Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 145
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 20(7): 835-851, 2019 07.
Article in English | MEDLINE | ID: mdl-31160797

ABSTRACT

How tumor cells genetically lose antigenicity and evade immune checkpoints remains largely elusive. We report that tissue-specific expression of the human long noncoding RNA LINK-A in mouse mammary glands initiates metastatic mammary gland tumors, which phenotypically resemble human triple-negative breast cancer (TNBC). LINK-A expression facilitated crosstalk between phosphatidylinositol-(3,4,5)-trisphosphate and inhibitory G-protein-coupled receptor (GPCR) pathways, attenuating protein kinase A-mediated phosphorylation of the E3 ubiquitin ligase TRIM71. Consequently, LINK-A expression enhanced K48-polyubiquitination-mediated degradation of the antigen peptide-loading complex (PLC) and intrinsic tumor suppressors Rb and p53. Treatment with LINK-A locked nucleic acids or GPCR antagonists stabilized the PLC components, Rb and p53, and sensitized mammary gland tumors to immune checkpoint blockers. Patients with programmed ccll death protein-1(PD-1) blockade-resistant TNBC exhibited elevated LINK-A levels and downregulated PLC components. Hence we demonstrate lncRNA-dependent downregulation of antigenicity and intrinsic tumor suppression, which provides the basis for developing combinational immunotherapy treatment regimens and early TNBC prevention.


Subject(s)
Antigen Presentation/immunology , Gene Expression Regulation, Neoplastic , Neoplasms/genetics , Neoplasms/immunology , Oncogenes , RNA, Long Noncoding/genetics , Tumor Escape/genetics , Tumor Escape/immunology , Adenoma/genetics , Adenoma/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Disease Models, Animal , Disease Progression , Humans , Mice , Neoplasms/metabolism , Neoplasms/pathology , Phosphorylation , Receptors, G-Protein-Coupled/antagonists & inhibitors , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Tumor Suppressor Protein p53/metabolism , Ubiquitination , Xenograft Model Antitumor Assays
2.
Cell ; 149(5): 1098-111, 2012 May 25.
Article in English | MEDLINE | ID: mdl-22632973

ABSTRACT

Akt kinase plays a central role in cell growth, metabolism, and tumorigenesis. The TRAF6 E3 ligase orchestrates IGF-1-mediated Akt ubiquitination and activation. Here, we show that Akt ubiquitination is also induced by activation of ErbB receptors; unexpectedly, and in contrast to IGF-1 induced activation, the Skp2 SCF complex, not TRAF6, is a critical E3 ligase for ErbB-receptor-mediated Akt ubiquitination and membrane recruitment in response to EGF. Skp2 deficiency impairs Akt activation, Glut1 expression, glucose uptake and glycolysis, and breast cancer progression in various tumor models. Moreover, Skp2 overexpression correlates with Akt activation and breast cancer metastasis and serves as a marker for poor prognosis in Her2-positive patients. Finally, Skp2 silencing sensitizes Her2-overexpressing tumors to Herceptin treatment. Our study suggests that distinct E3 ligases are utilized by diverse growth factors for Akt activation and that targeting glycolysis sensitizes Her2-positive tumors to Herceptin treatment.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Breast Neoplasms/drug therapy , Cell Transformation, Neoplastic , F-Box Proteins/metabolism , Glycolysis , S-Phase Kinase-Associated Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Breast Neoplasms/metabolism , Disease Models, Animal , Drug Resistance, Neoplasm , Female , Humans , Mice , Receptor, ErbB-2/metabolism , S-Phase Kinase-Associated Proteins/genetics , Trastuzumab , Ubiquitination
3.
Cell Mol Life Sci ; 80(10): 297, 2023 Sep 20.
Article in English | MEDLINE | ID: mdl-37728789

ABSTRACT

Human brain is characterized by extremely sparse extracellular matrix (ECM). Despite its low abundance, the significance of brain ECM in both physiological and pathological conditions should not be underestimated. Brain metastasis is a serious complication of cancer, and recent findings highlighted the contribution of ECM in brain metastasis development. In this review, we provide a comprehensive outlook on how ECM proteins promote brain metastasis seeding. In particular, we discuss (1) disruption of the blood-brain barrier in brain metastasis; (2) role of ECM in modulating brain metastasis dormancy; (3) regulation of brain metastasis seeding by ECM-activated integrin signaling; (4) functions of brain-specific ECM protein reelin in brain metastasis. Lastly, we consider the possibility of targeting ECM for brain metastasis management.


