1
|
Young O, Ngo N, Lin L, Stanbery L, Creeden JF, Hamouda D, Nemunaitis J. Folate Receptor as a Biomarker and Therapeutic Target in Solid Tumors. Curr Probl Cancer 2023; 47:100917. [PMID: 36508886 DOI: 10.1016/j.currproblcancer.2022.100917] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 10/21/2022] [Accepted: 10/27/2022] [Indexed: 11/18/2022]
Abstract
Folate is a B vitamin necessary for basic biological functions, including rapid cell turnover occurring in cancer cell proliferation. Though the role of folate as a causative versus protective agent in carcinogenesis is debated, several studies have indicated that the folate receptor (FR), notably subtype folate receptor alpha (FRα), could be a viable biomarker for diagnosis, progression, and prognosis. Several cancers, including gastrointestinal, gynecological, breast, lung, and squamous cell head and neck cancers overexpress FR and are currently under investigation to correlate receptor status to disease state. Traditional chemotherapies have included antifolate medications, such as methotrexate and pemetrexed, which generate anticancer activity during the synthesis phase of the cell cycle. Increasingly, the repertoire of pharmacotherapies is expanding to include FR as a target, with a heterogenous pool of directed therapies. Here we discuss the FR, expression and effect in cancer biology, and relevant pharmacologic inhibitors.
Collapse
Affiliation(s)
- Olivia Young
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Nealie Ngo
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Leslie Lin
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | | | - Justin Fortune Creeden
- Department of Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Danae Hamouda
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | | |
Collapse
|
2
|
Gunaratne PH, Pan Y, Rao AK, Lin C, Hernandez‐Herrera A, Liang K, Rait AS, Venkatanarayan A, Benham AL, Rubab F, Kim SS, Rajapakshe K, Chan CK, Mangala LS, Lopez‐Berestein G, Sood AK, Rowat AC, Coarfa C, Pirollo KF, Flores ER, Chang EH. Activating p53 family member TAp63: A novel therapeutic strategy for targeting p53-altered tumors. Cancer 2019; 125:2409-2422. [PMID: 31012964 PMCID: PMC6617807 DOI: 10.1002/cncr.32053] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 11/25/2018] [Accepted: 12/17/2018] [Indexed: 01/13/2023]
Abstract
BACKGROUND Over 96% of high-grade ovarian carcinomas and 50% of all cancers are characterized by alterations in the p53 gene. Therapeutic strategies to restore and/or reactivate the p53 pathway have been challenging. By contrast, p63, which shares many of the downstream targets and functions of p53, is rarely mutated in cancer. METHODS A novel strategy is presented for circumventing alterations in p53 by inducing the tumor-suppressor isoform TAp63 (transactivation domain of tumor protein p63) through its direct downstream target, microRNA-130b (miR-130b), which is epigenetically silenced and/or downregulated in chemoresistant ovarian cancer. RESULTS Treatment with miR-130b resulted in: 1) decreased migration/invasion in HEYA8 cells (p53 wild-type) and disruption of multicellular spheroids in OVCAR8 cells (p53-mutant) in vitro, 2) sensitization of HEYA8 and OVCAR8 cells to cisplatin (CDDP) in vitro and in vivo, and 3) transcriptional activation of TAp63 and the B-cell lymphoma (Bcl)-inhibitor B-cell lymphoma 2-like protein 11 (BIM). Overexpression of TAp63 was sufficient to decrease cell viability, suggesting that it is a critical downstream effector of miR-130b. In vivo, combined miR-130b plus CDDP exhibited greater therapeutic efficacy than miR-130b or CDDP alone. Mice that carried OVCAR8 xenograft tumors and were injected with miR-130b in 1,2-dioleoyl-sn-glycero-3-phosphatidylcholine (DOPC) liposomes had a significant decrease in tumor burden at rates similar to those observed in CDDP-treated mice, and 20% of DOPC-miR-130b plus CDDP-treated mice were living tumor free. Systemic injections of scL-miR-130b plus CDDP in a clinically tested, tumor-targeted nanocomplex (scL) improved survival in 60% and complete remissions in 40% of mice that carried HEYA8 xenografts. CONCLUSIONS The miR-130b/TAp63 axis is proposed as a new druggable pathway that has the potential to uncover broad-spectrum therapeutic options for the majority of p53-altered cancers.
Collapse
Affiliation(s)
- Preethi H. Gunaratne
- Department of Biochemistry and BiologyUniversity of HoustonHoustonTexas
- Department of Molecular and Cell BiologyBaylor College of MedicineHoustonTexas
- Human Genome Sequencing CenterBaylor College of MedicineHoustonTexas
- Lester and Sue Smith Breast CenterBaylor College of MedicineHoustonTexas
| | - Yinghong Pan
- Department of Biochemistry and BiologyUniversity of HoustonHoustonTexas
- UPMC Genome CenterPittsburghPennsylvania
| | - Abhi K. Rao
- Department of Oncology, Lombardi Comprehensive Cancer CenterGeorgetown UniversityWashingtonDistrict of Columbia
| | - Chunru Lin
- Department of Molecular and Cellular Oncology, Division of Basic ScienceThe University of Texas MD Anderson Cancer CenterHoustonTexas
| | | | - Ke Liang
- Department of Molecular and Cellular Oncology, Division of Basic ScienceThe University of Texas MD Anderson Cancer CenterHoustonTexas
| | - Antonina S. Rait
- Department of Oncology, Lombardi Comprehensive Cancer CenterGeorgetown UniversityWashingtonDistrict of Columbia
| | - Avinashnarayan Venkatanarayan
- Department of Molecular and Cellular Oncology, Division of Basic ScienceThe University of Texas MD Anderson Cancer CenterHoustonTexas
- Genentech, Inc.South San FranciscoCalifornia
| | - Ashley L. Benham
- Department of Biochemistry and BiologyUniversity of HoustonHoustonTexas
- 10X Genomics Inc.PleasantonCalifornia
| | | | - Sang Soo Kim
- Department of Oncology, Lombardi Comprehensive Cancer CenterGeorgetown UniversityWashingtonDistrict of Columbia
- SynerGene Therapeutics, Inc.PotomacMaryland
| | - Kimal Rajapakshe
- Department of Molecular and Cell BiologyBaylor College of MedicineHoustonTexas
| | - Clara K. Chan
- Department of Integrative Biology and PhysiologyUniversity of CaliforniaLos AngelesCalifornia
| | - Lingegowda S. Mangala
- Gynecologic Oncology and Reproductive MedicineThe University of Texas MD Anderson Cancer CenterHoustonTexas
- Center for RNAi and Non-Coding RNAsThe University of Texas MD Anderson Cancer CenterHoustonTexas
| | - Gabriel Lopez‐Berestein
- Center for RNAi and Non-Coding RNAsThe University of Texas MD Anderson Cancer CenterHoustonTexas
- Department of Experimental TherapeuticsThe University of Texas MD Anderson Cancer CenterHoustonTexas
| | - Anil K. Sood
- Gynecologic Oncology and Reproductive MedicineThe University of Texas MD Anderson Cancer CenterHoustonTexas
- Center for RNAi and Non-Coding RNAsThe University of Texas MD Anderson Cancer CenterHoustonTexas
| | - Amy C. Rowat
- Department of Integrative Biology and PhysiologyUniversity of CaliforniaLos AngelesCalifornia
| | - Cristian Coarfa
- Department of Molecular and Cell BiologyBaylor College of MedicineHoustonTexas
| | - Kathleen F. Pirollo
- Department of Oncology, Lombardi Comprehensive Cancer CenterGeorgetown UniversityWashingtonDistrict of Columbia
| | - Elsa R. Flores
- Department of Molecular and Cellular Oncology, Division of Basic ScienceThe University of Texas MD Anderson Cancer CenterHoustonTexas
- Department of Molecular OncologyCancer Biology and Evolution Program, Moffitt Cancer CenterTampaFlorida
| | - Esther H. Chang
- Department of Oncology, Lombardi Comprehensive Cancer CenterGeorgetown UniversityWashingtonDistrict of Columbia
- SynerGene Therapeutics, Inc.PotomacMaryland
| |
Collapse
|
3
|
Zhao Y, Chen H, Chen X, Hollett G, Gu Z, Wu J, Liu X. Targeted nanoparticles for head and neck cancers: overview and perspectives. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2017; 9. [PMID: 28387452 DOI: 10.1002/wnan.1469] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 02/14/2017] [Accepted: 02/25/2017] [Indexed: 11/11/2022]
Abstract
Head and neck cancer (HNC) is common in several regions and is associated with high morbidity and mortality worldwide. This review summarizes the recent progress in the development of targeted nanoparticle systems for HNC therapy. WIREs Nanomed Nanobiotechnol 2017, 9:e1469. doi: 10.1002/wnan.1469 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Yuying Zhao
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, PR China.,Key Laboratory of Sensing Technology and Biomedical Instrument of Guangdong Province, School of Engineering, Sun Yat-sen University, Guangzhou, PR China
| | - Haolin Chen
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, PR China.,Key Laboratory of Sensing Technology and Biomedical Instrument of Guangdong Province, School of Engineering, Sun Yat-sen University, Guangzhou, PR China
| | - Xing Chen
- Key Laboratory of Sensing Technology and Biomedical Instrument of Guangdong Province, School of Engineering, Sun Yat-sen University, Guangzhou, PR China
| | - Geoffrey Hollett
- Materials Science and Engineering Program, University of California San Diego, La Jolla, CA, USA
| | - Zhipeng Gu
- Key Laboratory of Sensing Technology and Biomedical Instrument of Guangdong Province, School of Engineering, Sun Yat-sen University, Guangzhou, PR China
| | - Jun Wu
- Key Laboratory of Sensing Technology and Biomedical Instrument of Guangdong Province, School of Engineering, Sun Yat-sen University, Guangzhou, PR China.,Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou, PR China
| | - Xiqiang Liu
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, PR China
| |
Collapse
|
4
|
Wu TT, Zhou SH. Nanoparticle-based targeted therapeutics in head-and-neck cancer. Int J Med Sci 2015; 12:187-200. [PMID: 25589895 PMCID: PMC4293184 DOI: 10.7150/ijms.10083] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 12/30/2014] [Indexed: 12/17/2022] Open
Abstract
Head-and-neck cancer is a major form of the disease worldwide. Treatment consists of surgery, radiation therapy and chemotherapy, but these have not resulted in improved survival rates over the past few decades. Versatile nanoparticles, with selective tumor targeting, are considered to have the potential to improve these poor outcomes. Application of nanoparticle-based targeted therapeutics has extended into many areas, including gene silencing, chemotherapeutic drug delivery, radiosensitization, photothermal therapy, and has shown much promise. In this review, we discuss recent advances in the field of nanoparticle-mediated targeted therapeutics for head-and-neck cancer, with an emphasis on the description of targeting points, including future perspectives.
Collapse
Affiliation(s)
- Ting-Ting Wu
- Department of Otolaryngology, The First Affiliated Hospital, College of Medicine, Zhejiang University, 310003, China
| | - Shui-Hong Zhou
- Department of Otolaryngology, The First Affiliated Hospital, College of Medicine, Zhejiang University, 310003, China
| |
Collapse
|
5
|
Liu K, Chen H, You Q, Shi H, Wang Z. The siRNA cocktail targeting VEGF and HER2 inhibition on the proliferation and induced apoptosis of gastric cancer cell. Mol Cell Biochem 2013; 386:117-24. [PMID: 24158524 PMCID: PMC3889296 DOI: 10.1007/s11010-013-1850-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 09/27/2013] [Indexed: 12/14/2022]
Abstract
The aim of this study was to investigate the inhibitory effect of a siRNA cocktail targeting Vascular endothelial growth factor (VEGF) and Human epidermal growth factor receptor 2 (HER2) on cell proliferation, induced apoptosis and the expression of VEGF and HER2 in human gastric carcinoma cell. The silencing rate of pre-designed siRNAs that targeted VEGF and HER2 was detected by Real-time Quantitative PCR (RT-QPCR) analysis. Furthermore, the best silencing siRNA that targeted VEGF and HER2 was prepared as a cocktail to co-knockdown VEGF and HER2 expression at both mRNA and protein levels which were detected by RT-QPCR and Western blot analysis. Cell proliferation inhibition rates were determined by CCK8 assay. The effect of siRNA cocktail on cell apoptosis was determined by flow cytometry. The migration inhibition of siRNA cocktail was analyzed by wound-healing assay. The ability of VEGF to induce endothelial cells to proliferate was examined in HUVECs by the method of tube formation assay. The pre-designed siRNAs could inhibit VEGF and HER2 mRNA level. siRNA cocktail, and co-downregulation of VEGF and HER2 result in significant inhibition of gastric cancer growth and migration in vitro. The inhibition of VEGF and HER2 expressions can induce apoptosis of SGC-7901 cells.
Collapse
Affiliation(s)
- Kun Liu
- Department of Cardiothoracic Surgery, The Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, Jiangsu, People's Republic of China
| | | | | | | | | |
Collapse
|
6
|
Tse GM, Yu KH, Chan AWH, King AD, Chen GG, Wong KT, Tsang RKY, Chan ABW. HER2 expression predicts improved survival in patients with cervical node-positive head and neck squamous cell carcinoma. Otolaryngol Head Neck Surg 2009; 141:467-73. [PMID: 19786214 DOI: 10.1016/j.otohns.2009.06.747] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2008] [Revised: 06/23/2009] [Accepted: 06/30/2009] [Indexed: 10/20/2022]
Abstract
OBJECTIVE To investigate the prognostic value of HER2 and p63 expression in head and neck squamous cell carcinoma (HNSCC). STUDY DESIGN A case review of 186 HNSCCs from the oral tongue, palate, maxillary sinus, floor of mouth, oropharynx, hypopharynx, and larynx. SUBJECTS AND METHODS All primary tumor specimens were evaluated by immunohistochemistry for HER2 and p63 expressions, which were correlated with clinical parameters including age, sex, grade, lymph node metastases, stage, and survival. RESULTS One hundred forty-one patients had stage III-IV disease and 109 had lymph node metastases. For all cases, T and N stages were significant prognostic predictors for both overall and disease-free survivals. In the node-positive subgroup, T stage and HER2 expression were significant prognostic predictors for both overall and disease-free survivals. CONCLUSION HER2 may be associated with longer survival in node-positive patients with HNSCC.
Collapse
Affiliation(s)
- Gary M Tse
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.
| | | | | | | | | | | | | | | |
Collapse
|
7
|
Nanodelivery of MRI contrast agent enhances sensitivity of detection of lung cancer metastases. Acad Radiol 2009; 16:627-37. [PMID: 19345904 DOI: 10.1016/j.acra.2008.12.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2007] [Revised: 12/05/2008] [Accepted: 12/15/2008] [Indexed: 11/21/2022]
Abstract
RATIONALE AND OBJECTIVES Early 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. MATERIALS AND METHODS We 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. RESULTS These 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. CONCLUSIONS Because 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.
Collapse
|
8
|
Puri A, Loomis K, Smith B, Lee JH, Yavlovich A, Heldman E, Blumenthal R. Lipid-based nanoparticles as pharmaceutical drug carriers: from concepts to clinic. Crit Rev Ther Drug Carrier Syst 2009; 26:523-80. [PMID: 20402623 PMCID: PMC2885142 DOI: 10.1615/critrevtherdrugcarriersyst.v26.i6.10] [Citation(s) in RCA: 553] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
In recent years, various nanotechnology platforms in the area of medical biology, including both diagnostics and therapy, have gained remarkable attention. Moreover, research and development of engineered multifunctional nanoparticles as pharmaceutical drug carriers have spurred exponential growth in applications to medicine in the last decade. Design principles of these nanoparticles, including nanoemulsions, dendrimers, nano-gold, liposomes, drug-carrier conjugates, antibody-drug complexes, and magnetic nanoparticles, are primarily based on unique assemblies of synthetic, natural, or biological components, including but not limited to synthetic polymers, metal ions, oils, and lipids as their building blocks. However, the potential success of these particles in the clinic relies on consideration of important parameters such as nanoparticle fabrication strategies, their physical properties, drug loading efficiencies, drug release potential, and, most importantly, minimum toxicity of the carrier itself. Among these, lipid-based nanoparticles bear the advantage of being the least toxic for in vivo applications, and significant progress has been made in the area of DNA/RNA and drug delivery using lipid-based nanoassemblies. In this review, we will primarily focus on the recent advances and updates on lipid-based nanoparticles for their projected applications in drug delivery. We begin with a review of current activities in the field of liposomes (the so-called honorary nanoparticles), and challenging issues of targeting and triggering will be discussed in detail. We will further describe nanoparticles derived from a novel class of amphipathic lipids called bolaamphiphiles with unique lipid assembly features that have been recently examined as drug/DNA delivery vehicles. Finally, an overview of an emerging novel class of particles (based on lipid components other than phospholipids), solid lipid nanoparticles and nanostructured lipid carriers will be presented. We conclude with a few examples of clinically successful formulations of currently available lipid-based nanoparticles.
Collapse
Affiliation(s)
- Anu Puri
- Center for Cancer Research Nanobiology Program, National Cancer Institute at Frederick, National Institutes of Health, Frederick, MD 21702-1201, USA.
| | | | | | | | | | | | | |
Collapse
|
9
|
Abstract
Targeted delivery via selective cellular markers can potentially increase the efficacy and reduce the toxicity of therapeutic agents. The folate receptor (FR) has two glycosyl phosphatidylinositol (GPI)-anchored isoforms, alpha and beta. FR-alpha expression is frequently amplified in epithelial cancers, whereas FR-beta expression is found in myeloid leukemia and activated macrophages associated with chronic inflammatory diseases. Conjugates of folic acid and anti-FR antibodies can be taken up by cancer cells via receptor-mediated endocytosis, thus providing a mechanism for targeted delivery to FR+ cells. The aim of this article is to provide a brief overview of applications of FR targeting in drug delivery, with an emphasis on the strategy of using folate as a targeting ligand. In order to do this, recent literature is surveyed on targeted delivery via both FR sub-types, as well as new findings on selective receptor upregulation in the targeted cells. A wide variety of molecules and drug carriers, including imaging agents, chemotherapeutic agents, oligonucleotides, proteins, haptens, liposomes, nanoparticles and gene transfer vectors have been conjugated to folate and evaluated for FR-targeted delivery. Substantial targeting efficacy has been found both in vitro and in vivo. In addition, mechanisms and methods for selective FR upregulation have been uncovered, which might enhance the effectiveness of the FR-targeted delivery strategy. FR-alpha serves as a useful marker for cancer, whereas FR-beta serves as a marker for myeloid leukemia and chronic inflammatory diseases. FR-targeted agents have shown promising efficacy in preclinical models and significant potential for future clinical application in a wide range of diseases.
Collapse
Affiliation(s)
- Xiaobin Zhao
- Abbott Laboratories, Global Pharmaceutics and Life Cycle Technology, Abbott Park, IL 60064, USA
| | | | | |
Collapse
|
10
|
Pirollo KF, Rait A, Zhou Q, Hwang SH, Dagata JA, Zon G, Hogrefe RI, Palchik G, Chang EH. Materializing the potential of small interfering RNA via a tumor-targeting nanodelivery system. Cancer Res 2007; 67:2938-43. [PMID: 17409398 DOI: 10.1158/0008-5472.can-06-4535] [Citation(s) in RCA: 160] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The field of small interfering RNA (siRNA) as potent sequence-selective inhibitors of transcription is rapidly developing. However, until now, low transfection efficiency, poor tissue penetration, and nonspecific immune stimulation by in vivo administered siRNAs have delayed their therapeutic application. Their potential as anticancer therapeutics hinges on the availability of a vehicle that can be systemically administered, safely and repeatedly, and will deliver the siRNA specifically and efficiently to the tumor, both primary tumors and metastases. We have developed a nanosized immunoliposome-based delivery complex (scL) that, when systemically administered, will preferentially target and deliver molecules useful in gene medicine, including plasmid DNA and antisense oligonucleotides, to tumor cells wherever they occur in the body. This tumor-targeting nanoparticle delivery vehicle can also deliver siRNA to both primary and metastatic disease. We have also enhanced the efficiency of this complex by the inclusion of a pH-sensitive histidine-lysine peptide in the complex (scL-HoKC) and by delivery of a modified hybrid (DNA-RNA) anti-HER-2 siRNA molecule. Scanning probe microscopy confirms that this modified complex maintains its nanoscale size. More importantly, we show that this nanoimmunoliposome anti-HER-2 siRNA complex can sensitize human tumor cells to chemotherapeutics, silence the target gene and affect its downstream pathway components in vivo, and significantly inhibit tumor growth in a pancreatic cancer model. Thus, this complex has the potential to help translate the potent effects of siRNA into a clinically viable anticancer therapeutic.
Collapse
Affiliation(s)
- Kathleen F Pirollo
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Molecular Mechanisms of ErbB2-Mediated Breast Cancer Chemoresistance. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 608:119-29. [DOI: 10.1007/978-0-387-74039-3_9] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
12
|
Hogrefe RI, Lebedev AV, Zon G, Pirollo KF, Rait A, Zhou Q, Yu W, Chang EH. Chemically modified short interfering hybrids (siHYBRIDS): nanoimmunoliposome delivery in vitro and in vivo for RNAi of HER-2. NUCLEOSIDES NUCLEOTIDES & NUCLEIC ACIDS 2006; 25:889-907. [PMID: 16901821 DOI: 10.1080/15257770600793885] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
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.
Collapse
|
13
|
Abstract
Despite significant advances that have been made in recent years, there is still an urgent need for novel, more effective and less toxic therapeutics for human cancer. Among many new molecular therapeutics being explored for cancer therapy, antisense oligonucleotides are a promising nucleic acid-based approach, with numerous antisense agents being evaluated in preclinical studies and several anticancer antisense drugs in clinical trials. Although there are still a few problems facing the development of antisense strategies for cancer therapy, with progress made in chemical modifications, target selection and drug delivery systems, antisense oligonucleotides are emerging as a novel approach to cancer therapy used alone or in combination with conventional treatments such as chemotherapy and radiation therapy.
Collapse
Affiliation(s)
- Elizabeth Rose Rayburn
- University of Alabama at Birmingham, Department of Pharmacology and Toxicology, Division of Clinical Pharmacology, VH 113, 1670 University Blvd., Birmingham, AL 35294, USA
| | | | | |
Collapse
|
14
|
Szumiel I. Epidermal growth factor receptor and DNA double strand break repair: the cell's self-defence. Cell Signal 2006; 18:1537-48. [PMID: 16713182 DOI: 10.1016/j.cellsig.2006.03.010] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2006] [Accepted: 03/21/2006] [Indexed: 01/27/2023]
Abstract
The purpose of this review is to discuss the relation between the repair of DNA double strand breaks (DSB)--the main lethal lesion inflicted by ionising radiation-and the function of receptors of epidermal growth factor (EGFR) and similar ligands (other members of the ERBB family). The reviewed experimental data support the assumption that in mammalian cells, one consequence of EGFR/ERBB activation by X-rays is its internalisation and nuclear translocation together with DNA-dependent protein kinase (DNA-PK) subunits present in lipid rafts or cytoplasm. The effect of EGFR/ERBB stimulation on DSB rejoining would be due to an increase in the nuclear content of DNA-PK subunits and hence, in activity increase of the DNA-PK dependent non-homologous end-joining (D-NHEJ) system. Such mechanism explains the radiosensitising action of "membrane-active drugs", hypertonic media, and other agents that affect nuclear translocation of proteins. Also, one radiosensitising effect of the recently introduced into clinical practice EGFR/ERBB inhibitors would consist on counteracting the nuclear translocation of DNA-PK subunits. In result, D-NHEJ may be less active in inhibitor-treated cells and this will contribute to an enhanced lethal effect of irradiation. The reviewed observations point to a heretofore not understood mechanism of the cell's self-defence against X-rays which can be exploited in combined radio- and chemotherapy.
Collapse
Affiliation(s)
- Irena Szumiel
- Department of Radiobiology and Health Protection, Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195 Warszawa, Poland.
| |
Collapse
|
15
|
Rayburn E, Wang W, Zhang R, Wang H. Antisense approaches in drug discovery and development. PROGRESS IN DRUG RESEARCH. FORTSCHRITTE DER ARZNEIMITTELFORSCHUNG. PROGRES DES RECHERCHES PHARMACEUTIQUES 2006; 63:227-74. [PMID: 16265883 DOI: 10.1007/3-7643-7414-4_10] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Elizabeth Rayburn
- Department of Pharmacology and Toxicology, Division of Clinical Pharmacology, University of Alabama at Birmingham, VH 112, Box 600, 1670 University Blvd., Birmingham, AL 35294-0019, USA
| | | | | | | |
Collapse
|
16
|
Pirollo KF, Dagata J, Wang P, Freedman M, Vladar A, Fricke S, Ileva L, Zhou Q, Chang EH. A Tumor-Targeted Nanodelivery System to Improve Early MRI Detection of Cancer. Mol Imaging 2006. [DOI: 10.2310/7290.2006.00005] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
| | - John Dagata
- National Institute of Standards and Technology
| | | | | | | | | | | | - Qi Zhou
- Georgetown University Medical Center
| | | |
Collapse
|
17
|
Pirollo KF, Zon G, Rait A, Zhou Q, Yu W, Hogrefe R, Chang EH. Tumor-Targeting Nanoimmunoliposome Complex for Short Interfering RNA Delivery. Hum Gene Ther 2006; 17:117-24. [PMID: 16409130 DOI: 10.1089/hum.2006.17.117] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The potential of short interfering RNA (siRNA) to be developed for therapeutic use against cancer depends on the availability of an efficient tumor-specific delivery vehicle. We have previously shown that a nanoscale nonviral liposome-based complex that includes an anti-transferrin receptor single-chain antibody fragment as the targeting moiety can, when systemically administered, specifically and efficiently target primary and metastatic tumors and deliver molecules useful in gene medicine, including plasmid DNA and antisense oligonucleotides. Here we explore the ability of this complex to deliver a fluorescein-labeled siRNA to tumor cells in vivo and examine the intracellular localization in vitro by confocal microscopy. We show that the immunoliposome--siRNA complex maintains its nanoscale size and, using three separate tumor models, can efficiently and specifically deliver siRNA to both primary and metastatic disease after systemic delivery, thus increasing the possibility for translating the potent effects of siRNA observed in vitro into clinically useful therapeutics.
Collapse
Affiliation(s)
- Kathleen F Pirollo
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | | | | | | | | | | | | |
Collapse
|
18
|
Pirollo KF, Zon G, Rait A, Zhou Q, Yu W, Hogrefe R, Chang EH. Tumor-Targeting Nanoimmunoliposome Complex for Short Interfering RNA Delivery. Hum Gene Ther 2005. [DOI: 10.1089/hum.2005.17.ft-155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
19
|
Abstract
Tumour cell-targeted liposomal delivery has the potential to enhance the therapeutic efficacy and reduce the toxicity of anticancer agents. Folate receptor (FR) expression is frequently amplified among human malignancies. FR is, therefore, potentially useful as a tumour marker for targeted drug delivery. FR-mediated liposomal delivery has been shown to enhance the antitumour efficacy of doxorubicin both in vitro and in vivo, and to overcome P-glycoprotein-mediated multi-drug resistance. In addition, FR-targeted liposomes have shown utility as effective delivery vehicles of genes and antisense oligodeoxyribonucleotides to FR(+) tumour cells. Both solid tumours and leukaemias can potentially benefit from FR-targeted drug delivery. Multiple mechanisms might contribute to greater therapeutic efficacy for FR-targeted liposomes, such as FR-dependent cytotoxicity and antiangiogenic activity. Further investigation of this promising drug delivery strategy is clearly warranted.
Collapse
Affiliation(s)
- Xiaogang Pan
- The Ohio State University, Division of Pharmaceutics, College of Pharmacy and Comprehensive Cancer Center, 500 W. 12th Avenue, Columbus, OH 43210, USA
| | | |
Collapse
|
20
|
Zhang Z, Li M, Rayburn ER, Hill DL, Zhang R, Wang H. Oncogenes as Novel Targets for Cancer Therapy (Part I). ACTA ACUST UNITED AC 2005; 5:173-90. [PMID: 15952871 DOI: 10.2165/00129785-200505030-00004] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In the past 10 years, progress made in cancer biology, genetics, and biotechnology has led to a major transition in cancer drug design and development. There has been a change from an emphasis on non-specific, cytotoxic agents to specific, molecular-based therapeutics. Mechanism-based therapy is designed to act on cellular and molecular targets that are causally involved in the formation, growth, and progression of human cancers. These agents, which may have greater selectivity for cancer versus normal cells, and which may produce better anti-tumor efficacy and lower host toxicity, can be small molecules, natural or engineered peptides, proteins, antibodies, or synthetic nucleic acids (e.g. antisense oligonucleotides, ribozymes, and siRNAs). Novel targets are identified and validated by state-of-the-art approaches, including high-throughput screening, combinatorial chemistry, and gene expression arrays, which increase the speed and efficiency of drug discovery and development. Examples of oncogene-based, molecular therapeutics that show promising clinical activity include trastuzumab (Herceptin), imatinib (Gleevec), and gefitinib (Iressa). However, the full potential of oncogenes as novel targets for cancer therapy has not been realized and many challenges remain, from the validation of novel targets, to the design of specific agents, to the evaluation of these agents in both preclinical and clinical settings. In maximizing the benefits of molecular therapeutics in monotherapy or combination therapy of cancer, it is necessary to have an understanding of the underlying molecular abnormalities and mechanisms involved. This is the first part of a four-part review in which we discuss progress made in the last decade as it relates to the discovery of novel oncogenes and signal transduction pathways, in the context of their potential as targets for cancer therapy. This part delineates the latest discoveries about the potential use of growth factors and protein tyrosine kinases as targets for therapy. Later parts focus on intermediate signaling pathways, transcription factors, and proteins involved in cell cycle, DNA damage, and apoptotic pathways.
Collapse
Affiliation(s)
- Zhuo Zhang
- Department of Pharmacology and Toxicology, and Division of Clinical Pharmacology, University of Alabama at Birmingham, Birmingham, Alabama 35294-0019, USA
| | | | | | | | | | | |
Collapse
|