Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 32
1.
Cell Rep ; 42(3): 112129, 2023 03 28.
Article En | MEDLINE | ID: mdl-36821441

TGF-ß induces senescence in embryonic tissues. Whether TGF-ß in the hypoxic tumor microenvironment (TME) induces senescence in cancer and how the ensuing senescence-associated secretory phenotype (SASP) remodels the cellular TME to influence immune checkpoint inhibitor (ICI) responses are unknown. We show that TGF-ß induces a deeper senescent state under hypoxia than under normoxia; deep senescence correlates with the degree of E2F suppression and is marked by multinucleation, reduced reentry into proliferation, and a distinct 14-gene SASP. Suppressing TGF-ß signaling in tumors in an immunocompetent mouse lung cancer model abrogates endogenous senescent cells and suppresses the 14-gene SASP and immune infiltration. Untreated human lung cancers with a high 14-gene SASP display immunosuppressive immune infiltration. In a lung cancer clinical trial of ICIs, elevated 14-gene SASP is associated with increased senescence, TGF-ß and hypoxia signaling, and poor progression-free survival. Thus, TME-induced senescence may represent a naturally occurring state in cancer, contributing to an immune-suppressive phenotype associated with immune therapy resistance.


Lung Neoplasms , Transforming Growth Factor beta , Mice , Animals , Humans , Phenotype , Disease Models, Animal , Cellular Microenvironment , Tumor Microenvironment , Cellular Senescence/physiology
2.
J Clin Invest ; 132(16)2022 08 15.
Article En | MEDLINE | ID: mdl-35708912

Aberrant expression of viral-like repeat elements is a common feature of epithelial cancers, and the substantial diversity of repeat species provides a distinct view of the cancer transcriptome. Repeatome profiling across ovarian, pancreatic, and colorectal cell lines identifies distinct clustering independent of tissue origin that is seen with coding gene analysis. Deeper analysis of ovarian cancer cell lines demonstrated that human satellite II (HSATII) satellite repeat expression was highly associated with epithelial-mesenchymal transition (EMT) and anticorrelated with IFN-response genes indicative of a more aggressive phenotype. SATII expression - and its correlation with EMT and anticorrelation with IFN-response genes - was also found in ovarian cancer RNA-Seq data and was associated with significantly shorter survival in a second independent cohort of patients with ovarian cancer. Repeat RNAs were enriched in tumor-derived extracellular vesicles capable of stimulating monocyte-derived macrophages, demonstrating a mechanism that alters the tumor microenvironment with these viral-like sequences. Targeting of HSATII with antisense locked nucleic acids stimulated IFN response and induced MHC I expression in ovarian cancer cell lines, highlighting a potential strategy of modulating the repeatome to reestablish antitumor cell immune surveillance.


Ovarian Neoplasms , RNA, Satellite , Carcinoma, Ovarian Epithelial/genetics , Cell Line, Tumor , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Ovarian Neoplasms/genetics , Phenotype , RNA , Tumor Microenvironment/genetics
3.
Clin Cancer Res ; 28(15): 3296-3307, 2022 08 02.
Article En | MEDLINE | ID: mdl-35363262

PURPOSE: Patient-derived organoids (PDO) are a promising technology to support precision medicine initiatives for patients with pancreatic ductal adenocarcinoma (PDAC). PDOs may improve clinical next-generation sequencing (NGS) and enable rapid ex vivo chemotherapeutic screening (pharmacotyping). EXPERIMENTAL DESIGN: PDOs were derived from tissues obtained during surgical resection and endoscopic biopsies and studied with NGS and pharmacotyping. PDO-specific pharmacotype is assessed prospectively as a predictive biomarker of clinical therapeutic response by leveraging data from a randomized controlled clinical trial. RESULTS: Clinical sequencing pipelines often fail to detect PDAC-associated somatic mutations in surgical specimens that demonstrate a good pathologic response to previously administered chemotherapy. Sequencing the PDOs derived from these surgical specimens, after biomass expansion, improves the detection of somatic mutations and enables quantification of copy number variants. The detection of clinically relevant mutations and structural variants is improved following PDO biomass expansion. On clinical trial, PDOs were derived from biopsies of treatment-naïve patients prior to treatment with FOLFIRINOX (FFX). Ex vivo PDO pharmacotyping with FFX components predicted clinical therapeutic response in these patients with borderline resectable or locally advanced PDAC treated in a neoadjuvant or induction paradigm. PDO pharmacotypes suggesting sensitivity to FFX components were associated with longitudinal declines of tumor marker, carbohydrate-antigen 19-9 (CA-19-9), and favorable RECIST imaging response. CONCLUSIONS: PDOs established from tissues obtained from patients previously receiving cytotoxic chemotherapies can be accomplished in a clinically certified laboratory. Sequencing PDOs following biomass expansion improves clinical sequencing quality. High in vitro sensitivity to standard-of-care chemotherapeutics predicts good clinical response to systemic chemotherapy in PDAC. See related commentary by Zhang et al., p. 3176.


Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/genetics , Biomarkers, Tumor/therapeutic use , Carcinoma, Pancreatic Ductal/diagnosis , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Humans , Organoids/pathology , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Precision Medicine , Pancreatic Neoplasms
4.
Cancer Discov ; 12(6): 1462-1481, 2022 06 02.
Article En | MEDLINE | ID: mdl-35320348

Altered RNA expression of repetitive sequences and retrotransposition are frequently seen in colorectal cancer, implicating a functional importance of repeat activity in cancer progression. We show the nucleoside reverse transcriptase inhibitor 3TC targets activities of these repeat elements in colorectal cancer preclinical models with a preferential effect in p53-mutant cell lines linked with direct binding of p53 to repeat elements. We translate these findings to a human phase II trial of single-agent 3TC treatment in metastatic colorectal cancer with demonstration of clinical benefit in 9 of 32 patients. Analysis of 3TC effects on colorectal cancer tumorspheres demonstrates accumulation of immunogenic RNA:DNA hybrids linked with induction of interferon response genes and DNA damage response. Epigenetic and DNA-damaging agents induce repeat RNAs and have enhanced cytotoxicity with 3TC. These findings identify a vulnerability in colorectal cancer by targeting the viral mimicry of repeat elements. SIGNIFICANCE: Colorectal cancers express abundant repeat elements that have a viral-like life cycle that can be therapeutically targeted with nucleoside reverse transcriptase inhibitors (NRTI) commonly used for viral diseases. NRTIs induce DNA damage and interferon response that provide a new anticancer therapeutic strategy. This article is highlighted in the In This Issue feature, p. 1397.


Colorectal Neoplasms , RNA-Directed DNA Polymerase , Animals , Antiviral Agents , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , DNA , Humans , Interferons/metabolism , Lamivudine , Life Cycle Stages , RNA , RNA-Directed DNA Polymerase/metabolism , Tumor Suppressor Protein p53/genetics
5.
Nat Cancer ; 2(11): 1124-1135, 2021 11.
Article En | MEDLINE | ID: mdl-35122060

Overcoming intrinsic resistance to immune checkpoint blockade for microsatellite stable (MSS) colorectal cancer (CRC) and pancreatic ductal adenocarcinoma (PDAC) remains challenging. We conducted a single-arm, non-randomized, phase II trial (NCT03104439) combining radiation, ipilimumab and nivolumab to treat patients with metastatic MSS CRC (n = 40) and PDAC (n = 25) with an Eastern Cooperative Oncology Group (ECOG) performance status of 0 or 1. The primary endpoint was disease control rate (DCR) by intention to treat. DCRs were 25% for CRC (ten of 40; 95% confidence interval (CI), 13-41%) and 20% for PDAC (five of 25; 95% CI, 7-41%). In the per-protocol analysis, defined as receipt of radiation, DCR was 37% (ten of 27; 95% CI, 19-58%) in CRC and 29% (five of 17; 95% CI, 10-56%) in PDAC. Pretreatment biopsies revealed low tumor mutational burden for all samples but higher numbers of natural killer (NK) cells and expression of the HERVK repeat RNA in patients with disease control. This study provides proof of concept of combining radiation with immune checkpoint blockade in immunotherapy-resistant cancers.


Adenocarcinoma , Carcinoma, Pancreatic Ductal , Colorectal Neoplasms , Pancreatic Neoplasms , Adenocarcinoma/genetics , Adenocarcinoma/therapy , Carcinoma, Pancreatic Ductal/therapy , Colorectal Neoplasms/therapy , Humans , Immune Checkpoint Inhibitors , Immunologic Factors/therapeutic use , Immunotherapy , Microsatellite Repeats/genetics , Pancreatic Neoplasms/therapy , Radiotherapy , Treatment Outcome , Pancreatic Neoplasms
6.
Am J Surg Pathol ; 45(1): 14-24, 2021 01.
Article En | MEDLINE | ID: mdl-32826529

Coronavirus disease-19 (COVID-19) is caused by a newly discovered coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Although SARS-CoV-2 is visualized on electron microscopy, there is an increasing demand for widely applicable techniques to visualize viral components within tissue specimens. Viral protein and RNA can be detected on formalin-fixed paraffin-embedded (FFPE) tissue using immunohistochemistry (IHC) and in situ hybridization (ISH), respectively. Herein, we evaluate the staining performance of ISH for SARS-CoV-2 and an IHC directed at the SARS-CoV nucleocapsid protein and compare these results to a gold standard, tissue quantitative real-time polymerase chain reaction (qRT-PCR). We evaluated FFPE sections from 8 COVID-19 autopsies, including 19 pulmonary and 39 extrapulmonary samples including the heart, liver, kidney, small intestine, skin, adipose tissue, and bone marrow. We performed RNA-ISH for SARS-CoV-2 on all cases with IHC for SARS-CoV and SARS-CoV-2 qRT-PCR performed on selected cases. Lungs from 37 autopsies performed before the COVID-19 pandemic served as negative controls. The ISH and IHC slides were reviewed by 4 observers to record a consensus opinion. Selected ISH and IHC slides were also reviewed by 4 independent observers. Evidence of SARS-CoV-2 was identified on both the IHC and ISH platforms. Within the postmortem lung, detected viral protein and RNA were often extracellular, predominantly within hyaline membranes in patients with diffuse alveolar damage. Among individual cases, there was regional variation in the amount of detectable virus in lung samples. Intracellular viral RNA and protein was localized to pneumocytes and immune cells. Viral RNA was detected on RNA-ISH in 13 of 19 (68%) pulmonary FFPE blocks from patients with COVID-19. Viral protein was detected on IHC in 8 of 9 (88%) pulmonary FFPE blocks from patients with COVID-19, although in 5 cases the stain was interpreted as equivocal. From the control cohort, FFPE blocks from all 37 patients were negative for SARS-CoV-2 RNA-ISH, whereas 5 of 13 cases were positive on IHC. Collectively, when compared with qRT-PCR on individual tissue blocks, the sensitivity and specificity for ISH was 86.7% and 100%, respectively, while those for IHC were 85.7% and 53.3%, respectively. The interobserver variability for ISH ranged from moderate to almost perfect, whereas that for IHC ranged from slight to moderate. All extrapulmonary samples from COVID-19-positive cases were negative for SARS-CoV-2 by ISH, IHC, and qRT-PCR. SARS-CoV-2 is detectable on both RNA-ISH and nucleocapsid IHC. In the lung, viral RNA and nucleocapsid protein is predominantly extracellular and within hyaline membranes in some cases, while intracellular locations are more prominent in others. The intracellular virus is detected within pneumocytes, bronchial epithelial cells, and possibly immune cells. The ISH platform is more specific, easier to analyze and the interpretation is associated with the improved interobserver agreement. ISH, IHC, and qRT-PCR failed to detect the virus in the heart, liver, and kidney.


COVID-19 Testing , COVID-19/diagnosis , Coronavirus Nucleocapsid Proteins/analysis , Immunohistochemistry , In Situ Hybridization , Lung/virology , RNA, Viral/analysis , SARS-CoV-2/chemistry , SARS-CoV-2/genetics , COVID-19/virology , Humans , Phosphoproteins/analysis , Predictive Value of Tests , Real-Time Polymerase Chain Reaction , Reproducibility of Results
7.
Nat Commun ; 11(1): 6319, 2020 12 09.
Article En | MEDLINE | ID: mdl-33298930

The relationship of SARS-CoV-2 pulmonary infection and severity of disease is not fully understood. Here we show analysis of autopsy specimens from 24 patients who succumbed to SARS-CoV-2 infection using a combination of different RNA and protein analytical platforms to characterize inter-patient and intra-patient heterogeneity of pulmonary virus infection. There is a spectrum of high and low virus cases associated with duration of disease. High viral cases have high activation of interferon pathway genes and a predominant M1-like macrophage infiltrate. Low viral cases are more heterogeneous likely reflecting inherent patient differences in the evolution of host response, but there is consistent indication of pulmonary epithelial cell recovery based on napsin A immunohistochemistry and RNA expression of surfactant and mucin genes. Using a digital spatial profiling platform, we find the virus corresponds to distinct spatial expression of interferon response genes demonstrating the intra-pulmonary heterogeneity of SARS-CoV-2 infection.


COVID-19 , Host Microbial Interactions , Interferons/metabolism , Lung , Adult , Aged , Aged, 80 and over , Aspartic Acid Endopeptidases/metabolism , Autopsy , COVID-19/immunology , COVID-19/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial Cells/virology , Female , Humans , Immunity , Immunohistochemistry , In Situ Hybridization , Interferons/genetics , Lung/pathology , Lung/virology , Macrophages/immunology , Male , Middle Aged , Mucins/genetics , Mucins/metabolism , Surface-Active Agents/metabolism , Transcriptome , Viral Load
8.
Nat Commun ; 11(1): 6311, 2020 12 09.
Article En | MEDLINE | ID: mdl-33298946

Blood-borne metastasis to the brain is a major complication of breast cancer, but cellular pathways that enable cancer cells to selectively grow in the brain microenvironment are poorly understood. We find that cultured circulating tumor cells (CTCs), derived from blood samples of women with advanced breast cancer and directly inoculated into the mouse frontal lobe, exhibit striking differences in proliferative potential in the brain. Derivative cell lines generated by serial intracranial injections acquire selectively increased proliferative competency in the brain, with reduced orthotopic tumor growth. Increased Hypoxia Inducible Factor 1A (HIF1A)-associated signaling correlates with enhanced proliferation in the brain, and shRNA-mediated suppression of HIF1A or drug inhibition of HIF-associated glycolytic pathways selectively impairs brain tumor growth while minimally impacting mammary tumor growth. In clinical specimens, brain metastases have elevated HIF1A protein expression, compared with matched primary breast tumors, and in patients with brain metastases, hypoxic signaling within CTCs predicts decreased overall survival. The selective activation of hypoxic signaling by metastatic breast cancer in the brain may have therapeutic implications.


Brain Neoplasms/secondary , Brain/pathology , Breast Neoplasms/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neoplastic Cells, Circulating/metabolism , Animals , Brain Neoplasms/blood , Brain Neoplasms/mortality , Breast Neoplasms/blood , Breast Neoplasms/mortality , Cell Hypoxia , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mammary Glands, Animal/pathology , Metabolomics , Mice , RNA, Small Interfering/metabolism , RNA-Seq , Signal Transduction/drug effects , Signal Transduction/genetics , Spheroids, Cellular , Stereotaxic Techniques , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
9.
medRxiv ; 2020 Aug 02.
Article En | MEDLINE | ID: mdl-32766600

The relationship of SARS-CoV-2 lung infection and severity of pulmonary disease is not fully understood. We analyzed autopsy specimens from 24 patients who succumbed to SARS-CoV-2 infection using a combination of different RNA and protein analytical platforms to characterize inter- and intra- patient heterogeneity of pulmonary virus infection. There was a spectrum of high and low virus cases that was associated with duration of disease and activation of interferon pathway genes. Using a digital spatial profiling platform, the virus corresponded to distinct spatial expression of interferon response genes and immune checkpoint genes demonstrating the intra-pulmonary heterogeneity of SARS-CoV-2 infection.

10.
Nat Commun ; 11(1): 3303, 2020 07 03.
Article En | MEDLINE | ID: mdl-32620742

Pancreatic ductal adenocarcinoma (PDAC) lethality is due to metastatic dissemination. Characterization of rare, heterogeneous circulating tumor cells (CTCs) can provide insight into metastasis and guide development of novel therapies. Using the CTC-iChip to purify CTCs from PDAC patients for RNA-seq characterization, we identify three major correlated gene sets, with stemness genes LIN28B/KLF4, WNT5A, and LGALS3 enriched in each correlated gene set; only LIN28B CTC expression was prognostic. CRISPR knockout of LIN28B-an oncofetal RNA-binding protein exerting diverse effects via negative regulation of let-7 miRNAs and other RNA targets-in cell and animal models confers a less aggressive/metastatic phenotype. This correlates with de-repression of let-7 miRNAs and is mimicked by silencing of downstream let-7 target HMGA2 or chemical inhibition of LIN28B/let-7 binding. Molecular characterization of CTCs provides a unique opportunity to correlated gene set metastatic profiles, identify drivers of dissemination, and develop therapies targeting the "seeds" of metastasis.