Subject(s)
Brain Neoplasms , Humans , Extracellular Matrix , Brain , Extracellular Matrix Proteins , Blood-Brain Barrier
4.
J Biol Chem ; 298(4): 101817, 2022 04.
Article in English | MEDLINE | ID: mdl-35278434

ABSTRACT

Expression of the receptor tyrosine kinase ephrin receptor A10 (EphA10), which is undetectable in most normal tissues except for the male testis, has been shown to correlate with tumor progression and poor prognosis in several malignancies, including triple-negative breast cancer (TNBC). Therefore, EphA10 could be a potential therapeutic target, likely with minimal adverse effects. However, no effective clinical drugs against EphA10 are currently available. Here, we report high expression levels of EphA10 in tumor regions of breast, lung, and ovarian cancers as well as in immunosuppressive myeloid cells in the tumor microenvironment. Furthermore, we developed anti-EphA10 monoclonal antibodies (mAbs) that specifically recognize cell surface EphA10, but not other EphA family isoforms, and target tumor regions precisely in vivo with no apparent accumulation in other organs. In syngeneic TNBC mouse models, we found that anti-EphA10 mAb clone #4 enhanced tumor regression, therapeutic response rate, and T cell-mediated antitumor immunity. Notably, the chimeric antigen receptor T cells derived from clone #4 significantly inhibited TNBC cell viability in vitro and tumor growth in vivo. Together, our findings suggest that targeting EphA10 via EphA10 mAbs and EphA10-specific chimeric antigen receptor-T cell therapy may represent a promising strategy for patients with EphA10-positive tumors.


Subject(s)
Antibodies, Monoclonal , Receptors, Chimeric Antigen , Receptors, Eph Family , T-Lymphocytes , Triple Negative Breast Neoplasms , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , Humans , Mice , Receptors, Eph Family/immunology , T-Lymphocytes/metabolism , Triple Negative Breast Neoplasms/drug therapy , Xenograft Model Antitumor Assays
6.
Acta Neuropathol ; 141(2): 303-321, 2021 02.
Article in English | MEDLINE | ID: mdl-33394124

ABSTRACT

The deadly complication of brain metastasis (BM) is largely confined to a relatively narrow cross-section of systemic malignancies, suggesting a fundamental role for biological mechanisms shared across commonly brain metastatic tumor types. To identify and characterize such mechanisms, we performed genomic, transcriptional, and proteomic profiling using whole-exome sequencing, mRNA-seq, and reverse-phase protein array analysis in a cohort of the lung, breast, and renal cell carcinomas consisting of BM and patient-matched primary or extracranial metastatic tissues. While no specific genomic alterations were associated with BM, correlations with impaired cellular immunity, upregulated oxidative phosphorylation (OXPHOS), and canonical oncogenic signaling pathways including phosphoinositide 3-kinase (PI3K) signaling, were apparent across multiple tumor histologies. Multiplexed immunofluorescence analysis confirmed significant T cell depletion in BM, indicative of a fundamentally altered immune microenvironment. Moreover, functional studies using in vitro and in vivo modeling demonstrated heightened oxidative metabolism in BM along with sensitivity to OXPHOS inhibition in murine BM models and brain metastatic derivatives relative to isogenic parentals. These findings demonstrate that pathophysiological rewiring of oncogenic signaling, cellular metabolism, and immune microenvironment broadly characterizes BM. Further clarification of this biology will likely reveal promising targets for therapeutic development against BM arising from a broad variety of systemic cancers.


Subject(s)
Brain Neoplasms/pathology , Brain Neoplasms/secondary , DNA Fingerprinting/methods , Genomics/methods , Animals , Base Sequence , Brain Neoplasms/immunology , Cell Survival , Female , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, SCID , Neoplasm Transplantation , Protein Array Analysis , Proteomics , Superoxide Dismutase/metabolism , Survival Analysis , Exome Sequencing
7.
Nature ; 527(7576): 100-104, 2015 Nov 05.
Article in English | MEDLINE | ID: mdl-26479035

ABSTRACT

The development of life-threatening cancer metastases at distant organs requires disseminated tumour cells' adaptation to, and co-evolution with, the drastically different microenvironments of metastatic sites. Cancer cells of common origin manifest distinct gene expression patterns after metastasizing to different organs. Clearly, the dynamic interaction between metastatic tumour cells and extrinsic signals at individual metastatic organ sites critically effects the subsequent metastatic outgrowth. Yet, it is unclear when and how disseminated tumour cells acquire the essential traits from the microenvironment of metastatic organs that prime their subsequent outgrowth. Here we show that both human and mouse tumour cells with normal expression of PTEN, an important tumour suppressor, lose PTEN expression after dissemination to the brain, but not to other organs. The PTEN level in PTEN-loss brain metastatic tumour cells is restored after leaving the brain microenvironment. This brain microenvironment-dependent, reversible PTEN messenger RNA and protein downregulation is epigenetically regulated by microRNAs from brain astrocytes. Mechanistically, astrocyte-derived exosomes mediate an intercellular transfer of PTEN-targeting microRNAs to metastatic tumour cells, while astrocyte-specific depletion of PTEN-targeting microRNAs or blockade of astrocyte exosome secretion rescues the PTEN loss and suppresses brain metastasis in vivo. Furthermore, this adaptive PTEN loss in brain metastatic tumour cells leads to an increased secretion of the chemokine CCL2, which recruits IBA1-expressing myeloid cells that reciprocally enhance the outgrowth of brain metastatic tumour cells via enhanced proliferation and reduced apoptosis. Our findings demonstrate a remarkable plasticity of PTEN expression in metastatic tumour cells in response to different organ microenvironments, underpinning an essential role of co-evolution between the metastatic cells and their microenvironment during the adaptive metastatic outgrowth. Our findings signify the dynamic and reciprocal cross-talk between tumour cells and the metastatic niche; importantly, they provide new opportunities for effective anti-metastasis therapies, especially of consequence for brain metastasis patients.


Subject(s)
Brain Neoplasms/pathology , Brain Neoplasms/secondary , Exosomes/genetics , Gene Expression Regulation, Neoplastic , Gene Silencing , MicroRNAs/genetics , PTEN Phosphohydrolase/deficiency , Tumor Microenvironment , Adaptation, Physiological/genetics , Animals , Astrocytes/cytology , Astrocytes/metabolism , Brain/metabolism , Brain/pathology , Brain Neoplasms/metabolism , Calcium-Binding Proteins , Cell Proliferation/genetics , Chemokine CCL2/metabolism , DNA-Binding Proteins/metabolism , Down-Regulation/genetics , Evolution, Molecular , Exosomes/metabolism , Female , Genes, Tumor Suppressor , Humans , Male , Mice , Microfilament Proteins , PTEN Phosphohydrolase/genetics , RNA, Messenger/analysis , RNA, Messenger/genetics , Tumor Microenvironment/genetics , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
8.
J Biomed Sci ; 27(1): 77, 2020 Jul 03.
Article in English | MEDLINE | ID: mdl-32620165

ABSTRACT

N-linked glycosylation is one of the most abundant posttranslational modifications of membrane-bound proteins in eukaryotes and affects a number of biological activities, including protein biosynthesis, protein stability, intracellular trafficking, subcellular localization, and ligand-receptor interaction. Accumulating evidence indicates that cell membrane immune checkpoint proteins, such as programmed death-ligand 1 (PD-L1), are glycosylated with heavy N-linked glycan moieties in human cancers. N-linked glycosylation of PD-L1 maintains its protein stability and interaction with its cognate receptor, programmed cell death protein 1 (PD-1), and this in turn promotes evasion of T-cell immunity. Studies have suggested targeting PD-L1 glycosylation as a therapeutic option by rational combination of cancer immunotherapies. Interestingly, structural hindrance by N-glycan on PD-L1 in fixed samples impedes its recognition by PD-L1 diagnostic antibodies. Notably, the removal of N-linked glycosylation enhances PD-L1 detection in a variety of bioassays and more accurately predicts the therapeutic efficacy of PD-1/PD-L1 inhibitors, suggesting an important clinical implication of PD-L1 N-linked glycosylation. A detailed understanding of the regulatory mechanisms, cellular functions, and diagnostic limits underlying PD-L1 N-linked glycosylation could shed new light on the clinical development of immune checkpoint inhibitors for cancer treatment and deepen our knowledge of biomarkers to identify patients who would benefit the most from immunotherapy. In this review, we highlight the effects of protein glycosylation on cancer immunotherapy using N-linked glycosylation of PD-L1 as an example. In addition, we consider the potential impacts of PD-L1 N-linked glycosylation on clinical diagnosis. The notion of utilizing the deglycosylated form of PD-L1 as a predictive biomarker to guide anti-PD-1/PD-L1 immunotherapy is also discussed.


Subject(s)
B7-H1 Antigen/metabolism , Immunotherapy , Neoplasms/diagnosis , Neoplasms/therapy , Nitrogen/metabolism , Glycosylation , Humans
9.
Biochim Biophys Acta Rev Cancer ; 1867(1): 49-57, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27939792

ABSTRACT

The metastasis of cancer to the central nervous system (CNS) remains a devastating clinical reality, carrying an estimated survival time of less than one year in spite of recent therapeutic breakthroughs for other disease contexts. Advances in brain metastasis research are hindered by a number of factors, including its complicated nature and the difficulty of modeling metastatic cancer growth in the unique brain microenvironment. In this review, we will discuss the clinical challenge, and compare the merits and limitations of the available models for brain metastasis research. Additionally, we will specifically address current knowledge on how brain metastases take advantage of the unique brain environment to benefit their own growth. Finally, we will explore the distinctive metabolic and chemical characteristics of the brain and how these paradoxically represent barriers to establishment of brain metastasis, but also provide ample supplies for metastatic cells' growth in the brain. We envision that multi-disciplinary innovative approaches will open opportunities for the field to make breakthroughs in tackling unique challenges of brain metastasis.


Subject(s)
Brain Neoplasms/pathology , Brain/pathology , Neoplasm Metastasis/pathology , Animals , Cell Proliferation/physiology , Humans
10.
Am J Physiol Gastrointest Liver Physiol ; 317(5): G625-G639, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31545917

ABSTRACT

Dissemination of primary tumors to distant anatomical sites has a substantial negative impact on patient prognosis. The liver is a common site for metastases from colorectal cancer, and patients with hepatic metastases have generally much shorter survival, raising a need to develop and implement novel strategies for targeting metastatic disease. The extracellular matrix (ECM) is a meshwork of highly crosslinked, insoluble high-molecular-mass proteins maintaining tissue integrity and establishing cell-cell interactions. Emerging evidence identifies the importance of the ECM in cancer cell migration, invasion, intravasation, and metastasis. Here, we isolated the ECM from MC38 mouse liver metastases using our optimized method of mild detergent solubilization followed by biochemical enrichment. The matrices were subjected to label-free quantitative mass spectrometry analysis, revealing proteins highly abundant in the metastatic matrisome. The resulting list of proteins upregulated in the ECM significantly predicted survival in patients with colorectal cancer but not other cancers with strong involvement of the ECM component. One of the proteins upregulated in liver metastatic ECM, annexin A1, was not previously studied in the context of cancer-associated matrisome. Here, we show that annexin A1 was markedly upregulated in colon cancer cell lines compared with cancer cells of other origin and also over-represented in human primary colorectal lesions, as well as hepatic metastases, compared with their adjacent healthy tissue counterparts. In conclusion, our study provides a comprehensive ECM characterization of MC38 experimental liver metastases and proposes annexin A1 as a putative target for this disease.NEW & NOTEWORTHY Here, the authors provide an extensive proteomics characterization of murine colorectal cancer liver metastasis matrisome (the ensemble of all extracellular matrix molecules). The findings presented in this study may enable identification of therapeutic targets or biomarkers of hepatic metastases.


Subject(s)
Colorectal Neoplasms/genetics , Extracellular Matrix Proteins/metabolism , Liver Neoplasms/genetics , Proteome/metabolism , Animals , Annexin A1/genetics , Annexin A1/metabolism , Colon/metabolism , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Extracellular Matrix Proteins/genetics , Female , Gene Expression Regulation, Neoplastic , HT29 Cells , Humans , Liver/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/secondary , Mice , Mice, Inbred C57BL , Proteome/genetics , Up-Regulation
11.
Cancer Metastasis Rev ; 35(4): 677-684, 2016 12.
Article in English | MEDLINE | ID: mdl-27873078

ABSTRACT

The past decade has witnessed impressive advances in cancer treatment ushered in by targeted and immunotherapies. However, with significantly prolonged survival, upon recurrence, more patients become inflicted by brain metastasis, which is mostly refractory to all currently available therapeutic regimens. Historically, brain metastasis is an understudied area in cancer research, partly due to the dearth of appropriate experimental models that closely simulate the special biological features of metastasis in the unique brain environment and to the sophistication of techniques required to perform in-depth studies of the extremely complex and challenging brain metastasis. Yet, with increasing clinical demand for more effective treatment options, brain metastasis research has rapidly advanced in recent years. The present review spotlights the recent major progresses in basic and translational studies of brain metastasis with focuses on new animal models, novel imaging technologies, omics "big data" resources, and some new and exciting biological insights on brain metastasis.


Subject(s)
Brain Neoplasms/secondary , Breast Neoplasms/pathology , Animals , Female , Humans
12.
Stem Cells ; 33(10): 2985-94, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26260958

ABSTRACT

The treatment of human epidermal growth factor receptor 2 (HER2)-overexpressing breast cancer has been revolutionized by trastuzumab. However, longer survival of these patients now predisposes them to forming HER2 positive brain metastases, as the therapeutic antibodies cannot cross the blood brain barrier. The current oncologic repertoire does not offer a rational, nontoxic targeted therapy for brain metastases. In this study, we used an established human neural stem cell line, HB1.F3 NSCs and generated a stable pool of cells secreting a high amount of functional full-length anti-HER2 antibody, equivalent to trastuzumab. Anti-HER2Ab secreted by the NSCs (HER2Ab-NSCs) specifically binds to HER2 overexpressing human breast cancer cells and inhibits PI3K-Akt signaling. This translates to HER2Ab-NSC inhibition of breast cancer cell growth in vitro. Preclinical in vivo experiments using HER2Ab overexpressing NSCs in a breast cancer brain metastases (BCBM) mouse model demonstrate that intracranial injection of HER2Ab-NSCs significantly improves survival. In effect, these NSCs provide tumor localized production of HER2Ab, minimizing any potential off-target side effects. Our results establish HER2Ab-NSCs as a novel, nontoxic, and rational therapeutic approach for the successful treatment of HER2 overexpressing BCBM, which now warrants further preclinical and clinical investigation.


Subject(s)
Antibodies, Anti-Idiotypic/biosynthesis , Brain Neoplasms/therapy , Breast Neoplasms/drug therapy , Neural Stem Cells/metabolism , Receptor, ErbB-2/biosynthesis , Animals , Antibodies, Anti-Idiotypic/immunology , Blood-Brain Barrier/drug effects , Brain Neoplasms/pathology , Brain Neoplasms/secondary , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Mice , Neural Stem Cells/immunology , Neural Stem Cells/transplantation , Receptor, ErbB-2/immunology , Trastuzumab/therapeutic use , Xenograft Model Antitumor Assays
13.
Clin Exp Metastasis ; 41(3): 187-198, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38430319

ABSTRACT

Brain metastasis, characterized by poor clinical outcomes, is a devastating disease. Despite significant mechanistic and therapeutic advances in recent years, pivotal improvements in clinical interventions have remained elusive. The heterogeneous nature of the primary tumor of origin, complications in drug delivery across the blood-brain barrier, and the distinct microenvironment collectively pose formidable clinical challenges in developing new treatments for patients with brain metastasis. Although current preclinical models have deepened our basic understanding of the disease, much of the existing research on brain metastasis has employed a reductionist approach. This approach, which often relies on either in vitro systems or in vivo injection models in young and treatment-naive mouse models, does not give sufficient consideration to the clinical context. Given the translational importance of brain metastasis research, we advocate for the design of preclinical experimental models that take into account these unique clinical challenges and align more closely with current clinical practices. We anticipate that aligning and simulating real-world patient conditions will facilitate the development of more translatable treatment regimens. This brief review outlines the most pressing clinical challenges, the current state of research in addressing them, and offers perspectives on innovative metastasis models and tools aimed at identifying novel strategies for more effective management of clinical brain metastasis.


Subject(s)
Brain Neoplasms , Translational Research, Biomedical , Brain Neoplasms/secondary , Humans , Animals , Disease Models, Animal , Blood-Brain Barrier , Tumor Microenvironment , Mice
14.
Nat Commun ; 15(1): 1009, 2024 Feb 02.
Article in English | MEDLINE | ID: mdl-38307859

ABSTRACT

Tumor-secreted factors contribute to the development of a microenvironment that facilitates the escape of cancer cells from immunotherapy. In this study, we conduct a retrospective comparison of the proteins secreted by hepatocellular carcinoma (HCC) cells in responders and non-responders among a cohort of ten patients who received Nivolumab (anti-PD-1 antibody). Our findings indicate that non-responders have a high abundance of secreted RNase1, which is associated with a poor prognosis in various cancer types. Furthermore, mice implanted with HCC cells that overexpress RNase1 exhibit immunosuppressive tumor microenvironments and diminished response to anti-PD-1 therapy. RNase1 induces the polarization of macrophages towards a tumor growth-promoting phenotype through activation of the anaplastic lymphoma kinase (ALK) signaling pathway. Targeting the RNase1/ALK axis reprograms the macrophage polarization, with increased CD8+ T- and Th1- cell recruitment. Moreover, simultaneous targeting of the checkpoint protein PD-1 unleashes cytotoxic CD8+ T-cell responses. Treatment utilizing both an ALK inhibitor and an anti-PD-1 antibody exhibits enhanced tumor regression and facilitates long-term immunity. Our study elucidates the role of RNase1 in mediating tumor resistance to immunotherapy and reveals an RNase1-mediated immunosuppressive tumor microenvironment, highlighting the potential of targeting RNase1 as a promising strategy for cancer immunotherapy in HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Humans , Mice , Anaplastic Lymphoma Kinase , Carcinoma, Hepatocellular/metabolism , CD8-Positive T-Lymphocytes , Immunosuppression Therapy , Liver Neoplasms/metabolism , Retrospective Studies , Ribonucleases , Tumor Microenvironment
15.
Nat Commun ; 15(1): 2132, 2024 Mar 08.
Article in English | MEDLINE | ID: mdl-38459011

ABSTRACT

Growth factor receptor-bound protein 2 (GRB2) is a cytoplasmic adapter for tyrosine kinase signaling and a nuclear adapter for homology-directed-DNA repair. Here we find nuclear GRB2 protects DNA at stalled replication forks from MRE11-mediated degradation in the BRCA2 replication fork protection axis. Mechanistically, GRB2 binds and inhibits RAD51 ATPase activity to stabilize RAD51 on stalled replication forks. In GRB2-depleted cells, PARP inhibitor (PARPi) treatment releases DNA fragments from stalled forks into the cytoplasm that activate the cGAS-STING pathway to trigger pro-inflammatory cytokine production. Moreover in a syngeneic mouse metastatic ovarian cancer model, GRB2 depletion in the context of PARPi treatment reduced tumor burden and enabled high survival consistent with immune suppression of cancer growth. Collective findings unveil GRB2 function and mechanism for fork protection in the BRCA2-RAD51-MRE11 axis and suggest GRB2 as a potential therapeutic target and an enabling predictive biomarker for patient selection for PARPi and immunotherapy combination.


