Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Kimberly Ann V. Zubris is active.

Publication


Featured researches published by Kimberly Ann V. Zubris.


Biomaterials | 2013

Prevention of nodal metastases in breast cancer following the lymphatic migration of paclitaxel-loaded expansile nanoparticles.

Rong Liu; Denis M. Gilmore; Kimberly Ann V. Zubris; Xiaoyin Xu; Paul J. Catalano; Robert F. Padera; Mark W. Grinstaff; Yolonda L. Colson

Although breast cancer patients with localized disease exhibit an excellent long-term prognosis, up to 40% of patients treated with local resection alone may harbor occult nodal metastatic disease leading to increased locoregional recurrence and decreased survival. Given the potential for targeted drug delivery to result in more efficacious locoregional control with less morbidity, the current study assessed the ability of drug-loaded polymeric expansile nanoparticles (eNP) to migrate from the site of tumor to regional lymph nodes, locally deliver a chemotherapeutic payload, and prevent primary tumor growth as well as lymph node metastases. Expansile nanoparticles entered tumor cells and paclitaxel-loaded eNP (Pax-eNP) exhibited dose-dependent cytotoxicity in vitro and significantly decreased tumor doubling time in vivo against human triple negative breast cancer in both microscopic and established murine breast cancer models. Furthermore, migration of Pax-eNP to axillary lymph nodes resulted in higher intranodal paclitaxel concentrations and a significantly lower incidence of lymph node metastases. These findings demonstrate that lymphatic migration of drug-loaded eNP provides regionally targeted delivery of chemotherapy to both decrease local tumor growth and strategically prevent the development of nodal metastases within the regional tumor-draining lymph node basin.


Biomaterials | 2011

The performance of expansile nanoparticles in a murine model of peritoneal carcinomatosis.

Yolonda L. Colson; Rong Liu; Emily Southard; Morgan D. Schulz; Jacqueline E. Wade; Aaron P. Griset; Kimberly Ann V. Zubris; Robert F. Padera; Mark W. Grinstaff

Carcinomatosis from peritoneal surface malignancies, such as mesothelioma, appendiceal carcinoma or ovarian metastases, significantly decreases survival and quality of life. Given a 60-80% locoregional recurrence rate after surgical debulking for mesothelioma, the current study explores the use of polymeric nanoparticles, specifically engineered to expand and locally deliver chemotherapeutic agents at endosomal pH, for the prevention of progressive carcinomatosis. Anti-tumor efficacy of paclitaxel-loaded pH-responsive expansile nanoparticles (Pax-eNP) was evaluated in vitro and in in vivo murine models of malignant peritoneal mesothelioma. Pax-eNP inhibited mesothelioma growth in vitro, markedly decreased tumor growth and disease severity in vivo, prevented initial intraperitoneal tumor implants, and significantly prolonged survival compared to other intraperitoneal drug delivery methods. These outcomes suggest that the mechanism of pH-triggered drug delivery and tumor affinity associated with eNP may effectively improve the local control of residual microscopic disease following surgical debulking of locoregionally aggressive malignancies.


Biomacromolecules | 2013

In vitro activity of Paclitaxel-loaded polymeric expansile nanoparticles in breast cancer cells.

Kimberly Ann V. Zubris; Rong Liu; Aaron H. Colby; Morgan D. Schulz; Yolonda L. Colson; Mark W. Grinstaff

Through a series of in vitro studies, the essential steps for intracellular drug delivery of paclitaxel using a pH-responsive nanoparticle system have been investigated in breast cancer cells. We successfully encapsulated paclitaxel within polymeric expansile nanoparticles (Pax-eNPs) at 5% loading via a miniemulsion polymerization procedure. Fluorescently tagged eNPs were readily taken up by MDA-MB-231 breast cancer cells grown in culture as confirmed by confocal microscopy and flow cytometry. The ability of the encapsulated paclitaxel to reach the cytoplasm was also observed using confocal microscopy and fluorescently labeled paclitaxel. Pax-eNPs were shown to be efficacious against three in vitro human breast adenocarcinoma cell lines (MDA-MB-231, MCF-7, and SK-BR-3) as well as cells isolated from the pleural effusions of two different breast cancer patients. Lastly, macropinocytosis was identified as the major cellular pathway responsible for eNP uptake, as confirmed using temperature-sensitive metabolic reduction, pharmacologic inhibitors, and fluid-phase marker colocalization.


Molecular Pharmaceutics | 2012

HYDROGELS AS INTRACELLULAR DEPOTS FOR DRUG DELIVERY

Kimberly Ann V. Zubris; Yolonda L. Colson; Mark W. Grinstaff

The intracellular activity and drug depot characteristics of micrometer-sized hydrogels are described. The hydrogel structure is formed after cellular uptake of a solid polymeric nanoparticle that swells in response to mildly acidic conditions as it transforms from a hydrophobic to a hydrophilic structure. These nanoparticles are rapidly taken up into A549 human non-small cell lung cancer cells with 88.3 ± 0.8% of cells experiencing uptake in 24 h, undergo expansion to release encapsulated drug and can effectively deliver chemotherapeutics in vitro. The anticancer drug paclitaxel was also shown to have a 3- to 4-fold increased affinity for the expanded nanoparticle state, allowing the expansile nanoparticles to act as intracellular drug depots and concentrate the drug locally.


Journal of The American College of Surgeons | 2012

Nanoparticle migration and delivery of Paclitaxel to regional lymph nodes in a large animal model.

Onkar V. Khullar; Aaron P. Griset; Summer L. Gibbs-Strauss; Lucian R. Chirieac; Kimberly Ann V. Zubris; John V. Frangioni; Mark W. Grinstaff; Yolonda L. Colson

BACKGROUND The aim of this study was to demonstrate feasibility of migration and in situ chemotherapy delivery to regional lymph nodes (LN) in a large animal model using an expansile polymer nanoparticle (eNP) delivery system. STUDY DESIGN Dual-labeled 50-nm and 100-nm eNP were prepared by encapsulating an IR-813 near-infrared (NIR) fluorescent dye within coumarin-conjugated expansile polymer nanoparticles (NIR-C-eNP). NIR imaging and fluorescent microscopy were used to identify intralymphatic migration of NIR-nanoparticles to draining inguinal or mesenteric LN after injection in swine hind legs or intestine. Nanoparticle-mediated intranodal delivery of chemotherapy was subsequently assessed with Oregon Green paclitaxel-loaded NIR-eNP (NIR-OGpax-eNP). RESULTS NIR imaging demonstrated direct lymphatic migration of 50-nm, but not 100-nm, NIR-C-eNP and NIR-OGpax-eNP to the draining regional LNs after intradermal injection in the hind leg or subserosal injection in intestine. Fluorescent microscopy demonstrated that IR-813 used for NIR real-time trafficking colocalized with both the coumarin-labeled polymer and paclitaxel chemotherapy and was identified within the subcapsular spaces of the draining LNs. These studies verify nodal migration of both nanoparticle and encapsulated payload, and confirm the feasibility of focusing chemotherapy delivery directly to regional nodes. CONCLUSIONS Regionally-targeted intranodal chemotherapy can be delivered to draining LNs for both skin and solid organs using 50-nm paclitaxel-loaded eNP.


The Annals of Thoracic Surgery | 2011

Paclitaxel-Loaded Expansile Nanoparticles Delay Local Recurrence in a Heterotopic Murine Non-Small Cell Lung Cancer Model

Rong Liu; Onkar V. Khullar; Aaron P. Griset; Jacqueline E. Wade; Kimberly Ann V. Zubris; Mark W. Grinstaff; Yolonda L. Colson

BACKGROUND Surgical resection remains the most effective treatment option for patients with early stage non-small cell lung cancer; however, comorbidities and poor pulmonary reserve often limit the extent of resection. Limited resections are associated with a twofold to threefold increase in locoregional recurrence, suggesting that microscopic disease remains near the resection margin. We hypothesized that local delivery of paclitaxel through 100-nm expansile polymer nanoparticles (pax-eNP) immediately after tumor resection could prevent local recurrence. METHODS Primary tumors, initiated on the dorsum of C57BL/6J mice through subcutaneous injection of 750,000 Lewis lung carcinoma cells, were excised when tumor volume reached 300 mm(3). After resection, animals were randomized to receive 300 μg paclitaxel intravenously or as pax-eNP locally at the tumor resection site versus unloaded eNP or saline controls. RESULTS In all mice receiving saline, unloaded eNP, or paclitaxel intravenously, visible local tumor recurrence developed at a median of 6 days. In contrast, tumor recurrence after pax-eNP was delayed to 10 days (pax-eNP versus all other groups, Kaplan-Meier, p < 0.05). Delay in local recurrence was associated with increased survival in the pax-eNP group (16 days) versus all other groups (11 and 12 days, p < 0.05). CONCLUSIONS The pax-eNP placed at the time of surgical resection delayed local tumor recurrence and modestly prolonged survival in a murine Lewis lung carcinoma recurrence model.


