1
|
Mitranovici MI, Caravia LG, Moraru L, Pușcașiu L. Targeting Cancer Stemness Using Nanotechnology in a Holistic Approach: A Narrative Review. Pharmaceutics 2025; 17:277. [PMID: 40142941 PMCID: PMC11945010 DOI: 10.3390/pharmaceutics17030277] [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: 01/02/2025] [Revised: 01/20/2025] [Accepted: 02/18/2025] [Indexed: 03/28/2025] Open
Abstract
Increasing evidence shows that a very small population of cancer stem cells (CSCs) is responsible for cancer recurrence, drug resistance, and metastasis. CSCs usually reside in hypoxic tumor regions and are characterized by high tumorigenicity. Their inaccessible nature allows them to avoid the effects of conventional treatments such as chemotherapy, radiotherapy, and surgery. In addition, conventional chemo- and radiotherapy is potentially toxic and could help CSCs to spread and survive. New therapeutic targets against CSCs are sought, including different signaling pathways and distinct cell surface markers. Recent advances in nanotechnology have provided hope for the development of new therapeutic avenues to eradicate CSCs. In this review, we present newly discovered nanoparticles that can be co-loaded with an apoptosis-inducing agent or differentiation-inducing agent, with high stability, cellular penetration, and drug release. We also summarize the molecular characteristics of CSCs and the signaling pathways responsible for their survival and maintenance. Controlled drug release targeting CSCs aims to reduce stemness-related drug resistance, suppress tumor growth, and prevent tumor relapse and metastases.
Collapse
Affiliation(s)
- Melinda-Ildiko Mitranovici
- Department of Anatomy, Faculty of Medicine, “George Emil Palade” University of Medicine, Pharmacy, Science and Technology, 540142 Targu Mures, Romania;
| | - Laura Georgiana Caravia
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Liviu Moraru
- Department of Anatomy, Faculty of Medicine, “George Emil Palade” University of Medicine, Pharmacy, Science and Technology, 540142 Targu Mures, Romania;
| | - Lucian Pușcașiu
- Department of Anatomy, Faculty of Medicine, “George Emil Palade” University of Medicine, Pharmacy, Science and Technology, 540142 Targu Mures, Romania;
| |
Collapse
|
2
|
Jiang X, Nik Nabil WN, Ze Y, Dai R, Xi Z, Xu H. Unlocking Natural Potential: Antibody-Drug Conjugates With Naturally Derived Payloads for Cancer Therapy. Phytother Res 2025; 39:789-874. [PMID: 39688127 DOI: 10.1002/ptr.8407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 11/06/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024]
Abstract
Natural compound-derived chemotherapies remain central to cancer treatment, however, they often cause off-target side effects that negatively impact patients' quality of life. In contrast, antibody-drug conjugates (ADCs) combine cytotoxic payloads with antibodies to specifically target cancer cells. Most approved and clinically investigated ADCs utilize naturally derived payloads, while those with conventional synthetic molecular payloads remain limited. This review focuses on approved ADCs that enhance the efficacy of naturally derived payloads by linking them with antibodies. We provide an overview of the core components of ADCs, their working mechanisms, and FDA-approved ADCs featuring naturally derived payloads, such as calicheamicin, camptothecin, dolastatin 10, maytansine, pyrrolbenzodiazepine (PBD), and the immunotoxin Pseudomonas exotoxin A. This review also explores recent clinical advancements aimed at broadening the therapeutic potential of ADCs, their applicability in treating heterogeneously composed tumors and their potential use beyond oncology. Additionally, this review highlights naturally derived payloads that are currently being clinically investigated but have not yet received approval. By summarizing the current landscape, this review provides insights into promising avenues for exploration and contributes to the refinement of treatment protocols for improved patient outcomes.
Collapse
Affiliation(s)
- Xue Jiang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, China
| | - Wan Najbah Nik Nabil
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- National Pharmaceutical Regulatory Agency, Ministry of Health, Selangor, Malaysia
| | - Yufei Ze
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, China
| | - Rongchen Dai
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, China
| | - Zhichao Xi
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, China
| | - Hongxi Xu
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
3
|
Fasih S, Welch S, Lohmann AE. Antibody-Drug Conjugates: A Start of a New Era in Gynecological Cancers. Curr Oncol 2024; 31:7088-7106. [PMID: 39590153 PMCID: PMC11593302 DOI: 10.3390/curroncol31110522] [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: 08/21/2024] [Revised: 10/30/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
Antibody-drug conjugates (ADCs) are a new class of therapeutic agents designed to target specific antigens on tumor cells, combining the specificity of monoclonal antibodies with the cytotoxicity of chemotherapy agents. ADCs have been available for over a decade, but in gynecological cancers, these agents are relatively new with great promise ahead. More than 80% of ongoing trials in gynecological cancers are evaluating ADCs' safety and efficacy, of which 40% are early-phase trials. Around twenty ADCs are currently under investigation, either alone or in combination with chemotherapies or immune checkpoint inhibitors. Among them, mirvetuximab soravtansine has been recently approved by the Food and Drug Administration (FDA) in platinum-resistant ovarian cancer with high folate-α receptor expression, as a single agent or in combination. Tisotumab vedotin and trastuzumab deruxtecan are also now approved by the FDA in patients with pre-treated cervical and uterine cancers and further investigation is ongoing. Overall, the toxicity profiles of ADCs are acceptable. Ocular toxicity is one of the specific side effects of some ADCs, but most of the cases are manageable with the use of prophylactic steroids and dose adjustments. This review aims to provide an overview of the fundamental and operational features of ADCs and examine the latest and most promising data, with a particular focus on the Canadian viewpoint.
Collapse
Affiliation(s)
- Samir Fasih
- Department of Oncology, Division of Medical Oncology, University of Western Ontario, London, ON N6A 5W9, Canada; (S.F.); (S.W.)
| | - Stephen Welch
- Department of Oncology, Division of Medical Oncology, University of Western Ontario, London, ON N6A 5W9, Canada; (S.F.); (S.W.)
| | - Ana Elisa Lohmann
- Department of Oncology, Division of Medical Oncology, University of Western Ontario, London, ON N6A 5W9, Canada; (S.F.); (S.W.)
- Department of Epidemiology and Biostatistics, University of Western Ontario, London, ON N6A 5W9, Canada
| |
Collapse
|
4
|
Parit S, Manchare A, Gholap AD, Mundhe P, Hatvate N, Rojekar S, Patravale V. Antibody-Drug Conjugates: A promising breakthrough in cancer therapy. Int J Pharm 2024; 659:124211. [PMID: 38750981 DOI: 10.1016/j.ijpharm.2024.124211] [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: 01/09/2024] [Revised: 04/30/2024] [Accepted: 05/06/2024] [Indexed: 06/03/2024]
Abstract
Antibody-drug conjugates (ADCs) provide effective cancer treatment through the selective delivery of cytotoxic payloads to the cancer cells. They offer unparalleled precision and specificity in directing drugs to cancer cells while minimizing off-target effects. Despite several advantages, there is a requirement for innovations in the molecular design of ADC owing to drug resistance, cancer heterogeneity along the adverse effects of treatment. The review critically analyses ADC function mechanisms, unraveling the intricate interplay between antibodies, linkers, and payloads in facilitating targeted drug delivery to cancer cells. The article also highlights notable advancements in antibody engineering, which aid in creating highly selective and potent ADCs. Additionally, the review details significant progress in clinical ADC development with an in-depth examination of pivotal trials and approved formulations. Antibody Drug Conjugates (ADCs) are a ground-breaking approach to targeted drug delivery, especially in cancer treatment. They offer unparalleled precision and specificity in directing drugs to cancer cells while minimizing off-target effects. This review provides a comprehensive examination of the current state of ADC development, covering their design, mechanisms of action, and clinical applications. The article emphasizes the need for greater precision in drug delivery and explains why ADCs are necessary.
Collapse
Affiliation(s)
- Swapnali Parit
- Institute of Chemical Technology, Marathwada Campus, Jalna 431203, Maharashtra, India
| | - Ajit Manchare
- Institute of Chemical Technology, Marathwada Campus, Jalna 431203, Maharashtra, India
| | - Amol D Gholap
- Department of Pharmaceutics, St. John Institute of Pharmacy and Research, Palghar 401404, Maharashtra, India
| | - Prashant Mundhe
- Institute of Chemical Technology, Marathwada Campus, Jalna 431203, Maharashtra, India
| | - Navnath Hatvate
- Institute of Chemical Technology, Marathwada Campus, Jalna 431203, Maharashtra, India
| | - Satish Rojekar
- Institute of Chemical Technology, Marathwada Campus, Jalna 431203, Maharashtra, India; Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Vandana Patravale
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai 400019, India.
| |
Collapse
|
5
|
Zhai C, Cui Y, Guo L, Chen C, Song Y, Zhong J, Wang Y. Progress in the study of antibody-drug conjugates for the treatment of cervical cancer. Front Oncol 2024; 14:1395784. [PMID: 38903711 PMCID: PMC11187480 DOI: 10.3389/fonc.2024.1395784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 05/21/2024] [Indexed: 06/22/2024] Open
Abstract
Cervical cancer is the second most prevalent malignancy affecting women's health globally, and the number of morbidity and mortality from cervical cancer continues to rise worldwide. The 5-year survival rate of patients with recurrent or metastatic cervical cancer is significantly reduced, and existing treatment modalities have low efficacy and high adverse effects, so there is a strong need for new, effective, and well-tolerated therapies. Antibody-drug conjugates (ADCs) are a new targeted therapeutic modality that can efficiently kill tumor cells. This review aims to summarize the composition, research, and development history and mechanism of action of ADCs, to review the research progress of ADCs in the treatment of cervical cancer, and to summarize and prospect the application of ADCs.
Collapse
Affiliation(s)
- Congcong Zhai
- Department of Oncology, Gannan Medical University, Ganzhou, China
- Department of Oncology, First Affiliated Hospital, Gannan Medical University, Ganzhou, China
| | - Yan Cui
- Department of Oncology, Bengbu Medical University, Lu’an, China
| | - Ling Guo
- Department of Oncology, Gannan Medical University, Ganzhou, China
| | - Cixiang Chen
- Department of Oncology, Gannan Medical University, Ganzhou, China
| | - Yanfang Song
- Department of Oncology, First Affiliated Hospital, Gannan Medical University, Ganzhou, China
| | - Jinghua Zhong
- Department of Oncology, First Affiliated Hospital, Gannan Medical University, Ganzhou, China
- Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
| | - Yili Wang
- Department of Oncology, First Affiliated Hospital, Gannan Medical University, Ganzhou, China
| |
Collapse
|
6
|
Nwabufo CK. Mirvetuximab soravtansine in ovarian cancer therapy: expert opinion on pharmacological considerations. Cancer Chemother Pharmacol 2024; 93:89-105. [PMID: 37594572 DOI: 10.1007/s00280-023-04575-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 08/03/2023] [Indexed: 08/19/2023]
Abstract
ImmunoGen developed mirvetuximab soravtansine as an antibody-drug conjugate comprising of a humanized anti-folate receptor-α (FRα) monoclonal antibody of IgG1k subtype, a cleavable linker, and a cytotoxic payload, DM4. Mirvetuximab soravtansine was granted accelerated approval by the US FDA on November 14, 2022, for the treatment of adult patients with FRα positive, platinum-resistant epithelial ovarian, fallopian tube or primary peritoneal cancer who have received 1-3 prior systemic treatment regimens. The approval of mirvetuximab soravtansine represents a breakthrough for addressing the unmet medical needs of ovarian cancer, especially for up to 80% of patients who relapse and become resistant to platinum-based chemotherapy, resulting in poor prognosis and limited treatment options. However, it is my impression that addressing several pharmacological factors could improve the safety and efficacy of mirvetuximab soravtansine. This article summarizes the current pharmacological profile of mirvetuximab soravtansine and provides an expert opinion on pharmacological strategies for optimizing its safety and efficacy profile for the treatment of platinum-resistant ovarian cancer.
Collapse
Affiliation(s)
- Chukwunonso K Nwabufo
- OneDrug, Toronto, ON, Canada.
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, ON, M5S 3M2, Canada.
- Program in Translational Medicine, Hospital for Sick Children, Toronto, ON, Canada.
| |
Collapse
|
7
|
Wang Y, Liu L, Jin X, Yu Y. Efficacy and safety of mirvetuximab soravtansine in recurrent ovarian cancer with FRa positive expression: A systematic review and meta-analysis. Crit Rev Oncol Hematol 2024; 194:104230. [PMID: 38122916 DOI: 10.1016/j.critrevonc.2023.104230] [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: 09/20/2023] [Revised: 11/18/2023] [Accepted: 12/05/2023] [Indexed: 12/23/2023] Open
Abstract
OBJECTIVE To evaluate the efficacy and safety of mirvetuximab soravtansine in treating recurrent ovarian cancer with folate receptor alpha (FRa) expression. METHODS A comprehensive search was conducted on online databases, including PubMed, Cochrane Library, and EMBASE, to identify relevant literature about the efficacy and safety of mirvetuximab soravtansine in recurrent ovarian cancer with FRa-positive expression. The keywords were the following: recurrent ovarian cancer, mirvetuximab soravtansine, FRa, and antibody-drug conjugate. Furthermore, studies that satisfied the necessary qualifications were carefully evaluated for further meta-analysis. RESULTS This meta-analysis involved the examination of seven trials with a total of 631 patients. According to the pooled data, the objective response rate (ORR) was 36% (95%CI: 27%-45%). Similarly, the disease control rate (DCR) was 88% (95% CI: 84-91%). Furthermore, the median progression-free survival (PFS) was determined to be 6.1 months (95% CI: 4.27-7.47). The overall response rate and PFS for platinum-resistant ovarian cancer were found to be 29% (95% CI: 25-32%) and 6.26 months (95% CI: 4.67-7.85), respectively. The most often observed adverse events (AEs) in patients with recurrent ovarian cancer (OC) receiving mirvetuximab soravtansine were blurred vision (all grades: 45%, Grade III: 2%), nausea (all grades: 42%, Grade III: 1%), and diarrhea (all grades: 42%, Grade III: 2%). These AEs were specifically associated with the safety profile of mirvetuximab soravtansine in this patient population. CONCLUSION The efficacy of mirvetuximab soravtansine in treating recurrent ovarian cancer with FRa-positive expression is satisfactory, and the safety is tolerable.
Collapse
Affiliation(s)
- Yicong Wang
- Department of Obstetrics and Gynecology,Central Hospital of Dalian University of Technology, Dalian, China
| | - Lifeng Liu
- Department of Obstetrics and Gynecology,Central Hospital of Dalian University of Technology, Dalian, China
| | - Xianyu Jin
- Department of Obstetrics and Gynecology,Central Hospital of Dalian University of Technology, Dalian, China
| | - Yongai Yu
- Department of Obstetrics and Gynecology,Central Hospital of Dalian University of Technology, Dalian, China.
| |
Collapse
|
8
|
Jaffry M, Choudhry H, Aftab OM, Dastjerdi MH. Antibody-Drug Conjugates and Ocular Toxicity. J Ocul Pharmacol Ther 2023; 39:675-691. [PMID: 37615544 DOI: 10.1089/jop.2023.0069] [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: 08/25/2023] Open
Abstract
Antibody-drug conjugates (ADCs) are a growing class of chemotherapeutic agents for the purpose of treating cancers that often have relapsed or failed first- and second-line treatments. ADCs are composed of extremely potent cytotoxins with a variety of side effects, one of the most significant being ocular toxicity. The available literature describes these toxicities as varying in severity and in incidence, although with disparate methods of evaluation and management. Some of the most common toxicities include microcyst-like epithelial keratopathy and dry eye. We discuss proposed mechanisms of ocular toxicity and describe the reports that mention these toxicities. We focus on ADCs with the most published literature and the most significant effects on ocular tissue. We propose areas for further investigation and possible ideas of future management. We provide a comprehensive look at the reports of ADCs in current literature to better inform clinicians on an expanding drug class.
Collapse
Affiliation(s)
- Mustafa Jaffry
- Department of Ophthalmology and Visual Science, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Hassaam Choudhry
- Department of Ophthalmology and Visual Science, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Owais M Aftab
- Department of Ophthalmology and Visual Science, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Mohammad H Dastjerdi
- Department of Ophthalmology and Visual Science, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| |
Collapse
|
9
|
Sasso J, Tenchov R, Bird R, Iyer KA, Ralhan K, Rodriguez Y, Zhou QA. The Evolving Landscape of Antibody-Drug Conjugates: In Depth Analysis of Recent Research Progress. Bioconjug Chem 2023; 34:1951-2000. [PMID: 37821099 PMCID: PMC10655051 DOI: 10.1021/acs.bioconjchem.3c00374] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/27/2023] [Indexed: 10/13/2023]
Abstract
Antibody-drug conjugates (ADCs) are targeted immunoconjugate constructs that integrate the potency of cytotoxic drugs with the selectivity of monoclonal antibodies, minimizing damage to healthy cells and reducing systemic toxicity. Their design allows for higher doses of the cytotoxic drug to be administered, potentially increasing efficacy. They are currently among the most promising drug classes in oncology, with efforts to expand their application for nononcological indications and in combination therapies. Here we provide a detailed overview of the recent advances in ADC research and consider future directions and challenges in promoting this promising platform to widespread therapeutic use. We examine data from the CAS Content Collection, the largest human-curated collection of published scientific information, and analyze the publication landscape of recent research to reveal the exploration trends in published documents and to provide insights into the scientific advances in the area. We also discuss the evolution of the key concepts in the field, the major technologies, and their development pipelines with company research focuses, disease targets, development stages, and publication and investment trends. A comprehensive concept map has been created based on the documents in the CAS Content Collection. We hope that this report can serve as a useful resource for understanding the current state of knowledge in the field of ADCs and the remaining challenges to fulfill their potential.
Collapse
Affiliation(s)
- Janet
M. Sasso
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Rumiana Tenchov
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Robert Bird
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | | | | | - Yacidzohara Rodriguez
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | | |
Collapse
|
10
|
Medina-Gutiérrez E, García-León A, Gallardo A, Álamo P, Alba-Castellón L, Unzueta U, Villaverde A, Vázquez E, Casanova I, Mangues R. Potent Anticancer Activity of CXCR4-Targeted Nanostructured Toxins in Aggressive Endometrial Cancer Models. Cancers (Basel) 2022; 15:cancers15010085. [PMID: 36612081 PMCID: PMC9818013 DOI: 10.3390/cancers15010085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 12/20/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022] Open
Abstract
Patients with advanced endometrial cancer (EC) show poor outcomes. Thus, the development of new therapeutic approaches to prevent metastasis development in high-risk patients is an unmet need. CXCR4 is overexpressed in EC tumor tissue, epitomizing an unexploited therapeutic target for this malignancy. The in vitro antitumor activity of two CXCR4-targeted nanoparticles, including either the C. diphtheriae (T22-DITOX-H6) or P. aeruginosa (T22-PE24-H6) toxin, was evaluated using viability assays. Apoptotic activation was assessed by DAPI and caspase-3 and PARP cleavage in cell blocks. Both nanotoxins were repeatedly administrated to a subcutaneous EC mouse model, whereas T22-DITOX-H6 was also used in a highly metastatic EC orthotopic model. Tumor burden was assessed through bioluminescence, while metastatic foci and toxicity were studied using histological or immunohistochemical analysis. We found that both nanotoxins exerted a potent antitumor effect both in vitro and in vivo via apoptosis and extended the survival of nanotoxin-treated mice without inducing any off-target toxicity. Repeated T22-DITOX-H6 administration in the metastatic model induced a dramatic reduction in tumor burden while significantly blocking peritoneal, lung and liver metastasis without systemic toxicity. Both nanotoxins, but especially T22-DITOX-H6, represent a promising therapeutic alternative for EC patients that have a dismal prognosis and lack effective therapies.
Collapse
Affiliation(s)
- Esperanza Medina-Gutiérrez
- Oncogenesis and Antitumor Drugs Group, Institut d’Investigació Biomèdica Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain
- Oncogenesis and Antitumor Drugs Group, Institut de Recerca Contra la Leucèmia Josep Carreras, 08025 Barcelona, Spain
| | - Annabel García-León
- Oncogenesis and Antitumor Drugs Group, Institut d’Investigació Biomèdica Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain
- Oncogenesis and Antitumor Drugs Group, Institut de Recerca Contra la Leucèmia Josep Carreras, 08025 Barcelona, Spain
| | - Alberto Gallardo
- Oncogenesis and Antitumor Drugs Group, Institut d’Investigació Biomèdica Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain
- Department of Pathology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain
| | - Patricia Álamo
- Oncogenesis and Antitumor Drugs Group, Institut d’Investigació Biomèdica Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain
- Oncogenesis and Antitumor Drugs Group, Institut de Recerca Contra la Leucèmia Josep Carreras, 08025 Barcelona, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
| | - Lorena Alba-Castellón
- Oncogenesis and Antitumor Drugs Group, Institut d’Investigació Biomèdica Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain
- Oncogenesis and Antitumor Drugs Group, Institut de Recerca Contra la Leucèmia Josep Carreras, 08025 Barcelona, Spain
| | - Ugutz Unzueta
- Oncogenesis and Antitumor Drugs Group, Institut d’Investigació Biomèdica Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain
- Oncogenesis and Antitumor Drugs Group, Institut de Recerca Contra la Leucèmia Josep Carreras, 08025 Barcelona, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Antonio Villaverde
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Esther Vázquez
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Isolda Casanova
- Oncogenesis and Antitumor Drugs Group, Institut d’Investigació Biomèdica Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain
- Oncogenesis and Antitumor Drugs Group, Institut de Recerca Contra la Leucèmia Josep Carreras, 08025 Barcelona, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
- Correspondence: (I.C.); (R.M.)
| | - Ramon Mangues
- Oncogenesis and Antitumor Drugs Group, Institut d’Investigació Biomèdica Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain
- Oncogenesis and Antitumor Drugs Group, Institut de Recerca Contra la Leucèmia Josep Carreras, 08025 Barcelona, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
- Correspondence: (I.C.); (R.M.)
| |
Collapse
|
11
|
Qi X, Li Y, Liu W, Wang Y, Chen Z, Lin L. Research Trend of Publications Concerning Antibody-Drug Conjugate in Solid Cancer: A Bibliometric Study. Front Pharmacol 2022; 13:921385. [PMID: 35795565 PMCID: PMC9252465 DOI: 10.3389/fphar.2022.921385] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 06/01/2022] [Indexed: 11/22/2022] Open
Abstract
Background: Antibody-drug conjugate (ADC) is a promising therapy for solid cancer that has raised global concern. Although several papers have reviewed the current state of ADCs in different solid cancers, a quantitative analysis of the publications in this field is scarce. Methods: Publications related to ADC in the field of solid cancer were obtained from the Web of Science Core Collection. Data analyses were performed with VOSviewer 1.6.9, HistCite 2.1, CiteSpace V and R package Bibliometrix. Results: A total of 3,482 records were obtained in the holistic field and 1,197 in the clinical field. Steady growth in the number of publications was observed. The United States was the leading contributor in this field. Krop IE was the most influential author. The most productive institution was Genentech Inc., while Mem Sloan Kettering Canc Ctr was the most cited one. The most impactful journal was the Journal of Clinical Oncology. A total of 37 burst references and five burst references were identified between 2017–2022 in the holistic and clinical fields, respectively. Keywords analysis indicated that ADCs research mainly involved breast cancer, triple-negative breast cancer, ovarian cancer, small cell lung cancer, prostate cancer, gastric cancer, and urothelial carcinoma. ADC agents including trastuzumab emtansine, trastuzumab deruxtecan, sacituzumab govitecan, enfortumab vedotin, and rovalpituzumab tesirine were highly studied. Targets including HER2, trophoblast cell-surface antigen, mesothelin, delta-like ligand 3, and nectin-4 were the major concerns. Conclusion: This study analyzed publications concerning ADCs in the field of solid cancer with bibliometric analysis. Further clinical trials of ADCs and designs of the next generation of ADCs are the current focuses of the field. Acquired resistance of ADCs and biomarkers for ADC therapy efficacy monitoring are future concerns.
Collapse
Affiliation(s)
- Xiangjun Qi
- The First Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yanlong Li
- The First Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wei Liu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yifan Wang
- School of Chinese Classics Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhuangzhong Chen
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lizhu Lin
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- *Correspondence: Lizhu Lin,
| |
Collapse
|
12
|
Actively Targeted Nanomedicines in Breast Cancer: From Pre-Clinal Investigation to Clinic. Cancers (Basel) 2022; 14:cancers14051198. [PMID: 35267507 PMCID: PMC8909490 DOI: 10.3390/cancers14051198] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/14/2022] [Accepted: 02/22/2022] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Despite all the efforts and advances made in the treatment of breast cancer, this pathology continues to be one of the main causes of cancer death in women, particularly triple-negative breast cancer (TNBC), and, although to a lesser degree, HER-2 receptor-positive tumors. Chemotherapy is one of the main treatments available. However, it shows numerous limitations due to its lack of selectivity. In this sense, the selective delivery of antineoplastics to cancer cells can reduce their adverse effects and increase their efficacy. The use of active targeted nanomedicine is a good strategy to achieve this selective chemotherapy. In fact, in recent decades, several active targeted nanoformulations have been approved or reached clinical investigation with excellent results. Among all nanomedicines, antibody-drug conjugates are the most promising. Abstract Breast cancer is one of the most frequently diagnosed tumors and the second leading cause of cancer death in women worldwide. The use of nanosystems specifically targeted to tumor cells (active targeting) can be an excellent therapeutic tool to improve and optimize current chemotherapy for this type of neoplasm, since they make it possible to reduce the toxicity and, in some cases, increase the efficacy of antineoplastic drugs. Currently, there are 14 nanomedicines that have reached the clinic for the treatment of breast cancer, 4 of which are already approved (Kadcyla®, Enhertu®, Trodelvy®, and Abraxane®). Most of these nanomedicines are antibody–drug conjugates. In the case of HER-2-positive breast cancer, these conjugates (Kadcyla®, Enhertu®, Trastuzumab-duocarmycin, RC48, and HT19-MMAF) target HER-2 receptors, and incorporate maytansinoid, deruxtecan, duocarmicyn, or auristatins as antineoplastics. In TNBC these conjugates (Trodelvy®, Glembatumumab-Vedotin, Ladiratuzumab-vedotin, Cofetuzumab-pelidotin, and PF-06647263) are directed against various targets, in particular Trop-2 glycoprotein, NMB glycoprotein, Zinc transporter LIV-1, and Ephrin receptor-4, to achieve this selective accumulation, and include campthotecins, calicheamins, or auristatins as drugs. Apart from the antibody–drug conjugates, there are other active targeted nanosystems that have reached the clinic for the treatment of these tumors such as Abraxane® and Nab-rapamicyn (albumin nanoparticles entrapping placlitaxel and rapamycin respectively) and various liposomes (MM-302, C225-ILS-Dox, and MM-310) loaded with doxorubicin or docetaxel and coated with ligands targeted to Ephrin A2, EPGF, or HER-2 receptors. In this work, all these active targeted nanomedicines are discussed, analyzing their advantages and disadvantages over conventional chemotherapy as well as the challenges involved in their lab to clinical translation. In addition, examples of formulations developed and evaluated at the preclinical level are also discussed.
Collapse
|