Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Sci Adv ; 10(27): eadl1197, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38959305

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is characterized by increasing fibrosis, which can enhance tumor progression and spread. Here, we undertook an unbiased temporal assessment of the matrisome of the highly metastatic KPC (Pdx1-Cre, LSL-KrasG12D/+, LSL-Trp53R172H/+) and poorly metastatic KPflC (Pdx1-Cre, LSL-KrasG12D/+, Trp53fl/+) genetically engineered mouse models of pancreatic cancer using mass spectrometry proteomics. Our assessment at early-, mid-, and late-stage disease reveals an increased abundance of nidogen-2 (NID2) in the KPC model compared to KPflC, with further validation showing that NID2 is primarily expressed by cancer-associated fibroblasts (CAFs). Using biomechanical assessments, second harmonic generation imaging, and birefringence analysis, we show that NID2 reduction by CRISPR interference (CRISPRi) in CAFs reduces stiffness and matrix remodeling in three-dimensional models, leading to impaired cancer cell invasion. Intravital imaging revealed improved vascular patency in live NID2-depleted tumors, with enhanced response to gemcitabine/Abraxane. In orthotopic models, NID2 CRISPRi tumors had less liver metastasis and increased survival, highlighting NID2 as a potential PDAC cotarget.


Subject(s)
Carcinoma, Pancreatic Ductal , Fibrosis , Pancreatic Neoplasms , Proteomics , Animals , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/genetics , Proteomics/methods , Mice , Humans , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/genetics , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Disease Models, Animal , Cell Line, Tumor , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/genetics , Gemcitabine , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Cell Adhesion Molecules
2.
Adv Sci (Weinh) ; 11(23): e2307963, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38602451

ABSTRACT

In recent decades, the role of tumor biomechanics on cancer cell behavior at the primary site has been increasingly appreciated. However, the effect of primary tumor biomechanics on the latter stages of the metastatic cascade, such as metastatic seeding of secondary sites and outgrowth remains underappreciated. This work sought to address this in the context of triple negative breast cancer (TNBC), a cancer type known to aggressively disseminate at all stages of disease progression. Using mechanically tuneable model systems, mimicking the range of stiffness's typically found within breast tumors, it is found that, contrary to expectations, cancer cells exposed to softer microenvironments are more able to colonize secondary tissues. It is shown that heightened cell survival is driven by enhanced metabolism of fatty acids within TNBC cells exposed to softer microenvironments. It is demonstrated that uncoupling cellular mechanosensing through integrin ß1 blocking antibody effectively causes stiff primed TNBC cells to behave like their soft counterparts, both in vitro and in vivo. This work is the first to show that softer tumor microenvironments may be contributing to changes in disease outcome by imprinting on TNBC cells a greater metabolic flexibility and conferring discrete cell survival advantages.


Subject(s)
Fatty Acids , Triple Negative Breast Neoplasms , Tumor Microenvironment , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Humans , Female , Fatty Acids/metabolism , Mice , Cell Line, Tumor , Animals , Biomechanical Phenomena , Disease Models, Animal , Neoplasm Metastasis
3.
Nat Cancer ; 4(9): 1326-1344, 2023 09.
Article in English | MEDLINE | ID: mdl-37640930

ABSTRACT

The lysyl oxidase family represents a promising target in stromal targeting of solid tumors due to the importance of this family in crosslinking and stabilizing fibrillar collagens and its known role in tumor desmoplasia. Using small-molecule drug-design approaches, we generated and validated PXS-5505, a first-in-class highly selective and potent pan-lysyl oxidase inhibitor. We demonstrate in vitro and in vivo that pan-lysyl oxidase inhibition decreases chemotherapy-induced pancreatic tumor desmoplasia and stiffness, reduces cancer cell invasion and metastasis, improves tumor perfusion and enhances the efficacy of chemotherapy in the autochthonous genetically engineered KPC model, while also demonstrating antifibrotic effects in human patient-derived xenograft models of pancreatic cancer. PXS-5505 is orally bioavailable, safe and effective at inhibiting lysyl oxidase activity in tissues. Our findings present the rationale for progression of a pan-lysyl oxidase inhibitor aimed at eliciting a reduction in stromal matrix to potentiate chemotherapy in pancreatic ductal adenocarcinoma.


Subject(s)
Pancreatic Diseases , Pancreatic Neoplasms , Humans , Gemcitabine , Protein-Lysine 6-Oxidase , Pancreatic Neoplasms/drug therapy
4.
Nat Commun ; 13(1): 4587, 2022 08 06.
Article in English | MEDLINE | ID: mdl-35933466

ABSTRACT

The tumour stroma, and in particular the extracellular matrix (ECM), is a salient feature of solid tumours that plays a crucial role in shaping their progression. Many desmoplastic tumours including breast cancer involve the significant accumulation of type I collagen. However, recently it has become clear that the precise distribution and organisation of matrix molecules such as collagen I is equally as important in the tumour as their abundance. Cancer-associated fibroblasts (CAFs) coexist within breast cancer tissues and play both pro- and anti-tumourigenic roles through remodelling the ECM. Here, using temporal proteomic profiling of decellularized tumours, we interrogate the evolving matrisome during breast cancer progression. We identify 4 key matrisomal clusters, and pinpoint collagen type XII as a critical component that regulates collagen type I organisation. Through combining our proteomics with single-cell transcriptomics, and genetic manipulation models, we show how CAF-secreted collagen XII alters collagen I organisation to create a pro-invasive microenvironment supporting metastatic dissemination. Finally, we show in patient cohorts that collagen XII may represent an indicator of breast cancer patients at high risk of metastatic relapse.


Subject(s)
Breast Neoplasms , Collagen Type XII/metabolism , Neoplasm Metastasis , Tumor Microenvironment , Breast Neoplasms/pathology , Collagen , Collagen Type I , Extracellular Matrix/pathology , Female , Humans , Neoplasm Metastasis/pathology , Neoplasm Recurrence, Local/pathology , Proteomics
5.
Cell Rep ; 36(11): 109689, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34525350

ABSTRACT

Assessing drug response within live native tissue provides increased fidelity with regards to optimizing efficacy while minimizing off-target effects. Here, using longitudinal intravital imaging of a Rac1-Förster resonance energy transfer (FRET) biosensor mouse coupled with in vivo photoswitching to track intratumoral movement, we help guide treatment scheduling in a live breast cancer setting to impair metastatic progression. We uncover altered Rac1 activity at the center versus invasive border of tumors and demonstrate enhanced Rac1 activity of cells in close proximity to live tumor vasculature using optical window imaging. We further reveal that Rac1 inhibition can enhance tumor cell vulnerability to fluid-flow-induced shear stress and therefore improves overall anti-metastatic response to therapy during transit to secondary sites such as the lung. Collectively, this study demonstrates the utility of single-cell intravital imaging in vivo to demonstrate that Rac1 inhibition can reduce tumor progression and metastases in an autochthonous setting to improve overall survival.


Subject(s)
Biosensing Techniques/methods , Breast Neoplasms/pathology , rac1 GTP-Binding Protein/metabolism , Aminoquinolines/pharmacology , Animals , Breast Neoplasms/diagnostic imaging , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Female , Fluorescence Resonance Energy Transfer , Humans , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Mice , Mice, Inbred BALB C , Pyrimidines/pharmacology , Shear Strength , Signal Transduction , rac1 GTP-Binding Protein/antagonists & inhibitors
6.
Sci Adv ; 7(40): eabh0363, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34586840

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a highly metastatic, chemoresistant malignancy and is characterized by a dense, desmoplastic stroma that modulates PDAC progression. Here, we visualized transient manipulation of focal adhesion kinase (FAK), which integrates bidirectional cell-environment signaling, using intravital fluorescence lifetime imaging microscopy of the FAK-based Förster resonance energy transfer biosensor in mouse and patient-derived PDAC models. Parallel real-time quantification of the FUCCI cell cycle reporter guided us to improve PDAC response to standard-of-care chemotherapy at primary and secondary sites. Critically, micropatterned pillar plates and stiffness-tunable matrices were used to pinpoint the contribution of environmental cues to chemosensitization, while fluid flow­induced shear stress assessment, patient-derived matrices, and personalized in vivo models allowed us to deconstruct how FAK inhibition can reduce PDAC spread. Last, stratification of PDAC patient samples via Merlin status revealed a patient subset with poor prognosis that are likely to respond to FAK priming before chemotherapy.

7.
Cancer Rep (Hoboken) ; 3(1): e1209, 2020 02.
Article in English | MEDLINE | ID: mdl-32671954

ABSTRACT

BACKGROUND: The use of in vitro cell cultures is a powerful tool for obtaining key insights into the behaviour and response of cells to interventions in normal and disease situations. Unlike in vivo settings, in vitro experiments allow a fine-tuned control of a range of microenvironmental elements independently within an isolated setting. The recent expansion in the use of three-dimensional (3D) in vitro assays has created a number of representative tools to study cell behaviour in a more physiologically 3D relevant microenvironment. Complex 3D in vitro models that can recapitulate human tissue biology are essential for understanding the pathophysiology of disease. AIM: The development of the 3D coculture collagen contraction and invasion assay, the "organotypic assay," has been widely adopted as a powerful approach to bridge the gap between standard two-dimensional tissue culture and in vivo mouse models. In the cancer setting, these assays can then be used to dissect how stromal cells, such as cancer-associated fibroblasts (CAFs), drive extracellular matrix (ECM) remodelling to alter cancer cell behaviour and response to intervention. However, to date, many of the published organotypic protocols are low-throughput, time-consuming (up to several weeks), and work-intensive with often limited scalability. Our aim was to develop a fast, high-throughput, scalable 3D organotypic assay for use in oncology screening and drug development. METHODS AND RESULTS: Here, we describe a modified 96-well organotypic assay, the "Mini-Organo," which can be easily completed within 5 days. We demonstrate its application in a wide range of mouse and human cancer biology approaches including evaluation of stromal cell 3D ECM remodelling, 3D cancer cell invasion, and the assessment of efficacy of potential anticancer therapeutic targets. Furthermore, the organotypic assay described is highly amenable to customisation using different cell types under diverse experimental conditions. CONCLUSIONS: The Mini-Organo high-throughput 3D organotypic assay allows the rapid screening of potential cancer therapeutics in human and mouse models in a time-efficient manner.


Subject(s)
Coculture Techniques/methods , Drug Development/methods , Drug Screening Assays, Antitumor/methods , Animals , Cancer-Associated Fibroblasts/physiology , High-Throughput Screening Assays , Humans , Mice , Neoplasm Invasiveness , Rats
8.
Nat Commun ; 10(1): 3637, 2019 08 12.
Article in English | MEDLINE | ID: mdl-31406163

ABSTRACT

Heterogeneous subtypes of cancer-associated fibroblasts (CAFs) coexist within pancreatic cancer tissues and can both promote and restrain disease progression. Here, we interrogate how cancer cells harboring distinct alterations in p53 manipulate CAFs. We reveal the existence of a p53-driven hierarchy, where cancer cells with a gain-of-function (GOF) mutant p53 educate a dominant population of CAFs that establish a pro-metastatic environment for GOF and null p53 cancer cells alike. We also demonstrate that CAFs educated by null p53 cancer cells may be reprogrammed by either GOF mutant p53 cells or their CAFs. We identify perlecan as a key component of this pro-metastatic environment. Using intravital imaging, we observe that these dominant CAFs delay cancer cell response to chemotherapy. Lastly, we reveal that depleting perlecan in the stroma combined with chemotherapy prolongs mouse survival, supporting it as a potential target for anti-stromal therapies in pancreatic cancer.


Subject(s)
Cancer-Associated Fibroblasts/pathology , Drug Resistance, Neoplasm/genetics , Heparan Sulfate Proteoglycans/metabolism , Pancreatic Neoplasms/pathology , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic/genetics , Mice , Mice, Inbred BALB C , Neoplasm Invasiveness/pathology , Pancreas/pathology , Pancreatic Neoplasms/genetics , Signal Transduction/physiology , Tumor Suppressor Protein p53/genetics
9.
Cell Rep ; 23(11): 3312-3326, 2018 06 12.
Article in English | MEDLINE | ID: mdl-29898401

ABSTRACT

Application of advanced intravital imaging facilitates dynamic monitoring of pathway activity upon therapeutic inhibition. Here, we assess resistance to therapeutic inhibition of the PI3K pathway within the hypoxic microenvironment of pancreatic ductal adenocarcinoma (PDAC) and identify a phenomenon whereby pronounced hypoxia-induced resistance is observed for three clinically relevant inhibitors. To address this clinical problem, we have mapped tumor hypoxia by both immunofluorescence and phosphorescence lifetime imaging of oxygen-sensitive nanoparticles and demonstrate that these hypoxic regions move transiently around the tumor. To overlay this microenvironmental information with drug response, we applied a FRET biosensor for Akt activity, which is a key effector of the PI3K pathway. Performing dual intravital imaging of drug response in different tumor compartments, we demonstrate an improved drug response to a combination therapy using the dual mTORC1/2 inhibitor AZD2014 with the hypoxia-activated pro-drug TH-302.


Subject(s)
Drug Resistance, Neoplasm , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , Animals , Benzamides , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Drug Therapy, Combination , Female , Fluorescence Resonance Energy Transfer , Humans , Hypoxia , Intravital Microscopy/methods , Mice , Mice, Inbred BALB C , Morpholines/pharmacology , Morpholines/therapeutic use , Nanoparticles/chemistry , Nitroimidazoles/pharmacology , Nitroimidazoles/therapeutic use , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Phosphoramide Mustards/pharmacology , Phosphoramide Mustards/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism , Pyrimidines , Signal Transduction/drug effects , Transplantation, Heterologous , Tumor Microenvironment
10.
Cancer Res ; 78(2): 501-515, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29180477

ABSTRACT

Radiotherapy is essential to the treatment of most solid tumors and acquired or innate resistance to this therapeutic modality is a major clinical problem. Here we show that miR-139-5p is a potent modulator of radiotherapy response in breast cancer via its regulation of genes involved in multiple DNA repair and reactive oxygen species defense pathways. Treatment of breast cancer cells with a miR-139-5p mimic strongly synergized with radiation both in vitro and in vivo, resulting in significantly increased oxidative stress, accumulation of unrepaired DNA damage, and induction of apoptosis. Several miR-139-5p target genes were also strongly predictive of outcome in radiotherapy-treated patients across multiple independent breast cancer cohorts. These prognostically relevant miR-139-5p target genes were used as companion biomarkers to identify radioresistant breast cancer xenografts highly amenable to sensitization by cotreatment with a miR-139-5p mimetic.Significance: The microRNA described in this study offers a potentially useful predictive biomarker of radiosensitivity in solid tumors and a generally applicable druggable target for tumor radiosensitization. Cancer Res; 78(2); 501-15. ©2017 AACR.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/radiotherapy , DNA Repair/radiation effects , Gene Regulatory Networks/radiation effects , MicroRNAs/genetics , Radiation Tolerance/genetics , Reactive Oxygen Species/metabolism , Animals , Apoptosis , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Case-Control Studies , Cell Proliferation , DNA Damage/radiation effects , Female , Follow-Up Studies , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/radiotherapy , Prognosis , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
11.
Sci Rep ; 7(1): 16887, 2017 12 04.
Article in English | MEDLINE | ID: mdl-29203823

ABSTRACT

Organotypic co-cultures bridge the gap between standard two-dimensional culture and mouse models. Such assays increase the fidelity of pre-clinical studies, to better inform lead compound development and address the increasing attrition rates of lead compounds within the pharmaceutical industry, which are often a result of screening in less faithful two-dimensional models. Using large-scale acid-extraction techniques, we demonstrate a step-by-step process to isolate collagen I from commercially available animal byproducts. Using the well-established rat tail tendon collagen as a benchmark, we apply our novel kangaroo tail tendon collagen as an alternative collagen source for our screening-ready three-dimensional organotypic co-culture platform. Both collagen sources showed equal applicability for invasive, proliferative or survival assessment of well-established cancer models and clinically relevant patient-derived cancer cell lines. Additional readouts were also demonstrated when comparing these alternative collagen sources for stromal contributions to stiffness, organization and ultrastructure via atomic force microscopy, second harmonic generation imaging and scanning electron microscopy, among other vital biological readouts, where only minor differences were found between the preparations. Organotypic co-cultures represent an easy, affordable and scalable model to investigate drug responses within a physiologically relevant 3D platform.


Subject(s)
Cell Culture Techniques/methods , Collagen/chemistry , Animals , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Coculture Techniques , Collagen/isolation & purification , Extracellular Matrix/metabolism , Gefitinib/pharmacology , Humans , Macropodidae/metabolism , Mice , Microscopy, Atomic Force , Rats , Tendons/metabolism
12.
Cell Rep ; 21(1): 274-288, 2017 Oct 03.
Article in English | MEDLINE | ID: mdl-28978480

ABSTRACT

The small GTPase RhoA is involved in a variety of fundamental processes in normal tissue. Spatiotemporal control of RhoA is thought to govern mechanosensing, growth, and motility of cells, while its deregulation is associated with disease development. Here, we describe the generation of a RhoA-fluorescence resonance energy transfer (FRET) biosensor mouse and its utility for monitoring real-time activity of RhoA in a variety of native tissues in vivo. We assess changes in RhoA activity during mechanosensing of osteocytes within the bone and during neutrophil migration. We also demonstrate spatiotemporal order of RhoA activity within crypt cells of the small intestine and during different stages of mammary gestation. Subsequently, we reveal co-option of RhoA activity in both invasive breast and pancreatic cancers, and we assess drug targeting in these disease settings, illustrating the potential for utilizing this mouse to study RhoA activity in vivo in real time.


Subject(s)
Biosensing Techniques , Fluorescence Resonance Energy Transfer/methods , Intravital Microscopy/methods , Time-Lapse Imaging/methods , rho GTP-Binding Proteins/genetics , Animals , Antineoplastic Agents/pharmacology , Bone and Bones/cytology , Bone and Bones/metabolism , Cell Movement/drug effects , Dasatinib/pharmacology , Erlotinib Hydrochloride/pharmacology , Female , Fluorescence Resonance Energy Transfer/instrumentation , Gene Expression Regulation , Intestine, Small/metabolism , Intestine, Small/ultrastructure , Intravital Microscopy/instrumentation , Mammary Glands, Animal/blood supply , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/ultrastructure , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/ultrastructure , Mechanotransduction, Cellular , Mice , Mice, Transgenic , Neutrophils/metabolism , Neutrophils/ultrastructure , Osteocytes/metabolism , Osteocytes/ultrastructure , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/ultrastructure , Time-Lapse Imaging/instrumentation , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein
13.
Sci Transl Med ; 9(384)2017 04 05.
Article in English | MEDLINE | ID: mdl-28381539

ABSTRACT

The emerging standard of care for patients with inoperable pancreatic cancer is a combination of cytotoxic drugs gemcitabine and Abraxane, but patient response remains moderate. Pancreatic cancer development and metastasis occur in complex settings, with reciprocal feedback from microenvironmental cues influencing both disease progression and drug response. Little is known about how sequential dual targeting of tumor tissue tension and vasculature before chemotherapy can affect tumor response. We used intravital imaging to assess how transient manipulation of the tumor tissue, or "priming," using the pharmaceutical Rho kinase inhibitor Fasudil affects response to chemotherapy. Intravital Förster resonance energy transfer imaging of a cyclin-dependent kinase 1 biosensor to monitor the efficacy of cytotoxic drugs revealed that priming improves pancreatic cancer response to gemcitabine/Abraxane at both primary and secondary sites. Transient priming also sensitized cells to shear stress and impaired colonization efficiency and fibrotic niche remodeling within the liver, three important features of cancer spread. Last, we demonstrate a graded response to priming in stratified patient-derived tumors, indicating that fine-tuned tissue manipulation before chemotherapy may offer opportunities in both primary and metastatic targeting of pancreatic cancer.


Subject(s)
Disease Progression , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , rho-Associated Kinases/antagonists & inhibitors , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/therapeutic use , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Albumin-Bound Paclitaxel/pharmacology , Albumin-Bound Paclitaxel/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biosensing Techniques , CDC2 Protein Kinase/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Collagen/metabolism , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Extracellular Matrix/metabolism , Humans , Liver/pathology , Mice , Neoplasm Invasiveness , Neoplasm Metastasis , Signal Transduction/drug effects , Treatment Outcome , rho-Associated Kinases/metabolism , src-Family Kinases/metabolism , Gemcitabine
SELECTION OF CITATIONS
SEARCH DETAIL