Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 257
Filter
1.
Article in English | MEDLINE | ID: mdl-38692741

ABSTRACT

Breast cancer kills hundreds of thousands of people every year. Rapid progress over the past two decades has increased our understanding of the genetic and environmental risk factors for disease. It has also shed light on drivers of tumor progression and the molecular landscape underpinning tumor heterogeneity, as well as the role of the microenvironment and the immune system. These strides forward should lead to more effective and tailored therapies for early- and late-stage patients.

2.
bioRxiv ; 2024 Apr 27.
Article in English | MEDLINE | ID: mdl-38712292

ABSTRACT

Tumor-associated neutrophils (TANs) have been shown to promote immunosuppression and tumor progression, and a high TAN frequency predicts poor prognosis in triple-negative breast cancer (TNBC). Dysregulation of CREB binding protein (CBP)/P300 function has been observed with multiple cancer types. The bromodomain (BRD) of CBP/P300 has been shown to regulate its activity. In this study, we found that IACS-70654, a novel and selective CBP/P300 BRD inhibitor, reduced TANs and inhibited the growth of neutrophil-enriched TNBC models. In the bone marrow, CBP/P300 BRD inhibition reduced the tumor-driven abnormal differentiation and proliferation of neutrophil progenitors. Inhibition of CBP/P300 BRD also stimulated the immune response by inducing an IFN response and MHCI expression in tumor cells and increasing tumor-infiltrated CTLs. Moreover, IACS-70654 improved the response of a neutrophil-enriched TNBC model to docetaxel and immune checkpoint blockade. This provides a rationale for combining a CBP/P300 BRD inhibitor with standard-of-care therapies in future clinical trials for neutrophil-enriched TNBC.

3.
JACC Case Rep ; 29(4): 102202, 2024 Feb 21.
Article in English | MEDLINE | ID: mdl-38379660

ABSTRACT

An 82-year-old man who had undergone Watchman FLX (Boston Scientific) device implantation presented with dyspnea. Multimodality evaluation demonstrated a small fistula from the proximal circumflex artery to the left atrial appendage. Anatomically, the left circumflex artery is close to the atrial appendage; therefore, it is plausible that fistula formation could be a late complication of implantation of the device.

4.
J Clin Invest ; 133(24)2023 Dec 15.
Article in English | MEDLINE | ID: mdl-37874652

ABSTRACT

Protein synthesis is frequently dysregulated in cancer and selective inhibition of mRNA translation represents an attractive cancer therapy. Here, we show that therapeutically targeting the RNA helicase eIF4A with zotatifin, the first-in-class eIF4A inhibitor, exerts pleiotropic effects on both tumor cells and the tumor immune microenvironment in a diverse cohort of syngeneic triple-negative breast cancer (TNBC) mouse models. Zotatifin not only suppresses tumor cell proliferation but also directly repolarizes macrophages toward an M1-like phenotype and inhibits neutrophil infiltration, which sensitizes tumors to immune checkpoint blockade. Mechanistic studies revealed that zotatifin reprograms the tumor translational landscape, inhibits the translation of Sox4 and Fgfr1, and induces an interferon (IFN) response uniformly across models. The induction of an IFN response is partially due to the inhibition of Sox4 translation by zotatifin. A similar induction of IFN-stimulated genes was observed in breast cancer patient biopsies following zotatifin treatment. Surprisingly, zotatifin significantly synergizes with carboplatin to trigger DNA damage and an even heightened IFN response, resulting in T cell-dependent tumor suppression. These studies identified a vulnerability of eIF4A in TNBC, potential pharmacodynamic biomarkers for zotatifin, and provide a rationale for new combination regimens consisting of zotatifin and chemotherapy or immunotherapy as treatments for TNBC.


Subject(s)
Antineoplastic Agents , Triple Negative Breast Neoplasms , Animals , Mice , Humans , Interferons , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Cell Proliferation , Tumor Microenvironment
5.
bioRxiv ; 2023 Sep 28.
Article in English | MEDLINE | ID: mdl-37808840

ABSTRACT

Protein synthesis is frequently dysregulated in cancer and selective inhibition of mRNA translation represents an attractive cancer therapy. Here, we show that therapeutically targeting the RNA helicase eIF4A by Zotatifin, the first-in-class eIF4A inhibitor, exerts pleiotropic effects on both tumor cells and the tumor immune microenvironment in a diverse cohort of syngeneic triple-negative breast cancer (TNBC) mouse models. Zotatifin not only suppresses tumor cell proliferation but also directly repolarizes macrophages towards an M1-like phenotype and inhibits neutrophil infiltration, which sensitizes tumors to immune checkpoint blockade. Mechanistic studies revealed that Zotatifin reprograms the tumor translational landscape, inhibits the translation of Sox4 and Fgfr1, and induces an interferon response uniformly across models. The induction of an interferon response is partially due to the inhibition of Sox4 translation by Zotatifin. A similar induction of interferon-stimulated genes was observed in breast cancer patient biopsies following Zotatifin treatment. Surprisingly, Zotatifin significantly synergizes with carboplatin to trigger DNA damage and an even heightened interferon response resulting in T cell-dependent tumor suppression. These studies identified a vulnerability of eIF4A in TNBC, potential pharmacodynamic biomarkers for Zotatifin, and provide a rationale for new combination regimens comprising Zotatifin and chemotherapy or immunotherapy as treatments for TNBC.

6.
Cancer Immunol Res ; 11(11): 1462-1479, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37603945

ABSTRACT

Long noncoding RNAs (lncRNA) play an important role in gene regulation in both normal tissues and cancer. Targeting lncRNAs is a promising therapeutic approach that has become feasible through the development of gapmer antisense oligonucleotides (ASO). Metastasis-associated lung adenocarcinoma transcript (Malat1) is an abundant lncRNA whose expression is upregulated in several cancers. Although Malat1 increases the migratory and invasive properties of tumor cells, its role in the tumor microenvironment (TME) is still not well defined. We explored the connection between Malat1 and the tumor immune microenvironment (TIME) using several immune-competent preclinical syngeneic Tp53-null triple-negative breast cancer (TNBC) mouse models that mimic the heterogeneity and immunosuppressive TME found in human breast cancer. Using a Malat1 ASO, we were able to knockdown Malat1 RNA expression resulting in a delay in primary tumor growth, decreased proliferation, and increased apoptosis. In addition, immunophenotyping of tumor-infiltrating lymphocytes revealed that Malat1 inhibition altered the TIME, with a decrease in immunosuppressive tumor-associated macrophages (TAM) and myeloid-derived suppressor cells (MDSC) as well as an increase in cytotoxic CD8+ T cells. Malat1 depletion in tumor cells, TAMs, and MDSCs decreased immunosuppressive cytokine/chemokine secretion whereas Malat1 inhibition in T cells increased inflammatory secretions and T-cell proliferation. Combination of a Malat1 ASO with chemotherapy or immune checkpoint blockade (ICB) improved the treatment responses in a preclinical model. These studies highlight the immunostimulatory effects of Malat1 inhibition in TNBC, the benefit of a Malat1 ASO therapeutic, and its potential use in combination with chemotherapies and immunotherapies.


Subject(s)
Adenocarcinoma , RNA, Long Noncoding , Triple Negative Breast Neoplasms , Humans , Animals , Mice , RNA, Long Noncoding/genetics , Triple Negative Breast Neoplasms/metabolism , Tumor Microenvironment , Gene Expression Regulation, Neoplastic , Cell Proliferation/physiology , Adenocarcinoma/genetics , Cell Line, Tumor
7.
Healthcare (Basel) ; 11(13)2023 Jun 28.
Article in English | MEDLINE | ID: mdl-37444709

ABSTRACT

As cognitive function is critical for muscle coordination, cognitive training may also improve neuromuscular control strategy and knee function following an anterior cruciate ligament reconstruction (ACLR). The purpose of this case-control study was to examine the effects of cognitive training on joint stiffness regulation in response to negative visual stimuli and knee function following ACLR. A total of 20 ACLR patients and 20 healthy controls received four weeks of online cognitive training. Executive function, joint stiffness in response to emotionally evocative visual stimuli (neutral, fearful, knee injury related), and knee function outcomes before and after the intervention were compared. Both groups improved executive function following the intervention (p = 0.005). The ACLR group had greater mid-range stiffness in response to fearful (p = 0.024) and injury-related pictures (p = 0.017) than neutral contents before the intervention, while no post-intervention stiffness differences were observed among picture types. The ACLR group showed better single-legged hop for distance after cognitive training (p = 0.047), while the healthy group demonstrated no improvement. Cognitive training enhanced executive function, which may reduce joint stiffness dysregulation in response to emotionally arousing images and improve knee function in ACLR patients, presumably by facilitating neural processing necessary for neuromuscular control.

8.
Am J Physiol Cell Physiol ; 324(3): C707-C717, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36717100

ABSTRACT

A better understanding of the mechanisms regulating cancer metastasis is critical to develop new therapies and decrease mortality. Emerging evidence suggests that the interactions between tumor cells and the host immune system play important roles in establishing metastasis. Tumor cells are able to recruit immune cells, which in turn promotes tumor cell invasion, intravasation, survival in circulation, extravasation, and colonization in different organs. The tumor-host immunological interactions also generate a premetastatic niche in distant organs which facilitates metastasis. In this review, we summarize the recent findings on how tumor cells and immune cells regulate each other to coevolve and promote the formation of metastases at the major organ sites of metastasis.


Subject(s)
Ecosystem , Neoplasms , Humans , Neoplasms/pathology , Neoplastic Stem Cells/pathology , Neoplasm Metastasis/pathology , Tumor Microenvironment/physiology
9.
Cancer Immunol Res ; 11(2): 171-183, 2023 02 03.
Article in English | MEDLINE | ID: mdl-36484736

ABSTRACT

Vα24-invariant natural killer T cells (NKT) possess innate antitumor properties that can be exploited for cancer immunotherapy. We have shown previously that the CD62L+ central memory-like subset of these cells drives the in vivo antitumor activity of NKTs, but molecular mediators of NKT central memory differentiation remain unknown. Here, we demonstrate that relative to CD62L- cells, CD62L+ NKTs express a higher level of the gene encoding the Wnt/ß-catenin transcription factor lymphoid enhancer binding factor 1 (LEF1) and maintain active Wnt/ß-catenin signaling. CRISPR/Cas9-mediated LEF1 knockout reduced CD62L+ frequency after antigenic stimulation, whereas Wnt/ß-catenin activator Wnt3a ligand increased CD62L+ frequency. LEF1 overexpression promoted NKT expansion and limited exhaustion following serial tumor challenge and was sufficient to induce a central memory-like transcriptional program in NKTs. In mice, NKTs expressing a GD2-specific chimeric-antigen receptor (CAR) with LEF1 demonstrated superior control of neuroblastoma xenograft tumors compared with control CAR-NKTs. These results identify LEF1 as a transcriptional activator of the NKT central memory program and advance development of NKT cell-based immunotherapy. See related Spotlight by Van Kaer, p. 144.


Subject(s)
Natural Killer T-Cells , Receptors, Chimeric Antigen , Humans , Animals , Mice , Natural Killer T-Cells/immunology , beta Catenin , Lymphoid Enhancer-Binding Factor 1/genetics , Lymphocyte Activation/immunology
10.
Semin Cancer Biol ; 87: 17-31, 2022 12.
Article in English | MEDLINE | ID: mdl-36354098

ABSTRACT

Metastatic cancer is almost always terminal, and more than 90% of cancer deaths result from metastatic disease. Combating cancer metastasis and post-therapeutic recurrence successfully requires understanding each step of metastatic progression. This review describes the current state of knowledge of the etiology and mechanism of cancer progression from primary tumor growth to the formation of new tumors in other parts of the body. Open questions, avenues for future research, and therapeutic approaches with the potential to prevent or inhibit metastasis through personalization to each patient's mutation and/or immune profile are also highlighted.


Subject(s)
Epithelial-Mesenchymal Transition , Neoplasms , Humans , Epithelial-Mesenchymal Transition/genetics , Neoplasms/genetics , Neoplasms/pathology , Neoplasm Metastasis
11.
Oncogene ; 41(48): 5214-5222, 2022 11.
Article in English | MEDLINE | ID: mdl-36261627

ABSTRACT

Signal transducer and activator of transcription 5 (STAT5) promotes cell survival and instigates breast tumor formation, and in the normal breast it also drives alveolar differentiation and lactogenesis. However, whether STAT5 drives a differentiated phenotype in breast tumorigenesis and therefore impacts cancer spread and metastasis is unclear. We found in two genetically engineered mouse models of breast cancer that constitutively activated Stat5a (Stat5aca) caused precancerous mammary epithelial cells to become lactogenic and evolve into tumors with diminished potential to metastasize. We also showed that STAT5aca reduced the migratory and invasive ability of human breast cancer cell lines in vitro. Furthermore, we demonstrated that STAT5aca overexpression in human breast cancer cells lowered their metastatic burden in xenografted mice. Moreover, RPPA, Western blotting, and studies of ChIPseq data identified several EMT drivers regulated by STAT5. In addition, bioinformatic studies detected a correlation between STAT5 activity and better prognosis of breast cancer patients. Together, we conclude that STAT5 activation during mammary tumorigenesis specifies a tumor phenotype of lactogenic differentiation, suppresses EMT, and diminishes potential for subsequent metastasis.


Subject(s)
Breast Neoplasms , STAT5 Transcription Factor , Animals , Female , Humans , Mice , Breast/pathology , Breast Neoplasms/pathology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Epithelial Cells/metabolism , Mammary Glands, Animal/pathology , STAT5 Transcription Factor/metabolism
12.
Front Syst Neurosci ; 16: 727054, 2022.
Article in English | MEDLINE | ID: mdl-35993088

ABSTRACT

Hyperexcitability in fear circuits is suggested to be important for development of pathological anxiety and trauma from adaptive mechanisms of fear. Hyperexcitability is proposed to be due to acquired sensitization in fear circuits that progressively becomes more severe over time causing changing symptoms in early and late pathology. We use the metaphor and mechanisms of kindling to examine gains and losses in function of one excitatory and one inhibitory neuropeptide, corticotrophin releasing factor and somatostatin, respectively, to explore this sensitization hypothesis. We suggest amygdala kindling induced hyperexcitability, hyper-inhibition and loss of inhibition provide clues to mechanisms for hyperexcitability and progressive changes in function initiated by stress and trauma.

13.
Mol Biol Cell ; 33(11): ar103, 2022 09 15.
Article in English | MEDLINE | ID: mdl-36001375

ABSTRACT

Cell-extracellular matrix (ECM) interactions represent fundamental exchanges during tumor progression, yet how particular signal-transduction factors prompt the conversion of tumor cells into migratory populations capable of systemic spread during metastasis remains elusive. We demonstrate that the noncanonical Wnt receptor, Ror2, regulates tumor cell-driven matrix remodeling and invasion in breast cancer. Ror2 loss-of-function (LOF) triggers the disruption of E-cadherin within tumor cells, accompanied by an increase in tumor cell invasion and collagen realignment in three-dimensional cultures. RNA sequencing of Ror2-deficient organoids further uncovered alterations in actin cytoskeleton, cell adhesion, and collagen cross-linking gene expression programs. Spatially, we pinpoint the up-regulation and redistribution of α5 and ß3 integrins together with the production of fibronectin in areas of invasion downstream of Ror2 loss. Wnt/ß-catenin-dependent and Wnt/Ror2 alternative Wnt signaling appear to regulate distinct functions for tumor cells regarding their ability to modify cell-ECM exchanges during invasion. Furthermore, blocking either integrin or focal adhesion kinase (FAK), a downstream mediator of integrin-mediated signal transduction, abrogates the enhanced migration observed upon Ror2 loss. These results reveal a critical function for the alternative Wnt receptor, Ror2, as a determinant of tumor cell-driven ECM exchanges during cancer invasion and metastasis.


Subject(s)
Breast Neoplasms , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Cell Adhesion , Cell Line, Tumor , Cell-Matrix Junctions , Female , Humans , Integrins , Wnt Signaling Pathway
14.
Cancer Res ; 82(12): 2281-2297, 2022 06 15.
Article in English | MEDLINE | ID: mdl-35442423

