Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Clin Invest ; 134(2)2024 Jan 16.
Article in English | MEDLINE | ID: mdl-38226620

ABSTRACT

The combination of targeted therapy with immune checkpoint inhibition (ICI) is an area of intense interest. We studied the interaction of fibroblast growth factor receptor (FGFR) inhibition with ICI in urothelial carcinoma (UC) of the bladder, in which FGFR3 is altered in 50% of cases. Using an FGFR3-driven, Trp53-mutant genetically engineered murine model (UPFL), we demonstrate that UPFL tumors recapitulate the histology and molecular subtype of their FGFR3-altered human counterparts. Additionally, UPFL1 allografts exhibit hyperprogression to ICI associated with an expansion of T regulatory cells (Tregs). Erdafitinib blocked Treg proliferation in vitro, while in vivo ICI-induced Treg expansion was fully abrogated by FGFR inhibition. Combined erdafitinib and ICI resulted in high therapeutic efficacy. In aggregate, our work establishes that, in mice, co-alteration of FGFR3 and Trp53 results in high-grade, non-muscle-invasive UC and presents a previously underappreciated role for FGFR inhibition in blocking ICI-induced Treg expansion.


Subject(s)
Carcinoma, Transitional Cell , Urinary Bladder Neoplasms , Animals , Humans , Mice , Carcinoma, Transitional Cell/drug therapy , Carcinoma, Transitional Cell/metabolism , Carcinoma, Transitional Cell/pathology , Immunosuppression Therapy , Receptor, Fibroblast Growth Factor, Type 3/genetics , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism
2.
Pharmaceutics ; 14(1)2022 Jan 05.
Article in English | MEDLINE | ID: mdl-35057024

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive malignancies and is the fourth leading cause of cancer-related deaths in the United States. Unfortunately, 80-85% of patients are diagnosed with unresectable, advanced stage tumors. These tumors are incurable and result in a median survival less than approximately six months and an overall 5-year survival rate of less than 7%. Whilst chemotherapy is a critical treatment, cure is not possible without surgical resection. The poor clinical outcomes in PDAC can be partially attributed to its dense desmoplastic stroma, taking up roughly 80% of the tumor mass. The stroma surrounding the tumor disrupts the normal architecture of pancreatic tissue leading to poor vascularization, high intratumoral pressure along with hypoxia and an acidic tumor microenvironment. This complicated microenvironment presents a significant challenge for drug delivery. The current manuscript discusses a novel approach to overcome many of these various obstacles. A complex of gemcitabine (GEM) and hemoglobin S (HbS) was formulated, which self-polymerizes under hypoxic and acidic conditions. When polymerized, HbS has the potential to break the tumor stroma, decrease intratumoral pressure, and therefore improve the treatment efficacy of standard therapy. Intratumoral injection of HbS with a fluorescent small molecule surrogate for GEM into a pancreatic tumor xenograft resulted in improved dissemination of the small molecule throughout the pancreatic tumor. The self-polymerization of HbS + GEM was significantly more effective than either agent individually at decreasing tumor size in an in vivo PDAC mouse model. These findings would suggest a clinical benefit from delivering the complex of GEM and HbS via direct injection by endoscopic ultrasound (EUS). With such a treatment option, patients with locally advanced disease would have the potential to become surgical candidates, offering them a chance for cure.

3.
Am Soc Clin Oncol Educ Book ; 39: e53-e58, 2019 Jan.
Article in English | MEDLINE | ID: mdl-31099672

ABSTRACT

Big data and predictive analytics have immense potential to improve risk stratification, particularly in data-rich fields like oncology. This article reviews the literature published on use cases and challenges in applying predictive analytics to improve risk stratification in oncology. We characterized evidence-based use cases of predictive analytics in oncology into three distinct fields: (1) population health management, (2) radiomics, and (3) pathology. We then highlight promising future use cases of predictive analytics in clinical decision support and genomic risk stratification. We conclude by describing challenges in the future applications of big data in oncology, namely (1) difficulties in acquisition of comprehensive data and endpoints, (2) the lack of prospective validation of predictive tools, and (3) the risk of automating bias in observational datasets. If such challenges can be overcome, computational techniques for clinical risk stratification will in short order improve clinical risk stratification for patients with cancer.


Subject(s)
Big Data , Data Mining , Medical Oncology/methods , Neoplasms/epidemiology , Algorithms , Decision Support Systems, Clinical , Electronic Health Records , Genomics/methods , Humans , Neoplasms/etiology , Precision Medicine , Public Health Surveillance , Reproducibility of Results , Risk Assessment
4.
ACS Appl Nano Mater ; 2(10): 6249-6257, 2019 Oct 25.
Article in English | MEDLINE | ID: mdl-33585803

ABSTRACT

Targeting therapeutic agents to specific organs in the body remains a challenge despite advances in the science of systemic drug delivery. We have engineered a programmable-bioinspired nanoparticle (P-BiNP) delivery system to simultaneously target the bone and increase uptake in homotypic tumor cells by coating polymeric nanoparticles with programmed cancer cell membranes. This approach is unique in that we have incorporated relevant clinical bioinformatics data to guide the design and enhancement of biological processes that these nanoparticles are engineered to mimic. To achieve this, an analysis of RNA expression from metastatic prostate cancer patients identified ITGB3 (a subunit of integrin α V ß 3) as overexpressed in patients with bone metastasis. Cancer cells were stimulated to increase this integrin expression on the cell surface, and these membranes were subsequently used to coat cargo carrying polymeric nanoparticles. Physicochemical optimization and characterization of the P-BiNPs showed desirable qualities regarding size, ζ potential, and stability. In vitro testing confirmed enhanced homotypic binding and uptake in cancer cells. P-BiNPs also demonstrated improved bone localization in vivo with a murine model. This novel approach of identifying clinically relevant targets for dual homotypic and bone targeting has potential as a strategy for treatment and imaging modalities in diseases that affect the bone as well as broader implications for delivering nanoparticles to other organs of interest.

5.
J Nanobiotechnology ; 16(1): 12, 2018 Feb 12.
Article in English | MEDLINE | ID: mdl-29433518

ABSTRACT

BACKGROUND: The process of optimization and fabrication of nanoparticle synthesis for preclinical studies can be challenging and time consuming. Traditional small scale laboratory synthesis techniques suffer from batch to batch variability. Additionally, the parameters used in the original formulation must be re-optimized due to differences in fabrication techniques for clinical production. Several low flow microfluidic synthesis processes have been reported in recent years for developing nanoparticles that are a hybrid between polymeric nanoparticles and liposomes. However, use of high flow microfluidic synthetic techniques has not been described for this type of nanoparticle system, which we will term as nanolipomer. In this manuscript, we describe the successful optimization and functional assessment of nanolipomers fabricated using a microfluidic synthesis method under high flow parameters. RESULTS: The optimal total flow rate for synthesis of these nanolipomers was found to be 12 ml/min and flow rate ratio 1:1 (organic phase: aqueous phase). The PLGA polymer concentration of 10 mg/ml and a DSPE-PEG lipid concentration of 10% w/v provided optimal size, PDI and stability. Drug loading and encapsulation of a representative hydrophobic small molecule drug, curcumin, was optimized and found that high encapsulation efficiency of 58.8% and drug loading of 4.4% was achieved at 7.5% w/w initial concentration of curcumin/PLGA polymer. The final size and polydispersity index of the optimized nanolipomer was 102.11 nm and 0.126, respectively. Functional assessment of uptake of the nanolipomers in C4-2B prostate cancer cells showed uptake at 1 h and increased uptake at 24 h. The nanolipomer was more effective in the cell viability assay compared to free drug. Finally, assessment of in vivo retention in mice of these nanolipomers revealed retention for up to 2 h and were completely cleared at 24 h. CONCLUSIONS: In this study, we have demonstrated that a nanolipomer formulation can be successfully synthesized and easily scaled up through a high flow microfluidic system with optimal characteristics. The process of developing nanolipomers using this methodology is significant as the same optimized parameters used for small batches could be translated into manufacturing large scale batches for clinical trials through parallel flow systems.


Subject(s)
Antineoplastic Agents/administration & dosage , Curcumin/administration & dosage , Drug Carriers/chemistry , Lab-On-A-Chip Devices , Liposomes/chemistry , Nanoparticles/chemistry , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Curcumin/pharmacokinetics , Curcumin/pharmacology , Drug Evaluation, Preclinical , Equipment Design , Humans , Lactic Acid/chemistry , Male , Mice , Phosphatidylethanolamines/chemistry , Polyethylene Glycols/chemistry , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Prostatic Neoplasms/drug therapy
6.
Nanomedicine (Lond) ; 12(17): 2083-2095, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28805551

ABSTRACT

AIM: The aim of this study was to develop a novel cabazitaxel bone targeted nanoparticle (NP) system for improved drug delivery to the bone microenvironment. MATERIALS & METHODS: Nanoparticles were developed using poly(D,L-lactic-co-glycolic acid) and cabazitaxel as the core with amino-bisphosphonate surface conjugation. Optimization of nanoparticle physiochemical properties, in vitro evaluation in prostate cancer cell lines and in vivo testing in an intraosseous model of metastatic prostate cancer was performed. RESULTS: This bone targeted cabazitaxel nanocarrier system showed significant reduction in tumor burden, while at the same time maintaining bone structure integrity and reducing pain in the mouse tumor limb. CONCLUSION: This bone microenvironment targeted nanoparticle system and clinically relevant approach of evaluation represents a promising advancement for treating bone metastatic cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Bone Neoplasms/drug therapy , Bone and Bones/drug effects , Nanoparticles/chemistry , Pain/drug therapy , Prostatic Neoplasms/drug therapy , Taxoids/pharmacology , Animals , Antineoplastic Agents/chemistry , Bone Neoplasms/secondary , Cell Line, Tumor , Cell Survival , Diphosphonates/chemistry , Diphosphonates/pharmacology , Drug Carriers/chemistry , Drug Liberation , Humans , Lactic Acid/chemistry , Male , Mice , Mice, Nude , Particle Size , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Prostatic Neoplasms/pathology , Surface Properties , Taxoids/chemistry , Xenograft Model Antitumor Assays
7.
J Exp Clin Cancer Res ; 36(1): 108, 2017 08 11.
Article in English | MEDLINE | ID: mdl-28800754

ABSTRACT

BACKGROUND: Elucidation of mechanisms regulating bone metastasis has progressed significantly in recent years and this has translated to many new therapeutic options for patients with bone metastatic cancers. However, the rapid rate of progress in both the basic science literature and therapies undergoing clinical trials makes staying abreast with current developments challenging. This review seeks to provide an update on the current state of the science in bone metastasis research and give a snap shot of therapies in clinical trials for bone metastatic cancer. MAIN BODY: Bone metastasis represents a difficult to treat clinical scenario due to pain, increased fracture risk, decreased quality of life and diminished overall survival outcomes. Multiple types of cancer have the specific ability to home to the bone microenvironment and cause metastatic lesions. This osteotropism was first described by Stephen Paget nearly 100 years ago as the 'seed and soil' hypothesis. Once cancer cells arrive at the bone they encounter a variety of cells native to the bone microenvironment which contribute to the establishment of bone metastatic lesions. In the first part of this review, the 'seed and soil' hypothesis is revisited while emphasizing recent developments in understanding the impact of native bone microenvironment cells on the metastatic process. Next, approved therapies for treating bone metastasis at the systemic level as well as those that target the bone microenvironment are discussed and current National Comprehensive Cancer Network (NCCN) guidelines relating to treatment of bone metastases are summarized. Finally, all open interventional clinical trials for therapies relating to treatment of bone metastasis have been complied and categorized. CONCLUSION: Understanding the recent advancements in bone metastasis research is important for continued development of novel bone targeted therapies. The plethora of ongoing clinical trials will hopefully translate into improved treatments options for patients suffering from bone metastatic cancers.


Subject(s)
Bone Neoplasms/therapy , Neoplasm Metastasis/therapy , Bone Neoplasms/pathology , Bone Neoplasms/secondary , Humans , Meta-Analysis as Topic , Neoplasm Metastasis/pathology , Quality of Life
8.
World J Surg ; 41(11): 2854-2857, 2017 11.
Article in English | MEDLINE | ID: mdl-28717906

ABSTRACT

BACKGROUND: Chronic obstruction of the biliary system may cause hepatic fibrosis and liver failure. The purpose of this study was to define the incidence of unrecognized liver fibrosis in patients undergoing pancreaticoduodenectomy (PD). METHODS: Retrospective data were collected on patients undergoing PD during a 21-month period. Each patient had a core needle biopsy at the time of surgery by a hepatobiliary surgeon. RESULTS: This study identified 36 consecutive patients who were referred to a tertiary center and underwent pancreatoduodenectomy during a period of 21 months. The majority of patients, 32 (88.8%), were diagnosed with pancreatic adenocarcinoma. Liver fibrosis was diagnosed in 23 (63.9%) patients. A total of 25 (69.4%) patients were found to have pathological evidence of cholestasis consistent with bile obstruction. Patients that were found to have evidence of obstruction had significantly increased odds that fibrosis stage 2 would be found on pathological diagnosis (OR 6.75, 95% CI 1.20-38.02, Fisher's exact test P value = 0.0312). There was no significant association in patients who were stented compared to non-stented patients and their diagnosis of high-grade fibrosis stage 2 (OR 1.5238, 95% CI 0.4019-5.7769, Fisher's exact test P value = 0.7360). CONCLUSIONS: An astonishing 63.9% of patients who underwent PD were diagnosed with stage 1-4 liver fibrosis and half (47.2%) had fibrosis stage of 2 or more. Further, stent status had no significant impact on the degree of liver fibrosis. Liver fibrosis is currently underrecognized in patients undergoing PD, which is important for physicians to be conscious of as it is known that liver fibrosis increases morbidity and mortality.


Subject(s)
Adenocarcinoma/surgery , Liver Cirrhosis/diagnosis , Pancreatic Neoplasms/surgery , Pancreaticoduodenectomy , Adenocarcinoma/complications , Aged , Cholestasis/etiology , Female , Humans , Liver Cirrhosis/complications , Male , Middle Aged , Pancreatic Neoplasms/complications , Retrospective Studies , Stents
9.
J Biomed Nanotechnol ; 12(4): 679-88, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27301194

ABSTRACT

Non-small-cell lung cancer therapy is a challenge due to poor prognosis and low survival rate. There is an acute need for advanced therapies having higher drug efficacy, low immunogenicity and fewer side effects which will markedly improve patient compliance and quality of life of cancer patients. The purpose of this study was to develop a novel hybrid curcumin nanoformulation (Curcumin-ER) and evaluate the therapeutic efficacy of this formulation on a non-small cell lung cancer xenograft model. Use of curcumin, a natural anticancer agent, is majorly limited due to its poor aqueous solubility and hence it's low systemic bioavailability. In this paper, we carried out the nanoformulation of Curcumin-ER, optimized the formulation process and determined the anticancer effects of Curcumin-ER against human A549 non-small cell lung cancer using in vitro and in vivo studies. Xenograft tumors in nude mice were treated with 20 mg/kg subcutaneous injection of Curcumin-ER and liposomal curcumin (Lipocurc) twice a week for seven weeks. Results showed that tumor growth was suppressed by 52.1% by Curcumin-ER treatment and only 32.2% by Lipocurc compared to controls. Tumor sections were isolated from murine xenografts and histology and immunohistochemistry was performed. A decrease in expression of NFκB-p65 subunit and proliferation marker, Ki-67 was observed in treated tumors. In addition, a potent anti-angiogenic effect, characterized by reduced expression of annexin A2 protein, was observed in treated tumors. These results establish the effectiveness of Curcumin-ER in regressing human non-small cell lung cancer growth in the xenograft model using subcutaneous route of administration. The therapeutic efficacy of Curcumin-ER highlights the potential of this hybrid nanoformulation in treating patients with non-small cell lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung/chemistry , Carcinoma, Non-Small-Cell Lung/drug therapy , Curcumin/administration & dosage , Curcumin/chemistry , Delayed-Action Preparations/administration & dosage , Liposomes/chemistry , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Delayed-Action Preparations/chemistry , Drug Compounding/methods , Female , Humans , Injections, Subcutaneous , Mice, Nude , Tissue Distribution
10.
Front Biosci (Landmark Ed) ; 21(4): 757-68, 2016 01 01.
Article in English | MEDLINE | ID: mdl-26709804

