Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Sci Adv ; 9(43): eadg6686, 2023 10 27.
Article in English | MEDLINE | ID: mdl-37889972

ABSTRACT

Mucosal melanoma (MM), an aggressive rare subtype of melanoma, is distinct from cutaneous melanoma and has poor prognoses. We addressed the lack of cell models for MM by establishing 30 organoids of human oral MM (OMM), which retained major histopathological and functional features of parental tumors. Organoid groups derived from chronologically or intratumorally distinct lesions within the same individual displayed heterogeneous genetics, expression profiles, and drug responses, indicating rapid tumor evolution and poor clinical response. Furthermore, transcriptome analysis revealed receptor tyrosine kinases (RTKs) signaling, particularly NGFR, a nerve growth factor receptor, was significantly up-regulated in OMMs and organoids from patients resistant to anti-programmed cell death protein 1 (anti-PD-1) therapy. Combining anti-PD-1 with anlotinib (a phase 2 multitarget RTK inhibitor for OMM) or NGFR knockdown enhanced the effective activity of immune cells in organoid-immune cell coculture systems. Together, our study suggested that OMM organoids serve as faithful models for exploring tumor evolution and immunotherapy combination strategies.


Subject(s)
Melanoma , Skin Neoplasms , Humans , Melanoma/drug therapy , Skin Neoplasms/pathology , Gene Expression Profiling , Immunotherapy , Organoids
2.
Cell Discov ; 9(1): 28, 2023 Mar 13.
Article in English | MEDLINE | ID: mdl-36914617

ABSTRACT

Precancerous lesions of the oral mucosa, especially those accompanied by moderate to severe dysplasia, contribute to the initiation of oral squamous cell carcinoma (OSCC). However, the cellular compositions and spatial organization of the precancerous stage and how these factors promote human OSCC initiation remain unclear. Here, we built a single-cell transcriptome atlas and a spatial transcriptome map after obtaining data from pairwise human oral mucosal biopsies of 9 individuals consisting of very early-stage OSCC, adjacent precancerous lesions with moderate to severe dysplasia, as well as a matched normal region. An altered epithelial gene-expression profile was identified which favored OSCC initiation. This observation was coupled with distinct fibroblast, monocytic, and regulatory T-cell subclusters involved in reshaping the microenvironment. In particular, a unique immune-inhibitory monocyte subtype and spatial-switching regulation of VEGF signaling were observed surrounding precancerous lesions, concertedly strengthening activities in promoting cancer initiation. Collectively, our work elucidated the cellular landscapes and roles of precancerous lesions underlying OSCC initiation, which is essential for understanding the entire OSCC initiation process and helps inform therapeutic strategies for cancer intervention.

3.
Int J Oral Sci ; 15(1): 9, 2023 02 10.
Article in English | MEDLINE | ID: mdl-36765028

ABSTRACT

Cancer cell membrane (CCM) derived nanotechnology functionalizes nanoparticles (NPs) to recognize homologous cells, exhibiting translational potential in accurate tumor therapy. However, these nanoplatforms are majorly generated from fixed cell lines and are typically evaluated in cell line-derived subcutaneous-xenografts (CDX), ignoring the tumor heterogeneity and differentiation from inter- and intra- individuals and microenvironments between heterotopic- and orthotopic-tumors, limiting the therapeutic efficiency of such nanoplatforms. Herein, various biomimetic nanoplatforms (CCM-modified gold@Carbon, i.e., Au@C-CCM) were fabricated by coating CCMs of head and neck squamous cell carcinoma (HNSCC) cell lines and patient-derived cells on the surface of Au@C NP. The generated Au@C-CCMs were evaluated on corresponding CDX, tongue orthotopic xenograft (TOX), immune-competent primary and distant tumor models, and patient-derived xenograft (PDX) models. The Au@C-CCM generates a photothermal conversion efficiency up to 44.2% for primary HNSCC therapy and induced immunotherapy to inhibit metastasis via photothermal therapy-induced immunogenic cell death. The homologous CCM endowed the nanoplatforms with optimal targeting properties for the highest therapeutic efficiency, far above those with mismatched CCMs, resulting in distinct tumor ablation and tumor growth inhibition in all four models. This work reinforces the feasibility of biomimetic NPs combining modular designed CMs and functional cores for customized treatment of HNSCC, can be further extended to other malignant tumors therapy.


