1
|
Zhou KXT, Bujold KE. The Emergence of Oligonucleotide Building Blocks in the Multispecific Proximity-Inducing Drug Toolbox of Destruction. ACS Chem Biol 2025; 20:3-18. [PMID: 39704048 DOI: 10.1021/acschembio.4c00311] [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: 12/21/2024]
Abstract
Oligonucleotides are a rapidly emerging class of therapeutics. Their most well-known examples are informational drugs that modify gene expression by binding mRNA. Despite inducing proximity between biological machinery and mRNA when applied to modulating gene expression, oligonucleotides are not typically labeled as "proximity-inducing" in literature. Yet, they have recently been explored as building blocks for multispecific proximity-inducing drugs (MPIDs). MPIDs are unique because they can direct endogenous biological machinery to destroy targeted molecules and cells, in contrast to traditional drugs that inhibit only their functions. The unique mechanism of action of MPIDs has enabled the targeting of previously "undruggable" molecular entities that cannot be effectively inhibited. However, the development of MPIDs must ensure that these molecules will selectively direct a potent, destruction-based mechanism of action toward intended targets over healthy tissues to avoid causing life-threatening toxicities. Oligonucleotides have emerged as promising building blocks for the design of MPIDs because they are sequence-controlled molecules that can be rationally designed to program multispecific binding interactions. In this Review, we examine the emergence of oligonucleotide-containing MPIDs in the proximity induction space, which has been dominated by antibody and small molecule MPID modalities. Moreover, examples of oligonucleotides developed as MPID candidates in immunotherapy and protein degradation are discussed to demonstrate the utility of oligonucleotides in expanding the scope and selectivity of the MPID toolbox. Finally, we discuss the utility of programming "AND" gates into oligonucleotide scaffolds to encode conditional responses that have the potential to be incorporated into MPIDs, which can further enhance their selectivity, thus increasing the scope of this drug category.
Collapse
Affiliation(s)
- Kevin Xiao Tong Zhou
- Department of Chemistry & Chemical Biology, McMaster University, 1280 Main Street West, Hamilton, ONL8S 4M1, Canada
| | - Katherine E Bujold
- Department of Chemistry & Chemical Biology, McMaster University, 1280 Main Street West, Hamilton, ONL8S 4M1, Canada
| |
Collapse
|
2
|
Blay V, Pandiella A. Strategies to boost antibody selectivity in oncology. Trends Pharmacol Sci 2024; 45:1135-1149. [PMID: 39609227 DOI: 10.1016/j.tips.2024.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/03/2024] [Accepted: 10/18/2024] [Indexed: 11/30/2024]
Abstract
Antibodies in oncology are being equipped with toxic cargoes and effector functions that can kill cells at very low concentrations. A key challenge is that most targets on cancer cells are also present on at least some healthy cells. Shared targets can result in off-tumor binding and compromise the safety and potential of therapeutic candidates. In this review, we survey strategies that can help direct biologics to cancer sites more selectively. These strategies are becoming increasingly feasible thanks to advances in molecular design and engineering. The objective is to create therapeutics that exploit changes in cancer and leverage the human body infrastructure, enabling therapeutics that discriminate not just self from non-self but diseased from healthy tissue.
Collapse
Affiliation(s)
- Vincent Blay
- University of California Santa Cruz, Department of Microbiology and Environmental Toxicology, Santa Cruz, CA 95064, USA.
| | - Atanasio Pandiella
- Centro de Investigación del Cáncer, CIBERONC and IBSAL, 37007 Salamanca, Spain
| |
Collapse
|
3
|
Sima Y, Ai L, Wang L, Zhang P, Zhang Q, Wu S, Xie S, Zhao Z, Tan W. A DNA Molecular Logic Circuit for Precise Tumor Identification. NANO LETTERS 2024; 24:12070-12079. [PMID: 39315658 DOI: 10.1021/acs.nanolett.4c02342] [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: 09/25/2024]
Abstract
Tumor-associated antigens (TAAs) are not exclusively expressed in cancer cells, inevitably causing the "on target, off tumor" effect of molecular recognition tools. To achieve precise recognition of cancer cells, by using protein tyrosine kinase 7 (PTK7) as a model TAA, a DNA molecular logic circuit Aisgc8 was rationally developed by arranging H+-binding i-motif, ATP-binding aptamer, and PTK7-targeting aptamer Sgc8c in a DNA sequence. Aisgc8 output the conformation of Sgc8c to recognize PTK7 on cells in a simulated tumor microenvironment characterized by weak acidity and abundant ATP, but not in a simulated physiological environment. Through in vitro and in vivo results, Aisgc8 demonstrated its ability to precisely recognize cancer cells and, as a result, displayed excellent performance in tumor imaging. Thus, our studies produced a simple and efficient strategy to construct DNA logic circuits, opening new possibilities to develop convenient and intelligent precision diagnostics by using DNA logic circuits.
Collapse
Affiliation(s)
- Yingyu Sima
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Lili Ai
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Linlin Wang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Pengge Zhang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Qiang Zhang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Shanchao Wu
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Sitao Xie
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Zilong Zhao
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Weihong Tan
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| |
Collapse
|
4
|
Liu J, Zhu J. Progresses of T-cell-engaging bispecific antibodies in treatment of solid tumors. Int Immunopharmacol 2024; 138:112609. [PMID: 38971103 DOI: 10.1016/j.intimp.2024.112609] [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: 05/27/2024] [Revised: 06/28/2024] [Accepted: 06/29/2024] [Indexed: 07/08/2024]
Abstract
T-cell-engaging bispecific antibody (TCB) therapies have emerged as a promising immunotherapeutic approach, effectively redirecting effector T cells to selectively eliminate tumor cells. The therapeutic potential of TCBs has been well recognized, particularly with the approval of multiple TCBs in recent years for the treatment of hematologic malignancies as well as some solid tumors. However, TCBs encounter multiple challenges in treating solid tumors, such as on-target off-tumor toxicity, cytokine release syndrome (CRS), and T cell dysfunction within the immunosuppressive tumor microenvironment, all of which may impact their therapeutic efficacy. In this review, we summarize clinical data on TCBs for solid tumor treatment, highlight the challenges faced, and discuss potential solutions based on emerging strategies from current clinical and preclinical research. These solutions include TCB structural optimization, target selection, and combination strategies. This comprehensive analysis aims to guide the development of TCBs from design to clinical application, addressing the evolving landscape of cancer immunotherapy.
