisplatin resistance. We also show that senescence is the major cellular response to cisplatin in wtp53 HNSCC cells and that cisplatin resistance in p53-null or -mutant TP53 cells is due to their lack of senescence. Given the dependence on checkpoint kinase (Chk)1/2 kinases to mediate the DNA damage response in p53-deficient cells, there is potential to exploit this to therapeutic advantage through targeted inhibition of the Chk1/2 kinases. Treatment of p53-deficient HNSCC cells with the Chk inhibitor AZD7762 sensitizes them to cisplatin through induction of mitotic cell death. This is the first report showing the ability of a Chk kinase inhibitor to sensitize TP53-deficient HNSCC to cisplatin in a synthetic lethal manner, which has significance given the frequency of TP53 mutations in this disease and because cisplatin has become part of standard therapy for aggressive HNSCC tumors. These preclinical data provide evidence that a personalized approach to the treatment of HNSCC based on Chk inhibition in p53-mutant tumors may be feasible.


Subject(s)
Antineoplastic Agents/pharmacology , Checkpoint Kinase 2/metabolism , Cisplatin/pharmacology , Head and Neck Neoplasms/pathology , Protein Kinases/metabolism , Thiophenes/pharmacology , Tumor Suppressor Protein p53/genetics , Urea/analogs & derivatives , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cellular Senescence/drug effects , Checkpoint Kinase 1 , Cisplatin/therapeutic use , DNA Damage , Drug Resistance, Neoplasm/genetics , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Humans , Mitosis , Molecular Targeted Therapy , Mutation , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Signal Transduction , Thiophenes/therapeutic use , Tumor Suppressor Protein p53/metabolism , Urea/pharmacology , Urea/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL