1
|
Li M, Chu K, Zhou Q, Wang H, Zhang W, Zhang Y, Lv J, Zhou H, An J, Wu Z, Li S. Dual-drug loaded hyaluronic acid conjugates coated polydopamine nanodrugs for synergistic chemo-photothermal therapy in triple negative breast cancer. Int J Biol Macromol 2025; 308:142559. [PMID: 40154698 DOI: 10.1016/j.ijbiomac.2025.142559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 03/06/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
Although combination of chemotherapy and photothermal therapy (PTT) holds significant promise for treating triple-negative breast cancer, the existing delivery systems for achieving synergistic antitumor activity remains unsatisfactory. Herein, we developed of dual-drug loaded hyaluronic acid (HA) nanodrugs, which exhibited pH, glutathione (GSH), and thermal triple-responsiveness and CD44-targeting capabilities for chemo-PTT synergistic therapy in breast cancer. Gemcitabine (GCB) and metformin (MET) were conjugated to HA via amide and disulfide bonds to form dual-drug loaded prodrugs (HSGM), which were then coated onto the surface of polydopamine nanoparticles (PDA NPs) to self-assemble into HSGM/PDA NPs. These NPs selectively accumulated at the tumor site through HA receptors and released GCB and MET in response to low pH and high GSH concentrations. The NPs demonstrated excellent photothermal performance, with heat generated from near-infrared (NIR)-laser irradiation accelerating drug release within tumor. Additionally, MET inhibited the production of heat shock protein 70 (HSP 70), mitigating thermotolerance induced by PTT, thereby enhancing the PTT effect. The combination of chemotherapy and PTT synergistically improved anti-tumor efficacy (tumor inhibition ratio: 99.11 %) while showing negligible systemic toxicity, effectively preventing tumor metastasis and recurrence. This integrated approach offers valuable insights for the clinical treatment of breast cancer and other malignant tumors.
Collapse
Affiliation(s)
- Min Li
- Department of Nuclear Medicine, The First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan 030001, Shanxi Province, PR China; Molecular Imaging Precision Medical Collaborative Innovation Center, Shanxi Medical University, Taiyuan 030001, Shanxi Province, PR China
| | - Kaile Chu
- Department of Nuclear Medicine, The First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan 030001, Shanxi Province, PR China; Molecular Imaging Precision Medical Collaborative Innovation Center, Shanxi Medical University, Taiyuan 030001, Shanxi Province, PR China
| | - Qin Zhou
- Department of Nuclear Medicine, The First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan 030001, Shanxi Province, PR China; Molecular Imaging Precision Medical Collaborative Innovation Center, Shanxi Medical University, Taiyuan 030001, Shanxi Province, PR China
| | - Hongliang Wang
- Department of Nuclear Medicine, The First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan 030001, Shanxi Province, PR China; Molecular Imaging Precision Medical Collaborative Innovation Center, Shanxi Medical University, Taiyuan 030001, Shanxi Province, PR China
| | - Wenjun Zhang
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian 116024, Liaoning Province, PR China; School of Chemical Engineering, Dalian University of Technology, Panjin 124221, Liaoning Province, PR China
| | - Yaqiong Zhang
- Department of Nuclear Medicine, The First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan 030001, Shanxi Province, PR China; Molecular Imaging Precision Medical Collaborative Innovation Center, Shanxi Medical University, Taiyuan 030001, Shanxi Province, PR China
| | - Junping Lv
- Department of Nuclear Medicine, The First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan 030001, Shanxi Province, PR China; Molecular Imaging Precision Medical Collaborative Innovation Center, Shanxi Medical University, Taiyuan 030001, Shanxi Province, PR China
| | - Haitao Zhou
- Department of Nuclear Medicine, The First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan 030001, Shanxi Province, PR China; Molecular Imaging Precision Medical Collaborative Innovation Center, Shanxi Medical University, Taiyuan 030001, Shanxi Province, PR China
| | - Jie An
- Department of Nuclear Medicine, The First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan 030001, Shanxi Province, PR China; Molecular Imaging Precision Medical Collaborative Innovation Center, Shanxi Medical University, Taiyuan 030001, Shanxi Province, PR China.
| | - Zhifang Wu
- Department of Nuclear Medicine, The First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan 030001, Shanxi Province, PR China; Molecular Imaging Precision Medical Collaborative Innovation Center, Shanxi Medical University, Taiyuan 030001, Shanxi Province, PR China.
| | - Sijin Li
- Department of Nuclear Medicine, The First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan 030001, Shanxi Province, PR China; Molecular Imaging Precision Medical Collaborative Innovation Center, Shanxi Medical University, Taiyuan 030001, Shanxi Province, PR China.
| |
Collapse
|
2
|
Yukimura T, Seki T, Seki T. Preparation and Characterization of a Glutathione-Responsive Doxorubicin Prodrug Modified by 2-Nitrobenzenesulfonamide Group-Its Selective Cytotoxicity Toward Cells with Enhanced Glutathione Production. Int J Mol Sci 2025; 26:4128. [PMID: 40362369 PMCID: PMC12071201 DOI: 10.3390/ijms26094128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 04/23/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025] Open
Abstract
GSH biosynthesis is enhanced in cancer cells that express the variant isoform of the surface antigen CD44 (CD44v), which is overexpressed in certain types of cancer. The GSH-responsive prodrug Ns-Dox was prepared by modifying the GSH-responsive group 2-nitrobenzene sulfonyl (Ns) with the model drug doxorubicin (Dox). Its function was evaluated based on its molecular interaction with model DNA in terms of its binding constant (Ka). The association constant of Ns-Dox was lower, and its interaction with model DNA was weaker compared to that of Dox, suggesting that Ns-Dox may act as a less toxic prodrug. HCT116 cells with high CD44v expression and GSH levels and BT474 cells with low CD44v expression and GSH levels were used. The addition of Ns-Dox to HCT116 cells produced cytotoxic effects similar to those of Dox. In contrast, a significant difference in viability was observed between Ns-Dox- and Dox-treated BT474 cells at low concentrations. These findings suggest that Ns-Dox functions as a prodrug with low environmental toxicity and a lower GSH concentration in cancer cells. It is efficiently activated to Dox in cells with high GSH production, demonstrating its cell-killing effects.
Collapse
Affiliation(s)
| | - Tomohiro Seki
- Faculty of Pharmacy and Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama 350-0295, Japan; (T.Y.); (T.S.)
| | | |
Collapse
|
3
|
Zhao G, Zeng Y, Cheng W, Karkampouna S, Papadopoulou P, Hu B, Zang S, Wezenberg E, Forn-Cuní G, Lopes-Bastos B, Julio MKD, Kros A, Snaar-Jagalska BE. Peptide-Modified Lipid Nanoparticles Boost the Antitumor Efficacy of RNA Therapeutics. ACS NANO 2025; 19:13685-13704. [PMID: 40176316 PMCID: PMC12004924 DOI: 10.1021/acsnano.4c14625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 03/25/2025] [Accepted: 03/26/2025] [Indexed: 04/04/2025]
Abstract
RNA therapeutics offer a promising approach to cancer treatment by precisely regulating cancer-related genes. While lipid nanoparticles (LNPs) are currently the most advanced nonviral clinically approved vectors for RNA therapeutics, their antitumor efficacy is limited by their unspecific hepatic accumulation after systemic administration. Thus, there is an urgent need to enhance the delivery efficiency of LNPs to target tumor-residing tissues. Here, we conjugated the cluster of differentiation 44 (CD44)-specific targeting peptide A6 (KPSSPPEE) to the cholesterol of LNPs via PEG, named AKPC-LNP, enabling specific tumor delivery. This modification significantly improved delivery to breast cancer cells both in vitro and in vivo, as shown by flow cytometry and confocal microscopy. We further used AKPC-siYT to codeliver siRNAs targeting the transcriptional coactivators YAP and TAZ, achieving potent gene silencing and increased cell death in both 2D cultures and 3D tumor spheroids, outperforming unmodified LNPs. In a breast tumor cell xenografted zebrafish model, systemically administered AKPC-siYT induced robust silencing of YAP/TAZ and downstream genes and significantly enhanced tumor suppression compared to unmodified LNPs. Additionally, AKPC-siYT effectively reduced proliferation in prostate cancer organoids and tumor growth in a patient-derived xenograft (PDX) model. Overall, we developed highly efficient AKPC-LNPs carrying RNA therapeutics for targeted cancer therapy.
Collapse
Affiliation(s)
- Gangyin Zhao
- Department
of Cellular Tumor Biology, Leiden Institute of Biology, Leiden University, Einsteinweg 55, Leiden 2333 CC, the Netherlands
- Shenzhen
Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 51800, China
| | - Ye Zeng
- Department
of Supramolecular & Biomaterials Chemistry, Leiden Institute of
Chemistry, Leiden University, Einsteinweg 55, Leiden 2333 CC, the Netherlands
| | - Wanli Cheng
- Urology
Research Laboratory, Department for BioMedical Research, University of Bern, Bern 3010, Switzerland
| | - Sofia Karkampouna
- Urology
Research Laboratory, Department for BioMedical Research, University of Bern, Bern 3010, Switzerland
- Department
of Urology, Inselspital, Bern University Hospital, University of Bern, Bern 3010, Switzerland
| | - Panagiota Papadopoulou
- Department
of Supramolecular & Biomaterials Chemistry, Leiden Institute of
Chemistry, Leiden University, Einsteinweg 55, Leiden 2333 CC, the Netherlands
| | - Bochuan Hu
- Department
of Supramolecular & Biomaterials Chemistry, Leiden Institute of
Chemistry, Leiden University, Einsteinweg 55, Leiden 2333 CC, the Netherlands
| | - Shuya Zang
- Department
of Supramolecular & Biomaterials Chemistry, Leiden Institute of
Chemistry, Leiden University, Einsteinweg 55, Leiden 2333 CC, the Netherlands
| | - Emma Wezenberg
- Department
of Supramolecular & Biomaterials Chemistry, Leiden Institute of
Chemistry, Leiden University, Einsteinweg 55, Leiden 2333 CC, the Netherlands
| | - Gabriel Forn-Cuní
- Department
of Cellular Tumor Biology, Leiden Institute of Biology, Leiden University, Einsteinweg 55, Leiden 2333 CC, the Netherlands
| | - Bruno Lopes-Bastos
- Department
of Cellular Tumor Biology, Leiden Institute of Biology, Leiden University, Einsteinweg 55, Leiden 2333 CC, the Netherlands
| | - Marianna Kruithof-de Julio
- Department
of Urology, Inselspital, Bern University Hospital, University of Bern, Bern 3010, Switzerland
| | - Alexander Kros
- Department
of Supramolecular & Biomaterials Chemistry, Leiden Institute of
Chemistry, Leiden University, Einsteinweg 55, Leiden 2333 CC, the Netherlands
| | - B. Ewa Snaar-Jagalska
- Department
of Cellular Tumor Biology, Leiden Institute of Biology, Leiden University, Einsteinweg 55, Leiden 2333 CC, the Netherlands
| |
Collapse
|
4
|
Gao Y, Shelling AN, Nolan E, Porter D, Leung E, Wu Z. Liposome-enabled bufalin and doxorubicin combination therapy for trastuzumab-resistant breast cancer with a focus on cancer stem cells. J Liposome Res 2024; 34:489-506. [PMID: 38269490 DOI: 10.1080/08982104.2024.2305866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 01/10/2024] [Indexed: 01/26/2024]
Abstract
Breast cancer stem cells (BCSCs) play a key role in therapeutic resistance in breast cancer treatments and disease recurrence. This study aimed to develop a combination therapy loaded with pH-sensitive liposomes to kill both BCSCs and the okbulk cancer cells using trastuzumab-sensitive and resistant human epidermal growth factor receptor 2 positive (HER2+) breast cancer cell models. The anti-BCSCs effect and cytotoxicity of all-trans retinoic acid, salinomycin, and bufalin alone or in combination with doxorubicin were compared in HER2+ cell line BT-474 and a validated trastuzumab-resistant cell line, BT-474R. The most potent anti-BCSC agent was selected and loaded into a pH-sensitive liposome system. The effects of the liposomal combination on BCSCs and bulk cancer cells were assessed. Compared with BT-474, the aldehyde dehydrogenase positive BCSC population was elevated in BT-474R (3.9 vs. 23.1%). Bufalin was the most potent agent and suppressed tumorigenesis of BCSCs by ∼50%, and showed strong synergism with doxorubicin in both BT-474 and BT-474R cell lines. The liposomal combination of bufalin and doxorubicin significantly reduced the BCSC population size by 85%, and inhibited both tumorigenesis and self-renewal, although it had little effect on the migration and invasiveness. The cytotoxicity against the bulk cancer cells was also enhanced by the liposomal combination than either formulation alone in both cell lines (p < 0.001). The liposomal bufalin and doxorubicin combination therapy may effectively target both BCSCs and bulk cancer cells for a better outcome in trastuzumab-resistant HER2+ breast cancer.
Collapse
Affiliation(s)
- Yu Gao
- Faculty of Medical and Health Sciences, School of Pharmacy, The University of Auckland, Auckland, New Zealand
| | - Andrew N Shelling
- Faculty of Medical and Health Sciences, School of Medicine, The University of Auckland, Auckland, New Zealand
| | - Emma Nolan
- Faculty of Medical and Health Sciences, Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand
| | - David Porter
- Auckland Regional Cancer and Blood Service, Auckland City Hospital, Auckland, New Zealand
| | - Euphemia Leung
- Faculty of Medical and Health Sciences, Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand
| | - Zimei Wu
- Faculty of Medical and Health Sciences, School of Pharmacy, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
5
|
Mungra N, Nsole Biteghe FA, Huysamen AM, Hardcastle NS, Bunjun R, Naran K, Lang D, Richter W, Hunter R, Barth S. An Investigation into the In Vitro Targeted Killing of CD44-Expressing Triple-Negative Breast Cancer Cells Using Recombinant Photoimmunotherapeutics Compared to Auristatin-F-Based Antibody-Drug Conjugates. Mol Pharm 2024; 21:4098-4115. [PMID: 39047292 DOI: 10.1021/acs.molpharmaceut.4c00449] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
Triple-negative breast cancer (TNBC) is the deadliest form of breast cancer with limited treatment options. The persistence of highly tumorigenic CD44-expressing subpopulation referred to as cancer stem cells (CSCs), endowed with the self-renewal capacity, has been associated with therapeutic resistance, hence clinical relapses. To mitigate these undesired events, targeted immunotherapies using antibody-photoconjugate (APC) or antibody-drug conjugate (ADC), were developed to specifically release cytotoxic payloads within targeted cells overexpressing cognate antigen receptors. Therefore, an αCD44(scFv)-SNAP-tag antibody fusion protein was engineered through genetic fusion of a single-chain antibody fragment (scFv) to a SNAPf-tag fusion protein, capable of self-conjugating with benzylguanine-modified light-sensitive near-infrared (NIR) phthalocyanine dye IRDye700DX (BG-IR700) or the small molecule toxin auristatin-F (BG-AURIF). Binding of the αCD44(scFv)-SNAPf-IR700 photoimmunoconjugate to antigen-positive cells was demonstrated by confocal microscopy and flow cytometry. By switching to NIR irradiation, CD44-expressing TNBC was selectively killed through induced phototoxic activities. Likewise, the αCD44(scFv)-SNAPf-AURIF immunoconjugate was able to selectively accumulate within targeted cells and significantly reduced cell viability through antimitotic activities at nano- to micromolar drug concentrations. This study provides an in vitro proof-of-concept for a future strategy to selectively destroy light-accessible superficial CD44-expressing TNBC tumors and their metastatic lesions which are inaccessible to therapeutic light.
Collapse
Affiliation(s)
- Neelakshi Mungra
- Institute of Infectious Disease and Molecular Medicine, Medical Biotechnology and Immunotherapy Research Unit, University of Cape Town, Cape Town 7700, South Africa
- Centre for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Washington 98101, United States
| | - Fleury A Nsole Biteghe
- College of Science, Department of Biotechnology, Northeastern University, Boston, Massachusetts 02115, United States
| | - Allan M Huysamen
- Department of Chemistry, University of Cape Town, PD Hahn Building, Cape Town 7700, South Africa
| | - Natasha S Hardcastle
- Institute of Infectious Disease and Molecular Medicine, Medical Biotechnology and Immunotherapy Research Unit, University of Cape Town, Cape Town 7700, South Africa
| | - Rubina Bunjun
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7700, South Africa
- Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town 7700, South Africa
| | - Krupa Naran
- Institute of Infectious Disease and Molecular Medicine, Medical Biotechnology and Immunotherapy Research Unit, University of Cape Town, Cape Town 7700, South Africa
| | - Dirk Lang
- Division of Physiological Sciences, Department of Human Biology, University of Cape Town, Cape Town 7700, South Africa
| | | | - Roger Hunter
- Department of Chemistry, University of Cape Town, PD Hahn Building, Cape Town 7700, South Africa
| | - Stefan Barth
- Institute of Infectious Disease and Molecular Medicine, Medical Biotechnology and Immunotherapy Research Unit, University of Cape Town, Cape Town 7700, South Africa
- Faculty of Health Sciences, Department of Integrative Biomedical Sciences, South African Research Chair in Cancer Biotechnology, University of Cape Town, Cape Town 7700, South Africa
| |
Collapse
|
6
|
Cao Z, Zuo X, Liu X, Xu G, Yong KT. Recent progress in stimuli-responsive polymeric micelles for targeted delivery of functional nanoparticles. Adv Colloid Interface Sci 2024; 330:103206. [PMID: 38823215 DOI: 10.1016/j.cis.2024.103206] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 05/20/2024] [Accepted: 05/27/2024] [Indexed: 06/03/2024]
Abstract
Stimuli-responsive polymeric micelles have emerged as a revolutionary approach for enhancing the in vivo stability, biocompatibility, and targeted delivery of functional nanoparticles (FNPs) in biomedicine. This article comprehensively reviews the preparation methods of these polymer micelles, detailing the innovative strategies employed to introduce stimulus responsiveness and surface modifications essential for precise targeting. We delve into the breakthroughs in utilizing these micelles to selectively deliver various FNPs including magnetic nanoparticles, upconversion nanoparticles, gold nanoparticles, and quantum dots, highlighting their transformative impact in the biomedical realm. Concluding, we present an insight into the current research landscape, addressing the challenges at hand, and envisioning the future trajectory in this burgeoning domain. Join us as we navigate the exciting confluence of polymer science and nanotechnology in reshaping biomedical solutions.
Collapse
Affiliation(s)
- Zhonglin Cao
- College of Materials Science and Engineering, Guizhou Minzu University, Guiyang 550025, China
| | - Xiaoling Zuo
- College of Materials Science and Engineering, Guizhou Minzu University, Guiyang 550025, China
| | - Xiaochen Liu
- School of Biomedical Engineering, The University of Sydney, Sydney, New South Wales 2006, Australia; The Biophotonics and Mechano-Bioengineering Lab, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Gaixia Xu
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University, Shenzhen 518060, China.
| | - Ken-Tye Yong
- School of Biomedical Engineering, The University of Sydney, Sydney, New South Wales 2006, Australia; The Biophotonics and Mechano-Bioengineering Lab, The University of Sydney, Sydney, New South Wales 2006, Australia.
| |
Collapse
|
7
|
Jin J, Barnett JD, Mironchik Y, Gross J, Kobayashi H, Levin A, Bhujwalla ZM. Photoimmunotheranostics of epithelioid sarcoma by targeting CD44 or EGFR. Transl Oncol 2024; 45:101966. [PMID: 38663219 PMCID: PMC11063645 DOI: 10.1016/j.tranon.2024.101966] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 04/05/2024] [Accepted: 04/16/2024] [Indexed: 05/05/2024] Open
Abstract
Epithelioid sarcoma (ES) is a rare soft tissue neoplasm with high recurrence rates. Wide surgical resection remains the only potential curative treatment. ES presents most commonly on the fingers, hands and forearm, making light-based cancer cell-targeted therapies such as near-infrared photoimmunotherapy (NIR-PIT) that is target-specific, but with limited penetration depth, suitable for ES treatment. We established that CD44 and EGFR were overexpressed in ES patient samples and in the VA-ES-BJ human ES cell line. NIR-PIT of VA-ES-BJ cells using antibody photosensitizer conjugates, prepared by conjugating a CD44 or EGFR monoclonal antibody to the photosensitizer IR700, confirmed that NIR-PIT with both conjugates resulted in cell death. Neither treatment with NIR light alone nor treatment with the conjugates but without NIR light were effective. CD44-IR700-PIT resulted in greater cell death than EGFR-IR700-PIT, consistent with the increased expression of CD44 by VA-ES-BJ cells. In tumors, EGFR-IR700 exhibited a higher tumor-to-normal ratio, as determined by in vivo fluorescence imaging, and a higher anti-tumor growth effect, compared to CD44-IR700. No antitumor effect of the EGFR antibody or the photosensitizer conjugate alone was observed in vivo. Our data support evaluating the use of EGFR-IR700-PIT in the management of ES for detecting and eliminating ES cells in surgical margins, and in the treatment of superficial recurrent tumors.
Collapse
Affiliation(s)
- Jiefu Jin
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - James D Barnett
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yelena Mironchik
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - John Gross
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hisataka Kobayashi
- Laboratory of Molecular Theranostics, Molecular Imaging Branch, NCI/NIH, Bethesda, MD, USA
| | - Adam Levin
- Orthopaedic Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zaver M Bhujwalla
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Radiation Oncology and Molecular Radiation Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
8
|
Yang JK, Kwon H, Kim S. Recent advances in light-triggered cancer immunotherapy. J Mater Chem B 2024; 12:2650-2669. [PMID: 38353138 DOI: 10.1039/d3tb02842a] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2024]
Abstract
Light-triggered phototherapies, such as photodynamic therapy (PDT) and photothermal therapy (PTT), have shown strong therapeutic efficacy with minimal invasiveness and systemic toxicity, offering opportunities for tumor-specific therapies. Phototherapies not only induce direct tumor cell killing, but also trigger anti-tumor immune responses by releasing various immune-stimulating factors. In recent years, conventional phototherapies have been combined with cancer immunotherapy as synergistic therapeutic modalities to eradicate cancer by exploiting the innate and adaptive immunity. These combined photoimmunotherapies have demonstrated excellent therapeutic efficacy in preventing tumor recurrence and metastasis compared to phototherapy alone. This review covers recent advancements in combined photoimmunotherapy, including photoimmunotherapy (PIT), PDT-combined immunotherapy, and PTT-combined immunotherapy, along with their underlying anti-tumor immune response mechanisms. In addition, the challenges and future research directions for light-triggered cancer immunotherapy are discussed.
Collapse
Affiliation(s)
- Jin-Kyoung Yang
- Department of Chemical Engineering, Dong-eui University, Busan, 47340, Republic of Korea.
| | - Hayoon Kwon
- Chemical & Biological integrative Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea.
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Sehoon Kim
- Chemical & Biological integrative Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea.
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| |
Collapse
|
9
|
Nakajima K, Ogawa M. Near-infrared photoimmunotherapy and anti-cancer immunity. Int Immunol 2024; 36:57-64. [PMID: 37843836 DOI: 10.1093/intimm/dxad042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 10/14/2023] [Indexed: 10/17/2023] Open
Abstract
The activation of the anti-cancer immune system is an important strategy to control cancer. A new form of cancer phototherapy, near-infrared photoimmunotherapy (NIR-PIT), was approved for clinical use in 2020 and uses IRDye® 700DX (IR700)-conjugated antibodies and NIR light. After irradiation with NIR light, the antibody-IR700 conjugate forms water-insoluble aggregations on the plasma membrane of target cells. This aggregation causes lethal damage to the plasma membrane, and effectively leads to immunogenic cell death (ICD). Subsequently, ICD activates anti-cancer immune cells such as dendritic cells and cytotoxic T cells. Combination therapy with immune-checkpoint blockade has synergistically improved the anti-cancer effects of NIR-PIT. Additionally, NIR-PIT can eliminate immunosuppressive immune cells in light-irradiated tumors by using specific antibodies against regulatory T cells and myeloid-derived suppressor cells. In addition to cancer-cell-targeted NIR-PIT, such immune-cell-targeted NIR-PIT has shown promising results by activating the anti-cancer immune system. Furthermore, NIR-PIT can be used to manipulate the tumor microenvironment by eliminating only targeted cells in the tumor, and thus it also can be used to gain insight into immunity in basic research.
Collapse
Affiliation(s)
- Kohei Nakajima
- Laboratory of Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido 060-0812, Japan
| | - Mikako Ogawa
- Laboratory of Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido 060-0812, Japan
| |
Collapse
|
10
|
Lee CE, Kim S, Park HW, Lee W, Jangid AK, Choi Y, Jeong WJ, Kim K. Tailoring tumor-recognizable hyaluronic acid-lipid conjugates to enhance anticancer efficacies of surface-engineered natural killer cells. NANO CONVERGENCE 2023; 10:56. [PMID: 38097911 PMCID: PMC10721593 DOI: 10.1186/s40580-023-00406-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 12/01/2023] [Indexed: 12/18/2023]
Abstract
Natural killer (NK) cells have clinical advantages in adoptive cell therapy owing to their inherent anticancer efficacy and their ability to identify and eliminate malignant tumors. However, insufficient cancer-targeting ligands on NK cell surfaces often inhibit their immunotherapeutic performance, especially in immunosuppressive tumor microenvironment. To facilitate tumor recognition and subsequent anticancer function of NK cells, we developed hyaluronic acid (HA, ligands to target CD44 overexpressed onto cancer cells)-poly(ethylene glycol) (PEG, cytoplasmic penetration blocker)-Lipid (molecular anchor for NK cell membrane decoration through hydrophobic interaction) conjugates for biomaterial-mediated ex vivo NK cell surface engineering. Among these major compartments (i.e., Lipid, PEG and HA), optimization of lipid anchors (in terms of chemical structure and intrinsic amphiphilicity) is the most important design parameter to modulate hydrophobic interaction with dynamic NK cell membranes. Here, three different lipid types including 1,2-dimyristoyl-sn-glycero-3-phosphati-dylethanolamine (C14:0), 1,2-distearoyl-sn-glycero-3-phosphatidylethanolamine (DSPE, C18:0), and cholesterol were evaluated to maximize membrane coating efficacy and associated anticancer performance of surface-engineered NK cells (HALipid-NK cells). Our results demonstrated that NK cells coated with HA-PEG-DSPE conjugates exhibited significantly enhanced anticancer efficacies toward MDA-MB-231 breast cancer cells without an off-target effect on human fibroblasts specifically via increased NK cell membrane coating efficacy and prolonged surface duration of HA onto NK cell surfaces, thereby improving HA-CD44 recognition. These results suggest that our HALipid-NK cells with tumor-recognizable HA-PEG-DSPE conjugates could be further utilized in various cancer immunotherapies.
Collapse
Affiliation(s)
- Chae Eun Lee
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul, 04620, Republic of Korea
| | - Sungjun Kim
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul, 04620, Republic of Korea
| | - Hee Won Park
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul, 04620, Republic of Korea
| | - Wonjeong Lee
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul, 04620, Republic of Korea
| | - Ashok Kumar Jangid
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul, 04620, Republic of Korea
| | - Yonghyun Choi
- Department of Chemical Science and Engineering, Tokyo Institute of Technology, Kanagawa, 226-8501, Japan
| | - Woo-Jin Jeong
- Department of Biological Engineering, Inha University, Incheon, 22212, Republic of Korea.
| | - Kyobum Kim
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul, 04620, Republic of Korea.
| |
Collapse
|
11
|
Mungra N, Biteghe FAN, Malindi Z, Huysamen AM, Karaan M, Hardcastle NS, Bunjun R, Chetty S, Naran K, Lang D, Richter W, Hunter R, Barth S. CSPG4 as a target for the specific killing of triple-negative breast cancer cells by a recombinant SNAP-tag-based antibody-auristatin F drug conjugate. J Cancer Res Clin Oncol 2023; 149:12203-12225. [PMID: 37432459 PMCID: PMC10465649 DOI: 10.1007/s00432-023-05031-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 06/27/2023] [Indexed: 07/12/2023]
Abstract
PURPOSE Triple-negative breast cancer (TNBC) is phenotypic of breast tumors lacking expression of the estrogen receptor (ER), the progesterone receptor (PgR), and the human epidermal growth factor receptor 2 (HER2). The paucity of well-defined molecular targets in TNBC, coupled with the increasing burden of breast cancer-related mortality, emphasizes the need to develop targeted diagnostics and therapeutics. While antibody-drug conjugates (ADCs) have emerged as revolutionary tools in the selective delivery of drugs to malignant cells, their widespread clinical use has been hampered by traditional strategies which often give rise to heterogeneous mixtures of ADC products. METHODS Utilizing SNAP-tag technology as a cutting-edge site-specific conjugation method, a chondroitin sulfate proteoglycan 4 (CSPG4)-targeting ADC was engineered, encompassing a single-chain antibody fragment (scFv) conjugated to auristatin F (AURIF) via a click chemistry strategy. RESULTS After showcasing the self-labeling potential of the SNAP-tag component, surface binding and internalization of the fluorescently labeled product were demonstrated on CSPG4-positive TNBC cell lines through confocal microscopy and flow cytometry. The cell-killing ability of the novel AURIF-based recombinant ADC was illustrated by the induction of a 50% reduction in cell viability at nanomolar to micromolar concentrations on target cell lines. CONCLUSION This research underscores the applicability of SNAP-tag in the unambiguous generation of homogeneous and pharmaceutically relevant immunoconjugates that could potentially be instrumental in the management of a daunting disease like TNBC.
Collapse
Affiliation(s)
- Neelakshi Mungra
- Institute of Infectious Disease and Molecular Medicine, Medical Biotechnology and Immunotherapy Research Unit, University of Cape Town, Cape Town, 7700 South Africa
- Centre for Immunity and Immunotherapies, Seattle Children’s Research Institute, Washington, 98101 USA
| | - Fleury A. N. Biteghe
- Department of Radiation Oncology and Biomedical Sciences, Cedars-Sinai Medical, Los Angeles, USA
| | - Zaria Malindi
- Institute of Infectious Disease and Molecular Medicine, Medical Biotechnology and Immunotherapy Research Unit, University of Cape Town, Cape Town, 7700 South Africa
- Faculty of Health Sciences, Laser Research Centre, University of Johannesburg, Doornfontein, Johannesburg, 2028 South Africa
| | - Allan M. Huysamen
- Department of Chemistry, PD Hahn Building, University of Cape Town, Cape Town, 7700 South Africa
| | - Maryam Karaan
- Institute of Infectious Disease and Molecular Medicine, Medical Biotechnology and Immunotherapy Research Unit, University of Cape Town, Cape Town, 7700 South Africa
| | - Natasha S. Hardcastle
- Institute of Infectious Disease and Molecular Medicine, Medical Biotechnology and Immunotherapy Research Unit, University of Cape Town, Cape Town, 7700 South Africa
| | - Rubina Bunjun
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, 7700 South Africa
- Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, 7700 South Africa
| | - Shivan Chetty
- Faculty of Health Sciences, School of Clinical Medicine, University of Witwatersrand, Braamfontein, Johannesburg, 2000 South Africa
| | - Krupa Naran
- Institute of Infectious Disease and Molecular Medicine, Medical Biotechnology and Immunotherapy Research Unit, University of Cape Town, Cape Town, 7700 South Africa
| | - Dirk Lang
- Division of Physiological Sciences, Department of Human Biology, University of Cape Town, Cape Town, 7700 South Africa
| | | | - Roger Hunter
- Department of Chemistry, PD Hahn Building, University of Cape Town, Cape Town, 7700 South Africa
| | - Stefan Barth
- Institute of Infectious Disease and Molecular Medicine, Medical Biotechnology and Immunotherapy Research Unit, University of Cape Town, Cape Town, 7700 South Africa
- Faculty of Health Sciences, Department of Integrative Biomedical Sciences, South African Research Chair in Cancer Biotechnology, University of Cape Town, Cape Town, 7700 South Africa
| |
Collapse
|
12
|
Cui Y, Xu Y, Li Y, Sun Y, Hu J, Jia J, Li X. Antibody Drug Conjugates of Near-Infrared Photoimmunotherapy (NIR-PIT) in Breast Cancers. Technol Cancer Res Treat 2023; 22:15330338221145992. [PMID: 36734039 PMCID: PMC9903039 DOI: 10.1177/15330338221145992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Worldwide, the incidence rate of breast cancer is the highest in women. Approximately 2.3 million people were newly diagnosed and 0.685 million were dead of breast cancer in 2020, which continues to grow. Triple-negative breast cancer (TNBC) is the most aggressive breast cancer subtype with a higher risk of recurrence and metastasis, but disappointly, there are no effective and specific therapies clinically, especially for patients presenting with metastatic diseases. Therefore, it is urgent to develop a new type of cancer therapy for survival improvisation and adverse effects alleviation of breast cancers. Near-infrared photoimmunotherapy (NIR-PIT) is a newly developed, photochemistry-based cancer therapy. It was drive by an antibody-photoabsorber conjugate (APC) which is triggered by near-infrared light. The key part of APC is a cancer-targeting monoclonal antibody (mAb) that can bind to receptors or antigens on the surface of tumor cells. Because of this targeted conjugate accumulation, subsequent deployment of focal NIR-light results in functional damage on the targeted cell membranes without harming the immediately adjacent receptor-negative cells and evokes a kind of photochemical, speedy, and highly specific immunogenic cell death (ICD) of cancer cells with corresponding antigens. Subsequently, immature dendritic cells adjacent to dying cancer cells will become mature, further inducing a host-oriented anti-cancer immune response, complicatedly and comprehensively. Currently, NIR-PIT has progressed into phase 3 clinical trial for recurrent head and neck cancer. And preclinical studies have illustrated strong therapeutic efficacy of NIR-PIT targeting various molecular receptors overexpressed in breast cancer cells, including EGFR, HER2, CD44c, CD206, ICAM-1 and FAP-α. Thereby, NIR-PIT is in early trials, but appears to be a promising breast cancer therapy and moving into the future. Here, we present the specific advantages and discuss the most recent preclinical studies against several transmembrane proteins of NIR-PIT in breast cancers.
Collapse
Affiliation(s)
- Yingshu Cui
- Department of Oncology, the Fifth Medical Center, Chinese PLA General Hospital, Beijing, China,Medical School of Chinese PLA, Beijing, China
| | - Yuanyuan Xu
- Department of Oncology, the Fifth Medical Center, Chinese PLA General Hospital, Beijing, China,Department of Laser, the First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yi Li
- Medical School of Chinese PLA, Beijing, China,Department of Laser, the First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yuanyuan Sun
- Department of Oncology, the Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jia Hu
- Department of Oncology, the Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jia Jia
- Department of Oncology, the Seventh Medical Center, Chinese PLA General Hospital, Beijing, China,Jia Jia, Department of Oncology, the Seventh Medical Centre, Chinese PLA General Hospital, Beijing 100700, China.
| | - Xiaosong Li
- Department of Oncology, the Fifth Medical Center, Chinese PLA General Hospital, Beijing, China,Xiaosong Li, Department of Oncology, the Fifth Medical Center, Chinese PLA General Hospital, Beijing 100071, China.
| |
Collapse
|
13
|
Jin J, Barnett JD, Krishnamachary B, Mironchik Y, Luo CK, Kobayashi H, Bhujwalla ZM. Evaluating near-infrared photoimmunotherapy for targeting fibroblast activation protein-α expressing cells in vitro and in vivo. Cancer Sci 2023; 114:236-246. [PMID: 36169301 PMCID: PMC9807523 DOI: 10.1111/cas.15601] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 08/25/2022] [Accepted: 09/02/2022] [Indexed: 01/07/2023] Open
Abstract
Photoimmunotherapy (PIT), carried out using an Ab conjugated to the near infrared dye IRDye700DX, is achieving significant success in target-specific elimination of cells. Fibroblast activation protein alpha (FAP-α) is an important target in cancer because of its expression by cancer-associated fibroblasts (CAFs) as well as by some cancer cells. Cancer-associated fibroblasts that express FAP-α have protumorigenic and immune suppressive functions. Using immunohistochemistry of human breast cancer tissue microarrays, we identified an increase of FAP-α+ CAFs in invasive breast cancer tissue compared to adjacent normal tissue. We found FAP-α expression increased in fibroblasts cocultured with cancer cells. In proof-of-principle studies, we engineered human FAP-α overexpressing MDA-MB-231 and HT-1080 cancer cells and murine FAP-α overexpressing NIH-3T3 fibroblasts to evaluate several anti-FAP-α Abs and selected AF3715 based on its high binding affinity with both human and mouse FAP-α. After conjugation of AF3715 with the phthalocyanine dye IR700, the resultant Ab conjugate, FAP-α-IR700, was evaluated in cells and tumors for its specificity and effectiveness in eliminating FAP-α expressing cell populations with PIT. Fibroblast activation protein-α-IR700-PIT resulted in effective FAP-α-specific cell killing in the engineered cancer cells and in two patient-derived CAFs in a dose-dependent manner. Following an intravenous injection, FAP-α-IR700 retention was three-fold higher than IgG-IR700 in FAP-α overexpressing tumors, and two-fold higher compared to WT tumors. Fibroblast activation protein-α-IR700-PIT resulted in significant growth inhibition of tumors derived from FAP-α overexpressing human cancer cells. A reduction of endogenous FAP-α+ murine CAFs was identified at 7 days after FAP-α-IR700-PIT. Fibroblast activation protein-α-targeted near infrared PIT presents a promising strategy to eliminate FAP-α+ CAFs.
Collapse
Affiliation(s)
- Jiefu Jin
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, Baltimore, Maryland, USA
| | - James D Barnett
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, Baltimore, Maryland, USA
| | - Balaji Krishnamachary
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, Baltimore, Maryland, USA
| | - Yelena Mironchik
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, Baltimore, Maryland, USA
| | - Catherine K Luo
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, Baltimore, Maryland, USA
| | - Hisataka Kobayashi
- Laboratory of Molecular Theranostics Molecular Imaging Branch, NCI/NIH, Bethesda, Maryland, USA
| | - Zaver M Bhujwalla
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, Baltimore, Maryland, USA.,Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Radiation Oncology and Molecular Radiation Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
14
|
Pramanik A, Xu Z, Ingram N, Coletta PL, Millner PA, Tyler AII, Hughes TA. Hyaluronic-Acid-Tagged Cubosomes Deliver Cytotoxics Specifically to CD44-Positive Cancer Cells. Mol Pharm 2022; 19:4601-4611. [PMID: 35938983 PMCID: PMC9727730 DOI: 10.1021/acs.molpharmaceut.2c00439] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Delivery of chemotherapy drugs specifically to cancer cells raises local drug doses in tumors and therefore kills more cancer cells while reducing side effects in other tissues, thereby improving oncological and quality of life outcomes. Cubosomes, liquid crystalline lipid nanoparticles, are potential vehicles for delivery of chemotherapy drugs, presenting the advantages of biocompatibility, stable encapsulation, and high drug loading of hydrophobic or hydrophilic drugs. However, active targeting of drug-loaded cubosomes to cancer cells, as opposed to passive accumulation, remains relatively underexplored. We formulated and characterized cubosomes loaded with potential cancer drug copper acetylacetonate and functionalized their surfaces using click chemistry coupling with hyaluronic acid (HA), the ligand for the cell surface receptor CD44. CD44 is overexpressed in many cancer types including breast and colorectal. HA-tagged, copper-acetylacetonate-loaded cubosomes have an average hydrodynamic diameter of 152 nm, with an internal nanostructure based on the space group Im3m. These cubosomes were efficiently taken up by two CD44-expressing cancer cell lines (MDA-MB-231 and HT29, representing breast and colon cancer) but not by two CD44-negative cell lines (MCF-7 breast cancer and HEK-293 kidney cells). HA-tagged cubosomes caused significantly more cell death than untargeted cubosomes in the CD44-positive cells, demonstrating the value of the targeting. CD44-negative cells were equally relatively resistant to both, demonstrating the specificity of the targeting. Cell death was characterized as apoptotic. Specific targeting and cell death were evident in both 2D culture and 3D spheroids. We conclude that HA-tagged, copper-acetylacetonate-loaded cubosomes show great potential as an effective therapeutic for selective targeting of CD44-expressing tumors.
Collapse
Affiliation(s)
- Arindam Pramanik
- School
of Medicine, University of Leeds, Leeds LS2 9JT, United Kingdom,
| | - Zexi Xu
- School
of Food Science and Nutrition, University
of Leeds, Leeds LS2 9JT, United Kingdom
| | - Nicola Ingram
- School
of Medicine, University of Leeds, Leeds LS2 9JT, United Kingdom
| | | | - Paul A Millner
- School
of Biomedical Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Arwen I I Tyler
- School
of Food Science and Nutrition, University
of Leeds, Leeds LS2 9JT, United Kingdom,
| | - Thomas A Hughes
- School
of Medicine, University of Leeds, Leeds LS2 9JT, United Kingdom,
| |
Collapse
|
15
|
Otohinoyi D, Kuchi A, Wu J, Hicks C. Integrating Genomic Information with Tumor-Immune Microenvironment in Triple-Negative Breast Cancer. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph192113901. [PMID: 36360779 PMCID: PMC9659069 DOI: 10.3390/ijerph192113901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/21/2022] [Accepted: 10/22/2022] [Indexed: 05/14/2023]
Abstract
BACKGROUND the development and progression of triple-negative breast cancer (TNBC) is driven by somatic driver mutations and the tumor-immune microenvironment. To date, data on somatic mutations has not been leveraged and integrated with information on the immune microenvironment to elucidate the possible oncogenic interactions and their potential effects on clinical outcomes. Here, we investigated possible oncogenic interactions between somatic mutations and the tumor-immune microenvironment, and their correlation with patient survival in TNBC. METHODS We performed analysis combining data on 7,875 somatic mutated genes with information on 1,751 immune-modulated genes, using gene-expression data as the intermediate phenotype, and correlated the resulting information with survival. We conducted functional analysis to identify immune-modulated molecular networks and signaling pathways enriched for somatic mutations likely to drive clinical outcomes. RESULTS We discovered differences in somatic mutation profiles between patients who died and those who survived, and a signature of somatic mutated immune-modulated genes transcriptionally associated with TNBC, predictive of survival. In addition, we discovered immune-modulated molecular networks and signaling pathways enriched for somatic mutations. CONCLUSIONS The investigation revealed possible oncogenic interactions between somatic mutations and the tumor-immune microenvironment in TNBC, likely to affect clinical outcomes.
Collapse
|
16
|
Wei D, Qi J, Hamblin MR, Wen X, Jiang X, Yang H. Near-infrared photoimmunotherapy: design and potential applications for cancer treatment and beyond. Am J Cancer Res 2022; 12:7108-7131. [PMID: 36276636 PMCID: PMC9576624 DOI: 10.7150/thno.74820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 09/28/2022] [Indexed: 11/22/2022] Open
Abstract
Near-infrared photoimmunotherapy (NIR-PIT) is a newly developed cancer treatment modality based on a target-specific photosensitizer conjugate (TSPC) composed of an NIR phthalocyanine photosensitizer and an antigen-specific recognition system. NIR-PIT has predominantly been used for targeted therapy of tumors via local irradiation with NIR light, following binding of TSPC to antigen-expressing cells. Physical stress-induced membrane damage is thought to be a major mechanism underlying NIR-PIT-triggered photokilling. Notably, NIR-PIT can rapidly induce immunogenic cell death and activate the adaptive immune response, thereby enabling its combination with immune checkpoint inhibitors. Furthermore, NIR-PIT-triggered “super-enhanced permeability and retention” effects can enhance drug delivery into tumors. Supported by its potential efficacy and safety, NIR-PIT is a rapidly developing therapeutic option for various cancers. Hence, this review seeks to provide an update on the (i) broad range of target molecules suitable for NIR-PIT, (ii) various types of receptor-selective ligands for designing the TSPC “magic bullet,” (iii) NIR light parameters, and (iv) strategies for enhancing the efficacy of NIR-PIT. Moreover, we review the potential application of NIR-PIT, including the specific design and efficacy in 19 different cancer types, and its clinical studies. Finally, we summarize possible NIR-PIT applications in noncancerous conditions, including infection, pain, itching, metabolic disease, autoimmune disease, and tissue engineering.
