1
|
Shajan I, Rochet LNC, Tracey SR, Benazza R, Jackowska B, Hernandez-Alba O, Cianférani S, Scott CJ, van Delft FL, Chudasama V, Albada B. Modular Semisynthetic Approach to Generate T Cell-Dependent Bispecific Constructs from Recombinant IgG1 Antibodies. Bioconjug Chem 2024; 35. [PMID: 39284580 PMCID: PMC11487494 DOI: 10.1021/acs.bioconjchem.4c00309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/09/2024] [Accepted: 09/09/2024] [Indexed: 10/20/2024]
Abstract
Redirecting T cells to tumor cells by bispecific antibodies is an effective approach to treat cancer, and T cell-dependent bispecific antibodies (TDBAs) are an emerging class of potent immunotherapeutic agents. By simultaneously targeting antigens on tumor cells and T cells, T cells are activated to kill tumor cells. Herein, we report a platform to generate a novel class of 2:1 structure of T cell-dependent bispecific antibody with bivalency for HER2 receptors on tumor cells and monovalency for CD3 receptors on T cells. For this, we use a biogenic inverse electron-demand Diels-Alder (IEDDA) click reaction on genetically encoded tyrosine residues to install one TCO handle on therapeutically approved antibody trastuzumab. Subsequent TCO-tetrazine click with a tetrazine-functionalized CD3-binding Fab yields a 2:1 HER2 × CD3 TDBA that exhibits a tumor-killing capability at picomolar concentrations. Monovalency toward the CD3 receptor on T cells can lower the chances of cytokine release syndrome, which is a common side effect of such agents. Our semisynthetic approach can generate highly potent TDBA constructs in a few chemoenzymatic and synthetic steps.
Collapse
Affiliation(s)
- Irene Shajan
- Laboratory
of Organic Chemistry, Wageningen University
& Research, Stippeneng 4, Wageningen 6807 WE, The Netherlands
| | - Léa N. C. Rochet
- Department
of Chemistry, University College London, 20 Gordon St, London WC1H 0AJ, United Kingdom
| | - Shannon R. Tracey
- Patrick
G. Johnston Centre for Cancer Research, School of Medicine, Dentistry
and Biomedical Sciences, Queen’s
University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Rania Benazza
- Laboratoire
de Spectrométrie de Masse BioOrganique, Université de Strasbourg, CNRS, IPHC UMR 7178, 67000 F-Strasbourg, France
- Infrastructure
Nationale de Protéomique ProFI—FR2048, 67087 Strasbourg, France
| | - Bianka Jackowska
- Patrick
G. Johnston Centre for Cancer Research, School of Medicine, Dentistry
and Biomedical Sciences, Queen’s
University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Oscar Hernandez-Alba
- Laboratoire
de Spectrométrie de Masse BioOrganique, Université de Strasbourg, CNRS, IPHC UMR 7178, 67000 F-Strasbourg, France
- Infrastructure
Nationale de Protéomique ProFI—FR2048, 67087 Strasbourg, France
| | - Sarah Cianférani
- Laboratoire
de Spectrométrie de Masse BioOrganique, Université de Strasbourg, CNRS, IPHC UMR 7178, 67000 F-Strasbourg, France
- Infrastructure
Nationale de Protéomique ProFI—FR2048, 67087 Strasbourg, France
| | - Christopher J. Scott
- Patrick
G. Johnston Centre for Cancer Research, School of Medicine, Dentistry
and Biomedical Sciences, Queen’s
University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Floris L. van Delft
- Laboratory
of Organic Chemistry, Wageningen University
& Research, Stippeneng 4, Wageningen 6807 WE, The Netherlands
- Synaffix
BV—A Lonza Company, Kloosterstraat 9, Oss 5349 AB, The Netherlands
| | - Vijay Chudasama
- Department
of Chemistry, University College London, 20 Gordon St, London WC1H 0AJ, United Kingdom
| | - Bauke Albada
- Laboratory
of Organic Chemistry, Wageningen University
& Research, Stippeneng 4, Wageningen 6807 WE, The Netherlands
| |
Collapse
|
2
|
Liu CH, Leu SJ, Lee CH, Lin CY, Wang WC, Tsai BY, Lee YC, Chen CL, Yang YY, Lin LT. Production and characterization of single-chain variable fragment antibodies targeting the breast cancer tumor marker nectin-4. Front Immunol 2024; 14:1292019. [PMID: 38288120 PMCID: PMC10822971 DOI: 10.3389/fimmu.2023.1292019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 12/11/2023] [Indexed: 01/31/2024] Open
Abstract
Background Nectin-4 is a novel biomarker overexpressed in various types of cancer, including breast cancer, in which it has been associated with poor prognosis. Current literature suggests that nectin-4 has a role in cancer progression and may have prognostic and therapeutic implications. The present study aims to produce nectin-4-specific single-chain variable fragment (scFv) antibodies and evaluate their applications in breast cancer cell lines and clinical specimens. Methods We generated recombinant nectin-4 ectodomain fragments as immunogens to immunize chickens and the chickens' immunoglobulin genes were amplified for construction of anti-nectin-4 scFv libraries using phage display. The binding capacities of the selected clones were evaluated with the recombinant nectin-4 fragments, breast cancer cell lines, and paraffin-embedded tissue sections using various laboratory approaches. The binding affinity and in silico docking profile were also characterized. Results We have selected two clones (S21 and L4) from the libraries with superior binding capacity. S21 yielded higher signals when used as the primry antibody for western blot analysis and flow cytometry, whereas clone L4 generated cleaner and stronger signals in immunofluorescence and immunohistochemistry staining. In addition, both scFvs could diminish attachment-free cell aggregation of nectin-4-positive breast cancer cells. As results from ELISA indicated that L4 bound more efficiently to fixed nectin-4 ectodomain, molecular docking analysis was further performed and demonstrated that L4 possesses multiple polar contacts with nectin-4 and diversity in interacting residues. Conclusion Overall, the nectin-4-specific scFvs could recognize nectin-4 expressed by breast cancer cells and have the merit of being further explored for potential diagnostic and therapeutic applications.
Collapse
Affiliation(s)
- Ching-Hsuan Liu
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Microbiology & Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Sy-Jye Leu
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chi-Hsin Lee
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Cheng-Yuan Lin
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Wei-Chu Wang
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | | | - Yu-Ching Lee
- The Center of Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chi-Long Chen
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University and Taipei Medical University Hospital, Taipei, Taiwan
| | - Yi-Yuan Yang
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Core Laboratory of Antibody Generation and Research, Taipei Medical University, Taipei, Taiwan
| | - Liang-Tzung Lin
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
3
|
Paul T, Palaniyandi K, Gnanasampanthapandian D. Therapeutic Approaches to Increase the Survival Rate of Cancer Patients in the Younger and Older Population. Curr Aging Sci 2024; 17:16-30. [PMID: 38062658 DOI: 10.2174/0118746098241507231127114248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 06/25/2023] [Accepted: 09/22/2023] [Indexed: 05/18/2024]
Abstract
Various developments have been observed in the treatment of cancer patients, such as higher survival rates and better treatment outcomes. However, expecting similar outcomes in older patients remains a challenge. The main reason for this conclusion is the exclusion of older people from clinical trials for cancer drugs, as well as other factors, such as comorbidity, side effects, age-related frailties and their willingness to undergo multiple treatments. However, the discovery of new techniques and drug combinations has led to a significant improvement in the survival of the elderly population after the onset of the disease. On the other hand, cancer treatments have not become more complex for the younger population when compared to the older population, as the younger population tends to respond well to treatment trials and their physiological conditions are stable in response to treatments. In summary, this review correlates recent cancer treatment strategies and the corresponding responses and survival outcomes of older and younger patients.
Collapse
Affiliation(s)
- Tharrun Paul
- Cancer Science Laboratory, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603203, Chengalpattu, India
| | - Kanagaraj Palaniyandi
- Cancer Science Laboratory, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603203, Chengalpattu, India
| | - Dhanavathy Gnanasampanthapandian
- Cancer Science Laboratory, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603203, Chengalpattu, India
| |
Collapse
|
4
|
Delgado M, Garcia-Sanz JA. Therapeutic Monoclonal Antibodies against Cancer: Present and Future. Cells 2023; 12:2837. [PMID: 38132155 PMCID: PMC10741644 DOI: 10.3390/cells12242837] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/06/2023] [Accepted: 12/12/2023] [Indexed: 12/23/2023] Open
Abstract
A series of monoclonal antibodies with therapeutic potential against cancer have been generated and developed. Ninety-one are currently used in the clinics, either alone or in combination with chemotherapeutic agents or other antibodies, including immune checkpoint antibodies. These advances helped to coin the term personalized medicine or precision medicine. However, it seems evident that in addition to the current work on the analysis of mechanisms to overcome drug resistance, the use of different classes of antibodies (IgA, IgE, or IgM) instead of IgG, the engineering of the Ig molecules to increase their half-life, the acquisition of additional effector functions, or the advantages associated with the use of agonistic antibodies, to allow a broad prospective usage of precision medicine successfully, a strategy change is required. Here, we discuss our view on how these strategic changes should be implemented and consider their pros and cons using therapeutic antibodies against cancer as a model. The same strategy can be applied to therapeutic antibodies against other diseases, such as infectious or autoimmune diseases.
Collapse
Affiliation(s)
| | - Jose A. Garcia-Sanz
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas (CIB-CSIC), 28040 Madrid, Spain;
| |
Collapse
|
5
|
Chen Z, Qiao S, Yang L, Sun M, Li B, Lu A, Li F. Mechanistic Insights into the Roles of the IL-17/IL-17R Families in Pancreatic Cancer. Int J Mol Sci 2023; 24:13539. [PMID: 37686343 PMCID: PMC10487659 DOI: 10.3390/ijms241713539] [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: 07/24/2023] [Revised: 08/17/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023] Open
Abstract
The members of the cytokine interleukin 17 (IL-17) family, along with their receptors (IL-17R), are vital players in a range of inflammatory diseases and cancer. Although generally regarded as proinflammatory, the effects they exhibit on cancer progression are a double-edged sword, with both antitumor and protumor activities being discovered. There is growing evidence that the IL-17 signaling pathways have significant impacts on the tumor microenvironment (TME), immune response, and inflammation in various types of cancer, including pancreatic cancer. However, the detailed mechanistic functions of the IL-17/IL-17R families in pancreatic cancer were rarely systematically elucidated. This review considers the role of the IL-17/IL-17R families in inflammation and tumor immunity and elaborates on the mechanistic functions and correlations of these members with pathogenesis, progression, and chemoresistance in pancreatic cancer. By summarizing the advanced findings on the role of IL-17/IL17R family members and IL-17 signaling pathways at the molecular level, cellular level, and disease level in pancreatic cancer, this review provides an in-depth discussion on the potential of IL-17/IL-17R as prognostic markers and therapeutic targets in pancreatic cancer.
Collapse
Affiliation(s)
- Zheng Chen
- Shum Yiu Foon Shum Bik Chuen Memorial Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China; (Z.C.); (S.Q.); (L.Y.); (M.S.); (B.L.)
- Institute of Precision Medicine and Innovative Drug Discovery (PMID), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Shuangying Qiao
- Shum Yiu Foon Shum Bik Chuen Memorial Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China; (Z.C.); (S.Q.); (L.Y.); (M.S.); (B.L.)
- Institute of Precision Medicine and Innovative Drug Discovery (PMID), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Liu Yang
- Shum Yiu Foon Shum Bik Chuen Memorial Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China; (Z.C.); (S.Q.); (L.Y.); (M.S.); (B.L.)
- Institute of Precision Medicine and Innovative Drug Discovery (PMID), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Meiheng Sun
- Shum Yiu Foon Shum Bik Chuen Memorial Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China; (Z.C.); (S.Q.); (L.Y.); (M.S.); (B.L.)
- Institute of Precision Medicine and Innovative Drug Discovery (PMID), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Boyue Li
- Shum Yiu Foon Shum Bik Chuen Memorial Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China; (Z.C.); (S.Q.); (L.Y.); (M.S.); (B.L.)
- Institute of Precision Medicine and Innovative Drug Discovery (PMID), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Aiping Lu
- Shum Yiu Foon Shum Bik Chuen Memorial Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China; (Z.C.); (S.Q.); (L.Y.); (M.S.); (B.L.)
- Institute of Precision Medicine and Innovative Drug Discovery (PMID), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Fangfei Li
- Shum Yiu Foon Shum Bik Chuen Memorial Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China; (Z.C.); (S.Q.); (L.Y.); (M.S.); (B.L.)
- Institute of Precision Medicine and Innovative Drug Discovery (PMID), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| |
Collapse
|
6
|
Joo V, Petrovas C, de Leval L, Noto A, Obeid M, Fenwick C, Pantaleo G. A CD64/FcγRI-mediated mechanism hijacks PD-1 from PD-L1/2 interaction and enhances anti-PD-1 functional recovery of exhausted T cells. Front Immunol 2023; 14:1213375. [PMID: 37622123 PMCID: PMC10446174 DOI: 10.3389/fimmu.2023.1213375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 07/24/2023] [Indexed: 08/26/2023] Open
Abstract
Therapeutic monoclonal antibodies (mAb) targeting the immune checkpoint inhibitor programmed cell death protein 1 (PD-1) have achieved considerable clinical success in anti-cancer therapy through relieving T cell exhaustion. Blockade of PD-1 interaction with its ligands PD-L1 and PD-L2 is an important determinant in promoting the functional recovery of exhausted T cells. Here, we show that anti-PD-1 mAbs act through an alternative mechanism leading to the downregulation of PD-1 surface expression on memory CD4+ and CD8+ T cells. PD-1 receptor downregulation is a distinct process from receptor endocytosis and occurs in a CD14+ monocyte dependent manner with the CD64/Fcγ receptor I acting as the primary factor for this T cell extrinsic process. Importantly, downregulation of surface PD-1 strongly enhances antigen-specific functional recovery of exhausted PD-1+CD8+ T cells. Our study demonstrates a novel mechanism for reducing cell surface levels of PD-1 and limiting the inhibitory targeting by PD-L1/2 and thereby enhancing the efficacy of anti-PD-1 Ab in restoring T cell functionality.
Collapse
Affiliation(s)
- Victor Joo
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Constantinos Petrovas
- Institute of Pathology, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Laurence de Leval
- Institute of Pathology, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Alessandra Noto
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Michel Obeid
- Lausanne Center for Immuno-oncology Toxicities (LCIT), Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Craig Fenwick
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Giuseppe Pantaleo
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
- Swiss Vaccine Research Institute, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
7
|
Ourailidou ME, Tsirigoti A, Kotsira G, Angelis S, Papadopoulos V, Gazouli M, Filippou DK. Oncology Clinical Trials in Greece: Progress in the Past Decade. J Long Term Eff Med Implants 2023; 33:79-88. [PMID: 36734930 DOI: 10.1615/jlongtermeffmedimplants.2022044793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Cancer is established as a major contributor to global burden as millions of deaths are reported every year. Advances in molecular, epidemiologic and clinical research have led to significant improvements in prevention, screening and treatment of tumors. The purpose of the study is to describe the progress of oncology clinical trials performed in Greece during the past decade and the obstacles that still need to be addressed in cancer research. A search was conducted in the public database EU Clinical Trials Register using the algorithm 'cancer AND Greece'. Results included relevant trials approved between 2010 and 2020. A total of 480 trials were approved for conduct in Greece from 2010 to 2020. The majority are multinational, phase III trials, exploring the efficacy and safety of agents in the management of lung cancer and multiple myeloma. A variety of small-molecules and monoclonal antibodies has and is being tested against key binding targets. Based on their promising effects on patients' responses and outcomes, many have been marketed for the treatment of several cancer types and are considered milestones in cancer discovery. It goes without saying that oncology research has made tremendous steps towards the development of potent and tolerable anticancer agents, with Greece having an active role. Current efforts focus on the use of alternative designs and tools aiming at further improving patients' survival and quality of life, while globalization of clinical research is also a matter of high importance.
Collapse
Affiliation(s)
- Maria Eleni Ourailidou
- Pharmaceutical Studies & Research Division, Clinical Trials Department, National Organization for Medicines, Athens, Greece
| | - Alexandra Tsirigoti
- School of Medicine, National and Kapodestrian University of Athens, Greece; Research and Educational Institute in Biomedical Sciences, Piraeus, Greece
| | - Georgia Kotsira
- School of Medicine, National and Kapodestrian University of Athens, Greece; Research and Educational Institute in Biomedical Sciences, Piraeus, Greece
| | - Stavros Angelis
- Second Orthopedic Department, Panagiotis & Aglaia Kyriakou Children's Hospital, Athens, Greece; Trauma and Orthopedic Department, Korgialenio-Benakio Hellenic Red Cross Hospital, Athens, Greece; Department of Anatomy, School of Medicine, National and Kapodistrian University of Athens, Greece
| | | | - Maria Gazouli
- School of Medicine, National and Kapodestrian University of Athens, Greece
| | - Dimitrios K Filippou
- Pharmaceutical Studies & Research Division, Clinical Trials Department, National Organization for Medicines, Athens, Greece; School of Medicine, National and Kapodestrian University of Athens, Greece; Department of Anatomy, School of Medicine, National and Kapodistrian University of Athens, Greece
| |
Collapse
|
8
|
The Mutually Mediated Chloride Intracellular Channel Protein 1 (CLIC1) Relationship between Malignant Cells and Tumor Blood Vessel Endothelium Exhibits a Significant Impact on Tumor Angiogenesis, Progression, and Metastasis in Clear Cell Renal Cell Carcinoma (ccRCC). Cancers (Basel) 2022; 14:cancers14235981. [PMID: 36497464 PMCID: PMC9740861 DOI: 10.3390/cancers14235981] [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: 10/16/2022] [Revised: 11/24/2022] [Accepted: 11/29/2022] [Indexed: 12/07/2022] Open
Abstract
Background: Overexpression of chloride intracellular channel protein 1 (CLIC1) in tumor cells has been confirmed, but it has received less attention in the tumor blood vessel endothelium. Aim: The assessment of CLIC1 expression in ccRCC tumor blood vessels and its relationship with TNM parameters and tumor cell CLIC1 expression. Methods: CLIC1 immunostaining in ccRCC was evaluated in 50 cases in both malignant cells and tumor blood vessels (CLIC1 microvessel density-CLIC1-MVD) and was correlated with TNM staging parameters. Results: CLIC1-MVD was observed in approximately 65% of cases, and CLIC1 co-localization in both tumor and endothelial cells was observed in 59% of cases. ccRCC was classified into four groups (Classes 0−3) based on the percentage of positive tumor cells, with each group including sub-groups defined by CLIC1 expression in the endothelium. Class 3 (60−100% positive tumor cells) had the highest CLIC1-MVD, with an impact on T and M parameters (p value = 0.007 for T, and p value = 0.006 for M). For cases with CLIC1 intracellular translocation, there was a strong correlation between CLIC1-MVD and M (p value < 0.001). Conclusions: Co-expression of ccRCC tumor and endothelial cells promotes tumor progression and metastasis and should be investigated further as a potential therapeutic target for ccRCC and other human malignancies.
Collapse
|
9
|
Lembo RR, Manna L, Froechlich G, Sasso E, Passariello M, De Lorenzo C. New Insights on the Role of Anti-PD-L1 and Anti-CTLA-4 mAbs on Different Lymphocytes Subpopulations in TNBC. Cancers (Basel) 2022; 14:5289. [PMID: 36358708 PMCID: PMC9656156 DOI: 10.3390/cancers14215289] [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: 09/22/2022] [Revised: 10/21/2022] [Accepted: 10/22/2022] [Indexed: 11/28/2022] Open
Abstract
Antibody-based cancer immunotherapy includes monoclonals against immune checkpoints (ICs), to modulate specific T cell responses against cancer. NK cells are a newly emerging target for immune checkpoint receptor inhibition in cancer immunotherapy, as ICs are also expressed on NK cells in various cancers. The latter cells are becoming attractive targets for cancer immunotherapy, as they are effector cells similar to CTLs, exerting natural cytotoxicity against primary tumor cells and metastasis, and they are able to distinguish tumor cells from healthy ones, leading to more specific anti-tumor cytotoxicity and reduced off-target effects. Thus, we decided to test the effects on isolated NK cells and T cell subpopulations of novel immunomodulatory mAbs, recently generated in our lab, in comparison with those in clinical use, such as ipilimumab and atezolizumab. Interestingly, we found that the novel anti-CTLA-4 (ID-1) and anti-PD-L1 (PD-L1_1) antibodies are able to induce NK cell activation and exert anti-tumor effects on TNBC cells co-cultured with NK cells more efficiently than the clinically validated ones, either when used as single agents or in combinatorial treatments. On the other hand, ipilimumab was found to be more effective in activating T cells with respect to ID-1. These findings indicate that antibodies targeting different epitopes can have differential effects on different lymphocytes subpopulations and that novel combinations of mAbs could be suitable for therapeutic approaches aimed at activating not only T cells but also NK cells, especially for tumors lacking MHC.
Collapse
Affiliation(s)
- Rosa Rapuano Lembo
- Ceinge—Biotecnologie Avanzate s.c.a.r.l., Via Gaetano Salvatore 486, 80145 Naples, Italy
- European School of Molecular Medicine, University of Milan, 20122 Milan, Italy
| | - Lorenzo Manna
- Ceinge—Biotecnologie Avanzate s.c.a.r.l., Via Gaetano Salvatore 486, 80145 Naples, Italy
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
| | - Guendalina Froechlich
- Ceinge—Biotecnologie Avanzate s.c.a.r.l., Via Gaetano Salvatore 486, 80145 Naples, Italy
- European School of Molecular Medicine, University of Milan, 20122 Milan, Italy
| | - Emanuele Sasso
- Ceinge—Biotecnologie Avanzate s.c.a.r.l., Via Gaetano Salvatore 486, 80145 Naples, Italy
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
| | - Margherita Passariello
- Ceinge—Biotecnologie Avanzate s.c.a.r.l., Via Gaetano Salvatore 486, 80145 Naples, Italy
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
| | - Claudia De Lorenzo
- Ceinge—Biotecnologie Avanzate s.c.a.r.l., Via Gaetano Salvatore 486, 80145 Naples, Italy
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
| |
Collapse
|
10
|
Jain M, Mishra A, Singh MK, Shyam H, Kumar S, Shankar P, Singh S. Immunotherapeutic and their immunological aspects: Current treatment strategies and agents. Natl J Maxillofac Surg 2022; 13:322-329. [PMID: 36683928 PMCID: PMC9851344 DOI: 10.4103/njms.njms_62_22] [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: 05/07/2022] [Revised: 07/26/2022] [Accepted: 07/29/2022] [Indexed: 01/24/2023] Open
Abstract
Cancer is often caused by the immune system's inability to deal with malignant cells and allows them to progress and proliferate. Emerging cancerous cells constantly evade the immune system, and as a result, these cancerous cells acquire more mutations and exhibit the deadliest characteristics among malignant tumors. The importance of understanding tumor immunology, particularly the functions of tumor antigens and the immunosuppressive tumor microenvironment, is highlighted by the effectiveness of cancer immunotherapy therapies. Many innovative immunotherapy drugs that effectively battle cancer have been produced since the 1980s. At present, in cancer treatment, immunotherapy appears as a paradigm that targets immune checkpoints of tumor cells such as CTLA-4, PD-1, and monoclonal antibodies (MABs), although the treatment of cancer is classified into non-specific and specific types. Specific types define the antibody targeting cell receptors as a new cancer treatment modality. For a number of malignancies, checkpoint inhibitors, MABs, and their derivatives have become standard-of-care therapy. Other immunotherapy techniques, such as most cancer vaccines and cell-based therapies, are still in the experimental stage. Many new immunotherapy techniques and agents are being explored and evaluated in clinical trials, which is a good thing. Thus, this review discusses the role of checkpoint inhibitors and MABs in the treatment of tumor cells. Moreover, these findings help us to understand the mechanism of action of this class of therapeutics and provide support for the management of cancer treatment.
Collapse
Affiliation(s)
- Mayank Jain
- Department of Thoracic Surgery, King George's Medical University, Lucknow, Uttar Pradesh, India
| | - Archana Mishra
- Department of Thoracic Surgery, King George's Medical University, Lucknow, Uttar Pradesh, India
| | - Mukul K. Singh
- Department of Urology, King George's Medical University, Lucknow, Uttar Pradesh, India
| | - Hari Shyam
- Department of Thoracic Surgery, King George's Medical University, Lucknow, Uttar Pradesh, India
| | - Shailendra Kumar
- Department of Thoracic Surgery, King George's Medical University, Lucknow, Uttar Pradesh, India
| | - Pratap Shankar
- Center for Advance Research, Lucknow, Uttar Pradesh, India
| | - Saumya Singh
- Department of Surgery, King George's Medical University, Lucknow, Uttar Pradesh, India
| |
Collapse
|
11
|
Raeisi H, Azimirad M, Nabavi-Rad A, Asadzadeh Aghdaei H, Yadegar A, Zali MR. Application of recombinant antibodies for treatment of Clostridioides difficile infection: Current status and future perspective. Front Immunol 2022; 13:972930. [PMID: 36081500 PMCID: PMC9445313 DOI: 10.3389/fimmu.2022.972930] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 08/03/2022] [Indexed: 11/13/2022] Open
Abstract
Clostridioides difficile (C. difficile), known as the major cause of antibiotic-associated diarrhea, is regarded as one of the most common healthcare-associated bacterial infections worldwide. Due to the emergence of hypervirulent strains, development of new therapeutic methods for C. difficile infection (CDI) has become crucially important. In this context, antibodies have been introduced as valuable tools in the research and clinical environments, as far as the effectiveness of antibody therapy for CDI was reported in several clinical investigations. Hence, production of high-performance antibodies for treatment of CDI would be precious. Traditional approaches of antibody generation are based on hybridoma technology. Today, application of in vitro technologies for generating recombinant antibodies, like phage display, is considered as an appropriate alternative to hybridoma technology. These techniques can circumvent the limitations of the immune system and they can be exploited for production of antibodies against different types of biomolecules in particular active toxins. Additionally, DNA encoding antibodies is directly accessible in in vitro technologies, which enables the application of antibody engineering in order to increase their sensitivity and specificity. Here, we review the application of antibodies for CDI treatment with an emphasis on recombinant fragment antibodies. Also, this review highlights the current and future prospects of the aforementioned approaches for antibody-mediated therapy of CDI.
Collapse
Affiliation(s)
- Hamideh Raeisi
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoumeh Azimirad
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Nabavi-Rad
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid Asadzadeh Aghdaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Yadegar
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
Venetoclax enhances the efficacy of therapeutic antibodies in B-cell malignancies by augmenting tumor cell phagocytosis. Blood Adv 2022; 6:4847-4858. [PMID: 35820018 PMCID: PMC9631674 DOI: 10.1182/bloodadvances.2022007364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 07/01/2022] [Indexed: 12/04/2022] Open
Abstract
Immunotherapy has evolved as a powerful tool for the treatment of B-cell malignancies, and patient outcomes have improved by combining therapeutic antibodies with conventional chemotherapy. Overexpression of antiapoptotic B-cell lymphoma 2 (Bcl-2) is associated with a poor prognosis, and increased levels have been described in patients with "double-hit" diffuse large B-cell lymphoma, a subgroup of Burkitt's lymphoma, and patients with pediatric acute lymphoblastic leukemia harboring a t(17;19) translocation. Here, we show that the addition of venetoclax (VEN), a specific Bcl-2 inhibitor, potently enhanced the efficacy of the therapeutic anti-CD20 antibody rituximab, anti-CD38 daratumumab, and anti-CD19-DE, a proprietary version of tafasitamab. This was because of an increase in antibody-dependent cellular phagocytosis by macrophages as shown in vitro and in vivo in cell lines and patient-derived xenograft models. Mechanistically, double-hit lymphoma cells subjected to VEN triggered phagocytosis in an apoptosis-independent manner. Our study identifies the combination of VEN and therapeutic antibodies as a promising novel strategy for the treatment of B-cell malignancies.
Collapse
|
13
|
Nejadghaderi SA, Balibegloo M, Saghazadeh A, Rezaei N. Clinical safety and efficacy of bispecific antibody in the treatment of solid tumors: A protocol for a systematic review. PLoS One 2022; 17:e0271506. [PMID: 35849585 PMCID: PMC9292075 DOI: 10.1371/journal.pone.0271506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 07/01/2022] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Cancers are among the most common causes of mortality and morbidity. Recently, bispecific antibodies (BsAbs) have been used for cancer treatment. The aim of this systematic review and meta-analysis will be to determine the safety and efficacy of BsAbs in the treatment of solid tumors. METHODS We will search five electronic databases, PubMed, EMBASE, Scopus, Web of Science, and CENTRAL, in addition to Clinical-Trials.gov and metaRegister of controlled trials and backward and forward citation searching of included studies. Eligible studies will be controlled clinical trials evaluating safety and/or efficacy of BsAbs in adult patients with solid tumors. The primary outcomes will be the incidence of safety and efficacy measures. Title and/or abstract screening, full text reviewing, data collection, and quality assessment will be done by two reviewers. We will use The Cochrane Collaboration's risk of bias tool 2 (RoB2) to assess the quality of included studies. If I-square heterogeneity was greater than 40%, we will implement random effect model. Subgroup analysis and meta-regression will be undertaken if applicable. The metaprop command of STATA will be used to calculate frequency of AEs. Funnel plot, Egger's and Peter's tests will be utilized to evaluate publication bias in case of including at least ten studies. We will use sensitivity analysis to evaluate the effects of funding sources and continuity correction on effects size. CONCLUSIONS The findings of the present study will provide information on safety and efficacy of BsAbs for physicians and researchers in the management of solid tumors. TRIAL REGISTRATION Registration on PROSPERO CRD42021227879 Also, important protocol amendments will be stated on PROSPERO registration.
Collapse
Affiliation(s)
- Seyed Aria Nejadghaderi
- Systematic Review and Meta-Analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Maryam Balibegloo
- Systematic Review and Meta-Analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Amene Saghazadeh
- Systematic Review and Meta-Analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Research Center for Immunodeficiencies, Children’s Medical Center, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Research Center for Immunodeficiencies, Children’s Medical Center, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| |
Collapse
|
14
|
Hambach J, Fumey W, Stähler T, Gebhardt AJ, Adam G, Weisel K, Koch-Nolte F, Bannas P. Half-Life Extended Nanobody-Based CD38-Specific Bispecific Killercell Engagers Induce Killing of Multiple Myeloma Cells. Front Immunol 2022; 13:838406. [PMID: 35651607 PMCID: PMC9150782 DOI: 10.3389/fimmu.2022.838406] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 04/19/2022] [Indexed: 01/14/2023] Open
Abstract
CD38 is a target for immunotherapy of multiple myeloma. Llama-derived CD38-specific nanobodies allow easy reformatting into mono-, bi- and multispecific proteins. To evaluate the utility of nanobodies for constructing CD38-specific nanobody-based killer cell engagers (nano-BiKEs), we generated half-life extended nano-BiKEs (HLE-nano-BiKEs) by fusing a CD38-specific nanobody to a CD16-specific nanobody for binding to the Fc-receptor on NK cells and further to an albumin-specific nanobody to extend the half-life in vivo. HLE-nano-BiKEs targeting three different epitopes (E1, E2, E3) of CD38 were expressed in transiently transfected HEK-6E cells. We verified specific and simultaneous binding to CD38 on myeloma cells, CD16 on NK cells, and to albumin. We tested the capacity of these HLE-nano-BiKEs to mediate cytotoxicity against CD38-expressing multiple myeloma cell lines and primary myeloma cells from human bone marrow biopsies in bioluminescence and flowcytometry assays with NK92 cells as effector cells. The results revealed specific time- and dose-dependent cytolysis of CD38+ myeloma cell lines and effective depletion of CD38-expressing multiple myeloma cells from primary human bone marrow samples. Our results demonstrate the efficacy of CD38-specific HLE-nano-BiKEs in vitro and ex vivo, warranting further preclinical evaluation in vivo of their therapeutic potential for the treatment of multiple myeloma.
Collapse
Affiliation(s)
- Julia Hambach
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - William Fumey
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias Stähler
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anna Josephine Gebhardt
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gerhard Adam
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katja Weisel
- Department of Oncology and Hematology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Friedrich Koch-Nolte
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Peter Bannas
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- *Correspondence: Peter Bannas,
| |
Collapse
|
15
|
Abu Khalaf S, Dandachi D, Granwehr BP, Rodriguez-Barradas MC. Cancer immunotherapy in adult patients with HIV. J Investig Med 2022; 70:883-891. [PMID: 35086858 DOI: 10.1136/jim-2021-002205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/07/2021] [Indexed: 11/03/2022]
Abstract
The availability of antiretroviral therapy (ART) has increased the life expectancy of people with HIV (PWH) and reduced the incidence of AIDS-associated malignancies, yet PWH have a significantly increased incidence of malignancy and less favorable outcomes of cancer treatment compared with the general population.Immunotherapy has revolutionized cancer therapy, becoming the standard of care for various malignancy treatments. However, PWH are an underserved population with limited access to clinical trials and cancer treatment.This review of the available evidence on different classes of cancer immunotherapy in PWH is mostly based on case reports, case series, but few prospective studies and clinical trials due to the exclusion of PWH from most oncologic clinical trials. The results of the available evidence support the safety of immunotherapy in PWH. Immunotherapy has similar effectiveness in PWH, an acceptable toxicity profile, and has no clinically significant impact on HIV viral load and CD4-T cell count. In addition, there is no reported change in the incidence of opportunistic infections and other complications for PWH with well-controlled viremia.This review aims to briefly summarize the current state of immunotherapy in cancer, guide clinicians in the management of immunotherapy in cancer PWH, and encourage the inclusion of PWH in clinical trials of cancer immunotherapy.
Collapse
Affiliation(s)
- Suha Abu Khalaf
- Department of Medicine, Division of Infectious Diseases, University of Missouri System, Columbia, Missouri, USA
| | - Dima Dandachi
- Department of Medicine, Division of Infectious Diseases, University of Missouri System, Columbia, Missouri, USA
| | - Bruno P Granwehr
- Department of Medicine, Division of Infectious Diseases, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Maria C Rodriguez-Barradas
- Infectious Diseases Section, Michael E DeBakey VAMC, Houston, Texas, USA.,Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
16
|
Chen F, Chen N, Gao Y, Jia L, Lyu Z, Cui J. Clinical Progress of PD-1/L1 Inhibitors in Breast Cancer Immunotherapy. Front Oncol 2022; 11:724424. [PMID: 35070955 PMCID: PMC8770281 DOI: 10.3389/fonc.2021.724424] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 12/06/2021] [Indexed: 12/20/2022] Open
Abstract
Breast cancer is a major killer of women's health worldwide. While breast cancer is thought to have lower immunogenicity compared with other solid tumors, combination therapy is able to improve the immunogenicity of the tumor and sensitize breast cancer cells to immunotherapy. Immunotherapy represented by immune checkpoint inhibitors (ICIs) has been largely explored in the field of breast cancer, including both early and advanced disease. Immunotherapy for triple-negative breast cancer (TNBC) has been the most studied, and the PD-L1 inhibitor atezolizumab combined with nab-paclitaxel has been used in the first-line treatment of TNBC. Immunotherapeutic data for human epidermal growth factor receptor-positive and hormone receptor-positive breast cancer are also accumulating. This review summarizes the clinical trial data of ICIs or ICI-containing therapies in different types and stages of breast cancer.
Collapse
Affiliation(s)
| | | | | | | | | | - Jiuwei Cui
- Cancer Center, the First Hospital of Jilin University, Changchun, China
| |
Collapse
|
17
|
Lynch KT, Squeo GC, Kane WJ, Meneveau MO, Petroni G, Olson WC, Chianese-Bullock KA, Slingluff CL, Foley EF, Friel CM. A pilot trial of vaccination with Carcinoembryonic antigen and Her2/neu peptides in advanced colorectal cancer. Int J Cancer 2022; 150:164-173. [PMID: 34480368 DOI: 10.1002/ijc.33793] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 06/03/2021] [Accepted: 06/23/2021] [Indexed: 12/14/2022]
Abstract
Checkpoint-blockade therapy (CBT) is approved for select colorectal cancer (CRC) patents, but additional immunotherapeutic options are needed. We hypothesized that vaccination with carcinoembryonic antigen (CEA) and Her2/neu (Her2) peptides would be immunogenic and well tolerated by participants with advanced CRC. A pilot clinical trial (NCT00091286) was conducted in HLA-A2+ or -A3+ Stage IIIC-IV CRC patients. Participants were vaccinated weekly with CEA and Her2 peptides plus tetanus peptide and GM-CSF emulsified in Montanide ISA-51 adjuvant for 3 weeks. Adverse events (AEs) were recorded per NIH Common Terminology Criteria for Adverse Events version 3. Immunogenicity was evaluated by interferon-gamma ELISpot assay of in vitro sensitized peripheral blood mononuclear cells and lymphocytes from the sentinel immunized node. Eleven participants were enrolled and treated; one was retrospectively found to be ineligible due to HLA type. All 11 participants were included in AEs and survival analyses, and the 10 eligible participants were evaluated for immunogenicity. All participants reported AEs: 82% were Grade 1-2, most commonly fatigue or injection site reactions. Two participants (18%) experienced treatment-related dose-limiting Grade 3 AEs; both were self-limiting. Immune responses to Her2 or CEA peptides were detected in 70% of participants. Median overall survival (OS) was 16 months; among those enrolled with no evidence of disease (n = 3), median OS was not reached after 10 years of follow-up. These data demonstrate that vaccination with CEA or Her2 peptides is well tolerated and immunogenic. Further study is warranted to assess potential clinical benefits of vaccination in advanced CRC either alone or in combination with CBT.
Collapse
Affiliation(s)
- Kevin T Lynch
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Gabriella C Squeo
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - William J Kane
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Max O Meneveau
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Gina Petroni
- Department of Public Health Sciences, University of Virginia Cancer Center, Charlottesville, Virginia, USA
| | - Walter C Olson
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | | | - Craig L Slingluff
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Eugene F Foley
- Department of Surgery, University of Wisconsin, Madison, Wisconsin, USA
| | - Charles M Friel
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
18
|
Larbouret C, Gros L, Pèlegrin A, Chardès T. Improving Biologics' Effectiveness in Clinical Oncology: From the Combination of Two Monoclonal Antibodies to Oligoclonal Antibody Mixtures. Cancers (Basel) 2021; 13:cancers13184620. [PMID: 34572847 PMCID: PMC8465647 DOI: 10.3390/cancers13184620] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 09/01/2021] [Accepted: 09/09/2021] [Indexed: 01/03/2023] Open
Abstract
Simple Summary The approval of the two antibody combinations trastuzumab/pertuzumab and ipilimumab/nivolumab in oncology has paved the way for novel antibody combinations or oligoclonal antibody mixtures to improve their efficacy in cancer. The underlying biological mechanisms and challenges of these strategies will be discussed using data from clinical trials listed in databases. These therapeutic combinations also lead to questions on how to optimize their formulation and delivery to induce a therapeutic polyclonal response in patients with cancer. Abstract Monoclonal antibodies have revolutionized the treatment of many diseases, but their clinical efficacy remains limited in some other cases. Pre-clinical and clinical trials have shown that combinations of antibodies that bind to the same target (homo-combinations) or to different targets (hetero-combinations) to mimic the polyclonal humoral immune response improve their therapeutic effects in cancer. The approval of the trastuzumab/pertuzumab combination for breast cancer and then of the ipilimumab/nivolumab combination for melanoma opened the way to novel antibody combinations or oligoclonal antibody mixtures as more effective biologics for cancer management. We found more than 300 phase II/III clinical trials on antibody combinations, with/without chemotherapy, radiotherapy, small molecules or vaccines, in the ClinicalTrials.gov database. Such combinations enhance the biological responses and bypass the resistance mechanisms observed with antibody monotherapy. Usually, such antibody combinations are administered sequentially as separate formulations. Combined formulations have also been developed in which separately produced antibodies are mixed before administration or are produced simultaneously in a single cell line or a single batch of different cell lines as a polyclonal master cell bank. The regulation, toxicity and injection sequence of these oligoclonal antibody mixtures still need to be addressed in order to optimize their delivery and their therapeutic effects.
Collapse
Affiliation(s)
- Christel Larbouret
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Institut Régional du Cancer de Montpellier (ICM), Inserm U1194, Université de Montpellier, 34298 Montpellier, France; (L.G.); (A.P.); (T.C.)
- Correspondence: ; Tel.: +33-411-283-110
| | - Laurent Gros
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Institut Régional du Cancer de Montpellier (ICM), Inserm U1194, Université de Montpellier, 34298 Montpellier, France; (L.G.); (A.P.); (T.C.)
- Centre National de la Recherche Scientifique (CNRS), 75016 Paris, France
| | - André Pèlegrin
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Institut Régional du Cancer de Montpellier (ICM), Inserm U1194, Université de Montpellier, 34298 Montpellier, France; (L.G.); (A.P.); (T.C.)
| | - Thierry Chardès
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Institut Régional du Cancer de Montpellier (ICM), Inserm U1194, Université de Montpellier, 34298 Montpellier, France; (L.G.); (A.P.); (T.C.)
- Centre National de la Recherche Scientifique (CNRS), 75016 Paris, France
| |
Collapse
|
19
|
De Meulenaere A, Vermassen T, Creytens D, De Keukeleire S, Delahaye T, Deron P, Duprez F, Fung S, Pauwels P, Ferdinande L, Rottey S. An open-label, nonrandomized, phase Ib feasibility study of cusatuzumab in patients with nasopharyngeal carcinoma. Clin Transl Sci 2021; 14:2300-2313. [PMID: 34405542 PMCID: PMC8604223 DOI: 10.1111/cts.13089] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 04/14/2021] [Accepted: 04/16/2021] [Indexed: 01/03/2023] Open
Abstract
CD70 is expressed in up to 80% of nasopharyngeal carcinoma (NPC) cases. Cusatuzumab is a humanized anti‐CD70 monoclonal antibody, with dual action mechanisms: induction of cytotoxicity against CD70+ tumor cells and reduction in CD70‐CD27 signaling mediated immune evasion. The aim of this study was to assess the safety, pharmacokinetic profile, immunogenicity, pharmacodynamic profile, and preliminary activity of cusatuzumab in advanced NPC. Eleven patients were enrolled: one patient was assigned to arm A (adjuvant cusatuzumab monotherapy after curative chemoradiation), nine patients to arm B (cusatuzumab monotherapy; noncurative setting), and one patient to arm C (cusatuzumab + chemotherapy; noncurative setting); irrespective of tumoral CD70 expression. Both patients in arms A and C completed the study. All patients in arm B discontinued at an early stage. Five patients experienced grade greater than or equal to 3 nondrug related treatment‐emergent adverse events, most commonly fatigue and pneumonia (18%). An infusion‐related reaction was observed in two of 11 patients. Laboratory results showed no trend over time. Seven patients were eligible for response evaluation. No objective response to cusatuzumab was observed with stable disease being the best response. The current study indicates that the safety profile of cusatuzumab (with or without concurrent chemotherapy) is manageable in patients with advanced NPC, which is consistent with known safety profile. Limited activity of cusatuzumab in advanced NPC was observed. Combination therapies of cusatuzumab and other types of therapy should be explored for the improvement of activity in NPC and other CD70‐expressing malignancies.
Collapse
Affiliation(s)
| | - Tijl Vermassen
- Department of Medical Oncology, University Hospital Ghent, Ghent, Belgium.,Drug Research Unit Ghent, University Hospital Ghent, Ghent, Belgium.,Cancer Research Unit Ghent (CRIG), Ghent, Belgium
| | - David Creytens
- Cancer Research Unit Ghent (CRIG), Ghent, Belgium.,Department of Pathology, University Hospital Ghent, Ghent, Belgium
| | | | | | - Philippe Deron
- Department Of Head and Neck Surgery, University Hospital Ghent, Ghent, Belgium
| | - Fréderic Duprez
- Department of Radiation Oncology, University Hospital Ghent, Ghent, Belgium
| | - Samson Fung
- argenx B.V., Ghent, Belgium.,Fung Consulting Healthcare and Life Sciences, Eching, Germany
| | - Patrick Pauwels
- Centre for Oncological Research (CORE), University of Antwerp, and University Hospital Antwerp, Edegem, Belgium
| | - Liesbeth Ferdinande
- Cancer Research Unit Ghent (CRIG), Ghent, Belgium.,Department of Pathology, University Hospital Ghent, Ghent, Belgium
| | - Sylvie Rottey
- Department of Medical Oncology, University Hospital Ghent, Ghent, Belgium.,Drug Research Unit Ghent, University Hospital Ghent, Ghent, Belgium.,Cancer Research Unit Ghent (CRIG), Ghent, Belgium
| |
Collapse
|
20
|
Triple negative breast cancer and non-small cell lung cancer: Clinical challenges and nano-formulation approaches. J Control Release 2021; 337:27-58. [PMID: 34273417 DOI: 10.1016/j.jconrel.2021.07.014] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 07/08/2021] [Accepted: 07/10/2021] [Indexed: 02/06/2023]
Abstract
Triple negative breast cancer (TNBC) and non-small cell lung cancer (NSCLC) are amongst the most aggressive forms of solid tumors. TNBC is highlighted by absence of genetic components of progesterone receptor, HER2/neu and estrogen receptor in breast cancer. NSCLC is characterized by integration of malignant carcinoma into respiratory system. Both cancers are associated with poor median and overall survival rates with low progression free survival with high incidences of relapse. These cancers are characterized by tumor heterogeneity, genetic mutations, generation of cancer-stem cells, immune-resistance and chemoresistance. Further, these neoplasms have been reported for tumor cross-talk into second primary cancers for each other. Current chemotherapeutic regimens include usage of multiple agents in tandem to affect tumor cells through multiple mechanisms with various such combinations being clinically tested. However, lack of controlled delivery and effective temporospatial presence of chemotherapeutics has resulted in suboptimal therapeutic response. Consequently, passive targeted albumin bound paclitaxel and PEGylated liposomal doxorubicin have been clinically used and tested with newer drugs for improved therapeutic efficacy in these cancers. Active targeting of nanocarriers against surface overexpressed proteins in both neoplasms have been explored. However, use of single agent nanoparticulate formulations against both cancers have failed to elicit desired outcomes. This review aims to identify clinical unmet need in these cancers while establishing a correlation with tested nano-formulation approaches and issues with preclinical to clinical translation. Lipid and polymer-based drug-drug and drug-gene combinatorial nanocarriers delivering multiple chemotherapeutics simultaneously to desired site of action have been detailed. Finally, emerging opportunities such as pharmacological targets (immune check point and epigentic modulators) as well as gene-based modulation (siRNA/CRISPR/Cas9) and the nano-formulation challenges for effective treatment of both cancers have been explored.
Collapse
|
21
|
Gurbatri CR, Lia I, Vincent R, Coker C, Castro S, Treuting PM, Hinchliffe TE, Arpaia N, Danino T. Engineered probiotics for local tumor delivery of checkpoint blockade nanobodies. Sci Transl Med 2021; 12:12/530/eaax0876. [PMID: 32051224 DOI: 10.1126/scitranslmed.aax0876] [Citation(s) in RCA: 259] [Impact Index Per Article: 86.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 10/25/2019] [Accepted: 01/13/2020] [Indexed: 12/25/2022]
Abstract
Checkpoint inhibitors have revolutionized cancer therapy but only work in a subset of patients and can lead to a multitude of toxicities, suggesting the need for more targeted delivery systems. Because of their preferential colonization of tumors, microbes are a natural platform for the local delivery of cancer therapeutics. Here, we engineer a probiotic bacteria system for the controlled production and intratumoral release of nanobodies targeting programmed cell death-ligand 1 (PD-L1) and cytotoxic T lymphocyte-associated protein-4 (CTLA-4) using a stabilized lysing release mechanism. We used computational modeling coupled with experimental validation of lysis circuit dynamics to determine the optimal genetic circuit parameters for maximal therapeutic efficacy. A single injection of this engineered system demonstrated an enhanced therapeutic response compared to analogous clinically relevant antibodies, resulting in tumor regression in syngeneic mouse models. Supporting the potentiation of a systemic immune response, we observed a relative increase in activated T cells, an abscopal effect, and corresponding increases in systemic T cell memory populations in mice treated with probiotically delivered checkpoint inhibitors. Last, we leveraged the modularity of our platform to achieve enhanced therapeutic efficacy in a poorly immunogenic syngeneic mouse model through effective combinations with a probiotically produced cytokine, granulocyte-macrophage colony-stimulating factor (GM-CSF). Together, these results demonstrate that our engineered probiotic system bridges synthetic biology and immunology to improve upon checkpoint blockade delivery.
Collapse
Affiliation(s)
- Candice R Gurbatri
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Ioana Lia
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Rosa Vincent
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Courtney Coker
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Samuel Castro
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Piper M Treuting
- Department of Comparative Medicine, University of Washington, Seattle, WA 98195, USA
| | - Taylor E Hinchliffe
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Nicholas Arpaia
- Department of Microbiology & Immunology, Vagelos College of Physicians and Surgeons of Columbia University, New York, NY 10032, USA.,Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10027, USA
| | - Tal Danino
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA. .,Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10027, USA.,Data Science Institute, Columbia University, New York, NY 10027, USA
| |
Collapse
|
22
|
Doostmohammadi M, Forootanfar H, Ramakrishna S. New Strategies for Safe Cancer Therapy Using Electrospun Nanofibers: A Short Review. Mini Rev Med Chem 2021; 20:1272-1286. [PMID: 32400330 DOI: 10.2174/1389557520666200513120924] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 12/10/2019] [Accepted: 02/14/2020] [Indexed: 12/26/2022]
Abstract
Electrospun nanofibers regarding their special features, including high drug loading capacity, high surface to volume area, flexibility, and ease of production and operation, are of great interest for being used in tissue engineering, and drug delivery approaches. In this context, several studies have been done for the production of biodegradable and biocompatible scaffolds containing different anticancer agents for fighting with solid tumors. Surprisingly, these scaffolds are able to deliver different combinations of drugs and agents, such as nanoparticles and release them in a time dependent manner. Here in this review, we summarize the principles of electrospinning and their uses in entrapment of drugs and anti-proliferative agents suitable for cancer therapy. The latest studies performed on treating cancer using electrospinning are mentioned and their advantages and disadvantages over conventional treatment methods are discussed.
Collapse
Affiliation(s)
- Mohsen Doostmohammadi
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Hamid Forootanfar
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Seeram Ramakrishna
- Department of Mechanical Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Singapore
| |
Collapse
|
23
|
Golpour M, Vatanpour P, Amini M, Saeedi M, Hafezi N, Rafiei A. The Perspective of Therapeutic Antibody Marketing in Iran: Trend and Estimation by 2025. Adv Pharmacol Pharm Sci 2021; 2021:5569590. [PMID: 33860229 PMCID: PMC8026318 DOI: 10.1155/2021/5569590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/11/2021] [Accepted: 03/17/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Monoclonal antibodies with high efficiency and specificity are one of the best strategies to diagnose and treat a variety of diseases such as cancer, autoimmunity, and inflammatory diseases. The market for monoclonal therapeutic antibodies (MTAs) has grown dramatically in the past decade. OBJECTIVE Given the importance of these issues, developing countries spend a high cost on importing or producing MTAs annually. This study intends to examine the market of monoclonal therapeutic antibodies in Iran and predict the future growth rate of this market using the obtained data. METHODS Data on the status of MTAs in the country (from 2008 to 2018) were obtained from the Food and Drug Deputy of Mazandaran University of Medical Sciences. The market status of MTAs was studied based on the dosage forms, application, and price. Then, the market outlook was predicted up to year 2025. RESULTS The results showed that 58.8% of all MTAs were humanized, and 86% of all antibody-based drugs were used to treat cancer. Sales of MTA-based medications will reach $454 million by 2025 and are projected to grow significantly in the future. CONCLUSION Given the increasing technology of the production of MTAs and their use in targeted therapies worldwide, their consumption market in Iran is expected to grow significantly.