ABSTRACT

The ultimate goal of cancer therapeutic research is to develop effective, targeted therapeutics that exploit the vulnerabilities of cancer cells. The three isoforms of Akt, also known as protein kinase B (PKB), are important mediators of various pathways that transmit mitogenic signals from the cell's exterior to the effector proteins of the cell's interior. Due to Akt\\\\\\\'s importance in cell functions such as growth, proliferation and cell survival, many cancer cells rely on this pathway to aid in their survival. This dependence can lead to chemoresistance and selection of more adapted populations of cancer cells. Thus, it is important to understand the functional significance of isoform specificity and its relation to chemoresistance. In this review, we have summarized recent studies on Akt isoform specific regulation as well as each isoform's role in chemoresistance, emphasizing their potential as targets for cancer therapy. We have also condensed ongoing clinical studies involving various types of Akt inhibitors while highlighting the type of study, rationale and co-therapies involved in identifying Akt isoforms as promising therapeutic targets.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism , Humans , Neoplasms/enzymology , Phosphorylation , Proto-Oncogene Proteins c-akt/antagonists & inhibitors
11.
Int J Mol Sci ; 16(2): 3990-5, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25690029

ABSTRACT

Treatments utilizing monoclonal antibody therapeutics against intracellular protein-protein interactions in cancer cells have been hampered by several factors, including poor intracellular uptake and rapid lysosomal degradation. Our current work examines the feasibility of encapsulating monoclonal antibodies within poly(lactic-co-glycolic acid) (PLGA) nanoparticles using a water/oil/water double emulsion solvent evaporation technique. This method can be used to prepare protective polymeric nanoparticles for transporting functional antibodies to the cytoplasmic compartment of cancer cells. Nanoparticles were formulated and then characterized using a number of physical and biological parameters. The average nanoparticle size ranged from 221 to 252 nm with a low polydispersity index. Encapsulation efficiency of 16%-22% and antibody loading of 0.3%-1.12% were observed. The antibody molecules were released from the nanoparticles in a sustained manner and upon release maintained functionality. Our studies achieved successful formulation of antibody loaded polymeric nanoparticles, thus indicating that a PLGA-based antibody nanoformulation is a promising intracellular delivery vehicle for a large number of new intracellular antibody targets in cancer cells.


Subject(s)
Antibodies, Monoclonal/chemistry , Biocompatible Materials/chemistry , Drug Carriers/chemistry , Nanoparticles/chemistry , Annexin A2/immunology , Annexin A2/metabolism , Antibodies, Monoclonal/immunology , Cell Line, Tumor , Humans , Lactic Acid/chemistry , Microscopy, Confocal , Particle Size , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer
12.
Adv Exp Med Biol ; 807: 33-58, 2014.
Article in English | MEDLINE | ID: mdl-24619617

ABSTRACT

The ability to detect many cancers at an early stage in its clinical course has the potential to improve patient outcomes in terms of morbidity and mortality. Nanosized components incorporated into existing clinical diagnostic and detection systems as well as novel nanobiosensors have demonstrated improved sensitivity and specificity compared with traditional cancer testing approaches. Nanoparticles, nanowires, nanotubes, and nanocantilevers are examples of four nanobiosensor systems that have been used experimentally in the context of detection and diagnosis of prostate, breast, pancreatic, lung, and brain cancers over the past few years. Nanobiosensors will begin to transition into clinically validated tests as experimental and engineering techniques advance. This paper presents examples of some such nanobiosensors for cancer diagnosis and detection.


Subject(s)
Biosensing Techniques/methods , Nanotechnology/methods , Neoplasms/diagnosis , Humans , Nanoparticles , Nanotubes, Carbon , Quantum Dots
SELECTION OF CITATIONS
SEARCH DETAIL
...