Network


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

Hotspot


Dive into the research topics where Kathleen F. Pirollo is active.

Publication


Featured researches published by Kathleen F. Pirollo.


Trends in Biotechnology | 2008

Does a targeting ligand influence nanoparticle tumor localization or uptake

Kathleen F. Pirollo; Esther H. Chang

Inclusion of a tumor-targeting molecule in nanosized delivery systems increases their in vivo efficacy. However, the biodistribution and pharmacokinetics of the uptake of such particles have not yet been well addressed. Several recent papers have suggested that tumor-targeting ligands function primarily to increase intracellular uptake of the nanocomplex and do not influence tumor localization. However, other reports indicate that they do play a role in the accumulation in the tumor. One difference might be the presence or absence of poly-[ethylene glycol] (PEG) in the complex and its impact on the enhanced permeability and retention (EPR) effect. Further studies are clearly needed to more fully elucidate the influence of composition on tumor-targeted, systemic delivery of nanoparticles.


Molecular Therapy | 2013

Phase I Study of a Systemically Delivered p53 Nanoparticle in Advanced Solid Tumors

Neil Senzer; John J. Nemunaitis; Derek Nemunaitis; Cynthia Bedell; Gerald Edelman; Minal Barve; Robert Nunan; Kathleen F. Pirollo; Antonina Rait; Esther H. Chang

Selective delivery of therapeutic molecules to primary and metastatic tumors is optimal for effective cancer therapy. A liposomal nanodelivery complex (scL) for systemic, tumor-targeting delivery of anticancer therapeutics has been developed. scL employs an anti-transferrin receptor (TfR), scFv as the targeting molecule. Loss of p53 suppressor function, through mutations or inactivation of the p53 pathway, is present in most human cancers. Rather than being transiently permissive for tumor initiation, persistence of p53 dysfunction is a continuing requirement for maintaining tumor growth. Herein, we report results of a first-in-man Phase I clinical trial of restoration of the normal human tumor suppressor gene p53 using the scL nanocomplex (SGT-53). Minimal side effects were observed in this trial in patients with advanced solid tumors. Furthermore, the majority of patients demonstrated stable disease. One patient with adenoid cystic carcinoma had his status changed from unresectable to resectable after one treatment cycle. More significantly, we observed an accumulation of the transgene in metastatic tumors, but not in normal skin tissue, in a dose-related manner. These results show not only that systemically delivered SGT-53 is well tolerated and exhibits anticancer activity, but also supply evidence of targeted tumor delivery of SGT-53 to metastatic lesions.


Molecular Medicine | 2001

Systemic p53 gene therapy of cancer with immunolipoplexes targeted by anti-transferrin receptor scFv.

Liang Xu; Wenhua Tang; Cheng-Cheng Huang; William Alexander; Lai-Man Xiang; Kathleen F. Pirollo; Antonina Rait; Esther H. Chang

BackgroundA long-standing goal in genetic therapy for cancer is a systemic gene delivery system that selectively targets tumor cells, including metastases. Here we describe a novel cationic immunolipoplex system that shows high in vivo gene transfer efficiency and antitumor efficacy when used for systemic p53 gene therapy of cancer.Materials and MethodsA cationic immunolipoplex incorporating a biosynthetically lipid-tagged, anti-transferrin receptor single-chain antibody (TfRscFv), was designed to target tumor cells both in vitro and in vivo. A human breast cancer metastasis model was employed to evaluate the in vivo efficacy of systemically administered, TfRscFv-immunolipoplex-mediated, p53 gene therapy in combination with docetaxel.ResultsThe TfRscFv-targeting cationic immunolipoplex had a size of 60–100 nm, showed enhanced tumor cell binding, and improved targeted gene delivery and transfection efficiencies, both in vitro and in vivo. The p53 tumor suppressor gene was not only systemically delivered by the immunolipoplex to human tumor xenografts in nude mice but also functionally expressed. In the nude mouse breast cancer metastasis model, the combination of the p53 gene delivered by the systemic administration of the TfRscFv-immunolipoplex and docetaxel resulted in significantly improved efficacy with prolonged survival.ConclusionsThis is the first report using scFv-targeting immunolipoplexes for systemic gene therapy. The TfRscFv has a number of advantages over the transferrin (Tf) molecule itself: (1) scFv has a much smaller size than Tf producing a smaller immunolipoplex giving better penetration into solid tumors; (2) unlike Tf, the scFv is a recombinant protein, not a blood product; (3) large scale production and strict quality control of the recombinant scFv, as well as scFv-immunolipoplex, are feasible. The sensitization of tumors to chemotherapy by this tumor-targeted and efficient p53 gene delivery method could lower the effective dose of the drug, correspondingly lessening the severe side effects, while decreasing the possibility of recurrence. Moreover, this approach is applicable to both primary and recurrent tumors, and more significantly, metastatic disease. The TfRscFv-targeting of cationic immunolipoplexes is a promising method of tumor targeted gene delivery that can be used for systemic gene therapy of cancer with the potential to critically impact the clinical management of cancer.


