1
|
Sweeney EE, Sekhri P, Muniraj N, Chen J, Feng S, Terao J, Chin SJ, Schmidt DE, Bollard CM, Cruz CRY, Fernandes R. Photothermal Prussian blue nanoparticles generate potent multi-targeted tumor-specific T cells as an adoptive cell therapy. Bioeng Transl Med 2024; 9:e10639. [PMID: 38818122 PMCID: PMC11135148 DOI: 10.1002/btm2.10639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 12/05/2023] [Accepted: 12/13/2023] [Indexed: 06/01/2024] Open
Abstract
Prussian blue nanoparticle-based photothermal therapy (PBNP-PTT) is an effective tumor treatment capable of eliciting an antitumor immune response. Motivated by the ability of PBNP-PTT to potentiate endogenous immune responses, we recently demonstrated that PBNP-PTT could be used ex vivo to generate tumor-specific T cells against glioblastoma (GBM) cell lines as an adoptive T cell therapy (ATCT). In this study, we further developed this promising T cell development platform. First, we assessed the phenotype and function of T cells generated using PBNP-PTT. We observed that PBNP-PTT facilitated CD8+ T cell expansion from healthy donor PBMCs that secreted IFNγ and TNFα and upregulated CD107a in response to engagement with target U87 cells, suggesting specific antitumor T cell activation and degranulation. Further, CD8+ effector and effector memory T cell populations significantly expanded after co-culture with U87 cells, consistent with tumor-specific effector responses. In orthotopically implanted U87 GBM tumors in vivo, PBNP-PTT-derived T cells effectively reduced U87 tumor growth and generated long-term survival in >80% of tumor-bearing mice by Day 100, compared to 0% of mice treated with PBS, non-specific T cells, or T cells expanded from lysed U87 cells, demonstrating an enhanced antitumor efficacy of this ATCT platform. Finally, we tested the generalizability of our approach by generating T cells targeting medulloblastoma (D556), breast cancer (MDA-MB-231), neuroblastoma (SH-SY5Y), and acute monocytic leukemia (THP-1) cell lines. The resulting T cells secreted IFNγ and exerted increased tumor-specific cytolytic function relative to controls, demonstrating the versatility of PBNP-PTT in generating tumor-specific T cells for ATCT.
Collapse
Affiliation(s)
- Elizabeth E. Sweeney
- Department of Biochemistry & Molecular Medicine, School of Medicine and Health SciencesGeorge Washington UniversityWashingtonDistrict of ColumbiaUSA
- Center for Cancer and Immunology ResearchChildren's National HospitalWashingtonDistrict of ColumbiaUSA
| | - Palak Sekhri
- Center for Cancer and Immunology ResearchChildren's National HospitalWashingtonDistrict of ColumbiaUSA
- The Integrated Biomedical Sciences Program, School of Medicine and Health SciencesGeorge Washington UniversityWashingtonDistrict of ColumbiaUSA
| | - Nethaji Muniraj
- The Integrated Biomedical Sciences Program, School of Medicine and Health SciencesGeorge Washington UniversityWashingtonDistrict of ColumbiaUSA
| | - Jie Chen
- Center for Cancer and Immunology ResearchChildren's National HospitalWashingtonDistrict of ColumbiaUSA
| | - Sally Feng
- Center for Cancer and Immunology ResearchChildren's National HospitalWashingtonDistrict of ColumbiaUSA
- George Washington Cancer Center, School of Medicine and Health SciencesGeorge Washington UniversityWashingtonDistrict of ColumbiaUSA
| | - Joshua Terao
- The Integrated Biomedical Sciences Program, School of Medicine and Health SciencesGeorge Washington UniversityWashingtonDistrict of ColumbiaUSA
| | - Samantha J. Chin
- Center for Cancer and Immunology ResearchChildren's National HospitalWashingtonDistrict of ColumbiaUSA
- George Washington Cancer Center, School of Medicine and Health SciencesGeorge Washington UniversityWashingtonDistrict of ColumbiaUSA
| | - Danielle E. Schmidt
- Center for Cancer and Immunology ResearchChildren's National HospitalWashingtonDistrict of ColumbiaUSA
| | - Catherine M. Bollard
- Center for Cancer and Immunology ResearchChildren's National HospitalWashingtonDistrict of ColumbiaUSA
- The Integrated Biomedical Sciences Program, School of Medicine and Health SciencesGeorge Washington UniversityWashingtonDistrict of ColumbiaUSA
| | - Conrad Russell Y. Cruz
- Center for Cancer and Immunology ResearchChildren's National HospitalWashingtonDistrict of ColumbiaUSA
- The Integrated Biomedical Sciences Program, School of Medicine and Health SciencesGeorge Washington UniversityWashingtonDistrict of ColumbiaUSA
| | - Rohan Fernandes
- Center for Cancer and Immunology ResearchChildren's National HospitalWashingtonDistrict of ColumbiaUSA
- George Washington Cancer Center, School of Medicine and Health SciencesGeorge Washington UniversityWashingtonDistrict of ColumbiaUSA
- Department of Medicine, School of Medicine and Health SciencesGeorge Washington UniversityWashingtonDistrict of ColumbiaUSA
| |
Collapse
|
2
|
Rechberger JS, Millesi E, Power EA, Wang H, Mardini S, Spinner RJ, Daniels DJ. Shaping Our Understanding of Malignant Peripheral Nerve Sheath Tumor: A Bibliometric Analysis of the 100 Most-Cited Articles. World Neurosurg 2024; 184:293-302.e11. [PMID: 38219800 DOI: 10.1016/j.wneu.2024.01.054] [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: 09/11/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 01/16/2024]
Abstract
BACKGROUND Malignant peripheral nerve sheath tumors (MPNSTs) are rare yet highly aggressive soft tissue sarcomas of mesenchymal origin, characterized by a heterogeneous pathological spectrum, limited therapeutic options, and high metastatic potential. METHODS Here, the authors conducted a comprehensive bibliometric analysis of the 100 most-cited MPNST articles by utilizing Elsevier's Scopus to identify all relevant published and indexed articles referring to MPNST, thereby aiming to elucidate the pertinent research findings regarding the disease's pathophysiology and therapeutic advancements. Articles were classified as basic science or clinical and analyzed for various bibliometric parameters. RESULTS The majority of articles (75%) focused on clinical aspects, reflecting the extensive clinicopathological characterization of MPNSTs. Notable studies investigated prognostic factors, histological and immunohistochemical features, and diagnostic modalities. The identification of loss of function mutations in the polycomb repressive complex 2 emerged as a pivotal role, as it opened avenues for potential targets for novel therapeutic interventions. Newer articles (published in or after 2006) demonstrated higher citation rates, suggesting evolving impact and collaboration. CONCLUSIONS This bibliometric analysis showed how developments in the understanding of MPNST pathophysiology and the creation of novel therapeutic strategies occurred throughout time. Changes that have been noticed recently could portend future innovative therapeutic approaches.
Collapse
Affiliation(s)
- Julian S Rechberger
- Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota, USA; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, Minnesota, USA
| | - Elena Millesi
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Mayo Clinic, Rochester, Minnesota, USA; Research Laboratory of the Division of Plastic and Reconstructive Surgery, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - Erica A Power
- Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois, USA
| | - Huan Wang
- Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Samir Mardini
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Robert J Spinner
- Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - David J Daniels
- Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota, USA; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, Minnesota, USA.
| |
Collapse
|
3
|
Millesi E, Rechberger JS, Wang H, Mardini S, Spinner RJ, Daniels DJ. Advancements in therapeutic approaches for malignant peripheral nerve sheath tumor. Ther Deliv 2023; 14:385-389. [PMID: 37464750 DOI: 10.4155/tde-2023-0014] [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: 07/20/2023] Open
Abstract
Tweetable abstract Emerging targeted therapies offer hope for malignant peripheral nerve sheath tumor. Innovative drug delivery enhances potential treatments. #MPNST #TargetedTherapies #TherapeuticDelivery.
Collapse
Affiliation(s)
- Elena Millesi
- Division of Plastic & Reconstructive Surgery, Department of Surgery, Mayo Clinic, Rochester, MN 55905, USA
- Research Laboratory of the Division of Plastic & Reconstructive Surgery, Department of Surgery, Medical University of Vienna, Vienna, 1090, Austria
| | - Julian S Rechberger
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, USA
| | - Huan Wang
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Samir Mardini
- Division of Plastic & Reconstructive Surgery, Department of Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Robert J Spinner
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - David J Daniels
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, USA
| |
Collapse
|
4
|
Saleh M, Dib A, Beaini S, Saad C, Faraj S, El Joueid Y, Kotob Y, Saoudi L, Emmanuel N. Neurofibromatosis type 1 system-based manifestations and treatments: a review. Neurol Sci 2023; 44:1931-1947. [PMID: 36826455 DOI: 10.1007/s10072-023-06680-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 02/11/2023] [Indexed: 02/25/2023]
Abstract
INTRODUCTION Neurofibromatosis type 1 (NF1) is a genetic disorder caused by a mutation in the NF1 gene. This disease presents with various system-based manifestations, including cardiac, musculoskeletal, and neuronal issues, which have been well-studied in previous research and have prompted the development of current and emerging treatments. These treatments, mainly medications targeting specific manifestations of NF1, aim to mitigate the negative impacts of the disease on patients' lives. NF1 is associated with an increased risk of malignancy and a significant decrease in life expectancy. In this paper, we review the current and emerging treatments for NF1 in relation to its system-based manifestations. METHODS We conducted an extensive literature search using specific keywords through databases such as PubMed, Scopus, and Cochrane. The articles we found were compiled and subjected to strict inclusion and exclusion criteria. RESULTS Pharmacological advances have led to the development of products that hold promise as future treatments for NF1. Given the diverse manifestations that can affect multiple organ systems in patients with NF1, it is important to consider a variety of treatment options to achieve optimal results. However, one of the major challenges in diagnosing and treating NF1 is that patients present asymptomatically, making it necessary to rely on clinical features for diagnosis. CONCLUSION In conclusion, NF1 is a complex disease with varying manifestations and a growing field of pharmacologic treatments. The information presented in this article synthesizes current knowledge and available therapies for NF1.
Collapse
Affiliation(s)
- Mustafa Saleh
- Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - AlFadel Dib
- Faculty of Medicine & Medical Sciences, University of Balamand, Koura, Lebanon
| | - Sarah Beaini
- Faculty of Medicine, Université Saint Joseph de Beirut, Damascus Street, Lebanon
| | - Charbel Saad
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Byblos, Lebanon
| | - Sary Faraj
- Department of Anatomy, Cell Biology, and Physiological Cell Sciences, AUB- FM and MC, Beirut, Lebanon
| | - Youssef El Joueid
- Department of Medicine, School of Health Sciences, University of Georgia, Tbilisi, Georgia
| | - Yasmine Kotob
- Faculty of Medicine, Université Saint Joseph de Beirut, Damascus Street, Lebanon
| | - Lara Saoudi
- Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Nancy Emmanuel
- Department of Dermatology, Faculty of Medicine of the University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
5
|
Ledezma DK, Balakrishnan PB, Shukla A, Medina JA, Chen J, Oakley E, Bollard CM, Shafirstein G, Miscuglio M, Fernandes R. Interstitial Photothermal Therapy Generates Durable Treatment Responses in Neuroblastoma. Adv Healthc Mater 2022; 11:e2201084. [PMID: 35943173 PMCID: PMC9588730 DOI: 10.1002/adhm.202201084] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 07/23/2022] [Indexed: 01/28/2023]
Abstract
Photothermal therapy (PTT) represents a promising modality for tumor control typically using infrared light-responsive nanoparticles illuminated by a wavelength-matched external laser. However, due to the constraints of light penetration, PTT is generally restricted to superficially accessible tumors. With the goal of extending the benefits of PTT to all tumor settings, interstitial PTT (I-PTT) is evaluated by the photothermal activation of intratumorally administered Prussian blue nanoparticles with a laser fiber positioned interstitially within the tumor. This interstitial fiber, which is fitted with a terminal diffuser, distributes light within the tumor microenvironment from the "inside-out" as compared to from the "outside-in" traditionally observed during superficially administered PTT (S-PTT). I-PTT improves the heating efficiency and heat distribution within a target treatment area compared to S-PTT. Additionally, I-PTT generates increased cytotoxicity and thermal damage at equivalent thermal doses, and elicits immunogenic cell death at lower thermal doses in targeted neuroblastoma tumor cells compared to S-PTT. In vivo, I-PTT induces significantly higher long-term tumor regression, lower rates of tumor recurrence, and improved long-term survival in multiple syngeneic murine models of neuroblastoma. This study highlights the significantly enhanced therapeutic benefit of I-PTT compared to traditional S-PTT as a promising treatment modality for solid tumors.
Collapse
Affiliation(s)
- Debbie K. Ledezma
- The George Washington Cancer CenterThe George Washington University800 22nd St NW, 8300 Science and Engineering HallWashingtonDC20052USA
- The Institute for Biomedical SciencesThe George Washington University2300 Eye Street NW, Ross Hall Room 561WashingtonDC20037USA
| | - Preethi B. Balakrishnan
- The George Washington Cancer CenterThe George Washington University800 22nd St NW, 8300 Science and Engineering HallWashingtonDC20052USA
| | - Anshi Shukla
- The George Washington Cancer CenterThe George Washington University800 22nd St NW, 8300 Science and Engineering HallWashingtonDC20052USA
| | - Jacob A. Medina
- The George Washington Cancer CenterThe George Washington University800 22nd St NW, 8300 Science and Engineering HallWashingtonDC20052USA
- The Institute for Biomedical SciencesThe George Washington University2300 Eye Street NW, Ross Hall Room 561WashingtonDC20037USA
| | - Jie Chen
- The George Washington Cancer CenterThe George Washington University800 22nd St NW, 8300 Science and Engineering HallWashingtonDC20052USA
| | - Emily Oakley
- Photodynamic Therapy CenterRoswell Park Comprehensive Cancer CenterDepartment of Cell Stress BiologyRoswell Park, Elm and Carlton StreetsBuffaloNY14263USA
| | - Catherine M. Bollard
- The George Washington Cancer CenterThe George Washington University800 22nd St NW, 8300 Science and Engineering HallWashingtonDC20052USA
- Center for Cancer and Immunology ResearchChildren's National Hospital111 Michigan Ave NWWashingtonDC20010USA
| | - Gal Shafirstein
- Photodynamic Therapy CenterRoswell Park Comprehensive Cancer CenterDepartment of Cell Stress BiologyRoswell Park, Elm and Carlton StreetsBuffaloNY14263USA
| | - Mario Miscuglio
- Department of Electrical and Computer EngineeringThe George Washington University800 22nd St NW, 5000 Science and Engineering HallWashingtonDC20052USA
| | - Rohan Fernandes
- The George Washington Cancer CenterThe George Washington University800 22nd St NW, 8300 Science and Engineering HallWashingtonDC20052USA
- Department of MedicineThe George Washington University2150 Pennsylvania Avenue, NW, Suite 8‐416WashingtonDC20037USA
| |
Collapse
|
6
|
Bonan NF, Ledezma DK, Tovar MA, Balakrishnan PB, Fernandes R. Anti-Fn14-Conjugated Prussian Blue Nanoparticles as a Targeted Photothermal Therapy Agent for Glioblastoma. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:2645. [PMID: 35957076 PMCID: PMC9370342 DOI: 10.3390/nano12152645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/26/2022] [Accepted: 07/28/2022] [Indexed: 12/10/2022]
Abstract
Prussian blue nanoparticles (PBNPs) are effective photothermal therapy (PTT) agents: they absorb near-infrared radiation and reemit it as heat via phonon-phonon relaxations that, in the presence of tumors, can induce thermal and immunogenic cell death. However, in the context of central nervous system (CNS) tumors, the off-target effects of PTT have the potential to result in injury to healthy CNS tissue. Motivated by this need for targeted PTT agents for CNS tumors, we present a PBNP formulation that targets fibroblast growth factor-inducible 14 (Fn14)-expressing glioblastoma cell lines. We conjugated an antibody targeting Fn14, a receptor abundantly expressed on many glioblastomas but near absent on healthy CNS tissue, to PBNPs (aFn14-PBNPs). We measured the attachment efficiency of aFn14 onto PBNPs, the size and stability of aFn14-PBNPs, and the ability of aFn14-PBNPs to induce thermal and immunogenic cell death and target and treat glioblastoma tumor cells in vitro. aFn14 remained stably conjugated to the PBNPs for at least 21 days. Further, PTT with aFn14-PBNPs induced thermal and immunogenic cell death in glioblastoma tumor cells. However, in a targeted treatment assay, PTT was only effective in killing glioblastoma tumor cells when using aFn14-PBNPs, not when using PBNPs alone. Our methodology is novel in its targeting moiety, tumor application, and combination with PTT. To the best of our knowledge, PBNPs have not been investigated as a targeted PTT agent in glioblastoma via conjugation to aFn14. Our results demonstrate a novel and effective method for delivering targeted PTT to aFn14-expressing tumor cells via aFn14 conjugation to PBNPs.
Collapse
Affiliation(s)
- Nicole F. Bonan
- George Washington Cancer Center, George Washington University, Washington, DC 20052, USA; (N.F.B.); (D.K.L.); (M.A.T.); (P.B.B.)
- Institute for Biomedical Sciences, George Washington University, Washington, DC 20052, USA
| | - Debbie K. Ledezma
- George Washington Cancer Center, George Washington University, Washington, DC 20052, USA; (N.F.B.); (D.K.L.); (M.A.T.); (P.B.B.)
- Institute for Biomedical Sciences, George Washington University, Washington, DC 20052, USA
| | - Matthew A. Tovar
- George Washington Cancer Center, George Washington University, Washington, DC 20052, USA; (N.F.B.); (D.K.L.); (M.A.T.); (P.B.B.)
- School of Medicine and Health Sciences, George Washington University, Washington, DC 20052, USA
| | - Preethi B. Balakrishnan
- George Washington Cancer Center, George Washington University, Washington, DC 20052, USA; (N.F.B.); (D.K.L.); (M.A.T.); (P.B.B.)
| | - Rohan Fernandes
- George Washington Cancer Center, George Washington University, Washington, DC 20052, USA; (N.F.B.); (D.K.L.); (M.A.T.); (P.B.B.)
- Institute for Biomedical Sciences, George Washington University, Washington, DC 20052, USA
- School of Medicine and Health Sciences, George Washington University, Washington, DC 20052, USA
- Department of Medicine, George Washington University, Washington, DC 20052, USA
| |
Collapse
|
7
|
Gonzalez-Muñoz T, Kim A, Ratner N, Peinado H. The need for new treatments targeting MPNST: the potential of strategies combining MEK inhibitors with antiangiogenic agents. Clin Cancer Res 2022; 28:3185-3195. [PMID: 35446392 DOI: 10.1158/1078-0432.ccr-21-3760] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 02/01/2022] [Accepted: 04/04/2022] [Indexed: 11/16/2022]
Abstract
Malignant Peripheral Nerve Sheath Tumors (MPNSTs) are aggressive soft tissue sarcomas that represent an important clinical challenge, particularly given their strong tendency to relapse and metastasize, and their relatively poor response to conventional therapies. To date, targeted, non-cytotoxic treatments have demonstrated limited clinical success with MPNSTs, highlighting the need to explore other key pathways in order to find novel, improved therapeutic approaches. Here, we review evidence supporting the crucial role of the RAS/MEK/ERK pathway and angiogenesis in MPNST pathogenesis, and we focus on the potential of therapies targeting these pathways to treat this disease. We also present works suggesting that the combination of MEK inhibitors and anti-angiogenic agents could represent a promising therapeutic strategy to manage MPNSTs. In support of this notion, we discuss the preclinical rational and clinical benefits of this combination therapy in other solid tumor types. Finally, we describe other emerging therapeutic approaches that could improve patient outcomes in MPNSTs, such as immune-based therapies.
Collapse
Affiliation(s)
| | - AeRang Kim
- Children's National Hospital, Washington, DC, United States
| | - Nancy Ratner
- Cincinnati Children's Hospital Medical Center, Cincinnati, United States
| | - Héctor Peinado
- Spanish National Cancer Research Centre, Madrid, Madrid, Spain
| |
Collapse
|
8
|
Balakrishnan PB, Ledezma DK, Cano-Mejia J, Andricovich J, Palmer E, Patel VA, Latham PS, Yvon ES, Villagra A, Fernandes R, Sweeney EE. CD137 agonist potentiates the abscopal efficacy of nanoparticle-based photothermal therapy for melanoma. NANO RESEARCH 2022; 15:2300-2314. [PMID: 36089987 PMCID: PMC9455608 DOI: 10.1007/s12274-021-3813-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Despite the promise of immunotherapy such as the immune checkpoint inhibitors (ICIs) anti-PD-1 and anti-CTLA-4 for advanced melanoma, only 26%-52% of patients respond, and many experience grade III/IV immune-related adverse events. Motivated by the need for an effective therapy for patients non-responsive to clinically approved ICIs, we have developed a novel nanoimmunotherapy that combines locally administered Prussian blue nanoparticle-based photothermal therapy (PBNP-PTT) with systemically administered agonistic anti-CD137 monoclonal antibody therapy (aCD137). PBNP-PTT was administered at various thermal doses to melanoma cells in vitro, and was combined with aCD137 in vivo to test treatment effects on melanoma tumor progression, animal survival, immunological protection against tumor rechallenge, and hepatotoxicity. When administered at a melanoma-specific thermal dose, PBNP-PTT elicits immunogenic cell death (ICD) in melanoma cells and upregulates markers associated with antigen presentation and immune cell co-stimulation in vitro. Consequently, PBNP-PTT eliminates primary melanoma tumors in vivo, yielding long-term tumor-free survival. However, the antitumor immune effects generated by PBNP-PTT cannot eliminate secondary tumors, despite significantly slowing their growth. The addition of aCD137 enables significant abscopal efficacy and improvement of survival, functioning through activated dendritic cells and tumor-infiltrating CD8+ T cells, and generates CD4+ and CD8+ T cell memory that manifests in the rejection of tumor rechallenge, with no long-term hepatotoxicity. This study describes for the first time a novel and effective nanoimmunotherapy combination of PBNP-PTT with aCD137 mAb therapy for melanoma.
Collapse
Affiliation(s)
- Preethi Bala Balakrishnan
- GW Cancer Center, Department of Medicine, School of Medicine and Health Sciences, George Washington University, Washington, DC 20052, USA
| | - Debbie K. Ledezma
- The Institute for Biomedical Sciences, School of Medicine and Health Sciences, George Washington University, Washington, DC 20052, USA
| | - Juliana Cano-Mejia
- GW Cancer Center, Department of Medicine, School of Medicine and Health Sciences, George Washington University, Washington, DC 20052, USA
| | - Jaclyn Andricovich
- The Institute for Biomedical Sciences, School of Medicine and Health Sciences, George Washington University, Washington, DC 20052, USA
| | - Erica Palmer
- GW Cancer Center, Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, George Washington University, Washington, DC 20052, USA
| | - Vishal A. Patel
- Department of Dermatology & Oncology, School of Medicine and Health Sciences, George Washington University, Washington, DC 20037, USA
| | - Patricia S. Latham
- Department of Pathology, School of Medicine and Health Sciences, George Washington University, Washington, DC 20037, USA
| | - Eric S. Yvon
- GW Cancer Center, Department of Medicine, School of Medicine and Health Sciences, George Washington University, Washington, DC 20052, USA
| | - Alejandro Villagra
- GW Cancer Center, Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, George Washington University, Washington, DC 20052, USA
| | - Rohan Fernandes
- GW Cancer Center, Department of Medicine, School of Medicine and Health Sciences, George Washington University, Washington, DC 20052, USA
- The Institute for Biomedical Sciences, School of Medicine and Health Sciences, George Washington University, Washington, DC 20052, USA
- ImmunoBlue, Bethesda, MD 20817, USA
| | - Elizabeth E. Sweeney
- GW Cancer Center, Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, George Washington University, Washington, DC 20052, USA
- ImmunoBlue, Bethesda, MD 20817, USA
| |
Collapse
|
9
|
Heshmati Aghda N, Torres Hurtado S, Abdulsahib SM, Lara EJ, Tunnell JW, Betancourt T. Dual Photothermal/Chemotherapy of Melanoma Cells with Albumin Nanoparticles Carrying Indocyanine Green and Doxorubicin Leads to Immunogenic Cell Death. Macromol Biosci 2021; 22:e2100353. [PMID: 34762334 DOI: 10.1002/mabi.202100353] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 11/04/2021] [Indexed: 12/11/2022]
Abstract
Recent focus on cancer immunotherapies has led to significant interest in the development of therapeutic strategies that can lead to immunogenic cell death (ICD), which can cause activation of an immune response against tumor cells and improve immunotherapy outcomes by enhancing the immunogenicity of the tumor microenvironment. In this work, a nanomedicine-mediated combination therapy is used to deliver the ICD inducers doxorubicin (Dox), a chemotherapeutic agent, and indocyanine green (ICG), a photothermal agent. These agents are loaded into nanoparticles (NPs) of bovine serum albumin (BSA) that are prepared through a desolvation process. The formulation of BSA NPs is optimized to achieve NPs of 102.6 nm in size and loadings of 8.55 % and 5.69 % (w/w) for ICG and Dox, respectively. The controlled release of these agents from the BSA NPs is confirmed. Upon laser irradiation for 2.5 min, NPs at a dose of 62.5 μg mL-1 are able to increase the temperature of the cells by 7 °C and thereby inhibit the growth of B16F10 melanoma cells in vitro. Surface presentation of heat shock proteins and calreticulin from the cells after treatment confirmed the ability of the Dox/ICG loaded BSA NPs to induce ICD in the melanoma cells.
Collapse
Affiliation(s)
- Niloofar Heshmati Aghda
- Materials Science, Engineering and Commercialization Program, Texas State University, 601 University Drive, San Marcos, TX, 78666, USA
| | - Susana Torres Hurtado
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W Dean Keeton Street, Austin, TX, 78712, USA
| | - Shahad M Abdulsahib
- Department of Chemistry and Biochemistry, Texas State University, 601 University Drive, San Marcos, TX, 78666, USA
| | - Emilio J Lara
- Department of Chemistry and Biochemistry, Texas State University, 601 University Drive, San Marcos, TX, 78666, USA
| | - James W Tunnell
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W Dean Keeton Street, Austin, TX, 78712, USA
| | - Tania Betancourt
- Materials Science, Engineering and Commercialization Program, Texas State University, 601 University Drive, San Marcos, TX, 78666, USA.,Department of Chemistry and Biochemistry, Texas State University, 601 University Drive, San Marcos, TX, 78666, USA
| |
Collapse
|
10
|
Shukla A, Cano-Mejia J, Andricovich J, Burga RA, Sweeney EE, Fernandes R. An Engineered Prussian Blue Nanoparticles-based Nanoimmunotherapy Elicits Robust and Persistent Immunological Memory in a TH-MYCN Neuroblastoma Model. ADVANCED NANOBIOMED RESEARCH 2021; 1. [PMID: 34435194 DOI: 10.1002/anbr.202100021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
A combination therapy using Prussian blue nanoparticles (PBNP) as photothermal therapy (PTT) agents coated with CpG oligodeoxynucleotides, an immunologic adjuvant, as a nanoimmunotherapy (CpG-PBNP-PTT) for neuroblastoma (NB) is described. NB driven by MYCN amplification confers high risk and correlates with a dismal prognosis, accounting for the majority of NB-related mortality. The efficacy of the CpG-PBNP-PTT nanoimmunotherapy in a clinically relevant, TH-MYCN murine NB model (9464D) overexpressing MYCN is tested. When administered to 9464D NB cells in vitro, CpG-PBNP-PTT triggers thermal dose-dependent immunogenic cell death and tumor cell priming for immune recognition in vitro, measured by the expression of specific costimulatory and antigen-presenting molecules. In vivo, intratumorally administered CpG-PBNP-PTT generates complete tumor regression and significantly higher long-term survival compared to controls. Furthermore, CpG-PBNP-PTT-treated mice reject tumor rechallenge. Ex vivo studies confirm these therapeutic responses result from the generation of robust T cell-mediated immunological memory. Consequently, in a synchronous 9464D tumor model, CpG-PBNP-PTT induces complete tumor regression on the treated flank and significantly slows tumor progression on the untreated flank, improving animal survival. These findings demonstrate that localized administration of the CpG-PBNP-PTT nanoimmunotherapy drives potent systemic T cell responses in solid tumors such as NB and therefore has therapeutic implications for NB.
Collapse
Affiliation(s)
- Anshi Shukla
- The George Washington Cancer Center, The George Washington University, 800 22nd St NW, Science and Engineering Hall 8 Floor, Washington, DC 20052, USA
| | - Juliana Cano-Mejia
- The George Washington Cancer Center, The George Washington University, 800 22nd St NW, Science and Engineering Hall 8 Floor, Washington, DC 20052, USA
| | - Jaclyn Andricovich
- The George Washington Cancer Center, The George Washington University, 800 22nd St NW, Science and Engineering Hall 8 Floor, Washington, DC 20052, USA.,The Institute for Biomedical Sciences, The George Washington University,2300 Eye Street NW, Ross Hall Room 561, Washington, DC 20037, USA
| | - Rachel A Burga
- The George Washington Cancer Center, The George Washington University, 800 22nd St NW, Science and Engineering Hall 8 Floor, Washington, DC 20052, USA.,The Institute for Biomedical Sciences, The George Washington University,2300 Eye Street NW, Ross Hall Room 561, Washington, DC 20037, USA
| | - Elizabeth E Sweeney
- The George Washington Cancer Center, The George Washington University, 800 22nd St NW, Science and Engineering Hall 8 Floor, Washington, DC 20052, USA
| | - Rohan Fernandes
- The George Washington Cancer Center, The George Washington University, 800 22nd St NW, Science and Engineering Hall 8 Floor, Washington, DC 20052, USA
| |
Collapse
|
11
|
Balakrishnan PB, Sweeney EE, Ramanujam AS, Fernandes R. Photothermal therapies to improve immune checkpoint blockade for cancer. Int J Hyperthermia 2021; 37:34-49. [PMID: 33426992 DOI: 10.1080/02656736.2020.1797190] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Immune checkpoint blockade (ICB) comprising monoclonal antibodies (mAbs) against immune 'checkpoints', such as CTLA-4 and the PD1/PDL1 axis have dramatically improved clinical outcomes for patients with cancer. However, ICB by itself has failed to provide benefit in a wide range of solid tumors, where recurrence still occurs with high incidence. These poor response rates may be due to the therapeutic shortcomings of ICB; namely, a lack of cancer-specific cytotoxicity and ability to debulk tumors. To overcome these limitations, effective ICB therapy may require the combination with other complementary therapeutic platforms. Here, we propose that photothermal therapy (PTT) is an ideal therapeutic modality for combination with ICB because it can generate both tumor-specific cytotoxicity and immunogenicity. PTT elicits these specific effects because it is a localized thermal ablation technique that utilizes light-responsive nanoparticles activated by a wavelength-matched laser. While ICB immunotherapy alone improves cancer immunogenicity but does not generate robust antitumor cytotoxicity, nanoparticle-based PTT elicits targeted and controlled cytotoxicity but sub-optimal long-term immunogenicity. Thus, the two platforms offer complementary and potentially synergistic antitumor effects, which will be detailed in this review. We highlight three classes of nanoparticles used as agents of PTT (i.e., metallic inorganic nanoparticles, carbon-based nanoparticles and organic dyes), and illustrate the potential for nanoparticle-based PTT to potentiate the effects of ICB in preclinical models. Through this discussion, we aim to present PTT combined with ICB as a potent synergistic combination treatment for diverse cancer types currently refractory to ICB as well as PTT monotherapies.
Collapse
Affiliation(s)
- Preethi B Balakrishnan
- The George Washington Cancer Center, The George Washington University, Washington, DC, USA
| | - Elizabeth E Sweeney
- The George Washington Cancer Center, The George Washington University, Washington, DC, USA
| | - Anvitha S Ramanujam
- The George Washington Cancer Center, The George Washington University, Washington, DC, USA.,Thomas Jefferson High School for Science and Technology, Alexandria, VA, USA
| | - Rohan Fernandes
- The George Washington Cancer Center, The George Washington University, Washington, DC, USA.,The Institute for Biomedical Sciences, The George Washington University, Washington, DC, USA.,Department of Medicine, The George Washington University, Washington, DC, USA
| |
Collapse
|
12
|
Sugumaran A, Mathialagan V. Colloidal Nanocarriers as Versatile Targeted Delivery Systems for Cervical Cancer. Curr Pharm Des 2021; 26:5174-5187. [PMID: 32586249 DOI: 10.2174/1381612826666200625110950] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Accepted: 04/01/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND The second most common malignant cancer of the uterus is cervical cancer, which is present worldwide, has a rising death rate and is predominant in developing countries. Different classes of anticancer agents are used to treat cervical carcinoma. The use of these agents results in severe untoward side-effects, toxicity, and multidrug resistance (MDR) with higher chances of recurrence and spread beyond the pelvic region. Moreover, the resulting clinical outcome remains very poor even after surgical procedures and treatment with conventional chemotherapy. Because of the nonspecificity of their use, the agents wipe out both cancerous and normal tissues. Colloidal nano dispersions have now been focusing on site-specific delivery for cervical cancer, and there has been much advancement. METHODS This review aims to highlight the problems in the current treatment of cervical cancer and explore the potential of colloidal nanocarriers for selective delivery of anticancer drugs using available literature. RESULTS In this study, we surveyed the role and potential of different colloidal nanocarriers in cervical cancer, such as nanoemulsion, nanodispersions, polymeric nanoparticles, and metallic nanoparticles and photothermal and photodynamic therapy. We found significant advancement in colloidal nanocarrier-based cervical cancer treatment. CONCLUSION Cervical cancer-targeted treatment with colloidal nanocarriers would hopefully result in minimal toxic side effects, reduced dosage frequency, and lower MDR incidence and enhance the patient survival rates. The future direction of the study should be focused more on the regulatory barrier of nanocarriers based on clinical outcomes for cervical cancer targeting with cost-effective analysis.
Collapse
Affiliation(s)
- Abimanyu Sugumaran
- Department of Pharmaceutics, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur 603203, India
| | - Vishali Mathialagan
- Department of Pharmaceutics, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur 603203, India
| |
Collapse
|
13
|
Deng X, Shao Z, Zhao Y. Solutions to the Drawbacks of Photothermal and Photodynamic Cancer Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2002504. [PMID: 33552860 PMCID: PMC7856884 DOI: 10.1002/advs.202002504] [Citation(s) in RCA: 279] [Impact Index Per Article: 69.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 09/24/2020] [Indexed: 05/11/2023]
Abstract
Phototherapy such as photothermal therapy and photodynamic therapy in cancer treatment has been developed quickly over the past few years for its noninvasive nature and high efficiency. However, there are still many drawbacks in phototherapy that prevent it from clinical applications. Thus, scientists have designed different systems to overcome the issues associated with phototherapy, including enhancing the targeting ability of phototherapy, low-temperature photothermal therapy, replacing near-infrared light with other excitation sources, and so on. This article discusses the problems and shortcomings encountered in the development of phototherapy and highlights possible solutions to address them so that phototherapy may become a useful cancer treatment approach in clinical practice. This article aims to give a brief summary about current research advancements in phototherapy research and provides a quick guideline toward future developments in the field.
Collapse
Affiliation(s)
- Xiangyu Deng
- Department of Orthopaedic SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Division of Chemistry and Biological ChemistrySchool of Physical and Mathematical SciencesNanyang Technological University21 Nanyang LinkSingapore637371Singapore
| | - Zengwu Shao
- Department of Orthopaedic SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Yanli Zhao
- Division of Chemistry and Biological ChemistrySchool of Physical and Mathematical SciencesNanyang Technological University21 Nanyang LinkSingapore637371Singapore
| |
Collapse
|
14
|
Cano-Mejia J, Shukla A, Ledezma DK, Palmer E, Villagra A, Fernandes R. CpG-coated prussian blue nanoparticles-based photothermal therapy combined with anti-CTLA-4 immune checkpoint blockade triggers a robust abscopal effect against neuroblastoma. Transl Oncol 2020; 13:100823. [PMID: 32652470 PMCID: PMC7348061 DOI: 10.1016/j.tranon.2020.100823] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/21/2020] [Accepted: 05/26/2020] [Indexed: 01/13/2023] Open
Abstract
High-risk neuroblastoma, which is associated with regional and systemic metastasis, is a leading cause of cancer-related mortality in children. Responding to this need for novel therapies for high-risk patients, we have developed a "nanoimmunotherapy," which combines photothermal therapy (PTT) using CpG oligodeoxynucleotide-coated Prussian blue nanoparticles (CpG-PBNPs) combined with anti-CTLA-4 (aCTLA-4) immunotherapy. Our in vitro studies demonstrate that in addition to causing ablative tumor cell death, our nanoimmunotherapy alters the surface levels of co-stimulatory, antigen-presenting, and co-inhibitory molecules on neuroblastoma tumor cells. When administered in a syngeneic, murine model of neuroblastoma bearing synchronous Neuro2a tumors, the CpG-PBNP-PTT plus aCTLA-4 nanoimmunotherapy elicits complete tumor regression in both primary (CpG-PBNP-PTT-treated) and secondary tumors, and long-term survival in a significantly higher proportion (55.5%) of treated-mice compared with the controls. Furthermore, the surviving, nanoimmunotherapy-treated animals reject Neuro2a rechallenge, suggesting that the therapy generates immunological memory. Additionally, the depletion of CD4+, CD8+, and NK+ populations abrogate the observed therapeutic responses of the nanoimmunotherapy. These findings demonstrate the importance of concurrent PTT-based cytotoxicity and the antitumor immune effects of PTT, CpG, and aCTLA-4 in generating a robust abscopal effect against neuroblastoma.
Collapse
Affiliation(s)
- Juliana Cano-Mejia
- The George Washington Cancer Center, The George Washington University, Washington, DC 20052, USA; Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Anshi Shukla
- The George Washington Cancer Center, The George Washington University, Washington, DC 20052, USA
| | - Debbie K Ledezma
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; The Institute for Biomedical Sciences, The George Washington University, Washington, DC 20037, USA
| | - Erica Palmer
- The George Washington Cancer Center, The George Washington University, Washington, DC 20052, USA
| | - Alejandro Villagra
- The George Washington Cancer Center, The George Washington University, Washington, DC 20052, USA
| | - Rohan Fernandes
- The George Washington Cancer Center, The George Washington University, Washington, DC 20052, USA; The Institute for Biomedical Sciences, The George Washington University, Washington, DC 20037, USA; Department of Medicine, The George Washington University, Washington, DC 20037, USA.
| |
Collapse
|
15
|
Non-cytotoxic systemic treatment in malignant peripheral nerve sheath tumors (MPNST): A systematic review from bench to bedside. Crit Rev Oncol Hematol 2019; 138:223-232. [DOI: 10.1016/j.critrevonc.2019.04.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 03/28/2019] [Accepted: 04/08/2019] [Indexed: 12/19/2022] Open
|
16
|
Cano-Mejia J, Bookstaver ML, Sweeney EE, Jewell CM, Fernandes R. Prussian blue nanoparticle-based antigenicity and adjuvanticity trigger robust antitumor immune responses against neuroblastoma. Biomater Sci 2019; 7:1875-1887. [PMID: 30789175 PMCID: PMC6491208 DOI: 10.1039/c8bm01553h] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
We describe the synthesis of CpG oligodeoxynucleotide-coated Prussian blue nanoparticles (CpG-PBNPs) that function as a nanoimmunotherapy for neuroblastoma, a common childhood cancer. These CpG-PBNPs increase the antigenicity and adjuvanticity of the treated tumors, ultimately driving robust antitumor immunity through a multi-pronged mechanism. CpG-PBNPs are synthesized using a facile layer-by-layer coating scheme resulting in nanoparticles that exhibit monodisperse size distributions and multiday stability without cytotoxicity. The strong intrinsic absorption of PBNPs in the CpG-PBNPs enables ablative photothermal therapy (CpG-PBNP-PTT) that triggers tumor cell death, as well as the release of tumor antigens to increase antigenicity. Simultaneously, the CpG coating functions as an exogenous molecular adjuvant that complements the endogenous adjuvants released by the CpG-PBNP-PTT (e.g. ATP, calreticulin, and HMGB1). In cell culture, coating NPs with CpG increases immunogenicity while maintaining the photothermal activity of PBNPs. When administered in a syngeneic, Neuro2a-based, murine model of neuroblastoma, CpG-PBNP-PTT results in complete tumor regression in a significantly higher proportion (70% at 60 days) of treated animals relative to controls. Furthermore, the long-term surviving, CpG-PBNP-PTT-treated animals reject Neuro2a rechallenge, suggesting that this therapy generates immunological memory. Our findings point to the importance of simultaneous cytotoxicity, antigenicity, and adjuvanticity to generate robust and persistent antitumor immune responses against neuroblastoma.
Collapse
Affiliation(s)
- Juliana Cano-Mejia
- The George Washington Cancer Center, The George Washington University, Washington, DC 20052, USA
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Michelle L. Bookstaver
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Elizabeth E. Sweeney
- The George Washington Cancer Center, The George Washington University, Washington, DC 20052, USA
| | - Christopher M. Jewell
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
- Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD 20742, USA
- United States Department of Veterans Affairs, Maryland VA Health Care System, Baltimore, MD 21201, USA
- Department of Microbiology and Immunology, University of Maryland Medical School, Baltimore, MD 21205, USA
- Marlene and Stewart Greenebaum Cancer Center, Baltimore, MD 21201, USA
| | - Rohan Fernandes
- The George Washington Cancer Center, The George Washington University, Washington, DC 20052, USA
- Department of Medicine, The George Washington University, Washington, DC, 20052, USA
| |
Collapse
|
17
|
Valcourt DM, Dang MN, Day ES. IR820-loaded PLGA nanoparticles for photothermal therapy of triple-negative breast cancer. J Biomed Mater Res A 2019; 107:1702-1712. [PMID: 30920169 DOI: 10.1002/jbm.a.36685] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 02/25/2019] [Accepted: 03/22/2019] [Indexed: 01/06/2023]
Abstract
Triple-negative breast cancer (TNBC) accounts for 15-25% of breast cancer cases and lacks expression of the three most common receptors seen on other subtypes of breast cancer. This lack of expression makes TNBC unsusceptible to currently available targeted or hormonal therapies, so new treatment strategies are desperately needed. Photothermal therapy (PTT), which utilizes nanoparticles (NPs) embedded in tumors as exogenous energy absorbers to convert externally applied near-infrared (NIR) light into heat to ablate cancer cells, has shown promise as an alternative strategy. However, it typically uses gold-based NPs that will remain in the body for extended period of time with unknown long-term health effects. To enable PTT with biodegradable, polymeric NPs, we encapsulated the NIR-absorbing dye IR820 in poly(lactic-co-glycolic acid) (PLGA) NPs. We characterized the physicochemical properties of these IR820-loaded PLGA NPs and evaluated their performance as PTT agents using both in vitro and in vivo models of TNBC. The results demonstrate that these NPs are potent mediators of PTT that induce cell death primarily through apoptosis to effectively hinder the growth of TNBC tumors. © 2019 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 107A: 1702-1712, 2019.
Collapse
Affiliation(s)
- Danielle M Valcourt
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, Delaware, 19716
| | - Megan N Dang
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, Delaware, 19716
| | - Emily S Day
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, Delaware, 19716.,Department of Materials Science & Engineering, University of Delaware, 201 DuPont Hall, Newark, Delaware, 19716.,Helen F. Graham Cancer Center & Research Institute, 4701 Ogletown Stanton Road, Newark, Delaware, 19713
| |
Collapse
|
18
|
Fischer-Huchzermeyer S, Chikobava L, Stahn V, Zangarini M, Berry P, Veal GJ, Senner V, Mautner VF, Harder A. Testing ATRA and MEK inhibitor PD0325901 effectiveness in a nude mouse model for human MPNST xenografts. BMC Res Notes 2018; 11:520. [PMID: 30055648 PMCID: PMC6064132 DOI: 10.1186/s13104-018-3630-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 07/20/2018] [Indexed: 01/29/2023] Open
Abstract
Objective Malignant peripheral nerve sheath tumors (MPNST) are aggressive sarcomas characterized by high recurrence rates and early metastases. These tumors arise more frequently within neurofibromatosis type 1 (NF1) and present with resistance during standard chemotherapy leading to increased mortality and morbidity in those patients. In vitro all-trans retinoic acid (ATRA) and MEK inhibitors (MEKi) were shown to inhibit tumor proliferation, especially when applied in combination. Therefore, we established a nude mouse model to investigate if treatment of xenografts derived from NF1 associated S462 and T265 MPNST cells respond to ATRA and the MEKi PD0325901. Results We demonstrated that human NF1 associated MPNST derived from S462 but not T265 cells form solid subcutaneous tumors in Foxn1 nude mice but not in Balb/c, SHO or Shorn mice. We verified a characteristic staining pattern of human MPNST xenografts by immunohistochemistry. Therapeutic effects of ATRA and/or MEKi PD0325901 on growth of S462 MPNST xenografts in Foxn1 nude mice were not demonstrated in vitro, as we did not observe significant suppression of MPNST growth compared with placebo treatment. Electronic supplementary material The online version of this article (10.1186/s13104-018-3630-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Levan Chikobava
- Institute of Neuropathology, University Hospital Münster, Münster, Germany
| | - Verena Stahn
- Institute of Neuropathology, University Hospital Münster, Münster, Germany
| | - Monique Zangarini
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, UK
| | - Philip Berry
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, UK
| | - Gareth J Veal
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, UK
| | - Volker Senner
- Institute of Neuropathology, University Hospital Münster, Münster, Germany
| | - Victor F Mautner
- Clinics and Polyclinics of Neurology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Anja Harder
- Institute of Neuropathology, University Hospital Münster, Münster, Germany. .,Institute of Pathology, Health Care Center, Brandenburg Hospital, Brandenburg Medical School Theodor Fontane, Brandenburg, Germany.
| |
Collapse
|
19
|
Evans ER, Bugga P, Asthana V, Drezek R. Metallic Nanoparticles for Cancer Immunotherapy. MATERIALS TODAY (KIDLINGTON, ENGLAND) 2018; 21:673-685. [PMID: 30197553 PMCID: PMC6124314 DOI: 10.1016/j.mattod.2017.11.022] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Cancer immunotherapy, or the utilization of the body's immune system to attack tumor cells, has gained prominence over the past few decades as a viable cancer treatment strategy. Recently approved immunotherapeutics have conferred remission upon patients with previously bleak outcomes and have expanded the number of tools available to treat cancer. Nanoparticles -including polymeric, liposomal, and metallic formulations - naturally traffic to the spleen and lymph organs and the relevant immune cells therein, making them good candidates for delivery of immunotherapeutic agents. Metallic nanoparticle formulations in particular are advantageous because of their potential for dense surface functionalization and their capability for optical or heat based therapeutic methods. Many research groups have investigated the potential of nanoparticle-mediated delivery platforms to improve the efficacy of immunotherapies. Despite the significant preclinical successes demonstrated by many of these platforms over the last twenty years, few metallic nanoparticles have successfully entered clinical trials with none achieving FDA approval for cancer therapy. In this review, we will discuss preclinical research and clinical trials involving metallic nanoparticles (MNPs) for cancer immunotherapy applications and discuss the potential for clinical translation of MNPs.
Collapse
Affiliation(s)
- Emily Reiser Evans
- Department of Bioengineering, Rice University, Houston, TX 77005, United States
| | - Pallavi Bugga
- Department of Bioengineering, Rice University, Houston, TX 77005, United States
| | - Vishwaratn Asthana
- Department of Bioengineering, Rice University, Houston, TX 77005, United States
| | - Rebekah Drezek
- Department of Bioengineering, Rice University, Houston, TX 77005, United States. Department of Electrical and Computer Engineering, Rice University, Houston, TX 77005, United States
| |
Collapse
|
20
|
Dacarro G, Taglietti A, Pallavicini P. Prussian Blue Nanoparticles as a Versatile Photothermal Tool. Molecules 2018; 23:E1414. [PMID: 29891819 PMCID: PMC6099709 DOI: 10.3390/molecules23061414] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 06/07/2018] [Accepted: 06/08/2018] [Indexed: 01/31/2023] Open
Abstract
Prussian blue (PB) is a coordination polymer studied since the early 18th century, historically known as a pigment. PB can be prepared in colloidal form with a straightforward synthesis. It has a strong charge-transfer absorption centered at ~700 nm, with a large tail in the Near-IR range. Irradiation of this band results in thermal relaxation and can be exploited to generate a local hyperthermia by irradiating in the so-called bio-transparent Near-IR window. PB nanoparticles are fully biocompatible (PB has already been approved by FDA) and biodegradable, this making them ideal candidates for in vivo use. While papers based on the imaging, drug-delivery and absorbing properties of PB nanoparticles have appeared and have been reviewed in the past decades, a very recent interest is flourishing with the use of PB nanoparticles as photothermal agents in biomedical applications. This review summarizes the syntheses and the optical features of PB nanoparticles in relation to their photothermal use and describes the state of the art of PB nanoparticles as photothermal agents, also in combination with diagnostic techniques.
Collapse
Affiliation(s)
- Giacomo Dacarro
- inLAB-Inorganic Nanochemistry Laboratory, Dipartimento di Chimica, Università di Pavia, 27100 Pavia, Italy.
| | - Angelo Taglietti
- inLAB-Inorganic Nanochemistry Laboratory, Dipartimento di Chimica, Università di Pavia, 27100 Pavia, Italy.
| | - Piersandro Pallavicini
- inLAB-Inorganic Nanochemistry Laboratory, Dipartimento di Chimica, Università di Pavia, 27100 Pavia, Italy.
- CHT, Centre for Health Technologies, Università di Pavia, 27100 Pavia, Italy.
| |
Collapse
|
21
|
Sweeney EE, Cano-Mejia J, Fernandes R. Photothermal Therapy Generates a Thermal Window of Immunogenic Cell Death in Neuroblastoma. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:e1800678. [PMID: 29665282 DOI: 10.1002/smll.201800678] [Citation(s) in RCA: 148] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Revised: 03/06/2018] [Indexed: 05/05/2023]
Abstract
A thermal "window" of immunogenic cell death (ICD) elicited by nanoparticle-based photothermal therapy (PTT) in an animal model of neuroblastoma is described. In studies using Prussian blue nanoparticles to administer photothermal therapy (PBNP-PTT) to established localized tumors in the neuroblastoma model, it is observed that PBNP-PTT conforms to the "more is better" paradigm, wherein higher doses of PBNP-PTT generates higher cell/local heating and thereby more cell death, and consequently improved animal survival. However, in vitro analysis of the biochemical correlates of ICD (ATP, high-motility group box 1, and calreticulin) elicited by PBNP-PTT demonstrates that PBNP-PTT triggers a thermal window of ICD. ICD markers are highly expressed within an optimal temperature (thermal dose) window of PBNP-PTT (63.3-66.4 °C) as compared with higher (83.0-83.5 °C) and lower PBNP-PTT (50.7-52.7 °C) temperatures, which both yield lower expression. Subsequent vaccination studies in the neuroblastoma model confirm the in vitro findings, wherein PBNP-PTT administered within the optimal temperature window results in long-term survival (33.3% at 100 d) compared with PBNP-PTT administered within the higher (0%) and lower (20%) temperature ranges, and controls (0%). The findings demonstrate a tunable immune response to heat generated by PBNP-PTT, which should be critically engaged in the administration of PTT for maximizing its therapeutic benefits.
Collapse
Affiliation(s)
- Elizabeth E Sweeney
- The George Washington Cancer Center, The George Washington University, 800 22nd St NW, Washington, 8th Floor Science and Engineering Hall, Washington, DC, 20052, USA
- The Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Health System, 111 Michigan Ave NW, 6th Floor Main Hospital, Washington, DC, 20010, USA
| | - Juliana Cano-Mejia
- The George Washington Cancer Center, The George Washington University, 800 22nd St NW, Washington, 8th Floor Science and Engineering Hall, Washington, DC, 20052, USA
- The Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Health System, 111 Michigan Ave NW, 6th Floor Main Hospital, Washington, DC, 20010, USA
- Fischell Department of Bioengineering, University of Maryland, Room 2330 Jeong H. Kim Engineering Building, College Park, MD, 20742, USA
| | - Rohan Fernandes
- The George Washington Cancer Center, The George Washington University, 800 22nd St NW, Washington, 8th Floor Science and Engineering Hall, Washington, DC, 20052, USA
- The Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Health System, 111 Michigan Ave NW, 6th Floor Main Hospital, Washington, DC, 20010, USA
- Fischell Department of Bioengineering, University of Maryland, Room 2330 Jeong H. Kim Engineering Building, College Park, MD, 20742, USA
- Department of Medicine, The George Washington University, 2150 Pennsylvania Ave NW, Suite 8-416, Washington, DC, 20037, USA
| |
Collapse
|
22
|
Neurofibromin level directs RAS pathway signaling and mediates sensitivity to targeted agents in malignant peripheral nerve sheath tumors. Oncotarget 2018; 9:22571-22585. [PMID: 29854299 PMCID: PMC5978249 DOI: 10.18632/oncotarget.25181] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 04/04/2018] [Indexed: 12/11/2022] Open
Abstract
Malignant peripheral nerve sheath tumor (MPNST) is a type of soft-tissue sarcoma strongly associated with dysfunction in neurofibromin; an inhibitor of the RAS pathway. We performed high-throughput screening of an array of FDA approved and promising agents in clinical development both alone and in combination at physiologically achievable concentrations against a panel of established MPNST cell line models. We found that drugs targeting a variety of factors in the RAS pathway can effectively lead to cell death in vitro with considerable drug combination synergy in regimens that target MEK or mTOR. We observed that the degree of relative sensitivity to chemotherapeutic agents was associated with the status of neurofibromin in these cell line models. Using a combination of agents that target MEK and mTORC1/2, we effectively silenced RAS/PI3K/MEK/mTOR signaling in vitro. Moreover, we employed RNAi against NF1 to establish that MPNST drug sensitivity is directly proportional to relative level of intracellular neurofibromin. Thus, two-drug combinations that target MEK and mTORC1/2 are most effective in halting the RAS signaling cascade, and the relative success of this and related small molecule interventions in MPNSTs may be predicated upon the molecular status of neurofibromin.
Collapse
|
23
|
Ali LMA, Mathlouthi E, Kajdan M, Daurat M, Long J, Sidi-Boulenouar R, Cardoso M, Goze-Bac C, Amdouni N, Guari Y, Larionova J, Gary-Bobo M. Multifunctional manganese-doped Prussian blue nanoparticles for two-photon photothermal therapy and magnetic resonance imaging. Photodiagnosis Photodyn Ther 2018; 22:65-69. [PMID: 29477814 DOI: 10.1016/j.pdpdt.2018.02.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 01/15/2018] [Accepted: 02/20/2018] [Indexed: 01/12/2023]
Abstract
Here we demonstrate for the first time that Mn2+-doped Prussian blue nanoparticles of c.a. 70 nm act as effective agents for photothermal therapy under two-photon excitation with an almost total eradication of malignant cells (97 and 98%) at a concentration of 100 μg mL-1 24 h after NIR excitation. This effect combined with interesting longitudinal NMR relaxivity values offer new perspectives for effective imaging and cancer treatment.
Collapse
Affiliation(s)
- Lamiaa M A Ali
- Institut des Biomolécules Max Mousseron, UMR5247, Université de Montpellier, CNRS, ENSCM, Faculté de Pharmacie, 15 Avenue Charles Flahault, 34093, Montpellier, Cedex 05, France
| | - Emna Mathlouthi
- Institut Charles Gerhardt, Equipe Ingénierie Moléculaire et Nano-Objets, Université de Montpellier, CNRS, ENSCM, Place Eugène Bataillon, 34095, Montpellier, Cedex 5, France; Université de Tunis el Manar, Faculté des Sciences, UR/11/ES/19, Physico-Chimie Des Matériaux à l'état Condensé, 2092, Tunis, Tunisie
| | - Marilyn Kajdan
- Institut Charles Gerhardt, Equipe Ingénierie Moléculaire et Nano-Objets, Université de Montpellier, CNRS, ENSCM, Place Eugène Bataillon, 34095, Montpellier, Cedex 5, France
| | - Morgane Daurat
- Institut des Biomolécules Max Mousseron, UMR5247, Université de Montpellier, CNRS, ENSCM, Faculté de Pharmacie, 15 Avenue Charles Flahault, 34093, Montpellier, Cedex 05, France; NanoMedSyn, 15 Avenue Charles Flahault, 34093, Montpellier, France
| | - Jérôme Long
- Institut Charles Gerhardt, Equipe Ingénierie Moléculaire et Nano-Objets, Université de Montpellier, CNRS, ENSCM, Place Eugène Bataillon, 34095, Montpellier, Cedex 5, France
| | - Rahima Sidi-Boulenouar
- L2C, UMR 5221 CNRS-UM2, Equipe BioNanoNMRI, Université de Montpellier, Place Eugène Bataillon, 34095, Montpellier, Cedex 5, France
| | - Maïda Cardoso
- L2C, UMR 5221 CNRS-UM2, Equipe BioNanoNMRI, Université de Montpellier, Place Eugène Bataillon, 34095, Montpellier, Cedex 5, France
| | - Christophe Goze-Bac
- L2C, UMR 5221 CNRS-UM2, Equipe BioNanoNMRI, Université de Montpellier, Place Eugène Bataillon, 34095, Montpellier, Cedex 5, France
| | - Nourredine Amdouni
- Université de Tunis el Manar, Faculté des Sciences, UR/11/ES/19, Physico-Chimie Des Matériaux à l'état Condensé, 2092, Tunis, Tunisie
| | - Yannick Guari
- Institut Charles Gerhardt, Equipe Ingénierie Moléculaire et Nano-Objets, Université de Montpellier, CNRS, ENSCM, Place Eugène Bataillon, 34095, Montpellier, Cedex 5, France
| | - Joulia Larionova
- Institut Charles Gerhardt, Equipe Ingénierie Moléculaire et Nano-Objets, Université de Montpellier, CNRS, ENSCM, Place Eugène Bataillon, 34095, Montpellier, Cedex 5, France.
| | - Magali Gary-Bobo
- Institut des Biomolécules Max Mousseron, UMR5247, Université de Montpellier, CNRS, ENSCM, Faculté de Pharmacie, 15 Avenue Charles Flahault, 34093, Montpellier, Cedex 05, France.
| |
Collapse
|
24
|
Xin Y, Yin M, Zhao L, Meng F, Luo L. Recent progress on nanoparticle-based drug delivery systems for cancer therapy. Cancer Biol Med 2017; 14:228-241. [PMID: 28884040 PMCID: PMC5570600 DOI: 10.20892/j.issn.2095-3941.2017.0052] [Citation(s) in RCA: 164] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Accepted: 05/24/2017] [Indexed: 12/19/2022] Open
Abstract
The development of cancer nanotherapeutics has attracted great interest in the recent decade. Cancer nanotherapeutics have overcome several limitations of conventional therapies, such as nonspecific biodistribution, poor water solubility, and limited bioavailability. Nanoparticles with tuned size and surface characteristics are the key components of nanotherapeutics, and are designed to passively or actively deliver anti-cancer drugs to tumor cells. We provide an overview of nanoparticle-based drug delivery methods and cancer therapies based on tumor-targeting delivery strategies that have been developed in recent years.
Collapse
Affiliation(s)
- Yanru Xin
- College of Life Science and Technology, National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Mingming Yin
- College of Life Science and Technology, National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Liyuan Zhao
- College of Life Science and Technology, National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Fanling Meng
- College of Life Science and Technology, National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Liang Luo
- College of Life Science and Technology, National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|