1
|
Longobardi G, Moore TL, Conte C, Ungaro F, Satchi‐Fainaro R, Quaglia F. Polyester nanoparticles delivering chemotherapeutics: Learning from the past and looking to the future to enhance their clinical impact in tumor therapy. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1990. [PMID: 39217459 PMCID: PMC11670051 DOI: 10.1002/wnan.1990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/20/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024]
Abstract
Polymeric nanoparticles (NPs), specifically those comprised of biodegradable and biocompatible polyesters, have been heralded as a game-changing drug delivery platform. In fact, poly(α-hydroxy acids) such as polylactide (PLA), poly(lactide-co-glycolide) (PLGA), and poly(ε-caprolactone) (PCL) have been heavily researched in the past three decades as the material basis of polymeric NPs for drug delivery applications. As materials, these polymers have found success in resorbable sutures, biodegradable implants, and even monolithic, biodegradable platforms for sustained release of therapeutics (e.g., proteins and small molecules) and diagnostics. Few fields have gained more attention in drug delivery through polymeric NPs than cancer therapy. However, the clinical translational of polymeric nanomedicines for treating solid tumors has not been congruent with the fervor or funding in this particular field of research. Here, we attempt to provide a comprehensive snapshot of polyester NPs in the context of chemotherapeutic delivery. This includes a preliminary exploration of the polymeric nanomedicine in the cancer research space. We examine the various processes for producing polyester NPs, including methods for surface-functionalization, and related challenges. After a detailed overview of the multiple factors involved with the delivery of NPs to solid tumors, the crosstalk between particle design and interactions with biological systems is discussed. Finally, we report state-of-the-art approaches toward effective delivery of NPs to tumors, aiming at identifying new research areas and re-evaluating the reasons why some research avenues have underdelivered. We hope our effort will contribute to a better understanding of the gap to fill and delineate the future research work needed to bring polyester-based NPs closer to clinical application. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
| | - Thomas Lee Moore
- Department of PharmacyUniversity of Naples Federico IINaplesItaly
| | - Claudia Conte
- Department of PharmacyUniversity of Naples Federico IINaplesItaly
| | - Francesca Ungaro
- Department of PharmacyUniversity of Naples Federico IINaplesItaly
| | - Ronit Satchi‐Fainaro
- Department of Physiology and Pharmacology, Faculty of MedicineTel Aviv UniversityTel AvivIsrael
- Sagol School of NeurosciencesTel Aviv UniversityTel AvivIsrael
| | - Fabiana Quaglia
- Department of PharmacyUniversity of Naples Federico IINaplesItaly
| |
Collapse
|
2
|
Medina Pérez VM, Baselga M, Schuhmacher AJ. Single-Domain Antibodies as Antibody-Drug Conjugates: From Promise to Practice-A Systematic Review. Cancers (Basel) 2024; 16:2681. [PMID: 39123409 PMCID: PMC11311928 DOI: 10.3390/cancers16152681] [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: 06/26/2024] [Revised: 07/23/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Antibody-drug conjugates (ADCs) represent potent cancer therapies that deliver highly toxic drugs to tumor cells precisely, thus allowing for targeted treatment and significantly reducing off-target effects. Despite their effectiveness, ADCs can face limitations due to acquired resistance and potential side effects. OBJECTIVES This study focuses on advances in various ADC components to improve both the efficacy and safety of these agents, and includes the analysis of several novel ADC formats. This work assesses whether the unique features of VHHs-such as their small size, enhanced tissue penetration, stability, and cost-effectiveness-make them a viable alternative to conventional antibodies for ADCs and reviews their current status in ADC development. METHODS Following PRISMA guidelines, this study focused on VHHs as components of ADCs, examining advancements and prospects from 1 January 2014 to 30 June 2024. Searches were conducted in PubMed, Cochrane Library, ScienceDirect and LILACS using specific terms related to ADCs and single-domain antibodies. Retrieved articles were rigorously evaluated, excluding duplicates and non-qualifying studies. The selected peer-reviewed articles were analyzed for quality and synthesized to highlight advancements, methods, payloads, and future directions in ADC research. RESULTS VHHs offer significant advantages for drug conjugation over conventional antibodies due to their smaller size and structure, which enhance tissue penetration and enable access to previously inaccessible epitopes. Their superior stability, solubility, and manufacturability facilitate cost-effective production and expand the range of targetable antigens. Additionally, some VHHs can naturally cross the blood-brain barrier or be easily modified to favor their penetration, making them promising for targeting brain tumors and metastases. Although no VHH-drug conjugates (nADC or nanoADC) are currently in the clinical arena, preclinical studies have explored various conjugation methods and linkers. CONCLUSIONS While ADCs are transforming cancer treatment, their unique mechanisms and associated toxicities challenge traditional views on bioavailability and vary with different tumor types. Severe toxicities, often linked to compound instability, off-target effects, and nonspecific blood cell interactions, highlight the need for better understanding. Conversely, the rapid distribution, tumor penetration, and clearance of VHHs could be advantageous, potentially reducing toxicity by minimizing prolonged exposure. These attributes make single-domain antibodies strong candidates for the next generation of ADCs, potentially enhancing both efficacy and safety.
Collapse
Affiliation(s)
- Víctor Manuel Medina Pérez
- Molecular Oncology Group, Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain;
| | - Marta Baselga
- Molecular Oncology Group, Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain;
| | - Alberto J. Schuhmacher
- Molecular Oncology Group, Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain;
- Fundación Aragonesa para la Investigación y el Desarrollo (ARAID), 50018 Zaragoza, Spain
| |
Collapse
|
3
|
Hu L, Zhang S, Sienkiewicz J, Zhou H, Berahovich R, Sun J, Li M, Ocampo A, Liu X, Huang Y, Harto H, Xu S, Golubovskaya V, Wu L. HER2-CD3-Fc Bispecific Antibody-Encoding mRNA Delivered by Lipid Nanoparticles Suppresses HER2-Positive Tumor Growth. Vaccines (Basel) 2024; 12:808. [PMID: 39066446 PMCID: PMC11281407 DOI: 10.3390/vaccines12070808] [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: 05/31/2024] [Revised: 07/10/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024] Open
Abstract
The human epidermal growth factor receptor 2 (HER2) is a transmembrane tyrosine kinase receptor and tumor-associated antigen abnormally expressed in various types of cancer, including breast, ovarian, and gastric cancer. HER2 overexpression is highly correlated with increased tumor aggressiveness, poorer prognosis, and shorter overall survival. Consequently, multiple HER2-targeted therapies have been developed and approved; however, only a subset of patients benefit from these treatments, and relapses are common. More potent and durable HER2-targeted therapies are desperately needed for patients with HER2-positive cancers. In this study, we developed a lipid nanoparticle (LNP)-based therapy formulated with mRNA encoding a novel HER2-CD3-Fc bispecific antibody (bsAb) for HER2-positive cancers. The LNPs efficiently transfected various types of cells, such as HEK293S, SKOV-3, and A1847, leading to robust and sustained secretion of the HER2-CD3-Fc bsAb with high binding affinity to both HER2 and CD3. The bsAb induced potent T-cell-directed cytotoxicity, along with secretion of IFN-λ, TNF-α, and granzyme B, against various types of HER2-positive tumor cells in vitro, including A549, NCI-H460, SKOV-3, A1847, SKBR3, and MDA-MB-231. The bsAb-mediated antitumor effect is highly specific and strictly dependent on its binding to HER2, as evidenced by the gained resistance of A549 and A1847 her2 knockout cells and the acquired sensitivity of mouse 4T1 cells overexpressing the human HER2 extracellular domain (ECD) or epitope-containing subdomain IV to the bsAb-induced T cell cytotoxicity. The bsAb also relies on its binding to CD3 for T-cell recruitment, as ablation of CD3 binding abolished the bsAb's ability to elicit antitumor activity. Importantly, intratumoral injection of the HER2-CD3-Fc mRNA-LNPs triggers a strong antitumor response and completely blocks HER2-positive tumor growth in a mouse xenograft model of human ovarian cancer. These results indicate that the novel HER2-CD3-Fc mRNA-LNP-based therapy has the potential to effectively treat HER2-positive cancer.
Collapse
Affiliation(s)
- Liang Hu
- Promab Biotechnologies, 2600 Hilltop Drive, Richmond, CA 94806, USA; (L.H.); (S.Z.); (J.S.); (H.Z.); (R.B.); (J.S.); (M.L.); (A.O.); (X.L.); (Y.H.); (H.H.); (S.X.)
| | - Shiming Zhang
- Promab Biotechnologies, 2600 Hilltop Drive, Richmond, CA 94806, USA; (L.H.); (S.Z.); (J.S.); (H.Z.); (R.B.); (J.S.); (M.L.); (A.O.); (X.L.); (Y.H.); (H.H.); (S.X.)
| | - John Sienkiewicz
- Promab Biotechnologies, 2600 Hilltop Drive, Richmond, CA 94806, USA; (L.H.); (S.Z.); (J.S.); (H.Z.); (R.B.); (J.S.); (M.L.); (A.O.); (X.L.); (Y.H.); (H.H.); (S.X.)
| | - Hua Zhou
- Promab Biotechnologies, 2600 Hilltop Drive, Richmond, CA 94806, USA; (L.H.); (S.Z.); (J.S.); (H.Z.); (R.B.); (J.S.); (M.L.); (A.O.); (X.L.); (Y.H.); (H.H.); (S.X.)
| | - Robert Berahovich
- Promab Biotechnologies, 2600 Hilltop Drive, Richmond, CA 94806, USA; (L.H.); (S.Z.); (J.S.); (H.Z.); (R.B.); (J.S.); (M.L.); (A.O.); (X.L.); (Y.H.); (H.H.); (S.X.)
| | - Jinying Sun
- Promab Biotechnologies, 2600 Hilltop Drive, Richmond, CA 94806, USA; (L.H.); (S.Z.); (J.S.); (H.Z.); (R.B.); (J.S.); (M.L.); (A.O.); (X.L.); (Y.H.); (H.H.); (S.X.)
| | - Michael Li
- Promab Biotechnologies, 2600 Hilltop Drive, Richmond, CA 94806, USA; (L.H.); (S.Z.); (J.S.); (H.Z.); (R.B.); (J.S.); (M.L.); (A.O.); (X.L.); (Y.H.); (H.H.); (S.X.)
| | - Adrian Ocampo
- Promab Biotechnologies, 2600 Hilltop Drive, Richmond, CA 94806, USA; (L.H.); (S.Z.); (J.S.); (H.Z.); (R.B.); (J.S.); (M.L.); (A.O.); (X.L.); (Y.H.); (H.H.); (S.X.)
| | - Xianghong Liu
- Promab Biotechnologies, 2600 Hilltop Drive, Richmond, CA 94806, USA; (L.H.); (S.Z.); (J.S.); (H.Z.); (R.B.); (J.S.); (M.L.); (A.O.); (X.L.); (Y.H.); (H.H.); (S.X.)
| | - Yanwei Huang
- Promab Biotechnologies, 2600 Hilltop Drive, Richmond, CA 94806, USA; (L.H.); (S.Z.); (J.S.); (H.Z.); (R.B.); (J.S.); (M.L.); (A.O.); (X.L.); (Y.H.); (H.H.); (S.X.)
| | - Hizkia Harto
- Promab Biotechnologies, 2600 Hilltop Drive, Richmond, CA 94806, USA; (L.H.); (S.Z.); (J.S.); (H.Z.); (R.B.); (J.S.); (M.L.); (A.O.); (X.L.); (Y.H.); (H.H.); (S.X.)
| | - Shirley Xu
- Promab Biotechnologies, 2600 Hilltop Drive, Richmond, CA 94806, USA; (L.H.); (S.Z.); (J.S.); (H.Z.); (R.B.); (J.S.); (M.L.); (A.O.); (X.L.); (Y.H.); (H.H.); (S.X.)
| | - Vita Golubovskaya
- Promab Biotechnologies, 2600 Hilltop Drive, Richmond, CA 94806, USA; (L.H.); (S.Z.); (J.S.); (H.Z.); (R.B.); (J.S.); (M.L.); (A.O.); (X.L.); (Y.H.); (H.H.); (S.X.)
| | - Lijun Wu
- Promab Biotechnologies, 2600 Hilltop Drive, Richmond, CA 94806, USA; (L.H.); (S.Z.); (J.S.); (H.Z.); (R.B.); (J.S.); (M.L.); (A.O.); (X.L.); (Y.H.); (H.H.); (S.X.)
- Forevertek Biotechnology, Janshan Road, Changsha Hi-Tech Industrial Development Zone, Changsha 410205, China
| |
Collapse
|
4
|
diZerega GS, Maulhardt HA, Verco SJ, Marin AM, Baltezor MJ, Mauro SA, Iacobucci MA. Intratumoral Injection of Large Surface Area Microparticle Taxanes in Carcinomas Increases Immune Effector Cell Concentrations, Checkpoint Expression, and Synergy with Checkpoint Inhibitors: A Review of Preclinical and Clinical Studies. Oncol Ther 2024; 12:31-55. [PMID: 38289576 PMCID: PMC10881942 DOI: 10.1007/s40487-024-00261-y] [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: 11/03/2023] [Accepted: 01/04/2024] [Indexed: 02/23/2024] Open
Abstract
This review summarizes development of large surface area microparticle paclitaxel (LSAM-PTX) and docetaxel (LSAM-DTX) for local treatment of primary carcinomas with emphasis on immunomodulation. Intratumoral (IT) delivery of LSAM-PTX and LSAM-DTX provides continuous, therapeutic drug levels for several weeks. Preclinical studies and clinical trials reported a reduction in tumor volume (TV) and immunomodulation in primary tumor and peripheral blood with increases in innate and adaptive immune cells and decreases in suppressor cells. Increased levels of checkpoint expression of immune cells occurred in clinical trials of high-risk non-muscle-invasive bladder cancer (LSAM-DTX) and unresectable localized pancreatic cancer (LSAM-PTX). TV reduction and increases in immune effector cells occurred following IT LSAM-DTX and IT LSAM-PTX together with anti-mCTLA-4 and anti-mPD-1, respectively. Synergistic benefits from combinatorial therapy in a 4T1-Luc breast cancer model included reduction of metastasis with IT LSAM-DTX + anti-mCTLA-4. IT LSAM-PTX and LSAM-DTX are tumoricidal, immune enhancing, and may improve solid tumor response to immune checkpoint inhibitors without additional systemic toxicity.
Collapse
Affiliation(s)
- Gere S diZerega
- US Biotest, Inc., 231 Bonetti Drive, Suite 240, San Luis Obispo, CA, 93401, USA.
- NanOlogy, LLC., 3909 Hulen Street, Fort Worth, TX, 76107, USA.
| | - Holly A Maulhardt
- US Biotest, Inc., 231 Bonetti Drive, Suite 240, San Luis Obispo, CA, 93401, USA
| | - Shelagh J Verco
- US Biotest, Inc., 231 Bonetti Drive, Suite 240, San Luis Obispo, CA, 93401, USA
| | - Alyson M Marin
- US Biotest, Inc., 231 Bonetti Drive, Suite 240, San Luis Obispo, CA, 93401, USA
| | | | - Samantha A Mauro
- US Biotest, Inc., 231 Bonetti Drive, Suite 240, San Luis Obispo, CA, 93401, USA
| | | |
Collapse
|
5
|
Shaha S, Rodrigues D, Mitragotri S. Locoregional drug delivery for cancer therapy: Preclinical progress and clinical translation. J Control Release 2024; 367:737-767. [PMID: 38325716 DOI: 10.1016/j.jconrel.2024.01.072] [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: 11/24/2023] [Revised: 01/26/2024] [Accepted: 01/31/2024] [Indexed: 02/09/2024]
Abstract
Systemic drug delivery is the current clinically preferred route for cancer therapy. However, challenges associated with tumor localization and off-tumor toxic effects limit the clinical effectiveness of this route. Locoregional drug delivery is an emerging viable alternative to systemic therapies. With the improvement in real-time imaging technologies and tools for direct access to tumor lesions, the clinical applicability of locoregional drug delivery is becoming more prominent. Theoretically, locoregional treatments can bypass challenges faced by systemic drug delivery. Preclinically, locoregional delivery of drugs has demonstrated enhanced therapeutic efficacy with limited off-target effects while still yielding an abscopal effect. Clinically, an array of locoregional strategies is under investigation for the delivery of drugs ranging in target and size. Locoregional tumor treatment strategies can be classified into two main categories: 1) direct drug infusion via injection or implanted port and 2) extended drug elution via injected or implanted depot. The number of studies investigating locoregional drug delivery strategies for cancer treatment is rising exponentially, in both preclinical and clinical settings, with some approaches approved for clinical use. Here, we highlight key preclinical advances and the clinical relevance of such locoregional delivery strategies in the treatment of cancer. Furthermore, we critically analyze 949 clinical trials involving locoregional drug delivery and discuss emerging trends.
Collapse
Affiliation(s)
- Suyog Shaha
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Allston, MA 02134, USA; Wyss Institute for Biologically Inspired Engineering, Boston, MA 02115, USA
| | - Danika Rodrigues
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Allston, MA 02134, USA; Wyss Institute for Biologically Inspired Engineering, Boston, MA 02115, USA
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Allston, MA 02134, USA; Wyss Institute for Biologically Inspired Engineering, Boston, MA 02115, USA.
| |
Collapse
|
6
|
Karan S, Jung E, Boone C, Steinmetz NF. Synergistic combination therapy using cowpea mosaic virus intratumoral immunotherapy and Lag-3 checkpoint blockade. Cancer Immunol Immunother 2024; 73:51. [PMID: 38349406 PMCID: PMC10864561 DOI: 10.1007/s00262-024-03636-2] [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: 11/30/2023] [Accepted: 01/15/2024] [Indexed: 02/15/2024]
Abstract
Immune checkpoint therapy (ICT) for cancer can yield dramatic clinical responses; however, these may only be observed in a minority of patients. These responses can be further limited by subsequent disease recurrence and resistance. Combination immunotherapy strategies are being developed to overcome these limitations. We have previously reported enhanced efficacy of combined intratumoral cowpea mosaic virus immunotherapy (CPMV IIT) and ICT approaches. Lymphocyte-activation gene-3 (LAG-3) is a next-generation inhibitory immune checkpoint with broad expression across multiple immune cell subsets. Its expression increases on activated T cells and contributes to T cell exhaustion. We observed heightened efficacy of a combined CPMV IIT and anti-LAG-3 treatment in a mouse model of melanoma. Further, LAG-3 expression was found to be increased within the TME following intratumoral CPMV administration. The integration of CPMV IIT with LAG-3 inhibition holds significant potential to improve treatment outcomes by concurrently inducing a comprehensive anti-tumor immune response, enhancing local immune activation, and mitigating T cell exhaustion.
Collapse
Affiliation(s)
- Sweta Karan
- Department of Nanoengineering, University of California, San Diego, La Jolla, CA, USA
| | - Eunkyeong Jung
- Department of Nanoengineering, University of California, San Diego, La Jolla, CA, USA
| | - Christine Boone
- Department of Radiology, University of California, San Diego, La Jolla, CA, USA.
| | - Nicole F Steinmetz
- Department of Nanoengineering, University of California, San Diego, La Jolla, CA, USA.
- Department of Radiology, University of California, San Diego, La Jolla, CA, USA.
- Shu and K.C. Chien and Peter Farrell Collaboratory, University of California, San Diego, La Jolla, CA, USA.
- Center for Nano-ImmunoEngineering, University of California, San Diego, La Jolla, CA, USA.
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA.
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA.
- Institute for Materials Discovery and Design, University of California, San Diego, La Jolla, CA, USA.
- Center for Engineering in Cancer, Institute of Engineering Medicine, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
7
|
Sandbank E, Eckerling A, Margalit A, Sorski L, Ben-Eliyahu S. Immunotherapy during the Immediate Perioperative Period: A Promising Approach against Metastatic Disease. Curr Oncol 2023; 30:7450-7477. [PMID: 37623021 PMCID: PMC10453707 DOI: 10.3390/curroncol30080540] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 08/26/2023] Open
Abstract
Tumor excision is a necessary life-saving procedure in most solid cancers. However, surgery and the days before and following it, known as the immediate perioperative period (IPP), entail numerous prometastatic processes, including the suppression of antimetastatic immunity and direct stimulation of minimal residual disease (MRD). Thus, the IPP is pivotal in determining long-term cancer outcomes, presenting a short window of opportunity to circumvent perioperative risk factors by employing several therapeutic approaches, including immunotherapy. Nevertheless, immunotherapy is rarely examined or implemented during this short timeframe, due to both established and hypothetical contraindications to surgery. Herein, we analyze how various aspects of the IPP promote immunosuppression and progression of MRD, and how potential IPP application of immunotherapy may interact with these deleterious processes. We discuss the feasibility and safety of different immunotherapies during the IPP with a focus on the latest approaches of immune checkpoint inhibition. Last, we address the few past and ongoing clinical trials that exploit the IPP timeframe for anticancer immunotherapy. Accordingly, we suggest that several specific immunotherapies can be safely and successfully applied during the IPP, alone or with supporting interventions, which may improve patients' resistance to MRD and overall survival.
Collapse
Affiliation(s)
- Elad Sandbank
- Neuro-Immunology Research Unit, School of Psychological Sciences, Tel Aviv University, Tel Aviv 69978, Israel; (E.S.); (A.E.); (L.S.)
| | - Anabel Eckerling
- Neuro-Immunology Research Unit, School of Psychological Sciences, Tel Aviv University, Tel Aviv 69978, Israel; (E.S.); (A.E.); (L.S.)
| | - Adam Margalit
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel;
| | - Liat Sorski
- Neuro-Immunology Research Unit, School of Psychological Sciences, Tel Aviv University, Tel Aviv 69978, Israel; (E.S.); (A.E.); (L.S.)
| | - Shamgar Ben-Eliyahu
- Neuro-Immunology Research Unit, School of Psychological Sciences, Tel Aviv University, Tel Aviv 69978, Israel; (E.S.); (A.E.); (L.S.)
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel;
| |
Collapse
|
8
|
Sunga GM, Hartgerink J, Sikora AG, Young S. Enhancement of Immunotherapies in Head and Neck Cancers Using Biomaterial-Based Treatment Strategies. Tissue Eng Part C Methods 2023; 29:257-275. [PMID: 37183412 PMCID: PMC10282827 DOI: 10.1089/ten.tec.2023.0090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 05/12/2023] [Indexed: 05/16/2023] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a challenging disease to treat because of typically late-stage diagnoses and tumor formation in difficult-to-treat areas, sensitive to aggressive or invasive treatments. To date, HNSCC treatments have been limited to surgery, radiotherapy, and chemotherapy, which may have significant morbidity and often lead to long-lasting side effects. The development of immunotherapies has revolutionized cancer treatment by providing a promising alternative to standard-of-care therapies. However, single-agent immunotherapy has been only modestly effective in the treatment of various cancers, including HNSCC, with most patients receiving no overall benefit or increased survival. In addition, single-agent immunotherapy's limitations, namely immune-related side effects and the necessity of multidose treatments, must be addressed to further improve treatment efficacy. Biocompatible biomaterials, in combination with cancer immunotherapies, offer numerous advantages in the concentration, localization, and controlled release of drugs, cancer antigens, and immune cells. Biomaterial structures are diverse, and their design can generally be customized to enhance immunotherapy response. In preclinical settings, the use of biomaterials has shown great promise in improving the efficacy of single-agent immunotherapy. Herein, we provide an overview of current immunotherapy treatments for HNSCC and their limitations, as well as the potential applications of biomaterials in enhancing cancer immunotherapies. Impact Statement Advances in anticancer immunotherapies for the past 30 years have yielded exciting clinical results and provided alternatives to long-standing standard-of-care treatments, which are associated with significant toxicities and long-term morbidity. However, patients with head and neck squamous cell carcinoma (HNSCC) have not benefited from immunotherapies as much as patients with other cancers. Immunotherapy limitations include systemic side effects, therapeutic resistance, poor delivery kinetics, and limited patient responses. Biomaterial-enhanced immunotherapies, as explored in this review, are a potentially powerful means of achieving localized drug delivery, sustained and controlled drug release, and immunomodulation. They may overcome current treatment limitations and improve patient outcomes and care.
Collapse
Affiliation(s)
- Gemalene M. Sunga
- Katz Department of Oral and Maxillofacial Surgery, The University of Texas Health Science Center at Houston School of Dentistry, Houston, Texas, USA
| | - Jeffrey Hartgerink
- Department of Chemistry, Rice University, Houston, Texas, USA
- Department of Bioengineering, Rice University, Houston, Texas, USA
| | - Andrew G. Sikora
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Simon Young
- Katz Department of Oral and Maxillofacial Surgery, The University of Texas Health Science Center at Houston School of Dentistry, Houston, Texas, USA
| |
Collapse
|