Collapse
Affiliation(s)
- Junjun Liu
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jianwei Zhu
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; Jecho Laboratories, Inc., Frederick, MD 21704, USA.
| |
Collapse
|
5
|
Zhao C, Zhang R, Yang H, Gao Y, Zou Y, Zhang X. Antibody-drug conjugates for non-small cell lung cancer: Advantages and challenges in clinical translation. Biochem Pharmacol 2024; 226:116378. [PMID: 38908529 DOI: 10.1016/j.bcp.2024.116378] [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: 04/07/2024] [Revised: 06/03/2024] [Accepted: 06/18/2024] [Indexed: 06/24/2024]
Abstract
Lung cancer is the leading cause of cancer death, with non-small cell lung cancer (NSCLC) accounting for approximately 85 % of all lung cancers and having a poor treatment and prognosis. Conventional clinical chemotherapy and immunotherapy are challenged by systemic toxicity and drug resistance, so researchers are increasingly focusing on antibody-drug conjugate (ADC), an innovative concept combining chemotherapy and targeted therapy, in which a drug selectively binds to antigens on the surface of a tumor cell via antibodies, which internalize the ADC, and then transfers the ADC to the lysosome via the endosomes to degrade the drug and kill the tumor cell. Despite the promising nature of ADCs, no ADC product for any indication including NSCLC has been approved for marketing by the FDA to date. In this review, we summarize the main advantages of ADCs and discuss in depth the design of the most desirable ADCs for NSCLC therapy. In addition to preclinical studies, we focus on the current state of clinical research on ADCs as interventions for the treatment of NSCLC by summarizing real-time clinical trial data from ClinicalTrials.gov, and reasonably speculate on the direction of the design of future generations of ADCs.
Collapse
Affiliation(s)
- Chenyu Zhao
- Department of China Medical University, The Queen's University of Belfast Joint College, School of Pharmacy, China Medical University, Shenyang 110122, China; School of Pharmacy, Queen's University Belfast, Belfast BT9 7BL, UK
| | - Ruihan Zhang
- Department of China Medical University, The Queen's University of Belfast Joint College, School of Pharmacy, China Medical University, Shenyang 110122, China; School of Pharmacy, Queen's University Belfast, Belfast BT9 7BL, UK
| | - Huazhe Yang
- School of Intelligent Medicine, China Medical University, Shenyang 110122, China
| | - Yiwei Gao
- Department of China Medical University, The Queen's University of Belfast Joint College, School of Pharmacy, China Medical University, Shenyang 110122, China; School of Pharmacy, Queen's University Belfast, Belfast BT9 7BL, UK
| | - Ying Zou
- Department of Rehabilitation Centre, Shengjing Hospital of China Medical University, Shenyang 110122, Liaoning, China.
| | - Xudong Zhang
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang 110000, Liaoning, China.
| |
Collapse
|
6
|
Fenis A, Demaria O, Gauthier L, Vivier E, Narni-Mancinelli E. New immune cell engagers for cancer immunotherapy. Nat Rev Immunol 2024; 24:471-486. [PMID: 38273127 DOI: 10.1038/s41577-023-00982-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2023] [Indexed: 01/27/2024]
Abstract
There have been major advances in the immunotherapy of cancer in recent years, including the development of T cell engagers - antibodies engineered to redirect T cells to recognize and kill cancer cells - for the treatment of haematological malignancies. However, the field still faces several challenges to develop agents that are consistently effective in a majority of patients and cancer types, such as optimizing drug dose, overcoming treatment resistance and improving efficacy in solid tumours. A new generation of T cell-targeted molecules was developed to tackle these issues that are potentially more effective and safer. In addition, agents designed to engage the antitumour activities of other immune cells, including natural killer cells and myeloid cells, are showing promise and have the potential to treat a broader range of cancers.
Collapse
Affiliation(s)
- Aurore Fenis
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
- Aix Marseille Université, Centre National de la Recherche Scientifique, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Olivier Demaria
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Laurent Gauthier
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
| | - Eric Vivier
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
- Aix Marseille Université, Centre National de la Recherche Scientifique, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
- Assistance Publique-Hôpitaux de Marseille, Hôpital de la Timone, Marseille Immunopôle, Marseille, France
| | - Emilie Narni-Mancinelli
- Aix Marseille Université, Centre National de la Recherche Scientifique, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France.
| |
Collapse
|
7
|
Hoff CO, Dal Lago EA, Siqueira JM, de Sousa LG, El-Naggar AK, Ahnert JR, Ferrarotto R. First Use of AXL Targeting in Metastatic, Refractory, Adenoid Cystic Carcinoma: A Case Report. JCO Precis Oncol 2024; 8:e2300633. [PMID: 38579194 PMCID: PMC11018191 DOI: 10.1200/po.23.00633] [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: 11/16/2023] [Revised: 01/14/2024] [Accepted: 02/14/2024] [Indexed: 04/07/2024] Open
Abstract
First use of AXL-targeting in adenoid cystic carcinoma (ACC); with positive results, ACC now included in AXL studies.
Collapse
Affiliation(s)
- Camilla O. Hoff
- Department of Thoracic Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Juliana Mota Siqueira
- Department of Thoracic Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Luana G. de Sousa
- Department of Thoracic Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Adel K. El-Naggar
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jordi Rodon Ahnert
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Renata Ferrarotto
- Department of Thoracic Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
8
|
Tsuchikama K, Anami Y, Ha SYY, Yamazaki CM. Exploring the next generation of antibody-drug conjugates. Nat Rev Clin Oncol 2024; 21:203-223. [PMID: 38191923 DOI: 10.1038/s41571-023-00850-2] [Citation(s) in RCA: 68] [Impact Index Per Article: 68.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2023] [Indexed: 01/10/2024]
Abstract
Antibody-drug conjugates (ADCs) are a promising cancer treatment modality that enables the selective delivery of highly cytotoxic payloads to tumours. However, realizing the full potential of this platform necessitates innovative molecular designs to tackle several clinical challenges such as drug resistance, tumour heterogeneity and treatment-related adverse effects. Several emerging ADC formats exist, including bispecific ADCs, conditionally active ADCs (also known as probody-drug conjugates), immune-stimulating ADCs, protein-degrader ADCs and dual-drug ADCs, and each offers unique capabilities for tackling these various challenges. For example, probody-drug conjugates can enhance tumour specificity, whereas bispecific ADCs and dual-drug ADCs can address resistance and heterogeneity with enhanced activity. The incorporation of immune-stimulating and protein-degrader ADCs, which have distinct mechanisms of action, into existing treatment strategies could enable multimodal cancer treatment. Despite the promising outlook, the importance of patient stratification and biomarker identification cannot be overstated for these emerging ADCs, as these factors are crucial to identify patients who are most likely to derive benefit. As we continue to deepen our understanding of tumour biology and refine ADC design, we will edge closer to developing truly effective and safe ADCs for patients with treatment-refractory cancers. In this Review, we highlight advances in each ADC component (the monoclonal antibody, payload, linker and conjugation chemistry) and provide more-detailed discussions on selected examples of emerging novel ADCs of each format, enabled by engineering of one or more of these components.
Collapse
Affiliation(s)
- Kyoji Tsuchikama
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Yasuaki Anami
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Summer Y Y Ha
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Chisato M Yamazaki
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
9
|
Frey G, Cugnetti APG, Liu H, Xing C, Wheeler C, Chang HW, Boyle WJ, Short JM. A novel conditional active biologic anti-EpCAM x anti-CD3 bispecific antibody with synergistic tumor selectivity for cancer immunotherapy. MAbs 2024; 16:2322562. [PMID: 38445633 PMCID: PMC10936661 DOI: 10.1080/19420862.2024.2322562] [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: 11/08/2023] [Accepted: 02/20/2024] [Indexed: 03/07/2024] Open
Abstract
Epithelial cell adhesion molecule (EpCAM) is a transmembrane glycoprotein that plays several roles in cancer biology. EpCAM is an attractive therapeutic target because of its expression in most solid tumors. However, targeting EpCAM has been challenging because it is also highly expressed in normal epithelial tissues. Initial attempts to develop EpCAM-specific T-cell engagers were unsuccessful due to severe cytokine release effects, as well as serious on-target, off-tumor drug-related toxicities. We developed novel, conditionally active biological (CAB) bispecific antibodies that bind to both EpCAM and CD3 in an acidic tumor microenvironment. In healthy tissues, binding to EpCAM and CD3 is greatly reduced by a novel, dual CAB selection, where each binding domain is independently blocked by the presence of physiological chemicals known as Protein-associated Chemical Switches (PaCS). The CAB anti-EpCAM T-cell engagers displayed the anticipated bispecific binding properties and mediated the potent lysis of EpCAM-positive cancer cell lines through the recruitment of T cells in the tumor microenvironment. Xenograft studies showed that the efficacy of CAB bispecific antibodies is similar to that of a non-CAB anti-EpCAM bispecific antibody, but they have markedly reduced toxicity in non-human primates, indicating an unprecedentedly widened therapeutic index of over 100-fold. These preclinical results indicate that the dual CAB bispecific antibody is potentially both a powerful and safe therapeutic platform and a promising T cell-engaging treatment for patients with EpCAM-expressing tumors.
Collapse
Affiliation(s)
- Gerhard Frey
- Research & Development, BioAtla Inc, San Diego, CA, USA
| | | | - Haizhen Liu
- Research & Development, BioAtla Inc, San Diego, CA, USA
| | - Charles Xing
- Research & Development, BioAtla Inc, San Diego, CA, USA
| | | | | | | | - Jay M. Short
- Research & Development, BioAtla Inc, San Diego, CA, USA
| |
Collapse
|
10
|
DeRyckere D, Huelse JM, Earp HS, Graham DK. TAM family kinases as therapeutic targets at the interface of cancer and immunity. Nat Rev Clin Oncol 2023; 20:755-779. [PMID: 37667010 DOI: 10.1038/s41571-023-00813-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/07/2023] [Indexed: 09/06/2023]
Abstract
Novel treatment approaches are needed to overcome innate and acquired mechanisms of resistance to current anticancer therapies in cancer cells and the tumour immune microenvironment. The TAM (TYRO3, AXL and MERTK) family receptor tyrosine kinases (RTKs) are potential therapeutic targets in a wide range of cancers. In cancer cells, TAM RTKs activate signalling pathways that promote cell survival, metastasis and resistance to a variety of chemotherapeutic agents and targeted therapies. TAM RTKs also function in innate immune cells, contributing to various mechanisms that suppress antitumour immunity and promote resistance to immune-checkpoint inhibitors. Therefore, TAM antagonists provide an unprecedented opportunity for both direct and immune-mediated therapeutic activity provided by inhibition of a single target, and are likely to be particularly effective when used in combination with other cancer therapies. To exploit this potential, a variety of agents have been designed to selectively target TAM RTKs, many of which have now entered clinical testing. This Review provides an essential guide to the TAM RTKs for clinicians, including an overview of the rationale for therapeutic targeting of TAM RTKs in cancer cells and the tumour immune microenvironment, a description of the current preclinical and clinical experience with TAM inhibitors, and a perspective on strategies for continued development of TAM-targeted agents for oncology applications.
Collapse
Affiliation(s)
- Deborah DeRyckere
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA
- Department of Paediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Justus M Huelse
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA
- Department of Paediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - H Shelton Earp
- Department of Medicine, UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Douglas K Graham
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA.
- Department of Paediatrics, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
11
|
Xu W, Chen H, Duan Y, Ge D, Ren D, Zhao S, Liu Y, Ma L, Zhang C. Bacterial Expression of Promelittin for Safe and Effective Tumor Therapy. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 37897422 DOI: 10.1021/acsami.3c11457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/30/2023]
Abstract
Bacteria-based tumor therapy has attracted much attention due to its unique mechanism and abundant application. With the rapid development of synthetic biology, utilizing gene technology to make bacteria express therapeutic agents has greatly innovated bacterial therapy paradigms. Herein, we constructed an Escherichia coli expressing promelittin protein system based on the Trojan horse strategy, which limited the toxicity of melittin through the fusion protein during melittin expression. After targeted colonization of bacteria in tumor tissues, promelittin was activated by matrix metalloproteinase, followed by causing tumor cell death through a membrane-lytic mechanism. Additionally, the released cytolytic melittin in turn killed the maternal bacteria, eliminating safety hazards and triggering host immunity. Detailed experiments revealed that the bacteria expressing the promelittin system could significantly inhibit the proliferation and metastasis of primitive tumors in a CT26-bearing mice model. This study sheds insights into the development of bacteria-based synergistic tumor therapy.
Collapse
Affiliation(s)
- Wenxuan Xu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan 430062, P. R. China
| | - Huanhuan Chen
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan 430062, P. R. China
| | - Yamin Duan
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan 430062, P. R. China
| | - Dejie Ge
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan 430062, P. R. China
| | - Debao Ren
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan 430062, P. R. China
| | - Shuai Zhao
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan 430062, P. R. China
| | - Yang Liu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan 430062, P. R. China
| | - Lixin Ma
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan 430062, P. R. China
| | - Cheng Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan 430062, P. R. China
| |
Collapse
|
12
|
Smith FD, Pierce RH, Thisted T, van der Horst EH. Conditionally Active, pH-Sensitive Immunoregulatory Antibodies Targeting VISTA and CTLA-4 Lead an Emerging Class of Cancer Therapeutics. Antibodies (Basel) 2023; 12:55. [PMID: 37753969 PMCID: PMC10525963 DOI: 10.3390/antib12030055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/24/2023] [Accepted: 08/28/2023] [Indexed: 09/28/2023] Open
Abstract
Immune checkpoints and other immunoregulatory targets can be difficult to precisely target due to expression on non-tumor immune cells critical to maintaining immune homeostasis in healthy tissues. On-target/off-tumor binding of therapeutics results in significant pharmacokinetic and pharmacodynamic problems. Target-mediated drug disposition (TMDD) significantly limits effective intratumoral drug levels and adversely affects anti-tumor efficacy. Target engagement outside the tumor environment may lead to severe immune-related adverse events (irAEs), resulting in a narrowing of the therapeutic window, sub-optimal dosing, or cessation of drug development altogether. Overcoming these challenges has become tractable through recent advances in antibody engineering and screening approaches. Here, we review the discovery and development of conditionally active antibodies with minimal binding to target at physiologic pH but high-affinity target binding at the low pH of the tumor microenvironment by focusing on the discovery and improved properties of pH-dependent mAbs targeting two T cell checkpoints, VISTA and CTLA-4.
Collapse
Affiliation(s)
- F. Donelson Smith
- Sensei Biotherapeutics, Inc., 1405 Research Blvd., Suite 125, Rockville, MD 20850, USA;
| | | | - Thomas Thisted
- Sensei Biotherapeutics, Inc., 1405 Research Blvd., Suite 125, Rockville, MD 20850, USA;
| | | |
Collapse
|
13
|
Sun Q, Hong Z, Zhang C, Wang L, Han Z, Ma D. Immune checkpoint therapy for solid tumours: clinical dilemmas and future trends. Signal Transduct Target Ther 2023; 8:320. [PMID: 37635168 PMCID: PMC10460796 DOI: 10.1038/s41392-023-01522-4] [Citation(s) in RCA: 132] [Impact Index Per Article: 66.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 05/11/2023] [Accepted: 05/28/2023] [Indexed: 08/29/2023] Open
Abstract
Immune-checkpoint inhibitors (ICBs), in addition to targeting CTLA-4, PD-1, and PD-L1, novel targeting LAG-3 drugs have also been approved in clinical application. With the widespread use of the drug, we must deeply analyze the dilemma of the agents and seek a breakthrough in the treatment prospect. Over the past decades, these agents have demonstrated dramatic efficacy, especially in patients with melanoma and non-small cell lung cancer (NSCLC). Nonetheless, in the field of a broad concept of solid tumours, non-specific indications, inseparable immune response and side effects, unconfirmed progressive disease, and complex regulatory networks of immune resistance are four barriers that limit its widespread application. Fortunately, the successful clinical trials of novel ICB agents and combination therapies, the advent of the era of oncolytic virus gene editing, and the breakthrough of the technical barriers of mRNA vaccines and nano-delivery systems have made remarkable breakthroughs currently. In this review, we enumerate the mechanisms of each immune checkpoint targets, associations between ICB with tumour mutation burden, key immune regulatory or resistance signalling pathways, the specific clinical evidence of the efficacy of classical targets and new targets among different tumour types and put forward dialectical thoughts on drug safety. Finally, we discuss the importance of accurate triage of ICB based on recent advances in predictive biomarkers and diagnostic testing techniques.
Collapse
Affiliation(s)
- Qian Sun
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Zhenya Hong
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Cong Zhang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Liangliang Wang
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Zhiqiang Han
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| | - Ding Ma
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| |
Collapse
|
14
|
Immunotherapeutic Approaches in Ovarian Cancer. Curr Issues Mol Biol 2023; 45:1233-1249. [PMID: 36826026 PMCID: PMC9955550 DOI: 10.3390/cimb45020081] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/27/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Ovarian cancer (OC) is gynecological cancer, and diagnosis and treatment are continuously advancing. Next-generation sequencing (NGS)-based diagnoses have emerged as novel methods for identifying molecules and pathways in cancer research. The NGS-based applications have expanded in OC research for early detection and identification of aberrant genes and dysregulation pathways, demonstrating comprehensive views of the entire transcriptome, such as fusion genes, genetic mutations, and gene expression profiling. Coinciding with advances in NGS-based diagnosis, treatment strategies for OC, such as molecular targeted therapy and immunotherapy, have also advanced. Immunotherapy is effective against many other cancers, and its efficacy against OC has also been demonstrated at the clinical phase. In this review, we describe several NGS-based applications for therapeutic targets of OC, and introduce current immunotherapeutic strategies, including vaccines, checkpoint inhibitors, and chimeric antigen receptor (CAR)-T cell transplantation, for effective diagnosis and treatment of OC.
Collapse
|
15
|
Fan J, Pung E, Lin Y, Wang Q. Recent development of hydrogen sulfide-releasing biomaterials as novel therapies: a narrative review. BIOMATERIALS TRANSLATIONAL 2022; 3:250-263. [PMID: 36846507 PMCID: PMC9947736 DOI: 10.12336/biomatertransl.2022.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/09/2022] [Accepted: 12/20/2022] [Indexed: 06/18/2023]
Abstract
Hydrogen sulfide (H2S) has been reported as an endogenous gasotransmitter that contributes to the modulation of a myriad of biological signalling pathways, which includes maintaining homeostasis in living organisms at physiological concentrations, controlling protein sulfhydration and persulfidation for signalling processes, mediating neurodegeneration, and regulating inflammation and innate immunity, etc. As a result, researchers are actively exploring effective approaches to evaluate the properties and the distribution of H2S in vivo. Furthermore, the regulation of the physiological conditions of H2S in vivo introduces the opportunity to further study the molecular mechanisms by which H2S regulates cellular functions. In recent years, many H2S-releasing compounds and biomaterials that can deliver H2S to various body systems have been developed to provide sustained and stable H2S delivery. Additionally, various designs of these H2S-releasing biomaterials have been proposed to aid in the normal conduction of physiological processes, such as cardioprotection and wound healing, by modulating different signalling pathways and cell functionalities. Using biomaterials as a platform to control the delivery of H2S introduces the opportunity to fine tune the physiological concentration of H2S in vivo, a key to many therapeutic applications. In this review, we highlight recent research works concerning the development and application of H2S-releasing biomaterials with a special emphasis to different release triggering conditions in in vivo studies. We believe that the further exploration of the molecular mechanisms underlying H2S donors and their function when incorporated with various biomaterials will potentially help us understand the pathophysiological mechanisms of different diseases and assist the development of H2S-based therapies.
Collapse
Affiliation(s)
- Jingyu Fan
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, USA
| | - Elizabeth Pung
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, USA
| | - Yuan Lin
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin Province, China
| | - Qian Wang
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, USA
| |
Collapse
|
16
|
Liu Y, Nguyen AW, Maynard JA. Engineering antibodies for conditional activity in the solid tumor microenvironment. Curr Opin Biotechnol 2022; 78:102809. [PMID: 36182870 DOI: 10.1016/j.copbio.2022.102809] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/25/2022] [Accepted: 08/30/2022] [Indexed: 12/14/2022]
Abstract
Antibody-based therapeutics enjoy considerable clinical and commercial successes as cancer treatments. However, they can also cause serious toxicities due to recognition of tumor-associated antigens in noncancerous tissues, which can prevent antibody use in certain patient populations and therapeutic modalities. Here, we discuss recent efforts to develop advanced antibody therapeutics with activities restricted to the solid tumor microenvironment. With the intent of decreasing toxicities and expanding therapeutic windows, protein engineering strategies can render ligand binding sensitive to multiple tumor-specific characteristics. These triggers can be intrinsic to solid tumor microenvironments, such as low pH, high extracellular ATP, and the presence of specific proteases. Emerging strategies rely instead on exogenous triggers such as light and ultrasound to provide spatial and temporal control over antibody activation. These multilayered approaches to targeting diseased tissues are expected to usher in a new generation of precision therapeutics.
Collapse
Affiliation(s)
- Yutong Liu
- Department of Chemical Engineering, University of Texas, Austin, TX 78712, USA
| | - Annalee W Nguyen
- Department of Chemical Engineering, University of Texas, Austin, TX 78712, USA.
| | - Jennifer A Maynard
- Department of Chemical Engineering, University of Texas, Austin, TX 78712, USA.
| |
Collapse
|
17
|
Furuya G, Katoh H, Atsumi S, Hashimoto I, Komura D, Hatanaka R, Senga S, Hayashi S, Akita S, Matsumura H, Miura A, Mita H, Nakakido M, Nagatoishi S, Sugiyama A, Suzuki R, Konishi H, Yamamoto A, Abe H, Hiraoka N, Aoki K, Kato Y, Seto Y, Yoshimura C, Miyadera K, Tsumoto K, Ushiku T, Ishikawa S. Nucleic acid-triggered tumoral immunity propagates pH-selective therapeutic antibodies through tumor-driven epitope spreading. Cancer Sci 2022; 114:321-338. [PMID: 36136061 PMCID: PMC9807517 DOI: 10.1111/cas.15596] [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: 08/11/2022] [Revised: 09/09/2022] [Accepted: 09/12/2022] [Indexed: 01/07/2023] Open
Abstract
Important roles of humoral tumor immunity are often pointed out; however, precise profiles of dominant antigens and developmental mechanisms remain elusive. We systematically investigated the humoral antigens of dominant intratumor immunoglobulin clones found in human cancers. We found that approximately half of the corresponding antigens were restricted to strongly and densely negatively charged polymers, resulting in simultaneous reactivities of the antibodies to both densely sulfated glycosaminoglycans (dsGAGs) and nucleic acids (NAs). These anti-dsGAG/NA antibodies matured and expanded via intratumoral immunological driving force of innate immunity via NAs. These human cancer-derived antibodies exhibited acidic pH-selective affinity across both antigens and showed specific reactivity to diverse spectrums of human tumor cells. The antibody-drug conjugate exerted therapeutic effects against multiple cancers in vivo by targeting cell surface dsGAG antigens. This study reveals that intratumoral immunological reactions propagate tumor-oriented immunoglobulin clones and demonstrates a new therapeutic modality for the universal treatment of human malignancies.
Collapse
Affiliation(s)
- Genta Furuya
- Department of Preventive medicine, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Hiroto Katoh
- Department of Preventive medicine, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Shinichiro Atsumi
- Department of Preventive medicine, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Itaru Hashimoto
- Department of Preventive medicine, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Daisuke Komura
- Department of Preventive medicine, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Ryo Hatanaka
- Discovery and Preclinical Research DivisionTaiho Pharmaceutical Co., Ltd.IbarakiJapan
| | - Shogo Senga
- Discovery and Preclinical Research DivisionTaiho Pharmaceutical Co., Ltd.IbarakiJapan
| | - Shuto Hayashi
- Department of Preventive medicine, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Shoji Akita
- Discovery and Preclinical Research DivisionTaiho Pharmaceutical Co., Ltd.IbarakiJapan
| | - Hirofumi Matsumura
- Discovery and Preclinical Research DivisionTaiho Pharmaceutical Co., Ltd.IbarakiJapan
| | - Akihiro Miura
- Discovery and Preclinical Research DivisionTaiho Pharmaceutical Co., Ltd.IbarakiJapan
| | - Hideaki Mita
- Department of Preventive medicine, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Makoto Nakakido
- Laboratory of Medical Proteomics, Institute of Medical ScienceThe University of TokyoTokyoJapan
| | - Satoru Nagatoishi
- Laboratory of Medical Proteomics, Institute of Medical ScienceThe University of TokyoTokyoJapan
| | - Akira Sugiyama
- Laboratory of Systems Biology and MedicineResearch Center for Advanced Science and Technology, The University of TokyoTokyoJapan
| | - Ryohei Suzuki
- Department of Preventive medicine, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Hiroki Konishi
- Department of Preventive medicine, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Asami Yamamoto
- Department of Preventive medicine, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Hiroyuki Abe
- Department of Pathology, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Nobuyoshi Hiraoka
- Department of Analytical PathologyNational Cancer Center Research InstituteTokyoJapan
| | - Kazunori Aoki
- Division of Molecular and Cellular MedicineNational Cancer Center Research InstituteTokyoJapan
| | - Yasumasa Kato
- Department of Oral Function and Molecular BiologyOhu University School of DentistryFukushimaJapan
| | - Yasuyuki Seto
- Department of Gastrointestinal SurgeryGraduate School of Medicine, The University of TokyoTokyoJapan
| | - Chihoko Yoshimura
- Discovery and Preclinical Research DivisionTaiho Pharmaceutical Co., Ltd.IbarakiJapan
| | - Kazutaka Miyadera
- Discovery and Preclinical Research DivisionTaiho Pharmaceutical Co., Ltd.IbarakiJapan
| | - Kouhei Tsumoto
- Laboratory of Medical Proteomics, Institute of Medical ScienceThe University of TokyoTokyoJapan
| | - Tetsuo Ushiku
- Department of Pathology, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Shumpei Ishikawa
- Department of Preventive medicine, Graduate School of MedicineThe University of TokyoTokyoJapan
| |
Collapse
|
18
|
Abstract
PURPOSE OF REVIEW T-cell-engaging antibodies or T-cell engagers (TCEs) can connect a patient's cytotoxic T cells with cancer cells, leading to potent redirected lysis. Until very recently, only one TCE was approved, the CD19/CD3-bispecific blinatumomab. Many new TCEs in late-stage clinical development target various hematopoietic lineage markers like CD20, BCMA, or CD123. Although very compelling single-agent activity of TCEs was observed with various blood-borne cancers, therapy of solid tumor indications has thus far been less successful. RECENT FINDINGS The approval in 2022 of the gp100 peptide-major histocompatibility complex (MHC)/CD3 bispecific TCE tebentafusp in uveal melanoma confirms that TCEs can also efficiently work against solid tumors. TCEs targeting peptide-MHC complexes will expand the target space for solid tumor therapy to intracellular targets. Likewise, early clinical trial data from TCEs targeting DLL3 in small cell lunger cancer showed promising antitumor activity. Various technologies for conditional activation of TCEs in the tumor microenvironment (TME) may expand the scope of conventional surface targets that suffer from a narrow therapeutic window. Finally, pharmacological enhancements for TCE therapies by engagement of certain costimulatory receptors and cytokines, or blockade of checkpoints, are showing promise. SUMMARY Targeting peptide-MHC complexes, conditional TCE technologies, and concepts enhancing TCE-activated T cells are paving the way towards overcoming challenges associated with solid tumor therapy.
