1
|
Hushmandi K, Einollahi B, Lee EHC, Sakaizawa R, Glaviano A, Reiter RJ, Saadat SH, Farani MR, Huh YS, Aref AR, Salimimoghadam S, Kumar AP. Bispecific antibodies as powerful immunotherapeutic agents for urological cancers: Recent innovations based on preclinical and clinical evidence. Int J Biol Sci 2025; 21:1410-1435. [PMID: 39990653 PMCID: PMC11844292 DOI: 10.7150/ijbs.96155] [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: 03/10/2024] [Accepted: 08/25/2024] [Indexed: 02/25/2025] Open
Abstract
Conventional immunotherapy has emerged as a key option for cancer treatment. However, its efficacy has been limited in urological cancers, especially prostate cancer, because of the immunosuppressive tumor microenvironment (TME), difficulty in drug delivery, aberrant immune response, and damage to normal cells. Bispecific antibodies (BsAbs) are engineered proteins with two different antigen-binding domains, designed using different technologies and in various formats. BsAb-based tumor immunotherapy has yielded optimistic results in preclinical and clinical investigations of many tumor types, including urological cancers. However, a series of challenges, including tumor heterogeneity, TME, Ab immunogenicity, adverse effects, serum half-life, low response rates, and drug resistance, hamper the application of BsAbs. In this review, we provide insights into the most common BsAb platforms with different mechanisms of action, which are under preclinical and clinical research, along with ways to overcome the challenges in BsAb administration for treating urological cancer.
Collapse
Affiliation(s)
- Kiavash Hushmandi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Behzad Einollahi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - E Hui Clarissa Lee
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Reo Sakaizawa
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Antonino Glaviano
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123 Palermo, Italy
| | - Russel J. Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, Long School of Medicine, San Antonio, Texas USA
| | - Seyed Hassan Saadat
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Marzieh Ramezani Farani
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Incheon 22212, Republic of Korea
| | - Yun Suk Huh
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Incheon 22212, Republic of Korea
| | - Amir Reza Aref
- Department of Vitro Vision, DeepkinetiX Inc., Boston, MA, USA
| | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
2
|
Massariol Pimenta T, Carlos de Souza J, da Silva Martins B, Silva Butzene SM, Simões Padilha JM, Ganho Marçal M, Dos Santos Elias G, Rangel LBA. Emerging strategies to overcome ovarian cancer: advances in immunotherapy. Front Pharmacol 2024; 15:1490896. [PMID: 39564107 PMCID: PMC11573523 DOI: 10.3389/fphar.2024.1490896] [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: 09/03/2024] [Accepted: 10/21/2024] [Indexed: 11/21/2024] Open
Abstract
Ovarian cancer is the second most common malignant neoplasm of gynecological origin and the leading cause of death from cancer in the female reproductive system worldwide. This scenario is largely due to late diagnoses, often in advanced stages, and the development of chemoresistance by cancer cells. These challenges highlight the need for alternative treatments, with immunotherapy being a promising option. Cancer immunotherapy involves triggering an anti-tumor immune response and developing immunological memory to eliminate malignant cells, prevent recurrence, and inhibit metastasis. Some ongoing research investigate potentially immunological advancements in the field of cancer vaccines, immune checkpoint blockade, CAR-T cell, and other strategies.
Collapse
Affiliation(s)
- Tatiana Massariol Pimenta
- Department of Pharmaceutical Sciences, Health Sciences Center, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Josiany Carlos de Souza
- Biotechnology Program/RENORBIO, Health Sciences Center, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Bárbara da Silva Martins
- Department of Pharmaceutical Sciences, Health Sciences Center, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Solenny Maria Silva Butzene
- Department of Pharmaceutical Sciences, Health Sciences Center, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | - José Matheus Simões Padilha
- Department of Pharmaceutical Sciences, Health Sciences Center, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Milleny Ganho Marçal
- Department of Pharmaceutical Sciences, Health Sciences Center, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Guilherme Dos Santos Elias
- Biochemistry Program, Health Sciences Center, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Leticia Batista Azevedo Rangel
- Department of Pharmaceutical Sciences, Health Sciences Center, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
- Biotechnology Program/RENORBIO, Health Sciences Center, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
- Biochemistry Program, Health Sciences Center, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| |
Collapse
|
3
|
Chelushkin MA, van Dorp J, van Wilpe S, Seignette IM, Mellema JJJ, Alkemade M, Gil-Jimenez A, Peters D, Brugman W, Stockem CF, Hooijberg E, Broeks A, van Rhijn BWG, Mertens LS, van der Heijden AG, Mehra N, van Montfoort ML, Wessels LFA, Vis DJ, van der Heijden MS. Platinum-Based Chemotherapy Induces Opposing Effects on Immunotherapy Response-Related Spatial and Stromal Biomarkers in the Bladder Cancer Microenvironment. Clin Cancer Res 2024; 30:4227-4239. [PMID: 39047168 DOI: 10.1158/1078-0432.ccr-24-0724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/08/2024] [Accepted: 07/23/2024] [Indexed: 07/27/2024]
Abstract
PURPOSE Platinum-based chemotherapy and immune checkpoint inhibitors are key components of systemic treatment for muscle-invasive and advanced urothelial cancer. The ideal integration of these two treatment modalities remains unclear as clinical trials have led to inconsistent results. Modulation of the tumor-immune microenvironment by chemotherapy is poorly characterized. We aimed to investigate this modulation, focusing on potential clinical implications for immune checkpoint inhibitor response. EXPERIMENTAL DESIGN We assessed immune cell densities, spatial relations, and tumor/stromal components from 116 patients with urothelial bladder cancer (paired data for 95 patients) before and after platinum-based chemotherapy. RESULTS Several published biomarkers for immunotherapy response changed upon chemotherapy treatment. The intratumoral CD8+ T-cell percentage increased after treatment and was associated with increased TNFα-via-NF-κB signaling. The percentage of PDL1+ immune cells was higher after chemotherapy. An increase in chemo-induced changes that potentially inhibit an antitumor immune response was also observed, including increased fibroblast-based TGFβ signaling and distances from immune cells to the nearest cancer cell. The latter two parameters correlated significantly in posttreatment samples, suggesting that TGFβ signaling in fibroblasts may play a role in spatially separating immune cells from cancer cells. We examined specific chemotherapy regimens and found that treatment with methotrexate, vinblastine, doxorubicin, and cisplatin was associated with an increase in the macrophage cell percentage. Gemcitabine-containing chemotherapy was associated with upregulation of fibroblast TGFβ signaling. CONCLUSIONS The opposing effects of platinum-based chemotherapy on the immune cell composition and stromal context of the tumor-immune microenvironment may explain the inconsistent results of clinical trials investigating chemotherapy and immune checkpoint inhibitor combinations in bladder cancer.
Collapse
Affiliation(s)
- Maksim A Chelushkin
- Department of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
| | - Jeroen van Dorp
- Department of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Sandra van Wilpe
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Iris M Seignette
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Jan-Jaap J Mellema
- Department of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Maartje Alkemade
- Core Facility Molecular Pathology & Biobanking, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Alberto Gil-Jimenez
- Department of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
| | - Dennis Peters
- Core Facility Molecular Pathology & Biobanking, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Wim Brugman
- Genomics Core Facility, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Chantal F Stockem
- Department of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Erik Hooijberg
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Annegien Broeks
- Core Facility Molecular Pathology & Biobanking, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Bas W G van Rhijn
- Department of Urology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Urology, Caritas St. Josef Medical Center, University of Regensburg, Regensburg, Germany
| | - Laura S Mertens
- Department of Urology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | | - Niven Mehra
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | | | - Lodewyk F A Wessels
- Department of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
- Faculty of Electrical Engineering, Mathematics and Computer Science, Delft University of Technology, Delft, the Netherlands
| | - Daniel J Vis
- Department of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
| | - Michiel S van der Heijden
- Department of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| |
Collapse
|
4
|
Qu F, Darji S, Thompson DH. Recent Advances in Drug Delivery Strategies for High-Risk BCG-Unresponsive Non-Muscle Invasive Bladder Cancer: A Brief Review from 2018 to 2024. Pharmaceutics 2024; 16:1154. [PMID: 39339191 PMCID: PMC11434993 DOI: 10.3390/pharmaceutics16091154] [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: 07/30/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/30/2024] Open
Abstract
High-risk BCG-unresponsive non-muscle invasive bladder cancer (NMIBC) is a condition that is typically treated with Bacillus Calmette-Guérin (BCG) therapy. Unfortunately, NMIBC is characterized by high recurrence, with a significant percentage of BCG patients ultimately requiring radical cystectomy. As a consequence, the development of effective new therapies to avoid RC has become a rapidly evolving field to address this unmet clinical need. To date, three biologics-Keytruda, Adstiladrin, and Anktiva-have been approved by the FDA, and multiple drug modalities, particularly gene therapies, have shown promising results in clinical trials. Advances in drug delivery strategies, such as targeted delivery, sustained release, and permeabilization of protective layers, are critical in overcoming the challenges posed by therapeutic intervention in bladder cancer. This review focuses on high-risk BCG-unresponsive NMIBC therapies that have been or are currently being investigated in clinical trials, offering a broad overview of the delivery system designs and up-to-date clinical outcomes that have been reported as of July 2024. It aims to inform the development of future drug delivery systems for second-line therapies in high-risk BCG-unresponsive NMIBC.
Collapse
Affiliation(s)
- Feng Qu
- Bindley Bioscience Center, Department of Chemistry, Multi-Disciplinary Cancer Research Facility, Purdue University, West Lafayette, IN 47907, USA
| | - Saloni Darji
- Bindley Bioscience Center, Department of Chemistry, Multi-Disciplinary Cancer Research Facility, Purdue University, West Lafayette, IN 47907, USA
| | - David H Thompson
- Bindley Bioscience Center, Department of Chemistry, Multi-Disciplinary Cancer Research Facility, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
5
|
Yi SY, Wei MZ, Zhao L. Targeted immunotherapy to cancer stem cells: A novel strategy of anticancer immunotherapy. Crit Rev Oncol Hematol 2024; 196:104313. [PMID: 38428702 DOI: 10.1016/j.critrevonc.2024.104313] [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: 10/14/2023] [Revised: 02/04/2024] [Accepted: 02/26/2024] [Indexed: 03/03/2024] Open
Abstract
Cancer is a major disease that endangers human health. Cancer drug resistance and relapse are the two main causes contributing to cancer treatment failure. Cancer stem cells (CSCs) are a small fraction of tumor cells that are responsible for tumorigenesis, metastasis, relapse, and resistance to conventional anticancer therapies. Therefore, CSCs are considered to be the root of cancer recurrence, metastasis, and drug resistance. Novel anticancer strategies need to face this new challenge and explore their efficacy against CSCs. Recently, immunotherapy has made rapid advances in cancer treatment, and its potential against CSCs is also an interesting area of research. Meanwhile, immunotherapy strategies are novel therapeutic modalities with promising results in targeting CSCs. In this review, we summarize the targeting of CSCs by various immunotherapy strategies such as monoclonal antibodies(mAb), tumor vaccines, immune checkpoint inhibitors, and chimeric antigen receptor-T cells(CAR-T) in pre-clinical and clinical studies. This review provides new insights into the application of these immunotherapeutic approaches to potential anti-tumor therapies in the future.
Collapse
Affiliation(s)
- Shan-Yong Yi
- Department of Oncology of the Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zheng Zhou, Henan Province 450007, China.
| | - Mei-Zhuo Wei
- Department of Oncology of the Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zheng Zhou, Henan Province 450007, China
| | - Ling Zhao
- Department of Oncology of the Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zheng Zhou, Henan Province 450007, China.
| |
Collapse
|
6
|
Hannouneh ZA, Hijazi A, Alsaleem AA, Hami S, Kheyrbek N, Tanous F, Khaddour K, Abbas A, Alshehabi Z. Novel immunotherapeutic options for BCG-unresponsive high-risk non-muscle-invasive bladder cancer. Cancer Med 2023; 12:21944-21968. [PMID: 38037752 PMCID: PMC10757155 DOI: 10.1002/cam4.6768] [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: 09/23/2023] [Revised: 11/10/2023] [Accepted: 11/17/2023] [Indexed: 12/02/2023] Open
Abstract
BACKGROUND High-risk non-muscle-invasive bladder cancer (HR-NMIBC) presents a challenge to many physicians due to its ability to resist Bacillus Calmette-Guérin (BCG) intravesical therapy and the substantial rate of progression into muscle-invasive bladder cancer (MIBC). Patients who are BCG-unresponsive have worse prognosis and thus require further management including radical cystectomy (RC), which significantly impacts quality of life. Moreover, the ongoing worldwide shortage of BCG warrants the need for policies that prioritize drug use and utilize alternative treatment strategies. Hence, there is a significant unmet need for bladder preserving therapy in this subset of patients. METHODS To address this issue, we searched the relevant literature in PUBMED for articles published from 2019 through May of 2023 using appropriate keywords. All clinical trials of patients with HR-NMIBC treated with immune-related agents were retrieved from clinicaltrials.gov. FINDINGS AND FUTURE PERSPECTIVES Exploratory treatments for BCG-Unresponsive HR-NMIBC included immune checkpoint inhibitors (ICI), oncolytic viral therapy, cytokine agonists, and other immunomodulators targeting TLR, EpCaM, FGFR, MetAP2, and IDO1. Some combination therapies have been found to work synergistically and are preferred therapeutically over monotherapy. Three drugs-pembrolizumab, valrubicin, and most recently, nadofaragene firadenovec-vncg-have been FDA approved for the treatment of BCG-unresponsive NMIBC in patients who are ineligible for or decline RC. However, all explored treatment options tend to postpone RC rather than provide long-term disease control. Additional combination strategies need to be studied to enhance the effects of immunotherapy. Despite the challenges faced in finding effective therapies, many potential treatments are currently under investigation. Addressing the landscape of biomarkers, mechanisms of progression, BCG resistance, and trial design challenges in HR-NMIBC is essential for the discovery of new targets and the development of effective treatments.
Collapse
Affiliation(s)
- Zein Alabdin Hannouneh
- Faculty of MedicineAl Andalus University for Medical SciencesTartusSyrian Arab Republic
- Cancer Research CenterTishreen UniversityLattakiaSyrian Arab Republic
| | - Amjad Hijazi
- Faculty of MedicineAl Andalus University for Medical SciencesTartusSyrian Arab Republic
- Cancer Research CenterTishreen UniversityLattakiaSyrian Arab Republic
| | - Alaa Aldeen Alsaleem
- Faculty of MedicineAl Andalus University for Medical SciencesTartusSyrian Arab Republic
- Cancer Research CenterTishreen UniversityLattakiaSyrian Arab Republic
| | - Siwan Hami
- Faculty of MedicineAl Andalus University for Medical SciencesTartusSyrian Arab Republic
- Cancer Research CenterTishreen UniversityLattakiaSyrian Arab Republic
| | - Nina Kheyrbek
- Cancer Research CenterTishreen UniversityLattakiaSyrian Arab Republic
- Faculty of MedicineTishreen UniversityLattakiaSyrian Arab Republic
| | - Fadi Tanous
- Cancer Research CenterTishreen UniversityLattakiaSyrian Arab Republic
- Faculty of MedicineAl‐Baath UniversityHomsSyrian Arab Republic
| | - Karam Khaddour
- Department of Medical OncologyDana‐Farber Cancer Institute, Harvard Medical SchoolBostonMassachusettsUSA
| | - Abdulfattah Abbas
- Professor of Nephrology, Faculty of MedicineAl Andalus University for Medical SciencesTartusSyrian Arab Republic
| | - Zuheir Alshehabi
- Cancer Research CenterTishreen UniversityLattakiaSyrian Arab Republic
- Department of PathologyTishreen University HospitalLattakiaSyrian Arab Republic
| |
Collapse
|
7
|
Chen C, Zhang H, Lin Y, Lu M, Liao Q, Zhang S, Chen W, Zheng X, Li Y, Ding R, Wan Z. Identification of potential therapeutic drugs targeting core genes for systemic lupus erythematosus (SLE) and coexisting COVID-19: Insights from bioinformatic analyses. Immun Inflamm Dis 2023; 11:e1087. [PMID: 38018597 PMCID: PMC10659756 DOI: 10.1002/iid3.1087] [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: 06/23/2023] [Revised: 10/24/2023] [Accepted: 11/02/2023] [Indexed: 11/30/2023] Open
Abstract
OBJECTIVE Systemic lupus erythematosus (SLE) patients are at risk during the COVID-19 pandemic, yet the underlying molecular mechanisms remain incompletely understood. This study sought to analyze the potential molecular connections between COVID-19 and SLE, employing a bioinformatics approach to identify effective drugs for both conditions. METHODS The data sets GSE100163 and GSE183071 were utilized to determine share differentially expressed genes (DEGs). These DEGs were later analyzed by various bioinformatic methods, including functional enrichment, protein-protein interaction (PPI) network analysis, regulatory network construction, and gene-drug interaction construction. RESULTS A total of 50 common DEGs were found between COVID-19 and SLE. Gene ontology (GO) functional annotation revealed that "immune response," "innate immune response," "plasma membrane," and "protein binding" were most enriched in. Additionally, the pathways that were enriched include "Th1 and Th2 cell differentiation." The study identified 48 genes/nodes enriched with 292 edges in the PPI network, of which the top 10 hub genes were CD4, IL7R, CD3E, CD5, CD247, KLRB1, CD40LG, CD7, CR2, and GZMK. Furthermore, the study found 48 transcription factors and 8 microRNAs regulating these hub genes. Finally, four drugs namely ibalizumab (targeted to CD4), blinatumomab (targeted to CD3E), muromonab-CD3 (targeted to CD3E), and catumaxomab (targeted to CD3E) were found in gene-drug interaction. CONCLUSION Four possible drugs that targeted two specific genes, which may be beneficial for COVID-19 patients with SLE.
Collapse
Affiliation(s)
- Chao Chen
- School of Medicine, Institute of GenomicsHuaqiao UniversityXiamenChina
| | - Hongjian Zhang
- Department of Oncology and Vascular Interventional RadiologyZhongshan Hospital Xiamen University, School of Medicine, Xiamen UniversityXiamenFujianChina
| | - Yanbin Lin
- Department of Oncology and Vascular Interventional RadiologyZhongshan Hospital Xiamen University, School of Medicine, Xiamen UniversityXiamenFujianChina
| | - Meiqi Lu
- Department of Oncology and Vascular Interventional RadiologyZhongshan Hospital Xiamen University, School of Medicine, Xiamen UniversityXiamenFujianChina
| | - Quan Liao
- Department of Oncology and Vascular Interventional RadiologyZhongshan Hospital Xiamen University, School of Medicine, Xiamen UniversityXiamenFujianChina
| | - Shichao Zhang
- Department of Oncology and Vascular Interventional RadiologyZhongshan Hospital Xiamen University, School of Medicine, Xiamen UniversityXiamenFujianChina
| | - Weibin Chen
- Department of Oncology and Vascular Interventional RadiologyZhongshan Hospital Xiamen University, School of Medicine, Xiamen UniversityXiamenFujianChina
| | - Xiongwei Zheng
- Department of Oncology and Vascular Interventional RadiologyZhongshan Hospital Xiamen University, School of Medicine, Xiamen UniversityXiamenFujianChina
| | - Yunpeng Li
- Department of Oncology and Vascular Interventional RadiologyZhongshan Hospital Xiamen University, School of Medicine, Xiamen UniversityXiamenFujianChina
| | - Rui Ding
- Department of Oncology and Vascular Interventional RadiologyZhongshan Hospital Xiamen University, School of Medicine, Xiamen UniversityXiamenFujianChina
| | - Zheng Wan
- Department of Oncology and Vascular Interventional RadiologyZhongshan Hospital Xiamen University, School of Medicine, Xiamen UniversityXiamenFujianChina
| |
Collapse
|
8
|
Rozenberg JM, Buzdin AA, Mohammad T, Rakitina OA, Didych DA, Pleshkan VV, Alekseenko IV. Molecules promoting circulating clusters of cancer cells suggest novel therapeutic targets for treatment of metastatic cancers. Front Immunol 2023; 14:1099921. [PMID: 37006265 PMCID: PMC10050392 DOI: 10.3389/fimmu.2023.1099921] [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: 11/16/2022] [Accepted: 02/28/2023] [Indexed: 03/17/2023] Open
Abstract
Treatment of metastatic disease remains among the most challenging tasks in oncology. One of the early events that predicts a poor prognosis and precedes the development of metastasis is the occurrence of clusters of cancer cells in the blood flow. Moreover, the presence of heterogeneous clusters of cancerous and noncancerous cells in the circulation is even more dangerous. Review of pathological mechanisms and biological molecules directly involved in the formation and pathogenesis of the heterotypic circulating tumor cell (CTC) clusters revealed their common properties, which include increased adhesiveness, combined epithelial-mesenchymal phenotype, CTC-white blood cell interaction, and polyploidy. Several molecules involved in the heterotypic CTC interactions and their metastatic properties, including IL6R, CXCR4 and EPCAM, are targets of approved or experimental anticancer drugs. Accordingly, analysis of patient survival data from the published literature and public datasets revealed that the expression of several molecules affecting the formation of CTC clusters predicts patient survival in multiple cancer types. Thus, targeting of molecules involved in CTC heterotypic interactions might be a valuable strategy for the treatment of metastatic cancers.
Collapse
Affiliation(s)
- Julian M. Rozenberg
- Laboratory of Translational Bioinformatics, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Anton A. Buzdin
- Laboratory of Translational Bioinformatics, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
- PathoBiology Group, European Organization for Research and Treatment of Cancer (EORTC), Brussels, Belgium
- Group for Genomic Analysis of Cell Signaling, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
- Laboratory for Clinical Genomic Bioinformatics, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Tharaa Mohammad
- Laboratory of Translational Bioinformatics, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Olga A. Rakitina
- Gene Immunooncotherapy Group, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | - Dmitry A. Didych
- Laboratory of human genes structure and functions, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | - Victor V. Pleshkan
- Gene Immunooncotherapy Group, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
- Gene oncotherapy sector, Institute of Molecular Genetics of National Research Centre (Kurchatov Institute), Moscow, Russia
| | - Irina V. Alekseenko
- Gene Immunooncotherapy Group, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
- Gene oncotherapy sector, Institute of Molecular Genetics of National Research Centre (Kurchatov Institute), Moscow, Russia
- Laboratory of Epigenetics, Institute of Oncogynecology and Mammology, National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov, Ministry of Healthcare of the Russian Federation, Moscow, Russia
| |
Collapse
|
9
|
Wei J, Yang Y, Wang G, Liu M. Current landscape and future directions of bispecific antibodies in cancer immunotherapy. Front Immunol 2022; 13:1035276. [PMID: 36389699 PMCID: PMC9650279 DOI: 10.3389/fimmu.2022.1035276] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 10/14/2022] [Indexed: 07/31/2023] Open
Abstract
Recent advances in cancer immunotherapy using monoclonal antibodies have dramatically revolutionized the therapeutic strategy against advanced malignancies, inspiring the exploration of various types of therapeutic antibodies. Bispecific antibodies (BsAbs) are recombinant molecules containing two different antigens or epitopes identifying binding domains. Bispecific antibody-based tumor immunotherapy has gained broad potential in preclinical and clinical investigations in a variety of tumor types following regulatory approval of newly developed technologies involving bispecific and multispecific antibodies. Meanwhile, a series of challenges such as antibody immunogenicity, tumor heterogeneity, low response rate, treatment resistance, and systemic adverse effects hinder the application of BsAbs. In this review, we provide insights into the various architecture of BsAbs, focus on BsAbs' alternative different mechanisms of action and clinical progression, and discuss relevant approaches to overcome existing challenges in BsAbs clinical application.
Collapse
Affiliation(s)
- Jing Wei
- Gastric Cancer Center/Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yueyao Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Gang Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Ming Liu
- Gastric Cancer Center/Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
10
|
Kang J, Sun T, Zhang Y. Immunotherapeutic progress and application of bispecific antibody in cancer. Front Immunol 2022; 13:1020003. [PMID: 36341333 PMCID: PMC9630604 DOI: 10.3389/fimmu.2022.1020003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 10/05/2022] [Indexed: 08/19/2023] Open
Abstract
Bispecific antibodies (bsAbs) are artificial antibodies with two distinct antigen-binding sites that can bind to different antigens or different epitopes on the same antigen. Based on a variety of technology platforms currently developed, bsAbs can exhibit different formats and mechanisms of action. The upgrading of antibody technology has promoted the development of bsAbs, which has been effectively used in the treatment of tumors. So far, 7 bsAbs have been approved for marketing in the world, and more than 200 bsAbs are in clinical and preclinical research stages. Here, we summarize the development process of bsAbs, application in tumor treatment and look forward to the challenges in future development.
Collapse
Affiliation(s)
- Jingyue Kang
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Tonglin Sun
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Division of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yan Zhang
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Division of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
11
|
Abd El-Salam MA, Smith CEP, Pan CX. Insights on recent innovations in bladder cancer immunotherapy. Cancer Cytopathol 2022; 130:667-683. [PMID: 35653623 DOI: 10.1002/cncy.22603] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/12/2022] [Accepted: 05/13/2022] [Indexed: 12/12/2022]
Abstract
Bladder carcinoma is the most common genitourinary cancer, with a high prevalence and global incidence. In addition to early detection by cytology, the management of bladder cancer has recently advanced, not only by improvements in conventional treatments such as surgery and chemotherapy, but also through the introduction of immunotherapeutic strategies. The number of approved immunotherapeutic agents has dramatically increased, with various preclinical and clinical applications in cancer drug discovery. Some bladder cancer immunotherapies include immune checkpoint inhibitors, adoptive cell therapy, cytokine-based therapy, bispecific antibodies, and antibody-drug conjugates. This review provides an overview of some of the innovative immunotherapeutic agents approved and in development that can potentially be used in the treatment of bladder cancer.
Collapse
Affiliation(s)
- Mohamed A Abd El-Salam
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA.,Department of Medicine, VA Boston Healthcare System, Boston, Massachusetts, USA.,Institute for Research in Biomedicine (IRB) Barcelona, The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.,Department of Pharmacognosy, Faculty of Pharmacy, Delta University for Science and Technology International Coastal Road, Gamasa, Egypt
| | - Claire E P Smith
- Department of Medicine, VA Boston Healthcare System, Boston, Massachusetts, USA.,Department of Hematology and Medical Oncology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Chong-Xian Pan
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA.,Department of Medicine, VA Boston Healthcare System, Boston, Massachusetts, USA
| |
Collapse
|
12
|
Valenza C, Antonarelli G, Giugliano F, Aurilio G, Verri E, Briganti A, Curigliano G, Necchi A. Emerging treatment landscape of non-muscle invasive bladder cancer. Expert Opin Biol Ther 2022; 22:717-734. [PMID: 35634893 DOI: 10.1080/14712598.2022.2082869] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Non-muscle invasive bladder cancer (NMIBC) accounts for 70-75% of all bladder cancers and is a heterogeneous disease characterized by a wide spectrum of recurrences and progression. Adjuvant treatment for intermediate- and high-risk NMIBC is mainly represented by Bacillus Calmette Guerin (BCG). However, 20%-40% of patients develop disease recurrences or persistence following BCG treatment and are classified as "BCG unresponsive' (BCGu), thus representing a therapeutic challenge due to their worse prognosis and unavailability of effective intravesical treatments. AREAS COVERED We provide an overview of completed and ongoing clinical trials assessing the role of innovative immunological and target agents in patients with BCGu and BCG naive (BCGn) NMIBCs. New treatment options are emerging, demonstrating promising clinical activity, namely, pembrolizumab, atezolizumab, oportuzumab monatox, nadofaragene firadenovec, and N-803. EXPERT OPINION The increasing number of newer therapeutic agents for patients with NMIBC poses challenges regarding the choice of the most suited treatment option for each patient and the best treatment sequence, given their diverse mechanisms of action and varying degrees of activity. Tailored treatment approaches are advocated, based on a deeper comprehension of disease features, available therapies, patient's characteristics, and consequently, on the identification and validation of prognostic and predictive biomarkers.
Collapse
Affiliation(s)
- Carmine Valenza
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, Milan, Italy.,(DIPO), University of MilanDepartment of Oncology and Hemato-Oncology, Milan, Italy
| | - Gabriele Antonarelli
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, Milan, Italy.,(DIPO), University of MilanDepartment of Oncology and Hemato-Oncology, Milan, Italy
| | - Federica Giugliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, Milan, Italy.,(DIPO), University of MilanDepartment of Oncology and Hemato-Oncology, Milan, Italy
| | - Gaetano Aurilio
- Division of Urogenital and Head and Neck Tumours, European Institute of Oncology, Milan, Italy
| | - Elena Verri
- Division of Urogenital and Head and Neck Tumours, European Institute of Oncology, Milan, Italy
| | - Alberto Briganti
- San Raffaele Department of Medical Oncology, IRCCS San Raffaele Hospital and Scientific InstituteUniversity Vita-Salute, Milan, Italy
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, Milan, Italy.,(DIPO), University of MilanDepartment of Oncology and Hemato-Oncology, Milan, Italy
| | - Andrea Necchi
- San Raffaele Department of Medical Oncology, IRCCS San Raffaele Hospital and Scientific InstituteUniversity Vita-Salute, Milan, Italy
| |
Collapse
|
13
|
Shin HG, Yang HR, Yoon A, Lee S. Bispecific Antibody-Based Immune-Cell Engagers and Their Emerging Therapeutic Targets in Cancer Immunotherapy. Int J Mol Sci 2022; 23:5686. [PMID: 35628495 PMCID: PMC9146966 DOI: 10.3390/ijms23105686] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/16/2022] [Accepted: 05/17/2022] [Indexed: 12/16/2022] Open
Abstract
Cancer is the second leading cause of death worldwide after cardiovascular diseases. Harnessing the power of immune cells is a promising strategy to improve the antitumor effect of cancer immunotherapy. Recent progress in recombinant DNA technology and antibody engineering has ushered in a new era of bispecific antibody (bsAb)-based immune-cell engagers (ICEs), including T- and natural-killer-cell engagers. Since the first approval of blinatumomab by the United States Food and Drug Administration (US FDA), various bsAb-based ICEs have been developed for the effective treatment of patients with cancer. Simultaneously, several potential therapeutic targets of bsAb-based ICEs have been identified in various cancers. Therefore, this review focused on not only highlighting the action mechanism, design and structure, and status of bsAb-based ICEs in clinical development and their approval by the US FDA for human malignancy treatment, but also on summarizing the currently known and emerging therapeutic targets in cancer. This review provides insights into practical considerations for developing next-generation ICEs.
Collapse
Affiliation(s)
- Ha Gyeong Shin
- Department of Biopharmaceutical Chemistry, College of Science and Technology, Kookmin University, Seoul 02707, Korea; (H.G.S.); (H.R.Y.)
| | - Ha Rim Yang
- Department of Biopharmaceutical Chemistry, College of Science and Technology, Kookmin University, Seoul 02707, Korea; (H.G.S.); (H.R.Y.)
| | - Aerin Yoon
- R&D Division, GC Biopharma, Yongin 16924, Korea
| | - Sukmook Lee
- Department of Biopharmaceutical Chemistry, College of Science and Technology, Kookmin University, Seoul 02707, Korea; (H.G.S.); (H.R.Y.)
- Biopharmaceutical Chemistry Major, School of Applied Chemistry, Kookmin University, Seoul 02707, Korea
- Antibody Research Institute, Kookmin University, Seoul 02707, Korea
| |
Collapse
|