Collapse
Affiliation(s)
- Monireh Golpour
- Molecular and Cell Biology Research Center, Student Research Committee, Faculty of Medicine, Mazandaran University of Medical Science, Sari, Iran
| | - Pouya Vatanpour
- Molecular and Cell Biology Research Center, Student Research Committee, Faculty of Medicine, Mazandaran University of Medical Science, Sari, Iran
- Oneocean Company, Oslo, Norway
| | - Mina Amini
- Food and Drug Deputy, Mazandaran University of Medical Science, Sari, Iran
| | - Majid Saeedi
- Departments of Pharmaceutics and Medicinal Chemistry, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Nasim Hafezi
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Alireza Rafiei
- Molecular and Cell Biology Research Center, Student Research Committee, Faculty of Medicine, Mazandaran University of Medical Science, Sari, Iran
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
24
|
Sun S, Ding Z, Yang X, Zhao X, Zhao M, Gao L, Chen Q, Xie S, Liu A, Yin S, Xu Z, Lu X. Nanobody: A Small Antibody with Big Implications for Tumor Therapeutic Strategy. Int J Nanomedicine 2021; 16:2337-2356. [PMID: 33790553 PMCID: PMC7997558 DOI: 10.2147/ijn.s297631] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/16/2021] [Indexed: 12/15/2022] Open
Abstract
The development of monoclonal antibody treatments for successful tumor-targeted therapies took several decades. However, the efficacy of antibody-based therapy is still confined and desperately needs further improvement. Nanobodies are the recombinant variable domains of heavy-chain-only antibodies, with many unique properties such as small size (~15kDa), excellent solubility, superior stability, ease of manufacture, quick clearance from blood, and deep tissue penetration, which gain increasing acceptance as therapeutical tools and are considered also as building blocks for chimeric antigen receptors as well as for targeted drug delivery. Thus, one of the promising novel developments that may address the deficiency of monoclonal antibody-based therapies is the utilization of nanobodies. This article provides readers the significant factors that the structural and biochemical properties of nanobodies and the research progress on nanobodies in the fields of tumor treatment, as well as their application prospect.
Collapse
Affiliation(s)
- Shuyang Sun
- International Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
- School of Stomatology, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Ziqiang Ding
- International Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Xiaomei Yang
- International Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
- School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Xinyue Zhao
- International Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
- School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Minlong Zhao
- International Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
- School of Stomatology, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Li Gao
- International Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
- School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Qu Chen
- International Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
- School of Stomatology, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Shenxia Xie
- International Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
- Department of Pharmacology, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Aiqun Liu
- International Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Shihua Yin
- International Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Zhiping Xu
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, QLD, 4072, Australia
| | - Xiaoling Lu
- International Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
- School of Stomatology, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| |
Collapse
|
25
|
Hatterer E, Chauchet X, Richard F, Barba L, Moine V, Chatel L, Broyer L, Pontini G, Bautzova T, Juan F, Calloud S, Bosson N, Charreton M, Masternak K, Buatois V, Shang L. Targeting a membrane-proximal epitope on mesothelin increases the tumoricidal activity of a bispecific antibody blocking CD47 on mesothelin-positive tumors. MAbs 2021; 12:1739408. [PMID: 32191151 PMCID: PMC7153835 DOI: 10.1080/19420862.2020.1739408] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Mesothelin (MSLN) is a cell surface glycoprotein overexpressed in several solid malignancies, including gastric, lung, mesothelioma, pancreatic and ovarian cancers. While several MSLN-targeting therapeutic approaches are in development, only limited efficacy has been achieved in patients. A potential shortcoming of several described antibody-based approaches is that they target the membrane distal region of MSLN and, additionally, are known to be handicapped by the high levels of circulating soluble MSLN in patients. We show here, using monoclonal antibodies (mAbs) targeting different MSLN-spanning epitopes, that the membrane-proximal region resulted in more efficient killing of MSLN-positive tumor cells in antibody-dependent cell-mediated cytotoxicity (ADCC) assays. Surprisingly, no augmented killing was observed in antibody-dependent cellular phagocytosis (ADCP) by mAbs targeting this membrane-proximal region. To further increase the ADCP potential, we, therefore, generated bispecific antibodies (bsAbs) coupling a high-affinity MSLN binding arm to a blocking CD47 arm. Here, targeting the membrane-proximal domain of MSLN demonstrated enhanced ADCP activity compared to membrane-distal domains when the bsAbs were used in in vitro phagocytosis killing assays. Importantly, the superior anti-tumor activity was also translated in xenograft tumor models. Furthermore, we show that the bsAb approach targeting the membrane-proximal epitope of MSLN optimized ADCC activity by augmenting FcγR-IIIA activation and enhanced ADCP via a more efficient blockade of the CD47/SIRPα axis.
Collapse
Affiliation(s)
- Eric Hatterer
- Light Chain Bioscience, Novimmune S.A., Plan-les-Ouates, Switzerland
| | - Xavier Chauchet
- Light Chain Bioscience, Novimmune S.A., Plan-les-Ouates, Switzerland
| | - Françoise Richard
- Light Chain Bioscience, Novimmune S.A., Plan-les-Ouates, Switzerland
| | - Leticia Barba
- Light Chain Bioscience, Novimmune S.A., Plan-les-Ouates, Switzerland
| | - Valéry Moine
- Light Chain Bioscience, Novimmune S.A., Plan-les-Ouates, Switzerland
| | - Laurence Chatel
- Light Chain Bioscience, Novimmune S.A., Plan-les-Ouates, Switzerland
| | - Lucile Broyer
- Light Chain Bioscience, Novimmune S.A., Plan-les-Ouates, Switzerland
| | | | - Tereza Bautzova
- Light Chain Bioscience, Novimmune S.A., Plan-les-Ouates, Switzerland
| | - Flora Juan
- Light Chain Bioscience, Novimmune S.A., Plan-les-Ouates, Switzerland
| | - Sebastien Calloud
- Light Chain Bioscience, Novimmune S.A., Plan-les-Ouates, Switzerland
| | - Nicolas Bosson
- Light Chain Bioscience, Novimmune S.A., Plan-les-Ouates, Switzerland
| | - Maud Charreton
- Light Chain Bioscience, Novimmune S.A., Plan-les-Ouates, Switzerland
| | | | - Vanessa Buatois
- Light Chain Bioscience, Novimmune S.A., Plan-les-Ouates, Switzerland
| | - Limin Shang
- Light Chain Bioscience, Novimmune S.A., Plan-les-Ouates, Switzerland
| |
Collapse
|
26
|
Anderson TS, Wooster AL, La-Beck NM, Saha D, Lowe DB. Antibody-drug conjugates: an evolving approach for melanoma treatment. Melanoma Res 2021; 31:1-17. [PMID: 33165241 DOI: 10.1097/cmr.0000000000000702] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Melanoma continues to be an aggressive and deadly form of skin cancer while therapeutic options are continuously developing in an effort to provide long-term solutions for patients. Immunotherapeutic strategies incorporating antibody-drug conjugates (ADCs) have seen varied levels of success across tumor types and represent a promising approach for melanoma. This review will explore the successes of FDA-approved ADCs to date compared to the ongoing efforts of melanoma-targeting ADCs. The challenges and opportunities for future therapeutic development are also examined to distinguish how ADCs may better impact individuals with malignancies such as melanoma.
Collapse
Affiliation(s)
| | | | - Ninh M La-Beck
- Departments of Immunotherapeutics and Biotechnology
- Pharmacy Practice, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, Texas, USA
| | | | - Devin B Lowe
- Departments of Immunotherapeutics and Biotechnology
| |
Collapse
|
27
|
Lu L, Jiang J, Zhan M, Zhang H, Wang QT, Sun SN, Guo XK, Yin H, Wei Y, Li SY, Liu JO, Li Y, He YW. Targeting Tumor-Associated Antigens in Hepatocellular Carcinoma for Immunotherapy: Past Pitfalls and Future Strategies. Hepatology 2021; 73:821-832. [PMID: 32767586 DOI: 10.1002/hep.31502] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/23/2020] [Accepted: 07/02/2020] [Indexed: 12/15/2022]
Affiliation(s)
- Ligong Lu
- Zhuhai Interventional Medical CenterZhuhai Precision Medical CenterZhuhai People's HospitalZhuhai Hospital Affiliated with Jinan UniversityZhuhaiGuangdong ProvinceP.R. China
| | - Jun Jiang
- Tricision Biotherapeutic Inc. Jinwan DistrictZhuhaiChina
| | - Meixiao Zhan
- Zhuhai Interventional Medical CenterZhuhai Precision Medical CenterZhuhai People's HospitalZhuhai Hospital Affiliated with Jinan UniversityZhuhaiGuangdong ProvinceP.R. China
| | - Hui Zhang
- First Affiliated HospitalChina Medical UniversityShenyangChina
| | - Qian-Ting Wang
- Tricision Biotherapeutic Inc. Jinwan DistrictZhuhaiChina
| | - Sheng-Nan Sun
- Tricision Biotherapeutic Inc. Jinwan DistrictZhuhaiChina
| | - Xiao-Kai Guo
- Tricision Biotherapeutic Inc. Jinwan DistrictZhuhaiChina
| | - Hua Yin
- Zhuhai Interventional Medical CenterZhuhai Precision Medical CenterZhuhai People's HospitalZhuhai Hospital Affiliated with Jinan UniversityZhuhaiGuangdong ProvinceP.R. China
| | - Yadong Wei
- Department of Pharmacology and Molecular SciencesJohns Hopkins University School of MedicineBaltimoreMD
| | - Shi-You Li
- Tricision Biotherapeutic Inc. Jinwan DistrictZhuhaiChina
| | - Jun O Liu
- Department of Pharmacology and Molecular SciencesJohns Hopkins University School of MedicineBaltimoreMD
| | - Yong Li
- Zhuhai Interventional Medical CenterZhuhai Precision Medical CenterZhuhai People's HospitalZhuhai Hospital Affiliated with Jinan UniversityZhuhaiGuangdong ProvinceP.R. China
| | - You-Wen He
- Department of ImmunologyDuke University Medical University Medical CenterDurhamNC
| |
Collapse
|
28
|
Cartaxo AL, Almeida J, Gualda EJ, Marsal M, Loza-Alvarez P, Brito C, Isidro IA. A computational diffusion model to study antibody transport within reconstructed tumor microenvironments. BMC Bioinformatics 2020; 21:529. [PMID: 33203360 PMCID: PMC7672975 DOI: 10.1186/s12859-020-03854-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Accepted: 10/30/2020] [Indexed: 12/11/2022] Open
Abstract
Background Antibodies revolutionized cancer treatment over the past decades. Despite their successfully application, there are still challenges to overcome to improve efficacy, such as the heterogeneous distribution of antibodies within tumors. Tumor microenvironment features, such as the distribution of tumor and other cell types and the composition of the extracellular matrix may work together to hinder antibodies from reaching the target tumor cells. To understand these interactions, we propose a framework combining in vitro and in silico models. We took advantage of in vitro cancer models previously developed by our group, consisting of tumor cells and fibroblasts co-cultured in 3D within alginate capsules, for reconstruction of tumor microenvironment features.
Results In this work, an experimental-computational framework of antibody transport within alginate capsules was established, assuming a purely diffusive transport, combined with an exponential saturation effect that mimics the saturation of binding sites on the cell surface. Our tumor microenvironment in vitro models were challenged with a fluorescent antibody and its transport recorded using light sheet fluorescence microscopy. Diffusion and saturation parameters of the computational model were adjusted to reproduce the experimental antibody distribution, with root mean square error under 5%. This computational framework is flexible and can simulate different random distributions of tumor microenvironment elements (fibroblasts, cancer cells and collagen fibers) within the capsule. The random distribution algorithm can be tuned to follow the general patterns observed in the experimental models. Conclusions We present a computational and microscopy framework to track and simulate antibody transport within the tumor microenvironment that complements the previously established in vitro models platform. This framework paves the way to the development of a valuable tool to study the influence of different components of the tumor microenvironment on antibody transport.
Collapse
Affiliation(s)
- Ana Luísa Cartaxo
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Jaime Almeida
- Departamento de Geologia, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal.,Instituto Dom Luiz, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - Emilio J Gualda
- ICFO, Institut de Ciències Fotòniques, The Barcelona Institute of Science and Technology, Castelldefels, Barcelona, Spain
| | - Maria Marsal
- ICFO, Institut de Ciències Fotòniques, The Barcelona Institute of Science and Technology, Castelldefels, Barcelona, Spain
| | - Pablo Loza-Alvarez
- ICFO, Institut de Ciències Fotòniques, The Barcelona Institute of Science and Technology, Castelldefels, Barcelona, Spain
| | - Catarina Brito
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Inês A Isidro
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal. .,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal.
| |
Collapse
|
29
|
Bose M, Mukherjee P. Potential of Anti-MUC1 Antibodies as a Targeted Therapy for Gastrointestinal Cancers. Vaccines (Basel) 2020; 8:E659. [PMID: 33167508 PMCID: PMC7712407 DOI: 10.3390/vaccines8040659] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/27/2020] [Accepted: 11/03/2020] [Indexed: 02/06/2023] Open
Abstract
Gastrointestinal cancers (GI) account for 26% of cancer incidences globally and 35% of all cancer-related deaths. The main challenge is to target cancer specific antigens. Mucins are heavily O-glycosylated proteins overexpressed in different cancers. The transmembrane glycoprotein MUC1 is the most likeable target for antibodies, owing to its specific overexpression and aberrant glycosylation in many types of cancers. For the past 30 years, MUC1 has remained a possible diagnostic marker and therapeutic target. Despite initiation of numerous clinical trials, a comprehensively effective therapy with clinical benefit is yet to be achieved. However, the interest in MUC1 as a therapeutic target remains unaltered. For all translational studies, it is important to incorporate updated relevant research findings into therapeutic strategies. In this review we present an overview of the antibodies targeting MUC1 in GI cancers, their potential role in immunotherapy (i.e., antibody-drug and radioimmunoconjugates, CAR-T cells), and other novel therapeutic strategies. We also present our perspectives on how the mechanisms of action of different anti-MUC1 antibodies can target specific hallmarks of cancer and therefore be utilized as a combination therapy for better clinical outcomes.
Collapse
Affiliation(s)
- Mukulika Bose
- Department of Biological Sciences, University of North Carolina, Charlotte, NC 28223, USA;
| | | |
Collapse
|
30
|
Sher YP, Chai KM, Chen WC, Shen KY, Chen IH, Lee MH, Chiu FF, Liu SJ. A Polypeptide of Tumor-Associated Antigen L6 with Intrinsic Adjuvant Activity Enhances Antitumor Immunity. Vaccines (Basel) 2020; 8:vaccines8040620. [PMID: 33096846 PMCID: PMC7711899 DOI: 10.3390/vaccines8040620] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/14/2020] [Accepted: 10/18/2020] [Indexed: 12/03/2022] Open
Abstract
Peptide vaccines are safe, and aim to elicit and expand tumor-specific immunity so as to eradicate tumors. However, achieving strong and long-lasting anti-tumor immunity with peptide vaccines for the antigen-specific treatment of cancer is challenging, in part because their efficacy depends on strong adjuvants or immunomodulators. We approached this problem by conjugating an epitope-based cancer vaccine with a lipidated sequence (an immunomodulator) to elicit a strong immune response. Lipidated and non-lipidated polyepitope proteins were generated that contained the universal T helper cell epitope (pan-DR), B cell epitopes, and the extended loop sequence of extracellular domain 2 of tumor-associated antigen L6 (TAL6). We show that the lipidated polyepitope cancer vaccine can activate bone marrow-derived dendritic cells, and trigger effective antigen-specific antibody and T helper cell responses, more effectively than the non-lipidated vaccine. Moreover, potent T cell immune responses were elicited in mice inoculated with the lipidated polyepitope cancer vaccine, providing protective antitumor immunity in mice bearing TAL6 tumors. Our study demonstrates that a lipidated polyepitope cancer vaccine could be employed to generate potent anti-tumor immune responses, including humoral and cellular immunity, which could be beneficial in the treatment of TAL6+ cancer.
Collapse
Affiliation(s)
- Yuh-Pyng Sher
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404, Taiwan
- Chinese Medicine Research Center, China Medical University, Taichung 404, Taiwan
- Research Center for Chinese Herbal Medicine, China Medical University, Taichung 404, Taiwan
- Center for Molecular Medicine, China Medical University Hospital, Taichung 404, Taiwan
| | - Kit Man Chai
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 350, Taiwan
| | - Wen-Ching Chen
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404, Taiwan
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 350, Taiwan
| | - Kuan-Yin Shen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 350, Taiwan
| | - I-Hua Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 350, Taiwan
| | - Ming-Hui Lee
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 350, Taiwan
| | - Fang-Feng Chiu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 350, Taiwan
| | - Shih-Jen Liu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404, Taiwan
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 350, Taiwan
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
31
|
Pourjafar M, Samadi P, Saidijam M. MUC1 antibody-based therapeutics: the promise of cancer immunotherapy. Immunotherapy 2020; 12:1269-1286. [PMID: 33019839 DOI: 10.2217/imt-2020-0019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Antibody-based targeted therapies have been able to target cancers with enhanced specificity and high efficacy. In this regard, identifying cancer markers (antigens) that are only present (tumor-specific antigens) or have an increased expression (tumor-associated antigen) on the surface of cancer cells is a crucial step for targeted cancer treatment. Various cancer antigens have already been used for therapeutic and diagnostic purposes. MUC1 is one of the most important tumor markers with high levels of expression in various solid tumors which makes it as a potential target for antibody-based therapies. This review discusses preclinical and clinical results from various platforms based on monoclonal antibodies, nanobodies as well as bispecific antibodies against MUC1. We also highlight unmet challenges that must be overcome to generate more effective cancer immunotherapy strategies.
Collapse
Affiliation(s)
- Mona Pourjafar
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.,Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Pouria Samadi
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.,Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Massoud Saidijam
- Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
32
|
Song X, Qi W, Guo J, Sun L, Ding A, Zhao G, Li H, Qiu W, Lv J. Immune checkpoint inhibitor combination therapy for gastric cancer: Research progress. Oncol Lett 2020; 20:46. [PMID: 32802168 PMCID: PMC7412728 DOI: 10.3892/ol.2020.11905] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 06/25/2020] [Indexed: 12/24/2022] Open
Abstract
Gastric cancer is one of the most common types of cancer; notably, gastric cancer is one of the top five malignancies with regards to incidence and mortality rates. The symptoms of early gastric cancer are not typical, exhibiting only slight upper abdominal discomfort. When the symptoms become more obvious, the lesion has usually progressed to an advanced stage. Notably, >90% of inpatients already have locally advanced or metastatic gastric cancer at the time of initial diagnosis, with limited treatment options for advanced gastric cancer. These options include chemotherapy, targeted therapy and immune checkpoint inhibitors (ICIs). With regards to ICIs, the clinical benefit of monotherapy for advanced gastric cancer is limited; however, combinations of ICIs and other therapies may have clinical benefit. Relevant clinical studies have demonstrated that combinations of ICIs with chemotherapy, anti-vascular targeted therapy or other molecular targeted therapies, and the use of two ICIs, improve outcomes for patients with advanced gastric cancer. This article is a review of progress in the use of ICIs in combination with other therapies for the treatment of gastric cancer. The purpose of this article was to advance gastric cancer immunotherapy and to improve the overall therapeutic benefit for patients with advanced gastric cancer.
Collapse
Affiliation(s)
- Xiaoxu Song
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Weiwei Qi
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Jing Guo
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Libin Sun
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Aiping Ding
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Guanghui Zhao
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Hui Li
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Wensheng Qiu
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Jing Lv
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| |
Collapse
|
33
|
Pecetta S, Finco O, Seubert A. Quantum leap of monoclonal antibody (mAb) discovery and development in the COVID-19 era. Semin Immunol 2020; 50:101427. [PMID: 33277154 PMCID: PMC7670927 DOI: 10.1016/j.smim.2020.101427] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/16/2020] [Accepted: 11/16/2020] [Indexed: 01/08/2023]
Abstract
In recent years the global market for monoclonal antibodies (mAbs) became a multi-billion-dollar business. This success is mainly driven by treatments in the oncology and autoimmune space. Instead, development of effective mAbs against infectious diseases has been lagging behind. For years the high production cost and limited efficacy have blocked broader application of mAbs in the infectious disease space, which instead has been dominated for almost a century by effective and cheap antibiotics and vaccines. Only very few mAbs against RSV, anthrax, Clostridium difficile or rabies have reached the market. This is about to change. The development of urgently needed and highly effective mAbs as preventive and therapeutic treatments against a variety of pathogens is gaining traction. Vast advances in mAb isolation, engineering and production have entirely shifted the cost-efficacy balance. MAbs against devastating diseases like Ebola, HIV and other complex pathogens are now within reach. This trend is further accelerated by ongoing or imminent health crises like COVID-19 and antimicrobial resistance (AMR), where antibodies could be the last resort. In this review we will retrace the history of antibodies from the times of serum therapy to modern mAbs and lay out how the current run for effective treatments against COVID-19 will lead to a quantum leap in scientific, technological and health care system innovation around mAb treatments for infectious diseases.