Carcinoma, Pancreatic Ductal/genetics , HMGA2 Protein/genetics , MicroRNAs/genetics , Neoplastic Cells, Circulating/metabolism , Pancreatic Neoplasms/genetics , RNA-Binding Proteins/genetics , Adult , Aged , Animals , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/metabolism , Cell Line, Tumor , Cell Movement/genetics , Female , Gene Expression Regulation, Neoplastic , HMGA2 Protein/metabolism , Humans , Kaplan-Meier Estimate , Kruppel-Like Factor 4 , Male , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Middle Aged , Neoplasm Metastasis , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , RNA-Binding Proteins/metabolism
11.
Ann Surg ; 272(3): 427-435, 2020 09 01.
Article En | MEDLINE | ID: mdl-32657929

OBJECTIVE: PDAC patients who undergo surgical resection and receive effective chemotherapy have the best chance of long-term survival. Unfortunately, we lack predictive biomarkers to guide optimal systemic treatment. Ex-vivo generation of PDO for pharmacotyping may serve as predictive biomarkers in PDAC. The goal of the current study was to demonstrate the clinical feasibility of a PDO-guided precision medicine framework of care. METHODS: PDO cultures were established from surgical specimens and endoscopic biopsies, expanded in Matrigel, and used for high-throughput drug testing (pharmacotyping). Efficacy of standard-of-care chemotherapeutics was assessed by measuring cell viability after drug exposure. RESULTS: A framework for rapid pharmacotyping of PDOs was established across a multi-institutional consortium of academic medical centers. Specimens obtained remotely and shipped to a central biorepository maintain viability and allowed generation of PDOs with 77% success. Early cultures maintain the clonal heterogeneity seen in PDAC with similar phenotypes (cystic-solid). Late cultures exhibit a dominant clone with a pharmacotyping profile similar to early passages. The biomass required for accurate pharmacotyping can be minimized by leveraging a high-throughput technology. Twenty-nine cultures were pharmacotyped to derive a population distribution of chemotherapeutic sensitivity at our center. Pharmacotyping rapidly-expanded PDOs was completed in a median of 48 (range 18-102) days. CONCLUSIONS: Rapid development of PDOs from patients undergoing surgery for PDAC is eminently feasible within the perioperative recovery period, enabling the potential for pharmacotyping to guide postoperative adjuvant chemotherapeutic selection. Studies validating PDOs as a promising predictive biomarker are ongoing.


Antineoplastic Agents/pharmacology , Neoplasm Staging/methods , Organoids/pathology , Pancreatic Neoplasms/therapy , Practice Guidelines as Topic , Precision Medicine/methods , Chemotherapy, Adjuvant , Humans , Pancreatectomy/methods , Pancreatic Neoplasms/diagnosis , Tumor Cells, Cultured
12.
Science ; 367(6485): 1468-1473, 2020 03 27.
Article En | MEDLINE | ID: mdl-32029688

Circulating tumor cells (CTCs) are shed into the bloodstream from primary tumors, but only a small subset of these cells generates metastases. We conducted an in vivo genome-wide CRISPR activation screen in CTCs from breast cancer patients to identify genes that promote distant metastasis in mice. Genes coding for ribosomal proteins and regulators of translation were enriched in this screen. Overexpression of RPL15, which encodes a component of the large ribosomal subunit, increased metastatic growth in multiple organs and selectively enhanced translation of other ribosomal proteins and cell cycle regulators. RNA sequencing of freshly isolated CTCs from breast cancer patients revealed a subset with strong ribosome and protein synthesis signatures; these CTCs expressed proliferation and epithelial markers and correlated with poor clinical outcome. Therapies targeting this aggressive subset of CTCs may merit exploration as potential suppressors of metastatic progression.