Journal of Controlled Release | 2001

Tumor-targeted p53-gene therapy enhances the efficacy of conventional chemo/radiotherapy

Liang Xu; Kathleen F. Pirollo; Esther H. Chang

A long-standing goal in gene therapy for cancer is a stable, low toxic, systemic gene delivery system that selectively targets tumor cells, including metastatic disease. Progress has been made toward developing non-viral, pharmaceutical formulations of genes for in vivo human therapy, particularly cationic liposome-mediated gene transfer systems. Ligand-directed tumor targeting of cationic liposome-DNA complexes (lipoplexes) is showing promise for targeted gene delivery and systemic gene therapy. Lipoplexes directed by ligands such as folate, transferrin or anti-transferrin receptor scFv, showed tumor-targeted gene delivery and expression in human breast, prostate, head and neck cancers. The two elements, ligand/receptor and liposome composition, work together to realize the goal of functional tumor targeting of gene therapeutics. The tumor suppressor gene, p53, has been shown to be involved in the control of DNA damage-induced apoptosis. Loss or malfunction of this p53-mediated apoptotic pathway has been proposed as one mechanism by which tumors become resistant to chemotherapy or radiation. The systemically delivered ligand-liposome-p53 gene therapeutics resulted in efficient expression of functional wild-type p53, sensitizing the tumors to chemotherapy and radiotherapy. This is a novel strategy combining current molecular medicine with conventional chemotherapy and radiotherapy for the treatment of cancer. The systemic delivery of normal tumor suppressor gene p53 by a non-viral, tumor-targeted delivery system as a new therapeutic intervention has the potential to critically impact the clinical management of cancer.


Pharmacology & Therapeutics | 2003

Antisense therapeutics: from theory to clinical practice

Kathleen F. Pirollo; Antonina Rait; Leanne S. Sleer; Esther H. Chang

The use of antisense (AS) oligonucleotides as therapeutic agents was proposed as far back as the 1960s/1970s when the AS strategy was initially developed. However, it has taken almost a quarter of a century for this potential to be realized. The last few years has seen a rapid increase in the number of AS molecules progressing past Phase I in clinical trials, due in part to our increased knowledge of their structure and chemistry. Here, we describe the most prominent of these modifications with respect to clinical applicability. However, the main focus of this review is clinical application, with a focus on cancer. We will discuss in detail both the status of the current AS clinical trials and the molecules that are likely to be the targets of the next group of AS molecules entering the clinic.


Nucleosides, Nucleotides & Nucleic Acids | 2006

Chemically modified short interfering hybrids (siHYBRIDS): nanoimmunoliposome delivery in vitro and in vivo for RNAi of HER-2.

Richard I. Hogrefe; Alexandre V. Lebedev; Gerald Zon; Kathleen F. Pirollo; Antonina Rait; Qi Zhou; Wei Yu; Esther H. Chang

A blunt-ended 19-mer short interfering hybrid (siHybrid) (H) comprised of sense-DNA/antisense-RNA targeting HER-2 mRNA was encapsulated in a liposomal nanoplex with anti-transferrin receptor single-chain antibody fragment (TfRscFv) as the targeting moiety for clinically relevant tumor-specific delivery. In vitro delivery to a human pancreatic cell line (PANC-1) was shown to exhibit sequence-specific inhibition of 48-h cell growth with an IC50 value of 37 nM. The inhibitory potency of this siHybrid was increased (IC50 value of 7.8 nM) using a homologous chemically modified siHybrid (mH) in which the 19-mer sense strand had the following pattern of 2 ′-deoxyinosine (dI) and 2 ′-O-methylribonucleotide (2 ′-OMe) residues: 5′-d(TITIT)-2′OMe(GCGGUGGUU)-d(GICIT). These modifications were intended to favor antisense strand-mediated RNAi while mitigating possible sense strand-mediated off-target effects and RNase H-mediated cleavage of the antisense RNA strand. The presently reported immunoliposomal delivery system was successfully used in vivo to inhibit HER-2 expression, and thus induce apoptosis in human breast carcinoma tumors (MDA-MB-435) in mice upon repeated i.v. treatment at a dose of 3 mg/kg of H or mH. The in vivo potency of modified siHybrid mH appeared to be qualitatively greater than that of H, as was the case in vitro.


Nanomedicine: Nanotechnology, Biology and Medicine | 2008

Nanoimmunoliposome delivery of superparamagnetic iron oxide markedly enhances targeting and uptake in human cancer cells in vitro and in vivo

Chengli Yang; Antonina Rait; Kathleen F. Pirollo; John A. Dagata; Natalia Farkas; Esther H. Chang

To circumvent the problem of reduction of the supermagnetic properties of superparamagnetic iron oxide (SPIO) nanoparticles after chemical modification to conjugate targeting molecules, we have adapted a tumor-targeting nanoimmunoliposome platform technology (scL) to encapsulate and deliver SPIO (scL-SPIO) in vitro and in vivo without chemical modification. Scanning probe microscopy, confocal microscopy, and Prussian blue staining were used to analyze the scL-SPIO and assess intracellular uptake and distribution of SPIO in vitro. In vivo targeting and tumor-specific uptake of scL-SPIO was examined using fluorescent-labeled SPIO. We demonstrated that SPIO encapsulation in the scL complex results in an approximately 11-fold increase in SPIO uptake in human cancer cells in vitro, with distribution to cytoplasm and nucleus. Moreover, the scL nanocomplex specifically and efficiently delivered SPIO into tumor cells after systemic administration, demonstrating the potential of this approach to enhance local tumor concentration and the utility of SPIO for clinical applications.