Collapse
Affiliation(s)
- Danfeng Wei
- Department of Dermatology and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China.,Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network West China Hospital, Sichuan University, Chengdu 610041, China.,NHC Key Lab of Transplant Engineering and Immunology, Organ Transplant Center, West China Hospital, Sichuan University, Chengdu, Chengdu 610041, China
| | - Jinxin Qi
- Department of Dermatology and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China.,Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network West China Hospital, Sichuan University, Chengdu 610041, China
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa
| | - Xiang Wen
- Department of Dermatology and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xian Jiang
- Department of Dermatology and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China.,Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hao Yang
- NHC Key Lab of Transplant Engineering and Immunology, Organ Transplant Center, West China Hospital, Sichuan University, Chengdu, Chengdu 610041, China.,Sichuan Provincial Engineering Laboratory of Pathology in Clinical Application, West China Hospital, Sichuan University
| |
Collapse
|
17
|
Inagaki FF, Kato T, Furusawa A, Okada R, Wakiyama H, Furumoto H, Okuyama S, Choyke PL, Kobayashi H. Disialoganglioside GD2-Targeted Near-Infrared Photoimmunotherapy (NIR-PIT) in Tumors of Neuroectodermal Origin. Pharmaceutics 2022; 14:2037. [PMID: 36297471 PMCID: PMC9612122 DOI: 10.3390/pharmaceutics14102037] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/18/2022] [Accepted: 09/22/2022] [Indexed: 10/21/2023] Open
Abstract
Disialoganglioside (GD2) is a subtype of glycolipids that is highly expressed in tumors of neuroectodermal origins, such as neuroblastoma and osteosarcoma. Its limited expression in normal tissues makes GD2 a potential target for precision therapy. Several anti-GD2 monoclonal antibodies are currently in clinical use and have had moderate success. Near-infrared photoimmunotherapy (NIR-PIT) is a cancer therapy that arms antibodies with IRDye700DX (IR700) and then exposes this antibody-dye conjugate (ADC) to NIR light at a wavelength of 690 nm. NIR light irradiation induces a profound photochemical response in IR700, resulting in protein aggregates that lead to cell membrane damage and death. In this study, we examined the feasibility of GD2-targeted NIR-PIT. Although GD2, like other glycolipids, is only located in the outer leaflet of the cell membrane, the aggregates formation exerted sufficient physical force to disrupt the cell membrane and kill target cells in vitro. In in vivo studies, tumor growth was significantly inhibited after GD2-targeted NIR-PIT, resulting in prolonged survival. Following GD2-targeted NIR-PIT, activation of host immunity was observed. In conclusion, GD2-targeted NIR-PIT was similarly effective to the conventional protein-targeted NIR-PIT. This study demonstrates that membrane glycolipid can be a new target of NIR-PIT.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Hisataka Kobayashi
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
18
|
Near-Infrared Photoimmunotherapy for Thoracic Cancers: A Translational Perspective. Biomedicines 2022; 10:biomedicines10071662. [PMID: 35884975 PMCID: PMC9312913 DOI: 10.3390/biomedicines10071662] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/23/2022] [Accepted: 07/07/2022] [Indexed: 12/18/2022] Open
Abstract
The conventional treatment of thoracic tumors includes surgery, anticancer drugs, radiation, and cancer immunotherapy. Light therapy for thoracic tumors has long been used as an alternative; conventional light therapy also called photodynamic therapy (PDT) has been used mainly for early-stage lung cancer. Recently, near-infrared photoimmunotherapy (NIR-PIT), which is a completely different concept from conventional PDT, has been developed and approved in Japan for the treatment of recurrent and previously treated head and neck cancer because of its specificity and effectiveness. NIR-PIT can apply to any target by changing to different antigens. In recent years, it has become clear that various specific and promising targets are highly expressed in thoracic tumors. In combination with these various specific targets, NIR-PIT is expected to be an ideal therapeutic approach for thoracic tumors. Additionally, techniques are being developed to further develop NIR-PIT for clinical practice. In this review, NIR-PIT is introduced, and its potential therapeutic applications for thoracic cancers are described.
Collapse
|
19
|
Mussini A, Uriati E, Bianchini P, Diaspro A, Cavanna L, Abbruzzetti S, Viappiani C. Targeted photoimmunotherapy for cancer. Biomol Concepts 2022; 13:126-147. [PMID: 35304984 DOI: 10.1515/bmc-2022-0010] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 02/24/2022] [Indexed: 12/12/2022] Open
Abstract
Photodynamic therapy (PDT) is a clinically approved procedure that can exert a curative action against malignant cells. The treatment implies the administration of a photoactive molecular species that, upon absorption of visible or near infrared light, sensitizes the formation of reactive oxygen species. These species are cytotoxic and lead to tumor cell death, damage vasculature, and induce inflammation. Clinical investigations demonstrated that PDT is curative and does not compromise other treatment options. One of the major limitations of the original method was the low selectivity of the photoactive compounds for malignant over healthy tissues. The development of conjugates with antibodies has endowed photosensitizing molecules with targeting capability, so that the compounds are delivered with unprecedented precision to the site of action. Given their fluorescence emission capability, these supramolecular species are intrinsically theranostic agents.
Collapse
Affiliation(s)
- Andrea Mussini
- Dipartimento di Scienze Matematiche, Fisiche e Informatiche, Università Degli Studi di Parma, Parma, Italy
| | - Eleonora Uriati
- Dipartimento di Scienze Matematiche, Fisiche e Informatiche, Università Degli Studi di Parma, Parma, Italy.,Department of Nanophysics, Nanoscopy, Istituto Italiano di Tecnologia, Genova, Italy
| | - Paolo Bianchini
- Dipartimento di Scienze Matematiche, Fisiche e Informatiche, Università Degli Studi di Parma, Parma, Italy.,Department of Nanophysics, Nanoscopy, Istituto Italiano di Tecnologia, Genova, Italy.,DIFILAB, Dipartimento di Fisica, Università Degli Studi di Genova, Genova, Italy
| | - Alberto Diaspro
- Department of Nanophysics, Nanoscopy, Istituto Italiano di Tecnologia, Genova, Italy.,DIFILAB, Dipartimento di Fisica, Università Degli Studi di Genova, Genova, Italy
| | - Luigi Cavanna
- Dipartimento di Oncologia-Ematologia, Azienda USL di Piacenza, Piacenza, Italy
| | - Stefania Abbruzzetti
- Dipartimento di Scienze Matematiche, Fisiche e Informatiche, Università Degli Studi di Parma, Parma, Italy
| | - Cristiano Viappiani
- Dipartimento di Scienze Matematiche, Fisiche e Informatiche, Università Degli Studi di Parma, Parma, Italy
| |
Collapse
|
20
|
PD-L1 near Infrared Photoimmunotherapy of Ovarian Cancer Model. Cancers (Basel) 2022; 14:cancers14030619. [PMID: 35158887 PMCID: PMC8833482 DOI: 10.3390/cancers14030619] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 01/22/2022] [Indexed: 12/14/2022] Open
Abstract
(1) Background: Despite advances in surgical approaches and drug development, ovarian cancer is still a leading cause of death from gynecological malignancies. Patients diagnosed with late-stage disease are treated with aggressive surgical resection and chemotherapy, but recurrence with resistant disease is often observed following treatment. There is a critical need for effective therapy for late-stage ovarian cancer. Photoimmunotherapy (PIT), using an antibody conjugated to a near infrared (NIR) dye, constitutes an effective theranostic strategy to detect and selectively eliminate targeted cell populations. (2) Methods: Here, we are targeting program death ligand 1 (PD-L1) using NIR-PIT in a syngeneic mouse model of ovarian cancer. PD-L1 PIT-mediated cytotoxicity was quantified in RAW264.7 macrophages and ID8-Defb29-VEGF cells in culture, and in vivo with orthotopic ID8-Defb29-VEGF tumors. (3) Results: Treatment efficacy was observed both in vitro and in vivo. (4) Conclusions: Our data highlight the need for further investigations to assess the potential of using NIR-PIT for ovarian cancer therapy to improve the treatment outcome of ovarian cancer.
Collapse
|
21
|
Furusawa A, Okada R, Inagaki F, Wakiyama H, Kato T, Furumoto H, Fukushima H, Okuyama S, Choyke PL, Kobayashi H. CD29 targeted near-infrared photoimmunotherapy (NIR-PIT) in the treatment of a pigmented melanoma model. Oncoimmunology 2022; 11:2019922. [PMID: 35003897 PMCID: PMC8741294 DOI: 10.1080/2162402x.2021.2019922] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 12/14/2021] [Accepted: 12/14/2021] [Indexed: 10/25/2022] Open
Abstract
Near-infrared photoimmunotherapy (NIR-PIT) is a newly developed cancer treatment that utilizes an antibody-photoabsorber-conjugate (AbPC) combined with NIR light. The AbPC is injected and binds to the tumor whereupon NIR light irradiation causes a photochemical reaction that selectively kills cancer cells. NIR-PIT is ideal for surface-located skin cancers such as melanoma. However, there is concern that the pigment in melanoma lesions could interfere with light delivery, rendering treatment ineffective. We investigated the efficacy of CD29- and CD44-targeted NIR-PIT (CD29-PIT and CD44-PIT, respectively) in the B16 melanoma model, which is highly pigmented. While CD29-PIT and CD44-PIT killed B16 cells invitro and invivo, CD29-PIT suppressed tumor growth more efficiently. Ki67 expression showed that cells surviving CD29-PIT were less proliferative, suggesting that CD29-PIT was selective for more proliferative cancer cells. CD29-PIT did not kill immune cells, whereas CD44-PIT killed both T and NK cells and most myeloid cells, including DCs, which could interfere with the immune response to NIR-PIT. The addition of anti-CTLA4 antibody immune checkpoint inhibitor (ICI) to CD29-PIT increased the infiltration of CD8 T cells and enhanced tumor suppression with prolonged survival. Such effects were less prominent when the anti-CTLA4 ICI was combined with CD44-PIT. The preservation of immune cells in the tumor microenvironment (TME) after CD29-PIT likely led to a better response when combined with anti-CTLA4 treatment. We conclude that NIR-PIT can be performed in pigmented melanomas and that CD29 is a promising target for NIR-PIT, which is amenable to combination therapy with other immunotherapies.
Collapse
Affiliation(s)
- Aki Furusawa
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Ryuhei Okada
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Fuyuki Inagaki
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Hiroaki Wakiyama
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Takuya Kato
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Hideyuki Furumoto
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Hiroshi Fukushima
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Shuhei Okuyama
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Peter L. Choyke
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Hisataka Kobayashi
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
22
|
Russo I, Fagotto L, Colombo A, Sartor E, Luisetto R, Alaibac M. Near-infrared photoimmunotherapy for the treatment of skin disorders. Expert Opin Biol Ther 2021; 22:509-517. [PMID: 34860146 DOI: 10.1080/14712598.2022.2012147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Near-Infrared Photoimmunotherapy (NIR-PIT) is a novel molecularly targeted phototherapy. This technique is based on a conjugate of a near-infrared photo-inducible molecule (antibody-photon absorber conjugate, APC) and a monoclonal antibody that targets a tumor-specific antigen. To date, this novel approach has been successfully applied to several types of cancer. AREAS COVERED The authors discuss the possible use of NIR-PIT for the management of skin diseases, with special attention given to squamous cell carcinomas, advanced melanomas, and primary cutaneous lymphomas. EXPERT OPINION NIR-PIT may be an attractive strategy for the treatment of skin disorders. The main advantage of NIR-PIT therapy is its low toxicity to healthy tissues. Cutaneous lymphocyte antigen is a potential molecular target for NIR-PIT for both cutaneous T-cell lymphomas and inflammatory skin disorders.
Collapse
Affiliation(s)
- Irene Russo
- Unit of Dermatology, University of Padua, Padova, Italy
| | - Laura Fagotto
- Unit of Dermatology, University of Padua, Padova, Italy
| | - Anna Colombo
- Unit of Dermatology, University of Padua, Padova, Italy
| | - Emma Sartor
- Unit of Dermatology, University of Padua, Padova, Italy
| | - Roberto Luisetto
- DISCOG-Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Mauro Alaibac
- Unit of Dermatology, University of Padua, Padova, Italy
| |
Collapse
|
23
|
Umar SM, Patra S, Kashyap A, Dev J R A, Kumar L, Prasad CP. Quercetin Impairs HuR-Driven Progression and Migration of Triple Negative Breast Cancer (TNBC) Cells. Nutr Cancer 2021; 74:1497-1510. [PMID: 34278888 DOI: 10.1080/01635581.2021.1952628] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
In the present study, we have explored the prognostic value of HuR gene as well as protein in breast cancers. Furthermore, we have also investigated the HuR therapeutic relevance in TNBCs, which is an aggressive breast cancer subtype. Using an online meta-analysis tool, we found that HuR protein overexpression positively correlates with reduced overall survival of TNBC patients (p = 0.028). Furthermore, we demonstrated that the TNBC breast cancer cell lines i.e., MDA-MB-231 and MDA-MB-468 are good model systems to study HuR protein, as they both exhibit a significant amount of cytoplasmic HuR (active form). Quercetin treatment significantly inhibited the cytoplasmic HuR in both TNBC cell lines. By using specific HuR siRNA, we established that quercetin-mediated inhibition of adhesion and migration of TNBC cells is dependent on HuR. Upon analyzing adhesion proteins i.e., β-catenin and CD44, we found that quercetin mediated effect on TNBC adhesion and migration was through the HuR-β-catenin axis and CD44, independently. Overall, the present results demonstrate that elevated HuR levels are associated with TNBC progression and relapse, and the ability of quercetin to inhibit cytoplasmic HuR protein provides a rationale for using it as an anticancer agent for the treatment of aggressive TNBCs.Supplemental data for this article is available online at at 10.1080/01635581.2021.1952628.
Collapse
Affiliation(s)
| | - Sushmita Patra
- Department of Medical Oncology, Dr. BRA IRCH, AIIMS, New Delhi, India
| | - Akanksha Kashyap
- Department of Medical Oncology, Dr. BRA IRCH, AIIMS, New Delhi, India
| | | | - Lalit Kumar
- Department of Medical Oncology, Dr. BRA IRCH, AIIMS, New Delhi, India
| | | |
Collapse
|
24
|
Ding Y, Yang R, Yu W, Hu C, Zhang Z, Liu D, An Y, Wang X, He C, Liu P, Tang Q, Chen D. Chitosan oligosaccharide decorated liposomes combined with TH302 for photodynamic therapy in triple negative breast cancer. J Nanobiotechnology 2021; 19:147. [PMID: 34011362 PMCID: PMC8136194 DOI: 10.1186/s12951-021-00891-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 05/11/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Triple negative breast cancer (TNBC) is an aggressive tumor with extremely high mortality that results from its lack of effective therapeutic targets. As an adhesion molecule related to tumorigenesis and tumor metastasis, cluster of differentiation-44 (also known as CD44) is overexpressed in TNBC. Moreover, CD44 can be effectively targeted by a specific hyaluronic acid analog, namely, chitosan oligosaccharide (CO). In this study, a CO-coated liposome was designed, with Photochlor (HPPH) as the 660 nm light mediated photosensitizer and evofosfamide (also known as TH302) as the hypoxia-activated prodrug. The obtained liposomes can help diagnose TNBC by fluorescence imaging and produce antitumor therapy by synergetic photodynamic therapy (PDT) and chemotherapy. RESULTS Compared with the nontargeted liposomes, the targeted liposomes exhibited good biocompatibility and targeting capability in vitro; in vivo, the targeted liposomes exhibited much better fluorescence imaging capability. Additionally, liposomes loaded with HPPH and TH302 showed significantly better antitumor effects than the other monotherapy groups both in vitro and in vivo. CONCLUSION The impressive synergistic antitumor effects, together with the superior fluorescence imaging capability, good biocompatibility and minor side effects confers the liposomes with potential for future translational research in the diagnosis and CD44-overexpressing cancer therapy, especially TNBC.