Subject(s)
Head and Neck Neoplasms , Photothermal Therapy , Animals , Humans , Squamous Cell Carcinoma of Head and Neck/therapy , Heterografts , Biomimetics , Disease Models, Animal , Head and Neck Neoplasms/therapy , Cell Line, Tumor , Tumor Microenvironment
4.
Adv Mater ; 35(12): e2210787, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36656993

ABSTRACT

Although immunotherapy has revolutionized oncotherapy, only ≈15% of head and neck squamous cell carcinoma (HNSCC) patients benefit from the current therapies. An immunosuppressive tumor microenvironment (TME) and dysregulation of the polycomb ring finger oncogene BMI1 are potential reasons for the failure. Herein, to promote immunotherapeutic efficacy against HNSCC, an injectable nanocomposite hydrogel is developed with a polymer framework (PLGA-PEG-PLGA) that is loaded with both imiquimod encapsulated CaCO3 nanoparticles (RC) and cancer cell membrane (CCM)-coated mesoporous silica nanoparticles containing a peptide-based proteolysis-targeting chimeras (PROTAC) for BMI1 and paclitaxel (PepM@PacC). Upon injection, this nanocomposite hydrogel undergoes in situ gelation, after which it degrades in the TME over time, releasing RC and PepM@PacC nanoparticles to respectively perform immunotherapy and chemotherapy. Specifically, the RC particles selectively manipulate tumor-associated macrophages and dendritic cells to activate a T-cell immune response, while CCM-mediated homologous targeting and endocytosis delivers the PepM@PacC particles into cancer cells, where endogenous glutathione promotes disulfide bond cleavage to release the PROTAC peptide for BMI1 degradation and frees the paclitaxel from the particle pores to elicit apoptosis meanwhile enhance immunotherapy. Thus, the nanocomposite hydrogel, which is designed to exploit multiple known vulnerabilities of HNSCC, succeeds in suppressing both growth and metastasis of HNSCC.


Subject(s)
Head and Neck Neoplasms , Immunotherapy , Humans , Squamous Cell Carcinoma of Head and Neck/therapy , Nanogels , Proteolysis , Head and Neck Neoplasms/drug therapy , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Tumor Microenvironment
5.
JCI Insight ; 7(22)2022 11 22.
Article in English | MEDLINE | ID: mdl-36509291

ABSTRACT

The aberrant activation of STAT3 is associated with the etiology and progression in a variety of malignant epithelial-derived tumors, including head and neck squamous cell carcinoma (HNSCC) and colorectal cancer (CRC). Due to the lack of an enzymatic catalytic site or a ligand-binding pocket, there are no small-molecule inhibitors directly targeting STAT3 that have been approved for clinical translation. Emerging proteolysis targeting chimeric (PROTAC) technology-based approach represents a potential strategy to overcome the limitations of conventional inhibitors and inhibit activation of STAT3 and downstream genes. In this study, the heterobifunctional small-molecule-based PROTACs are successfully prepared from toosendanin (TSN), with 1 portion binding to STAT3 and the other portion binding to an E3 ubiquitin ligase. The optimized lead PROTAC (TSM-1) exhibits superior selectivity, potency, and robust antitumor effects in STAT3-dependent HNSCC and CRC - especially in clinically relevant patient-derived xenografts (PDX) and patient-derived organoids (PDO). The following mechanistic investigation identifies the reduced expression of critical downstream STAT3 effectors, through which TSM-1 promotes cell cycle arrest and apoptosis in tumor cells. These findings provide the first demonstration to our knowledge of a successful PROTAC-targeting strategy in STAT3-dependent epithelial cancer.


Subject(s)
Head and Neck Neoplasms , Ubiquitin-Protein Ligases , Humans , Proteolysis , Squamous Cell Carcinoma of Head and Neck/drug therapy , Ubiquitin-Protein Ligases/metabolism , Head and Neck Neoplasms/drug therapy , STAT3 Transcription Factor/metabolism
6.
Sci Transl Med ; 14(661): eabo5987, 2022 09 07.
Article in English | MEDLINE | ID: mdl-36070368

ABSTRACT

Head and neck squamous cell carcinoma (HNSCC) is a common and frequently lethal cancer with few therapeutic options. In particular, there are few effective targeted therapies. Development of highly effective therapeutic strategies tailored to patients with HNSCC remains a pressing challenge. To address this, we present a pharmacogenomic study to facilitate precision treatments for patients with HNSCC. We established a large collection of 56 HNSCC patient-derived cells (PDCs), which recapitulated the molecular features of the original tumors. Pharmacological assessment of HNSCCs was conducted using a three-tiered high-throughput drug screening using 2248 compounds across these PDC models and an additional 18 immortalized cell lines. We integrated genomic, transcriptomic, and pharmacological analysis to predict biomarkers, gene-drug associations, and validated biomarkers. These results supported drug repurposing for multiple HNSCC subtypes, including the JAK2 inhibitor fedratinib, for low KRT18-expressing HNSCC cases, and the topoisomerase inhibitor mitoxantrone, for IL6R-activated HNSCC cases. Our results demonstrated concordance between susceptibility predictions from the PDCs and the matched patients' responses to standard clinical medication. Moreover, we identified and experimentally confirmed that high expression of ITGB1 elicited therapeutic resistance to docetaxel and high SOD1 expression conferred resistance to afatinib. We further validated ITGB1 as a predictive biomarker for the efficacy of docetaxel therapy in a phase 2 clinical trial. In summary, our study shows that this HNSCC cell resource, as well as the resulting pharmacogenomic profiles, is effective for biomarker discovery and for guiding precision oncology therapies in HNSCCs.


Subject(s)
Head and Neck Neoplasms , Pharmacogenetics , Docetaxel , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/genetics , Humans , Precision Medicine , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/genetics
7.
Signal Transduct Target Ther ; 7(1): 73, 2022 03 08.
Article in English | MEDLINE | ID: mdl-35260570

ABSTRACT

Cetuximab is a widely used drug for treating head and neck squamous cell carcinomas (HNSCCs); however, it provides restricted clinical benefits, and its response duration is limited by drug resistance. Here, we conducted randomized "Phase II-like clinical trials" of 49 HNSCC PDX models and reveal multiple informative biomarkers for intrinsic resistance to cetuximab (e.g., amplification of ANKH, up-regulation of PARP3). After validating these intrinsic resistance biomarkers in another HNSCC PDX cohort (61 PDX models), we generated acquired cetuximab resistance PDX models and analyzed them to uncover resistance mechanisms. Whole exome sequencing and transcriptome sequencing revealed diverse patterns of clonal selection in acquired resistant PDXs, including the emergence of subclones with strongly activated RAS/MAPK. Extending these insights, we show that a combination of a RAC1/RAC3 dual-target inhibitor and cetuximab could overcome acquired cetuximab resistance in vitro and in vivo. Beyond revealing intrinsic resistance biomarkers, our PDX-based study shows how clonal architecture changes underlying acquired resistance can be targeted to expand the therapeutic utility of this important drug to more HNSCC patients.


Subject(s)
Drug Resistance, Neoplasm , Head and Neck Neoplasms , Squamous Cell Carcinoma of Head and Neck , Biomarkers , Cetuximab/pharmacology , Drug Resistance, Neoplasm/genetics , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/genetics , Humans , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/genetics
8.
Onco Targets Ther ; 13: 541-548, 2020.
Article in English | MEDLINE | ID: mdl-32021296

ABSTRACT

AIM: To identify new biomarkers of prostate cancer (PCa) for the diagnosis and prediction of clinical outcomes. MATERIALS AND METHODS: Existing microarray data of PCa tissues in the Oncomine database were analyzed and candidate differentially expressed genes (DEGs) that may be novel and noninvasive biomarkers were obtained. On this basis, plasma mRNA was extracted from PCa patients and healthy donors. Furthermore, plasma mRNA expression of DEGs was evaluated by qRT-PCR. Finally, the diagnostic power of the biomarkers was evaluated in comparison to the clinical characteristics of the patients. RESULTS: In this study, the top five significantly overexpressed mRNA (AMACR, PPP1R14b, PCA3, DLX1, and RPL22L1) and the top five significantly underexpressed mRNA (DUOX1, EFS, GSTP1, S100A16, and NCRNA00087) were selected for further validation in PCa patients and healthy donors by qRT-PCR. The results showed that AMACR, DLX1, PCA3, DUOX1, and GSTP1 mRNA were stably amplified in plasma. Additionally, DLX1, PCA3, DUOX1, and GSTP1 mRNA expression was significantly different between PCa circulating free mRNA samples and healthy donors. These mRNAs may be useful biomarkers for PCa diagnosis. CONCLUSION: Analysis of the expression of genes in the Oncomine database showed that DLX1, PCA3, and DUOX1 expressions have a cancer specific pattern in PCa. Collectively, DLX1, PCA3, and DUOX1 may be useful candidate biomarkers for PCa diagnosis.

9.
Adv Sci (Weinh) ; 6(20): 1901461, 2019 Oct 16.
Article in English | MEDLINE | ID: mdl-31637173

ABSTRACT

Stimuli-responsive nanomedicines have become a recent research focus as a candidate for cancer treatment because of their effectiveness, sensibility, and minimal invasiveness. In this work, a novel nanosystem is developed based on Cu2- x S@MnS core-shell nanoparticles (CSNPs) in which the Cu2- x S core serves as a photosensitizer to generate hyperthermia and reactive oxygen species (ROS), and the MnS shell is used in H2O2-responsive O2 production. Cu2 -x S@MnS CSNPs with an independent core and shell ratio are synthesized by a controllable hot-injection method, resulting in an optimal photothermal (PT) effect with a PT conversion efficiency of up to 47.9%. An enhanced photodynamic (PD) effect also occurs in an H2O2 environment. More significantly, in vivo experiments demonstrate that Cu2 -x S@MnS CSNPs can mediate tumor shrinkage in both HeLa tumor cell line-derived xenograft (CDX) and head and neck squamous cell carcinoma (HNSCC) patient-derived xenograft (PDX) models, with the capability of being used as a T1-enhanced magnetic resonance (MR) contrast agent. These results suggest the great potential of as-prepared Cu2 -x S@MnS CSNPs as photo/H2O2-responsive therapeutic-agents against tumors, even in a complicated and heterogeneous environment, thus promoting the clinical translation of nanomedicine.

10.
Clin Cancer Res ; 25(12): 3548-3560, 2019 Jun 15.
Article in English | MEDLINE | ID: mdl-30782616

ABSTRACT

PURPOSE: Unlike advances in the genomics-driven precision treatment of cutaneous melanomas, the current poor understanding of the molecular basis of mucosal melanomas (MM) has hindered such progress for MM patients. Thus, we sought to characterize the genomic landscape of MM to identify genomic alterations with prognostic and/or therapeutic implications. EXPERIMENTAL DESIGN: Whole-genome sequencing (WGS) was performed on 65 MM samples, including 63 paired tumor blood samples and 2 matched lymph node metastases, with a further droplet digital PCR-based validation study of an independent MM cohort (n = 80). Guided by these molecular insights, the FDA-approved CDK4/6 inhibitor palbociclib was tested in an MM patient-derived xenograft (PDX) trial. RESULTS: Besides the identification of well-recognized driver mutations of BRAF (3.1%), RAS family (6.2%), NF1 (7.8%), and KIT (23.1%) in MMs, our study also found that (i) mutations and amplifications in the transmembrane nucleoporin gene POM121 (30.8%) defined a patient subgroup with higher tumor proliferation rates; (ii) enrichment of structural variations between chromosomes 5 and 12 defined a patient subgroup with significantly worse clinical outcomes; (iii) over 50% of the MM patients harbored recurrent focal amplification of several oncogenes (CDK4, MDM2, and AGAP2) at 12q13-15, and this co-occurred significantly with amplification of TERT at 5p15, which was verified in the validation cohort; (iv) the PDX trial demonstrated robust antitumor effects of palbociclib in MMs harboring CDK4 amplification. CONCLUSIONS: Our largest-to-date cohort WGS analysis of MMs defines the genomic landscape of this deadly cancer at unprecedented resolution and identifies genomic aberrations that could facilitate the delivery of precision cancer treatments.See related commentary by Shoushtari, p. 3473.


Subject(s)
Melanoma/genetics , Skin Neoplasms/genetics , Genomics , Humans , Mutation , Prognosis
SELECTION OF CITATIONS
SEARCH DETAIL
...