1
|
Mungra N, Nsole Biteghe FA, Huysamen AM, Hardcastle NS, Bunjun R, Naran K, Lang D, Richter W, Hunter R, Barth S. An Investigation into the In Vitro Targeted Killing of CD44-Expressing Triple-Negative Breast Cancer Cells Using Recombinant Photoimmunotherapeutics Compared to Auristatin-F-Based Antibody-Drug Conjugates. Mol Pharm 2024; 21:4098-4115. [PMID: 39047292 DOI: 10.1021/acs.molpharmaceut.4c00449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
Triple-negative breast cancer (TNBC) is the deadliest form of breast cancer with limited treatment options. The persistence of highly tumorigenic CD44-expressing subpopulation referred to as cancer stem cells (CSCs), endowed with the self-renewal capacity, has been associated with therapeutic resistance, hence clinical relapses. To mitigate these undesired events, targeted immunotherapies using antibody-photoconjugate (APC) or antibody-drug conjugate (ADC), were developed to specifically release cytotoxic payloads within targeted cells overexpressing cognate antigen receptors. Therefore, an αCD44(scFv)-SNAP-tag antibody fusion protein was engineered through genetic fusion of a single-chain antibody fragment (scFv) to a SNAPf-tag fusion protein, capable of self-conjugating with benzylguanine-modified light-sensitive near-infrared (NIR) phthalocyanine dye IRDye700DX (BG-IR700) or the small molecule toxin auristatin-F (BG-AURIF). Binding of the αCD44(scFv)-SNAPf-IR700 photoimmunoconjugate to antigen-positive cells was demonstrated by confocal microscopy and flow cytometry. By switching to NIR irradiation, CD44-expressing TNBC was selectively killed through induced phototoxic activities. Likewise, the αCD44(scFv)-SNAPf-AURIF immunoconjugate was able to selectively accumulate within targeted cells and significantly reduced cell viability through antimitotic activities at nano- to micromolar drug concentrations. This study provides an in vitro proof-of-concept for a future strategy to selectively destroy light-accessible superficial CD44-expressing TNBC tumors and their metastatic lesions which are inaccessible to therapeutic light.
Collapse
Affiliation(s)
- Neelakshi Mungra
- Institute of Infectious Disease and Molecular Medicine, Medical Biotechnology and Immunotherapy Research Unit, University of Cape Town, Cape Town 7700, South Africa
- Centre for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Washington 98101, United States
| | - Fleury A Nsole Biteghe
- College of Science, Department of Biotechnology, Northeastern University, Boston, Massachusetts 02115, United States
| | - Allan M Huysamen
- Department of Chemistry, University of Cape Town, PD Hahn Building, Cape Town 7700, South Africa
| | - Natasha S Hardcastle
- Institute of Infectious Disease and Molecular Medicine, Medical Biotechnology and Immunotherapy Research Unit, University of Cape Town, Cape Town 7700, South Africa
| | - Rubina Bunjun
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7700, South Africa
- Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town 7700, South Africa
| | - Krupa Naran
- Institute of Infectious Disease and Molecular Medicine, Medical Biotechnology and Immunotherapy Research Unit, University of Cape Town, Cape Town 7700, South Africa
| | - Dirk Lang
- Division of Physiological Sciences, Department of Human Biology, University of Cape Town, Cape Town 7700, South Africa
| | | | - Roger Hunter
- Department of Chemistry, University of Cape Town, PD Hahn Building, Cape Town 7700, South Africa
| | - Stefan Barth
- Institute of Infectious Disease and Molecular Medicine, Medical Biotechnology and Immunotherapy Research Unit, University of Cape Town, Cape Town 7700, South Africa
- Faculty of Health Sciences, Department of Integrative Biomedical Sciences, South African Research Chair in Cancer Biotechnology, University of Cape Town, Cape Town 7700, South Africa
| |
Collapse
|
2
|
Coskun A, Kayhan H, Senturk F, Esmekaya MA, Canseven AG. The Efficacy of Electrochemotherapy with Dacarbazine on Melanoma Cells. Bioelectricity 2024; 6:118-125. [PMID: 39119570 PMCID: PMC11305008 DOI: 10.1089/bioe.2023.0041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2024] Open
Abstract
Electrochemotherapy (ECT) involves locally applying electrical pulses to permeabilize cell membranes, using electroporation (EP). This process enhances the uptake of low-permeant chemotherapeutic agents, consequently amplifying their cytotoxic effects. In melanoma treatment, dacarbazine (DTIC) is a cornerstone, but it faces limitations because of poor cell membrane penetration, necessitating the use of high doses, which, in turn, leads to increased side effects. In our study, we investigated the effects of DTIC and EP, both individually and in combination, on the melanoma cell line (SK-MEL-30) as well as human dermal fibroblasts (HDF) using in vitro assays. First, the effects of different DTIC concentrations on the viability of SK-MEL-30 and HDF cells were determined, revealing that DTIC was more effective against melanoma cells at lower concentrations, whereas its cytotoxicity at 1000 μM was similar in both cell types. Next, an ideal electric field strength of 1500 V/cm achieved a balance between permeability (84%) and melanoma cell viability (79%), paving the way for effective ECT. The combined DTIC-EP (ECT) application reduced IC50 values by 2.2-fold in SK-MEL-30 cells and 2.7-fold in HDF cells compared with DTIC alone. In conclusion, ECT not only increased DTIC's cytotoxicity against melanoma cells but also affected healthy fibroblasts. These findings emphasize the need for cautious, targeted ECT management in melanoma therapy.
Collapse
Affiliation(s)
- Alaaddin Coskun
- Department of Biophysics, Faculty of Medicine, Gazi University, Ankara, Turkey
| | - Handan Kayhan
- Department of Adult Hematology, Faculty of Medicine, Gazi University, Ankara, Turkey
| | - Fatih Senturk
- Department of Biophysics, Faculty of Medicine, Duzce University, Duzce, Turkey
| | - Meric Arda Esmekaya
- Department of Biophysics, Faculty of Medicine, Gazi University, Ankara, Turkey
| | | |
Collapse
|
3
|
Fu C, Tong W, Yu L, Miao Y, Wei Q, Yu Z, Chen B, Wei M. When will the immune-stimulating antibody conjugates (ISACs) be transferred from bench to bedside? Pharmacol Res 2024; 203:107160. [PMID: 38547937 DOI: 10.1016/j.phrs.2024.107160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 03/17/2024] [Accepted: 03/25/2024] [Indexed: 04/30/2024]
Abstract
Immunostimulatory antibody conjugates (ISACs) as a promising new generation of targeted therapeutic antibody-drug conjugates (ADCs), that not only activate innate immunity but also stimulate adaptive immunity, providing a dual therapeutic effect to eliminate tumor cells. However, several ISACs are still in the early stages of clinical development or have already failed. Therefore, it is crucial to design ISACs more effectively to overcome their limitations, including high toxicity, strong immunogenicity, long development time, and poor pharmacokinetics. This review aims to summarize the composition and function of ISACs, incorporating current design considerations and ongoing clinical trials. Additionally, the review delves into the current issues with ISACs and potential solutions, such as adjusting the drug-antibody ratio (DAR) to improve the bioavailability of ISACs. By leveraging the affinity and bioavailability-enhancing properties of bispecific antibodies, the utility between antibodies and immunostimulatory agents can be balanced. Commonly used immunostimulatory agents may induce systemic immune reactions, and BTK (Bruton's tyrosine kinase) inhibitors can regulate immunogenicity. Finally, the concept of grafting ADC's therapeutic principles is simple, but the combination of payload, linker, and targeted functional molecules is not a simple permutation and combination problem. The development of conjugate drugs faces more complex pharmacological and toxicological issues. Standing on the shoulders of ADC, the development and application scenarios of ISAC are endowed with broader space.
Collapse
Affiliation(s)
- Chen Fu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, PR China; Liaoning Key Laboratory of Molecular Targeted Anti-tumor Drug Development and Evaluation, China Medical University, Shenyang 110122, PR China
| | - Weiwei Tong
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang 110122, PR China
| | - Lifeng Yu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, PR China; Liaoning Key Laboratory of Molecular Targeted Anti-tumor Drug Development and Evaluation, China Medical University, Shenyang 110122, PR China
| | - Yuxi Miao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, PR China; Liaoning Key Laboratory of Molecular Targeted Anti-tumor Drug Development and Evaluation, China Medical University, Shenyang 110122, PR China
| | - Qian Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, PR China; Liaoning Key Laboratory of Molecular Targeted Anti-tumor Drug Development and Evaluation, China Medical University, Shenyang 110122, PR China
| | - Zhaojin Yu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, PR China; Liaoning Key Laboratory of Molecular Targeted Anti-tumor Drug Development and Evaluation, China Medical University, Shenyang 110122, PR China.
| | - Bo Chen
- Department of Breast Surgery, The First Hospital of China Medical University, No. 155, Nanjing North Street, Shenyang 110122, PR China.
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, PR China; Liaoning Key Laboratory of Molecular Targeted Anti-tumor Drug Development and Evaluation, China Medical University, Shenyang 110122, PR China.
| |
Collapse
|
4
|
Fortuna A, Amaral T. Multidisciplinary approach and treatment of acral and mucosal melanoma. Front Oncol 2024; 14:1340408. [PMID: 38469235 PMCID: PMC10926023 DOI: 10.3389/fonc.2024.1340408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 01/29/2024] [Indexed: 03/13/2024] Open
Abstract
Acral and mucosal melanoma are uncommon variants of melanoma. Acral melanoma has an age-adjusted incidence of approximately 1.8 cases per million individuals per year, accounting for about 2% to 3% of all melanoma cases. On the other hand, mucosal melanoma, with an incidence of 2.2 cases per million per year, makes up around 1.3% of all melanoma cases. These melanomas, in addition to being biologically and clinically distinct from cutaneous melanoma, share certain clinical and pathologic characteristics. These include a more aggressive nature and a less favorable prognosis. Furthermore, they exhibit a different mutational pattern, with KIT mutations being more prevalent in acral and mucosal melanomas. This divergence in mutational patterns may partially account for the relatively poorer prognosis, particularly to immune checkpoint inhibitors. This review explores various aspects of acral and mucosal melanoma, including their clinical presentation, pathologic features, mutational profiles, current therapeutic approaches, outcomes associated with systemic therapy, and potential strategies to address resistance to existing treatments.
Collapse
Affiliation(s)
- Ana Fortuna
- Oncology Department, Centro Hospitalar Universitário do Algarve, Faro, Portugal
| | - Teresa Amaral
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University of Tübingen, Tübingen, Germany
- Cluster of Excellence Image-Guided and Functionally Instructed Tumor Therapies (iFIT) (EXC 2180), Tübingen, Germany
| |
Collapse
|
5
|
Mungra N, Biteghe FAN, Malindi Z, Huysamen AM, Karaan M, Hardcastle NS, Bunjun R, Chetty S, Naran K, Lang D, Richter W, Hunter R, Barth S. CSPG4 as a target for the specific killing of triple-negative breast cancer cells by a recombinant SNAP-tag-based antibody-auristatin F drug conjugate. J Cancer Res Clin Oncol 2023; 149:12203-12225. [PMID: 37432459 PMCID: PMC10465649 DOI: 10.1007/s00432-023-05031-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 06/27/2023] [Indexed: 07/12/2023]
Abstract
PURPOSE Triple-negative breast cancer (TNBC) is phenotypic of breast tumors lacking expression of the estrogen receptor (ER), the progesterone receptor (PgR), and the human epidermal growth factor receptor 2 (HER2). The paucity of well-defined molecular targets in TNBC, coupled with the increasing burden of breast cancer-related mortality, emphasizes the need to develop targeted diagnostics and therapeutics. While antibody-drug conjugates (ADCs) have emerged as revolutionary tools in the selective delivery of drugs to malignant cells, their widespread clinical use has been hampered by traditional strategies which often give rise to heterogeneous mixtures of ADC products. METHODS Utilizing SNAP-tag technology as a cutting-edge site-specific conjugation method, a chondroitin sulfate proteoglycan 4 (CSPG4)-targeting ADC was engineered, encompassing a single-chain antibody fragment (scFv) conjugated to auristatin F (AURIF) via a click chemistry strategy. RESULTS After showcasing the self-labeling potential of the SNAP-tag component, surface binding and internalization of the fluorescently labeled product were demonstrated on CSPG4-positive TNBC cell lines through confocal microscopy and flow cytometry. The cell-killing ability of the novel AURIF-based recombinant ADC was illustrated by the induction of a 50% reduction in cell viability at nanomolar to micromolar concentrations on target cell lines. CONCLUSION This research underscores the applicability of SNAP-tag in the unambiguous generation of homogeneous and pharmaceutically relevant immunoconjugates that could potentially be instrumental in the management of a daunting disease like TNBC.
Collapse
Affiliation(s)
- Neelakshi Mungra
- Institute of Infectious Disease and Molecular Medicine, Medical Biotechnology and Immunotherapy Research Unit, University of Cape Town, Cape Town, 7700 South Africa
- Centre for Immunity and Immunotherapies, Seattle Children’s Research Institute, Washington, 98101 USA
| | - Fleury A. N. Biteghe
- Department of Radiation Oncology and Biomedical Sciences, Cedars-Sinai Medical, Los Angeles, USA
| | - Zaria Malindi
- Institute of Infectious Disease and Molecular Medicine, Medical Biotechnology and Immunotherapy Research Unit, University of Cape Town, Cape Town, 7700 South Africa
- Faculty of Health Sciences, Laser Research Centre, University of Johannesburg, Doornfontein, Johannesburg, 2028 South Africa
| | - Allan M. Huysamen
- Department of Chemistry, PD Hahn Building, University of Cape Town, Cape Town, 7700 South Africa
| | - Maryam Karaan
- Institute of Infectious Disease and Molecular Medicine, Medical Biotechnology and Immunotherapy Research Unit, University of Cape Town, Cape Town, 7700 South Africa
| | - Natasha S. Hardcastle
- Institute of Infectious Disease and Molecular Medicine, Medical Biotechnology and Immunotherapy Research Unit, University of Cape Town, Cape Town, 7700 South Africa
| | - Rubina Bunjun
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, 7700 South Africa
- Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, 7700 South Africa
| | - Shivan Chetty
- Faculty of Health Sciences, School of Clinical Medicine, University of Witwatersrand, Braamfontein, Johannesburg, 2000 South Africa
| | - Krupa Naran
- Institute of Infectious Disease and Molecular Medicine, Medical Biotechnology and Immunotherapy Research Unit, University of Cape Town, Cape Town, 7700 South Africa
| | - Dirk Lang
- Division of Physiological Sciences, Department of Human Biology, University of Cape Town, Cape Town, 7700 South Africa
| | | | - Roger Hunter
- Department of Chemistry, PD Hahn Building, University of Cape Town, Cape Town, 7700 South Africa
| | - Stefan Barth
- Institute of Infectious Disease and Molecular Medicine, Medical Biotechnology and Immunotherapy Research Unit, University of Cape Town, Cape Town, 7700 South Africa
- Faculty of Health Sciences, Department of Integrative Biomedical Sciences, South African Research Chair in Cancer Biotechnology, University of Cape Town, Cape Town, 7700 South Africa
| |
Collapse
|
6
|
Sioud M, Zhang Q. Precision Killing of M2 Macrophages with Phage-Displayed Peptide-Photosensitizer Conjugates. Cancers (Basel) 2023; 15:cancers15072009. [PMID: 37046671 PMCID: PMC10093000 DOI: 10.3390/cancers15072009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 03/24/2023] [Indexed: 03/30/2023] Open
Abstract
Among the immunosuppressive cells recruited to the tumor microenvironment, macrophages are particularly abundant and involved in angiogenesis, metastasis, and resistance to current cancer therapies. A strategy that simultaneously targets tumor cells and macrophages, particularly pro-tumoral M2 macrophages, would have significant clinical impact for various types of solid malignancies. By the use of phage display technology, we have recently developed a synthetic peptide, named NW, which binds to M1 and M2 macrophages with high affinity. Additional affinity selection on M2 macrophages identified only dominant peptides whose binding motifs are similar to that of the NW peptide. To reduce the frequency of selecting such dominating peptides, the peptide library was affinity selected on M2 macrophages blocked with NW peptide. This approach resulted in the selection of peptides that bind to M2, but not M1 macrophages. To explore the therapeutic potential of the selected peptides, the M13 phage-displayed peptides were conjugated to the photosensitizer IR700, which has been used for cancer photoimmunotherapy. The phage displaying a dominant peptide (SPILWLNAPPWA) killed both M1 and M2 macrophages, while those displaying the M2-specific peptides killed M2 macrophages only upon near-infrared light exposure. A significant fraction of the M2 macrophages were also killed with the untargeted M13 phage-IR700 conjugates. Hence, M2 macrophages can also be selectively targeted by the wild type M13 phage, which displayed a significant tropism to these cells. The benefits of this photoimmunotherapy include an automatic self-targeting ability of the wild type M13 phage, and the option of genetic manipulation of the phage genome to include tumor targeting peptides, allowing the killing of both M2 macrophages and cancer cells.
Collapse
Affiliation(s)
- Mouldy Sioud
- Department of Cancer Immunology, Division of Cancer Medicine, Oslo University Hospital, Radiumhospitalet, Ullernchausseen 70, 0379 Oslo, Norway
- Correspondence:
| | - Qindong Zhang
- Department of Cancer Immunology, Division of Cancer Medicine, Oslo University Hospital, Radiumhospitalet, Ullernchausseen 70, 0379 Oslo, Norway
- Department of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Blindern, 0316 Oslo, Norway
| |
Collapse
|
7
|
Advanced Acral Melanoma Therapies: Current Status and Future Directions. Curr Treat Options Oncol 2022; 23:1405-1427. [PMID: 36125617 PMCID: PMC9526689 DOI: 10.1007/s11864-022-01007-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2022] [Indexed: 11/17/2022]
Abstract
Melanoma is one of the deadliest malignancies. Its incidence has been significantly increasing in most countries in recent decades. Acral melanoma (AM), a peculiar subgroup of melanoma occurring on the palms, soles, and nails, is the main subtype of melanoma in people of color and is extremely rare in Caucasians. Although great progress has been made in melanoma treatment in recent years, patients with AM have shown limited benefit from current therapies and thus consequently have worse overall survival rates. Achieving durable therapeutic responses in this high-risk melanoma subtype represents one of the greatest challenges in the field. The frequency of BRAF mutations in AM is much lower than that in cutaneous melanoma, which prevents most AM patients from receiving treatment with BRAF inhibitors. However, AM has more frequent mutations such as KIT and CDK4/6, so targeted therapy may still improve the survival of some AM patients in the future. AM may be less susceptible to immune checkpoint inhibitors because of the poor immunogenicity. Therefore, how to enhance the immune response to the tumor cells may be the key to the application of immune checkpoint inhibitors in advanced AM. Anti-angiogenic drugs, albumin paclitaxel, or interferons are thought to enhance the effectiveness of immune checkpoint inhibitors. Combination therapies based on the backbone of PD-1 are more likely to provide greater clinical benefits. Understanding the molecular landscapes and immune microenvironment of AM will help optimize our combinatory strategies.
Collapse
|
8
|
Synergistic effects of radiotherapy and targeted immunotherapy in improving tumor treatment efficacy: a review. CLINICAL & TRANSLATIONAL ONCOLOGY : OFFICIAL PUBLICATION OF THE FEDERATION OF SPANISH ONCOLOGY SOCIETIES AND OF THE NATIONAL CANCER INSTITUTE OF MEXICO 2022; 24:2255-2271. [PMID: 35913663 DOI: 10.1007/s12094-022-02888-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 07/05/2022] [Indexed: 10/16/2022]
Abstract
Radiotherapy (RT), unlike chemotherapy, is one of the most routinely used and effective genotoxic and immune response inducing cancer therapies with an advantage of reduced side effects. However, cancer can relapse after RT owing to multiple factors, including acquired tumor resistance, immune suppressive microenvironment buildup, increased DNA repair, thus favoring tumor metastasis. Efforts to mitigate these undesirable effects have drawn interest in combining RT with immunotherapy, particularly the use of immune checkpoint inhibitors, to tilt the pre-existing tumor stromal microenvironment into long-lasting therapy-induced antitumor immunity at multiple metastatic sites (abscopal effects). This multimodal therapeutic strategy can alleviate the increased T cell priming and decrease tumor growth and metastasis, thus emerging as a significant approach to sustain as long-term antitumor immunity. To understand more about this synergism, a detailed cellular mechanism underlying the dynamic interaction between tumor and immune cells within the irradiated tumor microenvironment needs to be explored. Hence, in the present review, we have attempted to evaluate various RT-inducible immune factors, which can be targeted by immunotherapy and provide detailed explanation to optimally maximize their synergy with immunotherapy for long-lasting antitumor immunity. Moreover, we have critically assessed various combinatorial approaches along with their challenges and described strategies to modify them in addition to providing approaches for optimal synergistic effects of the combination.
Collapse
|
9
|
Sadraeian M, Bahou C, da Cruz EF, Janini LMR, Sobhie Diaz R, Boyle RW, Chudasama V, Eduardo Gontijo Guimarães F. Photoimmunotherapy Using Cationic and Anionic Photosensitizer-Antibody Conjugates against HIV Env-Expressing Cells. Int J Mol Sci 2020; 21:E9151. [PMID: 33271741 PMCID: PMC7730620 DOI: 10.3390/ijms21239151] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/20/2020] [Accepted: 11/21/2020] [Indexed: 12/11/2022] Open
Abstract
Different therapeutic strategies have been investigated to target and eliminate HIV-1-infected cells by using armed antibodies specific to viral proteins, with varying degrees of success. Herein, we propose a new strategy by combining photodynamic therapy (PDT) with HIV Env-targeted immunotherapy, and refer to it as HIV photoimmunotherapy (PIT). A human anti-gp41 antibody (7B2) was conjugated to two photosensitizers (PSs) with different charges through different linking strategies; "Click" conjugation by using an azide-bearing porphyrin attached via a disulfide bridge linker with a drug-to-antibody ratio (DAR) of exactly 4, and "Lysine" conjugation by using phthalocyanine IRDye 700DX dye with average DARs of 2.1, 3.0 and 4.4. These photo-immunoconjugates (PICs) were compared via biochemical and immunological characterizations regarding the dosimetry, solubility, and cell targeting. Photo-induced cytotoxicity of the PICs were compared using assays for apoptosis, reactive oxygen species (ROS), photo-cytotoxicity, and confocal microscopy. Targeted phototoxicity seems to be primarily dependent on the binding of PS-antibody to the HIV antigen on the cell membrane, whilst being independent of the PS type. This is the first report of the application of PIT for HIV immunotherapy by killing HIV Env-expressing cells.
Collapse
Affiliation(s)
- Mohammad Sadraeian
- São Carlos Institute of Physics, University of São Paulo, São Carlos, SP 13566-590, Brazil;
| | - Calise Bahou
- Department of Chemistry, University College London, London WC1H 0AJ, UK;
| | - Edgar Ferreira da Cruz
- Laboratório de Retrovirologia, Disciplina de Microbiologia, Departamento de Microbiologia Imunologia Parasitologia, Universidade Federal de São Paulo, São Paulo, SP 04039-032, Brazil; (E.F.d.C.); (L.M.R.J.); (R.S.D.)
| | - Luíz Mário Ramos Janini
- Laboratório de Retrovirologia, Disciplina de Microbiologia, Departamento de Microbiologia Imunologia Parasitologia, Universidade Federal de São Paulo, São Paulo, SP 04039-032, Brazil; (E.F.d.C.); (L.M.R.J.); (R.S.D.)
| | - Ricardo Sobhie Diaz
- Laboratório de Retrovirologia, Disciplina de Microbiologia, Departamento de Microbiologia Imunologia Parasitologia, Universidade Federal de São Paulo, São Paulo, SP 04039-032, Brazil; (E.F.d.C.); (L.M.R.J.); (R.S.D.)
| | - Ross W. Boyle
- Department of Chemistry, University of Hull, Cottingham Road, Hull HU6 7RX, UK;
| | - Vijay Chudasama
- Department of Chemistry, University College London, London WC1H 0AJ, UK;
| | | |
Collapse
|