1
|
Lee JH, Lee CG, Kim MS, Kim S, Song M, Zhang H, Yang E, Kwon YH, Jung YH, Hyeon DY, Choi YJ, Oh S, Joe DJ, Kim TS, Jeon S, Huang Y, Kwon TH, Lee KJ. Deeply Implantable, Shape-Morphing, 3D MicroLEDs for Pancreatic Cancer Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024:e2411494. [PMID: 39679727 DOI: 10.1002/adma.202411494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 11/07/2024] [Indexed: 12/17/2024]
Abstract
Controlled photooxidation-mediated disruption of collagens in the tumor microenvironment can reduce desmoplasia and enhance immune responsiveness. However, achieving effective light delivery to solid tumors, particularly those with dynamic volumetric changes like pancreatic ductal adenocarcinoma (PDAC), remains challenging and limits the repeated and sustained photoactivation of drugs. Here, 3D, shape-morphing, implantable photonic devices (IPDs) are introduced that enable tumor-specific and continuous light irradiation for effective metronomic photodynamic therapy (mPDT). This IPD adheres seamlessly to the surface of orthotopic PDAC tumors, mitigating issues related to mechanical mismatch, delamination, and internal lesions. In freely moving mouse models, mPDT using the IPD with close adhesion significantly reduces desmoplastic tumor volume without causing cytotoxic effects in healthy tissues. These promising in vivo results underscore the potential of an adaptable and unidirectional IPD design in precisely targeting cancerous organs, suggesting a meaningful advance in light-based therapeutic technologies.
Collapse
Affiliation(s)
- Jae Hee Lee
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, 60208, USA
| | - Chae Gyu Lee
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Min Seo Kim
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Seungyeob Kim
- School of Electrical Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Myoung Song
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Haohui Zhang
- Department of Civil and Environmental Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Eunbyeol Yang
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Yoon Hee Kwon
- O2MEDi Incorporation, Ulsan, 44919, Republic of Korea
| | - Young Hoon Jung
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Dong Yeol Hyeon
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Yoon Ji Choi
- In Vivo Research Center, UNIST Central Research Facilities, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Seyong Oh
- Division of Electrical Engineering, Hanyang University ERICA, Ansan, 15588, Republic of Korea
| | - Daniel J Joe
- Division of Biomedical Metrology, Korea Research Institute of Standards and Science (KRISS), Daejeon, 34113, Republic of Korea
| | - Taek-Soo Kim
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Sanghun Jeon
- School of Electrical Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Yonggang Huang
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, 60208, USA
- Department of Civil and Environmental Engineering, Northwestern University, Evanston, IL, 60208, USA
- Departments of Mechanical Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Tae-Hyuk Kwon
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
- O2MEDi Incorporation, Ulsan, 44919, Republic of Korea
| | - Keon Jae Lee
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| |
Collapse
|
2
|
Bhandari C, Moffat A, Fakhry J, Malkoochi A, Nguyen A, Trinh B, Hoyt K, Story MD, Hasan T, Obaid G. A single photodynamic priming protocol augments delivery of ⍺-PD-L1 mAbs and induces immunogenic cell death in head and neck tumors. Photochem Photobiol 2024; 100:1647-1658. [PMID: 37818742 PMCID: PMC11006828 DOI: 10.1111/php.13865] [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: 08/14/2023] [Revised: 09/26/2023] [Accepted: 09/27/2023] [Indexed: 10/13/2023]
Abstract
Photodynamic priming (PDP) leverages the photobiological effects of subtherapeutic photodynamic therapy (PDT) regimens to modulate the tumor vasculature and stroma. PDP also sensitizes tumors to secondary therapies, such as immunotherapy by inducing a cascade of molecular events, including immunogenic cell death (ICD). We and others have shown that PDP improves the delivery of antibodies, among other theranostic agents. However, it is not known whether a single PDP protocol is capable of both inducing ICD in vivo and augmenting the delivery of immune checkpoint inhibitors. In this rapid communication, we show for the first time that a single PDP protocol using liposomal benzoporphyrin derivative (Lipo-BPD, 0.25 mg/kg) with 690 nm light (75 J/cm2, 100 mW/cm2) simultaneously doubles the delivery of ⍺-PD-L1 antibodies in murine AT-84 head and neck tumors and induces ICD in vivo. ICD was observed as a 3-11 fold increase in tumor cell exposure of damage-associated molecular patterns (Calreticulin, HMGB1, and HSP70). These findings suggest that this single, highly translatable PDP protocol using clinically relevant Lipo-BPD holds potential for improving immunotherapy outcomes in head and neck cancer. It can do so by simultaneously overcoming physical barriers to the delivery of immune checkpoint inhibitors, and biochemical barriers that contribute to immunosuppression.