Collapse
Affiliation(s)
- Yinan Ding
- Medical School of Southeast University, Nanjing, 210009, China
| | - Rui Yang
- Research Institute for Reproductive Health and Genetic Diseases, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi, 214002, China
| | - Weiping Yu
- Medical School of Southeast University, Nanjing, 210009, China
| | - Chunmei Hu
- Department of Tuberculosis, The Second Affiliated Hospital of Southeast University (The Second Hospital of Nanjing), Nanjing, 210009, China
| | - Zhiyuan Zhang
- Department of Neurosurgery, Nanjing Jinling Hospital, Nanjing University, Nanjing, 210002, China
| | - Dongfang Liu
- Medical School of Southeast University, Nanjing, 210009, China
| | - Yanli An
- Affiliated Zhongda Hospital of Southeast University, Nanjing, 210009, China
| | - Xihui Wang
- Medical School of Southeast University, Nanjing, 210009, China
| | - Chen He
- Medical School of Southeast University, Nanjing, 210009, China
| | - Peidang Liu
- Medical School of Southeast University, Nanjing, 210009, China
| | - Qiusha Tang
- Medical School of Southeast University, Nanjing, 210009, China.
| | - Daozhen Chen
- Research Institute for Reproductive Health and Genetic Diseases, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi, 214002, China.
| |
Collapse
|
25
|
Paraboschi I, Turnock S, Kramer-Marek G, Musleh L, Barisa M, Anderson J, Giuliani S. Near-InfraRed PhotoImmunoTherapy (NIR-PIT) for the local control of solid cancers: Challenges and potentials for human applications. Crit Rev Oncol Hematol 2021; 161:103325. [PMID: 33836238 PMCID: PMC8177002 DOI: 10.1016/j.critrevonc.2021.103325] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/29/2021] [Accepted: 03/26/2021] [Indexed: 01/10/2023] Open
Abstract
Near-InfraRed PhotoImmunoTherapy (NIR-PIT) is a novel cancer-targeted treatment effected by a chemical conjugation between a photosensitiser (e.g. the NIR phthalocyanine dye IRDye700DX) and a cancer-targeting moiety (e.g. a monoclonal antibody, moAb). Delivery of a conjugate in vivo leads to accumulation at the tumour cell surface by binding to cell surface receptors or antigens. Upon deployment of focal NIR-light, irradiation of the conjugate results in a rapid, targeted cell death. However, the mechanisms of action to produce the cytotoxic effects have yet to be fully understood. Herein, we bring together the current knowledge of NIR-PIT from preclinical and clinical studies in a variety of cancers highlighting the key unanswered research questions. Furthermore, we discuss how to enhance the local control of solid cancers using this novel treatment regimen.
Collapse
Affiliation(s)
- Irene Paraboschi
- Wellcome/EPSRC Centre for Interventional & Surgical Sciences, University College London, London, UK
| | - Stephen Turnock
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK
| | | | - Layla Musleh
- Department of Specialist Neonatal and Pediatric Surgery, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Marta Barisa
- Cancer Section, Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, UK
| | - John Anderson
- Cancer Section, Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, UK; Department of Oncology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, England, UK
| | - Stefano Giuliani
- Wellcome/EPSRC Centre for Interventional & Surgical Sciences, University College London, London, UK; Department of Specialist Neonatal and Pediatric Surgery, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK.
| |
Collapse
|
26
|
Hirata H, Kuwatani M, Nakajima K, Kodama Y, Yoshikawa Y, Ogawa M, Sakamoto N. Near-infrared photoimmunotherapy (NIR-PIT) on cholangiocarcinoma using a novel catheter device with light emitting diodes. Cancer Sci 2021; 112:828-838. [PMID: 33345417 PMCID: PMC7894014 DOI: 10.1111/cas.14780] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 12/14/2020] [Accepted: 12/17/2020] [Indexed: 12/11/2022] Open
Abstract
Near-infrared photoimmunotherapy (NIR-PIT) is a novel therapy for cancers that uses NIR light and antibody-photosensitizer (IR700) conjugates. However, it is difficult to deliver NIR light into the bile duct for cholangiocarcinoma (CCA) from the conventional extracorporeal apparatus. Thus, in this study, we developed a dedicated catheter with light emitting diodes (LEDs) that supersedes conventional external irradiation devices; we investigated the therapeutic effect of NIR-PIT for CCA using the novel catheter. The new catheter was designed to be placed in the bile duct and a temperature sensor was attached to the tip of the catheter to avoid thermal burn. An anti-epidermal growth factor receptor (EGFR) antibody, Panitumumab-IR700 conjugate or anti-human epidermal growth factor receptor type 2 (HER2) antibody, Trastuzumab-IR700 conjugate, was used with EGFR- or HER2-expressing cell lines, respectively. The in vitro efficacy of NIR-PIT was confirmed in cultured cells; the capability of the new catheter for NIR-PIT was then tested in a mouse tumor model. NIR-PIT via the developed catheter treated CCA xenografts in mice. NIR-PIT had an effect in Panitumumab-IR700 conjugate- and Trastuzumab-IR700 conjugate-treated CCA cells that depended on the receptor expression level. Tumor growth was significantly suppressed in mice treated with NIR-PIT using the novel catheter compared with controls (P < .01). NIR-PIT was an effective treatment for EGFR- and HER2-expressing CCA cells, and the novel catheter with mounted LEDs was useful for NIR-PIT of CCA.
Collapse
Affiliation(s)
- Hajime Hirata
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Masaki Kuwatani
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Kohei Nakajima
- Laboratory of Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | | | | | - Mikako Ogawa
- Laboratory of Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Naoya Sakamoto
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
27
|
Walker E, Turaga SM, Wang X, Gopalakrishnan R, Shukla S, Basilion JP, Lathia JD. Development of near-infrared imaging agents for detection of junction adhesion molecule-A protein. Transl Oncol 2021; 14:101007. [PMID: 33421750 PMCID: PMC7804988 DOI: 10.1016/j.tranon.2020.101007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/22/2020] [Accepted: 12/26/2020] [Indexed: 11/29/2022] Open
Abstract
Anti-junctional adhesion molecule-A (JAM-A) monoclonal antibodies (mAb) conjugated with near infra-red fluorescent dye, IR700 – as a JAM-A mAb/IR700 agent was developed. An in vivo JAM-A mAb/IR700-specific near infra-red imaging of human-derived prostate and breast cancer xenograft is presented. A single injection of the agent is diminished number of mitotic cells in cancerous tissue of mice bearing heterotopic tumors. Since, our agent depicts the specific accumulation within the targeted tumors, this agent may be adapted to solid tumor targeted photoimmunotherapy.
Introduction Prostate and breast cancer are the most prevalent primary malignant human tumors globally. Prostatectomy and breast conservative surgery remain the most common definitive treatment option for the >500,000 men and women newly diagnosed with localized prostate and breast cancer each year only in the US. Morphological examination is the mainstay of diagnosis but margin under-sampling of the excised cancer tissue may lead to local recurrence. In despite of the progress of non-invasive optical imaging, there is still a clinical need for targeted optical imaging probes that could rapidly and globally visualize cancerous tissues. Methods Elevated expression of junctional adhesion molecule-A (JAM-A) on tumor cells and its multiple pro-tumorigenic activity make the JAM-A a candidate for molecular imaging. Near-infrared imaging probe, which employed anti-JAM-A monoclonal antibody (mAb) phthalocyanine dye IR700 conjugates (JAM-A mAb/IR700), was synthesized and used to identify and visualize heterotopic human prostate and breast tumor mouse xenografts in vivo. Results The intravenously injected JAM-A mAb/IR700 conjugates enabled the non-invasive detection of prostate and breast cancerous tissue by fluorescence imaging. A single dose of JAM-A mAb/IR700 reduced number of mitotic cancer cells in vivo, indicating theranostic ability of this imaging agent. The JAM-A mAb/IR700 conjugates allowed us to image a specific receptor expression in prostate and breast tumors without post-image processing. Conclusion This agent demonstrates promise as a method to image the extent of prostate and breast cancer in vivo and could assist with real-time visualization of extracapsular extension of cancerous tissue.
Collapse
Affiliation(s)
- E Walker
- Department of Biomedical Engineering, Case Western Reserve University, Wearn Building, 11100 Euclid Ave., Cleveland, OH 44106-5056, USA; Case Comprehensive Cancer Center, Cleveland, OH 44106, USA.
| | - S M Turaga
- Lerner Research Institute, 9500 Euclid Avenue, NC10, Cleveland, OH 44195, USA; Department of Biological, Geological, and Environmental Sciences, Cleveland State University, 2121 Euclid Ave., Cleveland, OH 44115, USA
| | - X Wang
- Department of Biomedical Engineering, Case Western Reserve University, Wearn Building, 11100 Euclid Ave., Cleveland, OH 44106-5056, USA
| | - R Gopalakrishnan
- Department of Radiology, Case Center for Imaging Research, Case Western Reserve University, 11100 Euclid Ave, Cleveland, OH 44106-7207, USA
| | - S Shukla
- Department of Urology at the University of Florida College of Medicine, Faculty Clinic, 653 West 8th Street, FC12, Jacksonville, FL 32209, USA
| | - J P Basilion
- Department of Biomedical Engineering, Case Western Reserve University, Wearn Building, 11100 Euclid Ave., Cleveland, OH 44106-5056, USA; Department of Radiology, Case Center for Imaging Research, Case Western Reserve University, 11100 Euclid Ave, Cleveland, OH 44106-7207, USA; Case Comprehensive Cancer Center, Cleveland, OH 44106, USA
| | - J D Lathia
- Lerner Research Institute, 9500 Euclid Avenue, NC10, Cleveland, OH 44195, USA; Department of Biological, Geological, and Environmental Sciences, Cleveland State University, 2121 Euclid Ave., Cleveland, OH 44115, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, 9500 Euclid Avenue, NC10, Cleveland, OH 44195, USA; Case Comprehensive Cancer Center, Cleveland, OH 44106, USA
| |
Collapse
|
28
|
Molecular and Functional Imaging and Theranostics of the Tumor Microenvironment. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00069-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
29
|
Muttaqien SE, Nomoto T, Dou X, Takemoto H, Matsui M, Nishiyama N. Photodynamic therapy using LCST polymers exerting pH-responsive isothermal phase transition. J Control Release 2020; 328:608-616. [DOI: 10.1016/j.jconrel.2020.09.036] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 09/07/2020] [Accepted: 09/18/2020] [Indexed: 12/11/2022]
|
30
|
Biteghe FAN, Mungra N, Chalomie NET, Ndong JDLC, Engohang-Ndong J, Vignaux G, Padayachee E, Naran K, Barth S. Advances in epidermal growth factor receptor specific immunotherapy: lessons to be learned from armed antibodies. Oncotarget 2020; 11:3531-3557. [PMID: 33014289 PMCID: PMC7517958 DOI: 10.18632/oncotarget.27730] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 08/11/2020] [Indexed: 12/12/2022] Open
Abstract
The epidermal growth factor receptor (EGFR) has been recognized as an important therapeutic target in oncology. It is commonly overexpressed in a variety of solid tumors and is critically involved in cell survival, proliferation, metastasis, and angiogenesis. This multi-dimensional role of EGFR in the progression and aggressiveness of cancer, has evolved from conventional to more targeted therapeutic approaches. With the advent of hybridoma technology and phage display techniques, the first anti-EGFR monoclonal antibodies (mAbs) (Cetuximab and Panitumumab) were developed. Due to major limitations including host immune reactions and poor tumor penetration, these antibodies were modified and used as guiding mechanisms for the specific delivery of readily available chemotherapeutic agents or plants/bacterial toxins, giving rise to antibody-drug conjugates (ADCs) and immunotoxins (ITs), respectively. Continued refinement of ITs led to deimmunization strategies based on depletion of B and T-cell epitopes or substitution of non-human toxins leading to a growing repertoire of human enzymes capable of inducing cell death. Similarly, the modification of classical ADCs has resulted in the first, fully recombinant versions. In this review, we discuss significant advancements in EGFR-targeting immunoconjugates, including ITs and recombinant photoactivable ADCs, which serve as a blueprint for further developments in the evolving domain of cancer immunotherapy.
Collapse
Affiliation(s)
- Fleury Augustin Nsole Biteghe
- Department of Radiation Oncology and Biomedical Sciences, Cedars-Sinai Medical, Los Angeles, CA, USA
- These authors contributed equally to this work
| | - Neelakshi Mungra
- Medical Biotechnology & Immunotherapy Research Unit, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- These authors contributed equally to this work
| | | | - Jean De La Croix Ndong
- Department of Orthopedic Surgery, New York University School of Medicine, New York, NY, USA
| | - Jean Engohang-Ndong
- Department of Biological Sciences, Kent State University at Tuscarawas, New Philadelphia, OH, USA
| | | | - Eden Padayachee
- Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Krupa Naran
- Medical Biotechnology & Immunotherapy Research Unit, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- These authors contributed equally to this work
| | - Stefan Barth
- Medical Biotechnology & Immunotherapy Research Unit, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- South African Research Chair in Cancer Biotechnology, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- These authors contributed equally to this work
| |
Collapse
|
31
|
Nakajima K, Ogawa M. Phototoxicity in near-infrared photoimmunotherapy is influenced by the subcellular localization of antibody-IR700. Photodiagnosis Photodyn Ther 2020; 31:101926. [DOI: 10.1016/j.pdpdt.2020.101926] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/16/2020] [Accepted: 07/20/2020] [Indexed: 10/23/2022]
|
32
|
Walcher L, Kistenmacher AK, Suo H, Kitte R, Dluczek S, Strauß A, Blaudszun AR, Yevsa T, Fricke S, Kossatz-Boehlert U. Cancer Stem Cells-Origins and Biomarkers: Perspectives for Targeted Personalized Therapies. Front Immunol 2020; 11:1280. [PMID: 32849491 PMCID: PMC7426526 DOI: 10.3389/fimmu.2020.01280] [Citation(s) in RCA: 571] [Impact Index Per Article: 114.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 05/20/2020] [Indexed: 02/06/2023] Open
Abstract
The use of biomarkers in diagnosis, therapy and prognosis has gained increasing interest over the last decades. In particular, the analysis of biomarkers in cancer patients within the pre- and post-therapeutic period is required to identify several types of cells, which carry a risk for a disease progression and subsequent post-therapeutic relapse. Cancer stem cells (CSCs) are a subpopulation of tumor cells that can drive tumor initiation and can cause relapses. At the time point of tumor initiation, CSCs originate from either differentiated cells or adult tissue resident stem cells. Due to their importance, several biomarkers that characterize CSCs have been identified and correlated to diagnosis, therapy and prognosis. However, CSCs have been shown to display a high plasticity, which changes their phenotypic and functional appearance. Such changes are induced by chemo- and radiotherapeutics as well as senescent tumor cells, which cause alterations in the tumor microenvironment. Induction of senescence causes tumor shrinkage by modulating an anti-tumorigenic environment in which tumor cells undergo growth arrest and immune cells are attracted. Besides these positive effects after therapy, senescence can also have negative effects displayed post-therapeutically. These unfavorable effects can directly promote cancer stemness by increasing CSC plasticity phenotypes, by activating stemness pathways in non-CSCs, as well as by promoting senescence escape and subsequent activation of stemness pathways. At the end, all these effects can lead to tumor relapse and metastasis. This review provides an overview of the most frequently used CSC markers and their implementation as biomarkers by focussing on deadliest solid (lung, stomach, liver, breast and colorectal cancers) and hematological (acute myeloid leukemia, chronic myeloid leukemia) cancers. Furthermore, it gives examples on how the CSC markers might be influenced by therapeutics, such as chemo- and radiotherapy, and the tumor microenvironment. It points out, that it is crucial to identify and monitor residual CSCs, senescent tumor cells, and the pro-tumorigenic senescence-associated secretory phenotype in a therapy follow-up using specific biomarkers. As a future perspective, a targeted immune-mediated strategy using chimeric antigen receptor based approaches for the removal of remaining chemotherapy-resistant cells as well as CSCs in a personalized therapeutic approach are discussed.