Cancer Gene Therapy | 2001

Downmodulation of bFGF-binding protein expression following restoration of p53 function.

Zaki A. Sherif; Shigeru Nakai; Kathleen F. Pirollo; Antonina Rait; Esther H. Chang

Angiogenesis is a requirement for solid tumor growth. Therefore, inhibition of this neovascularization is one mechanism by which restoration of wtp53 function may lead to tumor regression. Here we report that adenoviral vector–mediated wild-type p53 transduction results in growth inhibition of squamous cell carcinoma of the head and neck tumor cells both in vitro and in a xenograft mouse model. This growth inhibition is associated with the down-regulation of the expression of fibroblast growth factor binding protein, a secreted protein required for the activation of angiogenic factor basic FGF. These findings suggest that wtp53-induced tumor regression is due, at least in part, to antiangiogenesis mediated by the downmodulation of fibroblast growth factor binding protein. Cancer Gene Therapy (2001) 8, 771–782


Molecular Therapy | 2014

The Clinical Potential of Targeted Nanomedicine: Delivering to Cancer Stem-like Cells

Sang-Soo Kim; Antonina Rait; Farwah Rubab; Abhi K Rao; Michael C Kiritsy; Kathleen F. Pirollo; Shangzi Wang; Louis M. Weiner; Esther H. Chang

Cancer stem-like cells (CSCs) have been implicated in recurrence and treatment resistance in many human cancers. Thus, a CSC-targeted drug delivery strategy to eliminate CSCs is a desirable approach for developing a more effective anticancer therapy. We have developed a tumor-targeting nanodelivery platform (scL) for systemic administration of molecular medicines. Following treatment with the scL nanocomplex carrying various payloads, we have observed exquisite tumor-targeting specificity and significant antitumor response with long-term survival benefit in numerous animal models. We hypothesized that this observed efficacy might be attributed, at least in part, to elimination of CSCs. Here, we demonstrate the ability of scL to target both CSCs and differentiated nonstem cancer cells (non-CSCs) in various mouse models including subcutaneous and intracranial xenografts, syngeneic, and chemically induced tumors. We also show that systemic administration of scL carrying the wtp53 gene was able to induce tumor growth inhibition and the death of both CSCs and non-CSCs in subcutaneous colorectal cancer xenografts suggesting that this could be an effective method to reduce cancer recurrence and treatment resistance. This scL nanocomplex is being evaluated in a number of clinical trials where it has been shown to be well tolerated with indications of anticancer activity.


Academic Radiology | 2009

Nanodelivery of MRI contrast agent enhances sensitivity of detection of lung cancer metastases.

Matthew L. Freedman; Esther H. Chang; Qi Zhou; Kathleen F. Pirollo

RATIONALE AND OBJECTIVESnEarly detection of lung cancer can be problematic. Although current imaging methods can identify lung cancers, they are limited in the size of detectable nodules. There is also lack of evidence that these methods can correctly classify nodules <7 mm as malignant because lung cancer can be mimicked in appearance by benign lesions that lower specificity. Therefore, there is a need for enhanced sensitivity/specificity of detection for small lung cancers.nnnMATERIALS AND METHODSnWe have developed a nanosized ( approximately 100 nm) immunoliposome complex for delivery of molecular medicines to tumors. In this complex, an anti-transferrin receptor single-chain antibody fragment (TfRscFv) decorates the surface of a cationic liposome encapsulating the payload. We have previously shown that this systemically administered complex (scL) selectively targets, and efficiently delivers its payload into, tumor cells. We have also encapsulated the magnetic resonance imaging (MRI) contrast agent gadopentetate dimeglumine (gad-d) within this complex, resulting in increased resolution and image intensity in a mouse model of primary cancer. Here we examine the ability of the scL-gad-d complex to increase the sensitivity of detection of lung metastases.nnnRESULTSnThese MRI studies show that the scL-gad-d nanocomplex is able to improve detection, and increase enhancement of, small lung cancers (400 microm and as small as 100 microm) compared to that of uncomplexed gad-d.nnnCONCLUSIONSnBecause of its tumor targeting specificity, deliver of an MRI contrast agent via this nanocomplex has potential for use as an agent that can identify small lung cancers, thus improving early detection and possibly increasing survival.

Collaboration


Dive into the Kathleen F. Pirollo's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Liang Xu

University of Kansas

View shared research outputs
Top Co-Authors

Avatar

John A. Dagata

National Institute of Standards and Technology

View shared research outputs
Top Co-Authors

Avatar

Qi Zhou

Georgetown University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Arlene O. Siefker-Radtke

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Charles C. Guo

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Natalia Farkas

National Institute of Standards and Technology

View shared research outputs
Top Co-Authors

Avatar

Randall E. Millikan

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge