651
|
Tian J, Ding L, Wang Q, Hu Y, Jia L, Yu JS, Ju H. Folate Receptor-Targeted and Cathepsin B-Activatable Nanoprobe for In Situ Therapeutic Monitoring of Photosensitive Cell Death. Anal Chem 2015; 87:3841-8. [DOI: 10.1021/acs.analchem.5b00429] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Jiangwei Tian
- State
Key Laboratory of Analytical Chemistry for Life Science, School of
Chemistry and Chemical Engineering, Nanjing University, Nanjing, Jiangsu 210093, P.R. China
- Jiangsu
Key Laboratory of TCM Evaluation and Translational Research, Department
of Complex Prescription of TCM, China Pharmaceutical University, Nanjing, Jiangsu 211198, P. R. China
| | - Lin Ding
- State
Key Laboratory of Analytical Chemistry for Life Science, School of
Chemistry and Chemical Engineering, Nanjing University, Nanjing, Jiangsu 210093, P.R. China
| | - Quanbo Wang
- State
Key Laboratory of Analytical Chemistry for Life Science, School of
Chemistry and Chemical Engineering, Nanjing University, Nanjing, Jiangsu 210093, P.R. China
| | - Yaoping Hu
- State
Key Laboratory of Analytical Chemistry for Life Science, School of
Chemistry and Chemical Engineering, Nanjing University, Nanjing, Jiangsu 210093, P.R. China
| | - Li Jia
- State
Key Laboratory of Analytical Chemistry for Life Science, School of
Chemistry and Chemical Engineering, Nanjing University, Nanjing, Jiangsu 210093, P.R. China
| | - Jun-Sheng Yu
- State
Key Laboratory of Analytical Chemistry for Life Science, School of
Chemistry and Chemical Engineering, Nanjing University, Nanjing, Jiangsu 210093, P.R. China
| | - Huangxian Ju
- State
Key Laboratory of Analytical Chemistry for Life Science, School of
Chemistry and Chemical Engineering, Nanjing University, Nanjing, Jiangsu 210093, P.R. China
| |
Collapse
|
652
|
Maawy AA, Hiroshima Y, Zhang Y, Garcia-Guzman M, Luiken GA, Kobayashi H, Hoffman RM, Bouvet M. Photoimmunotherapy lowers recurrence after pancreatic cancer surgery in orthotopic nude mouse models. J Surg Res 2015; 197:5-11. [PMID: 25799527 DOI: 10.1016/j.jss.2015.02.037] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 01/26/2015] [Accepted: 02/13/2015] [Indexed: 01/06/2023]
Abstract
BACKGROUND Photoimmunotherapy (PIT) is based on the use of a monoclonal antibody specific to cancer epitopes conjugated to a photosensitizer near-infrared phthalocyanine dye (IR700). In this study, PIT with IR700 conjugated to anti-carcinoembryonic antigen (CEA) was used as an adjunct to surgery in orthotopically-implanted human pancreatic cancer in a nude mouse model to eliminate microscopic disease in the post-surgical tumor bed and prevent local as well as metastatic recurrence. MATERIALS AND METHODS Athymic nude mice were orthotopically implanted with the human pancreatic cancer cell line BxPC3 expressing green fluorescent protein. After tumor engraftment, the mice were divided into two groups as follows: bright light surgery (BLS) + anti-CEA-IR700 + 690 nm laser (PIT); and BLS only. Anti-CEA-IR700 (100 μg) was administered to the treatment group via tail-vein injection 24 h before therapy. Tumors were resected, and the surgical bed was treated with intraoperative phototherapy at an intensity of 150 mW/cm(2) for 30 min. Mice were imaged noninvasively for 8 wk using an OV-100 small animal fluorescence imager. RESULTS BLS + PIT reduced local recurrence to 1/7 mice from 7/7 mice with BLS-only (P = 0.001) and metastatic recurrence to 2/7 mice compared with 6/7 mice with BLS-only (P = 0.03). Local tumor growth continued at a rapid rate after BLS-only compared with BLS + PIT where almost no local growth occurred. There was a significant difference in tumor size between mice in the BLS + PIT (2.14 mm(2), 95% confidence interval [CI] [-2.06 to 6.34] and BLS-only groups (115.2 mm(2), 95% CI [88.8-141.6]) at 6 wk after surgery (P < 0.001). There was also a significant difference in tumor weight between the BLS + PIT group (6.65 mg, 95% CI [-6.35 to 19.65] and BLS-only group (1100 mg, 95% CI [794-1406] at 8 wk after surgery (P < 0.001). CONCLUSIONS PIT holds promise in the treatment of pancreatic cancer and may serve as a useful adjunct to surgery in the eradication of microscopic residual disease that can lead to both local and metastatic recurrence. Further studies are warranted to investigate the potential toxicities of PIT, especially with regard to anastomoses, such as those involved in pancreaticoduodenectomy.
Collapse
Affiliation(s)
- Ali A Maawy
- Department of Surgery, University of California San Diego, San Diego, California
| | - Yukihiko Hiroshima
- Department of Surgery, University of California San Diego, San Diego, California; AntiCancer, Inc, San Diego, California; Department of Surgery, Yokohama City University, Yokohama City, Japan
| | | | | | | | | | - Robert M Hoffman
- Department of Surgery, University of California San Diego, San Diego, California; AntiCancer, Inc, San Diego, California
| | - Michael Bouvet
- Department of Surgery, University of California San Diego, San Diego, California; Department of Surgery, VA Healthcare System, San Diego, California.
| |
Collapse
|
653
|
InCVAX--a novel strategy for treatment of late-stage, metastatic cancers through photoimmunotherapy induced tumor-specific immunity. Cancer Lett 2015; 359:169-77. [PMID: 25633839 DOI: 10.1016/j.canlet.2015.01.029] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 01/20/2015] [Accepted: 01/21/2015] [Indexed: 12/31/2022]
Abstract
A novel, promising potential cancer vaccine strategy was proposed to use a two-injection procedure for solid tumors to prompt the immune system to identify and systemically eliminate primary and metastatic cancers. The two-injection procedure consists of local photothermal application on a selected tumor intended to liberate whole cell tumor antigens, followed by a local injection of an immunoadjuvant that consists of a semi-synthetic functionalized glucosamine polymer, N-dihydro-galacto-chitosan (GC), which is intended to activate antigen presenting cells and facilitate an increased uptake of tumor antigens. This strategy is thus proposed as an in situ autologous cancer vaccine (inCVAX) that may activate antigen presenting cells and expose them to tumor antigens in situ, with the intention of inducing a systemic tumor specific T-cell response. Here, the development of inCVAX for the treatment of metastatic cancers in the past decades is systematically reviewed. The antitumor immune responses of local photothermal treatment and immunological stimulation with GC are also discussed. This treatment approach is also commonly referred to as laser immunotherapy (LIT).
Collapse
|
654
|
Viral transduction of the HER2-extracellular domain expands trastuzumab-based photoimmunotherapy for HER2-negative breast cancer cells. Breast Cancer Res Treat 2015; 149:597-605. [PMID: 25616354 DOI: 10.1007/s10549-015-3265-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 12/26/2014] [Indexed: 10/23/2022]
Abstract
The prognosis of HER2-positive breast cancer has been improved by trastuzumab therapy, which features high specificity and limited side effects. However, trastuzumab-based therapy has shortcomings. Firstly, HER2-targeted therapy is only applicable to HER2-expressing tumors, which comprise only 20-25% of primary breast cancers. Secondly, many patients who initially respond to trastuzumab ultimately develop disease progression. To overcome these problems, we employed virus-mediated HER2 transduction and photoimmunotherapy (PIT) which involves trastuzumab conjugated with a photosensitizer, trastuzumab-IR700, and irradiation of near-infrared light. We hypothesized that the gene transduction technique together with PIT would expand the range of tumor entities suitable for trastuzumab-based therapy and improve its antitumor activity. The HER2-extracellular domain (ECD) was transduced by the adenoviral vector, Ad-HER2-ECD, and PIT with trastuzumab-IR700 was applied in the HER2-negative cancer cells. Ad-HER2-ECD can efficiently transduce HER2-ECD into HER2-negative human cancer cells. PIT with trastuzumab-IR700 induced direct cell membrane destruction of Ad-HER2-ECD-transduced HER2-negative cancer cells. Novel combination of viral transduction of a target antigen and an antibody-based PIT would expand and potentiate molecular-targeted therapy even for target-negative or attenuated cancer cells.
Collapse
|
655
|
Chang HY, Li MH, Huang TC, Hsu CL, Tsai SR, Lee SC, Huang HC, Juan HF. Quantitative proteomics reveals middle infrared radiation-interfered networks in breast cancer cells. J Proteome Res 2015; 14:1250-62. [PMID: 25556991 DOI: 10.1021/pr5011873] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Breast cancer is one of the leading cancer-related causes of death worldwide. Treatment of triple-negative breast cancer (TNBC) is complex and challenging, especially when metastasis has developed. In this study, we applied infrared radiation as an alternative approach for the treatment of TNBC. We used middle infrared (MIR) with a wavelength range of 3-5 μm to irradiate breast cancer cells. MIR significantly inhibited cell proliferation in several breast cancer cells but did not affect the growth of normal breast epithelial cells. We performed iTRAQ-coupled LC-MS/MS analysis to investigate the MIR-triggered molecular mechanisms in breast cancer cells. A total of 1749 proteins were identified, quantified, and subjected to functional enrichment analysis. From the constructed functionally enriched network, we confirmed that MIR caused G2/M cell cycle arrest, remodeled the microtubule network to an astral pole arrangement, altered the actin filament formation and focal adhesion molecule localization, and reduced cell migration activity and invasion ability. Our results reveal the coordinative effects of MIR-regulated physiological responses in concentrated networks, demonstrating the potential implementation of infrared radiation in breast cancer therapy.
Collapse
Affiliation(s)
- Hsin-Yi Chang
- Department of Life Science, National Taiwan University , Taipei 10617, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
656
|
Bumah VV, Masson-Meyers DS, Cashin S, Enwemeka CS. Optimization of the antimicrobial effect of blue light on methicillin-resistant Staphylococcus aureus (MRSA) in vitro. Lasers Surg Med 2015; 47:266-72. [PMID: 25639752 DOI: 10.1002/lsm.22327] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2014] [Indexed: 01/02/2023]
Abstract
BACKGROUND AND OBJECTIVE In previous studies, we showed that irradiation with 405 nm or 470 nm light suppresses up to 92% methicillin-resistant Staphylococcus aureus (MRSA) growth in vitro and that the remaining bacteria re-colonize. In this study, the aim was to develop a protocol that yields 100% MRSA growth suppression. MATERIALS AND METHODS We cultured 3 × 10(6) and 5 × 10(6) CFU/ml USA300 strain of MRSA and then irradiated each plate with varying fluences of 1-60 J/cm2 of 405 nm or 470 nm light, either once or twice at 6 hours intervals. Next, we plated 7 × 10(6) CFU/ml and irradiated it with 45, 50, 55, or 60 J/cm2 fluence, once, twice, or thrice at the same 6 hours intervals. In a third experiment, the same culture density was irradiated with 0, 165, 180, 220, or 240 J/cm(2) , either once, twice, or thrice. RESULTS Irradiation with either wavelength significantly reduced the bacterial colonies regardless of bacterial density (P < 0.05). At 3 × 10(6) CFU/ml density, nearly 40% and 50% growth of MRSA were suppressed with as little as 3 J/cm2 of 405 nm and 470 nm wavelengths, respectively. Moreover, 100% of the colonies were suppressed with a single exposure to 55 or 60 J/cm2 of 470 nm light or double treatment with 50, 55, or 60 J/cm2 of 405 nm wavelength. At 5 × 10(6) CFU/ml density, irradiating twice with 50, 55, or 60 J/cm2 of either wavelength suppressed bacterial growth completely, lower fluences did not. The denser 7 × 10(6) CFU/ml culture required higher doses to achieve 100% suppression, either one shot with 220 J/cm2 of 470 nm light or two shots of the same dose using 405 nm. CONCLUSION The bactericidal effect of blue light can be optimized to yield 100% bacterial growth suppression, but with relatively high fluences for dense bacterial cultures, such as 7 × 10(6) CFU/ml.
Collapse
Affiliation(s)
- Violet V Bumah
- College of Health Sciences, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, 53211
| | | | | | | |
Collapse
|
657
|
Sato K, Nakajima T, Choyke PL, Kobayashi H. Selective cell elimination in vitro and in vivo from tissues and tumors using antibodies conjugated with a near infrared phthalocyanine. RSC Adv 2015; 5:25105-25114. [PMID: 25866624 DOI: 10.1039/c4ra13835j] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Cell cultures and tissues often contain cellular subpopulations that potentially interfere with or contaminate other cells of interest. However, it is difficult to eliminate unwanted cells without damaging the very cell population one is seeking to protect, especially established tissue. Here, we report a method of eliminating a specific subpopulation of cells from a mixed 2D or 3D cell culture in vitro and a mixed-population in vivo tumor model by using antibody-photosensitizer conjugates (APC) with a near infrared (NIR) phthalocyanine-derivative (IRdye700DX, IR700) combined with NIR light exposure with minimal damage to non-targeted cells. Thus, APC combined with NIR light exposure holds promise as a method of removing specific cells from mixed cell cultures and tumors.
Collapse
Affiliation(s)
- Kazuhide Sato
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute
| | - Takahito Nakajima
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute
| | - Peter L Choyke
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute
| | - Hisataka Kobayashi
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute
| |
Collapse
|
658
|
Planas O, Gallavardin T, Nonell S. A novel fluoro-chromogenic click reaction for the labelling of proteins and nanoparticles with near-IR theranostic agents. Chem Commun (Camb) 2015; 51:5586-9. [DOI: 10.1039/c4cc09070e] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Binding of red-absorbing porphycene isothiocyanates to proteins and nanoparticles leads to near-IR fluorescent and photosensitising conjugates.
Collapse
Affiliation(s)
- Oriol Planas
- Institut Químic de Sarrià
- Universitat Ramon Llull
- Barcelona
- Spain
| | | | - Santi Nonell
- Institut Químic de Sarrià
- Universitat Ramon Llull
- Barcelona
- Spain
| |
Collapse
|
659
|
Watanabe R, Hanaoka H, Sato K, Nagaya T, Harada T, Mitsunaga M, Kim I, Paik CH, Wu AM, Choyke PL, Kobayashi H. Photoimmunotherapy targeting prostate-specific membrane antigen: are antibody fragments as effective as antibodies? J Nucl Med 2015; 56:140-4. [PMID: 25500827 PMCID: PMC6484861 DOI: 10.2967/jnumed.114.149526] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED Photoimmunotherapy is a highly cell-selective cancer therapy based on an armed antibody conjugate with a phthalocyanine-based photosensitizer, IR700. Photoimmunotherapy induces rapid and highly specific necrosis in targeted cancer cells after exposure to near-infrared (NIR) light. Cells not expressing the antigen are not affected. To date, photoimmunotherapy has been demonstrated only with full antibody-IR700 conjugates. In this study, small and bivalent antibody fragments, including anti-prostate-specific membrane antigen (PSMA) diabody (Db) and minibody (Mb), were compared with intact IgG for their effectiveness as photoimmunotherapy agents. METHODS Radioiodinated antibody and antibody fragments with (125)I were used to determine the timing of maximum binding of each anti-PSMA antibody fragment on the cell surface in vivo in mice bearing either PSMA-positive or -negative PC3 tumors. Then therapeutic efficacy of photoimmunotherapy was examined by exposing mice to NIR light at 2 time points based on the time of maximum cell surface binding at 6 h after injection for Db-IR700 and 24 h after injection for Mb-IR700 and IgG-IR700 as well as 24 h after the peak uptake times. RESULTS Photoimmunotherapy with the same molar concentration of PSMA-Db-IR700, PSMA-Mb-IR700, and PSMA-IgG-IR700 conjugate showed similar therapeutic effects in vitro and in vivo on PSMA-positive PC3 tumor xenografts in cytotoxicity and survival curves (P > 0.05). CONCLUSION The use of PSMA-Db-IR700 conjugate results in the shortest time interval between injection and NIR exposure without compromising therapeutic effects of photoimmunotherapy.
Collapse
Affiliation(s)
- Rira Watanabe
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Hirofumi Hanaoka
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Kazuhide Sato
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Tadanobu Nagaya
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Toshiko Harada
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Makoto Mitsunaga
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Insook Kim
- Applied/Developmental Research Directorate, Leidos Biomedical Research Inc., Frederick National Laboratory, Frederick, Maryland
| | - Chang H Paik
- Nuclear Medicine Department, Radiology and Imaging Science, Warren Grant Magnuson Clinical Center, National Institutes of Health, Bethesda, Maryland; and
| | - Anna M Wu
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Peter L Choyke
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Hisataka Kobayashi
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
660
|
Hanaoka H, Nakajima T, Sato K, Watanabe R, Phung Y, Gao W, Harada T, Kim I, Paik CH, Choyke PL, Ho M, Kobayashi H. Photoimmunotherapy of hepatocellular carcinoma-targeting Glypican-3 combined with nanosized albumin-bound paclitaxel. Nanomedicine (Lond) 2015; 10:1139-47. [PMID: 25929570 PMCID: PMC4420156 DOI: 10.2217/nnm.14.194] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
AIM Effectiveness of Glypican-3 (GPC3)-targeted photoimmunotherapy (PIT) combined with the nanoparticle albumin-bound paclitaxel (nab-paclitaxel) for hepatocellular carcinoma was evaluated. MATERIALS & METHODS GPC3 expressing A431/G1 cells were incubated with a phthalocyanine-derivative, IRDye700DX (IR700), conjugated to an anti-GPC3 antibody, IR700-YP7 and exposed to near-infrared light. Therapeutic experiments combining GPC3-targeted PIT with nab-paclitaxel were performed in A431/G1 tumor-bearing mice. RESULTS IR700-YP7 bound to A431/G1 cells and induced rapid target-specific necrotic cell death by near-infrared light exposure in vitro. IR700-YP7 accumulated in A431/G1 tumors. Tumor growth was inhibited by PIT compared with nontreated control. Additionally, PIT dramatically increased nab-paclitaxel delivery and enhanced the therapeutic effect. CONCLUSION PIT targeting GPC3 combined with nab-paclitaxel is a promising method for treating hepatocellular carcinoma.
Collapse
Affiliation(s)
- Hirofumi Hanaoka
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, NIH, Building 10, Room B3B69, MSC1088, Bethesda, MD 20892, USA
| | - Takahito Nakajima
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, NIH, Building 10, Room B3B69, MSC1088, Bethesda, MD 20892, USA
| | - Kazuhide Sato
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, NIH, Building 10, Room B3B69, MSC1088, Bethesda, MD 20892, USA
| | - Rira Watanabe
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, NIH, Building 10, Room B3B69, MSC1088, Bethesda, MD 20892, USA
| | - Yen Phung
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Wei Gao
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Toshiko Harada
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, NIH, Building 10, Room B3B69, MSC1088, Bethesda, MD 20892, USA
| | - Insook Kim
- Applied/Developmental Research Directorate, Leidos Biomedical Research Inc, Frederick National Laboratory, Frederick, MD 21702, USA
| | - Chang H Paik
- Nuclear Medicine Department, Warren Grant Magnuson Clinical Center, Radiology & Imaging Science, Warren Grant Magnuson Clinical Center, NIH, Bethesda, MD 20892, USA
| | - Peter L Choyke
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, NIH, Building 10, Room B3B69, MSC1088, Bethesda, MD 20892, USA
| | - Mitchell Ho
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Hisataka Kobayashi
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, NIH, Building 10, Room B3B69, MSC1088, Bethesda, MD 20892, USA
| |
Collapse
|
661
|
Yuan A, Hu Y, Ming X. Dendrimer Conjugates for Light-activated Delivery of Antisense Oligonucleotides. RSC Adv 2015; 5:35195-35200. [PMID: 26146545 DOI: 10.1039/c5ra04091d] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Therapeutic oligonucleotides, such as splice switching ONs (SSOs), provide opportunities for treating serious, life-threatening diseases. However, the development of ONs as therapeutic agents has progressed slowly, because difficult cytosolic delivery of SSOs into the cytosol and nucleus remains a major barrier. Photochemical internalization (PCI), a promising strategy for endosomal escape, was introduced to disrupt the endosomal membrane using light and a photosensitizer. Here we constructed Poly(amido amine) (PAMAM) dendrimer conjugates to simultaneously deliver SSOs and photosensitizers into endo/lysosomal compartments. After photo-irradiation, considerable ONs were observed to diffuse into the cytosol and accumulate in the nucleus. Furthermore, the PCI mediated cytosolic delivery of SSOs effectively enhanced their nuclear splice switching activity.
Collapse
Affiliation(s)
- Ahu Yuan
- Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA ; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210093, China
| | - Yiqiao Hu
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210093, China
| | - Xin Ming
- Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
662
|
Lamb BM, Barbas III CF. Selective arylthiolane deprotection by singlet oxygen: a promising tool for sensors and prodrugs. Chem Commun (Camb) 2015; 51:3196-9. [DOI: 10.1039/c4cc09040c] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A routine thioketal protecting group reacts rapidly and selectively with singlet oxygen to reveal ketone products in good (aryl 1,3-dithiolane) to excellent (aryl 1,3-oxathiolane) yields. Arylthiolanes are stable to biologically relevant reactive oxygen species and can be used as a light-activated gating mechanism for activating fluorescent sensors or small molecule prodrugs.
Collapse
Affiliation(s)
- Brian M. Lamb
- The Skaggs Institute for Chemical Biology
- The Scripps Research Institute
- La Jolla
- USA
- Department of Chemistry
| | - Carlos F. Barbas III
- The Skaggs Institute for Chemical Biology
- The Scripps Research Institute
- La Jolla
- USA
- Department of Chemistry
| |
Collapse
|
663
|
Hou B, Zheng B, Gong X, Wang H, Wang S, Liao Z, Li X, Zhang X, Chang J. A UCN@mSiO2@cross-linked lipid with high steric stability as a NIR remote controlled-release nanocarrier for photodynamic therapy. J Mater Chem B 2015; 3:3531-3540. [DOI: 10.1039/c5tb00240k] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In clinics, the application of photodynamic therapy (PDT) in deep tissue is severely constrained by the limited penetration depth of visible light that was used for activating the photosensitizer (PS).
Collapse
Affiliation(s)
- Beibei Hou
- School of Life Sciences
- Tianjin University
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin)
- Tianjin 300072
- P. R. China
| | - Bin Zheng
- School of Life Sciences
- Tianjin University
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin)
- Tianjin 300072
- P. R. China
| | - Xiaoqun Gong
- School of Life Sciences
- Tianjin University
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin)
- Tianjin 300072
- P. R. China
| | - Hanjie Wang
- School of Life Sciences
- Tianjin University
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin)
- Tianjin 300072
- P. R. China
| | - Sheng Wang
- School of Life Sciences
- Tianjin University
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin)
- Tianjin 300072
- P. R. China
| | - Zhenyu Liao
- The National Center of Supervision and Inspection for Quality of Food
- Tianjin Product Quality Inspection Technology Research Institute
- Tianjin 300384
- People's Republic of China
| | - Xiaodong Li
- The Second Hospital of Tianjin Medical University
- Tianjin, 300211
- China
| | - Xuening Zhang
- The Second Hospital of Tianjin Medical University
- Tianjin, 300211
- China
| | - Jin Chang
- School of Life Sciences
- Tianjin University
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin)
- Tianjin 300072
- P. R. China
| |
Collapse
|
664
|
Bian Y, Jiang J. Recent Advances in Phthalocyanine-Based Functional Molecular Materials. STRUCTURE AND BONDING 2015. [DOI: 10.1007/430_2015_194] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
665
|
Tekdaş DA, Gürek AG, Ahsen V. Asymmetric zinc phthalocyanines substituted with a single carboxyl and triethyleneoxysulfonyl groups: synthesis, characterization and validation for photodynamic therapy. J PORPHYR PHTHALOCYA 2014. [DOI: 10.1142/s1088424614500709] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
This work describes modular pathway towards the synthesis of new specific unsymmetrically (AB3) zinc phthalocyanines that contains three polyoxy ethylene and monocarboxylic acid groups as photosensitizers. Their photophysical and photochemical properties are studied. General trends are described for quantum yields of fluorescence, photodegradation and singlet oxygen quantum yields of these compounds. Therefore, these novel phthalocyanines could potentially be good photosensitizers for photodynamic therapy.
Collapse
Affiliation(s)
- Duygu Aydın Tekdaş
- Gebze Institute of Technology, Department of Chemistry, P.O. Box 141, Gebze, Kocaeli 41400, Turkey
| | - Ayşe G. Gürek
- Gebze Institute of Technology, Department of Chemistry, P.O. Box 141, Gebze, Kocaeli 41400, Turkey
| | - Vefa Ahsen
- Gebze Institute of Technology, Department of Chemistry, P.O. Box 141, Gebze, Kocaeli 41400, Turkey
| |
Collapse
|
666
|
Ali T, Nakajima T, Sano K, Sato K, Choyke PL, Kobayashi H. Dynamic fluorescent imaging with indocyanine green for monitoring the therapeutic effects of photoimmunotherapy. CONTRAST MEDIA & MOLECULAR IMAGING 2014; 9:276-82. [PMID: 24706611 DOI: 10.1002/cmmi.1570] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Revised: 08/13/2013] [Accepted: 08/26/2013] [Indexed: 01/01/2023]
Abstract
A new type of monoclonal antibody (mAb)-based, highly specific phototherapy (photoimmunotherapy; PIT) that uses a near-infrared (NIR) phthalocyanine dye, IRDye700DX (IR700) conjugated with an mAb, has recently been described. NIR light exposure leads to immediate, target-selective necrotic cell death. However, tumor shrinkage takes several days to occur, making it difficult to detect earlier changes in the tumor. In this study, Panitumumab targeting the epidermal growth factor receptor (EGFR1) conjugated to IR700 was used to treat EGFR-expressing A431 tumor cells and in vivo xenografts. PIT was performed at varying doses of NIR light (10, 30, 50 and 100 J cm(-2)) in xenograft tumors in mice. Indocyanine green (ICG) dynamic imaging was evaluated for monitoring cytotoxic effects for the first hour after PIT. Our results demonstrated a statistical difference (p < 0.05) in ICG intensity between control and PIT treated tumors in the higher light exposure groups (50 J cm(-2): 2.94 ± 0.35 vs 5.22 ± 0.92, p = 0.02; and 100 J cm(-2) : 3.56 ± 0.96 vs 5.71 ± 1.43, p = 0.008) as early as 20 min post ICG injection. However, no significant difference (p > 0.05) in ICG intensity between control and PIT treated tumors was evident in the lower light exposure group at any time points up to 60 min (10 J cm(-2) : 1.92 ± 0.49 vs 1.71 ± 0.3, p = 0.44; and 30 J cm(-2): 1.57 ± 0.35 vs 2.75 ± 0.59, p = 0.07). Similarly, the retention index (background to corrected uptake ratio of ICG) varied with light exposure. In conclusion, ICG may serve as a potential indicator of acute cytotoxic effects of mAb-IR700-induced PIT even before morphological changes can be seen in targeted tumors.
Collapse
Affiliation(s)
- Towhid Ali
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, 20892-1088, USA
| | | | | | | | | | | |
Collapse
|
667
|
Phthalocyanine-based photosensitizers: more efficient photodynamic therapy? Future Med Chem 2014; 6:1991-3. [DOI: 10.4155/fmc.14.139] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
668
|
Sato K, Hanaoka H, Watanabe R, Nakajima T, Choyke PL, Kobayashi H. Near infrared photoimmunotherapy in the treatment of disseminated peritoneal ovarian cancer. Mol Cancer Ther 2014; 14:141-50. [PMID: 25416790 DOI: 10.1158/1535-7163.mct-14-0658] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Near infrared photoimmunotherapy (NIR-PIT) is a new cancer treatment that combines the specificity of intravenously injected antibodies for targeting tumors with the toxicity induced by photosensitizers after exposure to near infrared (NIR) light. Herein, we evaluate the efficacy of NIR-PIT in a mouse model of disseminated peritoneal ovarian cancer. In vitro and in vivo experiments were conducted with a HER2-expressing, luciferase-expressing, ovarian cancer cell line (SKOV-luc). An antibody-photosensitizer conjugate (APC) consisting of trastuzumab and a phthalocyanine dye, IRDye-700DX, was synthesized (tra-IR700) and cells or tumors were exposed to NIR light. In vitro PIT cytotoxicity was assessed with dead staining and luciferase activity in freely growing cells and in a three-dimensional (3D) spheroid model. In vivo NIR-PIT was performed in mice with tumors implanted in the peritoneum and in the flank and these were assessed by tumor volume and/or bioluminescence. In vitro NIR-PIT-induced cytotoxicity was light dose dependent. Repeated light exposures induced complete tumor cell killing in the 3D spheroid model. In vivo the antitumor effects of NIR-PIT were confirmed by significant reductions in both tumor volume and luciferase activity in the flank model (NIR-PIT vs. control in tumor volume changes at day 10, P = 0.0001; NIR-PIT vs. control in luciferase activity at day 4, P = 0.0237), and the peritoneal model (NIR-PIT vs. control in luciferase activity at day 7, P = 0.0037). NIR-PIT provided effective cell killing in this HER2-positive model of disseminated peritoneal ovarian cancer. Thus, NIR-PIT is a promising new therapy for the treatment of disseminated peritoneal tumors.
Collapse
Affiliation(s)
- Kazuhide Sato
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute/NIH, Bethesda, Maryland
| | - Hirofumi Hanaoka
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute/NIH, Bethesda, Maryland
| | - Rira Watanabe
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute/NIH, Bethesda, Maryland
| | - Takahito Nakajima
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute/NIH, Bethesda, Maryland
| | - Peter L Choyke
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute/NIH, Bethesda, Maryland
| | - Hisataka Kobayashi
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute/NIH, Bethesda, Maryland.
| |
Collapse
|
669
|
Kobayashi H, Turkbey B, Watanabe R, Choyke PL. Cancer drug delivery: considerations in the rational design of nanosized bioconjugates. Bioconjug Chem 2014; 25:2093-100. [PMID: 25385142 PMCID: PMC4275162 DOI: 10.1021/bc500481x] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
![]()
In
order to efficiently deliver anticancer agents to tumors, biocompatible
nanoparticles or bioconjugates, including antibody–drug conjugates
(ADCs), have recently been designed, synthesized, and tested, some
even in clinical trials. Controlled delivery can be enhanced by changing
specific design characteristics of the bioconjugate such as its size,
the nature of the payload, and the surface features. The delivery
of macromolecular drugs to cancers largely relies on the leaky nature
of the tumor vasculature compared with healthy vessels in normal organs.
When administered intravenously, macromolecular bioconjugates and
nanosized agents tend to circulate for prolonged times, unless they
are small enough to be excreted by the kidney or stealthy enough to
evade the macrophage phagocytic system (MPS), formerly the reticulo-endothelial
system (RES). Therefore, macromolecular bioconjugates and nanosized
agents with long circulation times leak preferentially into tumor
tissue through permeable tumor vessels and are then retained in the
tumor bed due to reduced lymphatic drainage. This process is known
as the enhanced permeability and retention (EPR) effect. However,
success of cancer drug delivery only relying on the EPR effect is
still limited. To cure cancer patients, further improvement of drug
delivery is required by both designing superior agents and enhancing
EPR effects. In this Review, we describe the basis of macromolecular
or nanosized bioconjugate delivery into cancer tissue and discuss
current diagnostic methods for evaluating leakiness of the tumor vasculature.
Then, we discuss methods to augment conventional “permeability
and retention” effects for macromolecular or nanosized bioconjugates
in cancer tissue.
Collapse
Affiliation(s)
- Hisataka Kobayashi
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health , Bethesda, Maryland 20892, United States
| | | | | | | |
Collapse
|
670
|
Photoimmunotherapy of gastric cancer peritoneal carcinomatosis in a mouse model. PLoS One 2014; 9:e113276. [PMID: 25401794 PMCID: PMC4234664 DOI: 10.1371/journal.pone.0113276] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 10/21/2014] [Indexed: 12/28/2022] Open
Abstract
Photoimmunotherapy (PIT) is a new cancer treatment that combines the specificity of antibodies for targeting tumors with the toxicity induced by photosensitizers after exposure to near infrared (NIR) light. We performed PIT in a model of disseminated gastric cancer peritoneal carcinomatosis and monitored efficacy with in vivo GFP fluorescence imaging. In vitro and in vivo experiments were conducted with a HER2-expressing, GFP-expressing, gastric cancer cell line (N87-GFP). A conjugate comprised of a photosensitizer, IR-700, conjugated to trastuzumab (tra-IR700), followed by NIR light was used for PIT. In vitro PIT was evaluated by measuring cytotoxicity with dead staining and a decrease in GFP fluorescence. In vivo PIT was evaluated in a disseminated peritoneal carcinomatosis model and a flank xenograft using tumor volume measurements and GFP fluorescence intensity. In vivo anti-tumor effects of PIT were confirmed by significant reductions in tumor volume (at day 15, p<0.0001 vs. control) and GFP fluorescence intensity (flank model: at day 3, PIT treated vs. control p<0.01 and peritoneal disseminated model: at day 3 PIT treated vs. control, p<0.05). Cytotoxic effects in vitro were shown to be dependent on the light dose and caused necrotic cell rupture leading to GFP release and a decrease in fluorescence intensity in vitro. Thus, loss of GFP fluorescence served as a useful biomarker of cell necrosis after PIT.
Collapse
|
671
|
Zheng YW, Chen SF, Zheng BY, Ke MR, Huang JD. A Silicon(IV) Phthalocyanine–Folate Conjugate as an Efficient Photosensitizer. CHEM LETT 2014. [DOI: 10.1246/cl.140607] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
672
|
Miyako E, Russier J, Mauro M, Cebrian C, Yawo H, Ménard-Moyon C, Hutchison JA, Yudasaka M, Iijima S, De Cola L, Bianco A. Photofunctional Nanomodulators for Bioexcitation. Angew Chem Int Ed Engl 2014. [DOI: 10.1002/ange.201407169] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
673
|
Miyako E, Russier J, Mauro M, Cebrian C, Yawo H, Ménard-Moyon C, Hutchison JA, Yudasaka M, Iijima S, De Cola L, Bianco A. Photofunctional Nanomodulators for Bioexcitation. Angew Chem Int Ed Engl 2014; 53:13121-5. [DOI: 10.1002/anie.201407169] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 09/01/2014] [Indexed: 12/31/2022]
|
674
|
Zhang FL, Huang Q, Liu JY, Huang MD, Xue JP. Molecular-Target-Based Anticancer Photosensitizer: Synthesis and in vitro Photodynamic Activity of Erlotinib-Zinc(II) Phthalocyanine Conjugates. ChemMedChem 2014; 10:312-20. [DOI: 10.1002/cmdc.201402373] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Indexed: 11/09/2022]
|
675
|
de Boer E, Warram JM, Hartmans E, Bremer PJ, Bijl B, Crane LMA, Nagengast WB, Rosenthal EL, van Dam GM. A standardized light-emitting diode device for photoimmunotherapy. J Nucl Med 2014; 55:1893-8. [PMID: 25315245 DOI: 10.2967/jnumed.114.142299] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED Antibody-based photodynamic therapy-photoimmunotherapy (PIT)-is an ideal modality to improve cancer treatment because of its selective and tumor-specific mode of therapy. Because the use of PIT for cancer treatment is continuing to be described, there is great need to characterize a standardized light source for PIT application. In this work, we designed and manufactured a light-emitting diode (LED)/PIT device and validated the technical feasibility, applicability, safety, and consistency of the system for cancer treatment. METHODS To outline the characteristics and photobiologic safety of the LED device, multiple optical measurements were performed in accordance with a photobiologic safety standard. A luciferase-transfected breast cancer cell line (2LMP-Luc) in combination with panitumumab-IRDye 700DX (pan-IR700) was used to validate the in vitro and in vivo performance of our LED device. RESULTS Testing revealed the light source to be safe, easy to use, and independent of illumination and power output (mW cm(-2)) variations over time. For in vitro studies, an LED dose (2, 4, 6 J cm(-2))-dependent cytotoxicity was observed using propidium iodide exclusion and annexin V staining. Dose-dependent blebbing was also observed during microscopic analysis. Bioluminescence signals of tumors treated with 0.3 mg of pan-IR700 and 50 J cm(-2) decreased significantly (>80%) compared with signals of contralateral nontreated sites at 4 h and at 1 d after PIT. CONCLUSION To our knowledge, a normalized and standardized LED device has not been explicitly described or developed. In this article, we introduce a standardized light source and validate its usability for PIT applications.
Collapse
Affiliation(s)
- Esther de Boer
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama Department of Surgery, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jason M Warram
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - Elmire Hartmans
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Peter J Bremer
- Philips Consumer Lifestyle, Drachten, The Netherlands; and
| | - Ben Bijl
- SurgVision, Heerenveen, The Netherlands
| | - Lucia M A Crane
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - Wouter B Nagengast
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Eben L Rosenthal
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - Gooitzen M van Dam
- Department of Surgery, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
676
|
Kim J, Santos OA, Park JH. Selective photosensitizer delivery into plasma membrane for effective photodynamic therapy. J Control Release 2014; 191:98-104. [DOI: 10.1016/j.jconrel.2014.05.049] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 05/21/2014] [Accepted: 05/24/2014] [Indexed: 11/30/2022]
|
677
|
Horiuchi H, Kano T, Uehara H, Okutsu T. Sono-activatable Photosensitizer for Photodynamic Therapy. CHEM LETT 2014. [DOI: 10.1246/cl.140403] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Hiroaki Horiuchi
- Division of Molecular Science, Faculty of Science and Technology, Gunma University
- International Education and Research Center for Silicon Science, Faculty of Science and Technology, Gunma University
| | - Takayuki Kano
- Division of Molecular Science, Faculty of Science and Technology, Gunma University
| | - Hiroki Uehara
- Division of Molecular Science, Faculty of Science and Technology, Gunma University
| | - Tetsuo Okutsu
- Division of Molecular Science, Faculty of Science and Technology, Gunma University
| |
Collapse
|
678
|
Liang CP, Nakajima T, Watanabe R, Sato K, Choyke PL, Chen Y, Kobayashi H. Real-time monitoring of hemodynamic changes in tumor vessels during photoimmunotherapy using optical coherence tomography. JOURNAL OF BIOMEDICAL OPTICS 2014; 19:98004. [PMID: 25253195 PMCID: PMC4174533 DOI: 10.1117/1.jbo.19.9.098004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 08/11/2014] [Accepted: 08/29/2014] [Indexed: 05/30/2023]
Abstract
Photoimmunotherapy (PIT) is a cell-specific cancer therapy based on an armed antibody conjugate that induces rapid and highly selective cancer cell necrosis after exposure to near-infrared (NIR) light. The PIT treatment also induces the superenhanced permeability and retention effect, which allows high concentrations of nanoparticles to accumulate in the tumor bed. In our pilot studies, optical coherence tomography (OCT) reveals dramatic hemodynamic changes during PIT. We developed and applied speckle variance analysis, Doppler flow measurement, bulk motion removal, and automatic region of interest selection to quantify vessel diameter and blood velocity within tumors in vivo. OCT imaging reveals that blood velocity in peripheral tumor vessels quickly drops below the detection limit while the vessel lumen remains open (4 vessels from 3 animals). On the other hand, control tumor vessels (receive NIR illumination but no PIT drug) do not show the sustained blood velocity drop (5 vessels from 3 animals). Ultraslow blood velocity could result in a long drug circulation time in tumor. Increase of the blood pool volume within the central tumor (shown in histology) may be the leading cause of the periphery blood velocity drop and could also increase the drug pool volume in tumor vessels.
Collapse
Affiliation(s)
- Chia-Pin Liang
- University of Maryland, Fischell Department of Bioengineering, 2218 Jeong H. Kim Engineering Building, College Park, Maryland 20742, United States
| | - Takahito Nakajima
- National Institute of Health, National Cancer Institute, Molecular Imaging Program, Bldg 10, Room B3B47, Bethesda, Maryland 20892-1088, United States
| | - Rira Watanabe
- National Institute of Health, National Cancer Institute, Molecular Imaging Program, Bldg 10, Room B3B47, Bethesda, Maryland 20892-1088, United States
| | - Kazuhide Sato
- National Institute of Health, National Cancer Institute, Molecular Imaging Program, Bldg 10, Room B3B47, Bethesda, Maryland 20892-1088, United States
| | - Peter L. Choyke
- National Institute of Health, National Cancer Institute, Molecular Imaging Program, Bldg 10, Room B3B47, Bethesda, Maryland 20892-1088, United States
| | - Yu Chen
- University of Maryland, Fischell Department of Bioengineering, 2218 Jeong H. Kim Engineering Building, College Park, Maryland 20742, United States
| | - Hisataka Kobayashi
- National Institute of Health, National Cancer Institute, Molecular Imaging Program, Bldg 10, Room B3B47, Bethesda, Maryland 20892-1088, United States
| |
Collapse
|
679
|
Débarre D, Olivier N, Supatto W, Beaurepaire E. Mitigating phototoxicity during multiphoton microscopy of live Drosophila embryos in the 1.0-1.2 µm wavelength range. PLoS One 2014; 9:e104250. [PMID: 25111506 PMCID: PMC4128758 DOI: 10.1371/journal.pone.0104250] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Accepted: 07/09/2014] [Indexed: 11/18/2022] Open
Abstract
Light-induced toxicity is a fundamental bottleneck in microscopic imaging of live embryos. In this article, after a review of photodamage mechanisms in cells and tissues, we assess photo-perturbation under illumination conditions relevant for point-scanning multiphoton imaging of live Drosophila embryos. We use third-harmonic generation (THG) imaging of developmental processes in embryos excited by pulsed near-infrared light in the 1.0-1.2 µm range. We study the influence of imaging rate, wavelength, and pulse duration on the short-term and long-term perturbation of development and define criteria for safe imaging. We show that under illumination conditions typical for multiphoton imaging, photodamage in this system arises through 2- and/or 3-photon absorption processes and in a cumulative manner. Based on this analysis, we derive general guidelines for improving the signal-to-damage ratio in two-photon (2PEF/SHG) or THG imaging by adjusting the pulse duration and/or the imaging rate. Finally, we report label-free time-lapse 3D THG imaging of gastrulating Drosophila embryos with sampling appropriate for the visualisation of morphogenetic movements in wild-type and mutant embryos, and long-term multiharmonic (THG-SHG) imaging of development until hatching.
Collapse
Affiliation(s)
- Delphine Débarre
- Laboratory for Optics and Biosciences, Ecole Polytechnique, CNRS UMR 7645, and INSERM U696, Palaiseau, France
- Univ. Grenoble Alpes, LIPhy, Grenoble, France
- CNRS, LIPhy, Grenoble, France
| | - Nicolas Olivier
- Laboratory for Optics and Biosciences, Ecole Polytechnique, CNRS UMR 7645, and INSERM U696, Palaiseau, France
| | - Willy Supatto
- Laboratory for Optics and Biosciences, Ecole Polytechnique, CNRS UMR 7645, and INSERM U696, Palaiseau, France
| | - Emmanuel Beaurepaire
- Laboratory for Optics and Biosciences, Ecole Polytechnique, CNRS UMR 7645, and INSERM U696, Palaiseau, France
| |
Collapse
|
680
|
Tian J, Ding L, Ju H, Yang Y, Li X, Shen Z, Zhu Z, Yu JS, Yang CJ. A Multifunctional Nanomicelle for Real-Time Targeted Imaging and Precise Near-Infrared Cancer Therapy. Angew Chem Int Ed Engl 2014; 53:9544-9. [DOI: 10.1002/anie.201405490] [Citation(s) in RCA: 165] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Indexed: 12/19/2022]
|
681
|
Tian J, Ding L, Ju H, Yang Y, Li X, Shen Z, Zhu Z, Yu JS, Yang CJ. A Multifunctional Nanomicelle for Real-Time Targeted Imaging and Precise Near-Infrared Cancer Therapy. Angew Chem Int Ed Engl 2014. [DOI: 10.1002/ange.201405490] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
682
|
Advances in imaging probes and optical microendoscopic imaging techniques for early in vivo cancer assessment. Adv Drug Deliv Rev 2014; 74:53-74. [PMID: 24120351 DOI: 10.1016/j.addr.2013.09.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Revised: 09/18/2013] [Accepted: 09/27/2013] [Indexed: 12/12/2022]
Abstract
A new chapter in the history of medical diagnosis happened when the first X-ray technology was invented in the late 1800s. Since then, many non-invasive and minimally invasive imaging techniques have been invented for clinical diagnosis to research in cellular biology, drug discovery, and disease monitoring. These imaging modalities have leveraged the benefits of significant advances in computer, electronics, and information technology and, more recently, targeted molecular imaging. The development of targeted contrast agents such as fluorescent and nanoparticle probes coupled with optical imaging techniques has made it possible to selectively view specific biological events and processes in both in vivo and ex vivo systems with great sensitivity and selectivity. Thus, the combination of targeted molecular imaging probes and optical imaging techniques have become a mainstay in modern medicinal and biological research. Many promising results have demonstrated great potentials to translate to clinical applications. In this review, we describe a discussion of employing imaging probes and optical microendoscopic imaging techniques for cancer diagnosis.
Collapse
|
683
|
Nakajima T, Sato K, Hanaoka H, Watanabe R, Harada T, Choyke PL, Kobayashi H. The effects of conjugate and light dose on photo-immunotherapy induced cytotoxicity. BMC Cancer 2014; 14:389. [PMID: 24885589 PMCID: PMC4055275 DOI: 10.1186/1471-2407-14-389] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 05/20/2014] [Indexed: 12/03/2022] Open
Abstract
Background Photoimmunotherapy (PIT) is a highly cell-selective cancer therapy, which employs monoclonal antibodies conjugated to a potent photosensitizer (mAb-IR700). Once the conjugate has bound to the target cell, exposure to near infrared (NIR) light induces necrosis only in targeted cells with minimal damage to adjacent normal cells in vivo. Herein, we report on the effect of altering mAb-IR700 and light power and dose on effectiveness of PIT. Methods For evaluating cytotoxicity, we employed ATP-dependent bioluminescence imaging using a luciferase-transfected MDA-MB-468luc cell line, which expresses EGFR and luciferase. In in vitro experiments, panitumumab-IR700 (Pan-IR700) concentration was varied in combination with varying NIR light doses administered by an LED at one of three power settings, 100 mA and 400 mA continuous wave and 1733 mA intermittent wave. For in vivo experiments, the MDA-MB-468luc orthotopic breast cancer was treated with varying doses of Pan-IR700 and light. Results The in vitro cell study demonstrated that PIT induced cytotoxicity depended on light dose, when the conjugate concentration was kept constant. Increasing the dose of Pan-IR700 allowed lowering of the light dose to achieve equal effects thus indicating that for a given level of efficacy, the conjugate concentration multiplied by the light dose was a constant. A similar relationship between conjugate and light dose was observed in vivo. Conclusions The efficacy of PIT is defined by the product of the number of bound antibody conjugates and the dose of NIR light and can be achieve equally with continuous and pulse wave LED light using different power densities.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Hisataka Kobayashi
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Bldg, 10, Room B3B69, MSC 1088, Bethesda, Maryland 20892-1088, USA.
| |
Collapse
|
684
|
Huang HC, Hasan T. The " Nano" World in Photodynamic Therapy. AUSTIN JOURNAL OF NANOMEDICINE & NANOTECHNOLOGY 2014; 2:1020. [PMID: 34350362 PMCID: PMC8329842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Photodynamic Therapy (PDT) is an externally activated, photochemistry-based approach that generates cytotoxic reactive molecular species (RMS), which kill or modulate biological targets. PDT provides unique opportunities for applications of nanotechnology where light activation can trigger both direct RMS-mediated cytotoxic activity and the release of contents within the nanoconstructs (Figure 1). This process allows several species, working via different mechanisms and molecular targets to be activated or released in the right place and time, thus providing a distinctive approach to combination therapy. With advances in the development of miniaturized, even biodegradable, light sources and delivery systems, exciting possibilities of anatomical reach with PDT are being made possible. This brief article introduces aspects of interfaces of PDT and nanotechnology but, due to space constraints, makes no attempt to be a comprehensive review.
Collapse
Affiliation(s)
| | - Tayyaba Hasan
- Harvard Medical School, Massachusetts General Hospital, USA
- Division of Health Sciences and Technology, Harvard-MIT, USA
| |
Collapse
|
685
|
Jia N, Zhang S, Shao P, Bagia C, Janjic JM, Ding Y, Bai M. Cannabinoid CB2 receptor as a new phototherapy target for the inhibition of tumor growth. Mol Pharm 2014; 11:1919-29. [PMID: 24779700 DOI: 10.1021/mp5001923] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The success of targeted cancer therapy largely relies upon the selection of target and the development of efficient therapeutic agents that specifically bind to the target. In the current study, we chose a cannabinoid CB2 receptor (CB2R) as a new target and used a CB2R-targeted photosensitizer, IR700DX-mbc94, for phototherapy treatment. IR700DX-mbc94 was prepared by conjugating a photosensitizer, IR700DX, to mbc94, whose binding specificity to CB2R has been previously demonstrated. We found that phototherapy treatment using IR700DX-mbc94 greatly inhibited the growth of CB2R positive tumors but not CB2R negative tumors. In addition, phototherapy treatment with nontargeted IR700DX did not show significant therapeutic effect. Similarly, treatment with IR700DX-mbc94 without light irradiation or light irradiation without the photosensitizer showed no tumor-inhibitory effect. Taken together, IR700DX-mbc94 is a promising phototherapy agent with high target-specificity. Moreover, CB2R appears to have great potential as a phototherapeutic target for cancer treatment.
