1
|
Nikanjam M, Pérez-Granado J, Gramling M, Larvol B, Kurzrock R. Nectin-4 expression patterns and therapeutics in oncology. Cancer Lett 2025; 622:217681. [PMID: 40209851 DOI: 10.1016/j.canlet.2025.217681] [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: 03/02/2025] [Revised: 03/23/2025] [Accepted: 03/26/2025] [Indexed: 04/12/2025]
Abstract
Nectin-4 is a transmembrane receptor that is implicated in migration, adhesion, and proliferation of tumor cells. It has minimal expression in healthy adult tissue but is overexpressed in a number of cancers, with higher expression levels seen in breast, bladder, colorectal, gallbladder, gastric, and non-small cell lung cancers. Enfortumab vedotin is an antibody-drug conjugate against nectin-4 that has been studied in a number of solid tumors and is FDA-approved for advanced urothelial cancers (including in combination with pembrolizumab). There are a number of other nectin-4-targeting agents under investigation. This review summarizes nectin-4 expression (membranous and cytoplasmic)/amplification in cancer and its potential as a response biomarker, as well as clinical trials of enfortumab vedotin and novel nectin-4 targeting agents. Biomarker-driven approaches merit investigation for nectin-4 directed across tumor types.
Collapse
Affiliation(s)
- Mina Nikanjam
- Division of Hematology-Oncology, University of California San Diego, La Jolla, San Diego, CA, USA.
| | | | | | | | - Razelle Kurzrock
- Medical College of Wisconsin Cancer Center, Milwaukee, WI, USA; WIN Consortium, France.
| |
Collapse
|
2
|
Khosravifarsani M, Njotu FN, Fon DA, Fonge H. Maximizing therapeutic potential and safety: Exploring multi/dual-payload antibody conjugates as cancer theranostics. Adv Drug Deliv Rev 2025; 222:115608. [PMID: 40393542 DOI: 10.1016/j.addr.2025.115608] [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: 03/15/2025] [Revised: 05/06/2025] [Accepted: 05/16/2025] [Indexed: 05/22/2025]
Abstract
Tumor heterogeneity greatly contributes to the failure of traditional cancer treatments. This leads to tumor relapse, recurrence, and ultimately metastasis, presenting serious clinical challenges. In recent decades, advances in antibody-based immunotherapy have emerged as promising new pillars to combat cancers. Although single payload antibody drug conjugates (ADCs) have resulted in drastic improvements in patient outcomes compared with unconjugated antibodies, multiple de novo and acquired resistance mechanisms inherent with cancer cells have left patients with less than desired outcomes. Newer studies are exploring the use of dual and multiple payload ADCs to enhance effectiveness. These payloads include chemotherapeutic and/or radiotherapeutic agents. The approaches leverage the synergistic effects of the different payloads alongside the immunotherapeutic properties of the antibody carriers. This review presents a comprehensive overview of dual-payload monoclonal antibody conjugates for cancer therapy and diagnosis (theranostics). Additionally, it explores the use of various imageable radiometals that are conjugated to the ADCs for imaging/diagnosis. It discusses the role of radioisotope decay schemes (such as alpha emission, beta emission, or Auger electron emission) along with factors such as linker type and chelator, as well as drug-to-antibody ratio (DAR), which are aimed at enhancing the synergistic effects between the therapeutic payloads while ensuring safety. Because none of these dual-payload ADCs have reached the clinic, this review employs a predictive method to estimate human equivalent dose (HED), maximum tolerable dose (MTD), and radiotoxicity in humans based on preclinical data. Additionally, it discusses the combinatorial behavior of two cytotoxic payloads linked to a monoclonal antibody.
Collapse
Affiliation(s)
- Meysam Khosravifarsani
- Faculté de Pharmacie, Université Laval, Ferdinand Vandry Pavillon, Québec, QC G1V 0A6, Canada; Axe Oncologie, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC G1J 5B3 Canada
| | - Fabrice Ngoh Njotu
- Faculté de Pharmacie, Université Laval, Ferdinand Vandry Pavillon, Québec, QC G1V 0A6, Canada; Axe Oncologie, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC G1J 5B3 Canada
| | - Dede Api Fon
- Faculté de Pharmacie, Université Laval, Ferdinand Vandry Pavillon, Québec, QC G1V 0A6, Canada; Axe Oncologie, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC G1J 5B3 Canada
| | - Humphrey Fonge
- Faculté de Pharmacie, Université Laval, Ferdinand Vandry Pavillon, Québec, QC G1V 0A6, Canada; Axe Oncologie, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC G1J 5B3 Canada.
| |
Collapse
|
3
|
Abdelhamid MS, Wadan AHS, Saad HA, El-Dakroury WA, Hageen AW, Mohammed DH, Mourad S, Mohammed OA, Abdel-Reheim MA, Doghish AS. Nanoparticle innovations in targeted cancer therapy: advancements in antibody-drug conjugates. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:6369-6389. [PMID: 39825965 DOI: 10.1007/s00210-024-03764-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 12/23/2024] [Indexed: 01/20/2025]
Abstract
Antibody-drug conjugates (ADCs) have emerged as a promising strategy in targeted cancer therapy, enabling the precise delivery of cytotoxic agents to tumor sites while minimizing systemic toxicity. However, traditional ADCs face significant limitations, including restricted drug loading capacity, where an optimal drug-to-antibody ratio (DAR) is crucial; low DARs may lead to insufficient potency, while high DARs can cause rapid clearance and increased toxicity. Additionally, ADCs often suffer from instability in circulation due to the potential for premature release of cytotoxic agents, resulting in off-target effects and reduced therapeutic efficacy. Furthermore, their large size can impede adequate penetration into solid tumors, particularly in heterogeneous environments with varying antigen expressions. This review explores the innovative use of nanoparticles as carriers for ADCs, which offers a multifaceted approach to enhance therapeutic efficacy. By leveraging the unique properties of nanoparticles, such as their small size and ability to exploit the enhanced permeability and retention (EPR) effect, researchers can improve drug stability, prolong circulation time, and achieve more effective tumor targeting. Recent studies demonstrate that nanoparticle-encapsulated ADCs can significantly enhance treatment outcomes while reducing off-target effects, as evidenced by improved targeting capabilities and reduced toxicity in preclinical models. Despite the promising advancements, challenges remain, including potential nanoparticle toxicity and manufacturing complexities. This review aims to provide a comprehensive overview of the current research on nanoparticle-encapsulated ADCs. It highlights their potential to transform cancer treatment and offers insights into future directions for optimizing these advanced therapeutic strategies.
Collapse
Affiliation(s)
| | - Al-Hassan Soliman Wadan
- Oral Biology Department, Faculty of Dentistry, Galala University, Galala Plateau, Attaka, 15888, Suez Governorate, Egypt
| | - Hager Adel Saad
- Faculty of Pharmacy, German University in Cairo (GUC), New Cairo, 11835, Cairo, Egypt
| | - Walaa A El-Dakroury
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Ahmed W Hageen
- Faculty of Medicine, Tanta University, Tanta, 31527, Egypt
| | | | - Sohaila Mourad
- Faculty of Medicine, Alexandria University, Alexandria, 21526, Egypt
| | - Osama A Mohammed
- Department of Pharmacology, College of Medicine, University of Bisha, 61922, Bisha, Saudi Arabia
| | | | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt.
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231, Cairo, Egypt.
| |
Collapse
|
4
|
Shen K, Yuan S, Su N, Tang F, Rehim S, Wang H, Guo H, Zhang Y, Wu Y, Wang H. Monotherapy and combination therapy using antibody‑drug conjugates for platinum‑resistant ovarian cancer. Oncol Rep 2025; 53:68. [PMID: 40242965 PMCID: PMC12046379 DOI: 10.3892/or.2025.8901] [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: 12/04/2024] [Accepted: 03/17/2025] [Indexed: 04/18/2025] Open
Abstract
Platinum‑resistant ovarian cancer (PROC) is a significant clinical challenge due to the limited number of treatment options and poor outcomes. Moreover, cytotoxic drugs have an unsatisfactory therapeutic efficacy, high toxicity and side effects. An antibody‑drug conjugate (ADC) is a novel cancer therapeutic strategy that combines an antibody, a linker and a payload. ADCs precisely target the tumor cells by binding to the antigen on the surface of tumor cells, thus accurately delivering the cytotoxic drugs and minimizing systemic toxicity. The approval of mirvetuximab soravtansine by the US Food and Drug Administration for treating folate receptor alpha‑positive, platinum‑resistant epithelial ovarian cancer has promoted studies on the use of ADCs in ovarian cancer. A phase III clinical trial showed that mirvetuximab soravtansine achieved an objective remission rate of 42.3% in platinum‑resistant, FRα‑positive ovarian cancer, compared with 15.9% using chemotherapy, demonstrating its immense potential for ADC development. The present review summarizes the research progress on the use of ADCs in PROC as a monotherapy and combination therapy and considers the future development direction of ADCs in PROC.
Collapse
Affiliation(s)
- Ke Shen
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan 610041, P.R. China
| | - Shuang Yuan
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan 610041, P.R. China
| | - Ning Su
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan 610041, P.R. China
- Department of Gynecologic Oncology, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan 450008, P.R. China
| | - Furong Tang
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan 610041, P.R. China
| | - Shamsnur Rehim
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan 610041, P.R. China
| | - Han Wang
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan 610041, P.R. China
| | - Huihui Guo
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan 610041, P.R. China
| | - Yu Zhang
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan 610041, P.R. China
| | - Yufeng Wu
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan 610041, P.R. China
| | - Hongjing Wang
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
5
|
Sweeney-Lasch S, Quillmann M, Hannewald J, Dickgiesser S, Rasche N, Shan M, Deutsch C, Hecht S, Anderl J, Kolmar H, Piater B. Elucidating Critical Factors of Internalization and Drug Release of Antibody-Drug Conjugates (ADCs) Using Kinetic Parameters Evaluated by a Novel Tool Named TORCH. Bioconjug Chem 2025; 36:945-959. [PMID: 40326736 DOI: 10.1021/acs.bioconjchem.4c00579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
During the past decade, antibody-drug conjugates (ADCs) have emerged as new drugs in cancer therapy with 15 ADCs already approved such as Kadcyla, Enhertu, and Adcetris. ADCs contain a cytotoxic drug that is linked to an antibody, allowing for specific delivery of the warhead to tumor cells. Typically, the antibody targets a tumor-specific antigen expressed on the cell surface. After the internalization of ADCs into cells, the linker is often cleaved by enzymes in the lysosomal compartment of the cell, releasing the warhead and thereby allowing for its interaction with, for example, the DNA or the tubulin cytoskeleton, which finally leads to cell death. Consequently, binding, internalization, and drug release are key attributes for the efficacy of ADCs. Here, we describe a novel molecule named TORCH (Turn On after Release by CatHepsins) that contains a fluorescence quencher system that is separated by a cathepsin B-cleavable linker. When conjugated to an antibody, the TORCH molecule allows one to gain valuable insights on the internalization and drug release of ADCs. While we cannot exclude the influence of other factors such as receptor recycling, we have found that the receptor density is directly related to the amount of payload released intracellularly, meaning that the internalization per receptor is very similar for all investigated antibodies and cell lines.
Collapse
Affiliation(s)
- Stanley Sweeney-Lasch
- Department of Discovery & Development Technologies, The Healthcare Business of Merck KGaA, Darmstadt 64293, Germany
| | - Marie Quillmann
- Department of Biochemistry, Technical University of Darmstadt, Darmstadt 64293, Germany
| | - Jens Hannewald
- Department of Discovery & Development Technologies, The Healthcare Business of Merck KGaA, Darmstadt 64293, Germany
| | - Stephan Dickgiesser
- Department of Discovery & Development Technologies, The Healthcare Business of Merck KGaA, Darmstadt 64293, Germany
| | - Nicolas Rasche
- Department of Discovery & Development Technologies, The Healthcare Business of Merck KGaA, Darmstadt 64293, Germany
| | - Min Shan
- Department of Discovery & Development Technologies, The Healthcare Business of Merck KGaA, Darmstadt 64293, Germany
| | - Carl Deutsch
- Department of Discovery & Development Technologies, The Healthcare Business of Merck KGaA, Darmstadt 64293, Germany
| | - Stefan Hecht
- Department of Discovery & Development Technologies, The Healthcare Business of Merck KGaA, Darmstadt 64293, Germany
| | - Jan Anderl
- Department of Discovery & Development Technologies, The Healthcare Business of Merck KGaA, Darmstadt 64293, Germany
| | - Harald Kolmar
- Department of Biochemistry, Technical University of Darmstadt, Darmstadt 64293, Germany
| | - Birgit Piater
- Department of Discovery & Development Technologies, The Healthcare Business of Merck KGaA, Darmstadt 64293, Germany
| |
Collapse
|
6
|
Han J, Xu K, Yang L, Ding Y, Wang X, Yin D, Wang J, Zhang H, Hong Z. Cleavable PEGylation Enhances the Antitumor Efficacy of Small-Sized Antibody-Drug Conjugates. Mol Pharm 2025. [PMID: 40394811 DOI: 10.1021/acs.molpharmaceut.5c00090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2025]
Abstract
Antibody-drug conjugates (ADCs) have emerged as a promising class of cancer therapeutics. However, traditional ADCs are often limited by poor tumor penetration due to their large molecular size. While the use of small-sized antibody fragments or analogues can improve tumor permeability, this approach typically results in an extremely shortened blood circulation half-life, which diminishes the therapeutic benefits and brings other metabolic challenges. In addition, the expression of target antigens on normal tissues often leads to unnecessary on-target/off-tumor toxicity. To address these issues, we developed a novel tumor site-specific cleavable PEGylation strategy for small-sized ADC design. The small ADC molecule ZHER2-MMAE was site-specifically PEGylated at its N-terminus with a 20 kDa polyethylene glycol (PEG) chain and a uPA (LSGRSDNH) cleavage sequence was inserted between them (PEG20k-U-ZHER2-MMAE). Our results showed that PEG20k-U-ZHER2-MMAE achieves a similar half-life extension (6.4 and 6.0 h) compared to the conventional PEG20k-ZHER2-MMAE, both representing about a 26-fold improvement compared to ZHER2-MMAE. Importantly, PEG20k-U-ZHER2-MMAE exhibited significantly higher drug accumulation at the tumor site, leading to the complete eradication of NCI-N87 and SK-OV-3 tumors at a dose of 5.5 mg/kg. Additionally, it demonstrated a maximum tolerated dose (MTD) exceeding 35 mg/kg, while the noncleavable PEG20k-ZHER2-MMAE could only slow tumor growth. In addition, compared to ZHER2-MMAE, the in vitro cytotoxic activity of PEG20k-ZHER2-MMAE or PEG20k-U-ZHER2-MMAE was reduced by about 50 times, with the latter expected to reduce the on-target/off-tumor side effects due to the specific activation by uPA at tumor sites. These data fully demonstrate the effectiveness and high safety of our tumor-specific cleavable PEGylation strategy, supporting the potential in the development of next-generation ADCs for cancer therapy.
Collapse
Affiliation(s)
- Jiani Han
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Cancer Biology Center, Frontiers Science Center for New Organic Matter, College of Life Sciences, Nankai University, Tianjin 300071, P. R. China
| | - Keyuan Xu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Cancer Biology Center, Frontiers Science Center for New Organic Matter, College of Life Sciences, Nankai University, Tianjin 300071, P. R. China
| | - Liu Yang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Cancer Biology Center, Frontiers Science Center for New Organic Matter, College of Life Sciences, Nankai University, Tianjin 300071, P. R. China
| | - Yu Ding
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Cancer Biology Center, Frontiers Science Center for New Organic Matter, College of Life Sciences, Nankai University, Tianjin 300071, P. R. China
| | - Xi Wang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Cancer Biology Center, Frontiers Science Center for New Organic Matter, College of Life Sciences, Nankai University, Tianjin 300071, P. R. China
| | - Dongming Yin
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Cancer Biology Center, Frontiers Science Center for New Organic Matter, College of Life Sciences, Nankai University, Tianjin 300071, P. R. China
| | - Jian Wang
- Department of Comprehensive Surgery, Tianjin First Central Hospital, No. 24 Fukang Road, Tianjin 300192, P. R. China
| | - Hongru Zhang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Cancer Biology Center, Frontiers Science Center for New Organic Matter, College of Life Sciences, Nankai University, Tianjin 300071, P. R. China
- Nankai International Advanced Research Institute (Shenzhen Futian), Shenzhen 518045, P. R. China
| | - Zhangyong Hong
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Cancer Biology Center, Frontiers Science Center for New Organic Matter, College of Life Sciences, Nankai University, Tianjin 300071, P. R. China
- Nankai International Advanced Research Institute (Shenzhen Futian), Shenzhen 518045, P. R. China
| |
Collapse
|
7
|
Wharton T, Spring DR. Methods for the Generation of Single-Payload Antibody-Drug Conjugates. ChemMedChem 2025; 20:e202500132. [PMID: 40052373 PMCID: PMC12091852 DOI: 10.1002/cmdc.202500132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 03/06/2025] [Indexed: 03/20/2025]
Abstract
Antibody-drug conjugates (ADCs) have emerged as a powerful form of targeted therapy that can deliver drugs with a high level of selectivity towards a specific cell type, reducing off-target effects and increasing the therapeutic window compared to small molecule therapeutics. However, creating ADCs that are stable, homogeneous, and with controlled drug-to-antibody ratio (DAR) remains a significant challenge. Whilst a myriad of methods have been reported to generate ADCs with a DAR of 2, 4, and 8, strategies to generate DAR 1 constructs are seldom reported despite the advantages of low drug loading to tune ADC properties or to allow access to antibody-antibody and antibody-protein constructs. This concept article highlights the diversity of methods that have been employed to access single-payload ADCs and explores the outlook for the field.
Collapse
Affiliation(s)
- Thomas Wharton
- Yusuf Hamied Department of ChemistryUniversity of CambridgeLensfield RoadCambridgeUKCB2 1EW
| | - David R. Spring
- Yusuf Hamied Department of ChemistryUniversity of CambridgeLensfield RoadCambridgeUKCB2 1EW
| |
Collapse
|
8
|
Hushmandi K, Imani Fooladi AA, Reiter RJ, Farahani N, Liang L, Aref AR, Nabavi N, Alimohammadi M, Liu L, Sethi G. Next-generation immunotherapeutic approaches for blood cancers: Exploring the efficacy of CAR-T and cancer vaccines. Exp Hematol Oncol 2025; 14:75. [PMID: 40382583 DOI: 10.1186/s40164-025-00662-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 04/25/2025] [Indexed: 05/20/2025] Open
Abstract
Recent advancements in immunotherapy, particularly Chimeric antigen receptor (CAR)-T cell therapy and cancer vaccines, have significantly transformed the treatment landscape for leukemia. CAR-T cell therapy, initially promising in hematologic cancers, faces notable obstacles in solid tumors due to the complex and immunosuppressive tumor microenvironment. Challenges include the heterogeneous immune profiles of tumors, variability in antigen expression, difficulties in therapeutic delivery, T cell exhaustion, and reduced cytotoxic activity at the tumor site. Additionally, the physical barriers within tumors and the immunological camouflage used by cancer cells further complicate treatment efficacy. To overcome these hurdles, ongoing research explores the synergistic potential of combining CAR-T cell therapy with cancer vaccines and other therapeutic strategies such as checkpoint inhibitors and cytokine therapy. This review describes the various immunotherapeutic approaches targeting leukemia, emphasizing the roles and interplay of cancer vaccines and CAR-T cell therapy. In addition, by discussing how these therapies individually and collectively contribute to tumor regression, this article aims to highlight innovative treatment paradigms that could enhance clinical outcomes for leukemia patients. This integrative approach promises to pave the way for more effective and durable treatment strategies in the oncology field. These combined immunotherapeutic strategies hold great promise for achieving more complete and lasting remissions in leukemia patients. Future research should prioritize optimizing treatment sequencing, personalizing therapeutic combinations based on individual patient and tumor characteristics, and developing novel strategies to enhance T cell persistence and function within the tumor microenvironment. Ultimately, these efforts will advance the development of more effective and less toxic immunotherapeutic interventions, offering new hope for patients battling this challenging disease.
Collapse
Affiliation(s)
- Kiavash Hushmandi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Islamic Republic of Iran.
| | - Abbas Ali Imani Fooladi
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, 78229, USA
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Liping Liang
- Guangzhou Key Laboratory of Digestive Diseases, Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Amir Reza Aref
- Department of Vitro Vision, DeepkinetiX, Inc, Boston, MA, USA
| | | | - Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Le Liu
- Integrated Clinical Microecology Center, Shenzhen Hospital, Southern Medical University, Shenzhen, 518000, China.
- Department of Gastroenterology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| | - Gautam Sethi
- Department of Pharmacology and NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
| |
Collapse
|
9
|
Hashii N, Haruyama Y, Hirayama R, Kajita R, Kishino Y, Mochizuki T, Inoue K, Goda R, Hoshino M, Kuroiwa I, Aikawa H, Ueda N, Nagumo K, Oda Y, Saito Y, Ishii-Watabe A. Versatile intact LC-MS method for evaluating the drug-antibody ratio and drug load distribution of antibody-drug conjugates in human plasma. J Chromatogr B Analyt Technol Biomed Life Sci 2025; 1258:124608. [PMID: 40267607 DOI: 10.1016/j.jchromb.2025.124608] [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: 02/08/2025] [Revised: 04/04/2025] [Accepted: 04/15/2025] [Indexed: 04/25/2025]
Abstract
The average drug-antibody ratio (DAR) and drug load distribution (DLD) of an antibody-drug conjugate (ADC) can be altered by biotransformation after administration. In addition, drug loading affects the clearance and exposure of the ADC. Evaluating alterations in the average DAR and DLD of an ADC in vivo would provide valuable information to better understand of the pharmacokinetic (PK) profile of the ADC. Although the quantitation of antibodies/ADCs using LC-MS is often coupled with affinity capture methods, here, we aimed to develop a versatile intact LC-MS method for evaluating the average DAR and DLD of ADCs in human plasma. The development of the affinity purification process and method validation were performed using healthy human pooled plasma spiked with the model ADCs, commercially available trastuzumab emtansine (T-DM1) and brentuximab vedotin (B-MMAE), and the recombinant proteins HER2 and CD30 were used to capture T-DM1 and B-MMAE, respectively. As unique points of this study, initially, a two-step gradient was established for the sensitive detection of a small amount of ADC. The ADC elution conditions after affinity capture were also optimized considering its application for LC-MS analysis. Furthermore, a validation study of the intact LC-MS approach for analyzing the average DAR and DLD of ADCs in human plasma sample was proposed for the first time. Using the validation study, our analytical method was validated by verifying its performance characteristics, including sensitivity, intermediate precision, accuracy, carryover and autosampler stability. In addition, the feasibility of applying our method was demonstrated by a collaborative study with six laboratories. Finally, our method was shown to be versatile for evaluating the average DAR and DLD of ADCs in human plasma.
Collapse
Affiliation(s)
- Noritaka Hashii
- National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki 210-9501, Japan.
| | - Yusuke Haruyama
- CMIC Pharma Science Co., Ltd., 17-18, Nakahata, Nishiwaki 677-0032, Japan
| | - Ryu Hirayama
- CMIC Pharma Science Co., Ltd., 17-18, Nakahata, Nishiwaki 677-0032, Japan
| | - Ryo Kajita
- Daiichi Sankyo Co., Ltd., 1-2-58, Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Yuki Kishino
- Daiichi Sankyo Co., Ltd., 1-2-58, Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | | | - Kazuko Inoue
- Eisai Co., Ltd, 5-1-3 Tokodai, Tsukuba 300-2635, Japan
| | - Ryoya Goda
- Future Peak Co., Ltd., 3-48, Imainakamachi, Nakahara-ku, Kawasaki 211-0065, Japan
| | - Masaki Hoshino
- Mediford Corp., 1-15-10 Shimura, Itabashi-ku, Tokyo 174-0056, Japan
| | - Itsuki Kuroiwa
- Mediford Corp., 1-15-10 Shimura, Itabashi-ku, Tokyo 174-0056, Japan
| | - Hiroaki Aikawa
- Mediford Corp., 1-15-10 Shimura, Itabashi-ku, Tokyo 174-0056, Japan
| | - Natsuki Ueda
- Ono Pharmaceutical Co., Ltd., 17-2 Wadai, Tsukuba 300-4247, Japan
| | - Kaori Nagumo
- Ono Pharmaceutical Co., Ltd., 17-2 Wadai, Tsukuba 300-4247, Japan
| | - Yuki Oda
- Ono Pharmaceutical Co., Ltd., 17-2 Wadai, Tsukuba 300-4247, Japan
| | - Yoshiro Saito
- National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki 210-9501, Japan
| | - Akiko Ishii-Watabe
- National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki 210-9501, Japan
| |
Collapse
|
10
|
Hartmann M, Rauscher M, Robinson J, Welsh J, Roush D. Integration of QSAR models with high throughput screening to accelerate the development of polishing chromatography unit operations. J Chromatogr A 2025; 1747:465818. [PMID: 40023049 DOI: 10.1016/j.chroma.2025.465818] [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: 12/12/2024] [Revised: 02/21/2025] [Accepted: 02/24/2025] [Indexed: 03/04/2025]
Abstract
The development of robust polishing chromatographic processes is a critical step in downstream bioprocess development that can be time-consuming and resource intensive. Recently, there has been an increase in diverse protein constructs that are not amenable to platform approaches, increasing the need for novel processes to be developed for effective purification. High throughput screening (HTS) is an important tool to parse chromatographic design space and identify promising conditions to continue development. Despite its utility, HTS capabilities are challenged by tight development timelines, material scarcity, and an increasingly complex pipeline of biotherapeutics. Predictive modeling can augment HTS by leveraging historical screening data to rapidly explore and prioritize process design space, effectively expanding the range of conditions considered without the need for additional experimental screening. Here we present the development of a quantitative structure activity relationship (QSAR) model, trained from internal HTS data, that predicts protein partitioning as a function of resin and mobile phase conditions. The training dataset contains a diverse collection of screening data and has more than 8000 datapoints, covering 29 therapeutic proteins and 44 resins. The model encodes partitioning by building descriptors of the mobile phase, parameters that describe the resin, and biophysical properties of the protein. Overall, the regression model has an R2=0.92 and shows 95% and 93% classification accuracy for predicting elution and strong binding conditions, respectively. Here, we highlight the model predictiveness and describe how in silico screening can be used as a first step in the HTS workflow to reduce design space and accelerate process development.
Collapse
Affiliation(s)
- Michael Hartmann
- Modeling and Informatics, Merck & Co., Inc., Rahway, NJ 07065, USA.
| | - Michael Rauscher
- Downstream Biologics Process Research and Development, Merck & Co., Inc. Rahway, NJ 07065, USA
| | - Julie Robinson
- Downstream Biologics Process Research and Development, Merck & Co., Inc. Rahway, NJ 07065, USA
| | - John Welsh
- Downstream Biologics Process Research and Development, Merck & Co., Inc. Rahway, NJ 07065, USA
| | - David Roush
- Downstream Biologics Process Research and Development, Merck & Co., Inc. Rahway, NJ 07065, USA
| |
Collapse
|
11
|
Sonar PV, Singh AK, Mandadi S, Sharma NK. Expanding horizons of cancer immunotherapy: hopes and hurdles. Front Oncol 2025; 15:1511560. [PMID: 40352591 PMCID: PMC12061710 DOI: 10.3389/fonc.2025.1511560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 03/31/2025] [Indexed: 05/14/2025] Open
Abstract
Background Tumor displays various forms of tumor heterogeneity including immune heterogeneity that allow cancer cells to survive during conventional anticancer drug interventions. Thus, there is a strong rationale for overcoming anticancer drug resistance by employing the components of immune cells. Using the immune system to target tumor cells has revolutionized treatment. Recently, significant progress has been achieved at preclinical and clinical levels to benefit cancer patients. Approach A review of literature from the past ten years across PubMed, Scopus, and Web of Science focused on immunotherapy strategies. These include immune checkpoint inhibitors (ICIs), tumor-infiltrating lymphocyte therapy, antibody-drug conjugates (ADCs), cancer vaccines, CAR T-cell therapy, and the role of the gut microbiome. Conclusion While immunotherapy outcomes have improved, particularly for tumor types such as melanoma and non-small cell lung cancer (NSCLC), challenges persist regarding predictive biomarker identification and better management. Ongoing research on modifiers of immune function like gut microbiome-derived metabolites, next-generation ADCs, and new classes of biologics is warranted. Overall, continued investigation toward optimizing synergistic immunotherapeutic combinations through strategic drug delivery systems is imperative for preclinical and clinical success in cancer patients.
Collapse
Affiliation(s)
- Priyanka Vijay Sonar
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune, Maharashtra, India
| | - Anuj Kumar Singh
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune, Maharashtra, India
- Ichnos Glenmark Innovation, Glenmark Pharmaceuticals Limited, Navi Mumbai, Maharashtra, India
| | - Sravan Mandadi
- Ichnos Glenmark Innovation, Glenmark Pharmaceuticals Limited, Navi Mumbai, Maharashtra, India
| | - Nilesh Kumar Sharma
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune, Maharashtra, India
| |
Collapse
|
12
|
Valentin ME, Abdelal Q, Isern S, Ashby T. Rapidly progressive pneumonitis days after receiving tisotumab vedotin: a new antibody-drug conjugate. BMJ Case Rep 2025; 18:e264150. [PMID: 40234078 DOI: 10.1136/bcr-2024-264150] [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: 04/17/2025] Open
Abstract
The emergence of antibody-drug conjugates (ADCs) shows promise for treating a variety of tumours by using target-specific monoclonal antibodies to deliver cytotoxic substances directly to cancer cells. Their clinical development, however, is often hindered by significant toxicities. We report a case of a woman in her late 50s who developed grade 4 pneumonitis after receiving tisotumab vedotin (TV) for recurrent, metastatic squamous cell carcinoma of the cervix. 5 days post infusion, the patient exhibited severe respiratory distress, marked by hypoxaemia and diffuse ground-glass opacities on imaging. After careful ruling out of infection and alveolar haemorrhage via bronchoscopy, a presumptive diagnosis of drug-induced pneumonitis was established given the temporal relationship to receiving TV. The patient was treated with high-dose intravenous steroids and immunoglobulin, leading to eventual recovery. This case exhibits the potential for severe pulmonary toxicity associated with TV and highlights the need for greater awareness and investigation of adverse effects related to ADCs.
Collapse
Affiliation(s)
- Marcos E Valentin
- Pulmonary, University of Florida College of Medicine-Jacksonville, Jacksonville, Florida, USA
| | | | | | - Tracy Ashby
- Pulmonary and Critical Care, University of Florida Health at Jacksonville, Jacksonville, Florida, USA
| |
Collapse
|
13
|
Xu D, Guo S, Qi Z, He P, Guo X, Li H, Zhou Z, Hu X, Wang C, Yu C, Wu H. Stability of intravenous antibody dilutions in clinical use: Differences across patient populations with varying body weights. Int J Pharm 2025; 674:125496. [PMID: 40112902 DOI: 10.1016/j.ijpharm.2025.125496] [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: 12/03/2024] [Revised: 02/25/2025] [Accepted: 03/16/2025] [Indexed: 03/22/2025]
Abstract
Antibody-drug conjugate (ADC) plays a crucial role in the treatment of various diseases and intravenous injection is the primary administration route for ADCs due to its high bioavailability. However, this method requires drug preparation and dilution, which can compromise the stability of the solution and increase the risk of aggregation as excipient concentrations are reduced during dilution. Antibody drug dosing is often based on patient body weight and typically diluted in either 0.9% (w/v) sodium chloride or 5% (w/v) dextrose injections. Variations in patient body weight lead to differences in the final concentrations of both the drug and excipients, affecting stability. In this study, we examined the stability of intravenous dilutions for patients with varying body weights using trastuzumab and ado-trastuzumab emtansine ADCs. Analytical techniques including size-exclusion chromatography (SEC), fluid imaging microscopy (FIM), bicinchoninic acid (BCA) assay, and turbidity testing were employed to assess monomer, oligomer, and aggregate levels before and after dilution. Machine learning was applied to distinguish the morphology of sub-visible particles. Our findings reveal that weight-based dosing leads to significant stability differences in intravenous preparations. This study offers important insights into the formulation stability of biopharmaceuticals and their clinical use.
Collapse
Affiliation(s)
- Dongze Xu
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning, China
| | - Sha Guo
- WHO Collaborating Centre for Standardization and Evaluation of Biologicals, State Key Laboratory of Drug Regulatory Science, NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| | - Zhiyun Qi
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning, China; WHO Collaborating Centre for Standardization and Evaluation of Biologicals, State Key Laboratory of Drug Regulatory Science, NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| | - Pengfei He
- WHO Collaborating Centre for Standardization and Evaluation of Biologicals, State Key Laboratory of Drug Regulatory Science, NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| | - Xiang Guo
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning, China
| | - Hanhan Li
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning, China
| | - Zhenhao Zhou
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning, China
| | - Xueyan Hu
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning, China
| | - Cui Wang
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning, China
| | - Chuanfei Yu
- WHO Collaborating Centre for Standardization and Evaluation of Biologicals, State Key Laboratory of Drug Regulatory Science, NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, National Institutes for Food and Drug Control, Beijing, China.
| | - Hao Wu
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning, China.
| |
Collapse
|
14
|
Park YH, Bianchini G, Cortés J, Licata L, Vidal M, Iihara H, Roeland EJ, Jordan K, Scotté F, Schwartzberg L, Navari RM, Aapro M, Rugo HS. Nausea and vomiting in an evolving anticancer treatment landscape: long-delayed and emetogenic antibody-drug conjugates. Future Oncol 2025; 21:1261-1272. [PMID: 40105595 PMCID: PMC11988240 DOI: 10.1080/14796694.2025.2479417] [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: 10/08/2024] [Accepted: 03/11/2025] [Indexed: 03/20/2025] Open
Abstract
Nausea and vomiting are common, distressing side effects associated with chemotherapeutic regimens, resulting in reduced quality of life and treatment adherence. Appropriate antiemetic prophylaxis strategies may reduce/prevent chemotherapy-induced nausea and vomiting (CINV). Historically, investigators assessed antiemetics up to 120 hours after chemotherapy. However, CINV can extend beyond this time. Thus, the effect of antiemetics during the long-delayed period (>120 hours) requires investigation. Emerging treatment options, including certain antibody-drug conjugates (ADCs), are associated with high rates of acute and late-onset nausea and vomiting that can last for extended duration. With the increasing number of ADCs approved and in development, there is urgency to control nausea and vomiting in patients receiving these new therapies. In this narrative review, we present the emetogenic potential of ADCs and CINV in the long-delayed period along with antiemetic prophylaxis strategies used to date. We also discuss the promising role of the fixed-combination antiemetic NEPA ([fos]netupitant plus palonosetron) in controlling long-delayed nausea and vomiting, addressing characteristics that may contribute to its longer efficacy duration compared to other antiemetics. Finally, we highlight encouraging results with NEPA in patients receiving the ADCs trastuzumab deruxtecan or sacituzumab govitecan, which suggest NEPA may be an effective antiemetic prophylaxis in these settings.
Collapse
Affiliation(s)
- Yeon Hee Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Giampaolo Bianchini
- Department of Medical Oncology, IRCCS San Raffaele Hospital, Milan, Italy
- School of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy
| | - Javier Cortés
- International Breast Cancer Center (IBCC), Pangaea Oncology, Quironsalud Group, Barcelona, Spain
- IOB Madrid, Hospital Beata María Ana, Madrid, Spain
- Faculty of Biomedical and Health Sciences, Department of Medicine, Universidad Europea de Madrid, Madrid, Spain
| | - Luca Licata
- Department of Medical Oncology, IRCCS San Raffaele Hospital, Milan, Italy
| | - María Vidal
- Department of Medical Oncology, Hospital Clinic, Translational Genomics and Targeted Therapies in Solid Tumors, IDIBAPS, Barcelona, Spain
- Department of Medicine, University of Barcelona, Barcelona, Spain
| | | | - Eric J. Roeland
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Karin Jordan
- Department of Hematology, Oncology and Palliative Medicine, Ernst von Bergmann Hospital, Potsdam, Germany
- Department of Medicine V, Hematology, Oncology and Rheumatology, University of Heidelberg, Heidelberg, Germany
| | - Florian Scotté
- Interdisciplinary Cancer Course Department, Gustave Roussy Cancer Institute, Villejuif, France
| | - Lee Schwartzberg
- Renown Health-Pennington Cancer Institute, University of Nevada, Reno, NV, USA
| | | | - Matti Aapro
- Genolier Cancer Centre, Clinique de Genolier, Genolier, Switzerland
| | - Hope S. Rugo
- University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, USA
| |
Collapse
|
15
|
Zhao Y, Yuan H, Li N, Wu L. A mini-overview of antibody-drug conjugates in platinum-resistant ovarian cancer: A preclinical and clinical perspective. Int J Biol Macromol 2025; 304:140767. [PMID: 39922351 DOI: 10.1016/j.ijbiomac.2025.140767] [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: 10/30/2024] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/10/2025]
Abstract
Ovarian cancer is one of the most lethal gynaecologic cancers in China. Although platinum-based chemotherapy, PARP inhibitors and bevacizumab have prolonged long term survival and increased the overall response rate for platinum-sensitive ovarian cancer (PSOC), the treatment options for platinum-resistant ovarian cancer (PROC) are still limited. Antibody-drug conjugates (ADCs) represent a novel form of precision medicine, covalently linking specific monoclonal antibodies with potent cytotoxic payloads. Since mirvetuximab soravtansine (MIRV) received approval by the US Food and Drug Administration (FDA) as the first ADC for PROC in 2022, the development of novel ADCs for various targets in PROC has accelerated. In this review, we summarise the recent evidence and future prospects of ADCs targeting Folate Receptor alpha (FRα), mesothelin, cadherin-6, NaPi2b, human epidermal growth factor receptor 2 (HER2), dipeptidase 3 (DPEP3), B7-H4 (VTCN1), claudin-6 (CLDN6) and trophoblast antigen protein 2 (TROP2), in order to enhance our understanding of the clinical applications of ADCs and offer new insights for clinical practice and further research.
Collapse
Affiliation(s)
- Yuxi Zhao
- Department of Gynaecological Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100000, China
| | - Hua Yuan
- Department of Gynaecological Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100000, China
| | - Ning Li
- Department of Gynaecological Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100000, China
| | - Lingying Wu
- Department of Gynaecological Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100000, China.
| |
Collapse
|
16
|
Huang H, Zhou Y, Shang C, Zhang Y, Shen Y. A novel anti-HER2/EGFR bispecific antibody-drug conjugate demonstrates promising antitumor efficacy and overcomes resistance to HER2- or EGFR-targeted ADCs. Invest New Drugs 2025; 43:262-275. [PMID: 39982632 DOI: 10.1007/s10637-025-01507-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 01/17/2025] [Indexed: 02/22/2025]
Abstract
HER2 and EGFR are frequently co-expressed in various tumors. While antibody-drug conjugates (ADCs) targeting HER2, such as T-DM1 and T-Dxd, have shown remarkable antitumor effects in clinical responses, their effectiveness is constrained by drug resistance. EGFR amplification or high expression is one of the factors that lead to resistance against HER2-targeted ADCs. Likewise, the amplification of HER2 may lead to the development of resistance to EGFR-targeted therapies. To overcome these challenges, we, therefore, developed a bispecific antibody (B2C4) that targets HER2 and EGFR. B2C4 exhibited strong binding affinity and internalization activity in tumor cells with high expression of HER2 and EGFR, as well as in those with high expression of either target. B2C4 was then conjugated with vc-MMAE to create a bispecific ADC (B2C4-MMAE) with an average DAR of 4.05. By effectively engaging both arms of the bispecific ADC, B2C4-MMAE demonstrated significant antitumor activity in tumor cells and animal models that were unresponsive HER2- or EGFR-targeted ADCs. B2C4-MMAE could serve as an alternative therapeutic option for tumors that are resistant to single-target treatments. Additionally, B2C4-MMAE exhibited potential in treating tumors resistant to T-Dxd, underscoring its promise as a treatment for challenging cases.
Collapse
Affiliation(s)
- Huoying Huang
- Anhui University of Chinese Medicine, Hefei, People's Republic of China
- Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co., Ltd, Beijing, People's Republic of China
- Joint Graduate School, Yangtze Delta Drug Advanced Research Institute, Nantong, People's Republic of China
| | - Yuxin Zhou
- Anhui University of Chinese Medicine, Hefei, People's Republic of China
- Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co., Ltd, Beijing, People's Republic of China
- Joint Graduate School, Yangtze Delta Drug Advanced Research Institute, Nantong, People's Republic of China
| | - Chengzhang Shang
- Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co., Ltd, Beijing, People's Republic of China
| | - Yifu Zhang
- Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co., Ltd, Beijing, People's Republic of China
| | - Yuelei Shen
- Institute of Innovative Medicine, Biocytogen Pharmaceuticals (Beijing) Co., Ltd, Beijing, People's Republic of China.
- Joint Graduate School, Yangtze Delta Drug Advanced Research Institute, Nantong, People's Republic of China.
| |
Collapse
|
17
|
Ieki K, Fukuda S, Miyawaki S, Hirowatari K. Regulated bioanalysis of antibody-drug conjugates using LC-MS. Bioanalysis 2025; 17:549-560. [PMID: 40205765 DOI: 10.1080/17576180.2025.2490468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 04/03/2025] [Indexed: 04/11/2025] Open
Abstract
Antibody-drug conjugates (ADCs) are emerging as powerful tools in cancer therapy. Evaluating their drug disposition requires the development and validation of analytical methods to obtain accurate quantitative results, which depend on understanding the ADC structural properties and selecting appropriate analytical platforms. Liquid chromatography-mass spectrometry (LC-MS) is a key technology for ADC bioanalysis, enabling the quantification of payloads, linkers, total antibodies, ADCs, and drug-to-antibody ratio (DAR). This review highlights the strategies and challenges in developing analytical methods for quantifying ADC components in biological samples using LC-MS with a focus on their constituent units. In addition, it addresses the validation requirements of these quantitative analytical methods during drug development.
Collapse
Affiliation(s)
- Katsunori Ieki
- Pharmacokinetics & Bioanalysis Center, Shin Nippon Biomedical Laboratories, Ltd., Kainan, Wakayama, Japan
| | - Suguru Fukuda
- Pharmacokinetics & Bioanalysis Center, Shin Nippon Biomedical Laboratories, Ltd., Kainan, Wakayama, Japan
| | - Shiori Miyawaki
- Pharmacokinetics & Bioanalysis Center, Shin Nippon Biomedical Laboratories, Ltd., Kainan, Wakayama, Japan
| | - Kazunari Hirowatari
- Pharmacokinetics & Bioanalysis Center, Shin Nippon Biomedical Laboratories, Ltd., Kainan, Wakayama, Japan
| |
Collapse
|
18
|
Ding M, Chen M, Cheng Z, Jin J, Lu W, Zhu S. Comparison of quaternary ammonium-based linkers for antibody-drug conjugates based on camptothecin derivatives. Bioorg Med Chem 2025; 120:118084. [PMID: 39893760 DOI: 10.1016/j.bmc.2025.118084] [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: 12/09/2024] [Revised: 01/17/2025] [Accepted: 01/22/2025] [Indexed: 02/04/2025]
Abstract
Antibody-drug conjugates (ADCs) with camptothecin derivatives as payloads have been a hot topic of interest and research since the launch of DS-8201a. As an important component of ADCs, the adequate stability of the linker during circulation and its rapid release at the target site are crucial for the efficient efficacy of ADCs. Although traditional quaternary ammonium ADCs based on dipeptide linkers were highly stable and could be released by specific enzymes, their poor in vitro anti-tumor activity had limited their further exploration. We applied a methylsulfonylethylamine-modified MAC self-elimination system to a valine-alanine linker and constructed a quaternary ammonium ADC (HER2-11) that combines both stability and cleavability. The optimization of the linker effectively improved the in vitro cellular activity of conventional quaternary ammonium ADCs, but the complex intracellular cleavage mechanism of HER2-11 resulted in a weaker anti-tumor activity compared to HER2-GGFG-DXd, which provides great reference value for the continued research of this type of linker in the future.
Collapse
Affiliation(s)
- Mengyuan Ding
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062 PR China
| | - Ming Chen
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062 PR China
| | - Zhiyang Cheng
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062 PR China
| | - Jiyu Jin
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062 PR China
| | - Wei Lu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062 PR China.
| | - Shulei Zhu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062 PR China; Innovation Center for AI and Drug Discovery, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062 PR China; ATLATL Innovation Center, 1077 Zhangheng Road, Shanghai 201203 PR China.
| |
Collapse
|
19
|
Shaaban AM, Kaur T, Provenzano E. HER2-Low Breast Cancer-Current Knowledge and Future Directions. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:644. [PMID: 40282933 PMCID: PMC12028887 DOI: 10.3390/medicina61040644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 03/19/2025] [Accepted: 03/21/2025] [Indexed: 04/29/2025]
Abstract
The concept of binary classification of HER2 status has recently been challenged following the DESTINY-Breast trial data showing a clinically meaningful response to antibody-drug conjugates (ADCs) in invasive breast cancer expressing low levels of HER2. HER2-low breast cancer is defined as an immunohistochemistry (IHC) score of 1+ and 2+ without HER2 gene amplification. While HER2-low breast cancer does not represent a biological entity, it encompasses both hormone receptor-positive and triple-negative breast cancer. Differences exist between this group and HER2-null breast cancer. In this review, we provide an update on HER2-low and HER2-ultralow breast cancer, including background trial data, the evolution of HER2-low expression, current clinical guidelines, quality issues, and future directions.
Collapse
Affiliation(s)
- Abeer M. Shaaban
- Department of Cellular Pathology, Queen Elizabeth Hospital Birmingham, Birmingham B15 2GW, UK
- Cancer and Genomic Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Tanvier Kaur
- Department of Cellular Pathology, New Cross Hospital, Wolverhampton WV10 0QP, UK;
| | - Elena Provenzano
- Department of Histopathology, Addenbrookes Hospital, Cambridge CB2 0QQ, UK;
- NIHR Cambridge Biomedical Research Centre, Cambridge CB2 0QQ, UK
| |
Collapse
|
20
|
Yamada T, Furusho A, Kojima K, Sugiyama E, Mizuno H, Tsukakoshi K, Hayashi H, Yamano T, Hasebe T, Toyo'oka T, Ikebukuro K, Todoroki K. Development of a mertansine-specific DNA aptamer and novel high-throughput sandwich enzyme-linked oligonucleotide assay for quantification and characterization of trastuzumab emtansine. Biosens Bioelectron 2025; 272:117108. [PMID: 39764977 DOI: 10.1016/j.bios.2024.117108] [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: 10/30/2024] [Revised: 12/25/2024] [Accepted: 12/27/2024] [Indexed: 01/22/2025]
Abstract
We developed a novel DNA aptamer, D8#24S1, which specifically recognizes mertansine (DM1), the cytotoxic payload of the antibody-drug conjugate (ADC) trastuzumab emtansine (T-DM1), and applied it for T-DM1 analysis. D8#24S1 was obtained through SELEX and was shown to specifically recognize DM1 with high affinity (dissociation constant, KD = 84.2 nM). By combining this anti-payload aptamer with the trastuzumab-specific anti-idiotype aptamer, CH1-S3, we developed a sandwich enzyme-linked oligonucleotide assay (sELONA) for evaluating T-DM1 content and drug-to-antibody ratio (DAR). The sELONA demonstrated an excellent fit to 4-parameter logistic curve model (R2 = 0.994) over a T-DM1 concentration range of 1-500 μg/mL, with a lower limit of quantification of 1 μg/mL, a precision within 23.9% (n = 3), and an accuracy within ±20.2% (n = 3). The sELONA also showed specificity when tested with other therapeutic monoclonal antibodies, such as trastuzumab and bevacizumab, as well as complex samples like serum. For DAR analysis, the sELONA exhibited high linearity (R2 = 0.988) and a strong correlation with hydrophobic interaction chromatography, a conventional method (R2 = 0.984). Unlike antibody-based assays, such as enzyme-linked immunosorbent assays, the sELONA employs chemically synthesized aptamers, offering superior robustness and cost-effectiveness. Additionally, when compared to conventional HIC, sELONA utilizes a 96-well microplate format, enabling high-throughput analysis. This study demonstrates the feasibility of aptamer-based assays as reliable alternatives to antibody-dependent methods, providing an efficient and adaptable approach for evaluating ADCs and potentially contributing to streamlined pharmaceutical development.
Collapse
Affiliation(s)
- Tomohiro Yamada
- Laboratory of Analytical and Bio-Analytical Chemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, 422-8526, Japan; Analytical Research, Pharmaceutical Science and Technology Unit, Pharmaceutical Profiling and Development Function, DHBL, Eisai Co. Ltd., Ibaraki, 22 Sunayama, Kamisu, Ibaraki, 314-0255, Japan
| | - Aogu Furusho
- Laboratory of Analytical and Bio-Analytical Chemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, 422-8526, Japan
| | - Kenji Kojima
- Laboratory of Analytical and Bio-Analytical Chemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, 422-8526, Japan
| | - Eiji Sugiyama
- Laboratory of Analytical Chemistry, Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku, Nagoya, 468-8503, Japan
| | - Hajime Mizuno
- Laboratory of Analytical Chemistry, Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku, Nagoya, 468-8503, Japan
| | - Kaori Tsukakoshi
- Department of Biotechnology and Life Science, Graduate School of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo, 184-8588, Japan
| | - Hideki Hayashi
- Laboratory of Community Pharmaceutical Practice and Science, Gifu Pharmaceutical University, Daigaku-nishi 1-25-4, Gifu, 501-1196, Japan
| | - Takeshi Yamano
- Analytical Research, Pharmaceutical Science and Technology Unit, Pharmaceutical Profiling and Development Function, DHBL, Eisai Co. Ltd., Ibaraki, 22 Sunayama, Kamisu, Ibaraki, 314-0255, Japan
| | - Takashi Hasebe
- Analytical Research, Pharmaceutical Science and Technology Unit, Pharmaceutical Profiling and Development Function, DHBL, Eisai Co. Ltd., Ibaraki, 22 Sunayama, Kamisu, Ibaraki, 314-0255, Japan
| | - Toshimasa Toyo'oka
- Laboratory of Analytical and Bio-Analytical Chemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, 422-8526, Japan
| | - Kazunori Ikebukuro
- Department of Biotechnology and Life Science, Graduate School of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo, 184-8588, Japan
| | - Kenichiro Todoroki
- Laboratory of Analytical and Bio-Analytical Chemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, 422-8526, Japan.
| |
Collapse
|
21
|
Drouin A, Durand L, Esnault C, Gaboriaud P, Leblond V, Karim S, Fouché M, Dhommée C, Baltus CB, Boursin F, Aubrey N, Houben R, Schrama D, Guyétant S, Desgranges A, Viaud-Massuard MC, Gouilleux-Gruart V, Samimi M, Kervarrec T, Touzé A. Optimization of Adcitmer, a Monomethyl-Auristatin E bearing antibody-drug conjugate for the treatment of CD56-expressing cancers. J Immunother Cancer 2025; 13:e010897. [PMID: 40086820 PMCID: PMC11907074 DOI: 10.1136/jitc-2024-010897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 02/03/2025] [Indexed: 03/16/2025] Open
Abstract
The cell adhesion protein CD56 has been identified as a potential therapeutic target in several solid tumors and hematological malignancies. Recently, we developed a CD56-directed antibody-drug conjugate (ADC), called Adcitmer and demonstrated its antitumor properties in preclinical models of the rare and aggressive skin cancer Merkel cell carcinoma (MCC).The present study aims to further optimize Adcitmer to overcome the therapeutic limitations observed with previously evaluated CD56-targeting ADCs, which were partially related to toxic effects on leukocytes. To this end, we aimed to avoid interaction of Adcitmer with immune cells via Fc gamma receptor (FcγR) binding. Since glycosylation is essential for FcγR binding, an aglycosylated form of Adcitmer was generated and evaluated on human leukocytes and MCC cell lines using cell death (annexin V/7-aminoactinomycine D) and proliferation (2,3-Bis-(2-methoxy-4Nitro-5-sulfophenyl)-2H-tetrazolium-5carboxanilide) assays. Finally, the therapeutic performance of Adcitmer and its aglycosylated form was assessed in an MCC xenograft mouse model.Investigating the Adcitmer interaction with immune cells demonstrated that it is mostly mediated by Fc recognition. Accordingly, Adcitmer aglycosylation led to reduced immune cell toxicity and strikingly also to improved therapeutic performance even in an MCC xenograft model using immunodeficient mice.Our study suggests that aglycosylated Adcitmer should be considered as a therapeutic option in patients with advanced MCC or other CD56-positive tumors.
Collapse
Affiliation(s)
- Aurelie Drouin
- Team "Biologie des Infections à Polyomavirus", UMR1282, Tours, France
| | - Laurine Durand
- Team "Biologie des Infections à Polyomavirus", UMR1282, Tours, France
| | - Clara Esnault
- Team "Biologie des Infections à Polyomavirus", UMR1282, Tours, France
| | - Pauline Gaboriaud
- Team "Biologie des Infections à Polyomavirus", UMR1282, Tours, France
| | - Valérie Leblond
- Team "Biologie des Infections à Polyomavirus", UMR1282, Tours, France
| | - Shawk Karim
- Team FRAME, EA 7501, Tours, Centre-Val de Loire, France
| | | | | | | | - Fanny Boursin
- Team BIOMAP, UMR1282, Tours, Centre-Val de Loire, France
| | - Nicolas Aubrey
- Team BIOMAP, UMR1282, Tours, Centre-Val de Loire, France
| | - Roland Houben
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Würzburg, Germany
| | - David Schrama
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Würzburg, Germany
| | - Serge Guyétant
- Team "Biologie des Infections à Polyomavirus", UMR1282, Tours, France
- Department of Pathology, CHRU Hôpitaux de Tours, Tours, Centre-Val de Loire, France
| | | | | | | | - Mahtab Samimi
- Team "Biologie des Infections à Polyomavirus", UMR1282, Tours, France
- Department of Pathology, CHRU Hôpitaux de Tours, Tours, Centre-Val de Loire, France
| | - Thibault Kervarrec
- Team "Biologie des Infections à Polyomavirus", UMR1282, Tours, France
- Department of Pathology, CHRU Hôpitaux de Tours, Tours, Centre-Val de Loire, France
| | - Antoine Touzé
- Team "Biologie des Infections à Polyomavirus", UMR1282, Tours, France
| |
Collapse
|
22
|
Buyukgolcigezli I, Tenekeci AK, Sahin IH. Opportunities and Challenges in Antibody-Drug Conjugates for Cancer Therapy: A New Era for Cancer Treatment. Cancers (Basel) 2025; 17:958. [PMID: 40149295 PMCID: PMC11939980 DOI: 10.3390/cancers17060958] [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: 02/16/2025] [Revised: 03/10/2025] [Accepted: 03/10/2025] [Indexed: 03/29/2025] Open
Abstract
The antibody, linker, and payload moieties all play a significant role in giving the ADC its unique therapeutic potential. The antibody subclass employed in ADCs is determined based on relative individual receptor affinities and pharmacokinetics. Meanwhile, the linker used in an ADC can either be cleavable or non-cleavable. ADC therapy comprises antibody-dependent mechanisms in addition to the direct cytotoxic effects of the payload. These include antibody-dependent cellular cytotoxicity, complement-dependent cytotoxicity, and antibody-dependent cellular phagocytosis, as well as the "bystander effect", which refers to the diffusion of a portion of the cytotoxic molecules out of the target cell, exerting its cytotoxic effect on the adjacent cells. Target antigens of ADCs are expected to be expressed on the membranes of the cancer cells facing the external matrix, although new approaches utilize antigens regarding tumor-associated cells, the tumor microenvironment, or the tumor vasculature. These target antigens of ADCs not only determine the efficacy of these agents but also impact the off-targets and related adverse effects. The majority of ADC-related toxicities are associated with off-targets. The proposed mechanisms of ADC resistance include disrupted intracellular drug trafficking, dysfunctional lysosomal processing, and the efflux of the cytotoxic molecule via ATP-binding cassette (ABC) transporters. The latter mechanism is especially prominent for multi-drug-resistant tumors. An important limitation of ADCs is the penetration of the conjugate into the tumor microenvironment and their delivery to target cancer cells. Cancerous tissues' vascular profile and the steric "binding site barrier" formed around the peripheral vessels of tumors stand as potential challenges of ADC therapy for solid tumors. As research efforts focus on reducing toxicities, overcoming resistance, and improving pharmacokinetics, ADC options for cancer therapy are expected to continue to diversify, including standalone approaches and combination therapies.
Collapse
Affiliation(s)
| | - Ates Kutay Tenekeci
- Department of Biochemistry, Faculty of Medicine, Hacettepe University, Ankara 06230, Turkey;
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ibrahim Halil Sahin
- Division of Hematology/Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| |
Collapse
|
23
|
Dernbach G, Eich ML, Dragomir MP, Anders P, Jurczok N, Stief C, Jurmeister P, Schlomm T, Klauschen F, Horst D, Schulz GB, Schallenberg S. Spatial Expression of HER2, NECTIN4, and TROP-2 in Muscle-Invasive Bladder Cancer and Metastases: Implications for Pathological and Clinical Management. Mod Pathol 2025; 38:100753. [PMID: 40086591 DOI: 10.1016/j.modpat.2025.100753] [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: 08/11/2024] [Revised: 03/05/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025]
Abstract
Muscle-invasive bladder cancer (MIBC) presents significant treatment challenges. Antibody-drug conjugates targeting human epidermal growth factor receptor 2 (HER2), trophoblast cell surface antigen 2 (TROP-2), and nectin cell adhesion molecule 4 (NECTIN4) offer promising therapeutic options. This study examined the spatial expression of HER2, TROP-2, and NECTIN4 in MIBC and metastases, their association with molecular subtypes, and clinical outcomes. Formalin-fixed, paraffin-embedded tissue samples from 251 patients with MIBC were analyzed using immunohistochemistry and tissue microarray analysis. Expression patterns between the tumor front and tumor center (TC) were compared, and statistical analyses assessed associations with molecular subtypes and clinical parameters. Additionally, 67 matched lymph node metastases and a secondary cohort comprising 16 distant metastases, including 7 matched primary tumors, were examined to explore the expression patterns in advanced tumor stages. In primary tumors, HER2 was predominantly negative (83%) but showed higher positivity in the TC. TROP-2 exhibited high overall expression (58% score 3+), whereas NECTIN4 displayed significant heterogeneity with stronger expression in the TC. Spatial overexpression of TROP-2 and NECTIN4 at the tumor front relative to the TC was associated with a better disease-free survival. Accurate assessment required 4 biopsies for HER2 and NECTIN4 and 3 for TROP-2. HER2 expression was associated with urothelial-like and genomically unstable molecular subtypes, whereas TROP-2 was widely expressed except in the mesenchymal-like subtype. NECTIN4 showed the absence of staining in basal, Mes-like, and Sc/NEC-like subtypes. Paired lymph node metastases showed higher expression scores for all 3 markers, whereas distant metastases showed reduced NECTIN4 expression. Additionally, lymph node metastases revealed a considerable heterogeneity for HER2 compared with their matched primary tumors. The spatial heterogeneity of HER2, TROP-2, and NECTIN4 expression necessitates multiple biopsies, particularly from the TC, for accurate evaluation. These findings underscore the need for personalized treatment strategies in MIBC, considering the increased risk of relapse associated with HER2 and NECTIN4 overexpression in the TC. Implementing a multibiopsy approach is critical to enhancing diagnostic accuracy.
Collapse
Affiliation(s)
- Gabriel Dernbach
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institue of Pathology, Berlin, Germany; German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Berlin, Heidelberg, Germany.
| | - Marie-Lisa Eich
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institue of Pathology, Berlin, Germany.
| | - Mihnea P Dragomir
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institue of Pathology, Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany; German Cancer Consortium (DKTK), Partner Site Berlin, Berlin, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Philipp Anders
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institue of Pathology, Berlin, Germany
| | - Nadia Jurczok
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institue of Pathology, Berlin, Germany
| | - Christian Stief
- Department of Urology, University Hospital, LMU Munich, Munich, Germany
| | - Philipp Jurmeister
- Institute of Pathology, Ludwig Maximilians University, Munich, Germany; German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Munich Partner Site, Heidelberg, Germany
| | - Thorsten Schlomm
- Department of Urology, Charité Universitätsmedizin Berlin, Humboldt-Universität Zu Berlin, Berlin, Germany
| | - Frederick Klauschen
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institue of Pathology, Berlin, Germany; German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Berlin, Heidelberg, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany; Institute of Pathology, Ludwig Maximilians University, Munich, Germany; German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Munich Partner Site, Heidelberg, Germany; BIFOLD-Berlin Institute for the Foundations of Learning and Data, Berlin, Germany
| | - David Horst
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institue of Pathology, Berlin, Germany
| | | | - Simon Schallenberg
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institue of Pathology, Berlin, Germany.
| |
Collapse
|
24
|
Tan R, Yang Z, Xie J, Wu Z, Guo S, Li L, Yin Z, Hua H, Liu M, Li R. Innovative design concepts in tumor-targeting peptide-drug conjugates: Insights into emerging applications. Chin Med J (Engl) 2025:00029330-990000000-01456. [PMID: 40033748 DOI: 10.1097/cm9.0000000000003438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Indexed: 03/05/2025] Open
Abstract
ABSTRACT Peptide-drug conjugates (PDCs) have emerged as a promising strategy in cancer therapy, offering improved therapeutic efficacy and reduced toxicity. Compared to antibody-drug conjugates (ADCs) and small molecule-drug conjugates (SMDCs), PDCs possess distinct advantages, such as lower immunogenicity, improved tumor penetration, and simpler synthesis. This review discusses the latest advancements in PDC design, including novel peptide targeting mechanisms, linker selection, and formulation improvements for increased stability. Additionally, it explores the expanding clinical applications of PDCs and examines their limitations. The aim of this review is to provide a comprehensive overview of current PDC progress and outline future directions for their role in cancer treatment.
Collapse
Affiliation(s)
- Ruirong Tan
- Translational Chinese Medicine Key Laboratory of Sichuan, Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, Sichuan 610041, China
| | - Zhenya Yang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, China
| | - Jun Xie
- Information Technology Center, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
- West China Sanya Hospital of Sichuan University, Sanya, Hainan 572000, China
| | - Zijun Wu
- Xiangya School of Public Health, Central South University, Changsha, Hunan 410013, China
| | - Shanshan Guo
- Translational Chinese Medicine Key Laboratory of Sichuan, Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, Sichuan 610041, China
| | - Li Li
- Translational Chinese Medicine Key Laboratory of Sichuan, Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, Sichuan 610041, China
| | - Zhujun Yin
- Translational Chinese Medicine Key Laboratory of Sichuan, Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, Sichuan 610041, China
| | - Hua Hua
- Translational Chinese Medicine Key Laboratory of Sichuan, Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, Sichuan 610041, China
| | - Miao Liu
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Rui Li
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, Sichuan 610041, China
| |
Collapse
|
25
|
Hoyek C, Zheng-Lin B, Jones J, Bekaii-Saab T. Tucatinib in the treatment of HER2-overexpressing gastrointestinal cancers: current insights and future prospects. Expert Opin Investig Drugs 2025; 34:161-168. [PMID: 40019490 DOI: 10.1080/13543784.2025.2472411] [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: 08/14/2024] [Accepted: 02/23/2025] [Indexed: 03/01/2025]
Abstract
INTRODUCTION Over the past 20 years, the treatment landscape of HER2-amplified tumors has considerably evolved. Until now, no approved targeted therapies were available for patients with HER2-amplified metastatic colorectal cancer (mCRC). Tucatinib, a highly selective tyrosine kinase inhibitor, demonstrated significant efficacy in combination with trastuzumab in patients with refractory mCRC, leading to its approval by the Food and Drug Administration (FDA). AREAS COVERED This review dives into the efficacy of tucatinib-based regimens in gastrointestinal malignancies, with a focus on the pivotal MOUNTAINEER trial, which led to the FDA approval of tucatinib plus trastuzumab in chemo-refractory HER2-amplified mCRC. Additionally, ongoing trials are exploring tucatinib in earlier treatment lines and across other gastrointestinal cancers, including biliary tract, gastric, and pancreatic malignancies. The mechanistic basis of dual HER2 inhibition and its implications for clinical practice are discussed. EXPERT COMMENTARY The future of tucatinib-based therapeutic strategies in GI malignancies depends on their integration into different treatment lines. Addressing acquired resistance using liquid biopsy-guided strategies and other TKIs like lapatinib will be paramount to improve outcomes.
Collapse
Affiliation(s)
- Celine Hoyek
- Department of Hematology and Oncology, Mayo Clinic, Arizona, AZ, USA
| | - Binbin Zheng-Lin
- Department of Hematology and Oncology, Mayo Clinic, Arizona, AZ, USA
| | - Jeremy Jones
- Department of Hematology and Oncology, Mayo Clinic, Florida, FL, USA
| | | |
Collapse
|
26
|
Cohen JP, DiCaprio A, He J, Reibarkh M, Small J, Schombs M. Method for Screening Sodium Cyanoborohydride for Free Cyanide Content and Its Impact on Bioconjugation Chemistry. Bioconjug Chem 2025; 36:245-252. [PMID: 39912422 PMCID: PMC11843608 DOI: 10.1021/acs.bioconjchem.4c00514] [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: 11/10/2024] [Revised: 01/24/2025] [Accepted: 01/28/2025] [Indexed: 02/07/2025]
Abstract
Sodium cyanoborohydride (CBH) is commonly used as a mild reducing agent in the reductive amination of aldehydes and free amines. Within the pharmaceutical industry, this reaction is employed in the bioconjugation of proteins and peptides. Free cyanide species such as HCN and NaCN are known residual impurities in CBH that can contribute to the formation of undesired side products including cyanoamines and cyanohydrins. In commercial processes, the potential for bound cyanated species requires an analytical control strategy to monitor and mitigate any risk to human health. Given these concerns, minimization of cyanated side products is of utmost priority and can be achieved through a robust control strategy of quantitative screening of starting materials for free cyanide. Alternative risk mitigation strategies such as purification of bound cyanide containing species to pure species are less effective due to minor chemical differences between the expected product and bound cyanide species. Herein, we present a simple chromatographic assay for the quantitation of free cyanide in the raw material sodium cyanoborohydride. Method development, robustness evaluation, and scientific soundness assessment are reported with excellent linearity, accuracy, precision, and specificity. Additionally, this method was applied for the evaluation of raw material supplied from 10 commercial sources, none of which report a specification for free cyanide within their certificate of analysis. The measured free cyanide from these vendors ranged from 8 to 80 mM concentration, thereby confirming the value of screening these raw materials. Finally, we demonstrate the impact of free cyanide on a model bioconjugation reaction between ornithine and glyceraldehyde.
Collapse
Affiliation(s)
- Jarrod P. Cohen
- Vaccine Analytical
Research & Development, Merck &
Co., Inc., 770 Sumneytown Pike, P.O. Box 4, West Point, Pennsylvania 19486, United States
| | - Adam DiCaprio
- Analytical
Enabling Capabilities, Merck & Co.,
Inc., 770 Sumneytown Pike, P.O. Box 4, West Point, Pennsylvania 19486, United States
| | - Jian He
- Vaccine Analytical
Research & Development, Merck &
Co., Inc., 770 Sumneytown Pike, P.O. Box 4, West Point, Pennsylvania 19486, United States
| | - Mikhail Reibarkh
- Analytical
Enabling Capabilities, Merck & Co.,
Inc., 770 Sumneytown Pike, P.O. Box 4, West Point, Pennsylvania 19486, United States
| | - James Small
- Analytical
Enabling Capabilities, Merck & Co.,
Inc., 770 Sumneytown Pike, P.O. Box 4, West Point, Pennsylvania 19486, United States
| | - Matthew Schombs
- Vaccine Analytical
Research & Development, Merck &
Co., Inc., 770 Sumneytown Pike, P.O. Box 4, West Point, Pennsylvania 19486, United States
| |
Collapse
|
27
|
Buiar PG, Junior JDS, Sales MR, Favero GM. Time to focus again on matrix metalloproteinases? Results of complex network analysis involving the pathophysiology of HER2-positive breast cancer. Ecancermedicalscience 2025; 19:1850. [PMID: 40259907 PMCID: PMC12010132 DOI: 10.3332/ecancer.2025.1850] [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: 07/21/2024] [Indexed: 04/23/2025] Open
Abstract
Breast cancer is the most common cancer in women worldwide, with significant advances in understanding its multifactorial nature in recent years. The complex structure of molecular and cellular interactions in cancer pathophysiology presents challenges for developing effective treatments. One theoretical model used to study these interactions is the Graph model or Complex Networks, which uses mathematical methods to create graphical figures by connecting vertices (factors) through edges (interactions). This study uses the graph model to determine the complex interactions within the tumour microenvironment of HER2-positive breast cancer. Through a narrative review, 37 factors involved in the pathophysiology of HER2-positive breast cancer were identified and incorporated into a complex network design, starting with the HER2 vertex. The impact of each vertex was determined by calculating the relative error, and a knockout (KO) analysis of vertices was performed to identify their influences within the network. The Wilcoxon test was used to analyze the statistical significance of each KO. Significant alterations in the network structure were observed with the KOs of matrix metalloproteinases (MMPMMP2, MMP9, cyclin-dependent kinases 4/6, TWIST, vascular endothelial growth factor and transforming growth factor-beta. Notably, the KOs of (MMPs) MMP2 and MMP9 significantly impacted the network structure and downregulated the HER2 vertex. This raises questions about the potential applicability of targeting MMPs, including the option of HER2-directed antibody-drug conjugates. Could a metalloprotease inhibitor be a good choice for conjugation? Despite the theoretical nature of this model, the results suggest potential avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Pedro G Buiar
- Medical Oncology Department, Instituto Sul Paranaense de Oncologia, Ponta Grossa, Brazil
- https://orcid.org/0000-0001-5144-1197
| | - José Danilo Szezech Junior
- Department of Mathematics and Statistics, State University of Ponta Grossa, Ponta Grossa, PR, Brazil
- https://orcid.org/0000-0001-8306-8315
| | - Matheus Rolim Sales
- Department of Mathematics and Statistics, State University of Ponta Grossa, Ponta Grossa, PR, Brazil
- https://orcid.org/0000-0002-1121-6371
| | - Giovani Marino Favero
- Biological and Health Science Multidisciplinary Laboratory, State University of Ponta Grossa, Ponta Grossa, Brazil
- https://orcid.org/0000-0002-1946-3262
| |
Collapse
|
28
|
Casteleijn MG, Abendroth U, Zemella A, Walter R, Rashmi R, Haag R, Kubick S. Beyond In Vivo, Pharmaceutical Molecule Production in Cell-Free Systems and the Use of Noncanonical Amino Acids Therein. Chem Rev 2025; 125:1303-1331. [PMID: 39841856 PMCID: PMC11826901 DOI: 10.1021/acs.chemrev.4c00126] [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: 02/12/2024] [Revised: 12/26/2024] [Accepted: 01/06/2025] [Indexed: 01/24/2025]
Abstract
Throughout history, we have looked to nature to discover and copy pharmaceutical solutions to prevent and heal diseases. Due to the advances in metabolic engineering and the production of pharmaceutical proteins in different host cells, we have moved from mimicking nature to the delicate engineering of cells and proteins. We can now produce novel drug molecules, which are fusions of small chemical drugs and proteins. Currently we are at the brink of yet another step to venture beyond nature's border with the use of unnatural amino acids and manufacturing without the use of living cells using cell-free systems. In this review, we summarize the progress and limitations of the last decades in the development of pharmaceutical protein development, production in cells, and cell-free systems. We also discuss possible future directions of the field.
Collapse
Affiliation(s)
| | - Ulrike Abendroth
- VTT
Technical Research Centre of Finland Ltd, 02150 Espoo, Finland
| | - Anne Zemella
- Fraunhofer
Institute for Cell Therapy and Immunology (IZI), Branch Bioanalytics
and Bioprocesses (IZI-BB), Am Mühlenberg, 14476 Potsdam, Germany
| | - Ruben Walter
- Fraunhofer
Institute for Cell Therapy and Immunology (IZI), Branch Bioanalytics
and Bioprocesses (IZI-BB), Am Mühlenberg, 14476 Potsdam, Germany
| | - Rashmi Rashmi
- Freie
Universität Berlin, Institute of Chemistry and Biochemistry, 14195 Berlin, Germany
| | - Rainer Haag
- Freie
Universität Berlin, Institute of Chemistry and Biochemistry, 14195 Berlin, Germany
| | - Stefan Kubick
- Freie
Universität Berlin, Institute of Chemistry and Biochemistry, 14195 Berlin, Germany
- Faculty
of Health Sciences, Joint Faculty of the
Brandenburg University of Technology Cottbus–Senftenberg, The
Brandenburg Medical School Theodor Fontane and the University of Potsdam, 14469 Potsdam, Germany
- B4 PharmaTech
GmbH, Altensteinstraße
40, 14195 Berlin, Germany
| |
Collapse
|
29
|
Koganemaru S, Fuchigami H, Morizono C, Shinohara H, Kuboki Y, Furuuchi K, Uenaka T, Doi T, Yasunaga M. Potential Mechanisms of Interstitial Lung Disease Induced by Antibody-Drug Conjugates Based on Quantitative Analysis of Drug Distribution. Mol Cancer Ther 2025; 24:242-250. [PMID: 39450538 PMCID: PMC11791479 DOI: 10.1158/1535-7163.mct-24-0267] [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: 04/17/2024] [Revised: 06/18/2024] [Accepted: 10/22/2024] [Indexed: 10/26/2024]
Abstract
Antibody-drug conjugates (ADC) are a rapidly advancing category of therapeutic agents with notable anticancer efficacy. However, the emergence of interstitial lung disease as a severe ADC-associated adverse event highlights the need to better understand the underlying mechanisms. In this study, xenograft model mice with tumors expressing different levels of the trophoblast antigen 2 (TROP2) were generated by subcutaneously transplanting the various TROP2-expressing cancer lines. The mice received different doses of TROP2-eribulin, a novel TROP2-targeting ADC, composed of an anti-TROP2 antibody and the eribulin payload, joined by a cleavable linker. The concentration and distribution of TROP2-eribulin, as well as the pharmacokinetics of eribulin release, were assessed in tumor and lung tissues. Analysis of tumor tissue showed that the concentration of released eribulin was approximately 10-fold higher in NCI-H2110 (high TROP2 expression) than in A549 (low TROP2 expression), whereas analysis of lung tissue showed that TROP2-eribulin was distributed in lung tissue in a dose-dependent manner, regardless of TROP2 expression, with significantly more eribulin released in the high-dose group than in the other dose groups (P < 0.05). Immunofluorescence assay analysis showed that TROP2-eribulin localized to alveolar macrophages. In the analysis using human leukemia monocytic cell, the concentration of eribulin released from TROP2-eribulin was significantly reduced by the use of an Fc receptor inhibitor (P < 0.05). These results revealed that Fcγ receptor-mediated uptake by alveolar macrophages releases the cytotoxic payload into lung tissue, helping to clarify the pathogenesis of ADC-induced interstitial lung disease.
Collapse
Affiliation(s)
- Shigehiro Koganemaru
- Department of Experimental Therapeutics, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hirobumi Fuchigami
- Division of Developmental Therapeutics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Chihiro Morizono
- Department of Experimental Therapeutics, National Cancer Center Hospital East, Kashiwa, Japan
- Division of Developmental Therapeutics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Hiroko Shinohara
- Department of Experimental Therapeutics, National Cancer Center Hospital East, Kashiwa, Japan
- Division of Developmental Therapeutics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Yasutoshi Kuboki
- Department of Experimental Therapeutics, National Cancer Center Hospital East, Kashiwa, Japan
| | - Keiji Furuuchi
- Department of Preclinical Development, EPAT (Epochal Precision Anti-Cancer Therapeutics), Eisai Inc., Exton, Pennsylvania
| | - Toshimitsu Uenaka
- Department of Planning and Operations, EPAT, Eisai Inc., Exton, Pennsylvania
| | - Toshihiko Doi
- Department of Experimental Therapeutics, National Cancer Center Hospital East, Kashiwa, Japan
| | - Masahiro Yasunaga
- Division of Developmental Therapeutics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| |
Collapse
|
30
|
Yang Y, Patel JM, Yang RS, Ma F, Niu X, Zhang Y, Niedringhaus T, Al-Sayah M, Yang X. Determination of the decapping efficiency of THIOMAB™ antibodies with the engineered cysteine in the Fc region for making antibody-drug conjugates by specific hinge fragmentation-liquid chromatography. Anal Bioanal Chem 2025; 417:847-859. [PMID: 39688670 DOI: 10.1007/s00216-024-05707-w] [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: 09/24/2024] [Revised: 12/02/2024] [Accepted: 12/06/2024] [Indexed: 12/18/2024]
Abstract
The site-specific antibody-drug conjugates (ADCs), particularly those utilizing the engineered cysteine in Fc fragments of mAbs (THIOMAB™ antibodies), have emerged as a novel class of biotherapeutics for cancer treatment. The engineered cysteine residues in these antibodies are capped by cysteine or glutathione through a disulfide bond. Prior to conjugation with linker-payloads, these caps need to be removed through a reduction process. However, monitoring the efficiency of the decapping process has been challenging due to the lack of effective analytical methods. Intact reversed-phase liquid chromatography-mass spectrometry and hydrophobic interaction chromatography methods failed to separate decapped and capped intact THIOMAB™ mAbs in our study. Instead the fragmentation of mAbs provided a novel strategy to analyze the decapping effiency. After cleavage using a hinge specific enzyme, the generated Fc fragments with and without cysteine and/or glutathione caps displayed different hydrophobicity and were well separated by RPLC, allowing quantitative determination of the decapping efficiency. Enzymes that cleave both above and below the hinge disulfide bonds were tested. The use of FabALATICA can determine percentages of molecules with 0, 1, and 2 cysteine and/or glutathione caps, respectively, regardless of whether the antibody contains the hinge LALA mutations. On the other hand, FabRICATOR enzyme can only be utilized for antibodies without LALA mutations for the overall decapping percentage and cannot be used to estimate intact antibody each with 0, 1, and 2 caps. Therefore, FabALACTICA cleavage followed by RPLC provides a wider application of monitoring the decapping efficiency of all antibodies with the engineered cysteine in Fc.
Collapse
Affiliation(s)
- Yun Yang
- Discovery Analytical Research, Merck & Co., Inc, 213 E Grand Ave, South San Francisco, CA, 94080, USA
| | - Jaymin M Patel
- Biologics Analytical Research and Development (BARD), Merck & Co., Inc., 126 E. Lincoln Avenue, Rahway, NJ, 07065, USA
| | - Rong-Sheng Yang
- Analytical Research and Development, Merck & Co., Inc, 126 E. Lincoln Avenue, Rahway, NJ, 07065, USA
| | - Fengfei Ma
- Discovery Analytical Research, Merck & Co., Inc, 213 E Grand Ave, South San Francisco, CA, 94080, USA
| | - Xiangfeng Niu
- Discovery Analytical Research, Merck & Co., Inc, 213 E Grand Ave, South San Francisco, CA, 94080, USA
| | - Yixiao Zhang
- Bioprocess Research & Development, Merck & Co., Inc, 126 E. Lincoln Avenue, Rahway, NJ, 07065, USA
| | - Thomas Niedringhaus
- Biologics Analytical Research and Development (BARD), Merck & Co., Inc., 126 E. Lincoln Avenue, Rahway, NJ, 07065, USA
| | - Mohammad Al-Sayah
- Discovery Analytical Research, Merck & Co., Inc, 213 E Grand Ave, South San Francisco, CA, 94080, USA
| | - Xiaoyu Yang
- Discovery Analytical Research, Merck & Co., Inc, 213 E Grand Ave, South San Francisco, CA, 94080, USA.
- Biologics Analytical Research and Development (BARD), Merck & Co., Inc., 126 E. Lincoln Avenue, Rahway, NJ, 07065, USA.
| |
Collapse
|
31
|
Tripathy RK, Pande AH. Nanobody-Oligonucleotide Conjugates (NucleoBodies): The Next Frontier in Oligonucleotide Therapy. Pharm Res 2025; 42:219-236. [PMID: 39953265 DOI: 10.1007/s11095-025-03829-z] [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: 10/08/2024] [Accepted: 01/31/2025] [Indexed: 02/17/2025]
Abstract
As of now, more than 15 oligonucleotide drugs, primarily small interfering RNAs and antisense oligonucleotide classes, have been approved by the US FDA for therapeutic use, and many more are under clinical trials. However, safe and effective delivery of the oligonucleotide-based drugs to the target tissue still remains a major challenge. For enhanced plasma half-life, effective endosomal release, and other multiple functionalities, various carrier molecules have been used over the years. The successful therapeutic application of antibody-drug conjugates has made antibodies a popular choice for the delivery of oligonucleotide payloads into the target tissues. Single-chain variable domains of heavy chain antibodies (nanobodies) have proven a promising alternative to antibodies in recent years due to their small size, high affinity for the target, cell-penetrating potency, simple and easy production. The present review highlights the oligonucleotide drug types and their conjugation with nanobodies called NucleoBodies for effective targeted delivery, detection and diagnostics.
Collapse
Affiliation(s)
- Rajan K Tripathy
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali, 160062, Punjab, India
| | - Abhay H Pande
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali, 160062, Punjab, India.
| |
Collapse
|
32
|
Tao J, Gu Y, Zhou W, Wang Y. Dual-payload antibody-drug conjugates: Taking a dual shot. Eur J Med Chem 2025; 281:116995. [PMID: 39481229 DOI: 10.1016/j.ejmech.2024.116995] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/02/2024]
Abstract
Antibody-drug conjugates (ADCs) enable the precise delivery of cytotoxic agents by conjugating small-molecule drugs with monoclonal antibodies (mAbs). Over recent decades, ADCs have demonstrated substantial clinical efficacy. However, conventional ADCs often encounter various clinical challenges, including suboptimal efficacy, significant adverse effects, and the development of drug resistance, limiting their broader clinical application. Encouragingly, a next-generation approach-dual-payload ADCs-has emerged as a pioneering strategy to address these challenges. Dual-payload ADCs are characterized by the incorporation of two distinct therapeutic payloads on the same antibody, enhancing treatment efficacy by promoting synergistic effects and reducing the risk of drug resistance. However, the synthesis of dual-payload ADCs is complex due to the presence of multiple functional groups on antibodies. In this review, we comprehensively summarize the construction strategies for dual-payload ADCs, ranging from the design of ADC components to orthogonal chemistry. The subsequent sections explore current challenges and propose prospective strategies, highlighting recent advancements in dual-payload ADC research, thereby laying the foundation for the development of next-generation ADCs.
Collapse
Affiliation(s)
- Junjie Tao
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yilin Gu
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Wei Zhou
- Mabwell (Shanghai) Bioscience Co., Ltd, Shanghai, 201210, China.
| | - Yuxi Wang
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, Sichuan, China.
| |
Collapse
|
33
|
Ahmed G, Hamadani M, Al-Juhaishi T. The potential of antibody-drug conjugates for effective therapy in diffuse large B-cell lymphoma. Expert Opin Biol Ther 2025; 25:161-173. [PMID: 39798075 DOI: 10.1080/14712598.2025.2453524] [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: 08/01/2024] [Revised: 01/08/2025] [Accepted: 01/10/2025] [Indexed: 01/13/2025]
Abstract
INTRODUCTION Antibody-drug conjugates (ADCs) are a rapidly evolving class of anti-cancer drugs with a significant impact on management of hematological malignancies including diffuse large B-cell lymphoma (DLBCL). ADCs combine a cytotoxic drug (a.k.a. payload) attached through a linker to a monoclonal antibody specific to a particular cancer antigen. Payloads include microtubule disruptors or DNA damaging chemicals. After attaching to the antigen, the ADCs are internalized, and the payload is dissociated from ADC by lysozymes and delivered to the intended site for exerting cytotoxic effects. This unique molecular design permits a better balance of efficacy and safety. Loncastuximab tesirine and polatuzumab vedotin are two ADCs approved in the U.S.A. for treatment of DLBCL. AREAS COVERED This review covers the efficacy and safety data of these two drugs. We will review new ADC-based combination regimens and novel constructs in development. EXPERT OPINION ADCs have made a significant impact in improving outcomes of DLBCL patients. Both polatuzumab vedotin and loncastuximab tesirine are established as useful therapeutics options, with polatuzumab vedotin currently approved in first line and relapsed/refractory setting, while loncastuximab tesirine is approved in relapsed setting. ADCs are effective with tolerable safety profile and currently many more ADCs are undergoing clinical trials.
Collapse
MESH Headings
- Humans
- Immunoconjugates/adverse effects
- Immunoconjugates/administration & dosage
- Immunoconjugates/therapeutic use
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, Large B-Cell, Diffuse/immunology
- Antibodies, Monoclonal/adverse effects
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/therapeutic use
- Animals
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antineoplastic Agents/adverse effects
- Antineoplastic Agents/therapeutic use
- Benzodiazepines
Collapse
Affiliation(s)
- Gulrayz Ahmed
- Division of Hematology & Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Mehdi Hamadani
- Division of Hematology & Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | | |
Collapse
|
34
|
Malemnganba T, Pandey AK, Mishra A, Mehrotra S, Prajapati VK. Exploring immunotherapy with antibody-drug conjugates in solid tumor oncology. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 144:259-286. [PMID: 39978968 DOI: 10.1016/bs.apcsb.2024.10.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
Immunotherapy has emerged as a hallmark of hope in the formidable battle against solid tumors such as breast cancer, colorectal cancer, etc., with antibody-drug conjugates (ADCs) starting a new era of precision medicine. This chapter delves into the dynamic landscape of immunotherapeutic strategies, focusing on the transformative potential of ADCs. ADCs represent a combination of chemotherapy and immunotherapy, more innovative chemotherapy. We emphasize the intricate interplay between tumor biology and therapeutic intervention, uncovering the mechanisms underlying ADC efficacy and the hurdles they must overcome. Each facet of ADC development is carefully examined, from the delicate balance between payload potency and safety to the quest for enhanced tumor penetration. We also elucidate the synergistic potential of combining ADCs with existing modalities, including chemotherapy and radiation therapy, to amplify therapeutic outcomes while mitigating adverse effects. As we navigate the complexities of solid tumor oncology, a profound understanding of the immunotherapeutic potential of ADCs is gained, offering hope for a cure for patients and clinicians alike. Henceforth, we delve into this transformative journey as we advance in solid tumor treatment regimens using immunotherapy with ADCs, poised at the forefront of oncological innovation.
Collapse
Affiliation(s)
- Takhellambam Malemnganba
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India
| | - Anurag Kumar Pandey
- School of Biochemical Engineering, Indian Institute of Technology BHU, Varanasi, UP, India
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Rajasthan, India
| | - Sanjana Mehrotra
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Vijay Kumar Prajapati
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India.
| |
Collapse
|
35
|
Mihaylova R, Momekova D, Elincheva V, Momekov G. Immunoconjugates as an Efficient Platform for Drug Delivery: A Resurgence of Natural Products in Targeted Antitumor Therapy. Pharmaceuticals (Basel) 2024; 17:1701. [PMID: 39770542 PMCID: PMC11677665 DOI: 10.3390/ph17121701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/11/2024] [Accepted: 12/14/2024] [Indexed: 01/11/2025] Open
Abstract
The present review provides a detailed and comprehensive discussion on antibody-drug conjugates (ADCs) as an evolving new modality in the current therapeutic landscape of malignant diseases. The principle concepts of targeted delivery of highly toxic agents forsaken as stand-alone drugs are examined in detail, along with the biochemical and technological tools for their successful implementation. An extensive analysis of ADCs' major components is conducted in parallel with their function and impact on the stability, efficacy, safety, and resistance profiles of the immunoconjugates. The scope of the article covers the major classes of currently validated natural compounds used as payloads, with an emphasis on their structural and mechanistic features, natural origin, and distribution. Future perspectives in ADCs' design are thoroughly explored, addressing their inherent or emerging challenges and limitations. The survey also provides a comprehensive overview of the molecular rationale for active tumor targeting of ADC-based platforms, exploring the cellular biology and clinical relevance of validated tumor markers used as a "homing" mechanism in both hematological and solid tumor malignancies.
Collapse
Affiliation(s)
- Rositsa Mihaylova
- Department “Pharmacology, Pharmacotherapy and Toxicology”, Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria; (V.E.); (G.M.)
| | - Denitsa Momekova
- Department “Pharmaceutical Technology and Biopharmaceutics”, Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria;
| | - Viktoria Elincheva
- Department “Pharmacology, Pharmacotherapy and Toxicology”, Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria; (V.E.); (G.M.)
| | - Georgi Momekov
- Department “Pharmacology, Pharmacotherapy and Toxicology”, Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria; (V.E.); (G.M.)
| |
Collapse
|
36
|
Su Z, Yin S, Wu Y. Rationalize the Functional Roles of Protein-Protein Interactions in Targeted Protein Degradation by Kinetic Monte Carlo Simulations. J Phys Chem B 2024; 128:12092-12100. [PMID: 39610271 DOI: 10.1021/acs.jpcb.4c06497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2024]
Abstract
Targeted protein degradation is a promising therapeutic strategy to tackle disease-causing proteins that lack binding pockets for traditional small-molecule inhibitors. Its first step is to trigger the proximity between a ubiquitin ligase complex and a target protein through a heterobifunctional molecule, such as proteolysis targeting chimeras (PROTACs), leading to the formation of a ternary complex. The properties of protein-protein interactions play an important regulatory role during this process, which can be reflected by binding cooperativity. Unfortunately, although computer-aided drug design has become a cornerstone of modern drug development, the endeavor to model-targeted protein degradation is still in its infancy. The development of computational tools to understand the impacts of protein-protein interactions on targeted protein degradation, therefore, is highly demanded. To reach this goal, we constructed a nonredundant structural benchmark of the most updated ternary complexes and applied a kinetic Monte Carlo method to simulate the association between ligases and PROTAC-targeted proteins in the benchmark. Our results show that proteins in most complexes with positive cooperativity tend to associate into native-like configurations more often. In contrast, proteins very likely failed to associate into native-like configurations in complexes with negative cooperativity. Moreover, we compared protein-protein association through different interfaces generated from molecular docking. The native-like binding interface shows a higher association probability than all the other alternative interfaces only in the complex with positive cooperativity. These observations support the idea that the formation of ternary complexes is closely regulated by the binary interactions between proteins. Finally, we applied our method to cyclin-dependent kinases 4 and 6 (CDK4/6). We found that their interactions with the ligase are not as similar as their structures. Altogether, our study paves the way for understanding the role of protein-protein interactions in the PROTAC-induced ternary complex formation. It can potentially help in searching for degraders that selectively target specific proteins.
Collapse
Affiliation(s)
- Zhaoqian Su
- Data Science Institute, Vanderbilt University, 1001 19th Ave S, Nashville, Tennessee 37212, United States
| | - Shanye Yin
- Department of Pathology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York 10461, United States
| | - Yinghao Wu
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York 10461, United States
| |
Collapse
|
37
|
Olarewaju O, Held F, Curtis P, Kenny CH, Maier U, Panavas T, du Plessis F. αFAP-specific nanobodies mediate a highly precise retargeting of modified AAV2 capsids thereby enabling specific transduction of tumor tissues. Mol Ther Methods Clin Dev 2024; 32:101378. [PMID: 39703904 PMCID: PMC11655695 DOI: 10.1016/j.omtm.2024.101378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 11/08/2024] [Indexed: 12/21/2024]
Abstract
Due to the refractiveness of tumor tissues to adeno-associated virus (AAV) transduction, AAV vectors are poorly explored for cancer therapy delivery. Here, we aimed to engineer AAVs to target tumors by enabling the specific engagement of fibroblast activation protein (FAP). FAP is a cell surface receptor distinctly upregulated in the reactive tumor stroma, but rarely expressed in healthy tissues. Thus, targeting FAP presents an opportunity to selectively transduce tumor tissues. To achieve this, we modified the capsid surface of AAV2 with an αFAP nanobody to retarget the capsid to engage FAP receptor. Following transduction, we observed a 23- to 80-fold increase in the selective transduction of FAP+ tumor cells in vitro, and greater than 5-fold transduction of FAP+ tumor tissues in vivo. Subsequent optimization of the VP1-nanobody expression cassette further enhanced the transduction efficiency of the modified capsids. Due to the limited αFAP nanobodies repertoires, we broadened the versatility of this high-fidelity platform by screening a naive VHH yeast display library, leading to the identification of several novel αFAP nanobody candidates (KD = 0.1 to >100 nM). Hence, our study offers new opportunity for the application of AAV vectors for highly selective delivery of therapeutics to the tumor stroma.
Collapse
Affiliation(s)
- Olaniyi Olarewaju
- AAV Gene Therapy Research Group, Research Beyond Borders (RBB), Boehringer Ingelheim Pharma GmbH & Co. KG, 88400 Biberach an der Riß, Germany
| | - Franziska Held
- AAV Gene Therapy Research Group, Research Beyond Borders (RBB), Boehringer Ingelheim Pharma GmbH & Co. KG, 88400 Biberach an der Riß, Germany
| | - Pamela Curtis
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT 06877, USA
| | - Cynthia Hess Kenny
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT 06877, USA
| | - Udo Maier
- AAV Gene Therapy Research Group, Research Beyond Borders (RBB), Boehringer Ingelheim Pharma GmbH & Co. KG, 88400 Biberach an der Riß, Germany
| | - Tadas Panavas
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT 06877, USA
| | - Francois du Plessis
- AAV Gene Therapy Research Group, Research Beyond Borders (RBB), Boehringer Ingelheim Pharma GmbH & Co. KG, 88400 Biberach an der Riß, Germany
| |
Collapse
|
38
|
Kashyap VK, Sharma BP, Pandey D, Singh AK, Peasah-Darkwah G, Singh B, Roy KK, Yallapu MM, Chauhan SC. Small Molecule with Big Impact: Metarrestin Targets the Perinucleolar Compartment in Cancer Metastasis. Cells 2024; 13:2053. [PMID: 39768145 PMCID: PMC11674295 DOI: 10.3390/cells13242053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 12/01/2024] [Accepted: 12/02/2024] [Indexed: 01/11/2025] Open
Abstract
Metarrestin (ML246) is a first-in-class pyrrole-pyrimidine-derived small molecule that selectively targets the perinucleolar compartment (PNC). PNC is a distinct subnuclear structure predominantly found in solid tumor cells. The occurrence of PNC demonstrates a positive correlation with malignancy, serving as an indicator of tumor aggressiveness, progression, and metastasis. Various promising preclinical results have led to the clinical translation of metarrestin into a first-in-human trial. This review aims to summarize (i) the current understanding of the structure and function of PNC and its role in cancer progression and metastasis, (ii) key findings from studies examining the effect of metarrestin on various cancers across the translational spectrum, including in vitro, in vivo, and human clinical trial studies, and (iii) the pharmaceutical relevance of metarrestin as a promising anticancer candidate. Furthermore, our molecular docking and MD simulation studies show that metarrestin binds to eEF1A1 and eEF1A2 with a strong and stable affinity and inhibits eEF1A2 more efficiently compared to eEF1A1. The promising results from preclinical studies suggest that metarrestin has the potential to revolutionize the treatment of cancer, heralding a paradigm shift in its therapeutic management.
Collapse
Affiliation(s)
- Vivek K. Kashyap
- Division of Cancer Immunology and Microbiology, Medicine, and Oncology Integrated Service Unit, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
- South Texas Center of Excellence in Cancer Research (ST-CECR), McAllen, TX 78504, USA
| | - Bhuvnesh P. Sharma
- Department of Biotechnology, Bhagwant University, Ajmer 305004, Rajasthan, India
| | - Divya Pandey
- Department of Pharmaceutical Sciences, School of Health Sciences and Technology, UPES, Dehradun 248007, Uttarakhand, India
| | - Ajay K. Singh
- Department of Oncology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Godwin Peasah-Darkwah
- Division of Cancer Immunology and Microbiology, Medicine, and Oncology Integrated Service Unit, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
- South Texas Center of Excellence in Cancer Research (ST-CECR), McAllen, TX 78504, USA
| | - Bhupesh Singh
- School of Applied Sciences, OM Sterling Global University, Hisar 125001, Haryana, India
| | - Kuldeep K. Roy
- Department of Pharmaceutical Sciences, School of Health Sciences and Technology, UPES, Dehradun 248007, Uttarakhand, India
| | - Murali M. Yallapu
- Division of Cancer Immunology and Microbiology, Medicine, and Oncology Integrated Service Unit, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
- South Texas Center of Excellence in Cancer Research (ST-CECR), McAllen, TX 78504, USA
| | - Subhash C. Chauhan
- Division of Cancer Immunology and Microbiology, Medicine, and Oncology Integrated Service Unit, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
- South Texas Center of Excellence in Cancer Research (ST-CECR), McAllen, TX 78504, USA
| |
Collapse
|
39
|
Lucas S, Thomas SN. Therapeutic Immunomodulation of Tumor-Lymphatic Crosstalk via Intratumoral Immunotherapy. Mol Pharm 2024; 21:5929-5943. [PMID: 39478434 PMCID: PMC11615947 DOI: 10.1021/acs.molpharmaceut.4c00692] [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: 07/02/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 12/06/2024]
Abstract
Intra- and peritumoral lymphatics and tumor-draining lymph nodes play major roles in mediating the adaptive immune response to cancer immunotherapy. Despite this, current paradigms of clinical cancer management seldom seek to therapeutically modulate tumor-lymphatic immune crosstalk. This review explores recent developments that set the stage for how this regulatory axis can be therapeutically manipulated, with a particular emphasis on tumor-localized immunomodulation. Building on this idea, the nature of tumor-lymphatic immune crosstalk and relevant immunotherapeutic targets and pathways are reviewed, with a focus on their translational potential. Engineered drug delivery systems that enhance intratumoral immunotherapy by improving drug delivery to both the tumor and lymph nodes are also highlighted.
Collapse
Affiliation(s)
- Samuel
N. Lucas
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
of America
| | - Susan N. Thomas
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
of America
- George
W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States of America
- Parker
H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, United States of America
- Winship
Cancer Institute, Emory University, Atlanta, Georgia 30322, United States
of America
| |
Collapse
|
40
|
Nesbitt C, Van Der Walt A, Butzkueven H, Devitt B, Jokubaitis VG. Multiple sclerosis and cancer: Navigating a dual diagnosis. Mult Scler 2024; 30:1714-1736. [PMID: 39347791 DOI: 10.1177/13524585241274523] [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: 10/01/2024]
Abstract
Healthcare breakthroughs are extending the lives of multiple sclerosis (MS) patients and cancer survivors, creating a growing cohort of individuals navigating a dual diagnosis. Determining the relationship between MS and cancer risk remains challenging, with inconclusive findings confounded by age, risk exposures, comorbidities, genetics and the ongoing introduction of new MS disease-modifying therapies (DMTs) across study periods.This research places significant emphasis on cancer survival, with less attention given to the impact on MS outcomes. Our review explores the existing literature on MS, cancer risk and the intersection of DMTs and cancer treatments. We aim to navigate the complexities of managing MS in cancer survivors to optimise outcomes for both conditions. Continuous research and the formulation of treatment guidelines are essential for guiding future care. Collaboration between neuro-immunology and oncology is crucial, with a need to establish databases for retrospective and ultimately prospective analysis of outcomes in these rapidly evolving fields.
Collapse
Affiliation(s)
- Cassie Nesbitt
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
- Department of Neurology, MSNI Multiple Sclerosis and Neuro-Immunology, Alfred Health, Melbourne, VIC, Australia
- Department of Neuroscience, Monash University, The Alfred Centre, Melbourne, VIC, Australia
| | - Anneke Van Der Walt
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
- Department of Neurology, MSNI Multiple Sclerosis and Neuro-Immunology, Alfred Health, Melbourne, VIC, Australia
| | - Helmut Butzkueven
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
- Department of Neurology, MSNI Multiple Sclerosis and Neuro-Immunology, Alfred Health, Melbourne, VIC, Australia
| | - Bianca Devitt
- Department of Oncology, Eastern Health Clinical School, Monash University, Melbourne, VIC, Australia
- Oncology Clinical Trials Unit, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Vilija G Jokubaitis
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
- Department of Neurology, MSNI Multiple Sclerosis and Neuro-Immunology, Alfred Health, Melbourne, VIC, Australia
- Department of Neuroscience, Monash University, The Alfred Centre, Melbourne, VIC, Australia
| |
Collapse
|
41
|
Fridy PC, Rout MP, Ketaren NE. Nanobodies: From High-Throughput Identification to Therapeutic Development. Mol Cell Proteomics 2024; 23:100865. [PMID: 39433212 PMCID: PMC11609455 DOI: 10.1016/j.mcpro.2024.100865] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 10/08/2024] [Accepted: 10/13/2024] [Indexed: 10/23/2024] Open
Abstract
The camelid single-domain antibody fragment, commonly referred to as a nanobody, achieves the targeting power of conventional monoclonal antibodies (mAbs) at only a fraction of their size. Isolated from camelid species (including llamas, alpacas, and camels), their small size at ∼15 kDa, low structural complexity, and high stability compared with conventional antibodies have propelled nanobody technology into the limelight of biologic development. Nanobodies are proving themselves to be a potent complement to traditional mAb therapies, showing success in the treatment of, for example, autoimmune diseases and cancer, and more recently as therapeutic options to treat infectious diseases caused by rapidly evolving biological targets such as the SARS-CoV-2 virus. This review highlights the benefits of applying a proteomic approach to identify diverse nanobody sequences against a single antigen. This proteomic approach coupled with conventional yeast/phage display methods enables the production of highly diverse repertoires of nanobodies able to bind the vast epitope landscape of an antigen, with epitope sampling surpassing that of mAbs. Additionally, we aim to highlight recent findings illuminating the structural attributes of nanobodies that make them particularly amenable to comprehensive antigen sampling and to synergistic activity-underscoring the powerful advantage of acquiring a large, diverse nanobody repertoire against a single antigen. Lastly, we highlight the efforts being made in the clinical development of nanobodies, which have great potential as powerful diagnostic reagents and treatment options, especially when targeting infectious disease agents.
Collapse
Affiliation(s)
- Peter C Fridy
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, New York, USA
| | - Michael P Rout
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, New York, USA
| | - Natalia E Ketaren
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, New York, USA.
| |
Collapse
|
42
|
Li M, Jin M, Peng H, Wang H, Shen Q, Zhang L. Current Status and Future Prospects of TROP-2 ADCs in Lung Cancer Treatment. Drug Des Devel Ther 2024; 18:5005-5021. [PMID: 39525044 PMCID: PMC11550919 DOI: 10.2147/dddt.s489234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Lung cancer is the leading cause of mortality worldwide, and non-small cell lung cancer accounts for the majority of lung cancer cases. Chemotherapy and radiotherapy constitute the mainstays of lung cancer treatment; however, their associated side effects involving the kidneys, nervous system, gastrointestinal tract, and liver further add to dismal outcomes. The advent of antibody‒drug conjugates (ADCs) could change this situation. Trophoblast surface antigen 2 (TROP-2), a human trophoblast surface antigen, is a tumor-associated antigen that is expressed at low levels in normal tissues and is overexpressed in a variety of malignant tumors. The differential expression of the TROP-2 protein in a variety of tumors makes tumor immunotherapy with ADCs targeting TROP-2 a promising approach. Previous studies have shown that the expression of TROP-2 is related to the prognosis of patients with lung cancer and that TROP-2 expression is different across different histological types; however, research on TROP-2 and TROP-2 ADCs in patients with lung cancer is not comprehensive. The aims of this study were to review the mechanism of action and clinical efficacy of TROP-2 and related drugs in the treatment of lung cancer, to elucidate the prognostic value of TROP-2 in lung cancer, and to discuss the future prospects of TROP-2 ADCs to provide a reference for the precise treatment of lung cancer.
Collapse
Affiliation(s)
- Mingyi Li
- Department of Pulmonary and Critical Care Medicine, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Meng Jin
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Hao Peng
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Haitao Wang
- Department of Pulmonary and Critical Care Medicine, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Qian Shen
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Lei Zhang
- Department of Pulmonary and Critical Care Medicine, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
- Xianning Medical College, Hubei University of Science & Technology, Xianning, Hubei, 437000, People’s Republic of China
| |
Collapse
|
43
|
Jiao J, Qian Y, Lv Y, Wei W, Long Y, Guo X, Buerliesi A, Ye J, Han H, Li J, Zhu Y, Zhang W. Overcoming limitations and advancing the therapeutic potential of antibody-oligonucleotide conjugates (AOCs): Current status and future perspectives. Pharmacol Res 2024; 209:107469. [PMID: 39433169 DOI: 10.1016/j.phrs.2024.107469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 10/15/2024] [Accepted: 10/15/2024] [Indexed: 10/23/2024]
Abstract
As cancer incidence rises due to an aging population, the importance of precision medicine continues to grow. Antibody-drug conjugates (ADCs) exemplify targeted therapies by delivering cytotoxic agents to specific antigens. Building on this concept, researchers have developed antibody-oligonucleotide conjugates (AOCs), which combine antibodies with oligonucleotides to regulate gene expression. This review highlights the mechanism of AOCs, emphasizing their unique ability to selectively target and modulate disease-causing proteins. It also explores the components of AOCs and their application in tumor therapy while addressing key challenges such as manufacturing complexities, endosomal escape, and immune response. The article underscores the significance of AOCs in precision oncology and discusses future directions, highlighting their potential in treating cancers driven by genetic mutations and abnormal protein expression.
Collapse
Affiliation(s)
- Jinlan Jiao
- Division of Breast Surgery, Department of General Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, China
| | - Yun Qian
- Dermatologic Surgery Department, Institute of Dermatology, Chinese Academy of Medical Science & Peking Union Medical College, Nanjing 210042, China
| | - Yinhua Lv
- Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210000, China
| | - Wenqian Wei
- Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210000, China
| | - Yongxuan Long
- Division of Breast Surgery, Department of General Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, China
| | - Xiaoling Guo
- Division of Breast Surgery, Department of General Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, China
| | - Anya Buerliesi
- Division of Breast Surgery, Department of General Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, China
| | - Jiahui Ye
- Division of Breast Surgery, Department of General Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, China
| | - Hao Han
- Department of Ultrasound, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Jinbo Li
- State Key Laboratory of Analytical Chemistry for Life Sciences, Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing 210023, China.
| | - Yun Zhu
- Department of Pharmacy, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing 210008, China.
| | - Weijie Zhang
- Division of Breast Surgery, Department of General Surgery, Affiliated Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, China.
| |
Collapse
|
44
|
Goyal A, Murkute SL, Bhowmik S, Prasad CP, Mohapatra P. Belling the "cat": Wnt/β-catenin signaling and its significance in future cancer therapies. Biochim Biophys Acta Rev Cancer 2024; 1879:189195. [PMID: 39413855 DOI: 10.1016/j.bbcan.2024.189195] [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: 05/30/2024] [Revised: 09/15/2024] [Accepted: 10/07/2024] [Indexed: 10/18/2024]
Abstract
The WNT/β-catenin is among one of the most extensively studied cellular signaling pathways involved in the initiation and progression of several deadly cancers. It is now understood that the WNT/β-catenin signaling, during tumor progression operates in a very complex fashion beyond the earlier assumed simple WNT 'On' or 'Off' mode as it recruits numerous WNT ligands, receptors, transcriptional factors and also cross-talks with other signaling molecules including the noncanonical WNT regulators. WNT/β-catenin signaling molecules are often mutated in different cancers which makes them very challenging to inhibit and sometimes ranks them among the undruggable targets. Furthermore, due to the evolutionary conservation of this pathway, inhibiting WNT/β-catenin has caused significant toxicity in normal cells. These challenges are reflected in clinical trial data, where the use of WNT/β-catenin inhibitors as standalone treatments remains limited. In this review, we have highlighted the crucial functional associations of diverse WNT/β-catenin signaling regulators with cancer progression and the phenotypic switching of tumor cells. Next, we have shed light on the roles of WNT/β-catenin signaling in drug resistance, clonal evolution, tumor heterogeneity, and immune evasion. The present review also focuses on various classes of routine and novel WNT/β-catenin therapeutic regimes while addressing the challenges associated with targeting the regulators of this complex pathway. In the light of multiple case studies on WNT/β-catenin inhibitors, we also highlighted the challenges and opportunities for future clinical trial strategies involving these treatments. Additionally, we have proposed strategies for future WNT/β-catenin-based drug discovery trials, emphasizing the potential of combination therapies and AI/ML-driven prediction approaches. Overall, here we showcased the opportunities, possibilities, and potentialities of WNT/β-catenin signaling modulatory therapeutic regimes as promising precision cancer medicines for the future.
Collapse
Affiliation(s)
- Akansha Goyal
- Department of Biotechnology, NIPER Guwahati, Sila Katamur, Changsari, 781101 Kamrup, Assam, India
| | - Satyajit Laxman Murkute
- Department of Biotechnology, NIPER Guwahati, Sila Katamur, Changsari, 781101 Kamrup, Assam, India
| | - Sujoy Bhowmik
- Department of Biotechnology, NIPER Guwahati, Sila Katamur, Changsari, 781101 Kamrup, Assam, India
| | - Chandra Prakash Prasad
- Department of Medical Oncology Lab, DR BRA-IRCH, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Purusottam Mohapatra
- Department of Biotechnology, NIPER Guwahati, Sila Katamur, Changsari, 781101 Kamrup, Assam, India.
| |
Collapse
|
45
|
Rot AE, Hrovatin M, Bokalj B, Lavrih E, Turk B. Cysteine cathepsins: From diagnosis to targeted therapy of cancer. Biochimie 2024; 226:10-28. [PMID: 39245316 DOI: 10.1016/j.biochi.2024.09.001] [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: 03/17/2024] [Revised: 08/23/2024] [Accepted: 09/03/2024] [Indexed: 09/10/2024]
Abstract
Cysteine cathepsins are a fascinating group of proteolytic enzymes that play diverse and crucial roles in numerous biological processes, both in health and disease. Understanding these proteases is essential for uncovering novel insights into the underlying mechanisms of a wide range of disorders, such as cancer. Cysteine cathepsins influence cancer biology by participating in processes such as extracellular matrix degradation, angiogenesis, immune evasion, and apoptosis. In this comprehensive review, we explore foundational research that illuminates the diverse and intricate roles of cysteine cathepsins as diagnostic markers and therapeutic targets for cancer. This review aims to provide valuable insights into the clinical relevance of cysteine cathepsins and explore their capacity to advance personalised and targeted medical interventions in oncology.
Collapse
Affiliation(s)
- Ana Ercegovič Rot
- Jožef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000, Ljubljana, Slovenia; Jožef Stefan International Postgraduate School, Jamova 39, SI-1000, Ljubljana, Slovenia
| | - Matija Hrovatin
- Jožef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000, Ljubljana, Slovenia; Jožef Stefan International Postgraduate School, Jamova 39, SI-1000, Ljubljana, Slovenia
| | - Bor Bokalj
- Jožef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000, Ljubljana, Slovenia; Jožef Stefan International Postgraduate School, Jamova 39, SI-1000, Ljubljana, Slovenia
| | - Ernestina Lavrih
- Jožef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000, Ljubljana, Slovenia; Jožef Stefan International Postgraduate School, Jamova 39, SI-1000, Ljubljana, Slovenia
| | - Boris Turk
- Jožef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000, Ljubljana, Slovenia; Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, SI-1000, Ljubljana, Slovenia.
| |
Collapse
|
46
|
Vasic V, Dickopf S, Spranger N, Rosenberger RS, Fischer M, Mayer K, Larraillet V, Bates JA, Maier V, Sela T, Nussbaum B, Duerr H, Dengl S, Brinkmann U. Generation of binder-format-payload conjugate-matrices by antibody chain-exchange. Nat Commun 2024; 15:9406. [PMID: 39477939 PMCID: PMC11525586 DOI: 10.1038/s41467-024-53730-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 10/21/2024] [Indexed: 11/02/2024] Open
Abstract
The generation of antibody-drug conjugates with optimal functionality depends on many parameters. These include binder epitope, antibody format, linker composition, conjugation site(s), drug-to-antibody ratio, and conjugation method. The production of matrices that cover all possible parameters is a major challenge in identifying optimal antibody-drug conjugates. To address this bottleneck, we adapted our Format Chain Exchange technology (FORCE), originally established for bispecific antibodies, toward the generation of binder-format-payload matrices (pair-FORCE). Antibody derivatives with exchange-enabled Fc-heterodimers are combined with payload-conjugated Fc donors, and subsequent chain-exchange transfers payloads to antibody derivatives in different formats. The resulting binder-format-conjugate matrices can be generated with cytotoxic payloads, dyes, haptens, and large molecules, resulting in versatile tools for ADC screening campaigns. We show the relevance of pair-FORCE for identifying optimal HER2-targeting antibody-drug conjugates. Analysis of this matrix reveals that the notion of format-defines-function applies not only to bispecific antibodies, but also to antibody-drug conjugates.
Collapse
Affiliation(s)
- Vedran Vasic
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, Penzberg, Germany
| | - Steffen Dickopf
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, Penzberg, Germany
- Veraxa Biotech, Heidelberg, Germany
| | - Nadine Spranger
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, Penzberg, Germany
- Institute of Molecular Immunology, School of Medicine and Health, Technical University Munich (TUM), Munich, Germany
| | - Rose-Sophie Rosenberger
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, Penzberg, Germany
| | - Michaela Fischer
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, Penzberg, Germany
| | - Klaus Mayer
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, Penzberg, Germany
| | - Vincent Larraillet
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, Penzberg, Germany
| | - Jack A Bates
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, Penzberg, Germany
| | - Verena Maier
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, Penzberg, Germany
| | - Tatjana Sela
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, Penzberg, Germany
| | - Bianca Nussbaum
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, Penzberg, Germany
| | - Harald Duerr
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, Penzberg, Germany
| | - Stefan Dengl
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, Penzberg, Germany
| | - Ulrich Brinkmann
- Roche Pharma Research and Early Development (pRED), Large Molecule Research (LMR), Roche Innovation Center Munich, Penzberg, Germany.
| |
Collapse
|
47
|
Masnikosa R, Cvetković Z, Pirić D. Tumor Biology Hides Novel Therapeutic Approaches to Diffuse Large B-Cell Lymphoma: A Narrative Review. Int J Mol Sci 2024; 25:11384. [PMID: 39518937 PMCID: PMC11545713 DOI: 10.3390/ijms252111384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 10/13/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is a malignancy of immense biological and clinical heterogeneity. Based on the transcriptomic or genomic approach, several different classification schemes have evolved over the years to subdivide DLBCL into clinically (prognostically) relevant subsets, but each leaves unclassified samples. Herein, we outline the DLBCL tumor biology behind the actual and potential drug targets and address the challenges and drawbacks coupled with their (potential) use. Therapeutic modalities are discussed, including small-molecule inhibitors, naked antibodies, antibody-drug conjugates, chimeric antigen receptors, bispecific antibodies and T-cell engagers, and immune checkpoint inhibitors. Candidate drugs explored in ongoing clinical trials are coupled with diverse toxicity issues and refractoriness to drugs. According to the literature on DLBCL, the promise for new therapeutic targets lies in epigenetic alterations, B-cell receptor and NF-κB pathways. Herein, we present putative targets hiding in lipid pathways, ferroptosis, and the gut microbiome that could be used in addition to immuno-chemotherapy to improve the general health status of DLBCL patients, thus increasing the chance of being cured. It may be time to devote more effort to exploring DLBCL metabolism to discover novel druggable targets. We also performed a bibliometric and knowledge-map analysis of the literature on DLBCL published from 2014-2023.
Collapse
Affiliation(s)
- Romana Masnikosa
- Department of Physical Chemistry, Vinca Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia;
| | - Zorica Cvetković
- Department of Hematology, Clinical Hospital Centre Zemun, Vukova 9, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Dr Subotića 8, 11000 Belgrade, Serbia
| | - David Pirić
- Department of Physical Chemistry, Vinca Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia;
| |
Collapse
|
48
|
Mukherjee A, Bandyopadhyay D. Targeted Therapy in Breast Cancer: Advantages and Advancements of Antibody-Drug Conjugates, a Type of Chemo-Biologic Hybrid Drugs. Cancers (Basel) 2024; 16:3517. [PMID: 39456611 PMCID: PMC11505910 DOI: 10.3390/cancers16203517] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 10/08/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
Cancer is a significant health challenge globally, with millions of people affected every year, resulting in high morbidity and mortality. Although other treatment options are available with limitations, chemotherapy, either standalone or combined with other therapeutic procedures, is the most commonly used practice of treating cancer. In chemotherapy, cancer cells/malignant tumors are targeted; however, due to less target specificity, along with malignant cells, normal cells are also affected, which leads to various off-target effects (side effects) that impact the patient quality of life. Out of all the different types of cancers, breast cancer is the most common type of cancer in humans worldwide. Current anticancer drug discovery research aims to develop therapeutics with higher potency and lower toxicity, which is only possible through target-specific therapy. Antibody-drug conjugates (ADCs) are explicitly designed to target malignant tumors and minimize off-target effects by reducing systemic cytotoxicity. Several ADCs have been approved for clinical use and have shown moderate to good efficacy so far. Considering various aspects, chemotherapy and ADCs are useful in treating cancer. However, ADCs provide a more focused and less toxic approach, which is especially helpful in cases where resistance to chemotherapy (drug resistance) occurs and in the type of malignancies in which specific antigens are overexpressed. Ongoing ADC research aims to develop more target-specific cancer treatments. In short, this study presents a concise overview of ADCs specific to breast cancer treatment. This study provides insight into the classifications, mechanisms of action, structural aspects, and clinical trial phases (current status) of these chemo-biologic drugs (ADCs).
Collapse
Affiliation(s)
- Attrayo Mukherjee
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Patia, Bhubaneswar 751024, Odisha, India;
| | - Debasish Bandyopadhyay
- School of Integrative Biological and Chemical Sciences (SIBCS), University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
- School of Earth, Environmental, and Marine Sciences (SEEMS), University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
| |
Collapse
|
49
|
Su Z, Yin S, Wu Y. Rationalize the Functional Roles of Protein-Protein Interactions in Targeted Protein Degradation by Kinetic Monte-Carlo Simulations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.26.615190. [PMID: 39386564 PMCID: PMC11463391 DOI: 10.1101/2024.09.26.615190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Targeted protein degradation is a promising therapeutic strategy to tackle disease-causing proteins that lack binding pockets for traditional small-molecule inhibitors. Its first step is to trigger the proximity between a ubiquitin ligase complex and a target protein through a heterobifunctional molecule, such as proteolysis targeting chimeras (PROTACs), leading to the formation of a ternary complex. The properties of protein-protein interactions play an important regulatory role during this process, which can be reflected by binding cooperativity. Unfortunately, although computer-aided drug design has become a cornerstone of modern drug development, the endeavor to model targeted protein degradation is still in its infancy. The development of computational tools to understand the impacts of protein-protein interactions on targeted protein degradation, therefore, is highly demanded. To reach this goal, we constructed a non-redundant structural benchmark of the most updated ternary complexes and applied a kinetic Monte-Carlo method to simulate the association between ligases and PROTAC-targeted proteins in the benchmark. Our results show that proteins in most complexes with positive cooperativity tend to associate into native-like configurations more often. In contrast, proteins very likely failed to associate into native-like configurations in complexes with negative cooperativity. Moreover, we compared the protein-protein association through different interfaces generated from molecular docking. The native-like binding interface shows a higher association probability than all the other alternative interfaces only in the complex with positive cooperativity. These observations support the idea that the formation of ternary complexes is closely regulated by the binary interactions between proteins. Finally, we applied our method to cyclin-dependent kinases 4 and 6 (CDK4/6). We found that their interactions with the ligase are not as similar as their structures. Altogether, our study paves the way for understanding the role of protein-protein interactions in PROTACE-induced ternary complex formation. It can potentially help in searching for degraders that selectively target specific proteins.
Collapse
Affiliation(s)
- Zhaoqian Su
- Data Science Institute, Vanderbilt University, 1001 19th Ave S, Nashville, TN, 37212
| | - Shanye Yin
- Department of Pathology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461
| | - Yinghao Wu
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461
| |
Collapse
|
50
|
Ghemrawi R, Abuamer L, Kremesh S, Hussien G, Ahmed R, Mousa W, Khoder G, Khair M. Revolutionizing Cancer Treatment: Recent Advances in Immunotherapy. Biomedicines 2024; 12:2158. [PMID: 39335671 PMCID: PMC11429153 DOI: 10.3390/biomedicines12092158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 09/20/2024] [Accepted: 09/21/2024] [Indexed: 09/30/2024] Open
Abstract
Cancer immunotherapy has emerged as a transformative approach in oncology, utilizing the body's immune system to specifically target and destroy malignant cells. This review explores the scope and impact of various immunotherapeutic strategies, including monoclonal antibodies, chimeric antigen receptor (CAR)-T cell therapy, checkpoint inhibitors, cytokine therapy, and therapeutic vaccines. Monoclonal antibodies, such as Rituximab and Trastuzumab, have revolutionized treatment paradigms for lymphoma and breast cancer by offering targeted interventions that reduce off-target effects. CAR-T cell therapy presents a potentially curative option for refractory hematologic malignancies, although challenges remain in effectively treating solid tumors. Checkpoint inhibitors have redefined the management of cancers like melanoma and lung cancer; however, managing immune-related adverse events and ensuring durable responses are critical areas of focus. Cytokine therapy continues to play a vital role in modulating the immune response, with advancements in cytokine engineering improving specificity and reducing systemic toxicity. Therapeutic vaccines, particularly mRNA-based vaccines, represent a frontier in personalized cancer treatment, aiming to generate robust, long-lasting immune responses against tumor-specific antigens. Despite these advancements, the field faces significant challenges, including immune resistance, tumor heterogeneity, and the immunosuppressive tumor microenvironment. Future research should address these obstacles through emerging technologies, such as next-generation antibodies, Clustered Regularly Interspaced Short Palindromic Repeat (CRISPR)-based gene editing, and AI-driven drug discovery. By integrating these novel approaches, cancer immunotherapy holds the promise of offering more durable, less toxic, and highly personalized treatment options, ultimately improving patient outcomes and survival rates.
Collapse
Affiliation(s)
- Rose Ghemrawi
- College of Pharmacy, Al Ain University, Abu Dhabi P.O. Box 112612, United Arab Emirates
- AAU Health and Biomedical Research Center, Al Ain University, Abu Dhabi P.O. Box 112612, United Arab Emirates
| | - Lama Abuamer
- College of Pharmacy, Al Ain University, Abu Dhabi P.O. Box 112612, United Arab Emirates
- AAU Health and Biomedical Research Center, Al Ain University, Abu Dhabi P.O. Box 112612, United Arab Emirates
| | - Sedra Kremesh
- College of Pharmacy, Al Ain University, Abu Dhabi P.O. Box 112612, United Arab Emirates
- AAU Health and Biomedical Research Center, Al Ain University, Abu Dhabi P.O. Box 112612, United Arab Emirates
| | - Ghadeer Hussien
- College of Pharmacy, Al Ain University, Abu Dhabi P.O. Box 112612, United Arab Emirates
- AAU Health and Biomedical Research Center, Al Ain University, Abu Dhabi P.O. Box 112612, United Arab Emirates
| | - Rahaf Ahmed
- College of Pharmacy, Al Ain University, Abu Dhabi P.O. Box 112612, United Arab Emirates
- AAU Health and Biomedical Research Center, Al Ain University, Abu Dhabi P.O. Box 112612, United Arab Emirates
| | - Walaa Mousa
- College of Pharmacy, Al Ain University, Abu Dhabi P.O. Box 112612, United Arab Emirates
- AAU Health and Biomedical Research Center, Al Ain University, Abu Dhabi P.O. Box 112612, United Arab Emirates
| | - Ghalia Khoder
- Department of Pharmaceutics and Pharmaceuticals Technology, College of Pharmacy, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
| | - Mostafa Khair
- Core Technology Platforms, New York University Abu Dhabi, Abu Dhabi P.O. Box 129188, United Arab Emirates
| |
Collapse
|