Collapse
Affiliation(s)
- Lia Walcher
- Department of Immunology, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Ann-Kathrin Kistenmacher
- Department of Immunology, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Huizhen Suo
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Reni Kitte
- Department of Immunology, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Sarah Dluczek
- Department of Immunology, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Alexander Strauß
- Department of Immunology, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - André-René Blaudszun
- Department of Immunology, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Tetyana Yevsa
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Stephan Fricke
- Department of Immunology, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Uta Kossatz-Boehlert
- Department of Immunology, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| |
Collapse
|
33
|
Poudel K, Banstola A, Tran TH, Thapa RK, Gautam M, Ou W, Pham LM, Maharjan S, Jeong JH, Ku SK, Choi HG, Yong CS, Kim JO. Hyaluronic acid wreathed, trio-stimuli receptive and on-demand triggerable nanoconstruct for anchored combinatorial cancer therapy. Carbohydr Polym 2020; 249:116815. [PMID: 32933663 DOI: 10.1016/j.carbpol.2020.116815] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 07/01/2020] [Accepted: 07/22/2020] [Indexed: 12/12/2022]
Abstract
Hyaluronic acid (HA) assisted effective internalization into CD44 receptor-overexpressing cancer cells, which could offer an excellent cytotoxic profile and tumor alterations. In this study, duo-photothermal agents (copper sulfide (CuS) and graphene oxide (GO)), chemotherapeutic drug (doxorubicin (DOX)), and targeting moiety (HA) were incorporated into a complexed nanoconstruct for trio-responsive chemo-phototherapy. The nanosystem (CuS(DOX)-GO-HA) was demonstrating its responsive drug release and escalated photothermal behavior. The hyperthermia and photodynamic effect were observed along with efficient ROS generation in the presence of dual photosensitizers. The in vivo biodistribution and photothermal profile reflected a high accumulation and retention of the nanoconstruct in the tumor. Importantly, nanoconstructs effectively inhibit tumor growth based on tumor volume analysis and the altered expression of apoptosis, cell proliferation, and angiogenesis markers. Collectively, these findings suggest that this nanoconstruct has excellent antitumor effects in CD44 overexpressed cells showing the potential for clinical translation in the future.
Collapse
Affiliation(s)
- Kishwor Poudel
- College of Pharmacy, Yeungnam University, 214-1, Dae-Dong, Gyeongsan 712-749, South Korea
| | - Asmita Banstola
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea
| | - Tuan Hiep Tran
- Faculty of Pharmacy, Phenikaa University, Yen Nghia, Ha Dong District, Hanoi 100803, Viet Nam; PHENIKAA Research and Technology Institute (PRATI), A&A Green Phoenix Group JSC, No.167 Hoang Ngan, Trung Hoa, Cau Giay, Hanoi 11313, Viet Nam
| | - Raj Kumar Thapa
- College of Pharmacy, Yeungnam University, 214-1, Dae-Dong, Gyeongsan 712-749, South Korea
| | - Milan Gautam
- College of Pharmacy, Yeungnam University, 214-1, Dae-Dong, Gyeongsan 712-749, South Korea
| | - Wenquan Ou
- College of Pharmacy, Yeungnam University, 214-1, Dae-Dong, Gyeongsan 712-749, South Korea
| | - Le Minh Pham
- College of Pharmacy, Yeungnam University, 214-1, Dae-Dong, Gyeongsan 712-749, South Korea
| | - Srijan Maharjan
- College of Pharmacy, Yeungnam University, 214-1, Dae-Dong, Gyeongsan 712-749, South Korea
| | - Jee-Heon Jeong
- College of Pharmacy, Yeungnam University, 214-1, Dae-Dong, Gyeongsan 712-749, South Korea
| | - Sae Kwang Ku
- Department of Anatomy and Histology, College of Korean Medicine, Daegu Haany University, Gyeongan 712-715, South Korea
| | - Han-Gon Choi
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, 55, Hanyangdaehak-ro, Sangnok-gu, Ansan 426-791, South Korea
| | - Chul Soon Yong
- College of Pharmacy, Yeungnam University, 214-1, Dae-Dong, Gyeongsan 712-749, South Korea.
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, 214-1, Dae-Dong, Gyeongsan 712-749, South Korea.
| |
Collapse
|
34
|
Zhao Z, Li Y, Liu H, Jain A, Patel PV, Cheng K. Co-delivery of IKBKE siRNA and cabazitaxel by hybrid nanocomplex inhibits invasiveness and growth of triple-negative breast cancer. SCIENCE ADVANCES 2020; 6:eabb0616. [PMID: 32832636 PMCID: PMC7439402 DOI: 10.1126/sciadv.abb0616] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 06/02/2020] [Indexed: 05/15/2023]
Abstract
IKBKE is an oncogene in triple-negative breast cancer (TNBC), and we demonstrate that IKBKE small interfering RNA (siRNA) inhibits the proliferation, migration, and invasion of TNBC cells. Despite the recent success of siRNA therapeutics targeting to the liver, there still remains a great challenge to deliver siRNAs to solid tumors. Here, we report a hybrid nanocomplex to co-deliver the IKBKE siRNA and cabazitaxel to TNBC to achieve an optimal antitumor effect. The nanocomplex is modified with hyaluronic acid to target CD44 on TNBC cells. The nanocomplex shows higher cellular uptake and better tumor penetration of the encapsulated cargos. The nanocomplex also exhibits high tumor accumulation and antitumor activity in an orthotopic TNBC mouse model. Encapsulation of cabazitaxel in the nanocomplex enhances the activity of the IKBKE siRNA. The hybrid nanocomplex provides a novel and versatile platform for combination therapies using siRNAs and chemotherapy.
Collapse
|
35
|
Poudel K, Gautam M, Maharjan S, Jeong JH, Choi HG, Khan GM, Yong CS, Kim JO. Dual stimuli-responsive ursolic acid-embedded nanophytoliposome for targeted antitumor therapy. Int J Pharm 2020; 582:119330. [PMID: 32298743 DOI: 10.1016/j.ijpharm.2020.119330] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 03/23/2020] [Accepted: 04/10/2020] [Indexed: 12/12/2022]
Abstract
The hindrances in achieving clinically translatable anticancer platforms are being tackled through nanotechnology-based formulations. In this study, stimuli-responsive, phytoactive constituent-loaded nanophytoliposomes were fabricated for designing a specific antitumor platform. Ursolic acid (UA)-loaded nanophytoliposomes (UA-PLL-HA.P) enwrapped in a poly-L-lysine (PLL) coat and hyaluronic acid (HA) were nanosized; these nanophytoliposomes had spherical morphology, slightly negative charge, and an in-range polydispersity index (~0.25). Successful fabrication of the nanosystem was proven through several characterization methods and the pH- and enzyme-responsiveness of the nanosystem was assessed through a release study. The cellular internalization in CD44 receptor-expressing cell lines was amplified by enhanced permeation and retention as well as by active targeting. In vitro antitumor behavior was confirmed through in vitro cytotoxic and apoptotic activity of the nanosystem. Similarly, in vivo imaging showed exceptional biodistribution in the tumor in agreement with the in vitro findings. Moreover, the tumor inhibitory rate of UA-PLL-HA.P was significantly higher, and was ascribed to the targeting potential and stimuli-responsiveness. In summary, UA-PLL-HA.P exhibited pronounced anticancer effect and could open a number of possibilities for discovering novel phytoconstituent-incorporated nanoformulations.
Collapse
Affiliation(s)
- Kishwor Poudel
- College of Pharmacy, Yeungnam University, 280 Daehak-Ro, Gyeongsan 712-749, Republic of Korea
| | - Milan Gautam
- College of Pharmacy, Yeungnam University, 280 Daehak-Ro, Gyeongsan 712-749, Republic of Korea
| | - Srijan Maharjan
- College of Pharmacy, Yeungnam University, 280 Daehak-Ro, Gyeongsan 712-749, Republic of Korea
| | - Jee-Heon Jeong
- College of Pharmacy, Yeungnam University, 280 Daehak-Ro, Gyeongsan 712-749, Republic of Korea
| | - Han-Gon Choi
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, 55, Hanyangdaehak-ro, Sangnok-gu, Ansan 426-791, Republic of Korea
| | - Gulam Muhammad Khan
- Pokhara University Research Centre (PURC), Pokhara University, Dhungepatan, Lekhnath-12, Kaski, Nepal
| | - Chul Soon Yong
- College of Pharmacy, Yeungnam University, 280 Daehak-Ro, Gyeongsan 712-749, Republic of Korea.
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, 280 Daehak-Ro, Gyeongsan 712-749, Republic of Korea.
| |
Collapse
|
36
|
Zou W, Yang Y, Zheng R, Wang Z, Zeng H, Chen Z, Yang F, Wang J. Association of CD44 and CD24 phenotype with lymph node metastasis and survival in triple-negative breast cancer. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2020; 13:1008-1016. [PMID: 32509072 PMCID: PMC7270704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 03/26/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND CD44+CD24-/low phenotypes are associated with poor outcome of triple-negative breast cancer (TNBC); however, the role of the CD44+CD24-/low phenotype in lymph node metastasis and survival has not been fully understood in TNBC. METHODS A total of 51 TNBC patients were included. CD44 and CD24 expression was determined using immunohistochemistry by which CD44 and CD24 were double-immunostained. Overall survival (OS) and disease-free survival (DFS) were estimated using the Kaplan-Meier method. RESULTS The proportion of the CD44+CD24-/low phenotype was 33.3% in TNBC specimens without lymph node metastases and 69.0% in those with lymph node metastases. In addition, the CD44+CD24-/low phenotype correlated significantly with tumor size, histologic classification, TNM stage, and lymph node metastasis (P < 0.05). The CD44+CD24-/low phenotype was detected in 69.0% of TNBC patients with lymph node metastases, and 51.7% of TNBC patients without lymph node metastases. In TNBC patients without lymph node metastases, the median DFS and OS were 18.2 and 28 months in cases with a CD44+CD24-/low phenotype and 26.5 and 42.5 months in those without a CD44+CD24-/low phenotype (P < 0.05), and in TNBC patients with lymph node metastases, the median DFS and OS were 17.2 and 25.7 months in cases with a CD44+CD24-/low phenotype and 24.5 and 39.3 months in those without a CD44+CD24-/low phenotype, respectively (P < 0.05). CONCLUSIONS CD44 and CD24 are independent prognostic markers for patients with TNBC. The CD44+CD24-/low phenotype correlates with more aggressive clinicopathologic features and is strongly associated with poor prognosis in patients with TNBC.
Collapse
Affiliation(s)
- Weiyan Zou
- Department of Histology and Embryology, Bengbu Medical CollegeBengbu 233004, Anhui Province, China
| | - Yan Yang
- Department of Oncology, The First Affiliated Hospital of Bengbu Medical CollegeBengbu 233004, Anhui Province, China
| | - Rongsheng Zheng
- Department of Oncology, The First Affiliated Hospital of Bengbu Medical CollegeBengbu 233004, Anhui Province, China
| | - Zishu Wang
- Department of Oncology, The First Affiliated Hospital of Bengbu Medical CollegeBengbu 233004, Anhui Province, China
| | - Huihui Zeng
- Department of Oncology, The First Affiliated Hospital of Bengbu Medical CollegeBengbu 233004, Anhui Province, China
| | - Zhelong Chen
- Department of Pathology, Bengbu Medical CollegeBengbu 233004, Anhui Province, China
| | - Fen Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanjing Medical UniversityNanjing 211166, Jiangsu Province, China
| | - Junbin Wang
- Department of Oncology, The First Affiliated Hospital of Bengbu Medical CollegeBengbu 233004, Anhui Province, China
| |
Collapse
|
37
|
A multifunctional magnetic nanosystem based on "two strikes" effect for synergistic anticancer therapy in triple-negative breast cancer. J Control Release 2020; 322:401-415. [PMID: 32246976 DOI: 10.1016/j.jconrel.2020.03.036] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/12/2020] [Accepted: 03/25/2020] [Indexed: 12/14/2022]
Abstract
Multifunctional magnetic nanoparticles (MNPs) were widely used for ablation of cancer cells because of their potential on physical treatment. Herein, we developed the "cell targeting destructive" multifunctional polymeric nanoparticles (named as HA-Olb-PPMNPs) based on PEI-PLGA co-loaded with the anticancer drug Olaparib (Olb) and superparamagnetic iron oxide nanoparticles (Fe3O4 NPs), and further coated with a low molecular weight hyaluronic acid (HA) on its surface. Due to the high affinity between HA and CD44-receptor on cell surface of triple negative breast cancer (TNBC), an active targeting can be achieved. Under a rotating magnetic field (RMF), HA-Olb-PPMNPs produced a physical transfer of mechanical force by incomplete rotation. This mechanical force could cause the "two strikes" effect on the cells, in which "First-strike" was to damage the cell membrane structure (magneto-cell-lysis), another "Second-strike" could activate the lysosome-mitochondrial pathway by injuring lysosomes to induce cell apoptosis (magneto-cell-apoptosis). Therefore, the mechanical force and Olb exert dual anti-tumor effect to achieve synergistic therapeutic in the presence of RMF. This study proposes a novel multi-therapeutic concept for TNBC, as well as provided evidences of new anti-tumor therapeutic effects induced by the magnetic nanoparticles drug system.