Collapse
Affiliation(s)
- Chanda Bhandari
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
| | - Azophi Moffat
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
| | - John Fakhry
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
| | - Ashritha Malkoochi
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
| | - Austin Nguyen
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
| | - Brian Trinh
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
| | - Kenneth Hoyt
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
- Present Address: Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Michael D. Story
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Tayyaba Hasan
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Health Sciences and Technology, Harvard University and Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Girgis Obaid
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
| |
Collapse
|
3
|
Liao X, Cao Y, Zhong W, Zheng D, Jin L, Yao Y, Yang C. A Multifunctional Nanoparticle Dual Loading with Chlorin e6 and STING Agonist for Combinatorial Therapy of Melanoma. ACS APPLIED BIO MATERIALS 2024; 7:6768-6779. [PMID: 39289781 DOI: 10.1021/acsabm.4c00896] [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] [Indexed: 09/20/2024]
Abstract
Photodynamic therapy (PDT) is a noninvasive therapeutic approach that is effective in killing primary tumors with minimal surgical trauma, but its usage in metastatic lesions of melanoma is restricted by spatial limitations. Recently, stimulator of interferon genes (STING) agoinst-mediated innate immunity can activate the STING pathway and further promote dendritic cell (DC) maturation, tumor-specific cytotoxic T lymphocyte, and natural killer cell infiltration and has emerged as a promising approach for cancer therapy. Herein, the authors intriduce facile nanoparticles named HTCS, which can co-deliver STING agonist (2'3'-cGAMP) and a mitochondrial targeting modified photosensitizer (TPP-PEI-Ce6). While HTCS were intravenously injected to mice, they were endocytosed into tumor cells through hyaluronic acid-mediated active targeting. Thereafter, TPP-PEI-Ce6 was delivered to mitochondria to generate a large variety of reactive oxygen species and killed tumor cells effectively. Then the tumor cell debris further gave rise to immunogenic cell death, which played a role in immunosuppression. Furthermore, 2'3'-cGAMP contained in cell debris activated the STING pathway to promote the release of inflammatory cytokines and the maturation of DCs. As a consequence, the HTCS could achieve photodynamic multiple immunotherapy for melanoma. This work demonstrates multifunctional nanoparticles that efficiently inhibit tumors by PDT and reversing their immunosuppression to realize a versatile therapeutic strategy.
Collapse
Affiliation(s)
- Xukun Liao
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, Guizhou 550025, China
| | - Yong Cao
- Department of Pharmacy, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, China
| | - Wen Zhong
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, Guizhou 550025, China
| | - Dan Zheng
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, Guizhou 550025, China
| | - Lin Jin
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, Guizhou 550025, China
| | - Yongchao Yao
- Precision Medicine Translational Research Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Chengli Yang
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, Guizhou 550025, China
- Department of Pharmacy, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, China
| |
Collapse
|
4
|
Shah N, Soma SR, Quaye MB, Mahmoud D, Ahmed S, Malkoochi A, Obaid G. A Physiochemical, In Vitro, and In Vivo Comparative Analysis of Verteporfin-Lipid Conjugate Formulations: Solid Lipid Nanoparticles and Liposomes. ACS APPLIED BIO MATERIALS 2024; 7:4427-4441. [PMID: 38934648 PMCID: PMC11253097 DOI: 10.1021/acsabm.4c00316] [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: 03/05/2024] [Revised: 06/10/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024]
Abstract
VisudyneⓇ, a liposomal formulation of verteporfin (benzoporphyrin derivative; BPD), is the only nanomedicine approved to date for photodynamic therapy (PDT). We have previously demonstrated that BPD conjugated to the lysophospholipid 1-arachidoyl-2-hydroxy-sn-glycero-3-phosphocholine (BPD-PC) exhibits the greatest physical stability in liposomes, while maintaining cancer cell phototoxicity, from a panel of BPD lipid conjugates evaluated. In this study, we prepared 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC)-based solid lipid nanoparticles (LNPs) that stably entrap BPD-PC, which resemble the composition of the SpikevaxⓇ Moderna COVID-19 vaccine, and compared them to a DPPC based liposomal formulation (Lipo BPD-PC). We evaluated the photochemical, optical, and phototherapeutic properties of both formulations. We also investigated the in vivo distribution and tumor microdistribution of both formulations. Our results demonstrated that Lipo BPD-PC is able to generate 17% more singlet oxygen than LNP BPD-PC, while interestingly, LNP BPD-PC is able to produce 76% more hydroxyl radicals and/or peroxynitrite anion. Importantly, only 28% of BPD-PC leaches out of the LNP BPD-PC formulation during 7 days of incubation in serum at 37 °C, while 100% of BPD-PC leaches out of the Lipo BPD-PC formulation under the same conditions. Despite these differences, there was no significant difference in cellular uptake of BPD-PC or phototoxicity in CT1BA5 murine pancreatic cancer cells (derived from a genetically engineered mouse model). Interestingly, PDT using LNP BPD-PC was more efficient at inducing immunogenic cell death (calreticulin membrane translocation) than Lipo BPD-PC when using IC25 and IC50 PDT doses. In vivo studies revealed that CT1BA5 tumor fluorescence signals from BPD-PC were 2.41-fold higher with Lipo BPD-PC than with LNP BPD-PC; however, no significant difference was observed in tumor tissue selectivity or tumor penetration. As such, we present LNP BPD-PC as a unique and more stable nanoplatform to carry BPD lipid conjugates, such as BPD-PC, with a potential for future photodynamic immune priming studies and multiagent drug delivery.
Collapse
Affiliation(s)
- Nimit Shah
- Department
of Bioengineering, University of Texas at
Dallas, Richardson, Texas 75080, United States
| | - Siddharth Reddy Soma
- Department
of Bioengineering, University of Texas at
Dallas, Richardson, Texas 75080, United States
| | - Maxwell Bortei Quaye
- Department
of Bioengineering, University of Texas at
Dallas, Richardson, Texas 75080, United States
| | - Doha Mahmoud
- Department
of Bioengineering, University of Texas at
Dallas, Richardson, Texas 75080, United States
| | - Sarah Ahmed
- Department
of Bioengineering, University of Texas at
Dallas, Richardson, Texas 75080, United States
| | - Ashritha Malkoochi
- Department
of Bioengineering, University of Texas at
Dallas, Richardson, Texas 75080, United States
| | - Girgis Obaid
- Department
of Bioengineering, University of Texas at
Dallas, Richardson, Texas 75080, United States
| |
Collapse
|
5
|
Chanda B, Azophi Moffat, Shah N, Khan A, Quaye M, Fakhry J, Soma S, Nguyen A, Eroy M, Malkoochi A, Brekken R, Hasan T, Ferruzzi J, Obaid G. PD-L1 Immune Checkpoint Targeted Photoactivable Liposomes (iTPALs) Prime the Stroma of Pancreatic Tumors and Promote Self-Delivery. Adv Healthc Mater 2024; 13:e2304340. [PMID: 38324463 PMCID: PMC11281872 DOI: 10.1002/adhm.202304340] [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: 12/06/2023] [Indexed: 02/09/2024]
Abstract
Desmoplasia in pancreatic ductal adenocarcinoma (PDAC) limits the penetration and efficacy of therapies. It has been previously shown that photodynamic priming (PDP) using EGFR targeted photoactivable multi-inhibitor liposomes remediates desmoplasia in PDAC and doubles overall survival. Here, bifunctional PD-L1 immune checkpoint targeted photoactivable liposomes (iTPALs) that mediate both PDP and PD-L1 blockade are presented. iTPALs also improve phototoxicity in PDAC cells and induce immunogenic cell death. PDP using iTPALs reduces collagen density, thereby promoting self-delivery by 5.4-fold in collagen hydrogels, and by 2.4-fold in syngeneic CT1BA5 murine PDAC tumors. PDP also reduces tumor fibroblast content by 39.4%. Importantly, iTPALs also block the PD-1/PD-L1 immune checkpoint more efficiently than free α-PD-L1 antibodies. Only a single sub-curative priming dose using iTPALs provides 54.1% tumor growth inhibition and prolongs overall survival in mice by 42.9%. Overall survival directly correlates with the extent of tumor iTPAL self-delivery following PDP (Pearson's r = 0.670, p = 0.034), while no relationship is found for sham non-specific IgG constructs activated with light. When applied over multiple cycles, as is typical for immune checkpoint therapy, PDP using iTPALs promises to offer durable tumor growth delay and significant survival benefit in PDAC patients, especially when used to promote self-delivery of integrated chemo-immunotherapy regimens.
Collapse
Affiliation(s)
- Bhandari Chanda
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
| | - Azophi Moffat
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
| | - Nimit Shah
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
| | - Adil Khan
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
| | - Maxwell Quaye
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
| | - John Fakhry
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
| | - Siddharth Soma
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
| | - Austin Nguyen
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
| | - Menitte Eroy
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
| | - Ashritha Malkoochi
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
| | - Rolf Brekken
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Cancer Biology Graduate Program, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Tayyaba Hasan
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Health Sciences and Technology, Harvard University and Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jacopo Ferruzzi
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
| | - Girgis Obaid
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
| |
Collapse
|
6
|
Rahman I, Liang B, Sajid A, Ambudkar SV, Huang HC. Photodynamic priming modulates cellular ATP levels to overcome P-glycoprotein-mediated drug efflux in chemoresistant triple-negative breast cancer. Photochem Photobiol 2024. [PMID: 38824410 DOI: 10.1111/php.13970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 05/13/2024] [Accepted: 05/14/2024] [Indexed: 06/03/2024]
Abstract
P-glycoprotein (P-gp, ABCB1) is a well-researched ATP-binding cassette (ABC) drug efflux transporter linked to the development of cancer multidrug resistance (MDR). Despite extensive studies, approved therapies to safely inhibit P-gp in clinical settings are lacking, necessitating innovative strategies beyond conventional inhibitors or antibodies to reverse MDR. Photodynamic therapy is a globally approved cancer treatment that uses targeted, harmless red light to activate non-toxic photosensitizers, confining its cytotoxic photochemical effects to disease sites while sparing healthy tissues. This study demonstrates that photodynamic priming (PDP), a sub-cytotoxic photodynamic therapy process, can inhibit P-gp function by modulating cellular respiration and ATP levels in light accessible regions. Using chemoresistant (VBL-MDA-MB-231) and chemosensitive (MDA-MB-231) triple-negative breast cancer cell lines, we showed that PDP decreases mitochondrial membrane potential by 54.4% ± 30.4 and reduces mitochondrial ATP production rates by 94.9% ± 3.46. Flow cytometry studies showed PDP can effectively improve the retention of P-gp substrates (calcein) by up to 228.4% ± 156.3 in chemoresistant VBL-MDA-MB-231 cells, but not in chemosensitive MDA-MB-231 cells. Further analysis revealed that PDP did not alter the cell surface expression level of P-gp in VBL-MDA-MB-231 cells. These findings indicate that PDP can reduce cellular ATP below the levels that is required for the function of P-gp and improve intracellular substrate retention. We propose that PDP in combination with chemotherapy drugs, might improve the efficacy of chemotherapy and overcome cancer MDR.
Collapse
Affiliation(s)
- Idrisa Rahman
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Barry Liang
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Andaleeb Sajid
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Suresh V Ambudkar
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Huang-Chiao Huang
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA
| |
Collapse
|
7
|
Yaya-Candela AP, Ravagnani FG, Dietrich N, Sousa R, Baptista MS. Specific photodamage on HT-29 cancer cells leads to endolysosomal failure and autophagy blockage by cathepsin depletion. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2024; 255:112919. [PMID: 38677261 DOI: 10.1016/j.jphotobiol.2024.112919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 04/18/2024] [Indexed: 04/29/2024]
Abstract
Endolysosomes perform a wide range of cellular functions, including nutrient sensing, macromolecule digestion and recycling, as well as plasma membrane repair. Because of their high activity in cancerous cells, endolysosomes are attractive targets for the development of novel cancer treatments. Light-activated compounds termed photosensitizers (PS) can catalyze the oxidation of specific biomolecules and intracellular organelles. To selectively damage endosomes and lysosomes, HT-29 colorectal cancer cells were incubated with nanomolar concentrations of meso-tetraphenylporphine disulfonate (TPPS2a), an amphiphilic PS taken up via endocytosis and activated by green light (522 nm, 2.1 J.cm-1). Several cellular responses were characterized by a combination of immunofluorescence and immunoblotting assays. We showed that TPPS2a photosensitization blocked autophagic flux without extensive endolysosomal membrane rupture. Nevertheless, there was a severe functional failure of endolysosomes due to a decrease in CTSD (cathepsin D, 55%) and CTSB (cathepsin B, 52%) maturation. PSAP (prosaposin) processing (into saposins) was also considerably impaired, a fact that could be detrimental to glycosphingolipid homeostasis. Therefore, photosensitization of HT-29 cells previously incubated with a low concentration of TPPS2a promotes endolysosomal dysfunction, an effect that can be used to improve cancer therapies.
Collapse
Affiliation(s)
| | | | - Natasha Dietrich
- Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Rafaela Sousa
- Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil
| | | |
Collapse
|
8
|
Shimizu K, Kahramanian A, Jabbar MADA, Turna Demir F, Gokyer D, Uthamacumaran A, Rajan A, Saad MA, Gorham J, Wakimoto H, Martuza RL, Rabkin SD, Hasan T, Wakimoto H. Photodynamic augmentation of oncolytic virus therapy for central nervous system malignancies. Cancer Lett 2023; 572:216363. [PMID: 37619813 PMCID: PMC10529118 DOI: 10.1016/j.canlet.2023.216363] [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: 02/27/2023] [Revised: 08/09/2023] [Accepted: 08/18/2023] [Indexed: 08/26/2023]
Abstract
Oncolytic viruses (OVs) have emerged as a clinical therapeutic modality potentially effective for cancers that evade conventional therapies, including central nervous system malignancies. Rationally designed combinatorial strategies can augment the efficacy of OVs by boosting tumor-selective cytotoxicity and modulating the tumor microenvironment (TME). Photodynamic therapy (PDT) of cancer not only mediates direct neoplastic cell death but also primes the TME to sensitize the tumor to secondary therapies, allowing for the combination of two potentially synergistic therapies with broader targets. Here, we created G47Δ-KR, clinical oncolytic herpes simplex virus G47Δ that expresses photosensitizer protein KillerRed (KR). Optical properties and cytotoxic effects of G47Δ-KR infection followed by amber LED illumination (peak wavelength: 585-595 nm) were examined in human glioblastoma (GBM) and malignant meningioma (MM) models in vitro. G47Δ-KR infection of tumor cells mediated KR expression that was activated by LED and produced reactive oxygen species, leading to cell death that was more robust than G47Δ-KR without light. In vivo, we tested photodynamic-oncolytic virus (PD-OV) therapy employing intratumoral injection of G47Δ-KR followed by laser light tumor irradiation (wavelength: 585 nm) in GBM and MM xenografts. PD-OV therapy was feasible in these models and resulted in potent anti-tumor effects that were superior to G47Δ-KR alone (without laser light) or laser light alone. RNA sequencing analysis of post-treatment tumor samples revealed PD-OV therapy-induced increases in TME infiltration of variable immune cell types. This study thus demonstrated the proof-of-concept that G47Δ-KR enables PD-OV therapy for neuro-oncological malignancies and warrants further research to advance potential clinical translation.
Collapse
Affiliation(s)
- Kazuhide Shimizu
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA; Department of Neurosurgery, Tokyo Medical and Dental University, Tokyo, Japan
| | - Andranik Kahramanian
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA; Department of Neurosurgery, Royal Melbourne Hospital, Melbourne, Australia
| | | | - Fatma Turna Demir
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA; Department of Medical Services and Techniques, Medical Laboratory Techniques Programme, Vocational School of Health Services, Antalya Bilim University, Antalya, Turkey
| | - Dilan Gokyer
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Abicumaran Uthamacumaran
- McGill University, McGill Genome Center, Montreal, Canada; Douglas Mental Health University Institute, Department of Psychiatry, Montreal, Canada
| | - Anant Rajan
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Mohammad Ahsan Saad
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Joshua Gorham
- Department of Genetics, Harvard Medical School, Boston, MA, 02115, USA
| | - Hiroko Wakimoto
- Department of Genetics, Harvard Medical School, Boston, MA, 02115, USA
| | - Robert L Martuza
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Samuel D Rabkin
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Tayyaba Hasan
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA; Division of Health Sciences and Technology, Harvard University and Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Hiroaki Wakimoto
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA.
| |
Collapse
|
9
|
Zhou H, Tang D, Yu Y, Zhang L, Wang B, Karges J, Xiao H. Theranostic imaging and multimodal photodynamic therapy and immunotherapy using the mTOR signaling pathway. Nat Commun 2023; 14:5350. [PMID: 37660174 PMCID: PMC10475087 DOI: 10.1038/s41467-023-40826-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/11/2023] [Indexed: 09/04/2023] Open
Abstract
Tumor metastases are considered the leading cause of cancer-associated deaths. While clinically applied drugs have demonstrated to efficiently remove the primary tumor, metastases remain poorly accessible. To overcome this limitation, herein, the development of a theranostic nanomaterial by incorporating a chromophore for imaging and a photosensitizer for treatment of metastatic tumor sites is presented. The mechanism of action reveals that the nanoparticles are able to intervene by local generation of cellular damage through photodynamic therapy as well as by systemic induction of an immune response by immunotherapy upon inhibition of the mTOR signaling pathway which is of crucial importance for tumor onset, progression and metastatic spreading. The nanomaterial is able to strongly reduce the volume of the primary tumor as well as eradicates tumor metastases in a metastatic breast cancer and a multi-drug resistant patient-derived hepatocellular carcinoma models in female mice.
Collapse
Affiliation(s)
- Huiling Zhou
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, 100190, Beijing, P. R. China
- University of Chinese Academy of Sciences Beijing, 100049, Beijing, P. R. China
| | - Dongsheng Tang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, 100190, Beijing, P. R. China
- University of Chinese Academy of Sciences Beijing, 100049, Beijing, P. R. China
| | - Yingjie Yu
- State Key Laboratory of Organic-Inorganic Composites; Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, P.R. China
| | - Lingpu Zhang
- College of Life Science and Technology and State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Bin Wang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, 100190, Beijing, P. R. China
- University of Chinese Academy of Sciences Beijing, 100049, Beijing, P. R. China
| | - Johannes Karges
- Faculty of Chemistry and Biochemistry, Ruhr-University Bochum, Universitätsstrasse 150, Bochum, 44780, Germany.
| | - Haihua Xiao
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, 100190, Beijing, P. R. China.
- University of Chinese Academy of Sciences Beijing, 100049, Beijing, P. R. China.
| |
Collapse
|
10
|
Krysko DV, Demuynck R, Efimova I, Naessens F, Krysko O, Catanzaro E. In Vitro Veritas: From 2D Cultures to Organ-on-a-Chip Models to Study Immunogenic Cell Death in the Tumor Microenvironment. Cells 2022; 11:3705. [PMID: 36429133 PMCID: PMC9688238 DOI: 10.3390/cells11223705] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022] Open
Abstract
Immunogenic cell death (ICD) is a functionally unique form of cell death that promotes a T-cell-dependent anti-tumor immune response specific to antigens originating from dying cancer cells. Many anticancer agents and strategies induce ICD, but despite their robust effects in vitro and in vivo on mice, translation into the clinic remains challenging. A major hindrance in antitumor research is the poor predictive ability of classic 2D in vitro models, which do not consider tumor biological complexity, such as the contribution of the tumor microenvironment (TME), which plays a crucial role in immunosuppression and cancer evasion. In this review, we describe different tumor models, from 2D cultures to organ-on-a-chip technology, as well as spheroids and perfusion bioreactors, all of which mimic the different degrees of the TME complexity. Next, we discuss how 3D cell cultures can be applied to study ICD and how to increase the translational potential of the ICD inducers. Finally, novel research directions are provided regarding ICD in the 3D cellular context which may lead to novel immunotherapies for cancer.
Collapse
Affiliation(s)
- Dmitri V. Krysko
- Cell Death Investigation and Therapy Laboratory, Department of Human Structure and Repair, Ghent University, 9000 Ghent, Belgium
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| | - Robin Demuynck
- Cell Death Investigation and Therapy Laboratory, Department of Human Structure and Repair, Ghent University, 9000 Ghent, Belgium
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
| | - Iuliia Efimova
- Cell Death Investigation and Therapy Laboratory, Department of Human Structure and Repair, Ghent University, 9000 Ghent, Belgium
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
| | - Faye Naessens
- Cell Death Investigation and Therapy Laboratory, Department of Human Structure and Repair, Ghent University, 9000 Ghent, Belgium
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
| | - Olga Krysko
- Cell Death Investigation and Therapy Laboratory, Department of Human Structure and Repair, Ghent University, 9000 Ghent, Belgium
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| | - Elena Catanzaro
- Cell Death Investigation and Therapy Laboratory, Department of Human Structure and Repair, Ghent University, 9000 Ghent, Belgium
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
| |
Collapse
|
11
|
Saad MA, Hasan T. Spotlight on Photoactivatable Liposomes beyond Drug Delivery: An Enabler of Multitargeting of Molecular Pathways. Bioconjug Chem 2022; 33:2041-2064. [PMID: 36197738 DOI: 10.1021/acs.bioconjchem.2c00376] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The potential of photoactivating certain molecules, photosensitizers (PS), resulting in photochemical processes, has long been realized in the form of photodynamic therapy (PDT) for the management of several cancerous and noncancerous pathologies. With an improved understanding of the photoactivation process and its broader implications, efforts are being made to exploit the various facets of photoactivation, PDT, and the associated phenomenon of photodynamic priming in enhancing treatment outcomes, specifically in cancer therapeutics. The parallel emergence of nanomedicine, specifically liposome-based nanoformulations, and the convergence of the two fields of liposome-based drug delivery and PDT have led to the development of unique hybrid systems, which combine the exciting features of liposomes with adequate complementation through the photoactivation process. While initially liposomes carrying photosensitizers (PSs) were developed for enhancing the pharmacokinetics and the general applicability of PSs, more recently, PS-loaded liposomes, apart from their utility in PDT, have found several applications including enhanced targeting of drugs, coloading multiple therapeutic agents to enhance synergistic effects, imaging, priming, triggering drug release, and facilitating the escape of therapeutic agents from the endolysosomal complex. This review discusses the design strategies, potential, and unique attributes of these hybrid systems, with not only photoactivation as an attribute but also the ability to encapsulate multiple agents for imaging, biomodulation, priming, and therapy referred to as photoactivatable multiagent/inhibitor liposomes (PMILS) and their targeted versions─targeted PMILS (TPMILS). While liposomes have formed their own niche in nanotechnology and nanomedicine with several clinically approved formulations, we try to highlight how using PS-loaded liposomes could address some of the limitations and concerns usually associated with liposomes to overcome them and enhance their preclinical and clinical utility in the future.
Collapse
Affiliation(s)
- Mohammad A Saad
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Tayyaba Hasan
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States.,Division of Health Sciences and Technology, Harvard University and Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
12
|
Obaid G, Bano S, Thomsen H, Callaghan S, Shah N, Swain JWR, Jin W, Ding X, Cameron CG, McFarland SA, Wu J, Vangel M, Stoilova‐McPhie S, Zhao J, Mino‐Kenudson M, Lin C, Hasan T. Remediating Desmoplasia with EGFR-Targeted Photoactivable Multi-Inhibitor Liposomes Doubles Overall Survival in Pancreatic Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104594. [PMID: 35748165 PMCID: PMC9404396 DOI: 10.1002/advs.202104594] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 05/31/2022] [Indexed: 05/20/2023]
Abstract
Desmoplasia is characteristic of pancreatic ductal adenocarcinoma (PDAC), which exhibits 5-year survival rates of 3%. Desmoplasia presents physical and biochemical barriers that contribute to treatment resistance, yet depleting the stroma alone is unsuccessful and even detrimental to patient outcomes. This study is the first demonstration of targeted photoactivable multi-inhibitor liposomes (TPMILs) that induce both photodynamic and chemotherapeutic tumor insult, while simultaneously remediating desmoplasia in orthotopic PDAC. TPMILs targeted with cetuximab (anti-EGFR mAb) contain lipidated benzoporphyrin derivative (BPD-PC) photosensitizer and irinotecan. The desmoplastic tumors comprise human PDAC cells and patient-derived cancer-associated fibroblasts. Upon photoactivation, the TPMILs induce 90% tumor growth inhibition at only 8.1% of the patient equivalent dose of nanoliposomal irinotecan (nal-IRI). Without EGFR targeting, PMIL photoactivation is ineffective. TPMIL photoactivation is also sixfold more effective at inhibiting tumor growth than a cocktail of Visudyne-photodynamic therapy (PDT) and nal-IRI, and also doubles survival and extends progression-free survival by greater than fivefold. Second harmonic generation imaging reveals that TPMIL photoactivation reduces collagen density by >90% and increases collagen nonalignment by >103 -fold. Collagen nonalignment correlates with a reduction in tumor burden and survival. This single-construct phototoxic, chemotherapeutic, and desmoplasia-remediating regimen offers unprecedented opportunities to substantially extend survival in patients with otherwise dismal prognoses.
Collapse
Affiliation(s)
- Girgis Obaid
- Department of DermatologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
- Present address:
Department of BioengineeringUniversity of Texas at DallasRichardsonTX75080USA
| | - Shazia Bano
- Department of DermatologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - Hanna Thomsen
- Department of DermatologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - Susan Callaghan
- Department of DermatologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - Nimit Shah
- Present address:
Department of BioengineeringUniversity of Texas at DallasRichardsonTX75080USA
| | - Joseph W. R. Swain
- Department of DermatologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - Wendong Jin
- Department of DermatologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - Xiadong Ding
- Department of DermatologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | | | | | - Juwell Wu
- Department of DermatologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - Mark Vangel
- Department of DermatologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | | | - Jie Zhao
- Department of DermatologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - Mari Mino‐Kenudson
- Department of DermatologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - Charles Lin
- Department of DermatologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - Tayyaba Hasan
- Department of DermatologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
- Division of Health Sciences and TechnologyHarvard University and Massachusetts Institute of TechnologyCambridgeMA02139USA
| |
Collapse
|
13
|
Bhandari C, Fakhry J, Eroy M, Song JJ, Samkoe K, Hasan T, Hoyt K, Obaid G. Towards Photodynamic Image-Guided Surgery of Head and Neck Tumors: Photodynamic Priming Improves Delivery and Diagnostic Accuracy of Cetuximab-IRDye800CW. Front Oncol 2022; 12:853660. [PMID: 35837101 PMCID: PMC9273965 DOI: 10.3389/fonc.2022.853660] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 05/06/2022] [Indexed: 11/17/2022] Open
Abstract
Fluorescence image-guided surgery (IGS) using antibody conjugates of the fluorophore IRDye800CW have revolutionized the surgical debulking of tumors. Cetuximab, an anti-epidermal growth factor receptor (EGFR) monoclonal antibody, conjugated to IRDye800CW (Cet-IRDye800) is the first molecular targeted antibody probe to be used for IGS in head and neck cancer patients. In addition to surgical debulking, Cetuximab-targeted photodynamic therapy (photoimmunotherapy; PIT) is emerging in the clinic as a powerful modality for head and neck tumor photodestruction. A plethora of other photoactivable agents are also in clinical trials for photodynamic-based therapies of head and neck cancer. Considering the vascular and stromal modulating effects of sub-therapeutic photodynamic therapy, namely photodynamic priming (PDP), this study explores the potential synergy between PDP and IGS for a novel photodynamic image-guided surgery (P-IGS) strategy. To the best of our knowledge, this is the first demonstration that PDP of the tumor microenvironment can augment the tumor delivery of full-length antibodies, namely Cet-IRDye800. In this study, we demonstrate a proof-of-concept that PDP primes orthotopic FaDu human head and neck tumors in mice for P-IGS by increasing the delivery of Cet-IRDye800 by up to 138.6%, by expediting its interstitial accumulation by 10.5-fold, and by increasing its fractional tumor coverage by 49.5% at 1 h following Cet-IRDye800 administration. Importantly, PDP improves the diagnostic accuracy of tumor detection by up to 264.2% with respect to vicinal salivary glands at 1 h. As such, PDP provides a time-to-surgery benefit by reducing the time to plateau 10-fold from 25.7 h to 2.5 h. We therefore propose that a pre-operative PDP regimen can expedite and augment the accuracy of IGS-mediated surgical debulking of head and neck tumors and reduce the time-to-IGS. Furthermore, this P-IGS regimen, can also enable a forward-looking post-operative protocol for the photodestruction of unresectable microscopic disease in the surgical bed. Beyond this scope, the role of PDP in the homogenous delivery of diagnostic, theranostic and therapeutic antibodies in solid tumors is of considerable significance to the wider community.
Collapse
Affiliation(s)
- Chanda Bhandari
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, United States
| | - John Fakhry
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, United States
| | - Menitte Eroy
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, United States
| | - Jane Junghwa Song
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, United States
| | - Kimberley Samkoe
- Thayer School of Engineering, Dartmouth College, Hanover, NH, United States
| | - Tayyaba Hasan
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Division of Health Sciences and Technology, Harvard University and Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Kenneth Hoyt
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, United States
| | - Girgis Obaid
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, United States
| |
Collapse
|
14
|
Nkune NW, Simelane NWN, Montaseri H, Abrahamse H. Photodynamic Therapy-Mediated Immune Responses in Three-Dimensional Tumor Models. Int J Mol Sci 2021; 22:12618. [PMID: 34884424 PMCID: PMC8657498 DOI: 10.3390/ijms222312618] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/16/2021] [Accepted: 11/16/2021] [Indexed: 12/12/2022] Open
Abstract
Photodynamic therapy (PDT) is a promising non-invasive phototherapeutic approach for cancer therapy that can eliminate local tumor cells and produce systemic antitumor immune responses. In recent years, significant efforts have been made in developing strategies to further investigate the immune mechanisms triggered by PDT. The majority of in vitro experimental models still rely on the two-dimensional (2D) cell cultures that do not mimic a three-dimensional (3D) cellular environment in the human body, such as cellular heterogeneity, nutrient gradient, growth mechanisms, and the interaction between cells as well as the extracellular matrix (ECM) and therapeutic resistance to anticancer treatments. In addition, in vivo animal studies are highly expensive and time consuming, which may also show physiological discrepancies between animals and humans. In this sense, there is growing interest in the utilization of 3D tumor models, since they precisely mimic different features of solid tumors. This review summarizes the characteristics and techniques for 3D tumor model generation. Furthermore, we provide an overview of innate and adaptive immune responses induced by PDT in several in vitro and in vivo tumor models. Future perspectives are highlighted for further enhancing PDT immune responses as well as ideal experimental models for antitumor immune response studies.
Collapse
Affiliation(s)
| | | | | | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Johannesburg 2028, South Africa; (N.W.N.); (N.W.N.S.); (H.M.)
| |
Collapse
|
15
|
Pigula M, Mai Z, Anbil S, Choi MG, Wang K, Maytin E, Pogue B, Hasan T. Dramatic Reduction of Distant Pancreatic Metastases Using Local Light Activation of Verteporfin with Nab-Paclitaxel. Cancers (Basel) 2021; 13:5781. [PMID: 34830934 PMCID: PMC8616053 DOI: 10.3390/cancers13225781] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/06/2021] [Accepted: 11/15/2021] [Indexed: 12/16/2022] Open
Abstract
Despite substantial drug development efforts, pancreatic adenocarcinoma (PDAC) remains a difficult disease to treat, and surgical resection is the only potentially curative option. Unfortunately, 80% of patients are ineligible for surgery due to the presence of invasive disease and/or distant metastases at the time of diagnosis. Treatment strategies geared towards reclassifying these patients as surgical candidates by reducing metastatic burden represents the most promising approach to improve long-term survival. We describe a photodynamic therapy (PDT) based approach that, in combination with the first-line chemotherapeutic nab-paclitaxel, effectively addresses distant metastases in three separate orthotopic PDAC models in immunodeficient mice. In addition to effectively controlling local tumor growth, PDT plus nab-paclitaxel primes the tumor to elicit systemic effects and reduce or abrogate metastases. This combination dramatically inhibits (up to 100%) the eventual development of metastases in models of early stage PDAC, and completely eliminates metastasis in 55% of animals with already established distant disease in late-stage models. Our findings suggest that this light activation process initiates local biological and/or physiological changes within the tumor microenvironment that can be leveraged to treat both localized and distant disease, and potentially reclassify patients with previously inoperable disease as surgical candidates.
Collapse
Affiliation(s)
- Michael Pigula
- Department of Chemistry, Scripps Research, La Jolla, CA 92037, USA;
| | - Zhiming Mai
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA;
| | - Sriram Anbil
- David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA;
| | - Myung-Gyu Choi
- Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul 137-040, Korea;
| | - Kenneth Wang
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55902, USA;
| | - Edward Maytin
- Department of Dermatology, Cleveland Clinic, Cleveland, OH 44195, USA;
| | - Brian Pogue
- Department of Engineering Sciences, Dartmouth College, Hanover, NH 03755, USA;
| | - Tayyaba Hasan
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA;
| |
Collapse
|
16
|
Sorrin AJ, Liu C, Cicalo J, Reader J, Najafali D, Zhang Y, Roque DM, Huang HC. Photodynamic Priming Improves the Anti-Migratory Activity of Prostaglandin E Receptor 4 Antagonist in Cancer Cells In Vitro. Cancers (Basel) 2021; 13:5259. [PMID: 34771424 PMCID: PMC8582354 DOI: 10.3390/cancers13215259] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 10/05/2021] [Accepted: 10/10/2021] [Indexed: 12/11/2022] Open
Abstract
The combination of photodynamic agents and biological inhibitors is rapidly gaining attention for its promise and approval in treating advanced cancer. The activity of photodynamic treatment is mainly governed by the formation of reactive oxygen species upon light activation of photosensitizers. Exposure to reactive oxygen species above a threshold dose can induce cellular damage and cancer cell death, while the surviving cancer cells are "photodynamically primed", or sensitized, to respond better to other drugs and biological treatments. Here, we report a new combination regimen of photodynamic priming (PDP) and prostaglandin E2 receptor 4 (EP4) inhibition that reduces the migration and invasion of two human ovarian cancer cell lines (OVCAR-5 and CAOV3) in vitro. PDP is achieved by red light activation of the FDA-approved photosensitizer, benzoporphyrin derivative (BPD), or a chemical conjugate composed of the BPD linked to cetuximab, an anti-epithelial growth factor receptor (EGFR) antibody. Immunoblotting data identify co-inhibition of EGFR, cAMP-response element binding protein (CREB), and extracellular signal-regulated kinase 1/2 (ERK1/2) as key in the signaling cascades modulated by the combination of EGFR-targeted PDP and EP4 inhibition. This study provides valuable insights into the development of a molecular-targeted photochemical strategy to improve the anti-metastatic effects of EP4 receptor antagonists.
Collapse
Affiliation(s)
- Aaron J. Sorrin
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; (A.J.S.); (C.L.); (J.C.); (D.N.)
| | - Cindy Liu
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; (A.J.S.); (C.L.); (J.C.); (D.N.)
| | - Julia Cicalo
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; (A.J.S.); (C.L.); (J.C.); (D.N.)
| | - Jocelyn Reader
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (J.R.); (D.M.R.)
- University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, MD 21201, USA;
| | - Daniel Najafali
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; (A.J.S.); (C.L.); (J.C.); (D.N.)
| | - Yuji Zhang
- University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, MD 21201, USA;
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Dana M. Roque
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (J.R.); (D.M.R.)
- University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, MD 21201, USA;
| | - Huang-Chiao Huang
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; (A.J.S.); (C.L.); (J.C.); (D.N.)
- University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, MD 21201, USA;
| |
Collapse
|