Subject(s)
DNA Replication , Neoplasms , Animals , Humans , Mice , DNA , Genomic Instability , GRB2 Adaptor Protein/genetics , GRB2 Adaptor Protein/metabolism , Immunity, Innate , MRE11 Homologue Protein/metabolism , Neoplasms/genetics , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism
16.
Cancer Metastasis Rev ; 31(3-4): 479-91, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22706845

ABSTRACT

Paget's "seed and soil" hypothesis stated that cancer metastasis requires permissive interactions between tumor cells and secondary organ microenvironments. Many of these "permissive interactions" are now known to be growth factor receptor and ligand interactions by which metastatic tumor cells coopt signaling pathways normally used by host organs. However, although cancer cell signaling pathways responsible for primary cancer growth have been extensively characterized, signaling pathways important in supporting tumor cell-secondary organ heterotypic interactions have been neglected. Even as targeted therapies have shown promise and efficacy in treating myriad primary tumors, metastatic cancer remains incurable. Here, we will discuss several growth factor signaling pathways known to be involved in both general and site-specific metastasis. We will address the complexity in generalizing the role of growth factor signaling in metastasis, as both pro- and antimetastatic roles for the same pathways have been demonstrated depending upon context. We will discuss the limitations of current usage of targeted therapies to pathways known to be dysregulated in metastasis. We propose that the future of cancer metastasis-targeted therapy will lie in better understanding of the interactions between tumor cells and the secondary organ microenvironments that may guide rationally designed personalized combinatorial targeted regimens. We hope to promote research to better understand the complex process of metastasis and ultimately better treatments for the abjectly underserved population of patients with metastatic cancer.


Subject(s)
Intercellular Signaling Peptides and Proteins/physiology , Neoplasm Metastasis/pathology , Signal Transduction/physiology , Animals , ErbB Receptors/physiology , Humans , Neoplasm Metastasis/physiopathology , Proto-Oncogene Proteins c-met/physiology , Receptors, Transforming Growth Factor beta/physiology , Vascular Endothelial Growth Factor A/physiology
17.
Cancer Cell ; 8(6): 437-9, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16338656

ABSTRACT

Akt is well known to enhance malignancy and is recognized as a key target for antineoplastic therapies. However, intriguing findings reported by Yoeli-Lerner et al. in the November 23, 2005 issue of Molecular Cell, suggest a novel, antimetastasis function of Akt: activation of Akt1 inhibited invasion in some cancer cells. One possible mechanism for this surprising phenotype was that Akt activated the E3 ubiquitin ligase HDM2, causing ubiquitination and degradation of NFAT, an invasion-promoting factor. These findings clearly justify further investigations and, if validated in vivo, call for reevaluation of some Akt-targeting therapeutic strategies currently under development.


Subject(s)
Neoplasm Metastasis/prevention & control , Proto-Oncogene Proteins c-akt/physiology , Tumor Suppressor Proteins/physiology , Animals , Humans , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Neoplasm Metastasis/physiopathology , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Proteins/genetics , Ubiquitin/metabolism
18.
Int J Biol Sci ; 19(10): 2957-2973, 2023.
Article in English | MEDLINE | ID: mdl-37416781

ABSTRACT

The secretory enzyme human ribonuclease 1 (RNase1) is involved in innate immunity and anti-inflammation, achieving host defense and anti-cancer effects; however, whether RNase1 contributes to adaptive immune response in the tumor microenvironment (TME) remains unclear. Here, we established a syngeneic immunocompetent mouse model in breast cancer and demonstrated that ectopic RNase1 expression significantly inhibited tumor progression. Overall changes in immunological profiles in the mouse tumors were analyzed by mass cytometry and showed that the RNase1-expressing tumor cells significantly induced CD4+ Th1 and Th17 cells and natural killer cells and reduced granulocytic myeloid-derived suppressor cells, supporting that RNase1 favors an antitumor TME. Specifically, RNase1 increased expression of T cell activation marker CD69 in a CD4+ T cell subset. Notably, analysis of cancer-killing potential revealed that T cell-mediated antitumor immunity was enhanced by RNase1, which further collaborated with an EGFR-CD3 bispecific antibody to protect against breast cancer cells across molecular subtypes. Our results uncover a tumor-suppressive role of RNase1 through adaptive immune response in breast cancer in vivo and in vitro, providing a potential treatment strategy of combining RNase1 with cancer immunotherapies for immunocompetent patients.