ChemMedChem | 2010

Ease of Synthesis, Controllable Sizes, and In Vivo Large Animal Lymph Migration of Polymeric Nanoparticles

Kimberly Ann V. Zubris; Onkar V. Khullar; Aaron P. Griset; Summer L. Gibbs-Strauss; John V. Frangioni; Yolonda L. Colson; Mark W. Grinstaff

Polymeric nanoparticles were synthesized using both a photoinduced and base-catalyzed free radical polymerization method. These mild room temperature approaches allow sensitive molecules, such as dyes, to be encapsulated. Using this method, near infrared dye loaded nanoparticles for lymphatic migration and lymph nodes localization were synthesized. After injection into a large animal, we observed migration of the nanoparticles over 20 cm to the sentinel lymph. Nanoparticle migration was observed for nanoparticles of 50 nm but not 100 nm in diameter. These investigations further support the development of new materials and clinical approaches to treat cancer including identification of nodal disease, characterization of the extent of disease, and establishment of treatment regimes.


The Annals of Thoracic Surgery | 2011

Paclitaxel-Loaded Expansile Nanoparticles in a Multimodal Treatment Model of Malignant Mesothelioma

Morgan D. Schulz; Kimberly Ann V. Zubris; Jacqueline E. Wade; Robert F. Padera; Xiaoyin Xu; Mark W. Grinstaff; Yolonda L. Colson

BACKGROUND Malignant mesothelioma has a poor prognosis even when treated aggressively with multimodal therapy. Traditional murine tumor models can be used to evaluate drug efficacy and toxicity in malignant mesothelioma, but not to assess the effect of a multimodal approach that includes the surgical resection of tumor. We therefore developed a murine model of multimodal therapy in which we evaluated paclitaxel-loaded expansile nanoparticles (Pax-eNP) for delivering intracavitary chemotherapy in malignant mesothelioma. METHODS Paclitaxel-loaded expansile nanoparticles (Pax-eNP) of 100 nm, designed to release drug at an endosomal pH below 5, were synthesized. Xenografts of human malignant mesothelioma were established intraperitoneally in nude mice, followed by cytoreductive surgery (CRS) via laparotomy, and with omentectomy and resection of abdominal fat pads done 14 days later. At fascial closure, 10 mg/kg paclitaxel was delivered as traditional paclitaxel/paclitaxel Cremophor-EL (Pax-CE) or Pax-eNP. Morbidity and survival were assessed over a period of 90 days. RESULTS Cytoreductive surgery in mice was feasible and reproducible, and incurred less than 5% operative mortality. By itself, CRS did not significantly prolong survival; however, the addition of intraoperative Pax-CE or Pax-eNP significantly increased survival as compared with that of mice with untreated disease. In the case of Pax-eNP, the increase in survival was also statistically significant as compared with that following resection alone. CONCLUSIONS A murine model of CRS for malignant mesothelioma allows the in vivo assessment of multimodal therapy, including nanoparticle delivery. Combination therapy was superior to no treatment or CRS alone in prolonging survival. Treatment with Pax-eNP improved overall survival in the setting of CRS, suggesting that Pax-eNP merits further evaluation for intracavitary drug delivery following the surgical resection of malignant mesothelioma.


Annals of Surgical Oncology | 2013

Cytoreductive surgery and intraoperative administration of paclitaxel-loaded expansile nanoparticles delay tumor recurrence in ovarian carcinoma.

Denis M. Gilmore; Morgan D. Schulz; Rong Liu; Kimberly Ann V. Zubris; Robert F. Padera; Paul J. Catalano; Mark W. Grinstaff; Yolonda L. Colson


Journal of Surgical Research | 2012

Mechanism for Expansile Nanoparticle Uptake in Mesothelioma

Denis M. Gilmore; Kimberly Ann V. Zubris; Mark W. Grinstaff; Yolonda L. Colson

Collaboration


Dive into the Kimberly Ann V. Zubris's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yolonda L. Colson

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Rong Liu

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jacqueline E. Wade

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Morgan D. Schulz

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Robert F. Padera

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Denis M. Gilmore

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

John V. Frangioni

Beth Israel Deaconess Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge