1
|
Um‐e‐Kalsoom, Wang S, Qu J, Liu L. Innovative optical imaging strategies for monitoring immunotherapy in the tumor microenvironments. Cancer Med 2024; 13:e70155. [PMID: 39387259 PMCID: PMC11465031 DOI: 10.1002/cam4.70155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 08/01/2024] [Accepted: 08/16/2024] [Indexed: 10/15/2024] Open
Abstract
BACKGROUND The tumor microenvironment (TME) plays a critical role in cancer progression and response to immunotherapy. Immunotherapy targeting the immune system has emerged as a promising treatment modality, but challenges in understanding the TME limit its efficacy. Optical imaging strategies offer noninvasive, real-time insights into the interactions between immune cells and the TME. OBJECTIVE This review assesses the progress of optical imaging technologies in monitoring immunotherapy within the TME and explores their potential applications in clinical trials and personalized cancer treatment. METHODS This is a comprehensive literature review based on the advances in optical imaging modalities including fluorescence imaging (FLI), bioluminescence imaging (BLI), and photoacoustic imaging (PAI). These modalities were analyzed for their capacity to provide high-resolution, real-time imaging of immune cell dynamics, tumor vasculature, and other critical components of the TME. RESULTS Optical imaging techniques have shown significant potential in tracking immune cell infiltration, assessing immune checkpoint inhibitors, and visualizing drug delivery within the TME. Technologies like FLI and BLI are pivotal in tracking immune responses in preclinical models, while PAI provides functional imaging with deeper tissue penetration. The integration of these modalities with immunotherapy holds promise for improving treatment monitoring and outcomes. CONCLUSION Optical imaging is a powerful tool for understanding the complexities of the TME and optimizing immunotherapy. Further advancements in imaging technologies, combined with nanomaterial-based approaches, could pave the way for enhanced diagnostic accuracy and therapeutic efficacy in cancer treatment.
Collapse
Affiliation(s)
- Um‐e‐Kalsoom
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic EngineeringShenzhen UniversityShenzhenChina
| | - Shiqi Wang
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic EngineeringShenzhen UniversityShenzhenChina
| | - Junle Qu
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic EngineeringShenzhen UniversityShenzhenChina
| | - Liwei Liu
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic EngineeringShenzhen UniversityShenzhenChina
| |
Collapse
|
2
|
Chenab KK, Malektaj H, Nadinlooie AAR, Mohammadi S, Zamani-Meymian MR. Intertumoral and intratumoral barriers as approaches for drug delivery and theranostics to solid tumors using stimuli-responsive materials. Mikrochim Acta 2024; 191:541. [PMID: 39150483 DOI: 10.1007/s00604-024-06583-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 07/15/2024] [Indexed: 08/17/2024]
Abstract
The solid tumors provide a series of biological barriers in cellular microenvironment for designing drug delivery methods based on advanced stimuli-responsive materials. These intertumoral and intratumoral barriers consist of perforated endotheliums, tumor cell crowding, vascularity, lymphatic drainage blocking effect, extracellular matrix (ECM) proteins, hypoxia, and acidosis. Triggering opportunities have been drawn for solid tumor therapies based on single and dual stimuli-responsive drug delivery systems (DDSs) that not only improved drug targeting in deeper sites of the tumor microenvironments, but also facilitated the antitumor drug release efficiency. Single and dual stimuli-responsive materials which are known for their lowest side effects can be categorized in 17 main groups which involve to internal and external stimuli anticancer drug carriers in proportion to microenvironments of targeted solid tumors. Development of such drug carriers can circumvent barriers in clinical trial studies based on their superior capabilities in penetrating into more inaccessible sites of the tumor tissues. In recent designs, key characteristics of these DDSs such as fast response to intracellular and extracellular factors, effective cytotoxicity with minimum side effect, efficient permeability, and rate and location of drug release have been discussed as core concerns of designing paradigms of these materials.
Collapse
Affiliation(s)
- Karim Khanmohammadi Chenab
- Department of Chemistry, Iran University of Science and Technology, Tehran, P.O. Box 16846-13114, Iran
- Department of Physics, Iran University of Science and Technology, Tehran, P.O. Box 16846-13114, Iran
| | - Haniyeh Malektaj
- Department of Materials and Production, Aalborg University, Fibigerstraede 16, 9220, Aalborg, Denmark
| | | | | | | |
Collapse
|
3
|
Arriaga JM, Ronaldson-Bouchard K, Picech F, Nunes de Almeida F, Afari S, Chhouri H, Vunjak-Novakovic G, Abate-Shen C. In vivo genome-wide CRISPR screening identifies CITED2 as a driver of prostate cancer bone metastasis. Oncogene 2024; 43:1303-1315. [PMID: 38454137 PMCID: PMC11101692 DOI: 10.1038/s41388-024-02995-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 02/21/2024] [Accepted: 02/26/2024] [Indexed: 03/09/2024]
Abstract
Most cancer deaths are due to metastatic dissemination to distant organs. Bone is the most frequently affected organ in metastatic prostate cancer and a major cause of prostate cancer deaths. Yet, our partial understanding of the molecular factors that drive bone metastasis has been a limiting factor for developing preventative and therapeutic strategies to improve patient survival and well-being. Although recent studies have uncovered molecular alterations that occur in prostate cancer metastasis, their functional relevance for bone metastasis is not well understood. Using genome-wide CRISPR activation and inhibition screens we have identified multiple drivers and suppressors of prostate cancer metastasis. Through functional validation, including an innovative organ-on-a-chip invasion platform for studying bone tropism, our study identifies the transcriptional modulator CITED2 as a novel driver of prostate cancer bone metastasis and uncovers multiple new potential molecular targets for bone metastatic disease.
Collapse
Affiliation(s)
- Juan M Arriaga
- Department of Molecular Pharmacology and Therapeutics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, 10032, USA.
- Department of Oncological Sciences, Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| | | | - Florencia Picech
- Department of Molecular Pharmacology and Therapeutics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Francisca Nunes de Almeida
- Department of Molecular Pharmacology and Therapeutics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Stephanie Afari
- Department of Molecular Pharmacology and Therapeutics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Houssein Chhouri
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering, Columbia University, New York, NY, 10032, USA
- Department of Medicine, Vagelos College of Physicians and Surgeons, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Cory Abate-Shen
- Departments of Molecular Pharmacology and Therapeutics, Urology, Medicine, Pathology & Cell Biology, and Systems Biology, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
4
|
Donelan W, Brisbane W, O'Malley P, Crispen P, Kusmartsev S. Hyaluronan Metabolism in Urologic Cancers. Adv Biol (Weinh) 2023; 7:e2300168. [PMID: 37615259 DOI: 10.1002/adbi.202300168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 07/06/2023] [Indexed: 08/25/2023]
Abstract
Hyaluronan (HA) is one of the major components of the extracellular matrix in tumor tissue. Recent reports have made it clear that the balance of HA synthesis and degradation is critical for tumor progression. HA is synthesized on the cytoplasmic surface of the plasma membrane by hyaluronan synthases (HAS) and extruded into the extracellular space. Excessive HA production in cancer is associated with enhanced HA degradation in the tumor microenvironment, leading to the accumulation of HA fragments with small molecular weight. These perturbations in both HA synthesis and degradation may play important roles in tumor progression. Recently, it has become increasingly clear that small HA fragments can induce a variety of biological events, such as angiogenesis, cancer-promoting inflammation, and tumor-associated immune suppression. Progression of urologic malignancies, particularly of prostate and bladder cancers, as well as of certain types of kidney cancer show markedly perturbed metabolism of tumor-associated HA. This review highlights the recent research findings regarding HA metabolism in tumor microenvironments with a special focus on urologic cancers. It also will discuss the potential implications of these findings for the development of novel therapeutic interventions for the treatment of prostate, bladder, and kidney cancers.
Collapse
Affiliation(s)
| | - Wayne Brisbane
- UCLA Medical Center, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | | | - Paul Crispen
- University of Florida, Gainesville, FL, 32611, USA
| | | |
Collapse
|
5
|
Sołtys A, Galanty A, Grabowska K, Paśko P, Zagrodzki P, Podolak I. Multidirectional Effects of Terpenoids from Sorbus intermedia (EHRH.) PERS Fruits in Cellular Model of Benign Prostate Hyperplasia. Pharmaceuticals (Basel) 2023; 16:965. [PMID: 37513877 PMCID: PMC10383022 DOI: 10.3390/ph16070965] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 07/03/2023] [Accepted: 07/04/2023] [Indexed: 07/30/2023] Open
Abstract
Benign prostatic hyperplasia (BPH) is a common urological disease affecting aging men. Its pathogenesis is regarded as complex and multifactorial, with sex hormones and inflammation as key contributory factors. In the current study, we investigated the anti-BPH potential of terpenoids present in the fruits of Sorbus intermedia (EHRH.) PERS. Not only the effects on testosterone-stimulated normal prostate epithelial PNT2 cells, namely suppression of 5-α-reductase activity, PSA secretion, and cell proliferation, were determined but also the inhibitory activity on heat-induced protein denaturation, hyaluronidase, as well as IL-6, TNF-α, and NO release in LPS-treated macrophages. Sorbus terpenoids significantly inhibited 5-α-reductase activity and reduced PSA secretion in PNT2 cells, reversing the stimulatory effect of testosterone. PNT2 cell proliferation was also found to be attenuated. Subsequently, all compounds reduced the release of pro-inflammatory mediators in RAW 264.7 cells. In addition, ursolic acid (UA) and its aldehyde (UAL) were the most potent hyaluronidase inhibitors of all compounds, with IC50 values of 225.75 µg/mL and 369.77 µg/mL, respectively. For better understanding and interpretation of the overall effect of Sorbus terpenoids on different aspects of BPH pathogenesis and development, cluster analysis was applied.
Collapse
Affiliation(s)
- Agnieszka Sołtys
- Department of Pharmacognosy, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Agnieszka Galanty
- Department of Pharmacognosy, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Karolina Grabowska
- Department of Pharmacognosy, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Paweł Paśko
- Department of Food Chemistry and Nutrition, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Paweł Zagrodzki
- Department of Food Chemistry and Nutrition, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Irma Podolak
- Department of Pharmacognosy, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| |
Collapse
|
6
|
Ikeda-Imafuku M, Gao Y, Shaha S, Wang LLW, Park KS, Nakajima M, Adebowale O, Mitragotri S. Extracellular matrix degrading enzyme with stroma-targeting peptides enhance the penetration of liposomes into tumors. J Control Release 2022; 352:1093-1103. [PMID: 36351520 DOI: 10.1016/j.jconrel.2022.11.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/29/2022] [Accepted: 11/03/2022] [Indexed: 11/21/2022]
Abstract
Various anti-tumor nanomedicines have been developed based on the enhanced permeability and retention effect. However, the dense extracellular matrix (ECM) in tumors remains a major barrier for the delivery and accumulation of nanoparticles into tumors. While ECM-degrading enzymes, such as collagenase, hyaluronidase, and bromelain, have been used to facilitate the accumulation of nanoparticles, serious side effects arising from the current non-tumor-specific delivery methods limit their clinical applications. Here, we report targeted delivery of bromelain into tumor tissues through its covalent attachment to a hyaluronic acid (HA)-peptide conjugate with tumor ECM targeting ability. The ECM targeting peptide, collagen type IV-binding peptide (C4BP), was chosen from six candidate-peptides based on their ability to bind to frozen sections of triple-negative breast cancer, 4T1 tumor ex vivo. The HA- C4BP conjugate showed a significant increase in tumor accumulation in 4T1-bearing mice after intravenous administration compared to unmodified HA. We further demonstrated that the systemic administration of bromelain conjugated C4BP-HA (C4BP-HA-Bro) potentiates the anti-tumor efficacy of liposomal doxorubicin. C4BP-HA-Bro decreased the number and length of collagen fibers and improved the distribution of doxorubicin within the tumor. No infusion reaction was noted after delivery of C4BP-HA-Bro. C4BP-HA thus offers a potential for effective and safe delivery of bromelain for improved intratumoral delivery of therapeutics.
Collapse
Affiliation(s)
- Mayumi Ikeda-Imafuku
- John A. Paulson School of Engineering & Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Cambridge, MA 20138, USA
| | - Yongsheng Gao
- John A. Paulson School of Engineering & Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Cambridge, MA 20138, USA
| | - Suyog Shaha
- John A. Paulson School of Engineering & Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Cambridge, MA 20138, USA
| | - Lily Li-Wen Wang
- John A. Paulson School of Engineering & Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Cambridge, MA 20138, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Kyung Soo Park
- John A. Paulson School of Engineering & Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Cambridge, MA 20138, USA
| | - Mayuka Nakajima
- John A. Paulson School of Engineering & Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Cambridge, MA 20138, USA
| | - Omokolade Adebowale
- John A. Paulson School of Engineering & Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Cambridge, MA 20138, USA
| | - Samir Mitragotri
- John A. Paulson School of Engineering & Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Cambridge, MA 20138, USA.
| |
Collapse
|
7
|
Huttala O, Loreth D, Staff S, Tanner M, Wikman H, Ylikomi T. Decellularized In Vitro Capillaries for Studies of Metastatic Tendency and Selection of Treatment. Biomedicines 2022; 10:biomedicines10020271. [PMID: 35203480 PMCID: PMC8869401 DOI: 10.3390/biomedicines10020271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/21/2022] [Accepted: 01/22/2022] [Indexed: 11/19/2022] Open
Abstract
Vascularization plays an important role in the microenvironment of the tumor. Therefore, it should be a key element to be considered in the development of in vitro cancer assays. In this study, we decellularized in vitro capillaries to remove genetic material and optimized the medium used to increase the robustness and versatility of applications. The growth pattern and drug responses of cancer cell lines and patient-derived primary cells were studied on decellularized capillaries. Interestingly, two distinct growth patterns were seen when cancer cells were grown on decellularized capillaries: “network” and “cluster”. Network formation correlated with the metastatic properties of the cells and cluster formation was observed in non-metastatic cells. Drug responses of patient-derived cells correlated better with clinical findings when cells were cultured on decellularized capillaries compared with those cultured on plastic. Decellularized capillaries provide a novel method for cancer cell culture applications. It bridges the gap between complex 3D culture methods and traditional 2D culture methods by providing the ease and robustness of 2D culture as well as an in vivo-like microenvironment and scaffolding for 3D cultures.
Collapse
Affiliation(s)
- Outi Huttala
- Cell Biology, Faculty of Medicine and Health Technology, Tampere University, 33100 Tampere, Finland;
- Tays Cancer Center, Tampere University Hospital, 33520 Tampere, Finland; (S.S.); (M.T.)
- Correspondence: ; Tel.: +358-401909721
| | - Desiree Loreth
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (D.L.); (H.W.)
| | - Synnöve Staff
- Tays Cancer Center, Tampere University Hospital, 33520 Tampere, Finland; (S.S.); (M.T.)
- Department of Obstetrics and Gynecology, Tampere University Hospital, 33520 Tampere, Finland
| | - Minna Tanner
- Tays Cancer Center, Tampere University Hospital, 33520 Tampere, Finland; (S.S.); (M.T.)
- Department of Oncology, Tampere University Hospital, 33520 Tampere, Finland
- Department of Oncology, Faculty of Medicine and Health Technology, Tampere University, 33100 Tampere, Finland
| | - Harriet Wikman
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (D.L.); (H.W.)
| | - Timo Ylikomi
- Cell Biology, Faculty of Medicine and Health Technology, Tampere University, 33100 Tampere, Finland;
- Tays Cancer Center, Tampere University Hospital, 33520 Tampere, Finland; (S.S.); (M.T.)
| |
Collapse
|
8
|
Abstract
The extracellular matrix is part of the microenvironment and its functions are associated with the physical and chemical properties of the tissue. Among the extracellular components, the glycosaminoglycan hyaluronan is a key component, defining both the physical and biochemical characteristics of the healthy matrices. The hyaluronan metabolism is strictly regulated in physiological conditions, but in the tumoral tissues, its expression, size and binding proteins interaction are dysregulated. Hyaluronan from the tumor microenvironment promotes tumor cell proliferation, invasion, immune evasion, stemness alterations as well as drug resistance. This chapter describes data regarding novel concepts of hyaluronan functions in the tumor. Additionally, we discuss potential clinical applications of targeting HA metabolism in cancer therapy.
Collapse
|
9
|
Zargar A, Chang S, Kothari A, Snijders AM, Mao JH, Wang J, Hernández AC, Keasling JD, Bivona TG. Overcoming the challenges of cancer drug resistance through bacterial-mediated therapy. Chronic Dis Transl Med 2019; 5:258-266. [PMID: 32055785 PMCID: PMC7004931 DOI: 10.1016/j.cdtm.2019.11.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Indexed: 12/23/2022] Open
Abstract
Despite tremendous efforts to fight cancer, it remains a major public health problem and a leading cause of death worldwide. With increased knowledge of cancer pathways and improved technological platforms, precision therapeutics that specifically target aberrant cancer pathways have improved patient outcomes. Nevertheless, a primary cause of unsuccessful cancer therapy remains cancer drug resistance. In this review, we summarize the broad classes of resistance to cancer therapy, particularly pharmacokinetics, the tumor microenvironment, and drug resistance mechanisms. Furthermore, we describe how bacterial-mediated cancer therapy, a bygone mode of treatment, has been revitalized by synthetic biology and is uniquely suited to address the primary resistance mechanisms that confound traditional therapies. Through genetic engineering, we discuss how bacteria can be potent anticancer agents given their tumor targeting potential, anti-tumor activity, safety, and coordinated delivery of anti-cancer drugs.
Collapse
Affiliation(s)
- Amin Zargar
- Joint BioEnergy Institute, Lawrence Berkeley National Laboratory, Emeryville, CA 94608, USA
- QB3 Institute, University of California-Berkeley, 174 Stanley Hall, Berkeley, CA 94720, USA
| | - Samantha Chang
- Joint BioEnergy Institute, Lawrence Berkeley National Laboratory, Emeryville, CA 94608, USA
| | - Ankita Kothari
- Biological Systems and Engineering, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Antoine M. Snijders
- Biological Systems and Engineering, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Jian-Hua Mao
- Biological Systems and Engineering, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Jessica Wang
- Joint BioEnergy Institute, Lawrence Berkeley National Laboratory, Emeryville, CA 94608, USA
| | - Amanda C. Hernández
- Joint BioEnergy Institute, Lawrence Berkeley National Laboratory, Emeryville, CA 94608, USA
- Department of Chemical & Biomolecular Engineering, Department of Bioengineering, University of California, Berkeley, CA 94720, USA
| | - Jay D. Keasling
- Joint BioEnergy Institute, Lawrence Berkeley National Laboratory, Emeryville, CA 94608, USA
- QB3 Institute, University of California-Berkeley, 174 Stanley Hall, Berkeley, CA 94720, USA
| | - Trever G. Bivona
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94143, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
10
|
Varallo GR, Jardim-Perassi BV, Alexandre PA, Fukumasu H, Zuccari DAPC. Global gene expression profile in canine mammary carcinomas. Vet J 2019; 254:105393. [PMID: 31836163 DOI: 10.1016/j.tvjl.2019.105393] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 09/30/2019] [Accepted: 10/01/2019] [Indexed: 11/26/2022]
Abstract
Mammary gland tumors are a heterogeneous group of neoplastic diseases. Genetic studies make it possible to determine genetic profiles and identify new molecular markers. The aim of the study was to evaluate the gene expression profile of canine mammary carcinomas and identify potential prognostic markers. Twelve mammary cancer samples from bitches were collected for the evaluation of global gene expression. Microarray assays were performed using commercial kits. Statistical analysis of the microarray was done using moderate t-statistic and adjusted using the Benjamini and Hochberg procedure. Differential connectivity analysis was also performed. Enrichment analyses were conducted using WebGestalt. P-values were calculated using hypergeometric statistics and adjusted using the Benjamini and Hochberg procedure. The HYAL-1 gene was validated using quantitative PCR (qPCR). There were 878 upregulated genes and 821 downregulated genes in the neoplasms studied. Enrichment analysis (individual analysis) identified the HYAL-1 gene as a potential marker of tumorigenesis and tumor recurrence. Differential connectivity analysis demonstrated 262 differentially connected genes.
Collapse
Affiliation(s)
- G R Varallo
- Postgraduate Program in Veterinary Surgery, UNESP-FCAV, Jaboticabal 14884-900, Brazil
| | - B V Jardim-Perassi
- Department of Molecular Biology, FAMERP, São José do Rio Preto 15090000, Brazil
| | - P A Alexandre
- Department of Veterinary Medicine, FZEA-USP, Pirassununga 13635900, Brazil
| | - H Fukumasu
- Department of Veterinary Medicine, FZEA-USP, Pirassununga 13635900, Brazil
| | - D A P C Zuccari
- Department of Molecular Biology, FAMERP, São José do Rio Preto 15090000, Brazil.
| |
Collapse
|
11
|
Abstract
In the last few decades, hyaluronic acid (HA) has become increasingly employed as a biomaterial in both clinical and research applications. The abundance of HA in many tissues, together with its amenability to chemical modification, has made HA an attractive material platform for a wide range of applications including regenerative medicine, drug delivery, and scaffolds for cell culture. HA has traditionally been appreciated to modulate tissue mechanics and remodeling through its distinctive biophysical properties and ability to organize other matrix proteins. However, HA can influence cell behavior in much more direct and specific ways by engaging cellular HA receptors, which can trigger signals that influence cell survival, proliferation, adhesion, and migration. In turn, cells modify HA by regulating synthesis and degradation through a dedicated arsenal of enzymes. Optimal design of HA-based biomaterials demands full consideration of these diverse modes of regulation. This review summarizes how HA-based signaling regulates cell behavior and discusses how these signals can be leveraged to create cell-instructive biomaterials.
Collapse
Affiliation(s)
- Kayla J. Wolf
- University of California, Berkeley – University of California, San Francisco Graduate Program in Bioengineering, Berkeley, California, 94720, USA
- Department of Bioengineering, University of California, Berkeley, Berkeley, California, 94720, USA
| | - Sanjay Kumar
- University of California, Berkeley – University of California, San Francisco Graduate Program in Bioengineering, Berkeley, California, 94720, USA
- Department of Bioengineering, University of California, Berkeley, Berkeley, California, 94720, USA
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, California, 94720, USA
| |
Collapse
|
12
|
Zhang Q, Jin H, Chen L, Chen Q, He Y, Yang Y, Ma S, Xiao S, Xi F, Luo Q, Liu J. Effect of Ultrasound Combined With Microbubble Therapy on Interstitial Fluid Pressure and VX2 Tumor Structure in Rabbit. Front Pharmacol 2019; 10:716. [PMID: 31293427 PMCID: PMC6606793 DOI: 10.3389/fphar.2019.00716] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 06/05/2019] [Indexed: 11/13/2022] Open
Abstract
Interstitial fluid pressure (IFP) in tumor tissue is significantly higher than that in normal tissue, which reduces the effectiveness of therapeutic drugs. There are several methods to decrease the IFP, such as normalizing blood vessel, decreasing hyaluronic acid and collagen fiber content in the extracellular matrix (ECM), and recovering lymphatic function. Reducing tumor IFP might be developed as a novel approach in cancer therapy. In this study, we aimed to elucidate the relationship between ultrasound combined with microbubble therapy and IFP, and the associated mechanism. VX2 tumor in rabbit was treated with ultrasound combined with microbubbles at different intensities. The IFP was measured using the wick-in-needle (WIN) method. The collagen and reticular fibers were stained by Masson and Gordon-Sweets, respectively. The results showed that low-frequency non-focus ultrasound combined with microbubbles therapy influences the IFP in tumor tissues; low-frequency non-focus ultrasound with low pressure increased the IFP, whereas middle-high pressure decreased the IFP. The results showed that the structure and content of collagen and reticular fibers in tumor tissue were rarely influenced by the treatment. Our study provides a novel approach of reduced IFP antitumor therapy.
Collapse
Affiliation(s)
- Qianyun Zhang
- Department of Medical Ultrasound, Guangzhou First People's Hospital, Guangzhou, China.,School of Medicine, South China University of Technology, Guangzhou, China.,Department of Medical Ultrasound, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Hai Jin
- Department of Medical Ultrasound, Guangzhou First People's Hospital, Guangzhou, China.,School of Medicine, South China University of Technology, Guangzhou, China
| | - Liping Chen
- Department of Medical Ultrasound, Guangzhou First People's Hospital, Guangzhou, China.,School of Medicine, South China University of Technology, Guangzhou, China
| | - Qiaoli Chen
- Department of Medical Ultrasound, Guangzhou First People's Hospital, Guangzhou, China.,School of Medicine, South China University of Technology, Guangzhou, China
| | - Yan He
- Department of Medical Ultrasound, Guangzhou First People's Hospital, Guangzhou, China.,School of Medicine, South China University of Technology, Guangzhou, China
| | - Yuwen Yang
- Department of Medical Ultrasound, Guangzhou First People's Hospital, Guangzhou, China.,School of Medicine, South China University of Technology, Guangzhou, China
| | - Suihong Ma
- Department of Medical Ultrasound, Guangzhou First People's Hospital, Guangzhou, China.,School of Medicine, South China University of Technology, Guangzhou, China
| | - Shuyi Xiao
- Department of Medical Ultrasound, Guangzhou First People's Hospital, Guangzhou, China.,School of Medicine, South China University of Technology, Guangzhou, China
| | - Fen Xi
- Department of Medical Ultrasound, Guangzhou First People's Hospital, Guangzhou, China.,School of Medicine, South China University of Technology, Guangzhou, China
| | - Qiong Luo
- Department of Medical Ultrasound, Guangzhou First People's Hospital, Guangzhou, China.,School of Medicine, South China University of Technology, Guangzhou, China
| | - Jianhua Liu
- Department of Medical Ultrasound, Guangzhou First People's Hospital, Guangzhou, China.,School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
13
|
El-Sawy HS, Al-Abd AM, Ahmed TA, El-Say KM, Torchilin VP. Stimuli-Responsive Nano-Architecture Drug-Delivery Systems to Solid Tumor Micromilieu: Past, Present, and Future Perspectives. ACS NANO 2018; 12:10636-10664. [PMID: 30335963 DOI: 10.1021/acsnano.8b06104] [Citation(s) in RCA: 277] [Impact Index Per Article: 39.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
The microenvironment characteristics of solid tumors, renowned as barriers that harshly impeded many drug-delivery approaches, were precisely studied, investigated, categorized, divided, and subdivided into a complex diverse of barriers. These categories were further studied with a particular perspective, which makes all barriers found in solid-tumor micromilieu turn into different types of stimuli, and were considered triggers that can increase and hasten drug-release targeting efficacy. This review gathers data concerning the nature of solid-tumor micromilieu. Past research focused on the treatment of such tumors, the recent efforts employed for engineering smart nanoarchitectures with the utilization of the specified stimuli categories, the possibility of combining more than one stimuli for much-greater targeting enhancement, examples of the approved nanoarchitectures that already translated clinically as well as the obstacles faced by the use of these nanostructures, and, finally, an overview of the possible future implementations of smart-chemical engineering for the design of more-efficient drug delivery and theranostic systems and for making nanosystems with a much-higher level of specificity and penetrability features.
Collapse
Affiliation(s)
- Hossam S El-Sawy
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy , Egyptian Russian University , Badr City , Cairo 63514 , Egypt
| | - Ahmed M Al-Abd
- Department of Pharmaceutical Sciences, College of Pharmacy , Gulf Medical University , Ajman , United Arab Emirates
- Pharmacology Department, Medical Division , National Research Centre , Giza 12622 , Egypt
| | - Tarek A Ahmed
- Department of Pharmaceutics, Faculty of Pharmacy , King Abdulaziz University , Jeddah 21589 , Saudi Arabia
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy , Al-Azhar University , Cairo 11651 , Egypt
| | - Khalid M El-Say
- Department of Pharmaceutics, Faculty of Pharmacy , King Abdulaziz University , Jeddah 21589 , Saudi Arabia
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy , Al-Azhar University , Cairo 11651 , Egypt
| | - Vladimir P Torchilin
- Department of Pharmaceutical Sciences Center for Pharmaceutical Biotechnology and Nanomedicine , Northeastern University , 140 The Fenway, Room 211/214, 360 Huntington Aveue , Boston , Massachusetts 02115 , United States
| |
Collapse
|
14
|
McAtee CO, Booth C, Elowsky C, Zhao L, Payne J, Fangman T, Caplan S, Henry MD, Simpson MA. Prostate tumor cell exosomes containing hyaluronidase Hyal1 stimulate prostate stromal cell motility by engagement of FAK-mediated integrin signaling. Matrix Biol 2018; 78-79:165-179. [PMID: 29753676 DOI: 10.1016/j.matbio.2018.05.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 03/28/2018] [Accepted: 05/08/2018] [Indexed: 01/22/2023]
Abstract
The hyaluronidase Hyal1 is clinically and functionally implicated in prostate cancer progression and metastasis. Elevated Hyal1 accelerates vesicular trafficking in prostate tumor cells, thereby enhancing their metastatic potential in an autocrine manner through increased motility and proliferation. In this report, we found Hyal1 protein is a component of exosomes produced by prostate tumor cell lines overexpressing Hyal1. We investigated the role of exosomally shed Hyal1 in modulating tumor cell autonomous functions and in modifying the behavior of prostate stromal cells. Catalytic activity of Hyal1 was necessary for enrichment of Hyal1 in the exosome fraction, which was associated with increased presence of LC3BII, an autophagic marker, in the exosomes. Hyal1-positive exosome contents were internalized from the culture medium by WPMY-1 prostate stromal fibroblasts. Treatment of prostate stromal cells with tumor exosomes did not affect proliferation, but robustly stimulated their migration in a manner dependent on Hyal1 catalytic activity. Increased motility of exosome-treated stromal cells was accompanied by enhanced adhesion to a type IV collagen matrix, as well as increased FAK phosphorylation and integrin engagement through dynamic membrane residence of β1 integrins. The presence of Hyal1 in tumor-derived exosomes and its ability to impact the behavior of stromal cells suggests cell-cell communication via exosomes is a novel mechanism by which elevated Hyal1 promotes prostate cancer progression.
Collapse
Affiliation(s)
- Caitlin O McAtee
- Department of Biochemistry, University of Nebraska, Lincoln, NE, United States
| | - Christine Booth
- Department of Biochemistry, University of Nebraska, Lincoln, NE, United States
| | - Christian Elowsky
- Morrison Microscopy Facility, University of Nebraska, Lincoln, NE, United States
| | - Lei Zhao
- Department of Molecular Physiology and Biophysics, Holden Comprehensive Cancer Center, University of Iowa Carver College of Medicine; Iowa City, IA, United States
| | - Jeremy Payne
- Department of Biochemistry, University of Nebraska, Lincoln, NE, United States
| | - Teresa Fangman
- Morrison Microscopy Facility, University of Nebraska, Lincoln, NE, United States
| | - Steve Caplan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States; Fred and Pamela Buffett Cancer Center, Omaha, NE, United States
| | - Michael D Henry
- Department of Molecular Physiology and Biophysics, Holden Comprehensive Cancer Center, University of Iowa Carver College of Medicine; Iowa City, IA, United States
| | - Melanie A Simpson
- Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, NC, United States.
| |
Collapse
|
15
|
Khalid A, Persano S, Shen H, Zhao Y, Blanco E, Ferrari M, Wolfram J. Strategies for improving drug delivery: nanocarriers and microenvironmental priming. Expert Opin Drug Deliv 2017; 14:865-877. [PMID: 27690153 PMCID: PMC5584706 DOI: 10.1080/17425247.2017.1243527] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION The ultimate goal in the field of drug delivery is to exclusively direct therapeutic agents to pathological tissues in order to increase therapeutic efficacy and eliminate side effects. This goal is challenging due to multiple transport obstacles in the body. Strategies that improve drug transport exploit differences in the characteristics of normal and pathological tissues. Within the field of oncology, these concepts have laid the groundwork for a new discipline termed transport oncophysics. Areas covered: Efforts to improve drug biodistribution have mainly focused on nanocarriers that enable preferential accumulation of drugs in diseased tissues. A less common approach to enhance drug transport involves priming strategies that modulate the biological environment in ways that favor localized drug delivery. This review discusses a variety of priming and nanoparticle design strategies that have been used for drug delivery. Expert opinion: Combinations of priming agents and nanocarriers are likely to yield optimal drug distribution profiles. Although priming strategies have yet to be widely implemented, they represent promising solutions for overcoming biological transport barriers. In fact, such strategies are not restricted to priming the tumor microenvironment but can also be directed toward healthy tissue in order to reduce nanoparticle uptake.
Collapse
Affiliation(s)
- Ayesha Khalid
- Medical Program, Weill Cornell Medicine-Qatar, Qatar Foundation, Doha, Qatar
| | - Stefano Persano
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Haifa Shen
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Yuliang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience & Technology of China, University of Chinese Academy of Sciences, Beijing 100190, China
- Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
| | - Elvin Blanco
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Mauro Ferrari
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Medicine, Weill Cornell Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Joy Wolfram
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience & Technology of China, University of Chinese Academy of Sciences, Beijing 100190, China
| |
Collapse
|
16
|
Simões RV, Veeraperumal S, Serganova IS, Kruchevsky N, Varshavsky J, Blasberg RG, Ackerstaff E, Koutcher JA. Inhibition of prostate cancer proliferation by Deferiprone. NMR IN BIOMEDICINE 2017; 30:10.1002/nbm.3712. [PMID: 28272795 PMCID: PMC5505495 DOI: 10.1002/nbm.3712] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 01/20/2017] [Accepted: 01/20/2017] [Indexed: 05/22/2023]
Abstract
Cancer growth and proliferation rely on intracellular iron availability. We studied the effects of Deferiprone (DFP), a chelator of intracellular iron, on three prostate cancer cell lines: murine, metastatic TRAMP-C2; murine, non-metastatic Myc-CaP; and human, non-metastatic 22rv1. The effects of DFP were evaluated at different cellular levels: cell culture proliferation and migration; metabolism of live cells (time-course multi-nuclear magnetic resonance spectroscopy cell perfusion studies, with 1-13 C-glucose, and extracellular flux analysis); and expression (Western blot) and activity of mitochondrial aconitase, an iron-dependent enzyme. The 50% and 90% inhibitory concentrations (IC50 and IC90 , respectively) of DFP for the three cell lines after 48 h of incubation were within the ranges 51-67 μM and 81-186 μM, respectively. Exposure to 100 μM DFP led to: (i) significant inhibition of cell migration after different exposure times, ranging from 12 h (TRAMP-C2) to 48 h (22rv1), in agreement with the respective cell doubling times; (ii) significantly decreased glucose consumption and glucose-driven tricarboxylic acid cycle activity in metastatic TRAMP-C2 cells, during the first 10 h of exposure, and impaired cellular bioenergetics and membrane phospholipid turnover after 23 h of exposure, consistent with a cytostatic effect of DFP. At this time point, all cell lines studied showed: (iii) significant decreases in mitochondrial functional parameters associated with the oxygen consumption rate, and (iv) significantly lower mitochondrial aconitase expression and activity. Our results indicate the potential of DFP to inhibit prostate cancer proliferation at clinically relevant doses and plasma concentrations.
Collapse
Affiliation(s)
- Rui V. Simões
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center
| | | | | | | | - Joseph Varshavsky
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center
| | - Ronald G. Blasberg
- Department of Neurology, Memorial Sloan Kettering Cancer Center
- Department of Medicine, Memorial Sloan Kettering Cancer Center
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center
| | - Ellen Ackerstaff
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center
| | - Jason A. Koutcher
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center
- Department of Radiology, Memorial Sloan Kettering Cancer Center
- Department of Medicine, Memorial Sloan Kettering Cancer Center
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center
- Weill Cornell Medical College, Cornell University. New York, NY 10065, USA
| |
Collapse
|
17
|
Abstract
OBJECTIVES Increased production and processing (degradation) of hyaluronan (HA) is critical for cancer invasion and metastasis. Although HA is known to be overexpressed in pancreatic ductal adenocarcinoma (PDAC), little is known about the expression and biological significance of HA-degrading enzymes, hyaluronidases (HYALs), in PDAC. METHODS Expression of HYALs mRNA was examined in PDAC cells by quantitative real-time RT-PCR. HYAL1 protein expression was examined in primary PDAC tumors by enzyme-linked immuno-sorbent assay. The migratory ability of PDAC cells was determined by a transwell cell migration assay. RESULTS Screening of mRNA expression of three major HYAL genes (HYAL1, 2, and 3) identified HYAL1 as a gene overexpressed in PDAC cells. Treatment of PDAC cells with 5-aza-2'-deoxycytidine and/or trichostatin A further increased the HYAL1 expression, suggesting a possible involvement of epigenetic mechanisms in the transcriptional regulation of this gene. HYAL1 protein concentrations were significantly higher in primary PDAC tissues as compared with nontumor pancreatic tissues (P = 0.049). Importantly, inhibition of HYAL activity by dextran sulfate significantly inhibited the migration of PDAC cells showing strong HYAL1 expression (P = 0.002). CONCLUSIONS These findings suggest that overexpression of HYAL1 is a common mechanism that may contribute to the aggressive phenotype of PDAC.
Collapse
|
18
|
Martelli C, Dico AL, Diceglie C, Lucignani G, Ottobrini L. Optical imaging probes in oncology. Oncotarget 2016; 7:48753-48787. [PMID: 27145373 PMCID: PMC5217050 DOI: 10.18632/oncotarget.9066] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 04/10/2016] [Indexed: 01/19/2023] Open
Abstract
Cancer is a complex disease, characterized by alteration of different physiological molecular processes and cellular features. Keeping this in mind, the possibility of early identification and detection of specific tumor biomarkers by non-invasive approaches could improve early diagnosis and patient management.Different molecular imaging procedures provide powerful tools for detection and non-invasive characterization of oncological lesions. Clinical studies are mainly based on the use of computed tomography, nuclear-based imaging techniques and magnetic resonance imaging. Preclinical imaging in small animal models entails the use of dedicated instruments, and beyond the already cited imaging techniques, it includes also optical imaging studies. Optical imaging strategies are based on the use of luminescent or fluorescent reporter genes or injectable fluorescent or luminescent probes that provide the possibility to study tumor features even by means of fluorescence and luminescence imaging. Currently, most of these probes are used only in animal models, but the possibility of applying some of them also in the clinics is under evaluation.The importance of tumor imaging, the ease of use of optical imaging instruments, the commercial availability of a wide range of probes as well as the continuous description of newly developed probes, demonstrate the significance of these applications. The aim of this review is providing a complete description of the possible optical imaging procedures available for the non-invasive assessment of tumor features in oncological murine models. In particular, the characteristics of both commercially available and newly developed probes will be outlined and discussed.
Collapse
Affiliation(s)
- Cristina Martelli
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
- Centre of Molecular and Cellular Imaging-IMAGO, Milan, Italy
| | - Alessia Lo Dico
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
- Umberto Veronesi Foundation, Milan, Italy
| | - Cecilia Diceglie
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
- Centre of Molecular and Cellular Imaging-IMAGO, Milan, Italy
- Tecnomed Foundation, University of Milan-Bicocca, Monza, Italy
| | - Giovanni Lucignani
- Centre of Molecular and Cellular Imaging-IMAGO, Milan, Italy
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Luisa Ottobrini
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
- Centre of Molecular and Cellular Imaging-IMAGO, Milan, Italy
- Institute for Molecular Bioimaging and Physiology (IBFM), National Research Council (CNR), Milan, Italy
| |
Collapse
|
19
|
The cancer-promoting gene fatty acid-binding protein 5 (FABP5) is epigenetically regulated during human prostate carcinogenesis. Biochem J 2016; 473:449-61. [DOI: 10.1042/bj20150926] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 11/27/2015] [Indexed: 12/12/2022]
Abstract
The DNA methylation status of CpG islands in the FABP5 promoter is critical for its expression. Epigenetic regulation of FABP5 gene expression plays an important role during human prostate carcinogenesis, along with up-regulation of c-Myc and Sp1.
Collapse
|
20
|
Tanaka N, Lajud SA, Ramsey A, Szymanowski AR, Ruffner R, O'Malley BW, Li D. Application of infrared-based molecular imaging to a mouse model with head and neck cancer. Head Neck 2015; 38 Suppl 1:E1351-7. [PMID: 26348614 DOI: 10.1002/hed.24226] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 07/08/2015] [Accepted: 07/20/2015] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND This study investigated whether near infrared (NIR) or visible fluorescent molecular imaging produced a better representation of a mouse model with head and neck squamous cell carcinoma (HNSCC). Additionally, the study explored whether epidermal growth factor receptor (EGFR)-targeted probes could play an important role in the diagnosis of HNSCC. METHODS An orthotopic mouse model of HNSCC labeled with the NIR fluorophore, infrared fluorescent protein (iRFP), was developed and monitored noninvasively in real time. The tumors were further evaluated using tumor-specific EGFR-targeted probes conjugated with an NIR dye (IRDye800), or a visible fluorescent protein. RESULTS The iRFP cell line produced better results than cells emitting visible light when studying local, distant, and deep tumors in the mouse model. The EGFR-targeted probe conjugated with IRDye800 accurately detected tumor perimeters. CONCLUSION This model has great potential as a unique tool in the study of HNSCC tumor development. © 2015 Wiley Periodicals, Inc. Head Neck 38: E1351-E1357, 2016.
Collapse
Affiliation(s)
- Nobuaki Tanaka
- Department of Otolaryngology - Head and Neck Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania.,Department of Otolaryngology - Head and Neck Surgery, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Shayanne A Lajud
- Department of Otolaryngology - Head and Neck Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Andrew Ramsey
- Department of Otolaryngology - Head and Neck Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Adam R Szymanowski
- Department of Otolaryngology - Head and Neck Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Randall Ruffner
- Department of Otolaryngology - Head and Neck Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Bert W O'Malley
- Department of Otolaryngology - Head and Neck Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Daqing Li
- Department of Otolaryngology - Head and Neck Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
21
|
Al-Abd AM, Aljehani ZK, Gazzaz RW, Fakhri SH, Jabbad AH, Alahdal AM, Torchilin VP. Pharmacokinetic strategies to improve drug penetration and entrapment within solid tumors. J Control Release 2015; 219:269-277. [PMID: 26342660 DOI: 10.1016/j.jconrel.2015.08.055] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 08/09/2015] [Accepted: 08/28/2015] [Indexed: 02/08/2023]
Abstract
Despite the discovery of a large number of anticancer agents, cancer still remains among the leading causes of death since the middle of the twentieth century. Solid tumors possess a high degree of genetic instability and emergence of treatment resistance. Tumor resistance has emerged for almost all approved anticancer drugs and will most probably emerge for newly discovered anticancer agents as well. The use of pharmacokinetic approaches to increase anticancer drug concentrations within the solid tumor compartment and prolong its entrapment might diminish the possibility of resistance emergence at the molecular pharmacodynamic level and might even reverse tumor resistance. Several novel treatment modalities such as metronomic therapy, angiogenesis inhibitors, vascular disrupting agents and tumor priming have been introduced to improve solid tumor treatment outcomes. In the current review we will discuss the pharmacokinetic aspect of these treatment modalities in addition to other older treatment modalities, such as extracellular matrix dissolving agents, extracellular matrix synthesis inhibitors, chemoembolization and cellular efflux pump inhibition. Many of these strategies showed variable degrees of success/failure; however, reallocating these modalities based on their influence on the intratumoral pharmacokinetics might improve their understanding and treatment outcomes.
Collapse
Affiliation(s)
- Ahmed M Al-Abd
- Department of Pharmacology, Medical Division, National Research Centre, Dokki, Giza, Egypt; Center for Pharmaceutical Biotechnology and Nanomedicine (CPBN), Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA; Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Zekra K Aljehani
- Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Rana W Gazzaz
- Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sarah H Fakhri
- Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Aisha H Jabbad
- Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | | | - Vladimir P Torchilin
- Center for Pharmaceutical Biotechnology and Nanomedicine (CPBN), Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA; Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| |
Collapse
|
22
|
Schmaus A, Bauer J, Sleeman JP. Sugars in the microenvironment: the sticky problem of HA turnover in tumors. Cancer Metastasis Rev 2015; 33:1059-79. [PMID: 25324146 DOI: 10.1007/s10555-014-9532-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The properties and behavior of tumor cells are closely regulated by their microenvironment. Accordingly, stromal cells and extracellular matrix components can have a pronounced effect on cancer initiation, growth, and progression. The linear glycosaminoglycan hyaluronan (HA) is a major component of the extracellular matrix. Altered synthesis and degradation of HA in the tumor context has been implicated in many aspects of tumor biology. In particular, the accumulation of small HA oligosaccharides (sHA) in the tumor interstitial space may play a decisive role, due to the ability of sHA to activate a number of biological processes that are not modulated by high molecular weight (HMW)-HA. In this article, we review the normal physiological role and metabolism of HA and then survey the evidence implicating HA in tumor growth and progression, focusing in particular on the potential contribution of sHA to these processes.
Collapse
Affiliation(s)
- Anja Schmaus
- Institut für Toxikologie und Genetik, Karlsruhe Institute for Technology (KIT), Campus Nord, Postfach 3640, 76021, Karlsruhe, Germany
| | | | | |
Collapse
|
23
|
Abstract
Hyaluronidases are a family of five human enzymes that have been differentially implicated in the progression of many solid tumor types, both clinically and in functional studies. Advances in the past 5 years have clarified many apparent contradictions: (1) by demonstrating that specific hyaluronidases have alternative substrates to hyaluronan (HA) or do not exhibit any enzymatic activity, (2) that high-molecular weight HA polymers elicit signaling effects that are opposite those of the hyaluronidase-digested HA oligomers, and (3) that it is actually the combined overexpression of HA synthesizing enzymes with hyaluronidases that confers tumorigenic potential. This review examines the literature supporting these conclusions and discusses novel mechanisms by which hyaluronidases impact invasive tumor cell processes. In addition, a detailed structural and functional comparison of the hyaluronidases is presented with insights into substrate selectivity and potential for therapeutic targeting. Finally, technological advances in targeting hyaluronidase for tumor imaging and cancer therapy are summarized.
Collapse
Affiliation(s)
- Caitlin O McAtee
- Department of Biochemistry, University of Nebraska, Lincoln, Nebraska, USA
| | - Joseph J Barycki
- Department of Biochemistry, University of Nebraska, Lincoln, Nebraska, USA
| | - Melanie A Simpson
- Department of Biochemistry, University of Nebraska, Lincoln, Nebraska, USA.
| |
Collapse
|
24
|
Mahata B, Banerjee A, Kundu M, Bandyopadhyay U, Biswas K. TALEN mediated targeted editing of GM2/GD2-synthase gene modulates anchorage independent growth by reducing anoikis resistance in mouse tumor cells. Sci Rep 2015; 5:9048. [PMID: 25762467 PMCID: PMC4357006 DOI: 10.1038/srep09048] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 02/16/2015] [Indexed: 01/08/2023] Open
Abstract
Complex ganglioside expression is highly deregulated in several tumors which is further dependent on specific ganglioside synthase genes. Here, we designed and constructed a pair of highly specific transcription-activator like effector endonuclease (TALENs) to disrupt a particular genomic locus of mouse GM2-synthase, a region conserved in coding sequence of all four transcript variants of mouse GM2-synthase. Our designed TALENs effectively work in different mouse cell lines and TALEN induced mutation rate is over 45%. Clonal selection strategy is undertaken to generate stable GM2-synthase knockout cell line. We have also demonstrated non-homologous end joining (NHEJ) mediated integration of neomycin cassette into the TALEN targeted GM2-synthase locus. Functionally, clonally selected GM2-synthase knockout clones show reduced anchorage-independent growth (AIG), reduction in tumor growth and higher cellular adhesion as compared to wild type Renca-v cells. Insight into the mechanism shows that, reduced AIG is due to loss in anoikis resistance, as both knockout clones show increased sensitivity to detachment induced apoptosis. Therefore, TALEN mediated precise genome editing at GM2-synthase locus not only helps us in understanding the function of GM2-synthase gene and complex gangliosides in tumorigenicity but also holds tremendous potential to use TALENs in translational cancer research and therapeutics.
Collapse
Affiliation(s)
- Barun Mahata
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Avisek Banerjee
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Manjari Kundu
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Uday Bandyopadhyay
- Department of Infectious Diseases and Immunology, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Kaushik Biswas
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| |
Collapse
|
25
|
Singha NC, Nekoroski T, Zhao C, Symons R, Jiang P, Frost GI, Huang Z, Shepard HM. Tumor-associated hyaluronan limits efficacy of monoclonal antibody therapy. Mol Cancer Ther 2014; 14:523-32. [PMID: 25512619 DOI: 10.1158/1535-7163.mct-14-0580] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Despite tremendous progress in cancer immunotherapy for solid tumors, clinical success of monoclonal antibody (mAb) therapy is often limited by poorly understood mechanisms associated with the tumor microenvironment (TME). Accumulation of hyaluronan (HA), a major component of the TME, occurs in many solid tumor types, and is associated with poor prognosis and treatment resistance in multiple malignancies. In this study, we describe that a physical barrier associated with high levels of HA (HA(high)) in the TME restricts antibody and immune cell access to tumors, suggesting a novel mechanism of in vivo resistance to mAb therapy. We determined that approximately 60% of HER2(3+) primary breast tumors and approximately 40% of EGFR(+) head and neck squamous cell carcinomas are HA(high), and hypothesized that HA(high) tumors may be refractory to mAb therapy. We found that the pericellular matrix produced by HA(high) tumor cells inhibited both natural killer (NK) immune cell access to tumor cells and antibody-dependent cell-mediated cytotoxicity (ADCC) in vitro. Depletion of HA by PEGPH20, a pegylated recombinant human PH20 hyaluronidase, resulted in increased NK cell access to HA(high) tumor cells, and greatly enhanced trastuzumab- or cetuximab-dependent ADCC in vitro. Furthermore, PEGPH20 treatment enhanced trastuzumab and NK cell access to HA(high) tumors, resulting in enhanced trastuzumab- and NK cell-mediated tumor growth inhibition in vivo. These results suggest that HA(high) matrix in vivo may form a barrier inhibiting access of both mAb and NK cells, and that PEGPH20 treatment in combination with anticancer mAbs may be an effective adjunctive therapy for HA(high) tumors.
Collapse
Affiliation(s)
| | | | - Chunmei Zhao
- Halozyme Therapeutics, Inc., San Diego, California
| | | | - Ping Jiang
- Halozyme Therapeutics, Inc., San Diego, California
| | | | | | | |
Collapse
|
26
|
Schmaus A, Sleeman JP. Hyaluronidase-1 expression promotes lung metastasis in syngeneic mouse tumor models without affecting accumulation of small hyaluronan oligosaccharides in tumor interstitial fluid. Glycobiology 2014; 25:258-68. [PMID: 25354852 DOI: 10.1093/glycob/cwu106] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Enhanced levels in tumors of hyaluronan, a glycosaminoglycan component of the extracellular matrix, and hyaluronidases such as hyaluronidase-1 (Hyal1) that degrade hyaluronan have both been linked to poor prognosis and metastasis, suggesting that the turnover of hyaluronan might contribute to tumor progression. Small hyaluronan oligosaccharides (sHA) can accumulate in tumor interstitial fluid (TIF), and have been implicated in a number of processes that drive tumor progression, including MMP expression and angiogenesis. The properties of Hyal1 suggest that it might contribute to the degradation of hyaluronan in tumors and the subsequent accumulation of sHA. Accumulation of Hyal1-produced sHA may therefore account for the association between Hyal1 and metastasis. Here we have investigated this hypothesis using mouse syngeneic breast tumor models. Specifically, we modulated Hyal1 expression and activity either in the tumor cells themselves, or in the stromal compartment by using Hyal1 knockout (KO) mice. These approaches did not change sHA levels in TIF, but nevertheless fostered metastasis to the lung in some of the models used in the study. Together, these data suggest that Hyal1 can promote lung metastasis in a manner that is not dependent on altered accumulation of sHA in TIF.
Collapse
Affiliation(s)
- Anja Schmaus
- Medical Faculty Mannheim, Centre for Biomedicine and Medical Technology Mannheim (CBTM), University of Heidelberg, Mannheim 68167, Germany Karlsruhe Institute of Technology (KIT), Campus Nord, Institut für Toxikologie und Genetik, Postfach 3640, Karlsruhe 76021, Germany
| | - Jonathan P Sleeman
- Medical Faculty Mannheim, Centre for Biomedicine and Medical Technology Mannheim (CBTM), University of Heidelberg, Mannheim 68167, Germany Karlsruhe Institute of Technology (KIT), Campus Nord, Institut für Toxikologie und Genetik, Postfach 3640, Karlsruhe 76021, Germany
| |
Collapse
|
27
|
Keereweer S, Van Driel PBAA, Robinson DJ, Lowik CWGM. Shifting focus in optical image-guided cancer therapy. Mol Imaging Biol 2014; 16:1-9. [PMID: 24037176 DOI: 10.1007/s11307-013-0688-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cancer patients could benefit from a surgical procedure that helps the surgeon to determine adequate tumor resection margins. Systemic injection of tumor-specific fluorescence agents with subsequent intraoperative optical imaging can guide the surgeon in this process. However, tumor heterogeneity hampers tumor-specific targeting. In addition, determination of adequate resection margins can be very challenging due to invasive tumor strands that are difficult to resolve and because of the confounding effect of variations in tissue optical properties in the surgical margin. We provide an overview of the "classic approach" of imaging tumor-specific targets or tumor-associated pathophysiological processes, and explain the limitations of these targeting strategies. It is proposed that problems of tumor heterogeneity can theoretically be circumvented by shifting focus of tumor targeting towards the follicle-stimulating hormone receptor (FSHR). Furthermore, we discuss why objective determination of resection margins is required to improve resection of the invasive strands, a goal that may be achieved by targeting the FSHR. When invasive strands would nevertheless extend beyond such a standardized resection margin, we suggest that adjuvant photodynamic therapy would be a very suitable therapeutic regimen. Finally, we describe how point optical spectroscopy can be used to scrutinize suspect tissue that is difficult to differentiate from normal tissue by measuring the local tissue optical properties to recover a local intrinsic fluorescence measurement.
Collapse
Affiliation(s)
- Stijn Keereweer
- Department of Molecular Imaging, Leiden University Medical Center, Leiden, The Netherlands,
| | | | | | | |
Collapse
|
28
|
Hyaluronan in aged collagen matrix increases prostate epithelial cell proliferation. In Vitro Cell Dev Biol Anim 2014; 51:50-8. [PMID: 25124870 DOI: 10.1007/s11626-014-9800-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 07/13/2014] [Indexed: 10/24/2022]
Abstract
The extracellular matrix (ECM) of the prostate, which is comprised primarily of collagen, becomes increasingly disorganized with age, a property that may influence the development of hyperplasia and cancer. Collageous ECM extracted from the tails of aged mice exhibits many characteristics of collagen in aged tissues, including the prostate. When polymerized into a 3-dimensional (3D) gel, these collagen extracts can serve as models for the study of specific cell-ECM interactions. In the present study, we examined the behaviors of human prostatic epithelial cell lines representing normal prostate epithelial cells (PEC), benign prostatic hyperplasia (BPH-1), and adenocarcinoma (LNCaP) cultured in contact with 3D gels made from collagen extracts of young and aged mice. We found that proliferation of PEC, BPH-1, and LNCaP cells were all increased by culture on aged collagen gels relative to young collagen gels. In examining age-associated differences in the composition of the collagen extracts, we found that aged and young collagen had a similar amount of several collagen-associated ECM components, but aged collagen had a much greater content of the glycosaminoglycan hyaluronan (HA) than young collagen. The addition of HA (of similar size and concentration to that found in aged collagen extracts) to cells placed in young collagen elicited significantly increased proliferation in BPH-1 cells, but not in PEC or LNCaP cells, relative to controls not exposed to HA. Of note, histochemical analyses of human prostatic tissues showed significantly higher expression of HA in BPH and prostate cancer stroma relative to stroma of normal prostate. Collectively, these results suggest that changes in ECM involving increased levels of HA contribute to the growth of prostatic epithelium with aging.
Collapse
|
29
|
Wang Y, Luo Y, Bing T, Chen Z, Lu M, Zhang N, Shangguan D, Gao X. DNA aptamer evolved by cell-SELEX for recognition of prostate cancer. PLoS One 2014; 9:e100243. [PMID: 24956390 PMCID: PMC4067300 DOI: 10.1371/journal.pone.0100243] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2014] [Accepted: 05/23/2014] [Indexed: 01/01/2023] Open
Abstract
Morbidity and mortality of prostate cancer (PCa) have increased in recent years worldwide. Currently existing methods for diagnosis and treatment do not make the situation improve, especially for hormone refractory prostate cancer (HRPC). The lack of molecular probes for PCa hindered the early diagnosis of metastasis and accurate staging for PCa. In this work, we have developed a new aptamer probe Wy-5a against PCa cell line PC-3 by cell-SELEX technique. Wy-5a shows high specificity to the target cells with dissociation constants in the nanomolar range, and does not recognize other tested PCa cell lines and other tested tumor cell lines. The staining of clinical tissue sections with fluorescent dye labeled Wy-5a shows that sections from high risk group with metastasis exhibited stronger fluorescence and sections from Benign Prostatic Hyperplasia (BPH) did not exhibit notable fluorescence, which suggests that aptamer Wy-5a may bind to protein related to the progression of PCa. The high affinity and specificity of Wy-5a makes this aptamer hold potential for application in diagnosis and target therapy of PCa.
Collapse
Affiliation(s)
- Yuanyuan Wang
- Department of Urology, the third affiliated hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yun Luo
- Department of Urology, the third affiliated hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Tao Bing
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China
| | - Zheng Chen
- Department of Urology, the third affiliated hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Minhua Lu
- Department of Urology, the third affiliated hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Nan Zhang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China
| | - Dihua Shangguan
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China
- * E-mail: (XG); (DS)
| | - Xin Gao
- Department of Urology, the third affiliated hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
- * E-mail: (XG); (DS)
| |
Collapse
|
30
|
Schmaus A, Klusmeier S, Rothley M, Dimmler A, Sipos B, Faller G, Thiele W, Allgayer H, Hohenberger P, Post S, Sleeman JP. Accumulation of small hyaluronan oligosaccharides in tumour interstitial fluid correlates with lymphatic invasion and lymph node metastasis. Br J Cancer 2014; 111:559-67. [PMID: 24937668 PMCID: PMC4119989 DOI: 10.1038/bjc.2014.332] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Revised: 04/09/2014] [Accepted: 05/15/2014] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Association studies have implicated the glycosaminoglycan hyaluronan (hyaluronic acid, HA) and its degrading enzymes the hyaluronidases in tumour progression and metastasis. Oligosaccharides of degraded HA have been ascribed a number of biological functions that are not exerted by high-molecular-weight HA (HMW-HA). However, whether these small HA oligosaccharides (sHA) have a role in tumour progression currently remains uncertain due to an inability to analyse their concentration in tumours. METHODS We report a novel method to determine the concentration of sHA ranging from 6 to 25 disaccharides in tumour interstitial fluid (TIF). Levels of sHA were measured in TIF from experimental rat tumours and human colorectal tumours. RESULTS While the majority of HA in TIF is HMW-HA, concentrations of sHA up to 6 μg ml(-1) were detected in a subset of tumours, but not in interstitial fluid from healthy tissues. In a cohort of 72 colorectal cancer patients we found that increased sHA concentrations in TIF are associated with lymphatic vessel invasion by tumour cells and the formation of lymph node metastasis. CONCLUSIONS These data document for the first time the pathophysiological concentration of sHA in tumours, and provide evidence of a role for sHA in tumour progression.
Collapse
Affiliation(s)
- A Schmaus
- 1] Medical Faculty Mannheim, University of Heidelberg, Ludolf-Krehl-Strasse 13-17, 68167 Mannheim, Germany [2] Karlsruhe Institute for Technology (KIT), Campus Nord, Institut für Toxikologie und Genetik, Postfach 3640, 76021 Karlsruhe, Germany
| | - S Klusmeier
- 1] Medical Faculty Mannheim, University of Heidelberg, Ludolf-Krehl-Strasse 13-17, 68167 Mannheim, Germany [2] Karlsruhe Institute for Technology (KIT), Campus Nord, Institut für Toxikologie und Genetik, Postfach 3640, 76021 Karlsruhe, Germany
| | - M Rothley
- 1] Medical Faculty Mannheim, University of Heidelberg, Ludolf-Krehl-Strasse 13-17, 68167 Mannheim, Germany [2] Karlsruhe Institute for Technology (KIT), Campus Nord, Institut für Toxikologie und Genetik, Postfach 3640, 76021 Karlsruhe, Germany
| | - A Dimmler
- Institut und Gemeinschaftspraxis für Pathologie an den St Vincentiuskliniken Karlsruhe, Südendstrasse 37, 76137 Karlsruhe, Germany
| | - B Sipos
- Universitätsklinikum Tübingen, Department of Pathology, Liebermeisterstrasse 8, 72076 Tübingen, Germany
| | - G Faller
- Institut und Gemeinschaftspraxis für Pathologie an den St Vincentiuskliniken Karlsruhe, Südendstrasse 37, 76137 Karlsruhe, Germany
| | - W Thiele
- 1] Medical Faculty Mannheim, University of Heidelberg, Ludolf-Krehl-Strasse 13-17, 68167 Mannheim, Germany [2] Karlsruhe Institute for Technology (KIT), Campus Nord, Institut für Toxikologie und Genetik, Postfach 3640, 76021 Karlsruhe, Germany
| | - H Allgayer
- Medical Faculty Mannheim, University of Heidelberg, Ludolf-Krehl-Strasse 13-17, 68167 Mannheim, Germany
| | - P Hohenberger
- Medical Faculty Mannheim, University of Heidelberg, Ludolf-Krehl-Strasse 13-17, 68167 Mannheim, Germany
| | - S Post
- Medical Faculty Mannheim, University of Heidelberg, Ludolf-Krehl-Strasse 13-17, 68167 Mannheim, Germany
| | - J P Sleeman
- 1] Medical Faculty Mannheim, University of Heidelberg, Ludolf-Krehl-Strasse 13-17, 68167 Mannheim, Germany [2] Karlsruhe Institute for Technology (KIT), Campus Nord, Institut für Toxikologie und Genetik, Postfach 3640, 76021 Karlsruhe, Germany
| |
Collapse
|
31
|
Hwang SG, Yang A, Kim SJ, Kim MK, Kim SS, Oh HJ, Lee JD, Lee EJ, Nam KW, Han MD. Screening of Hyaluronidase Inhibitor in Korean Medicinal Plants. ACTA ACUST UNITED AC 2014. [DOI: 10.5352/jls.2014.24.5.498] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
32
|
Pharmacokinetic and Biodistribution Assessment of a Near Infrared-Labeled PSMA-Specific Small Molecule in Tumor-Bearing Mice. Prostate Cancer 2014; 2014:104248. [PMID: 24804103 PMCID: PMC3997074 DOI: 10.1155/2014/104248] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2013] [Revised: 02/17/2014] [Accepted: 03/07/2014] [Indexed: 02/05/2023] Open
Abstract
Prostate cancer is the most frequently diagnosed cancer in men and often requires surgery. Use of near infrared (NIR) technologies to perform image-guided surgery may improve accurate delineation of tumor margins. To facilitate preclinical testing of such outcomes, here we developed and characterized a PSMA-targeted small molecule, YC-27. IRDye 800CW was conjugated to YC-27 or an anti-PSMA antibody used for reference. Human 22Rv1, PC3M-LN4, and/or LNCaP prostate tumor cells were exposed to the labeled compounds. In vivo targeting and clearance properties were determined in tumor-bearing mice. Organs and tumors were excised and imaged to assess probe localization. YC-27 exhibited a dose dependent increase in signal upon binding. Binding specificity and internalization were visualized by microscopy. In vitro and in vivo blocking studies confirmed YC-27 specificity. In vivo, YC-27 showed good tumor delineation and tissue contrast at doses as low as 0.25 nmole. YC-27 was cleared via the kidneys but bound the proximal tubules of the renal cortex and epididymis. Since PSMA is also broadly expressed on the neovasculature of most tumors, we expect YC-27 will have clinical utility for image-guided surgery and tumor resections.
Collapse
|
33
|
Gong H, Kovar JL, Cheung L, Rosenthal EL, Olive DM. A comparative study of affibody, panitumumab, and EGF for near-infrared fluorescence imaging of EGFR- and EGFRvIII-expressing tumors. Cancer Biol Ther 2013; 15:185-93. [PMID: 24100437 DOI: 10.4161/cbt.26719] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Aberrant overexpression and/or activation of epidermal growth factor receptor (EGFR) is associated with many types of cancers. EGFR variant III (EGFRvIII) is a common in-frame deletion mutant, which lacks a large part of the extracellular portion (exons 2-7), including components of the ligand-binding domain. Although EGFR has been extensively studied as a molecular imaging target, information about EGFRvIII-targeted molecular imaging is lacking. In this study, the EGFR-specific affibody, therapeutic antibody panitumumab, and ligand EGF were labeled with IRDye 800CW (Ex/Em: 774/789 nm), yielding Aff800, Pan800, and EGF800, respectively. The binding affinities of the labeled agents were compared in cell-based assays using a rat glioma cell line F98 parental (F98-p) lacking EGFR expression, and 2 F98-derived transgenic cell lines expressing EGFR or EGFRvIII (designated as F98-EGFR and F98-vIII, respectively). Results showed that all agents could bind to F98-EGFR, with Pan800 having the highest binding affinity, followed by Aff800 and EGF800. Pan800 and Aff800, but not EGF800, also bound to F98-vIII. In vivo animal imaging demonstrated that compared with F98-p tumors, F98-EGFR tumors generated higher signals with all three agents. However, in the case of F98-vIII, only Pan800 and Aff800 signals were higher. Analysis of tissue lysates showed that a large portion of Pan800 was degraded into small fragments in F98-EGFR and F98-vIII tumors, possibly due to proteolytic digestion after its specific binding and internalization. In conclusion, Pan800 and Aff800 could be used as imaging agents for both wild-type EGFR and EGFRvIII, whereas EGF800 only targets wild-type EGFR.
Collapse
Affiliation(s)
| | | | | | - Eben L Rosenthal
- Division of Otolaryngology; Head and Neck Surgery; University of Alabama at Birmingham; Birmingham, AL USA
| | | |
Collapse
|
34
|
Roca H, Hernandez J, Weidner S, McEachin RC, Fuller D, Sud S, Schumann T, Wilkinson JE, Zaslavsky A, Li H, Maher CA, Daignault-Newton S, Healy PN, Pienta KJ. Transcription factors OVOL1 and OVOL2 induce the mesenchymal to epithelial transition in human cancer. PLoS One 2013; 8:e76773. [PMID: 24124593 PMCID: PMC3790720 DOI: 10.1371/journal.pone.0076773] [Citation(s) in RCA: 185] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Accepted: 08/28/2013] [Indexed: 12/03/2022] Open
Abstract
Cell plasticity regulated by the balance between the mesenchymal to epithelial transition (MET) and the opposite program, EMT, is critical in the metastatic cascade. Several transcription factors (TFs) are known to regulate EMT, though the mechanisms of MET remain unclear. We demonstrate a novel function of two TFs, OVOL1 and OVOL2, as critical inducers of MET in human cancers. Our findings indicate that the OVOL-TFs control MET through a regulatory feedback loop with EMT-inducing TF ZEB1, and the regulation of mRNA splicing by inducing Epithelial Splicing Regulatory Protein 1 (ESRP1). Using mouse prostate tumor models we show that expression of OVOL-TFs in mesenchymal prostate cancer cells attenuates their metastatic potential. The role of OVOL-TFs as inducers of MET is further supported by expression analyses in 917 cancer cell lines, suggesting their role as crucial regulators of epithelial-mesenchymal cell plasticity in cancer.
Collapse
Affiliation(s)
- Hernan Roca
- Department of Urology, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
- * E-mail:
| | - James Hernandez
- Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
| | - Savannah Weidner
- Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
| | - Richard C. McEachin
- Department of Computational Medicine and Bioinformatics, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
| | - David Fuller
- Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
| | - Sudha Sud
- Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
| | - Taibriana Schumann
- Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
| | - John E. Wilkinson
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
| | - Alexander Zaslavsky
- Department of Urology, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
| | - Hangwen Li
- Department of Urology, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
| | - Christopher A. Maher
- The Genome Institute, Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Stephanie Daignault-Newton
- Division of Biostatistics, Comprehensive Cancer Center, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
| | - Patrick N. Healy
- Division of Biostatistics, Comprehensive Cancer Center, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
| | - Kenneth J. Pienta
- Department of Urology, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
- Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
| |
Collapse
|
35
|
Yim H, Park SJ, Bae YH, Na K. Biodegradable cationic nanoparticles loaded with an anticancer drug for deep penetration of heterogeneous tumours. Biomaterials 2013; 34:7674-82. [DOI: 10.1016/j.biomaterials.2013.06.058] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 06/26/2013] [Indexed: 12/21/2022]
|
36
|
Cohen G, Lecht S, Oron-Herman M, Momic T, Nissan A, Lazarovici P. Near infrared optical visualization of epidermal growth factor receptors levels in COLO205 colorectal cell line, orthotopic tumor in mice and human biopsies. Int J Mol Sci 2013; 14:14669-88. [PMID: 23857061 PMCID: PMC3742266 DOI: 10.3390/ijms140714669] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 07/05/2013] [Indexed: 01/05/2023] Open
Abstract
In this study, we present the applicability of imaging epidermal growth factor (EGF) receptor levels in preclinical models of COLO205 carcinoma cells in vitro, mice with orthotopic tumors and ex vivo colorectal tumor biopsies, using EGF-labeled with IRDye800CW (EGF-NIR). The near infrared (NIR) bio-imaging of COLO205 cultures indicated specific and selective binding, reflecting EGF receptors levels. In vivo imaging of tumors in mice showed that the highest signal/background ratio between tumor and adjacent tissue was achieved 48 hours post-injection. Dissected colorectal cancer tissues from different patients demonstrated ex vivo specific imaging using the NIR bio-imaging platform of the heterogeneous distributed EGF receptors. Moreover, in the adjacent gastrointestinal tissue of the same patients, which by Western blotting was demonstrated as EGF receptor negative, no labeling with EGF-NIR probe was detected. Present results support the concept of tumor imaging by measuring EGF receptor levels using EGF-NIR probe. This platform is advantageous for EGF receptor bio-imaging of the NCI-60 recommended panel of tumor cell lines including 6-9 colorectal cell lines, since it avoids radioactive probes and is appropriate for use in the clinical setting using NIR technologies in a real-time manner.
Collapse
Affiliation(s)
- Gadi Cohen
- School of Pharmacy, Institute for Drug Research, Faculty of Medicine, the Hebrew University of Jerusalem, Jerusalem 91120, Israel; E-Mails: (G.C.); (S.L.); (T.M.)
| | - Shimon Lecht
- School of Pharmacy, Institute for Drug Research, Faculty of Medicine, the Hebrew University of Jerusalem, Jerusalem 91120, Israel; E-Mails: (G.C.); (S.L.); (T.M.)
| | - Mor Oron-Herman
- Advanced Technology Center, the Chaim Sheba Medical Center, Tel-Hashomer 52621, Israel; E-Mail:
| | - Tatjana Momic
- School of Pharmacy, Institute for Drug Research, Faculty of Medicine, the Hebrew University of Jerusalem, Jerusalem 91120, Israel; E-Mails: (G.C.); (S.L.); (T.M.)
| | - Aviram Nissan
- Department of Surgery, Hadassah-Hebrew University Medical Center, Jerusalem 24035, Israel; E-Mail:
| | - Philip Lazarovici
- School of Pharmacy, Institute for Drug Research, Faculty of Medicine, the Hebrew University of Jerusalem, Jerusalem 91120, Israel; E-Mails: (G.C.); (S.L.); (T.M.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +972-2-675-8729; Fax: +972-2-675-7490
| |
Collapse
|
37
|
Cohen G, Lecht S, Arien-Zakay H, Ettinger K, Amsalem O, Oron-Herman M, Yavin E, Prus D, Benita S, Nissan A, Lazarovici P. Bio-imaging of colorectal cancer models using near infrared labeled epidermal growth factor. PLoS One 2012; 7:e48803. [PMID: 23144978 PMCID: PMC3493605 DOI: 10.1371/journal.pone.0048803] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2012] [Accepted: 10/01/2012] [Indexed: 01/11/2023] Open
Abstract
Novel strategies that target the epidermal growth factor receptor (EGFR) have led to the clinical development of monoclonal antibodies, which treat metastatic colorectal cancer (mCRC) but only subgroups of patients with increased wild type KRAS and EGFR gene copy, respond to these agents. Furthermore, resistance to EGFR blockade inevitably occurred, making future therapy difficult. Novel bio-imaging (BOI) methods may assist in quantization of EGFR in mCRC tissue thus complementing the immunohistochemistry methodology, in guiding the future treatment of these patients. The aim of the present study was to explore the usefulness of near infrared-labeled EGF (EGF-NIR) for bio-imaging of CRC using in vitro and in vivo orthotopic tumor CRC models and ex vivo human CRC tissues. We describe the preparation and characterization of EGF-NIR and investigate binding, using BOI of a panel of CRC cell culture models resembling heterogeneity of human CRC tissues. EGF-NIR was specifically and selectively bound by EGFR expressing CRC cells, the intensity of EGF-NIR signal to background ratio (SBR) reflected EGFR levels, dose-response and time course imaging experiments provided optimal conditions for quantization of EGFR levels by BOI. EGF-NIR imaging of mice with HT-29 orthotopic CRC tumor indicated that EGF-NIR is more slowly cleared from the tumor and the highest SBR between tumor and normal adjacent tissue was achieved two days post-injection. Furthermore, images of dissected tissues demonstrated accumulation of EGF-NIR in the tumor and liver. EGF-NIR specifically and strongly labeled EGFR positive human CRC tissues while adjacent CRC tissue and EGFR negative tissues expressed weak NIR signals. This study emphasizes the use of EGF-NIR for preclinical studies. Combined with other methods, EGF-NIR could provide an additional bio-imaging specific tool in the standardization of measurements of EGFR expression in CRC tissues.
Collapse
Affiliation(s)
- Gadi Cohen
- School of Pharmacy Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Shimon Lecht
- School of Pharmacy Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Hadar Arien-Zakay
- School of Pharmacy Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Keren Ettinger
- School of Pharmacy Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Orit Amsalem
- School of Pharmacy Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Mor Oron-Herman
- Advanced Technology Center, The Chaim Sheba Medical Center, Tel-Hashomer, Israel
| | - Eylon Yavin
- School of Pharmacy Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Diana Prus
- Department of Pathology and Surgical Oncology Laboratory, Hadassah-Hebrew University Medical Center, Mount Scopus, Jerusalem, Israel
| | - Simon Benita
- School of Pharmacy Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Aviram Nissan
- Department of Surgery, Hadassah-Hebrew University Medical Center, Mount Scopus, Jerusalem, Israel
| | - Philip Lazarovici
- School of Pharmacy Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
- * E-mail:
| |
Collapse
|
38
|
Sankpal UT, Abdelrahim M, Connelly SF, Lee CM, Madero-Visbal R, Colon J, Smith J, Safe S, Maliakal P, Basha R. Small molecule tolfenamic acid inhibits PC-3 cell proliferation and invasion in vitro, and tumor growth in orthotopic mouse model for prostate cancer. Prostate 2012; 72:1648-58. [PMID: 22473873 DOI: 10.1002/pros.22518] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Accepted: 02/24/2012] [Indexed: 02/02/2023]
Abstract
BACKGROUND Specificity protein (Sp) transcription factors are implicated in critical cellular and molecular processes associated with cancer that impact tumor growth and metastasis. The non-steroidal anti-inflammatory drug, tolfenamic acid (TA) is known to inhibit Sp proteins in some human cancer cells and laboratory animal models. We evaluated the anti-cancer activity of TA using in vitro and in vivo models for prostate cancer. METHODS The anti-proliferative efficacy of TA was evaluated using DU-145, PC-3, and LNCaP cells. PC-3 cells were treated with DMSO or 50 µM TA for 48 hr. Whole cell lysates were evaluated for the expression of Sp1, survivin, c-PARP, Akt/p-Akt, c-Met, cdk4, cdc2, cyclin D3, and E2F1 by Western blot analysis. Cell invasion was assessed by Boyden-chamber assay and flow cytometry analysis was used to study apoptosis and cell cycle distribution. An orthotopic mouse model for prostate cancer with PC-3-Luc cells was used to study the in vivo effect of TA. RESULTS TA inhibited the expression of Sp1, c-Met, p-Akt, and survivin; increased c-PARP expression and caspases activity in PC-3 cells. TA caused cell arrest at G(0) /G(1) phase accompanied by a decrease in cdk4, cdc2, cyclin D3, and E2F1 and an increase in critical apoptotic markers. TA augmented annexin-V staining, caspase activity, and c-PARP expression indicating the activation of apoptotic pathways. TA also decreased PC-3 cell invasion. TA significantly decreased the tumor weight and volume which was associated with low expression of Sp1 and survivin in tumor sections. CONCLUSION TA targets critical pathways associated with tumorigenesis and invasion. These pre-clinical data strongly demonstrated the anti-cancer activity of TA in prostate cancer.
Collapse
Affiliation(s)
- Umesh T Sankpal
- Cancer Research Institute, MD Anderson Cancer Center Orlando, Orlando, Florida 32827, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
el Filali M, Ly LV, Luyten GP, Versluis M, Grossniklaus HE, van der Velden PA, Jager MJ. Bevacizumab and intraocular tumors: an intriguing paradox. Mol Vis 2012; 18:2454-67. [PMID: 23077404 PMCID: PMC3472924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2011] [Accepted: 10/03/2012] [Indexed: 11/25/2022] Open
Abstract
PURPOSE Bevacizumab, a humanized monoclonal antibody to vascular endothelial growth factor-A (VEGF-A), was originally developed as an anti-tumor treatment. In ocular oncology, it is being used to treat macular edema due to radiation retinopathy, but it may also be useful for the treatment of primary uveal melanoma (UM) or its metastases. We determined the effect of bevacizumab on the growth of B16F10 cells inside the eye and on B16F10 and UM cells cultured in vitro. METHODS B16F10 melanoma cells were placed into the anterior chamber of the eye of C57Bl/6 mice and tumor growth was monitored after injection of different doses of bevacizumab or mock injection. In addition, the effect of bevacizumab on in vitro growth of B16F10 and human UM cells and on the expression of VEGF-A, GLUT-1, and HIF-1α was evaluated. RESULTS Following intraocular injection of bevacizumab into murine B16 tumor-containing eyes, an acceleration of tumor growth was observed, with the occurrence of anterior chamber hemorrhages. Bevacizumab did not affect proliferation of B16F10 cells in vitro, while it inhibited UM cell proliferation. Expression analysis demonstrated that addition of bevacizumab under hypoxic conditions induced VEGF-A, GLUT-1 and HIF-1α in B16F10 cells as well as in UM cell lines and two of four primary UM tumor cultures. CONCLUSIONS In contrast with expectations, intraocular injection of bevacizumab stimulated B16F10 melanoma growth in murine eyes. In vitro exposure of B16 and human UM cells to bevacizumab led to paradoxical VEGF-A upregulation. The use of VEGF inhibitors for treatment of macular edema (due to radiation retinopathy) after irradiation of UM should be considered carefully, because of the possible adverse effects on residual UM cells.
Collapse
Affiliation(s)
| | - Long V. Ly
- Department of Ophthalmology, LUMC, Leiden, the Netherlands
| | | | | | - Hans E. Grossniklaus
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, GA,Department of Pathology, Emory University School of Medicine, Atlanta, GA
| | | | | |
Collapse
|
40
|
Jose J, Maas RM, Teese MG. Autodisplay of enzymes—Molecular basis and perspectives. J Biotechnol 2012; 161:92-103. [DOI: 10.1016/j.jbiotec.2012.04.001] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2011] [Revised: 02/14/2012] [Accepted: 04/04/2012] [Indexed: 11/16/2022]
|
41
|
Samkoe KS, Sexton K, Tichauer KM, Hextrum SK, Pardesi O, Davis SC, O’Hara JA, Hoopes PJ, Hasan T, Pogue BW. High vascular delivery of EGF, but low receptor binding rate is observed in AsPC-1 tumors as compared to normal pancreas. Mol Imaging Biol 2012; 14:472-9. [PMID: 21847690 PMCID: PMC3529005 DOI: 10.1007/s11307-011-0503-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
PURPOSE Cellular receptor targeted imaging agents present the potential to target extracellular molecular expression in cancerous lesions; however, the image contrast in vivo does not reflect the magnitude of overexpression expected from in vitro data. Here, the in vivo delivery and binding kinetics of epidermal growth factor receptor (EGFR) was determined for normal pancreas and AsPC-1 orthotopic pancreatic tumors known to overexpress EGFR. PROCEDURES EGFR in orthotopic xenograft AsPC-1 tumors was targeted with epidermal growth factor (EGF) conjugated with IRDye800CW. The transfer rate constants (k(e), K₁₂, k₂₁, k₂₃, and k₃₂) associated with a three-compartment model describing the vascular delivery, leakage rate and binding of targeted agents were determined experimentally. The plasma excretion rate, k (e), was determined from extracted blood plasma samples. K₁₂, k₂₁, and k₃₂ were determined from ex vivo tissue washing studies at time points ≥ 24 h. The measured in vivo uptake of IRDye800CW-EGF and a non-targeted tracer dye, IRDye700DX-carboxylate, injected simultaneously was used to determined k₂₃. RESULTS The vascular exchange of IRDye800CW-EGF in the orthotopic tumor (K₁₂ and k₂₁) was higher than in the AsPC-1 tumor as compared to normal pancreas, suggesting that more targeted agent can be taken up in tumor tissue. However, the cellular associated (binding) rate constant (k₂₃) was slightly lower for AsPC-1 pancreatic tumor (4.1 × 10(-4) s(-1)) than the normal pancreas (5.5 × 10(-4) s(-1)), implying that less binding is occurring. CONCLUSIONS Higher vascular delivery but low cellular association in the AsPC-1 tumor compared to the normal pancreas may be indicative of low receptor density due to low cellular content. This attribute of the AsPC-1 tumor may indicate one contributing cause of the difficulty in treating pancreatic tumors with cellular targeted agents.
Collapse
Affiliation(s)
- Kimberley S. Samkoe
- Thayer School of Engineering, Dartmouth College, 8000 Cummings Hall, Hanover, NH 03755, USA
| | - Kristian Sexton
- Thayer School of Engineering, Dartmouth College, 8000 Cummings Hall, Hanover, NH 03755, USA
| | - Kenneth M. Tichauer
- Thayer School of Engineering, Dartmouth College, 8000 Cummings Hall, Hanover, NH 03755, USA
| | - Shannon K. Hextrum
- Thayer School of Engineering, Dartmouth College, 8000 Cummings Hall, Hanover, NH 03755, USA
| | - Omar Pardesi
- Thayer School of Engineering, Dartmouth College, 8000 Cummings Hall, Hanover, NH 03755, USA
| | - Scott C. Davis
- Thayer School of Engineering, Dartmouth College, 8000 Cummings Hall, Hanover, NH 03755, USA
| | - Julia A. O’Hara
- Thayer School of Engineering, Dartmouth College, 8000 Cummings Hall, Hanover, NH 03755, USA
| | - P. Jack Hoopes
- Thayer School of Engineering, Dartmouth College, 8000 Cummings Hall, Hanover, NH 03755, USA
- Department of Surgery, Dartmouth Medical School, Hanover, NH 03755, USA
| | - Tayyaba Hasan
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Brian W. Pogue
- Thayer School of Engineering, Dartmouth College, 8000 Cummings Hall, Hanover, NH 03755, USA
- Department of Surgery, Dartmouth Medical School, Hanover, NH 03755, USA
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
| |
Collapse
|
42
|
Wang J, Lu Z, Gao Y, Wientjes MG, Au JLS. Improving delivery and efficacy of nanomedicines in solid tumors: role of tumor priming. Nanomedicine (Lond) 2012; 6:1605-20. [PMID: 22077464 DOI: 10.2217/nnm.11.141] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Effectiveness of nanomedicines in cancer therapy is limited in part by inadequate delivery and transport in tumor interstitium. This article reviews the experimental approaches to improve nanomedicine delivery and transport in solid tumors. These approaches include tumor vasculature normalization, interstitial fluid pressure modulation, enzymatic extracellular matrix degradation, and apoptosis-inducing tumor priming technology. We advocate the latter approach due to its ease and practicality (accomplished with standard-of-care chemotherapy, such as paclitaxel) and tumor selectivity. Examples of applying tumor priming to deliver nanomedicines and to design drug/RNAi-loaded carriers are discussed.
Collapse
Affiliation(s)
- Jie Wang
- Optimum Therapeutics LLC, The Ohio State University Science Tech Village, Columbus, OH 43212, USA
| | | | | | | | | |
Collapse
|
43
|
Keereweer S, Mol IM, Vahrmeijer AL, Van Driel PBAA, Baatenburg de Jong RJ, Kerrebijn JDF, Löwik CWGM. Dual wavelength tumor targeting for detection of hypopharyngeal cancer using near-infrared optical imaging in an animal model. Int J Cancer 2012; 131:1633-40. [PMID: 22234729 DOI: 10.1002/ijc.27430] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2011] [Accepted: 12/21/2011] [Indexed: 12/29/2022]
Abstract
Optical imaging is a promising technique to visualize cancer tissue during surgery. In this study, we explored the use of combinations of near-infrared (NIR) fluorescence agents that emit fluorescence signal at different wavelengths and each target specific tumor characteristics. Two combinations of agents (ProSense680 combined with 2DG CW800 and MMPSense680 combined with EGF CW800) were used to detect hypopharyngeal cancer in an animal model. ProSense680 and MMPSense680 detect increased activity of cathepsins and matrix metalloproteinases, respectively. These enzymes are mainly found in the invasive tumor border due to degradation of the extracellular matrix. 2DG CW800 detects tumor cells with high glucose metabolism and EGF CW800 is internalized by the epidermal growth factor receptor of tumor cells. Whole-body imaging revealed clear demarcation of tumor tissue using all four agents. The tumor-to-background ratio (standard deviation, p-value) was 3.69 (0.72, p < 0.001) for ProSense680; 4.26 (1.33, p < 0.001) for MMPSense680; 5.81 (3.59, p = 0.02) for 2DG CW800 and 4.84 (1.56, p < 0.001) for EGF CW800. Fluorescence signal corresponded with histopathology and immunohistochemistry, demonstrating signal of ProSense680 and MMPSense680 in the invasive tumor border, and signal of 2DG CW800 and EGF CW800 in the tumor tissue. In conclusion, we demonstrated the feasibility of dual wavelength tumor detection using different targeting strategies simultaneously in an animal model. Combined targeting at different wavelengths allowed simultaneous imaging of different tumor characteristics. NIR fluorescence optical imaging has the potential to be translated into the clinic in order to improve the complete removal of tumors by real-time image-guided surgery.
Collapse
Affiliation(s)
- Stijn Keereweer
- Department of Otorhinolaryngology Head and Neck Surgery, Erasmus Medical Center, Rotterdam, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
44
|
Xie BW, Mol IM, Keereweer S, van Beek ER, Que I, Snoeks TJA, Chan A, Kaijzel EL, Löwik CWGM. Dual-wavelength imaging of tumor progression by activatable and targeting near-infrared fluorescent probes in a bioluminescent breast cancer model. PLoS One 2012; 7:e31875. [PMID: 22348134 PMCID: PMC3278453 DOI: 10.1371/journal.pone.0031875] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Accepted: 01/13/2012] [Indexed: 12/29/2022] Open
Abstract
Bioluminescence imaging (BLI) has shown its appeal as a sensitive technique for in vivo whole body optical imaging. However, the development of injectable tumor-specific near-infrared fluorescent (NIRF) probes makes fluorescence imaging (FLI) a promising alternative to BLI in situations where BLI cannot be used or is unwanted (e.g., spontaneous transgenic tumor models, or syngeneic mice to study immune effects). In this study, we addressed the questions whether it is possible to detect tumor progression using FLI with appropriate sensitivity and how FLI correlates with BLI measurements. In addition, we explored the possibility to simultaneously detect multiple tumor characteristics by dual-wavelength FLI (∼700 and ∼800 nm) in combination with spectral unmixing. Using a luciferase-expressing 4T1-luc2 mouse breast cancer model and combinations of activatable and targeting NIRF probes, we showed that the activatable NIRF probes (ProSense680 and MMPSense680) and the targeting NIRF probes (IRDye 800CW 2-DG and IRDye 800CW EGF) were either activated by or bound to 4T1-luc2 cells. In vivo, we implanted 4T1-luc2 cells orthotopically in nude mice and were able to follow tumor progression longitudinally both by BLI and dual-wavelength FLI. We were able to reveal different probe signals within the tumor, which co-localized with immuno-staining. Moreover, we observed a linear correlation between the internal BLI signals and the FLI signals obtained from the NIRF probes. Finally, we could detect pulmonary metastases both by BLI and FLI and confirmed their presence histologically. Taken together, these data suggest that dual-wavelength FLI is a feasible approach to simultaneously detect different features of one tumor and to follow tumor progression with appropriate specificity and sensitivity. This study may open up new perspectives for the detection of tumors and metastases in various experimental models and could also have clinical applications, such as image-guided surgery.
Collapse
Affiliation(s)
- Bang-Wen Xie
- Department of Endocrinology and Metabolic Diseases, Leiden University Medical Center, Leiden, The Netherlands.
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Yao Y, Jiang Y, Sheng Z, Zhang Y, An Y, Yan F, Ma G, Liu N, Teng G, Cheng Z. Analysis of in situ and ex vivo αVβ3 integrin expression during experimental carotid atherogenesis. Int J Nanomedicine 2012; 7:641-9. [PMID: 22334786 PMCID: PMC3278228 DOI: 10.2147/ijn.s28065] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
OBJECTIVE Mural inflammation has been shown to contribute to the development of plaque, with the α(V)β(3) integrin highly expressed in atherosclerotic plaques. We herein examined α(V)β(3) integrin expression as a function of carotid atherosclerosis formation in the apolipoprotein E-deficient (apoE(-/-)) mouse. METHODS AND RESULTS Constrictive collars were placed around the left common carotid arteries of apo E(-/-) mice maintained on a high-fat diet (n = 14). Before and 21 days following collar placement, in vivo serial magnetic resonance imaging (MRI) measurements of the carotid aortic diameter were performed using a 7T magnetic resonance (MR) scanner. Near- infrared fluorescence (NIRF) imaging was performed (n = 6) using an in vivo imaging system 0-24 hours following administration of 1.0 nmol c(RGDyK)-Cy5.5 via the tail vein. A competition experiment was performed by the co-injection of a saturating dose of bicyclic RGD peptide H-Glu[cyclo(Arg-Gly-Asp-D-Tyr-Lys)]2 (n = 3). Following image acquisition and sacrifice at 24 hours after injection, carotid arteries were harvested for histological analyses. Neointima formation and arterial remodeling in the carotid arteries of apoE(-/-) mice were induced by the placement of a constrictive collar. Significantly greater fluorescent signals were obtained from constrictive collar left common carotid arteries as compared to uninvolved aortic segments in constrictive collar mice. Binding to stenotic lesions was efficiently blocked in competition experiments. Immunostaining confirmed the presence of mural α(V)β(3) integrin expression in macrophages in the neointima. Signal intensity increased in a macrophage density-dependent fashion in the stenotic segments. CONCLUSION Mural α(V)β(3) integrin expression, as determined using RGD-Cy5.5 near-infrared optical imaging, was increased in carotid arteries with constrictive collars in experimental mice. This expression can estimate the macrophage-bound inflammatory activity of atherosclerotic lesions.
Collapse
Affiliation(s)
- Yuyu Yao
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, Jiangsu, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Histone deacetylase inhibitors restore cell surface expression of the coxsackie adenovirus receptor and enhance CMV promoter activity in castration-resistant prostate cancer cells. Prostate Cancer 2012; 2012:137163. [PMID: 22288017 PMCID: PMC3263646 DOI: 10.1155/2012/137163] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Accepted: 09/27/2011] [Indexed: 12/20/2022] Open
Abstract
Adenoviral gene therapy using the death receptor ligand TRAIL as the therapeutic transgene can be safely administered via intraprostatic injection but has not been evaluated for efficacy in patients. Here we investigated the efficacy of adenoviral TRAIL gene therapy in a model of castration resistant prostate cancer and found that intratumoral injections can significantly delay tumor growth but cannot eliminate established lesions. We hypothesized that an underlying cause is inefficient adenoviral delivery. Using the LNCaP progression model of prostate cancer we show that surface CAR expression decreases with increasing tumorigenicity and that castration resistant C4-2b cells were more difficult to transduce with adenovirus than castration sensitive LNCaP cells. Many genes, including CAR, are epigenetically silenced during transformation but a new class of chemotherapeutic agents, known as histone deacetylase inhibitors (HDACi), can reverse this process. We demonstrate that HDACi restore CAR expression and infectivity in C4-2b cells and enhance caspase activation in response to infection with a TRAIL adenovirus. We also show that in cells with high surface CAR expression, HDACi further enhance transgene expression from the CMV promoter. Thus HDACi have multiple beneficial effects, which may enhance not only viral but also non-viral gene therapy of castration resistant prostate cancer.
Collapse
|
47
|
Nioka S, Chen Y. Optical tecnology developments in biomedicine: history, current and future. Transl Med UniSa 2011; 1:51-150. [PMID: 23905030 PMCID: PMC3728850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Biomedical optics is a rapidly emerging field for medical imaging and diagnostics. This paper reviews several biomedical optical technologies that have been developed and translated for either clinical or pre-clinical applications. Specifically, we focus on the following technologies: 1) near-infrared spectroscopy and tomography, 2) optical coherence tomography, 3) fluorescence spectroscopy and imaging, and 4) optical molecular imaging. There representative biomedical applications are also discussed here.
Collapse
Affiliation(s)
- Shoko Nioka
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA -
| | - Yu Chen
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742 USA -
| |
Collapse
|
48
|
Keereweer S, Kerrebijn JDF, Mol IM, Mieog JSD, Van Driel PBAA, Baatenburg de Jong RJ, Vahrmeijer AL, Löwik CWGM. Optical imaging of oral squamous cell carcinoma and cervical lymph node metastasis. Head Neck 2011; 34:1002-8. [PMID: 21987435 DOI: 10.1002/hed.21861] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2011] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND In oral cancer surgery, intraoperative optical imaging could help the surgeon to determine adequate tumor-free margins. METHODS Tumor-specific near-infrared fluorescence agents targeting epidermal growth factor receptor (CW800 EGF) or glucose transporter system (CW800 2-DG) were administered to mice with tongue carcinoma and cervical lymph node metastases. Tumor growth was followed by bioluminescence imaging. Fluorescence signals were compared with a control group of healthy animals. RESULTS Significantly higher fluorescence was found in tongue tumors and cervical lymph node metastases compared with that in control animals. Fluorescence correlated with histopathology. Tumor-to-background ratio of CW800 EGF in the tongue was 13.8 (SD = 6.1) and in the lymph nodes 15.7 (SD = 8.8). For CW800 2-DG, the tumor-to-background ratio in the tongue was 4.6 (SD = 2.1) and in the lymph nodes 33.9 (SD = 18.4). CONCLUSIONS Optical imaging can be used to detect oral cancer and cervical lymph node metastases and could potentially improve complete surgical resection by real-time image-guided surgery.
Collapse
Affiliation(s)
- Stijn Keereweer
- Erasmus Medical Center, Department of Otorhinolaryngology Head and Neck Surgery, Rotterdam, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Kovar JL, Xu X, Draney D, Cupp A, Simpson MA, Michael Olive D. Near-infrared-labeled tetracycline derivative is an effective marker of bone deposition in mice. Anal Biochem 2011; 416:167-73. [DOI: 10.1016/j.ab.2011.05.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2011] [Revised: 03/30/2011] [Accepted: 05/06/2011] [Indexed: 12/31/2022]
|
50
|
Linder KE, Metcalfe E, Nanjappan P, Arunachalam T, Ramos K, Skedzielewski TM, Marinelli ER, Tweedle MF, Nunn AD, Swenson RE. Synthesis, in vitro evaluation, and in vivo metabolism of fluor/quencher compounds containing IRDye 800CW and Black Hole Quencher-3 (BHQ-3). Bioconjug Chem 2011; 22:1287-97. [PMID: 21639144 DOI: 10.1021/bc100457s] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Protease-cleavable peptides containing a suitable fluor/quencher (Fl/Q) pair are optically dark until cleaved by their target protease, generating fluorescence. This approach has been used with many Fl/Q pairs, but little has been reported with IRDye 800CW, a popular near-infrared (NIR) fluor. We explored the use of the azo-bond-containing Black Hole Quencher 3 (BHQ-3) as a quencher for IRDye 800CW and found that IRDye 800CW/BHQ-3 is a suitable Fl/Q pair, despite the lack of proper spectral overlap for fluorescence resonance energy transfer (FRET) applications. Cleavage of IRDye 800CW-PLGLK(BHQ-3)AR-NH(2) (8) and its D-arginine (Darg) analogue (9) by matrix metalloproteinases (MMPs) in vitro yielded the expected cleavage fragments. In vivo, extensive metabolism was found. Significant decomposition of a "non-cleavable" control IRDye 800CW-(1,13-diamino-4,7,10-trioxatridecane)-BHQ-3 (10) was evident in plasma of normal mice by 3 min post injection. The major metabolite showed a m/z and UV/vis spectrum consistent with azo bond cleavage in the BHQ-3 moiety. Preparation of an authentic standard of this metabolite (11) confirmed the assignment. Although the IRDye 800CW/BHQ-3 constructs showed efficient contact quenching prior to enzymatic cleavage, BHQ-3 should be used with caution in vivo, due to instability of its azo bond.
Collapse
Affiliation(s)
- Karen E Linder
- Ernst Felder Laboratories, Bracco Research USA, 305 College Road East, Princeton, New Jersey 08540, United States.
| | | | | | | | | | | | | | | | | | | |
Collapse
|