ABSTRACT

Immunosuppressive elements within the tumor microenvironment, such as tumor-associated macrophages (TAM), can present a barrier to successful antitumor responses by cytolytic T cells. Here we employed preclinical syngeneic p53 null mouse models of triple-negative breast cancer (TNBC) to develop a treatment regimen that harnessed the immunostimulatory effects of low-dose cyclophosphamide coupled with the pharmacologic inhibition of TAMs using either a small-molecule CSF1R inhibitor or an anti-CSF1R antibody. This therapeutic combination was effective in treating several highly aggressive TNBC murine mammary tumor and lung metastasis models. Single-cell RNA sequencing characterized tumor-infiltrating lymphocytes including Th cells and antigen-presenting B cells that were highly enriched in responders to combination therapy. In one model that exhibited long-term posttreatment tumor regression, high-dimensional imaging techniques identified the close spatial localization of B220+/CD86+-activated B cells and CD4+ T cells in tertiary lymphoid structures that were present up to 6 weeks posttreatment. The transcriptional and metabolic heterogeneity of TAMs was also characterized in two closely related claudin-low/mesenchymal subtype tumor models with differential treatment responses. A murine TAM signature derived from the T12 model was highly conserved in human claudin-low breast cancers, and high expression of the TAM signature correlated with reduced overall survival in patients with breast cancer. This TAM signature may help identify human patients with claudin-low breast cancer that will benefit from the combination of cyclophosphamide and anti-CSF1R therapy. These studies illustrate the complexity of the tumor immune microenvironment and highlight different immune responses that result from rational immunotherapy combinations. SIGNIFICANCE: Immunostimulatory chemotherapy combined with pharmacologic inhibition of TAMs results in durable treatment responses elicited by Th cells and B cells in claudin-low TNBC models.


Subject(s)
Triple Negative Breast Neoplasms , Animals , B-Lymphocytes , Claudins/metabolism , Claudins/therapeutic use , Cyclophosphamide/pharmacology , Cyclophosphamide/therapeutic use , Humans , Macrophages/metabolism , Mice , T-Lymphocytes, Cytotoxic/pathology , Triple Negative Breast Neoplasms/pathology , Tumor Microenvironment
15.
Cancer Res ; 82(5): 885-899, 2022 Mar 01.
Article in English | MEDLINE | ID: mdl-34965936

ABSTRACT

Tumor dormancy is a stage in which residual cancer cells remain inactive, but regrowth of dormant cancer cells contributes to recurrence. The complex ecosystem in cancer that promotes cell survival and the factors that eventually overcome growth constraints and result in proliferation remain to be fully elucidated. Doing so may provide new insights and help identify novel strategies to prolong cancer dormancy and prevent disease recurrence. To dissect the molecular pathways and the microenvironments involved in regulation of dormancy, we utilized a novel immunocompetent transgenic model to study minimal residual disease and relapse. This model revealed a significant reorganization of cancer cell structures, stroma, and immune cells, with cancer cells showing dormant cell signatures. Single-cell RNA sequencing uncovered remodeling of myeloid and lymphoid compartments. In addition, the Jagged-1/Notch signaling pathway was shown to regulate many aspects of tumorigenesis, including stem cell development, epithelial-to-mesenchymal transition, and immune cell homeostasis during minimal residual disease. Treatment with an anti-Jagged-1 antibody inhibited the Jagged-1/Notch signaling pathway in tumor cells and the microenvironment, delaying tumor recurrence. These findings uncover a cascade of regulatory changes in the microenvironment during dormancy and identify a therapeutic strategy to undercut these changes. SIGNIFICANCE: Single-cell RNA-sequencing analysis reveals dormancy-associated changes in immune and stromal cells and demonstrates a rationale to pursue Jagged-1/Notch pathway inhibition as a viable therapeutic strategy to reduce disease recurrence.


Subject(s)
Ecosystem , Single-Cell Analysis , Humans , Jagged-1 Protein/genetics , Jagged-1 Protein/metabolism , Neoplasm Recurrence, Local/pathology , Neoplasm, Residual/genetics , Signal Transduction/physiology , Tumor Microenvironment/physiology
16.
Semin Cancer Biol ; 82: 3-10, 2022 07.
Article in English | MEDLINE | ID: mdl-34274486

ABSTRACT

Breast cancer ecosystems are composed of complex cell types, including tumor, stromal and immune cells, each of which can assume diverse phenotypes. Both the heterogeneous composition and spatially distinct tumor microenvironment impact breast cancer progression, treatment response and therapeutic resistance. Thus, a deeper understanding of breast cancer heterogeneity may help facilitate the development of novel therapies and improve outcomes for patients. The advent of paradigm shifting single-cell analysis and spatial pathologies allows for a comprehensive analysis of the tumor ecosystem as well as the interactions between its components at unprecedented resolution. In this review, we discuss the insights gained through single-cell analysis and spatial pathologies on breast cancer heterogeneity.


Subject(s)
Breast Neoplasms , Single-Cell Analysis , Breast Neoplasms/genetics , Breast Neoplasms/therapy , Ecosystem , Female , Humans , Tumor Microenvironment
17.
Sci Transl Med ; 13(617): eabe6201, 2021 Oct 27.
Article in English | MEDLINE | ID: mdl-34705519

ABSTRACT

Treatment with immune checkpoint blockade (ICB) has resulted in durable responses for a subset of patients with cancer, with predictive biomarkers for ICB response originally identified largely in the context of hypermutated cancers. Although recent clinical data have demonstrated clinical responses to ICB in certain patients with nonhypermutated cancers, previously established ICB response biomarkers have failed to accurately identify which of these patients may benefit from ICB. Here, we demonstrated that a replication stress response (RSR) defect gene expression signature, but not other proposed biomarkers, is associated with ICB response in 12 independent cohorts of patients with nonhypermutated cancer across seven tumor types, including those of the breast, prostate, kidney, and brain. Induction or suppression of RSR deficiencies was sufficient to modulate response to ICB in preclinical models of breast and renal cancers. Mechanistically, we found that despite robust activation of checkpoint kinase 1 signaling in RSR-deficient cancer cells, aberrant replication origin firing caused exhaustion of replication protein A, resulting in accumulation of immunostimulatory cytosolic DNA. We further found that deficient RSR coincided with increased intratumoral dendritic cells in both mouse cancer models and human tumors. Together, this work demonstrates that the RSR defect gene signature can accurately identify patients who may benefit from ICB across numerous nonhypermutated tumor types, and pharmacological induction of RSR defects may further expand the benefits of ICB to more patients.


Subject(s)
Immune Checkpoint Inhibitors , Neoplasms , Humans , Neoplasms/drug therapy
18.
J Long Term Eff Med Implants ; 31(3): 45-50, 2021.
Article in English | MEDLINE | ID: mdl-34369721

ABSTRACT

BACKGROUND: The primary objective of this study was to determine the effect of single versus multiple rounds of intra-articular hyaluronic acid (IA-HA) in delaying the need for total knee arthroplasty (TKA) in patients with knee OA, and if additional benefits were seen when used in conjunction with other multimodal treatment options. METHODS: This study was a retrospective claims analysis of a large commercial database containing more than 100 million patients with continuous coverage from October 1, 2010 through September 30, 2015. Time to TKA for patients who received one course of Euflexxa (IA-BioHA) were compared to patients who received two or more courses of IA-BioHA and patients who received no IA-HA. Assessment of multimodal treatment effects was done between the following groups: IA-BioHA injections alone, IA-BioHA and bracing, IA-BioHA and corticosteroid injection, and IA-BioHA with both corticosteroids and bracing. RESULTS: A total of 26,727 patients were included in the analysis of treatment courses, and 31,034 in the analysis of multimodal treatment combinations. The use of IA-BioHA demonstrated a delay of TKA that was prolonged with repeated courses of treatment (1.411 years, interquartile range [IQR]: 1.44). The greatest delay to TKA was observed for the patients who had received all three treatment options (1.5 years, IQR: 1.52) in the multimodal analysis. CONCLUSIONS: These results confirm that treatment of knee OA should consider the use of multimodal therapy instead of focusing on individual treatment options. Additionally, the use of repeated courses of IA-BioHA should be considered for prolonged benefit for patients with symptomatic knee OA.


Subject(s)
Arthroplasty, Replacement, Knee , Osteoarthritis, Knee , Humans , Hyaluronic Acid/therapeutic use , Injections, Intra-Articular , Molecular Weight , Osteoarthritis, Knee/drug therapy , Osteoarthritis, Knee/surgery , Retrospective Studies
19.
Nat Commun ; 12(1): 4262, 2021 07 12.
Article in English | MEDLINE | ID: mdl-34253738

ABSTRACT

The epithelial-mesenchymal transition (EMT) has been implicated in conferring stem cell properties and therapeutic resistance to cancer cells. Therefore, identification of drugs that can reprogram EMT may provide new therapeutic strategies. Here, we report that cells derived from claudin-low mammary tumors, a mesenchymal subtype of triple-negative breast cancer, exhibit a distinctive organoid structure with extended "spikes" in 3D matrices. Upon a miR-200 induced mesenchymal-epithelial transition (MET), the organoids switch to a smoother round morphology. Based on these observations, we developed a morphological screening method with accompanying analytical pipelines that leverage deep neural networks and nearest neighborhood classification to screen for EMT-reversing drugs. Through screening of a targeted epigenetic drug library, we identified multiple class I HDAC inhibitors and Bromodomain inhibitors that reverse EMT. These data support the use of morphological screening of mesenchymal mammary tumor organoids as a platform to identify drugs that reverse EMT.


Subject(s)
Antineoplastic Agents/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Mammary Neoplasms, Animal/pathology , Mesoderm/pathology , Organoids/pathology , Animals , Azacitidine/pharmacology , Benzamides/pharmacology , Drug Screening Assays, Antitumor , Epigenesis, Genetic , Female , Gene Expression Regulation, Neoplastic , Image Processing, Computer-Assisted , Mammary Neoplasms, Animal/genetics , Mice, Inbred BALB C , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasm Proteins/metabolism , Organoids/drug effects , Pyrimidines/pharmacology , Reproducibility of Results , Small Molecule Libraries/pharmacology
20.
Article in English | MEDLINE | ID: mdl-33814937

ABSTRACT

BACKGROUND: Total knee arthroplasty (TKA) is a surgical treatment for patients with knee osteoarthritis (KOA) that no longer experience symptom relief from non-operative or pharmacologic treatments. Non-operative KOA management aims to address patient symptoms and improve function, as well as forestall or mitigate the large costs associated with TKA. The primary objective of this study was to examine the relationship between intra-articular hyaluronic acid (IA-HA) treatment and delaying TKA in patients with KOA compared to patients not receiving IA-HA, as well as to identify differences in KOA-related costs incurred among patients who received or did not receive IA-HA. METHODS: This was a retrospective analysis of an administrative claims database from October 1st, 2010 through September 30th, 2015. Kaplan-Meier survival analysis was conducted to determine the TKA-free survival of patients who received IA-HA, stratified by the number of injection courses received versus those who did not receive any IA-HA. Median KOA-related costs per year were calculated for 2 comparisons: (1) patients who received IA-HA versus patients who did not receive IA-HA, among patients who eventually had TKA, and (2) patients who received IA-HA versus patients who did not receive IA-HA, among patients who did not have TKA. RESULTS: A total of 744 734 patients were included in the analysis. A delay to TKA was observed after IA-HA treatment for patients treated with IA-HA compared to those who did not receive IA-HA. At 1 year, the TKA-free survival was 85.8% (95% CI: 85.6%-86.0%) for patients who received IA-HA and 74.1% (95% CI: 74.0%-74.3%) for those who did not receive IA-HA. At 2 years, the TKA free survival was 70.8% (70.5%-71.1%) and 63.7% (63.5%-63.9%) in the 2 groups, respectively. Patients treated with multiple courses of IA-HA demonstrated an incremental increase in delay to TKA with more courses of IA-HA, suggesting that the risk of TKA over the study time period is reduced with additional IA-HA courses. The hazard ratio for the need of TKA was 0.85 (95% CI 0.84-0.86) for a single course and 0.27 (95% CI 0.25-0.28) for ⩾5 courses, both compared to the no IA-HA group. In patients that eventually had TKA, the median KOA-related costs were lower among those who received IA-HA before their TKA ($860.24, 95% CI: 446.65-1722.20), compared to those who did not receive IA-HA ($2659.49, 95% CI: 891.04-7480.38). For patients who did not have TKA, the median and interquartile range (IQR) KOA-related costs per year were similar for patients who received IA-HA compared with those who did not. CONCLUSION: These results demonstrate that within a large cohort of KOA patients, individuals who received multiple courses of IA-HA had a progressively greater delay to TKA compared to patients who did not receive IA-HA treatment. Also, for patients who progressed to TKA, IA-HA treatment was associated with a large reduction in KOA-related healthcare costs. Based on these results, multiple, repeat courses of IA-HA may be beneficial in substantially delaying TKA in KOA patients, as well as minimizing KOA-related healthcare costs.

SELECTION OF CITATIONS
SEARCH DETAIL
...