Collapse
Affiliation(s)
- Ningyang Jia
- Department of Radiology, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University , Shanghai 200438, P. R. China
| | | | | | | | | | | | | |
Collapse
|
686
|
Sato K, Watanabe R, Hanaoka H, Harada T, Nakajima T, Kim I, Paik CH, Choyke PL, Kobayashi H. Photoimmunotherapy: comparative effectiveness of two monoclonal antibodies targeting the epidermal growth factor receptor. Mol Oncol 2014; 8:620-32. [PMID: 24508062 PMCID: PMC4004687 DOI: 10.1016/j.molonc.2014.01.006] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Revised: 12/05/2013] [Accepted: 01/13/2014] [Indexed: 01/13/2023] Open
Abstract
Photoimmunotherapy (PIT) is a new cancer treatment that combines the specificity of antibodies for targeting tumors with the toxicity induced by photosensitizers after exposure to near infrared (NIR) light. Herein we compare two commonly available anti-EGFR monoclonal antibodies, cetuximab and panitumumab, for their effectiveness as PIT agents in EGFR positive tumor models. A photosensitizer, IR-700, conjugated to either cetuximab (cet-IR700) or panitumumab (pan-IR700), was evaluated using EGFR-expressing A431 and MDAMB468-luc cells in 2D- and 3D-culture. PIT was conducted with irradiation of NIR light after exposure of the sample or animal to each conjugate. In vivo PIT was performed with fractionated exposure of NIR light after injection of each agent into A431 xenografts or a MDAMB468-luc orthotopic tumor bearing model. Cet-IR700 and pan-IR700 bound with equal affinity to the cells in 2D-culture and penetrated equally into the 3D-spheroid, resulting in identical PIT cytotoxic effects in vitro. In contrast, in vivo anti-tumor effects of PIT with cet-IR700 were inferior to that of pan-IR700. Assessment of the biodistribution showed lower accumulation into the tumors and more rapid hepatic catabolism of cet-IR700 compared to pan-IR700. Although cet-IR700 and pan-IR700 showed identical in vitro characteristics, pan-IR700 showed better therapeutic tumor responses than cet-IR700 in in vivo mice models due to the prolonged retention of the conjugate in the circulation, suggesting that retention in the circulation is advantageous for tumor responses to PIT. These results suggest that the choice of monoclonal antibody in photosensitizer conjugates may influence the effectiveness of PIT.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/pharmacokinetics
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized/chemistry
- Antibodies, Monoclonal, Humanized/pharmacokinetics
- Antibodies, Monoclonal, Humanized/therapeutic use
- Cell Line, Tumor
- Cetuximab
- ErbB Receptors/antagonists & inhibitors
- Female
- Humans
- Immunotherapy/methods
- Mice
- Neoplasms/pathology
- Neoplasms/therapy
- Panitumumab
- Photosensitizing Agents/chemistry
- Photosensitizing Agents/pharmacokinetics
- Photosensitizing Agents/therapeutic use
- Phototherapy/methods
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Kazuhide Sato
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, MD 20892, United States
| | - Rira Watanabe
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, MD 20892, United States
| | - Hirofumi Hanaoka
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, MD 20892, United States
| | - Toshiko Harada
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, MD 20892, United States
| | - Takahito Nakajima
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, MD 20892, United States
| | - Insook Kim
- Applied/Developmental Research Directorate, Leidos Biomedical Research Inc., Frederick, MD 20892, United States
| | - Chang H Paik
- Nuclear Medicine Department, Warren Grant Magnuson Clinical Center, MD 20892, United States
| | - Peter L Choyke
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, MD 20892, United States
| | - Hisataka Kobayashi
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, MD 20892, United States.
| |
Collapse
|
687
|
Guo M, Mao H, Li Y, Zhu A, He H, Yang H, Wang Y, Tian X, Ge C, Peng Q, Wang X, Yang X, Chen X, Liu G, Chen H. Dual imaging-guided photothermal/photodynamic therapy using micelles. Biomaterials 2014; 35:4656-66. [PMID: 24613048 PMCID: PMC4568826 DOI: 10.1016/j.biomaterials.2014.02.018] [Citation(s) in RCA: 174] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 02/10/2014] [Indexed: 11/26/2022]
Abstract
We report a type of photosensitizer (PS)-loaded micelles integrating cyanine dye as potential theranostic micelles for precise anatomical tumor localization via dual photoacoustic (PA)/near-infrared fluorescent (NIRF) imaging modalities, and simultaneously superior cancer therapy via sequential synergistic photothermal therapy (PTT)/photodynamic therapy (PDT). The micelles exhibit enhanced photostability, cell internalization and tumor accumulation. The dual NIRF/PA imaging modalities of the micelles cause the high imaging contrast and spatial resolution of tumors, which provide precise anatomical localization of the tumor and its inner vasculature for guiding PTT/PDT treatments. Moreover, the micelles can generate severe photothermal damage on cancer cells and destabilization of the lysosomes upon PTT photoirradiation, which subsequently facilitate synergistic photodynamic injury via PS under PDT treatment. The sequential treatments of PTT/PDT trigger the enhanced cytoplasmic delivery of PS, which contributes to the synergistic anticancer efficacy of PS. Our strategy provides a dual-modal cancer imaging with high imaging contrast and spatial resolution, and subsequent therapeutic synergy of PTT/PDT for potential multimodal theranostic application.
Collapse
Affiliation(s)
- Miao Guo
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Huajian Mao
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Yanli Li
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Aijun Zhu
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Hui He
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Hong Yang
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Yangyun Wang
- School for Radiological & Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou 215123, China; School of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China
| | - Xin Tian
- School for Radiological & Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou 215123, China; School of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China
| | - Cuicui Ge
- School for Radiological & Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou 215123, China; School of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China
| | - Qiaoli Peng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Xiaoyong Wang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Xiangliang Yang
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health, Bethesda, MD 20892, United States
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China.
| | - Huabing Chen
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; School for Radiological & Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou 215123, China; School of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China.
| |
Collapse
|
688
|
Liu J, Li J, Zhang Z, Weng Y, Chen G, Yuan B, Yang K, Ma Y. Encapsulation of Hydrophobic Phthalocyanine with Poly( N-isopropylacrylamide)/Lipid Composite Microspheres for Thermo-Responsive Release and Photodynamic Therapy. MATERIALS (BASEL, SWITZERLAND) 2014; 7:3481-3493. [PMID: 28788630 PMCID: PMC5453233 DOI: 10.3390/ma7053481] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 03/26/2014] [Accepted: 04/21/2014] [Indexed: 12/18/2022]
Abstract
Phthalocyanine (Pc) is a type of promising sensitizer molecules for photodynamic therapy (PDT), but its hydrophobicity substantially prevents its applications. In this study, we efficiently encapsulate Pc into poly(N-isopropylacrylamide) (pNIPAM) microgel particles, without or with lipid decoration (i.e., Pc@pNIPAM or Pc@pNIPAM/lipid), to improve its water solubility and prevent aggregation in aqueous medium. The incorporation of lipid molecules significantly enhances the Pc loading efficiency of pNIPAM. These Pc@pNIPAM and Pc@pNIPAM/lipid composite microspheres show thermo-triggered release of Pc and/or lipid due to the phase transition of pNIPAM. Furthermore, in the in vitro experiments, these composite particles work as drug carriers for the hydrophobic Pc to be internalized into HeLa cells. After internalization, the particles show efficient fluorescent imaging and PDT effect. Our work demonstrates promising candidates in promoting the use of hydrophobic drugs including photosensitizers in tumor therapies.
Collapse
Affiliation(s)
- Jiaojiao Liu
- Center for Soft Condensed Matter Physics and Interdisciplinary Research, Soochow University, Suzhou 215006, Jiangsu, China.
| | - Jingliang Li
- Institute for Frontier Materials, Deakin University, Waurn Ponds, Vic 3216, Australia.
| | - Zexin Zhang
- Center for Soft Condensed Matter Physics and Interdisciplinary Research, Soochow University, Suzhou 215006, Jiangsu, China.
| | - Yuyan Weng
- Center for Soft Condensed Matter Physics and Interdisciplinary Research, Soochow University, Suzhou 215006, Jiangsu, China.
| | - Gaojian Chen
- Center for Soft Condensed Matter Physics and Interdisciplinary Research, Soochow University, Suzhou 215006, Jiangsu, China.
| | - Bing Yuan
- Center for Soft Condensed Matter Physics and Interdisciplinary Research, Soochow University, Suzhou 215006, Jiangsu, China.
| | - Kai Yang
- Center for Soft Condensed Matter Physics and Interdisciplinary Research, Soochow University, Suzhou 215006, Jiangsu, China.
| | - Yuqiang Ma
- Center for Soft Condensed Matter Physics and Interdisciplinary Research, Soochow University, Suzhou 215006, Jiangsu, China.
- National Laboratory of Solid State Microstructures, Department of Physics, Nanjing University, Nanjing 210093, Jiangsu, China.
| |
Collapse
|
689
|
Shirasu N, Yamada H, Shibaguchi H, Kuroki M, Kuroki M. Potent and specific antitumor effect of CEA-targeted photoimmunotherapy. Int J Cancer 2014; 135:2697-710. [PMID: 24740257 DOI: 10.1002/ijc.28907] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 04/02/2014] [Indexed: 12/27/2022]
Abstract
Conventional photodynamic therapy (PDT) for cancer is limited by the insufficient efficacy and specificity of photosensitizers. We herein describe a highly effective and selective tumor-targeted PDT using a near-infrared (NIR) photosensitizer, IRDye700DX, conjugated to a human monoclonal antibody (Ab) specific for carcinoembryonic antigen (CEA). The antitumor effects of this Ab-assisted PDT, called photoimmunotherapy (PIT), were investigated in vitro and in vivo. The Ab-IRDye conjugate induced potent cytotoxicity against CEA-positive tumor cells after NIR-irradiation, whereas CEA-negative cells were not affected at all, even in the presence of excess photoimmunoconjugate. We found an equivalent phototoxicity and a predominant plasma membrane localization of Ab-IRDye after both one and six hours of incubation. Either no or little caspase activation and membrane peroxidation were observed in PIT-treated cells and a panel of scavengers for reactive oxygen species showed only partial inhibition of the phototoxic effect. Strikingly, Ab-IRDye retained significant phototoxicity even under hypoxia. We established a xenograft model, which allowed us to sensitively investigate the therapeutic efficacy of PIT by non-invasive bioluminescence imaging. Luciferase-expressing MKN-45-luc human gastric carcinoma cells were subcutaneously implanted into both flanks of nude mice. NIR-irradiation was performed for only the tumor on one side. In vivo imaging and measurement of the tumor size revealed that a single PIT treatment, with intraperitoneal administration of Ab-IRDye and subsequent NIR-irradiation, caused rapid cell death and significant inhibition of tumor growth, but only on the irradiated side. Together, these data suggest that Ab-IRDye-mediated PIT has great potential as an anticancer therapeutics targeting CEA-positive tumors.
Collapse
Affiliation(s)
- Naoto Shirasu
- Department of Biochemistry, Faculty of Medicine, Fukuoka University, Japan
| | | | | | | | | |
Collapse
|
690
|
Horiuchi H, Hosaka M, Mashio H, Terata M, Ishida S, Kyushin S, Okutsu T, Takeuchi T, Hiratsuka H. Silylation improves the photodynamic activity of tetraphenylporphyrin derivatives in vitro and in vivo. Chemistry 2014; 20:6054-60. [PMID: 24710805 DOI: 10.1002/chem.201303120] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 01/29/2014] [Indexed: 11/06/2022]
Abstract
The effects of silyl and hydrophilic groups on the photodynamic properties of tetraphenylporphyrin (TPP) derivatives have been studied in vitro and in vivo. Silylation led to an improvement in the quantum yield of singlet oxygen sensitization for both sulfo and carboxy derivatives, although the silylation did not affect other photophysical properties. Silylation also improved the cellular uptake efficiency for both sulfo and carboxy derivatives, enhancing the in vitro photodynamic activity of the photosensitizer in U251 human glioma cells. The carboxy derivative (SiTPPC4 ) was found to show higher cellular uptake efficiency and in vitro photodynamic activity than the corresponding sulfo derivative (SiTPPS4 ), which indicates that the carboxy group is a more promising hydrophilic group than the sulfo group in the silylated porphyrin. SiTPPC4 was found to show high selective accumulation efficiency in tumors, although almost no tumor selectivity was observed for the nonsilylated porphyrin. The concentration of SiTPPC4 in tumors was 13 times higher than that in muscle 12 h after drug administration. We also studied tumor response after treatment and found that silylation enhanced in vivo photodynamic activity significantly. SiTPPC4 shows higher photodynamic activity than NPe6 with white light irradiation.
Collapse
Affiliation(s)
- Hiroaki Horiuchi
- Division of Molecular Science and International Education and Research Center for Silicon Science, Faculty of Science and Technology, Gunma University, Kiryu (Japan), Fax: (+81) 277-30-1244.
| | | | | | | | | | | | | | | | | |
Collapse
|
691
|
Zhang L, Zhou H, Belzile O, Thorpe P, Zhao D. Phosphatidylserine-targeted bimodal liposomal nanoparticles for in vivo imaging of breast cancer in mice. J Control Release 2014; 183:114-23. [PMID: 24698945 DOI: 10.1016/j.jconrel.2014.03.043] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 03/24/2014] [Indexed: 11/30/2022]
Abstract
Phosphatidylserine (PS) that is normally constrained to the inner plasma membrane becomes exposed on the surface of endothelial cells (ECs) in tumor vasculature. In the present study, we report the development of a novel tumor vasculature-targeted liposomal nanoprobe by conjugating a human monoclonal antibody, PGN635 that specifically targets PS to polyethylene glycol-coated liposomes. MR contrast, superparamagnetic iron oxide nanoparticles (SPIO) were packed into the core of liposomes, while near-infrared dye, DiR was incorporated into the lipophilic bilayer. The liposomal nanoprobe PGN-L-IO/DiR was fully characterized, and its binding specificity and subsequent internalization into PS-exposed vascular ECs was confirmed by in vitro MRI and histological staining. In vivo longitudinal MRI and optical imaging were performed after i.v. injection of the liposomal nanoprobes into mice bearing breast MDA-MB231 tumors. At 9.4T, T2-weighted MRI detected drastic reduction on signal intensity and T2 values of tumors at 24h. Ionizing radiation significantly increased PS exposure on tumor vascular ECs, resulting in a further MRI signal loss of tumors. Concurrent with MRI, optical imaging revealed a clear tumor contrast at 24h. Intriguingly, PGN-L-IO/DiR exhibited distinct pharmacokinetics and biodistribution with significantly reduced accumulations in liver or spleen. Localization of PGN-L-IO/DiR to tumor was antigen specific, since a control probe of irrelevant specificity showed minimal accumulation in the tumors. Our studies indicate that PS-targeted liposomes may provide a useful platform for tumor-targeted delivery of imaging contrast agents or potentially anti-cancer drugs for cancer theranostics.
Collapse
Affiliation(s)
- Liang Zhang
- Radiology, UT Southwestern Medical Center, Dallas, USA
| | - Heling Zhou
- Radiology, UT Southwestern Medical Center, Dallas, USA
| | | | - Philip Thorpe
- Pharmacology, UT Southwestern Medical Center, Dallas, USA
| | - Dawen Zhao
- Radiology, UT Southwestern Medical Center, Dallas, USA.
| |
Collapse
|
692
|
Wang H, Liu Z, Wang S, Dong C, Gong X, Zhao P, Chang J. MC540 and upconverting nanocrystal coloaded polymeric liposome for near-infrared light-triggered photodynamic therapy and cell fluorescent imaging. ACS APPLIED MATERIALS & INTERFACES 2014; 6:3219-3225. [PMID: 24511877 DOI: 10.1021/am500097f] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
In clinic, the application of photodynamic therapy (PDT) in deep tissue is severely constrained by the limited penetration depth of visible light needed for activating the photosensitizer (PS). In this Article, a merocyanine 540 (MC540) and upconverting nanoparticle (UCN) coloaded functional polymeric liposome nanocarrier, (MC540 + UCN)/FPL, was designed and constructed successfully for solving this problem in PDT. Compared with the conventional approaches using UCNs absorbing PSs directly, the combination of UCN and polymeric liposome has unique advantages. The UCN core as a transducer can convert deep-penetrating near-infrared light to visible light for activating MC540. The functional polymeric liposome shell decorated with folate as a nanoshield can keep the UCN and MC540 stable, protect them from being attacked, and help them get into cells. The results show that (MC540 + UCN)/FPL is an individual nanosphere with an average size of 26 nm. MC540 can be activated to produce singlet oxygen successfully by upconverting fluorescence emitted from UCNs. After (MC540 + UCN)/FPL was modified with folate, the cell uptake efficiency increased obviously. More interestingly, in the PDT effect test, the (MC540 + UCN)/FPL nanocarrier further improved the inhibition effect on tumor cells by anchoring targeting folate and transactivating transduction peptide. Our data suggest that the (MC540 + UCN)/FPL nanocarrier may be a useful nanoplatform for future PDT treatment in deep-cancer therapy based on upconversion mechanism.
Collapse
Affiliation(s)
- Hanjie Wang
- Institute of Nanobiotechnology, School of Materials Science and Engineering, Tianjin University and Tianjin Key Laboratory of Composites and Functional Materials , Tianjin 300072, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
693
|
Zhang S, Jia N, Shao P, Tong Q, Xie XQ, Bai M. Target-selective phototherapy using a ligand-based photosensitizer for type 2 cannabinoid receptor. ACTA ACUST UNITED AC 2014; 21:338-44. [PMID: 24583052 DOI: 10.1016/j.chembiol.2014.01.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2013] [Revised: 12/29/2013] [Accepted: 01/23/2014] [Indexed: 12/31/2022]
Abstract
Phototherapy is a powerful, noninvasive approach for cancer treatment, with several agents currently in clinical use. Despite the progress and promise, most current phototherapy agents have serious side effects as they can lead to damage to healthy tissue, even when the photosensitizers are fused to targeting molecules due to nonspecific light activation of the unbound photosensitizer. To overcome these limitations, we developed a phototherapy agent that combines a functional ligand and a near infrared phthalocyanine dye. Our target is type 2 cannabinoid receptor (CB2R), considered an attractive therapeutic target for phototherapy given it is overexpressed by many types of cancers that are located at a surface or can be reached by an endoscope. We show that our CB2R-targeted phototherapy agent, IR700DX-mbc94, is specific for CB2R and effective only when bound to the target receptor. Overall, this opens up the opportunity for development of an alternative treatment option for CB2R-positive cancers.
Collapse
Affiliation(s)
- Shaojuan Zhang
- Department of Radiology, University of Pittsburgh, 100 Technology Drive, Pittsburgh, PA 15219, USA; Department of Diagnostic Radiology, First Hospital of Medical School, Xi'an Jiaotong University, 277 Yanta West Road, 710061 Xi'an, PRC
| | - Ningyang Jia
- Department of Radiology, University of Pittsburgh, 100 Technology Drive, Pittsburgh, PA 15219, USA; Department of Radiology, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 225 Changhai Road, 200438 Shanghai, PRC
| | - Pin Shao
- Department of Radiology, University of Pittsburgh, 100 Technology Drive, Pittsburgh, PA 15219, USA
| | - Qin Tong
- Department of Pharmaceutical Sciences, Computational Chemical Genomics Screening Center, School of Pharmacy and Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Xiang-Qun Xie
- Department of Pharmaceutical Sciences, Computational Chemical Genomics Screening Center, School of Pharmacy and Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Mingfeng Bai
- Department of Radiology, University of Pittsburgh, 100 Technology Drive, Pittsburgh, PA 15219, USA; University of Pittsburgh Cancer Institute, 5150 Centre Avenue, Pittsburgh, PA 15232, USA.
| |
Collapse
|
694
|
Selective treatment and monitoring of disseminated cancer micrometastases in vivo using dual-function, activatable immunoconjugates. Proc Natl Acad Sci U S A 2014; 111:E933-42. [PMID: 24572574 DOI: 10.1073/pnas.1319493111] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Drug-resistant micrometastases that escape standard therapies often go undetected until the emergence of lethal recurrent disease. Here, we show that it is possible to treat microscopic tumors selectively using an activatable immunoconjugate. The immunoconjugate is composed of self-quenching, near-infrared chromophores loaded onto a cancer cell-targeting antibody. Chromophore phototoxicity and fluorescence are activated by lysosomal proteolysis, and light, after cancer cell internalization, enabling tumor-confined photocytotoxicity and resolution of individual micrometastases. This unique approach not only introduces a therapeutic strategy to help destroy residual drug-resistant cells but also provides a sensitive imaging method to monitor micrometastatic disease in common sites of recurrence. Using fluorescence microendoscopy to monitor immunoconjugate activation and micrometastatic disease, we demonstrate these concepts of "tumor-targeted, activatable photoimmunotherapy" in a mouse model of peritoneal carcinomatosis. By introducing targeted activation to enhance tumor selectively in complex anatomical sites, this study offers prospects for catching early recurrent micrometastases and for treating occult disease.
Collapse
|
695
|
Kirui DK, Mai J, Palange AL, Qin G, van de Ven AL, Liu X, Shen H, Ferrari M. Transient mild hyperthermia induces E-selectin mediated localization of mesoporous silicon vectors in solid tumors. PLoS One 2014; 9:e86489. [PMID: 24558362 PMCID: PMC3928046 DOI: 10.1371/journal.pone.0086489] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 12/09/2013] [Indexed: 01/03/2023] Open
Abstract
Background Hyperthermia treatment has been explored as a strategy to overcome biological barriers that hinder effective drug delivery in solid tumors. Most studies have used mild hyperthermia treatment (MHT) to target the delivery of thermo-sensitive liposomes carriers. Others have studied its application to permeabilize tumor vessels and improve tumor interstitial transport. However, the role of MHT in altering tumor vessel interfacial and adhesion properties and its relationship to improved delivery has not been established. In the present study, we evaluated effects of MHT treatment on tumor vessel flow dynamics and expression of adhesion molecules and assessed enhancement in particle localization using mesoporous silicon vectors (MSVs). We also determined the optimal time window at which maximal accumulation occur. Results In this study, using intravital microscopy analyses, we showed that temporal mild hyperthermia (∼1 W/cm2) amplified delivery and accumulation of MSVs in orthotopic breast cancer tumors. The number of discoidal MSVs (1000×400 nm) adhering to tumor vasculature increased 6-fold for SUM159 tumors and 3-fold for MCF-7 breast cancer tumors. By flow chamber experiments and Western blotting, we established that a temporal increase in E-selectin expression correlated with enhanced particle accumulation. Furthermore, MHT treatment was shown to increase tumor perfusion in a time-dependent fashion. Conclusions Our findings reveal that well-timed mild hyperthermia treatment can transiently elevate tumor transport and alter vascular adhesion properties and thereby provides a means to enhance tumor localization of non-thermally sensitive particles such as MSVs. Such enhancement in accumulation could be leveraged to increase therapeutic efficacy and reduce drug dosing in cancer therapy.
Collapse
Affiliation(s)
- Dickson K. Kirui
- Department of NanoMedicine, Houston Methodist Research Institute, Houston, Texas, United States of America
| | - Juahua Mai
- Department of NanoMedicine, Houston Methodist Research Institute, Houston, Texas, United States of America
| | - Anna-Lisa Palange
- Department of NanoMedicine, Houston Methodist Research Institute, Houston, Texas, United States of America
| | - Guoting Qin
- Department of NanoMedicine, Houston Methodist Research Institute, Houston, Texas, United States of America
| | - Anne L. van de Ven
- Department of NanoMedicine, Houston Methodist Research Institute, Houston, Texas, United States of America
| | - Xuewu Liu
- Department of NanoMedicine, Houston Methodist Research Institute, Houston, Texas, United States of America
| | - Haifa Shen
- Department of NanoMedicine, Houston Methodist Research Institute, Houston, Texas, United States of America
- Department of Cell and Development Biology, Weill Cornell Medical College, New York, New York, United States of America
| | - Mauro Ferrari
- Department of NanoMedicine, Houston Methodist Research Institute, Houston, Texas, United States of America
- Department of Internal Medicine, Weill Cornell Medical College, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
696
|
Yin J, Gan P, Zhou F, Wang J. Sensitive detection of transcription factors using near-infrared fluorescent solid-phase rolling circle amplification. Anal Chem 2014; 86:2572-9. [PMID: 24475783 DOI: 10.1021/ac403758p] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
This study describes a method for analyzing transcription factor (TF) activity, near-infrared fluorescent solid-phase rolling circle amplification (NIRF-sRCA). This method analyzes TF activity in four steps: (i) incubate DNA with protein sample and isolate TF-bound DNA, (ii) hybridize the TF-bound DNA and rolling circle to DNA microarray, (iii) amplify the TF-bound DNA with sRCA that contains biotin-labeled dUTP, and (iv) detect sRCA products by binding of NIRF-labeled streptavidin and NIRF imaging. This method was validated by proof-of-concept detection of purified TF protein and cell nuclear extract. Detection of purified TF protein demonstrated that NIRF-sRCA could quantitatively detect NF-κB p50 protein, and as little as 6.94 ng (∼140 fmol) of this protein was detected. Detection of nuclear extract revealed that NIRF-sRCA could specifically and quantitatively detect NF-κB p50 activity in HeLa cell nuclear extracts, and the activity of this TF in as little as 0.625 μg of nuclear extracts could be detected. Detection of nuclear extract also revealed that NIRF-sRCA could detect the relative activities of multiple TFs in HeLa cell nuclear extracts and the fold induction of multiple TFs in the TNFα-induced HeLa cell nuclear extracts. Therefore, this study provides a new tool for studying TFs.
Collapse
Affiliation(s)
- Junhuan Yin
- State Key Laboratory of Bioelectronics, Southeast University , Nanjing 210096, China
| | | | | | | |
Collapse
|
697
|
Min Y, Li J, Liu F, Padmanabhan P, Yeow EKL, Xing B. Recent Advance of Biological Molecular Imaging Based on Lanthanide-Doped Upconversion-Luminescent Nanomaterials. NANOMATERIALS 2014; 4:129-154. [PMID: 28348288 PMCID: PMC5304614 DOI: 10.3390/nano4010129] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 01/23/2014] [Accepted: 01/27/2014] [Indexed: 11/16/2022]
Abstract
Lanthanide-doped upconversion-luminescent nanoparticles (UCNPs), which can be excited by near-infrared (NIR) laser irradiation to emit multiplex light, have been proven to be very useful for in vitro and in vivo molecular imaging studies. In comparison with the conventionally used down-conversion fluorescence imaging strategies, the NIR light excited luminescence of UCNPs displays high photostability, low cytotoxicity, little background auto-fluorescence, which allows for deep tissue penetration, making them attractive as contrast agents for biomedical imaging applications. In this review, we will mainly focus on the latest development of a new type of lanthanide-doped UCNP material and its main applications for in vitro and in vivo molecular imaging and we will also discuss the challenges and future perspectives.
Collapse
Affiliation(s)
- Yuanzeng Min
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, Singapore 637371, Singapore.
| | - Jinming Li
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, Singapore 637371, Singapore.
| | - Fang Liu
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, Singapore 637371, Singapore.
| | - Parasuraman Padmanabhan
- The Lee Kong Chian School of Medicine, Nanyang Technological University, 50 Nanyang Drive, Research Techno Plaza, Singapore 637553, Singapore.
| | - Edwin K L Yeow
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, Singapore 637371, Singapore.
| | - Bengang Xing
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, Singapore 637371, Singapore.
| |
Collapse
|
698
|
Noninvasive positron emission tomography and fluorescence imaging of CD133+ tumor stem cells. Proc Natl Acad Sci U S A 2014; 111:E692-701. [PMID: 24469819 DOI: 10.1073/pnas.1314189111] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
A technology that visualizes tumor stem cells with clinically relevant tracers could have a broad impact on cancer diagnosis and treatment. The AC133 epitope of CD133 currently is one of the best-characterized tumor stem cell markers for many intra- and extracranial tumor entities. Here we demonstrate the successful noninvasive detection of AC133(+) tumor stem cells by PET and near-infrared fluorescence molecular tomography in subcutaneous and orthotopic glioma xenografts using antibody-based tracers. Particularly, microPET with (64)Cu-NOTA-AC133 mAb yielded high-quality images with outstanding tumor-to-background contrast, clearly delineating subcutaneous tumor stem cell-derived xenografts from surrounding tissues. Intracerebral tumors as small as 2-3 mm also were clearly discernible, and the microPET images reflected the invasive growth pattern of orthotopic cancer stem cell-derived tumors with low density of AC133(+) cells. These data provide a basis for further preclinical and clinical use of the developed tracers for high-sensitivity and high-resolution monitoring of AC133(+) tumor stem cells.
Collapse
|
699
|
Synthesis, bioanalysis and biodistribution of photosensitizer conjugates for photodynamic therapy. Bioanalysis 2014; 5:1099-114. [PMID: 23641699 DOI: 10.4155/bio.13.37] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Photodynamic therapy (PDT) was discovered in 1900 by Raab, and has since emerged as a promising tool for treating diseases characterized by unwanted cells or hyperproliferating tissue (e.g., cancer or infectious disease). PDT consists of the light excitation of a photosensitizer (PS) in the presence of O(2) to yield highly reactive oxygen species. In recent years, PDT has been improved by the synthesis of targeted bioconjugates between monoclonal antibodies and PS, and by investigating PS biodistribution and PD. Here, we provide a comprehensive review of major developments in PS-immunoconjugate-based PDT and the bioanalysis of these agents, with a specific emphasis on anticancer and antimicrobial PDT.
Collapse
|
700
|
Rong P, Yang K, Srivastan A, Kiesewetter DO, Yue X, Wang F, Nie L, Bhirde A, Wang Z, Liu Z, Niu G, Wang W, Chen X. Photosensitizer loaded nano-graphene for multimodality imaging guided tumor photodynamic therapy. Theranostics 2014; 4:229-39. [PMID: 24505232 PMCID: PMC3915087 DOI: 10.7150/thno.8070] [Citation(s) in RCA: 177] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 12/01/2013] [Indexed: 12/11/2022] Open
Abstract
Graphene, a 2-dimensional carbon nanomaterial, has attracted wide attention in biomedical applications, owing to its intrinsic physical and chemical properties. In this work, a photosensitizer molecule, 2-(1-hexyloxyethyl)-2-devinyl pyropheophorbide-alpha (HPPH or Photochlor®), is loaded onto polyethylene glycol (PEG)-functionalized graphene oxide (GO) via supramolecular π-π stacking. The obtained GO-PEG-HPPH complex shows high HPPH loading efficiency. The in vivo distribution and delivery were tracked by fluorescence imaging as well as positron emission tomography (PET) after radiolabeling of HPPH with (64)Cu. Compared with free HPPH, GO-PEG-HPPH offers dramatically improved photodynamic cancer cell killing efficacy due to the increased tumor delivery of HPPH. Our study identifies a role for graphene as a carrier of PDT agents to improve PDT efficacy and increase long-term survival following treatment.
Collapse
|