Breast Neoplasms/pathology , Neoplasm Metastasis , Neoplastic Cells, Circulating/pathology , Ribosomal Proteins/genetics , Animals , Breast Neoplasms/genetics , CRISPR-Cas Systems , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Neoplasm Transplantation , Sequence Analysis, RNA
13.
Proc Natl Acad Sci U S A ; 116(52): 26835-26845, 2019 Dec 26.
Article En | MEDLINE | ID: mdl-31843922

Transcriptional profiling has defined pancreatic ductal adenocarcinoma (PDAC) into distinct subtypes with the majority being classical epithelial (E) or quasi-mesenchymal (QM). Despite clear differences in clinical behavior, growing evidence indicates these subtypes exist on a continuum with features of both subtypes present and suggestive of interconverting cell states. Here, we investigated the impact of different therapies being evaluated in PDAC on the phenotypic spectrum of the E/QM state. We demonstrate using RNA-sequencing and RNA-in situ hybridization (RNA-ISH) that FOLFIRINOX combination chemotherapy induces a common shift of both E and QM PDAC toward a more QM state in cell lines and patient tumors. In contrast, Vitamin D, another drug under clinical investigation in PDAC, induces distinct transcriptional responses in each PDAC subtype, with augmentation of the baseline E and QM state. Importantly, this translates to functional changes that increase metastatic propensity in QM PDAC, but decrease dissemination in E PDAC in vivo models. These data exemplify the importance of both the initial E/QM subtype and the plasticity of E/QM states in PDAC in influencing response to therapy, which highlights their relevance in guiding clinical trials.

14.
Pancreatology ; 14(5): 356-60, 2014.
Article En | MEDLINE | ID: mdl-25278304

OBJECTIVES: To evaluate the relationship between exocrine pancreatic insufficiency and the level of glycemic control in diabetes (DM). METHODS: Patients with type 2 DM treated in our clinic were prospectively recruited into the study. Pancreatic diabetes was excluded. Cases with HbA1c ≥7% formed Group A (n = 59), and with HbA1c <7% Group B (n = 42). The fecal level of pancreatic elastase (PE-1) was measured and morphological examinations of the pancreas were performed. RESULTS: The PE-1 level was significantly lower in Group A than in Group B (385.9 ± 171.1 µg/g, vs. 454.6 ± 147.3 µg/g, p = 0.038). The PE-1 level was not correlated with HbA1c (r = -0.132, p = 0.187), the duration of DM (r = -0.046, p = 0.65), age (r = 0.010, p = 0.921), BMI (r = 0.203, p = 0.059), or pancreatic steatosis (r = 0.117, p = 0.244). The size of the pancreas did not differ significantly between Groups A and B. CONCLUSIONS: An exocrine pancreatic insufficiency demonstrated by fecal PE-1 determination is more frequent in type 2 DM patients with poor glycemic control. The impaired exocrine pancreatic function cannot be explained by an alteration in the size of the pancreas or by pancreatic steatosis.


Diabetes Mellitus, Type 2/complications , Exocrine Pancreatic Insufficiency/etiology , Glycated Hemoglobin/metabolism , Adult , Aged , Aged, 80 and over , Biomarkers/metabolism , Blood Glucose/metabolism , Carrier Proteins/metabolism , Diabetes Mellitus, Type 2/blood , Exocrine Pancreatic Insufficiency/diagnosis , Exocrine Pancreatic Insufficiency/epidemiology , Exocrine Pancreatic Insufficiency/metabolism , Female , Follow-Up Studies , Humans , Male , Middle Aged , Pancreas/pathology , Pancreatic Elastase , Prevalence , Prospective Studies
16.
PLoS One ; 8(3): e57522, 2013.
Article En | MEDLINE | ID: mdl-23483913

BACKGROUND: Pancreatic cancer is one of the deadliest of all human malignancies with limited options for therapy. Here, we report the development of an optimized targeted drug delivery system to inhibit advanced stage pancreatic tumor growth in an orthotopic mouse model. METHODPRINCIPAL FINDINGS: Targeting specificity in vitro was confirmed by preincubation of the pancreatic cancer cells with C225 as well as Nitrobenzylthioinosine (NBMPR - nucleoside transporter (NT) inhibitor). Upon nanoconjugation functional activity of gemcitabine was retained as tested using a thymidine incorporation assay. Significant stability of the nanoconjugates was maintained, with only 12% release of gemcitabine over a 24-hour period in mouse plasma. Finally, an in vivo study demonstrated the inhibition of tumor growth through targeted delivery of a low dose of gemcitabine in an orthotopic model of pancreatic cancer, mimicking an advanced stage of the disease. CONCLUSION: We demonstrated in this study that the gold nanoparticle-based therapeutic containing gemcitabine inhibited tumor growth in an advanced stage of the disease in an orthotopic model of pancreatic cancer. Future work would focus on understanding the pharmacokinetics and combining active targeting with passive targeting to further improve the therapeutic efficacy and increase survival.