Subject(s)
Breast Neoplasms , Humans , Animals , Mice , Female , Breast Neoplasms/pathology , Ribonucleases/pharmacology , Adaptive Immunity , Lymphocyte Activation , T-Lymphocytes , Tumor Microenvironment , Cell Line, Tumor
19.
Eur J Breast Health ; 19(4): 267-273, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37795002

ABSTRACT

Objective: Risk-reducing therapy with selective estrogen receptor (ER) modulators and aromatase inhibitors reduce breast cancer risk. However, the effects are limited to ER-positive breast cancer. Therefore, new agents with improved toxicity profiles that reduce the risk in ER-negative breast cancers are urgently needed. The aim of this prospective, short-term, prevention study was to evaluate the effect of dasatinib, an inhibitor of the tyrosine kinase Src, on biomarkers in normal (but increased risk) breast tissue and serum of women at high risk for a second, contralateral primary breast cancer. Materials and Methods: Women with a history of unilateral stage I, II, or III ER-negative breast cancer, having no active disease, and who completed all adjuvant therapies were eligible. Patients underwent baseline fine-needle aspiration (FNA) of the contralateral breast and serum collection for biomarker analysis and were randomized to receive either no treatment (control) or dasatinib at 40 or 80 mg/day for three months. After three months, serum collection and breast FNA were repeated. Planned biomarker analysis consisted of changes in cytology and Ki-67 on breast FNA, and changes in serum levels of insulin-like growth factor 1 (IGF-1), IGF-binding protein 1, and IGF-binding protein 3. The primary objective was to evaluate changes in Ki-67 and secondary objective included changes in cytology in breast tissue and IGF-related serum biomarkers. Toxicity was also evaluated. Results: Twenty-three patients started their assigned treatments. Compliance during the study was high, with 86.9% (20/23) of patients completing their assigned doses. Dasatinib was well tolerated and no drug-related grade 3 and 4 adverse events were observed. Since only one patient met the adequacy criteria for the paired FNA sample, we could not evaluate Ki-67 level or cytological changes. No significant change in serum biomarkers was observed among the three groups. Conclusion: Dasatinib was well tolerated but did not induce any significant changes in serum biomarkers. The study could not fulfill its primary objective due to an inadequate number of paired FNA samples. Further, larger studies are needed to evaluate the effectiveness of Src inhibitors in breast cancer prevention.

20.
Cell Metab ; 35(8): 1457-1473.e13, 2023 08 08.
Article in English | MEDLINE | ID: mdl-37329887

ABSTRACT

Obesity, in which the functional importance of small nucleolar RNAs (snoRNAs) remains elusive, correlates with risk for many cancer types. Here, we identify that the serum copies of adipocyte-expressed SNORD46 correlate with body mass index (BMI), and serum SNORD46 antagonizes interleukin-15 (IL-15) signaling. Mechanically, SNORD46 binds IL-15 via G11, and G11A (a mutation that significantly enhances binding affinity) knockin drives obesity in mice. Functionally, SNORD46 blocks IL-15-induced, FER kinase-dependent phosphorylation of platelet glycoprotein 4 (CD36) and monoglyceride lipase (MGLL) in adipocytes, leading to inhibited lipolysis and browning. In natural killer (NK) cells, SNORD46 suppresses the IL-15-dependent autophagy, leading to reduced viability of obese NK. SNORD46 power inhibitors exhibit anti-obesity effects, concurring with improved viability of obese NK and anti-tumor immunity of CAR-NK cell therapy. Hence, our findings demonstrate the functional importance of snoRNAs in obesity and the utility of snoRNA power inhibitors for antagonizing obesity-associated immune resistance.


Subject(s)
Lipolysis , RNA, Small Nucleolar , Animals , Mice , RNA, Small Nucleolar/genetics , RNA, Small Nucleolar/metabolism , Interleukin-15/metabolism , Rejuvenation , Adipocytes/metabolism , Obesity/metabolism , Killer Cells, Natural
SELECTION OF CITATIONS
SEARCH DETAIL