1
|
Kim HU, Kim YK. Bispecific antibodies and CLEM: an analytical approach to advanced cell imaging for therapeutic strategies. Appl Microsc 2025; 55:1. [PMID: 39828773 PMCID: PMC11743405 DOI: 10.1186/s42649-024-00106-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 12/15/2024] [Indexed: 01/22/2025] Open
Abstract
The development of bispecific antibodies (BsAbs) represents a significant advancement in therapeutic antibody design, enabling the simultaneous targeting of two different antigens. This dual-targeting capability enhances therapeutic efficacy, particularly in complex diseases like cancer, where tumor heterogeneity presents a significant challenge for traditional treatments. By bridging two distinct pathways, BsAbs can improve specificity and minimize off-target effects, making them invaluable in therapeutic contexts. Integrating advanced imaging techniques, particularly Correlative Light and Electron Microscopy (CLEM), offers a unique opportunity to visualize the dynamic interactions of BsAbs within cellular environments. CLEM combines the strengths of optical and electron microscopy, allowing researchers to observe real-time antibody-antigen interactions at nanoscale resolution. This synergy not only deepens our understanding of BsAbs' mechanisms of action but also provides critical insights into their spatial distribution, binding kinetics, and functional dynamics in live cells. In this review, the integration of BsAbs and CLEM paves the way for targeted therapeutic strategies, fostering the development of more effective treatments that can adapt to the complexities of disease pathology.
Collapse
Affiliation(s)
- Han-Ul Kim
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Kangwon Center for Systems Imaging, Kangwondaehak-gil, Chuncheon-si, Gangwon-do, 24341, Republic of Korea
| | - Young Kwan Kim
- Kangwon Center for Systems Imaging, Kangwondaehak-gil, Chuncheon-si, Gangwon-do, 24341, Republic of Korea.
| |
Collapse
|
2
|
Manoharan
Nair Sudha Kumari S, Thankappan Suryabai X. Sensing the Future-Frontiers in Biosensors: Exploring Classifications, Principles, and Recent Advances. ACS OMEGA 2024; 9:48918-48987. [PMID: 39713646 PMCID: PMC11656264 DOI: 10.1021/acsomega.4c07991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/29/2024] [Accepted: 11/18/2024] [Indexed: 12/24/2024]
Abstract
Biosensors are transforming healthcare by delivering swift, precise, and economical diagnostic solutions. These analytical instruments combine biological indicators with physical transducers to identify and quantify biomarkers, thereby improving illness detection, management, and patient surveillance. Biosensors are widely utilized in healthcare for the diagnosis of chronic and infectious diseases, tailored treatment, and real-time health monitoring. This thorough overview examines several categories of biosensors and their uses in the detection of numerous biomarkers, including glucose, proteins, nucleic acids, and infections. Biosensors are commonly classified based on the type of transducer employed or the specific biorecognition element utilized. This review introduces a novel classification based on substrate morphology, offering a comprehensive perspective on biosensor categorization. Considerable emphasis is placed on the advancement of point-of-care biosensors, facilitating decentralized diagnostics and alleviating the strain on centralized healthcare systems. Recent advancements in nanotechnology have significantly improved the sensitivity, selectivity, and downsizing of biosensors, rendering them more efficient and accessible. The study examines problems such as stability, reproducibility, and regulatory approval that must be addressed to enable the widespread implementation of biosensors in clinical environments. The study examines the amalgamation of biosensors with wearable devices and smartphones, emphasizing the prospects for ongoing health surveillance and individualized medical care. This viewpoint clarifies the distinct types of biosensors and their particular roles, together with recent developments in the "smart biosensor" sector, facilitated by artificial intelligence and the Internet of Medical Things (IoMT). This novel approach seeks to deliver a comprehensive evaluation of the present condition of biosensor technology in healthcare, recent developments, and prospective paths, emphasizing their significance in influencing the future of medical diagnostics and patient care.