Collapse
|
38
|
Redox-Sensitive and Hyaluronic Acid-Functionalized Nanoparticles for Improving Breast Cancer Treatment by Cytoplasmic 17α-Methyltestosterone Delivery. Molecules 2020; 25:molecules25051181. [PMID: 32151062 PMCID: PMC7179432 DOI: 10.3390/molecules25051181] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 02/24/2020] [Accepted: 02/25/2020] [Indexed: 11/30/2022] Open
Abstract
Novel reduction-responsive hyaluronic acid–chitosan–lipoic acid nanoparticles (HACSLA-NPs) were designed and synthesized for effective treatment of breast cancer by targeting Cluster of Differentiation 44 (CD44)-overexpressing cells and reduction-triggered 17α-Methyltestosterone (MT) release for systemic delivery. The effectiveness of these nanoparticles was investigated by different assays, including release rate, 3-(4,5-Dimethylthiazol-2-Yl)-2,5-Diphenyltetrazolium Bromide (MTT), lactate dehydrogenase (LDH), caspase-3 activity, Rhodamine 123 (RH-123), and Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL). In vitro experiments revealed that Methyltestosterone/Hyaluronic acid–chitosan–lipoic acid nanoparticles (MT/HACSLA-NPs) illustrated a sustained drug release in the absence of glutathione (GSH), while the presence of GSH led to fast MT release. HACSLA-NPs also showed high cellular internalization via CD44 receptors, quick drug release inside the cells, and amended cytotoxicity against positive CD44 BT-20 breast cancer cell line as opposed to negative CD44, Michigan Cancer Foundation-7 (MCF-7) cell line. These findings supported that these novel reduction-responsive NPs can be promising candidates for efficient targeted delivery of therapeutics in cancer therapy.
Collapse
|
39
|
Abstract
This Account is the first comprehensive review article on the newly developed, photochemistry-based cancer therapy near-infrared (NIR) photoimmunotherapy (PIT). NIR-PIT is a molecularly targeted phototherapy for cancer that is based on injecting a conjugate of a near-infrared, water-soluble, silicon-phthalocyanine derivative, IRdye700DX (IR700), and a monoclonal antibody (mAb) that targets an expressed antigen on the cancer cell surface. Subsequent local exposure to NIR light turns on this photochemical "death" switch, resulting in the rapid and highly selective immunogenic cell death (ICD) of targeted cancer cells. ICD occurs as early as 1 min after exposure to NIR light and results in irreversible morphologic changes only in target-expressing cells based on the newly discovered photoinduced ligand release reaction that induces physical changes on conjugated antibody/antigen complex resulting in functional damage on cell membrane. Meanwhile, immediately adjacent receptor-negative cells are totally unharmed. Because of its highly targeted nature, NIR-PIT carries few side effects and healing is rapid. Evaluation of the tumor microenvironment reveals that ICD induced by NIR-PIT results in rapid maturation of immature dendritic cells adjacent to dying cancer cells initiating a host anticancer immune response, resulting in repriming of polyclonal CD8+T cells against various released cancer antigens, which amplifies the therapeutic effect of NIR-PIT. NIR-PIT can target and treat virtually any cell surface antigens including cancer stem cell markers, that is, CD44 and CD133. A first-in-human phase 1/2 clinical trial of NIR-PIT using cetuximab-IR700 (RM1929) targeting EGFR in inoperable recurrent head and neck cancer patients successfully concluded in 2017 and led to "fast tracking" by the FDA and a phase 3 trial ( https://clinicaltrials.gov/ct2/show/NCT03769506 ) that is currently underway in 3 countries in Asia, US/Canada, and 4 countries in EU. The next step for NIR-PIT is to further exploit the immune response. Preclinical research in animals with intact immune systems has shown that NIT-PIT targeting of immunosuppressor cells within the tumor, such as regulatory T-cells, can further enhance tumor-cell-selective systemic host-immunity leading to significant responses in distant metastatic tumors, which are not treated with light. By combining cancer-targeting NIR-PIT and immune-activating NIR-PIT or other cancer immunotherapies, NIR-PIT of a local tumor, could lead to responses in distant metastases and may also inhibit recurrences due to activation of systemic anticancer immunity and long-term immune memory without the systemic autoimmune adverse effects often associated with immune checkpoint inhibitors. Furthermore, NIR-PIT also enhances nanodrug delivery into tumors up to 24-fold superior to untreated tumors with conventional EPR effects by intensively damaging cancer cells behind tumor vessels. We conclude by describing future advances in this novel photochemical cancer therapy that are likely to further enhance the efficacy of NIR-PIT.
Collapse
Affiliation(s)
- Hisataka Kobayashi
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10, Room B3B69, MSC1088, Bethesda, Maryland 20892-1088, United States
| | - Peter L. Choyke
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10, Room B3B69, MSC1088, Bethesda, Maryland 20892-1088, United States
| |
Collapse
|
40
|
Fernandes SRG, Fernandes R, Sarmento B, Pereira PMR, Tomé JPC. Photoimmunoconjugates: novel synthetic strategies to target and treat cancer by photodynamic therapy. Org Biomol Chem 2019; 17:2579-2593. [PMID: 30648722 DOI: 10.1039/c8ob02902d] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Photodynamic therapy (PDT) combines a photosensitizer (PS) with the physical energy of non-ionizing light to trigger cell death pathways. PDT has potential as a therapeutic modality to be used in alternative or in combination with other conventional cancer treatment protocols (e.g. surgery, chemotherapy and radiotherapy). Still, due to the lack of specificity of the current PSs to target the tumor cells, several studies have exploited their conjugation with targeting moieties. PSs conjugated with antibodies (Abs) or their fragments, able to bind antigens overexpressed in the tumors, have demonstrated potential in PDT of tumors. This review provides an overview of the most recent advances on photoimmunoconjugates (PICs) for cancer PDT, which involve the first and second-generation PSs conjugated to Abs. This is an update of our previous review "Antibodies armed with photosensitizers: from chemical synthesis to photobiological applications", published in 2015 in Org. Biomol. Chem.
Collapse
Affiliation(s)
- Sara R G Fernandes
- CQE, Departamento de Engenharia Química, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal.
| | | | | | | | | |
Collapse
|
41
|
Moamer A, Hachim IY, Binothman N, Wang N, Lebrun JJ, Ali S. A role for kinesin-1 subunits KIF5B/KLC1 in regulating epithelial mesenchymal plasticity in breast tumorigenesis. EBioMedicine 2019; 45:92-107. [PMID: 31204277 PMCID: PMC6642081 DOI: 10.1016/j.ebiom.2019.06.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 06/03/2019] [Accepted: 06/06/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Epithelial mesenchymal plasticity (EMP) is deemed vital in breast cancer progression, metastasis, stemness and resistance to therapy. Therefore, characterizing molecular mechanisms contributing to EMP are in need enabling the development of more advanced therapeutics against breast cancer. While kinesin superfamily proteins (KIFs) are well known for their role in intracellular cargo movement, our knowledge of their function in breast tumorigenesis is still limited. METHODS Various breast cancer cell lines representing different molecular subtypes were used to determine the role of kinesine-1 subunits KIF5B/KLC1 in regulation of EMP. FINDINGS In breast cancer, we show that kinesin family member 5B (KIF5B) and its partner protein kinesin light chain 1 (KLC1), subunits of kinesin-1, to play differential roles in regulating EMP and tumorigenesis. Indeed, we found KIF5B to be expressed in triple negative (TN)-basal-like/claudin low breast cancer subtype and to be an inducer of epithelial-mesenchymal transition (EMT), stemness, invasiveness, tumor formation and metastatic colonization. Whereas, we found KLC1 to be expressed in epithelial/luminal breast cancer subtypes and to be a suppressor of EMT, invasion, metastasis and stem cell markers expression as well as to be an inducer of epithelial/luminal phenotype. Interestingly, in TN-basal-like/claudin low cells we found a novel nuclear accumulation of KIF5B and its interaction with the EMT transcriptional regulator Snail1 independent of KLC1. In addition, TGF-β mediated pro-invasive activity was found to be dependent on KIF5B expression. In contrast, the epithelial differentiation factor and EMT suppressor prolactin (PRL) was found to repress KIF5B gene expression and KIF5B-Snail1 nuclear accumulation, but enhanced KLC1 gene expression and KIF5B-KLC1 interaction. INTERPRETATION Together, these results highlight a new paradigm for kinesin-1 function in breast tumorigenesis by regulating EMP programing and aggressiveness. FUND: This work was supported by the Canadian Institutes of Health Research (operating grants #233437 and 233438) granted to Suhad Ali.
Collapse
Affiliation(s)
- Alaa Moamer
- Department of Medicine, Cancer Research Program, Centre for Translational Biology, McGill University Health Centre, McGill University, Canada.
| | - Ibrahim Y Hachim
- Department of Medicine, Cancer Research Program, Centre for Translational Biology, McGill University Health Centre, McGill University, Canada.
| | - Najat Binothman
- Department of Medicine, Cancer Research Program, Centre for Translational Biology, McGill University Health Centre, McGill University, Canada.
| | - Ni Wang
- Department of Medicine, Cancer Research Program, Centre for Translational Biology, McGill University Health Centre, McGill University, Canada.
| | - Jean-Jacques Lebrun
- Department of Medicine, Cancer Research Program, Centre for Translational Biology, McGill University Health Centre, McGill University, Canada.
| | - Suhad Ali
- Department of Medicine, Cancer Research Program, Centre for Translational Biology, McGill University Health Centre, McGill University, Canada.
| |
Collapse
|
42
|
Synergistic triple-combination therapy with hyaluronic acid-shelled PPy/CPT nanoparticles results in tumor regression and prevents tumor recurrence and metastasis in 4T1 breast cancer. Biomaterials 2019; 217:119264. [PMID: 31260883 DOI: 10.1016/j.biomaterials.2019.119264] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 06/06/2019] [Accepted: 06/07/2019] [Indexed: 12/14/2022]
Abstract
Breast cancer is characterized by high aggression, poor prognosis, and high recurrence rate. Early detection and specific targeted treatment with less toxicity are the ultimate goals for breast cancer therapy. To improve antitumor therapeutic effects, we developed a novel polypyrrole nanoparticle using the near infrared dye IRDye800CW with camptothecin (CPT)-conjugated hyaluronic acid (HA) shell (PPy@CPT-HA-IRDye800CW) and performed a photothermal therapy (PTT), along with chemotherapy, guided by fluorescence and photoacoustic dual-modality imaging, in combination with immunotherapy. Irradiation with near infrared (NIR) light offered a strong PTT effect and promoted CPT drug release in tumors. Moreover, we found that chemo-photothermal therapy with PPy@CPT-HA-IRDye800CW NPs, in combination with immune checkpoint inhibitor anti-PD-L1 immunotherapy, synergistically enhanced the anti-tumor immune response, thereby eliminating primary breast cancer and preventing tumor metastases and recurrences in 4T1 tumor-bearing mice. This approach may provide important clues for the clinical management of breast cancer and other malignant tumors.
Collapse
|
43
|
Pramanik N, Ranganathan S, Rao S, Suneet K, Jain S, Rangarajan A, Jhunjhunwala S. A Composite of Hyaluronic Acid-Modified Graphene Oxide and Iron Oxide Nanoparticles for Targeted Drug Delivery and Magnetothermal Therapy. ACS OMEGA 2019; 4:9284-9293. [PMID: 31460017 PMCID: PMC6648023 DOI: 10.1021/acsomega.9b00870] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 05/15/2019] [Indexed: 05/27/2023]
Abstract
Graphene oxide (GO) nanoparticles have been developed for a variety of biomedical applications as a number of different therapeutic modalities may be added onto them. Here, we report the development and testing of such a multifunctional GO nanoparticle platform that contains a grafted cell-targeting functionality, active pharmaceutical ingredients, and particulates that enable the use of magnetothermal therapy. Specifically, we demonstrate the ability to covalently attach hyaluronic acid (HA) onto GO, and the resultant nanoparticulates (GO-HA) exhibited low inherent toxicity toward two different breast cancer cell lines, BT-474 and MDA-MB-231. Doxorubicin (Dox) and paclitaxel (Ptx) were successfully loaded onto GO-HA with high and moderate efficiencies, respectively. A GO-HA-Dox/Ptx system was significantly better than the GO-Dox/Ptx system at specifically killing CD44-expressing MDA-MB-231 cells but not BT-474 cells that do not express CD44. Further, modified iron oxide nanoparticles were loaded onto the GO-HA-Dox system, enabling the use of magnetic hyperthermia. Hyperthermia in combination with Dox treatment through the GO-HA system showed significantly better performance in reducing viable tumor cell numbers when compared to the individual systems. In summary, we showcase a multifunctional GO nanoparticle system that demonstrates improved efficacy in killing tumor cells.
Collapse
Affiliation(s)
- Nilkamal Pramanik
- Centre
for BioSystems Science and Engineering and Molecular Reproduction, Development
and Genetics, Indian Institute of Science, Bengaluru 560012, India
| | - Santhalakshmi Ranganathan
- Centre
for BioSystems Science and Engineering and Molecular Reproduction, Development
and Genetics, Indian Institute of Science, Bengaluru 560012, India
| | - Sunaina Rao
- Centre
for BioSystems Science and Engineering and Molecular Reproduction, Development
and Genetics, Indian Institute of Science, Bengaluru 560012, India
| | - Kaushik Suneet
- Centre
for BioSystems Science and Engineering and Molecular Reproduction, Development
and Genetics, Indian Institute of Science, Bengaluru 560012, India
| | - Shilpee Jain
- Centre
for BioSystems Science and Engineering and Molecular Reproduction, Development
and Genetics, Indian Institute of Science, Bengaluru 560012, India
| | - Annapoorni Rangarajan
- Centre
for BioSystems Science and Engineering and Molecular Reproduction, Development
and Genetics, Indian Institute of Science, Bengaluru 560012, India
| | - Siddharth Jhunjhunwala
- Centre
for BioSystems Science and Engineering and Molecular Reproduction, Development
and Genetics, Indian Institute of Science, Bengaluru 560012, India
| |
Collapse
|
44
|
Varshosaz J, Sarrami N, Aghaei M, Aliomrani M, Azizi R. LHRH Targeted Chonderosomes of Mitomycin C in Breast Cancer: An In Vitro/ In Vivo Study. Anticancer Agents Med Chem 2019; 19:1405-1417. [PMID: 30987576 DOI: 10.2174/1871520619666190415165849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 03/04/2019] [Accepted: 04/02/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Mitomycin C (MMC) is an anti-cancer drug used for the treatment of breast cancer with limited therapeutic index, extreme gastric adverse effects and bone marrow suppression. The purpose of the present study was the preparation of a dual-targeted delivery system of MMC for targeting CD44 and LHRH overexpressed receptors of breast cancer. METHODS MMC loaded LHRH targeted chonderosome was prepared by precipitation method and was characterized for their physicochemical properties. Cell cycle arrest and cytotoxicity tests were studied on cell lines of MCF-7, MDA-MB231 and 4T1 (as CD44 and LHRH positive cells) and BT-474 cell line (as CD44 negative receptor cells). The in vivo histopathology and antitumor activity of MMC-loaded chonderosomes were compared with free MMC in 4T1 cells inducing breast cancer in Balb-c mice. RESULTS MMC loaded LHRH targeted chonderosomes caused 3.3 and 5.5 fold more cytotoxicity on MCF-7 and 4T1 cells than free MMC at concentrations of 100μM and 10μM, respectively. However, on BT-474 cells the difference was insignificant. The cell cycle test showed no change for MMC mechanism of action when it was loaded in chonderosomes compared to free MMC. The in vivo antitumor studies showed that MMC loaded LHRH targeted chonderosomes were 6.5 fold more effective in the reduction of tumor volume than free MMC with the most severe necrosis compared to non-targeted chonderosomes in pathological studies on harvested tumors. CONCLUSION The developed MMC loaded LHRH targeted chonderosomes were more effective in tumor growth suppression and may be promising for targeted delivery of MMC in breast cancer.