Collapse
|
19
|
Designing antibodies as therapeutics. Cell 2022; 185:2789-2805. [PMID: 35868279 DOI: 10.1016/j.cell.2022.05.029] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/18/2022] [Accepted: 05/31/2022] [Indexed: 12/25/2022]
Abstract
Antibody therapeutics are a large and rapidly expanding drug class providing major health benefits. We provide a snapshot of current antibody therapeutics including their formats, common targets, therapeutic areas, and routes of administration. Our focus is on selected emerging directions in antibody design where progress may provide a broad benefit. These topics include enhancing antibodies for cancer, antibody delivery to organs such as the brain, gastrointestinal tract, and lungs, plus antibody developability challenges including immunogenicity risk assessment and mitigation and subcutaneous delivery. Machine learning has the potential, albeit as yet largely unrealized, for a transformative future impact on antibody discovery and engineering.
Collapse
|
20
|
Chen TT. Conditionally active T cell engagers for the treatment of solid tumors: rationale and clinical development. Expert Opin Biol Ther 2022; 22:955-963. [PMID: 35857922 DOI: 10.1080/14712598.2022.2098674] [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: 11/04/2022]
Abstract
INTRODUCTION T cell engagers are a class of bispecific molecules that induce highly potent T cell-dependent cytotoxicity by bringing T cell activating receptors into proximity with cancer-associated cell surface antigens. However, because of their high potency, there is a greater risk of on-target/off-tumor toxicity owing to normal tissues having tumor antigen expression even at low levels. To reduce these adverse events, the dysregulated activity of proteases within the tumor microenvironment has recently been explored to create inert prodrugs that become conditionally active engagers after their cleavage by these enzymes. AREAS COVERED T-cell engagers that have been introduced for clinical use, and their respective successes and failures are reviewed. The unique challenges of these bispecific molecules for treating solid tumors and prior technologies used to exploit the proteolytic tumor microenvironment to create better-tolerated prodrugs and how that experience has led to the current series of conditionally active T-cell engagers, are discussed. EXPERT OPINION Methods for modulating the serum half-life of both inert and activated T cell engagers could have important ramifications in how they infiltrate tumors and prevent toxicity. Alternative features of the tumor microenvironment can also be leveraged in the development of conditional T cell engagers.
Collapse
Affiliation(s)
- T Timothy Chen
- Maverick Therapeutics, Inc., a wholly owned subsidiary of Takeda Development Center Americas, Inc
| |
Collapse
|
21
|
The Role of Metabolic Plasticity of Tumor-Associated Macrophages in Shaping the Tumor Microenvironment Immunity. Cancers (Basel) 2022; 14:cancers14143331. [PMID: 35884391 PMCID: PMC9316955 DOI: 10.3390/cancers14143331] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 06/22/2022] [Accepted: 07/01/2022] [Indexed: 02/07/2023] Open
Abstract
Cancer cells possess a high metabolic demand for their rapid proliferation, survival, and progression and thus create an acidic and hypoxic tumor microenvironment (TME) deprived of nutrients. Moreover, acidity within the TME is the central regulator of tumor immunity that influences the metabolism of the immune cells and orchestrates the local and systemic immunity, thus, the TME has a major impact on tumor progression and resistance to anti-cancer therapy. Specifically, myeloid cells, which include myeloid-derived suppressor cells (MDSC), dendritic cells, and tumor-associated macrophages (TAMs), often reprogram their energy metabolism, resulting in stimulating the angiogenesis and immunosuppression of tumors. This review summarizes the recent findings of glucose, amino acids, and fatty acid metabolism changes of the tumor-associated macrophages (TAMs), and how the altered metabolism shapes the TME and anti-tumor immunity. Multiple proton pumps/transporters are involved in maintaining the alkaline intracellular pH which is necessary for the glycolytic metabolism of the myeloid cells and acidic TME. We highlighted the roles of these proteins in modulating the cellular metabolism of TAMs and their potential as therapeutic targets for improving immune checkpoint therapy.
Collapse
|
22
|
Li M, Mei S, Yang Y, Shen Y, Chen L. Strategies to mitigate the on- and off-target toxicities of recombinant immunotoxins: an antibody engineering perspective. Antib Ther 2022; 5:164-176. [PMID: 35928456 PMCID: PMC9344849 DOI: 10.1093/abt/tbac014] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 10/14/2021] [Accepted: 06/14/2022] [Indexed: 11/14/2022] Open
Abstract
Abstract
Targeted cancer therapies using immunotoxins has achieved remarkable efficacies in hematological malignancies. However, the clinical development of immunotoxins is also faced with many challenges like anti-drug antibodies and dose-limiting toxicity issues. Such a poor efficacy/safety ratio is also the major hurdle in the research and development of antibody-drug conjugates. From an antibody engineering perspective, various strategies were summarized/proposed to tackle the notorious on target off tumor toxicity issues, including passive strategy (XTENylation of immunotoxins) and active strategies (modulating the affinity and valency of the targeting moiety of immunotoxins, conditionally activating immunotoxins in the tumor microenvironments and reconstituting split toxin to reduce systemic toxicity etc.). By modulating the functional characteristics of the targeting moiety and the toxic moiety of immunotoxins, selective tumor targeting can be augmented while sparing the healthy cells in normal tissues expressing the same target of interest. If successful, the improved therapeutic index will likely help to address the dose-limiting toxicities commonly observed in the clinical trials of various immunotoxins.
Collapse
Affiliation(s)
- Mengyu Li
- Department of Postgraduate , Jiangxi University of Traditional Chinese Medicine, Nanchang, P.R. China
- Joint Graduate School , Yangtze Delta Drug Advanced Research Institute, Nantong, P.R. China
- Joint Graduate School , Yangtze Delta Pharmaceutical College, Nantong, P.R. China
| | - Sen Mei
- Biotherapeutics , Biocytogen Jiangsu Co. Ltd, Nantong, P.R. China
| | - Yi Yang
- Joint Graduate School , Yangtze Delta Drug Advanced Research Institute, Nantong, P.R. China
- Joint Graduate School , Yangtze Delta Pharmaceutical College, Nantong, P.R. China
- Institute of Innovative Medicine , Biocytogen Pharmaceuticals (Beijing) Co, Ltd, Beijing, P.R. China
| | - Yuelei Shen
- Joint Graduate School , Yangtze Delta Drug Advanced Research Institute, Nantong, P.R. China
- Joint Graduate School , Yangtze Delta Pharmaceutical College, Nantong, P.R. China
- Biotherapeutics , Biocytogen Pharmaceuticals (Beijing) Co, Ltd, Beijing, P.R. China
- Institute of Innovative Medicine , Biocytogen Pharmaceuticals (Beijing) Co, Ltd, Beijing, P.R. China
| | - Lei Chen
- Biotherapeutics , Biocytogen Jiangsu Co. Ltd, Nantong, P.R. China
- Biotherapeutics , Biocytogen Pharmaceuticals (Beijing) Co, Ltd, Beijing, P.R. China
| |
Collapse
|
23
|
Holder PG, Lim SA, Huang CS, Sharma P, Dagdas YS, Bulutoglu B, Sockolosky JT. Engineering interferons and interleukins for cancer immunotherapy. Adv Drug Deliv Rev 2022; 182:114112. [PMID: 35085624 DOI: 10.1016/j.addr.2022.114112] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/07/2022] [Accepted: 01/12/2022] [Indexed: 02/08/2023]
Abstract
Cytokines are a class of potent immunoregulatory proteins that are secreted in response to various stimuli and act locally to regulate many aspects of human physiology and disease. Cytokines play important roles in cancer initiation, progression, and elimination, and thus, there is a long clinical history associated with the use of recombinant cytokines to treat cancer. However, the use of cytokines as therapeutics has been limited by cytokine pleiotropy, complex biology, poor drug-like properties, and severe dose-limiting toxicities. Nevertheless, cytokines are crucial mediators of innate and adaptive antitumor immunity and have the potential to enhance immunotherapeutic approaches to treat cancer. Development of immune checkpoint inhibitors and combination immunotherapies has reinvigorated interest in cytokines as therapeutics, and a variety of engineering approaches are emerging to improve the safety and effectiveness of cytokine immunotherapy. In this review we highlight recent advances in cytokine biology and engineering for cancer immunotherapy.
Collapse
|
24
|
Lee PS, MacDonald KG, Massi E, Chew PV, Bee C, Perkins P, Chau B, Thudium K, Lohre J, Nandi P, Deyanova EG, Barman I, Gudmundsson O, Dollinger G, Sproul T, Engelhardt JJ, Strop P, Rajpal A. Improved therapeutic index of an acidic pH-selective antibody. MAbs 2022; 14:2024642. [PMID: 35192429 PMCID: PMC8865267 DOI: 10.1080/19420862.2021.2024642] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Although therapeutically efficacious, ipilimumab can exhibit dose-limiting toxicity that prevents maximal efficacious clinical outcomes and can lead to discontinuation of treatment. We hypothesized that an acidic pH-selective ipilimumab (pH Ipi), which preferentially and reversibly targets the acidic tumor microenvironment over the neutral periphery, may have a more favorable therapeutic index. While ipilimumab has pH-independent CTLA-4 affinity, pH Ipi variants have been engineered to have up to 50-fold enhanced affinity to CTLA-4 at pH 6.0 compared to pH 7.4. In hCTLA-4 knock-in mice, these variants have maintained anti-tumor activity and reduced peripheral activation, a surrogate marker for toxicity. pH-sensitive therapeutic antibodies may be a differentiating paradigm and a novel modality for enhanced tumor targeting and improved safety profiles.
Collapse
Affiliation(s)
- Peter S Lee
- Discovery Biotherapeutics, Bristol Myers Squibb, Redwood City, CA, USA
| | | | - Evan Massi
- Immuno-Oncology Research, Bristol Myers Squibb, Redwood City, CA, USA
| | - Pamela V Chew
- Oncology Biology, Gilead Sciences, Foster City, CA, USA
| | - Christine Bee
- Discovery Biology, Frontier Medicines, South San Francisco, CA, USA
| | - Padma Perkins
- Immuno-Oncology Research, Bristol Myers Squibb, Redwood City, CA, USA
| | - Bryant Chau
- Kyverna, Synthetic Biology, Emeryville, CA, USA
| | - Kent Thudium
- Immuno-Oncology Research, Bristol Myers Squibb, Redwood City, CA, USA
| | - Jack Lohre
- In Vivo Pharmacology, Bristol Myers Squibb, Redwood City, CA, USA
| | - Pradyot Nandi
- Pharmaceutical Candidate Optimization, Bristol Myers Squibb, Lawrenceville, NJ, USA
| | - Ekaterina G Deyanova
- Pharmaceutical Candidate Optimization, Bristol Myers Squibb, Lawrenceville, NJ, USA
| | - Ishita Barman
- Therapeutic Discovery, 3T Biosciences, South San Francisco, CA, USA
| | - Olafur Gudmundsson
- Pharmaceutical Candidate Optimization, Bristol Myers Squibb, Lawrenceville, NJ, USA
| | - Gavin Dollinger
- Discovery Biotherapeutics, Bristol Myers Squibb, Redwood City, CA, USA
| | - Tim Sproul
- In Vivo Pharmacology, UNITY Biotechnology, South San Francisco, CA, USA
| | | | - Pavel Strop
- Biologics Discovery, Tallac Therapeutics, Burlingame, CA, USA
| | - Arvind Rajpal
- Large Molecule Drug Discovery, Genentech Research and Early Development, South San Francisco, CA, USA
| |
Collapse
|
25
|
Ceci C, Lacal PM, Graziani G. Antibody-drug conjugates: Resurgent anticancer agents with multi-targeted therapeutic potential. Pharmacol Ther 2022; 236:108106. [PMID: 34990642 DOI: 10.1016/j.pharmthera.2021.108106] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 12/23/2021] [Accepted: 12/29/2021] [Indexed: 12/18/2022]
Abstract
Antibody-drug conjugates (ADCs) constitute a relatively new group of anticancer agents, whose first appearance took place about two decades ago, but a renewed interest occurred in recent years, following the success of anti-cancer immunotherapy with monoclonal antibodies. Indeed, an ADC combines the selectivity of a monoclonal antibody with the cell killing properties of a chemotherapeutic agent (payload), joined together through an appropriate linker. The antibody moiety targets a specific cell surface antigen expressed by tumor cells and/or cells of the tumor microenvironment and acts as a carrier that delivers the cytotoxic payload within the tumor mass. Despite advantages in terms of selectivity and potency, the development of ADCs is not devoid of challenges, due to: i) low tumor selectivity when the target antigens are not exclusively expressed by cancer cells; ii) premature release of the cytotoxic drug into the bloodstream as a consequence of linker instability; iii) development of tumor resistance mechanisms to the payload. All these factors may result in lack of efficacy and/or in no safety improvement compared to unconjugated cytotoxic agents. Nevertheless, the development of antibodies engineered to remain inert until activated in the tumor (e.g., antibodies activated proteolytically after internalization or by the acidic conditions of the tumor microenvironment) together with the discovery of innovative targets and cytotoxic or immunomodulatory payloads, have allowed the design of next-generation ADCs that are expected to possess improved therapeutic properties. This review provides an overview of approved ADCs, with related advantages and limitations, and of novel targets exploited by ADCs that are presently under clinical investigation.
Collapse
Affiliation(s)
- Claudia Ceci
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | | | - Grazia Graziani
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; IDI-IRCCS, Via Monti di Creta 104, 00167 Rome, Italy.
| |
Collapse
|
26
|
Enhancing immunotherapy in cancer by targeting emerging immunomodulatory pathways. Nat Rev Clin Oncol 2022; 19:37-50. [PMID: 34580473 DOI: 10.1038/s41571-021-00552-7] [Citation(s) in RCA: 414] [Impact Index Per Article: 138.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/13/2021] [Indexed: 02/08/2023]
Abstract
The discovery and clinical implementation of immune-checkpoint inhibitors (ICIs) targeting CTLA4, PD-1 and PD-L1 has revolutionized the treatment of cancer, as recognized by the 2018 Nobel Prize for Medicine and Physiology. This groundbreaking new approach has improved the outcomes of patients with various forms of advanced-stage cancer; however, the majority of patients receiving these therapies, even in combination, do not derive clinical benefit. Further development of agents targeting additional immune checkpoints, co-stimulatory receptors and/or co-inhibitory receptors that control T cell function is therefore critical. In this Review, we discuss the translational potential and clinical development of agents targeting both co-stimulatory and co-inhibitory T cell receptors. Specifically, we describe their mechanisms of action, and provide an overview of ongoing clinical trials involving novel ICIs including those targeting LAG3, TIM3, TIGIT and BTLA as well as agonists of the co-stimulatory receptors GITR, OX40, 41BB and ICOS. We also discuss several additional approaches, such as harnessing T cell metabolism, in particular via adenosine signalling, inhibition of IDO1, and targeting changes in glucose and fatty acid metabolism. We conclude that further efforts are needed to optimize the timing of combination ICI approaches and, most importantly, to individualize immunotherapy based on both patient-specific and tumour-specific characteristics.
Collapse
|
27
|
Zou W, Huang C, Sun Q, Zhao K, Gao H, Su R, Li Y. A stepwise mutagenesis approach using histidine and acidic amino acid to engineer highly pH-dependent protein switches. 3 Biotech 2022; 12:21. [PMID: 34956814 PMCID: PMC8686790 DOI: 10.1007/s13205-021-03079-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 11/26/2021] [Indexed: 10/26/2022] Open
Abstract
Antibody-based drugs can be highly toxic, because they target normal tissue as well as tumor tissue. The pH value of the extracellular microenvironments around tumor tissues is lower than that of normal tissues. Therefore, antibodies that engage in pH-dependent binding at slightly acidic pH are crucial for improving the safety of antibody-based drugs. Thus, we implemented a stepwise mutagenesis approach to engineering pH-dependent antibodies capable of selective binding in the acidic microenvironment in this study. The first step involved single-residue histidine scanning mutagenesis of the antibody's complementarity-determining regions to prescreen for pH-dependent mutants and identify ionizable sensitive hot-spot residues that could be substituted by acidic amino acids to obtain pH-dependent antibodies. The second step involved single-acidic amino acid residue substitutions of the identified residues and the assessment of pH-dependent binding. We identified six ionizable sensitive hot-spot residues using single-histidine scanning mutagenesis. Nine pH-dependent antibodies were isolated using single-acidic amino acid residue mutagenesis at the six hot-spot residue positions. Relative to wild-type anti-CEA chimera antibody, the binding selectivity of the best performing mutant was improved by approximately 32-fold according to ELISA and by tenfold according to FACS assay. The mutant had a high affinity in the pH range of 5.5-6.0. This study supports the development of pH-dependent protein switches and increases our understanding of the role of ionizable residues in protein interfaces. The stepwise mutagenesis approach is rapid, general, and robust and is expected to produce pH-sensitive protein affinity reagents for various applications.
Collapse
Affiliation(s)
- Wenjun Zou
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049 China
- Key Laboratory of Interdisciplinary Research, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101 China
| | - Chuncui Huang
- Key Laboratory of Interdisciplinary Research, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101 China
| | - Qing Sun
- Key Laboratory of Interdisciplinary Research, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101 China
| | - Keli Zhao
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049 China
- Key Laboratory of Interdisciplinary Research, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101 China
| | - Huanyu Gao
- Key Laboratory of Interdisciplinary Research, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101 China
| | - Rong Su
- Department of Clinical Laboratory, Foshan Hospital of Traditional Chinese Medicine, Foshan, 528000 Guangdong China
| | - Yan Li
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049 China
- Key Laboratory of Interdisciplinary Research, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101 China
| |
Collapse
|
28
|
Pinho JO, da Silva IV, Amaral JD, Rodrigues CMP, Casini A, Soveral G, Gaspar MM. Therapeutic potential of a copper complex loaded in pH-sensitive long circulating liposomes for colon cancer management. Int J Pharm 2021; 599:120463. [PMID: 33711474 DOI: 10.1016/j.ijpharm.2021.120463] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 12/13/2022]
Abstract
Colorectal carcinoma is a complex malignancy and current therapies are hampered by systemic toxicity and tumor resistance to treatment. In the field of cancer therapy, copper (Cu) compounds hold great promise, with some reaching clinical trials. However, the anticancer potential of Cu complexes has not yet been fully disclosed due to speciation in biological systems, leading to inactivation and/or potential side effects. This is the case of the widely studied Cu(II) complexes featuring phenanthroline ligands, with potent antiproliferative effects in vitro, but often failing in vivo. Aiming to overcome these limitations and maximize its anticancer effects in vivo, the Cu(II) complex (Cu(1,10-phenanthroline)Cl2) (Cuphen), displaying IC50 values <6 μM against different tumor cell lines, was loaded in long circulating liposomes with pH-sensitive properties (F1, DMPC:CHEMS:DSPE-PEG; F2, DOPE:CHEMS:DMPC:DSPE-PEG). This enabled a pH-dependent Cuphen release, with F1 and F2 releasing 36/78% and 47/94% of Cuphen at pH 6/4.5, respectively. The so formed nanoformulations preserved Cuphen effects towards cancer cell lines, with F2 presenting IC50 of 2.7 μM and 4.9 μM towards colon cancer CT-26 and HCT-116 cells, respectively. Additional in vitro studies confirmed that Cuphen antiproliferative activity towards colon cancer cells does not rely on cell cycle effect. Furthermore, in these cells, Cuphen reduced glycerol permeation and impaired cell migration. At 24 h incubation, wound closure was reduced by Cuphen, with migration values of 29% vs 54% (control) and 45% (1,10-phenanthroline) in CT-26 cells, and 33% vs ~44% (control and 1,10-phenanthroline) in HCT-116 cells. These effects were probably due to inhibition of aquaglyceroporins, membrane water and glycerol channels that are often abnormally expressed in tumors. In a syngeneic murine colon cancer model, F2 significantly reduced tumor progression, compared to the control group and to mice treated with free Cuphen or with the ligand, 1,10-phenanthroline, without eliciting toxic side effects. F2 led to a tumor volume reduction of ca. 50%. This was confirmed by RTV analysis, where F2 reached a value of 1.3 vs 4.4 (Control), 5.8 (Phen) and 3.8 (free Cuphen). These results clearly demonstrated the important role of the Cu(II) for the observed biological activity that was maximized following the association to a lipid-based nanosystem. Overall, this study represents a step forward in the development of pH-sensitive nanotherapeutic strategies of metallodrugs for colon cancer management.
Collapse
Affiliation(s)
- Jacinta O Pinho
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Inês V da Silva
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Joana D Amaral
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Cecília M P Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Angela Casini
- Department of Chemistry, Technical University of Munich (TUM), Lichtenbergstr. 4, 85748 Garching b. München, Germany.
| | - Graça Soveral
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
| | - M Manuela Gaspar
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
| |
Collapse
|