Collapse
Affiliation(s)
- Sumitha Manoharan
Nair Sudha Kumari
- Centre for
Advanced Materials Research, Department of Physics, Government College for Women, Thiruvananthapuram, University of Kerala, Kerala 695014, India
| | - Xavier Thankappan Suryabai
- Centre for
Advanced Materials Research, Department of Physics, Government College for Women, Thiruvananthapuram, University of Kerala, Kerala 695014, India
| |
Collapse
|
3
|
Mundhara N, Sadhukhan P. Cracking the Codes behind Cancer Cells' Immune Evasion. Int J Mol Sci 2024; 25:8899. [PMID: 39201585 PMCID: PMC11354234 DOI: 10.3390/ijms25168899] [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: 07/12/2024] [Revised: 08/03/2024] [Accepted: 08/09/2024] [Indexed: 09/02/2024] Open
Abstract
Immune evasion is a key phenomenon in understanding tumor recurrence, metastasis, and other critical steps in tumor progression. The tumor microenvironment (TME) is in constant flux due to the tumor's ability to release signals that affect it, while immune cells within it can impact cancer cell behavior. Cancer cells undergo several changes, which can change the enrichment of different immune cells and modulate the activity of existing immune cells in the tumor microenvironment. Cancer cells can evade immune surveillance by downregulating antigen presentation or expressing immune checkpoint molecules. High levels of tumor-infiltrating lymphocytes (TILs) correlate with better outcomes, and robust immune responses can control tumor growth. On the contrary, increased enrichment of Tregs, myeloid-derived suppressor cells, and M2-like anti-inflammatory macrophages can hinder effective immune surveillance and predict poor prognosis. Overall, understanding these immune evasion mechanisms guides therapeutic strategies. Researchers aim to modulate the TME to enhance immune surveillance and improve patient outcomes. In this review article, we strive to summarize the composition of the tumor immune microenvironment, factors affecting the tumor immune microenvironment (TIME), and different therapeutic modalities targeting the immune cells. This review is a first-hand reference to understand the basics of immune surveillance and immune evasion.
Collapse
Affiliation(s)
| | - Pritam Sadhukhan
- Department of Oncology, Johns Hopkins University, Baltimore, MD 21287, USA
| |
Collapse
|
4
|
Rolin C, Zimmer J, Seguin-Devaux C. Bridging the gap with multispecific immune cell engagers in cancer and infectious diseases. Cell Mol Immunol 2024; 21:643-661. [PMID: 38789528 PMCID: PMC11214628 DOI: 10.1038/s41423-024-01176-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 04/23/2024] [Indexed: 05/26/2024] Open
Abstract
By binding to multiple antigens simultaneously, multispecific antibodies are expected to substantially improve both the activity and long-term efficacy of antibody-based immunotherapy. Immune cell engagers, a subclass of antibody-based constructs, consist of engineered structures designed to bridge immune effector cells to their target, thereby redirecting the immune response toward the tumor cells or infected cells. The increasing number of recent clinical trials evaluating immune cell engagers reflects the important role of these molecules in new therapeutic approaches for cancer and infections. In this review, we discuss how different immune cell types (T and natural killer lymphocytes, as well as myeloid cells) can be bound by immune cell engagers in immunotherapy for cancer and infectious diseases. Furthermore, we explore the preclinical and clinical advancements of these constructs, and we discuss the challenges in translating the current knowledge from cancer to the virology field. Finally, we speculate on the promising future directions that immune cell engagers may take in cancer treatment and antiviral therapy.
Collapse
Affiliation(s)
- Camille Rolin
- Department of Infection and Immunity, Luxembourg Institute of Health, 29 Rue Henri Koch, L-4354, Esch-Sur-Alzette, Luxembourg.
- University of Luxembourg, 2 Place de l'Université, L-4365, Esch-sur-Alzette, Luxembourg.
| | - Jacques Zimmer
- Department of Infection and Immunity, Luxembourg Institute of Health, 29 Rue Henri Koch, L-4354, Esch-Sur-Alzette, Luxembourg
| | - Carole Seguin-Devaux
- Department of Infection and Immunity, Luxembourg Institute of Health, 29 Rue Henri Koch, L-4354, Esch-Sur-Alzette, Luxembourg
| |
Collapse
|
5
|
Conti V, Polcaro G, De Bellis E, Donnarumma D, De Rosa F, Stefanelli B, Corbi G, Sabbatino F, Filippelli A. Natural Health Products for Anti-Cancer Treatment: Evidence and Controversy. J Pers Med 2024; 14:685. [PMID: 39063939 PMCID: PMC11278393 DOI: 10.3390/jpm14070685] [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: 05/20/2024] [Revised: 06/14/2024] [Accepted: 06/21/2024] [Indexed: 07/28/2024] Open
Abstract
Natural Health Products (NHPs) have long been considered a valuable therapeutic approach for the prevention and treatment of various diseases, including cancer. However, research on this topic has led to inconclusive and often controversial results. This review aims to provide a comprehensive update of the effects and mechanisms related to the use of NHPs, to describe the results of randomized clinical trials (RCTs) on their effects in cancer patients, and to critically discuss factors influencing clinical outcomes. RCTs available in the literature, even those studying the same NHP, are very heterogeneous in terms of indications, doses, route and timing of administration, and outcomes evaluated. Silymarin, ginsenoside, and vitamin E appear to be useful in attenuating adverse events related to radiotherapy or chemotherapy, and curcumin and lycopene might provide some benefit in patients with prostate cancer. Most RCTs have not clarified whether NHP supplementation provides any real benefit, while harmful effects have been shown in some cases. Overall, the available data suggest that although there is some evidence to support the benefits of NHPs in the management of cancer patients, further clinical trials with the same design are needed before their introduction into clinical practice can be considered.
Collapse
Affiliation(s)
- Valeria Conti
- Department of Medicine, Surgery, and Dentistry, Scuola Medica Salernitana, University of Salerno, 84081 Baronissi, Italy; (V.C.); (G.P.); (F.D.R.); (B.S.); (F.S.); (A.F.)
- Clinical Pharmacology Unit, San Giovanni di Dio e Ruggi d’Aragona University Hospital, 84131 Salerno, Italy
| | - Giovanna Polcaro
- Department of Medicine, Surgery, and Dentistry, Scuola Medica Salernitana, University of Salerno, 84081 Baronissi, Italy; (V.C.); (G.P.); (F.D.R.); (B.S.); (F.S.); (A.F.)
| | - Emanuela De Bellis
- PhD School “Clinical and Translational Oncology (CTO)”, Scuola Superiore Meridionale, University of Naples “Federico II”, 80138 Naples, Italy;
| | - Danilo Donnarumma
- PhD School “Clinical and Translational Oncology (CTO)”, Scuola Superiore Meridionale, University of Naples “Federico II”, 80138 Naples, Italy;
| | - Federica De Rosa
- Department of Medicine, Surgery, and Dentistry, Scuola Medica Salernitana, University of Salerno, 84081 Baronissi, Italy; (V.C.); (G.P.); (F.D.R.); (B.S.); (F.S.); (A.F.)
| | - Berenice Stefanelli
- Department of Medicine, Surgery, and Dentistry, Scuola Medica Salernitana, University of Salerno, 84081 Baronissi, Italy; (V.C.); (G.P.); (F.D.R.); (B.S.); (F.S.); (A.F.)
| | - Graziamaria Corbi
- Department of Translational Medical Sciences, University of Naples “Federico II”, 80131 Naples, Italy;
| | - Francesco Sabbatino
- Department of Medicine, Surgery, and Dentistry, Scuola Medica Salernitana, University of Salerno, 84081 Baronissi, Italy; (V.C.); (G.P.); (F.D.R.); (B.S.); (F.S.); (A.F.)
- Oncology Unit, University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy
| | - Amelia Filippelli
- Department of Medicine, Surgery, and Dentistry, Scuola Medica Salernitana, University of Salerno, 84081 Baronissi, Italy; (V.C.); (G.P.); (F.D.R.); (B.S.); (F.S.); (A.F.)
- Clinical Pharmacology Unit, San Giovanni di Dio e Ruggi d’Aragona University Hospital, 84131 Salerno, Italy
| |
Collapse
|
6
|
Wang L, Jiang H, Yin X, Liang T, Li G, Ding C, Yang M, Zhang L, Liu J, Xu Y. PHE1-based IgG-like antibody platform provides a novel strategy for enhanced T-cell immunotherapy. Front Immunol 2024; 15:1415834. [PMID: 38933272 PMCID: PMC11201533 DOI: 10.3389/fimmu.2024.1415834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
Introduction Bispecific antibodies (BsAbs) can simultaneously target two epitopes of different antigenic targets, bringing possibilities for diversity in antibody drug design and are promising tools for the treatment of cancers and other diseases. T-cell engaging bsAb is an important application of the bispecific antibody, which could promote T cell-mediated tumor cell killing by targeting tumor-associated antigen (TAA) and CD3 at the same time. Methods This study comprised antibodies purification, Elisa assay for antigen binding, cytotoxicity assays, T cell activation by flow cytometry in vitro and xenogenic tumor model in vivo. Results We present a novel bsAb platform named PHE-Ig technique to promote cognate heavy chain (HC)-light chain (LC) pairing by replacing the CH1/CL regions of different monoclonal antibodies (mAbs) with the natural A and B chains of PHE1 fragment of Integrin β2 based on the knob-in-hole (KIH) technology. We had also verified that PHE-Ig technology can be effectively used as a platform to synthesize different desired bsAbs for T-cell immunotherapy. Especially, BCMA×CD3 PHE-Ig bsAbs exhibited robust anti-multiple myeloma (MM) activity in vitro and in vivo. Discussion Moreover, PHE1 domain was further shortened with D14G and R41S mutations, named PHE-S, and the PHE-S-based BCMA×CD3 bsAbs also showed anti BCMA+ tumor effect in vitro and in vivo, bringing more possibilities for the development and optimization of different bsAbs. To sum up, PHE1-based IgG-like antibody platform for bsAb construction provides a novel strategy for enhanced T-cell immunotherapy.
Collapse
Affiliation(s)
- Lingbin Wang
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haojie Jiang
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuying Yin
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tingting Liang
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guoming Li
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Ding
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mina Yang
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lin Zhang
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junling Liu
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Synvida Biotechnology Co., Ltd, Shanghai, China
| | - Yanyan Xu
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
7
|
Mehlhaff E, Miller D, Ebben JD, Dobrzhanskyi O, Uboha NV. Targeted Agents in Esophagogastric Cancer Beyond Human Epidermal Growth Factor Receptor-2. Hematol Oncol Clin North Am 2024; 38:659-675. [PMID: 38485551 DOI: 10.1016/j.hoc.2024.02.006] [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] [Indexed: 05/06/2024]
Abstract
Gastroesophageal cancers are highly diverse tumors in terms of their anatomic and molecular characteristics, making drug development challenging. Recent advancements in understanding the molecular profiles of these cancers have led to the identification of several new biomarkers. Ongoing clinical trials are investigating new targeted agents with promising results. CLDN18.2 has emerged as a biomarker with established activity of associated targeted therapies. Other targeted agents, such as bemarituzumab and DKN-01, are under active investigation. As new agents are incorporated into the treatment continuum, the questions of biomarker overlap, tumor heterogeneity, and toxicity management will need to be addressed.
Collapse
Affiliation(s)
- Eric Mehlhaff
- Division of Hematology, Medical Oncology and Palliative Care, Department of Medicine, University of Wisconsin School of Medicine and Public Health, University of Wisconsin, 600 Highland Avenue, Madison, WI 53792, USA
| | - Devon Miller
- Division of Hematology, Medical Oncology and Palliative Care, Department of Medicine, University of Wisconsin School of Medicine and Public Health, University of Wisconsin, 600 Highland Avenue, Madison, WI 53792, USA
| | - Johnathan D Ebben
- Division of Hematology, Medical Oncology and Palliative Care, Department of Medicine, University of Wisconsin School of Medicine and Public Health, University of Wisconsin, 600 Highland Avenue, Madison, WI 53792, USA
| | - Oleksii Dobrzhanskyi
- Upper Gastrointestinal Tumors Department, National Cancer Institute, Kyiv, Ukraine
| | - Nataliya V Uboha
- Division of Hematology, Medical Oncology and Palliative Care, Department of Medicine, University of Wisconsin School of Medicine and Public Health, University of Wisconsin, 600 Highland Avenue, Madison, WI 53792, USA; University of Wisconsin, Carbone Cancer Center, Madison, WI, USA.
| |
Collapse
|
8
|
Li T, Niu M, Zhou J, Wu K, Yi M. The enhanced antitumor activity of bispecific antibody targeting PD-1/PD-L1 signaling. Cell Commun Signal 2024; 22:179. [PMID: 38475778 PMCID: PMC10935874 DOI: 10.1186/s12964-024-01562-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 03/05/2024] [Indexed: 03/14/2024] Open
Abstract
The programmed cell death 1 (PD-1) signaling pathway, a key player in immune checkpoint regulation, has become a focal point in cancer immunotherapy. In the context of cancer, upregulated PD-L1 on tumor cells can result in T cell exhaustion and immune evasion, fostering tumor progression. The advent of PD-1/PD-L1 inhibitor has demonstrated clinical success by unleashing T cells from exhaustion. Nevertheless, challenges such as resistance and adverse effects have spurred the exploration of innovative strategies, with bispecific antibodies (BsAbs) emerging as a promising frontier. BsAbs offer a multifaceted approach to cancer immunotherapy by simultaneously targeting PD-L1 and other immune regulatory molecules. We focus on recent advancements in PD-1/PD-L1 therapy with a particular emphasis on the development and potential of BsAbs, especially in the context of solid tumors. Various BsAb products targeting PD-1 signaling are discussed, highlighting their unique mechanisms of action and therapeutic potential. Noteworthy examples include anti-TGFβ × PD-L1, anti-CD47 × PD-L1, anti-VEGF × PD-L1, anti-4-1BB × PD-L1, anti-LAG-3 × PD-L1, and anti-PD-1 × CTLA-4 BsAbs. Besides, we summarize ongoing clinical studies evaluating the efficacy and safety of these innovative BsAb agents. By unraveling the intricacies of the tumor microenvironment and harnessing the synergistic effects of anti-PD-1/PD-L1 BsAbs, there exists the potential to elevate the precision and efficacy of cancer immunotherapy, ultimately enabling the development of personalized treatment strategies tailored to individual patient profiles.
Collapse
Affiliation(s)
- Tianye Li
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, People's Republic of China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, China
| | - Mengke Niu
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China
| | - Jianwei Zhou
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, People's Republic of China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, China
| | - Kongming Wu
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China.
| | - Ming Yi
- Department of Breast Surgery, College of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, 310000, People's Republic of China.
| |
Collapse
|
9
|
Cappuzzello E, Vigolo E, D’Accardio G, Astori G, Rosato A, Sommaggio R. How can Cytokine-induced killer cells overcome CAR-T cell limits. Front Immunol 2023; 14:1229540. [PMID: 37675107 PMCID: PMC10477668 DOI: 10.3389/fimmu.2023.1229540] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 08/02/2023] [Indexed: 09/08/2023] Open
Abstract
The successful treatment of patients affected by B-cell malignancies with Chimeric Antigen Receptor (CAR)-T cells represented a breakthrough in the field of adoptive cell therapy (ACT). However, CAR-T therapy is not an option for every patient, and several needs remain unmet. In particular, the production of CAR-T cells is expensive, labor-intensive and logistically challenging; additionally, the toxicities deriving from CAR-T cells infusion, such as cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS), have been documented extensively. Alternative cellular therapy products such as Cytokine-induced killer (CIK) cells have the potential to overcome some of these obstacles. CIK cells are a heterogeneous population of polyclonal CD3+CD56+ T cells with phenotypic and functional properties of NK cells. CIK cell cytotoxicity is exerted in a major histocompatibility complex (MHC)-unrestricted manner through the engagement of natural killer group 2 member D (NKG2D) molecules, against a wide range of hematological and solid tumors without the need for prior antigen exposure or priming. The foremost potential of CIK cells lies in the very limited ability to induce graft-versus-host disease (GvHD) reactions in the allogeneic setting. CIK cells are produced with a simple and extremely efficient expansion protocol, which leads to a massive expansion of effector cells and requires a lower financial commitment compared to CAR-T cells. Indeed, CAR-T manufacturing involves the engineering with expensive GMP-grade viral vectors in centralized manufacturing facilities, whereas CIK cell production is successfully performed in local academic GMP facilities, and CIK cell treatment is now licensed in many countries. Moreover, the toxicities observed for CAR-T cells are not present in CIK cell-treated patients, thus further reducing the costs associated with hospitalization and post-infusion monitoring of patients, and ultimately encouraging the delivery of cell therapies in the outpatient setting. This review aims to give an overview of the limitations of CAR-T cell therapy and outline how the use of CIK cells could overcome such drawbacks thanks to their unique features. We highlight the undeniable advantages of using CIK cells as a therapeutic product, underlying the opportunity for further research on the topic.
Collapse
Affiliation(s)
- Elisa Cappuzzello
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
| | - Emilia Vigolo
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
| | - Giulia D’Accardio
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Giuseppe Astori
- Advanced Cellular Therapy Laboratory, Department of Hematology, San Bortolo Hospital of Vicenza, Vicenza, Italy
| | - Antonio Rosato
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Roberta Sommaggio
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| |
Collapse
|
10
|
Mercogliano MF, Bruni S, Mauro FL, Schillaci R. Emerging Targeted Therapies for HER2-Positive Breast Cancer. Cancers (Basel) 2023; 15:cancers15071987. [PMID: 37046648 PMCID: PMC10093019 DOI: 10.3390/cancers15071987] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/23/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023] Open
Abstract
Breast cancer is the most common cancer in women and the leading cause of death. HER2 overexpression is found in approximately 20% of breast cancers and is associated with a poor prognosis and a shorter overall survival. Tratuzumab, a monoclonal antibody directed against the HER2 receptor, is the standard of care treatment. However, a third of the patients do not respond to therapy. Given the high rate of resistance, other HER2-targeted strategies have been developed, including monoclonal antibodies such as pertuzumab and margetuximab, trastuzumab-based antibody drug conjugates such as trastuzumab-emtansine (T-DM1) and trastuzumab-deruxtecan (T-DXd), and tyrosine kinase inhibitors like lapatinib and tucatinib, among others. Moreover, T-DXd has proven to be of use in the HER2-low subtype, which suggests that other HER2-targeted therapies could be successful in this recently defined new breast cancer subclassification. When patients progress to multiple strategies, there are several HER2-targeted therapies available; however, treatment options are limited, and the potential combination with other drugs, immune checkpoint inhibitors, CAR-T cells, CAR-NK, CAR-M, and vaccines is an interesting and appealing field that is still in development. In this review, we will discuss the highlights and pitfalls of the different HER2-targeted therapies and potential combinations to overcome metastatic disease and resistance to therapy.
Collapse
|