Antineoplastic Agents/therapeutic use , Designer Drugs/therapeutic use , Pancreatic Neoplasms/drug therapy , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Animals , Antineoplastic Agents/pharmacology , Body Fluids/drug effects , Body Fluids/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Disease Models, Animal , Gold/therapeutic use , Humans , Immunohistochemistry , Light , Male , Metal Nanoparticles/therapeutic use , Metal Nanoparticles/ultrastructure , Mice , Mice, Nude , Molecular Targeted Therapy , Nanoconjugates/therapeutic use , Nanoconjugates/ultrastructure , Pancreatic Neoplasms/pathology , Scattering, Radiation , Static Electricity , Treatment Outcome , Gemcitabine
17.
PLoS One ; 6(6): e20347, 2011.
Article En | MEDLINE | ID: mdl-21738572

BACKGROUND: Pancreatic cancer is the fourth leading cause of cancer related deaths in America. Monoclonal antibodies are a viable treatment option for inhibiting cancer growth. Tumor specific drug delivery could be achieved utilizing these monoclonal antibodies as targeting agents. This type of designer therapeutic is evolving and with the use of gold nanoparticles it is a promising approach to selectively deliver chemotherapeutics to malignant cells. Gold nanoparticles (GNPs) are showing extreme promise in current medicinal research. GNPs have been shown to non-invasively kill tumor cells by hyperthermia using radiofrequency. They have also been implemented as early detection agents due to their unique X-ray contrast properties; success was revealed with clear delineation of blood capillaries in a preclinical model by CT (computer tomography). The fundamental parameters for intelligent design of nanoconjugates are on the forefront. The goal of this study is to define the necessary design parameters to successfully target pancreatic cancer cells. METHODOLOGY/PRINCIPAL FINDINGS: The nanoconjugates described in this study were characterized with various physico-chemical techniques. We demonstrate that the number of cetuximab molecules (targeting agent) on a GNP, the hydrodynamic size of the nanoconjugates, available reactive surface area and the ability of the nanoconjugates to sequester EGFR (epidermal growth factor receptor), all play critical roles in effectively targeting tumor cells in vitro and in vivo in an orthotopic model of pancreatic cancer. CONCLUSION: Our results suggest the specific targeting of tumor cells depends on a number of crucial components 1) targeting agent to nanoparticle ratio 2) availability of reactive surface area on the nanoparticle 3) ability of the nanoconjugate to bind the target and 4) hydrodynamic diameter of the nanoconjugate. We believe this study will help define the design parameters for formulating better strategies for specifically targeting tumors with nanoparticle conjugates.


Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Gold/chemistry , Metal Nanoparticles/chemistry , Nanoconjugates/chemistry , Pancreatic Neoplasms/drug therapy , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cetuximab , Humans , Mice , Mice, Nude , Microscopy, Electron, Transmission , Xenograft Model Antitumor Assays
18.
PLoS One ; 6(3): e17918, 2011 Mar 21.
Article En | MEDLINE | ID: mdl-21445297

Undoubtedly ovarian cancer is a vexing, incurable disease for patients with recurrent cancer and therapeutic options are limited. Although the polycomb group gene, Bmi-1 that regulates the self-renewal of normal stem and progenitor cells has been implicated in the pathogenesis of many human malignancies, yet a role for Bmi-1 in influencing chemotherapy response has not been addressed before. Here we demonstrate that silencing Bmi-1 reduces intracellular GSH levels and thereby sensitizes chemoresistant ovarian cancer cells to chemotherapeutics such as cisplatin. By exacerbating ROS production in response to cisplatin, Bmi-1 silencing activates the DNA damage response pathway, caspases and cleaves PARP resulting in the induction apoptosis in ovarian cancer cells. In an in vivo orthotopic mouse model of chemoresistant ovarian cancer, knockdown of Bmi-1 by nanoliposomal delivery significantly inhibits tumor growth. While cisplatin monotherapy was inactive, combination of Bmi-1 silencing along with cisplatin almost completely abrogated ovarian tumor growth. Collectively these findings establish Bmi-1 as an important new target for therapy in chemoresistant ovarian cancer.


Antineoplastic Agents/therapeutic use , Cisplatin/therapeutic use , Gene Silencing , Nuclear Proteins/genetics , Ovarian Neoplasms/drug therapy , Proto-Oncogene Proteins/genetics , Repressor Proteins/genetics , Apoptosis , Cell Proliferation , Female , Gene Knockdown Techniques , Glutathione/biosynthesis , Humans , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Polycomb Repressive Complex 1 , Reactive Oxygen Species/metabolism
19.
J Biol Chem ; 285(41): 31840-8, 2010 Oct 08.
Article En | MEDLINE | ID: mdl-20675371

The transforming growth factor-beta (TGF-ß) superfamily is one of the most diversified cell signaling pathways and regulates many physiological and pathological processes. Recently, neuropilin-1 (NRP-1) was reported to bind and activate the latent form of TGF-ß1 (LAP-TGF-ß1). We investigated the role of NRP-1 on Smad signaling in stromal fibroblasts upon TGF-ß stimulation. Elimination of NRP-1 in stromal fibroblast cell lines increases Smad1/5 phosphorylation and downstream responses as evidenced by up-regulation of inhibitor of differentiation (Id-1). Conversely, NRP-1 loss decreases Smad2/3 phosphorylation and its responses as shown by down-regulation of α-smooth muscle actin (α-SMA) and also cells exhibit more quiescent phenotypes and growth arrest. Moreover, we also observed that NRP-1 expression is increased during the culture activation of hepatic stellate cells (HSCs), a liver resident fibroblast. Taken together, our data suggest that NRP-1 functions as a key determinant of the diverse responses downstream of TGF-ß1 that are mediated by distinct Smad proteins and promotes myofibroblast phenotype.


Fibroblasts/metabolism , Myoblasts/metabolism , Neuropilin-1/metabolism , Signal Transduction/physiology , Smad Proteins, Receptor-Regulated/metabolism , Actins/genetics , Actins/metabolism , Animals , Cell Line, Transformed , Fibroblasts/cytology , Gene Expression Regulation/physiology , Hepatic Stellate Cells/cytology , Hepatic Stellate Cells/metabolism , Humans , Liver/metabolism , Mice , Mice, Knockout , Myoblasts/cytology , Neuropilin-1/genetics , Phosphorylation/physiology , Smad Proteins, Receptor-Regulated/genetics , Transforming Growth Factor beta1/metabolism
20.
Eur J Pharmacol ; 616(1-3): 270-4, 2009 Aug 15.
Article En | MEDLINE | ID: mdl-19486901

The proteosome inhibitor bortezomib is used in the treatment of patients with multiple myeloma. Proteosomes are responsible for the degradation of I-kappaB, the inhibitory protein of transcription factor nuclear factor kappa B (Nf-kappaB). The heat shock protein (HSP) inducing effect of bortezomib is also documented. The aim of our work was to test the anti-inflammatory effect of bortezomib in cholecystokinin-octapeptide (CCK-8)-induced acute pancreatitis. Male Wistar rats were divided into three groups (n=8 in each). Group P received an i.p. injection of physiological saline (p.s.) 60 min. before the induction of acute pancreatitis by three hourly s.c. injections of 100 microg/kg CCK-8. Group BP received 1 mg/kg bortezomib dissolved in p.s. 1 h previous to pancreatitis induction. Group C was treated with the vehicle (p.s.). Animals were exsanguinated 4 h after the last injection of CCK-8. Bortezomib pre-treatment significantly reduced the pancreatic weight/body weight ratio, and improved the histology by decreasing the extent of vacuolization and infiltration. Bortezomib pre-treatment inhibited I-kappaBbeta degradation, and induced the synthesis of HSP72. The results confirmed the anti-inflammatory effect of bortezomib in acute experimental pancreatitis. This effect of the drug is presumably mediated by the inhibition of Nf-kappaB activation and induction of HSP synthesis.


Boronic Acids/pharmacology , Heat-Shock Proteins/metabolism , Multienzyme Complexes/antagonists & inhibitors , Pancreatitis/metabolism , Protease Inhibitors/pharmacology , Pyrazines/pharmacology , Animals , Boronic Acids/therapeutic use , Bortezomib , Male , Pancreas/drug effects , Pancreas/metabolism , Pancreas/pathology , Pancreatitis/drug therapy , Pancreatitis/enzymology , Pancreatitis/pathology , Protease Inhibitors/therapeutic use , Pyrazines/therapeutic use , Rats , Rats, Wistar
...