Collapse
Affiliation(s)
- Jaleh Varshosaz
- Novel Drug Delivery Systems Research Center, Department of Pharmaceutics, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nasim Sarrami
- Novel Drug Delivery Systems Research Center, Department of Pharmaceutics, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahmoud Aghaei
- Department of Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mehdi Aliomrani
- Department of Pharmacology and Toxicology, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Reza Azizi
- Department of Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
45
|
Das A, Narayanam MK, Paul S, Mukhnerjee P, Ghosh S, Dastidar DG, Chakrabarty S, Ganguli A, Basu B, Pal M, Chatterji U, Banerjee SK, Karmakar P, Kumar D, Chakrabarti G. A novel triazole, NMK-T-057, induces autophagic cell death in breast cancer cells by inhibiting γ-secretase-mediated activation of Notch signaling. J Biol Chem 2019; 294:6733-6750. [PMID: 30824542 DOI: 10.1074/jbc.ra119.007671] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Indexed: 01/05/2023] Open
Abstract
Notch signaling is reported to be deregulated in several malignancies, including breast, and the enzyme γ-secretase plays an important role in the activation and nuclear translocation of Notch intracellular domain (NICD). Hence, pharmacological inhibition of γ-secretase might lead to the subsequent inhibition of Notch signaling in cancer cells. In search of novel γ-secretase inhibitors (GSIs), we screened a series of triazole-based compounds for their potential to bind γ-secretase and observed that 3-(3'4',5'-trimethoxyphenyl)-5-(N-methyl-3'-indolyl)-1,2,4-triazole compound (also known as NMK-T-057) can bind to γ-secretase complex. Very interestingly, NMK-T-057 was found to inhibit proliferation, colony-forming ability, and motility in various breast cancer (BC) cells such as MDA-MB-231, MDA-MB-468, 4T1 (triple-negative cells), and MCF-7 (estrogen receptor (ER)/progesterone receptor (PR)-positive cell line) with negligible cytotoxicity against noncancerous cells (MCF-10A and peripheral blood mononuclear cells). Furthermore, significant induction of apoptosis and inhibition of epithelial-to-mesenchymal transition (EMT) and stemness were also observed in NMK-T-057-treated BC cells. The in silico study revealing the affinity of NMK-T-057 toward γ-secretase was further validated by a fluorescence-based γ-secretase activity assay, which confirmed inhibition of γ-secretase activity in NMK-T-057-treated BC cells. Interestingly, it was observed that NMK-T-057 induced significant autophagic responses in BC cells, which led to apoptosis. Moreover, NMK-T-057 was found to inhibit tumor progression in a 4T1-BALB/c mouse model. Hence, it may be concluded that NMK-T-057 could be a potential drug candidate against BC that can trigger autophagy-mediated cell death by inhibiting γ-secretase-mediated activation of Notch signaling.
Collapse
Affiliation(s)
- Amlan Das
- From the Department of Biotechnology and Dr. B. C. Guha Centre for Genetic Engineering and Biotechnology and .,Department of Chemistry, National Institute of Technology, Ravangla, South Sikkim 737139, India
| | - Maruthi Kumar Narayanam
- Department of Chemistry, Birla Institute of Technology and Science, Pilani, Rajasthan 333031, India.,Department of Molecular and Medical Pharmacology and Crump Institute for Molecular Imaging, David Geffen School of Medicine, UCLA, Los Angeles, California 90095, and
| | - Santanu Paul
- From the Department of Biotechnology and Dr. B. C. Guha Centre for Genetic Engineering and Biotechnology and
| | - Pritha Mukhnerjee
- Department of Zoology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata 700019, West Bengal, India
| | - Suvranil Ghosh
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Road, Scheme VIIM, Kankurgachi, Kolkata 700054, West Bengal, India
| | - Debabrata Ghosh Dastidar
- From the Department of Biotechnology and Dr. B. C. Guha Centre for Genetic Engineering and Biotechnology and.,Division of Pharmaceutics, Guru Nanak Institute of Pharmaceutical Science and Technology, 157/F Nilgunj Road, Panihati, Kolkata 700114, West Bengal, India
| | - Subhendu Chakrabarty
- From the Department of Biotechnology and Dr. B. C. Guha Centre for Genetic Engineering and Biotechnology and
| | - Arnab Ganguli
- From the Department of Biotechnology and Dr. B. C. Guha Centre for Genetic Engineering and Biotechnology and
| | - Biswarup Basu
- Department of Experimental Hematology and Neuroendocrinology, Chittaranjan National Cancer Institute, 37 Shyama Prasad Mukherjee Road, Kolkata 700026, West Bengal, India
| | - Mahadeb Pal
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Road, Scheme VIIM, Kankurgachi, Kolkata 700054, West Bengal, India
| | - Urmi Chatterji
- Department of Zoology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata 700019, West Bengal, India
| | - Sushanta K Banerjee
- Cancer Research Unit, Veterans Affairs Medical Center, Kansas City, Missouri 64128.,Departments of Anatomy and Cell Biology and Pathology, University of Kansas Medical Center, Kansas City, Kansas 66160
| | - Parimal Karmakar
- Department of Life Science and Biotechnology, Jadavpur University, 188 Raja S. C. Mullick Road, Kolkata 700032, Western Bengal, India
| | - Dalip Kumar
- Department of Chemistry, Birla Institute of Technology and Science, Pilani, Rajasthan 333031, India,
| | - Gopal Chakrabarti
- From the Department of Biotechnology and Dr. B. C. Guha Centre for Genetic Engineering and Biotechnology and
| |
Collapse
|
46
|
Aung W, Tsuji AB, Sugyo A, Takashima H, Yasunaga M, Matsumura Y, Higashi T. Near-infrared photoimmunotherapy of pancreatic cancer using an indocyanine green-labeled anti-tissue factor antibody. World J Gastroenterol 2018; 24:5491-5504. [PMID: 30622378 PMCID: PMC6319132 DOI: 10.3748/wjg.v24.i48.5491] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 11/07/2018] [Accepted: 11/16/2018] [Indexed: 02/07/2023] Open
Abstract
AIM To investigate near-infrared photoimmunotherapeutic effect mediated by an anti-tissue factor (TF) antibody conjugated to indocyanine green (ICG) in a pancreatic cancer model. METHODS Near-infrared photoimmunotherapy (NIR-PIT) is a highly selective tumor treatment that utilizes an antibody-photosensitizer conjugate administration, followed by NIR light exposure. Anti-TF antibody 1849-ICG conjugate was synthesized by labeling of rat IgG2b anti-TF monoclonal antibody 1849 (anti-TF 1849) to a NIR photosensitizer, ICG. The expression levels of TF in two human pancreatic cancer cell lines were examined by western blotting. Specific binding of the 1849-ICG to TF-expressing BxPC-3 cells was examined by fluorescence microscopy. NIR-PIT-induced cell death was determined by cell viability imaging assay. In vivo longitudinal fluorescence imaging was used to explore the accumulation of 1849-ICG conjugate in xenograft tumors. To examine the effect of NIR-PIT, tumor-bearing mice were separated into 5 groups: (1) 100 μg of 1849-ICG i.v. administration followed by NIR light exposure (50 J/cm2) on two consecutive days (Days 1 and 2); (2) NIR light exposure (50 J/cm2) only on two consecutive days (Days 1 and 2); (3) 100 μg of 1849-ICG i.v. administration; (4) 100 μg of unlabeled anti-TF 1849 i.v. administration; and (5) the untreated control. Semiweekly tumor volume measurements, accompanied with histological and immunohistochemical (IHC) analyses of tumors, were performed 3 d after the 2nd irradiation with NIR light to monitor the effect of treatments. RESULTS High TF expression in BxPC-3 cells was observed via western blot analysis, concordant with the observed preferential binding with intracellular localization of 1849-ICG via fluorescence microscopy. NIR-PIT-induced cell death was observed by performing cell viability imaging assay. In contrast to the other test groups, tumor growth was significantly inhibited by NIR-PIT with a statistically significant difference in relative tumor volumes for 27 d after the treatment start date [2.83 ± 0.38 (NIR-PIT) vs 5.42 ± 1.61 (Untreated), vs 4.90 ± 0.87 (NIR), vs 4.28 ± 1.87 (1849-ICG), vs 4.35 ± 1.42 (anti-TF 1849), at Day 27, P < 0.05]. Tumors that received NIR-PIT showed evidence of necrotic cell death-associated features upon hematoxylin-eosin staining accompanied by a decrease in Ki-67-positive cells (a cell proliferation marker) by IHC examination. CONCLUSION The TF-targeted NIR-PIT with the 1849-ICG conjugate can potentially open a new platform for treatment of TF-expressing pancreatic cancer.
Collapse
Affiliation(s)
- Winn Aung
- Department of Molecular Imaging and Theranostics, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology (QST-NIRS), Chiba 263-8555, Japan
| | - Atsushi B Tsuji
- Department of Molecular Imaging and Theranostics, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology (QST-NIRS), Chiba 263-8555, Japan
| | - Aya Sugyo
- Department of Molecular Imaging and Theranostics, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology (QST-NIRS), Chiba 263-8555, Japan
| | - Hiroki Takashima
- Division of Developmental Therapeutics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Chiba 277-8577, Japan
| | - Masahiro Yasunaga
- Division of Developmental Therapeutics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Chiba 277-8577, Japan
| | - Yasuhiro Matsumura
- Division of Developmental Therapeutics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Chiba 277-8577, Japan
| | - Tatsuya Higashi
- Department of Molecular Imaging and Theranostics, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology (QST-NIRS), Chiba 263-8555, Japan
| |
Collapse
|
47
|
Sato K, Ando K, Okuyama S, Moriguchi S, Ogura T, Totoki S, Hanaoka H, Nagaya T, Kokawa R, Takakura H, Nishimura M, Hasegawa Y, Choyke PL, Ogawa M, Kobayashi H. Photoinduced Ligand Release from a Silicon Phthalocyanine Dye Conjugated with Monoclonal Antibodies: A Mechanism of Cancer Cell Cytotoxicity after Near-Infrared Photoimmunotherapy. ACS CENTRAL SCIENCE 2018; 4:1559-1569. [PMID: 30555909 PMCID: PMC6276043 DOI: 10.1021/acscentsci.8b00565] [Citation(s) in RCA: 176] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Indexed: 05/23/2023]
Abstract
Photochemical reactions can dramatically alter physical characteristics of reacted molecules. In this study, we demonstrate that near-infrared (NIR) light induces an axial ligand-releasing reaction, which dramatically alters hydrophilicity of a silicon phthalocyanine derivative (IR700) dye leading to a change in the shape of the conjugate and its propensity to aggregate in aqueous solution. This photochemical reaction is proposed as a major mechanism of cell death induced by NIR photoimmunotherapy (NIR-PIT), which was recently developed as a molecularly targeted cancer therapy. Once the antibody-IR700 conjugate is bound to its target, activation by NIR light causes physical changes in the shape of antibody antigen complexes that are thought to induce physical stress within the cellular membrane leading to increases in transmembrane water flow that eventually lead to cell bursting and necrotic cell death.
Collapse
Affiliation(s)
- Kazuhide Sato
- Molecular
Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892-1088, United States
- Institute
for Advanced Research, Nagoya University, Nagoya, Aichi 464-0814, Japan
- Department
of Respiratory Medicine, Nagoya University
Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Kanta Ando
- Laboratory
for Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical
Sciences, Hokkaido University, Sapporo, Hokkaido 060-0812, Japan
| | - Shuhei Okuyama
- Molecular
Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892-1088, United States
- Shimadzu
Corporation, Kyoto 604-8511, Japan
| | | | | | | | - Hirofumi Hanaoka
- Molecular
Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892-1088, United States
| | - Tadanobu Nagaya
- Molecular
Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892-1088, United States
| | | | - Hideo Takakura
- Laboratory
for Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical
Sciences, Hokkaido University, Sapporo, Hokkaido 060-0812, Japan
| | | | - Yoshinori Hasegawa
- Department
of Respiratory Medicine, Nagoya University
Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Peter L. Choyke
- Molecular
Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892-1088, United States
| | - Mikako Ogawa
- Laboratory
for Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical
Sciences, Hokkaido University, Sapporo, Hokkaido 060-0812, Japan
| | - Hisataka Kobayashi
- Molecular
Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892-1088, United States
| |
Collapse
|
48
|
Gotov O, Battogtokh G, Shin D, Ko YT. Hyaluronic acid-coated cisplatin conjugated gold nanoparticles for combined cancer treatment. J IND ENG CHEM 2018. [DOI: 10.1016/j.jiec.2018.04.034] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
49
|
CSCs: regenerating optimism for osteosarcoma treatment. Oncotarget 2018; 9:31562-31563. [PMID: 30167075 PMCID: PMC6114969 DOI: 10.18632/oncotarget.25820] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 07/16/2018] [Indexed: 01/21/2023] Open
|
50
|
Nakajima K, Takakura H, Shimizu Y, Ogawa M. Changes in plasma membrane damage inducing cell death after treatment with near-infrared photoimmunotherapy. Cancer Sci 2018; 109:2889-2896. [PMID: 29949672 PMCID: PMC6125438 DOI: 10.1111/cas.13713] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 06/25/2018] [Indexed: 12/12/2022] Open
Abstract
Near‐infrared photoimmunotherapy (NIR‐PIT) is a new cancer phototherapy modality using an antibody conjugated to a photosensitizer, IRDye700DX. When the conjugate binds to the plasma membrane and is exposed to NIR light, NIR‐PIT‐treated cells undergo swelling, and target‐selective necrotic/immunogenic cell death is induced. However, the cytotoxic mechanism of NIR‐PIT has not been elucidated. In order to understand the mechanism, it is important to elucidate how the damage to the plasma membrane induced by NIR light irradiation changes over time. Thus, in the present study, we investigated the changes in plasma membrane permeability using ions and molecules of various sizes. Na+ flowed into cells immediately after NIR light irradiation, even when the function of transporters or channels was blocked. Subsequently, fluorescent molecules larger than Na+ entered the cells, but the damage was not large enough for dextran to pass through at early time points. To assess these phenomena quantitatively, membrane permeability was estimated using radiolabeled ions and molecules: 111InCl3, 111In‐DTPA, and 3H‐H2O, and comparable results were obtained. Although minute plasma membrane perforations usually do not induce cell death, our results suggest that the minute damage induced by NIR‐PIT was irreversibly extended with time. In conclusion, minute plasma membrane damage is a trigger for the increase in plasma membrane permeability, cell swelling, and necrotic/immunogenic cell death in NIR‐PIT. Our findings provide new insight into the cytotoxic mechanism of NIR‐PIT.
Collapse
Affiliation(s)
- Kohei Nakajima
- Laboratory of Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Hideo Takakura
- Laboratory of Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Yoichi Shimizu
- Laboratory of Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Mikako Ogawa
- Laboratory of Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan.,PRESTO, Japan Science and Technology Agency, Kawaguchi, Japan
| |
Collapse
|