Collapse
|
34
|
Zahavi D, Weiner L. Monoclonal Antibodies in Cancer Therapy. Antibodies (Basel) 2020; 9:E34. [PMID: 32698317 PMCID: PMC7551545 DOI: 10.3390/antib9030034] [Citation(s) in RCA: 307] [Impact Index Per Article: 76.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/11/2020] [Accepted: 07/04/2020] [Indexed: 12/19/2022] Open
Abstract
Monoclonal antibody-based immunotherapy is now considered to be a main component of cancer therapy, alongside surgery, radiation, and chemotherapy. Monoclonal antibodies possess a diverse set of clinically relevant mechanisms of action. In addition, antibodies can directly target tumor cells while simultaneously promoting the induction of long-lasting anti-tumor immune responses. The multifaceted properties of antibodies as a therapeutic platform have led to the development of new cancer treatment strategies that will have major impacts on cancer care. This review focuses on the known mechanisms of action, current clinical applications for the treatment of cancer, and mechanisms of resistance of monoclonal antibody therapy. We further discuss how monoclonal antibody-based strategies have moved towards enhancing anti-tumor immune responses by targeting immune cells instead of tumor antigens as well as some of the current combination therapies.
Collapse
Affiliation(s)
- David Zahavi
- Tumor Biology Training Program, Lombardi Comprehensive Cancer Center, Georgetown University, Medical Center, 3800 Reservoir Rd NW, Washington, DC 20007, USA;
| | - Louis Weiner
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Medical Center, 3800 Reservoir Rd NW, Washington, DC 20007, USA
| |
Collapse
|
35
|
Balibegloo M, Rezaei N. Development and clinical application of bispecific antibody in the treatment of colorectal cancer. Expert Rev Clin Immunol 2020; 16:689-709. [PMID: 32536227 DOI: 10.1080/1744666x.2020.1783249] [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: 12/24/2022]
Abstract
INTRODUCTION Treatment of colorectal cancer as one of the most commonly diagnosed and a frequent cause of cancer-related deaths is of great challenges in health-related issues. AREAS COVERED Immunotherapy is the fourth pillar of cancer treatment which provides more novel therapeutic options with expanding investigational potentials. One of the modalities in immunotherapy is the use of bispecific antibodies. Despite demonstrating many promising roles, it still needs more advanced studies to identify the actual pros and cons. In this review, the application of bispecific antibody in the treatment of colorectal cancer has been explained, based on preclinical and clinical studies. The literature search was conducted mainly through PubMed in June and September 2019. EXPERT OPINION Bispecific antibody is in its early stages in colorectal cancer treatment, requiring modern technologies in manufacturing, better biomarkers and more specific target antigens, more studies on individual genetic variations, and conducting later phase clinical trials and systematic reviews to achieve better survival benefits.
Collapse
Affiliation(s)
- Maryam Balibegloo
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences , Tehran, Iran.,Cancer Immunology Project (CIP), Universal Scientific Education & Research Network (USERN) , Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences , Tehran, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences , Tehran, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN) , Tehran, Iran
| |
Collapse
|
36
|
Gauzy-Lazo L, Sassoon I, Brun MP. Advances in Antibody–Drug Conjugate Design: Current Clinical Landscape and Future Innovations. SLAS DISCOVERY 2020; 25:843-868. [DOI: 10.1177/2472555220912955] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The targeted delivery of potent cytotoxic molecules into cancer cells is considered a promising anticancer strategy. The design of clinically effective antibody–drug conjugates (ADCs), in which biologically active drugs are coupled through chemical linkers to monoclonal antibodies, has presented challenges for pharmaceutical researchers. After 30 years of intensive research and development activities, only seven ADCs have been approved for clinical use; two have received fast-track designation and two breakthrough therapy designation from the Food and Drug Administration. There is continued interest in the field, as documented by the growing number of candidates in clinical development. This review aims to summarize the most recent innovations that have been applied to the design of ADCs undergoing early- and late-stage clinical trials. Discovery and rational optimization of new payloads, chemical linkers, and antibody formats have improved the therapeutic index of next-generation ADCs, ultimately resulting in improved clinical benefit for the patients.
Collapse
Affiliation(s)
| | - Ingrid Sassoon
- Immuno-Oncology Therapeutic Area, Sanofi, Vitry-sur-Seine, France
| | | |
Collapse
|
37
|
Srikoti M, Bolgar MS, Kazakevich Y. Hybrid mode of hydrophobic interaction chromatography of monoclonal antibodies and related biomolecules: Influence of elution conditions on chromatographic performance using poly (alkyl aspartamide) silica columns. J Chromatogr B Analyt Technol Biomed Life Sci 2020; 1140:121984. [DOI: 10.1016/j.jchromb.2020.121984] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 01/09/2020] [Accepted: 01/13/2020] [Indexed: 11/24/2022]
|
38
|
Khanna V, Panyam J, Griffith TS. Exploiting antibody biology for the treatment of cancer. Immunotherapy 2020; 12:255-267. [DOI: 10.2217/imt-2019-0118] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Over the last decade, antibodies have become an important component in the arsenal of cancer therapeutics. High-specificity, low off-target effects, desirable pharmacokinetics and high success rate are a few of the many attributes that make antibodies amenable for development as drugs. To design antibodies for successful clinical applications, however, it is critical to have an understanding of their structure, functions, mechanisms of action and pharmacokinetic/pharmacodynamic properties. This review highlights some of these key aspects, as well as certain limitations encountered, with monoclonal antibody therapy. Further, we discuss rational combination therapies for clinical applications, some of which could help overcome the limitations.
Collapse
Affiliation(s)
- Vidhi Khanna
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jayanth Panyam
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- School of Pharmacy, Temple University, Philadelphia, PA 19140, USA
| | - Thomas S Griffith
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Urology, Universityof Minnesota, Minneapolis, MN 55455, USA
- Center for Immunology, Universityof Minnesota, Minneapolis, MN 55455, USA
- Microbiology, Immunology, & Cancer Biology Graduate Program, University of Minnesota,Minneapolis, MN 55455, USA
| |
Collapse
|
39
|
Saab S, Zalzale H, Rahal Z, Khalifeh Y, Sinjab A, Kadara H. Insights Into Lung Cancer Immune-Based Biology, Prevention, and Treatment. Front Immunol 2020; 11:159. [PMID: 32117295 PMCID: PMC7026250 DOI: 10.3389/fimmu.2020.00159] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 01/21/2020] [Indexed: 12/12/2022] Open
Abstract
Lung cancer is the number one cause of cancer-related deaths. The malignancy is characterized by dismal prognosis and poor clinical outcome mostly due to advanced-stage at diagnosis, thereby inflicting a heavy burden on public health worldwide. Recent breakthroughs in immunotherapy have greatly benefited a subset of lung cancer patients, and more importantly, they are undauntedly bringing forth a paradigm shift in the drugs approved for cancer treatment, by introducing "tumor-type agnostic therapies". Yet, and to fulfill immunotherapy's potential of personalized cancer treatment, demarcating the immune and genomic landscape of cancers at their earliest possible stages will be crucial to identify ideal targets for early treatment and to predict how a particular patient will fare with immunotherapy. Recent genomic surveys of premalignant lung cancer have shed light on early alterations in the evolution of lung cancer. More recently, the advent of immunogenomic technologies has provided prodigious opportunities to study the multidimensional landscape of lung tumors as well as their microenvironment at the molecular, genomic, and cellular resolution. In this review, we will summarize the current state of immune-based therapies for cancer, with a focus on lung malignancy, and highlight learning outcomes from clinical and preclinical studies investigating the naïve immune biology of lung cancer. The review also collates immunogenomic-based evidence from seminal reports which collectively warrant future investigations of premalignancy, the tumor-adjacent normal-appearing lung tissue, pulmonary inflammatory conditions such as chronic obstructive pulmonary disease, as well as systemic microbiome imbalance. Such future directions enable novel insights into the evolution of lung cancers and, thus, can provide a low-hanging fruit of targets for early immune-based treatment of this fatal malignancy.
Collapse
Affiliation(s)
- Sara Saab
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Hussein Zalzale
- School of Medicine, American University of Beirut, Beirut, Lebanon
| | - Zahraa Rahal
- School of Medicine, American University of Beirut, Beirut, Lebanon
| | - Yara Khalifeh
- School of Medicine, American University of Beirut, Beirut, Lebanon
| | - Ansam Sinjab
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Humam Kadara
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
40
|
Jones S, King PJ, Antonescu CN, Sugiyama MG, Bhamra A, Surinova S, Angelopoulos N, Kragh M, Pedersen MW, Hartley JA, Futter CE, Hochhauser D. Targeting of EGFR by a combination of antibodies mediates unconventional EGFR trafficking and degradation. Sci Rep 2020; 10:663. [PMID: 31959764 PMCID: PMC6970994 DOI: 10.1038/s41598-019-57153-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 12/02/2019] [Indexed: 01/17/2023] Open
Abstract
Antibody combinations targeting cell surface receptors are a new modality of cancer therapy. The trafficking and signalling mechanisms regulated by such therapeutics are not fully understood but could underlie differential tumour responses. We explored EGFR trafficking upon treatment with the antibody combination Sym004 which has shown promise clinically. Sym004 promoted EGFR endocytosis distinctly from EGF: it was asynchronous, not accompanied by canonical signalling events and involved EGFR clustering within detergent-insoluble plasma mebrane-associated tubules. Sym004 induced lysosomal degradation independently of EGFR ubiquitylation but dependent upon Hrs/Tsg101 that are required for the formation of intraluminal vesicles (ILVs) within late endosomes. We propose Sym004 cross-links EGFR physically triggering EGFR endocytosis and incorporation onto ILVs and so Sym004 sensitivity correlates with EGFR numbers available for binding, rather than specific signalling events. Consistently Sym004 efficacy and potentiation of cisplatin responses correlated with EGFR surface expression in head and neck cancer cells. These findings will have implications in understanding the mode of action of this new class of cancer therapeutics.
Collapse
Affiliation(s)
- Sylwia Jones
- Cancer Research UK Drug-DNA Interactions Research Group, UCL Cancer Institute, Paul O'Gorman Building, University College London, London, WC1E 6DD, UK
| | - Peter J King
- Cancer Research UK Drug-DNA Interactions Research Group, UCL Cancer Institute, Paul O'Gorman Building, University College London, London, WC1E 6DD, UK
| | | | | | - Amandeep Bhamra
- Proteomics Research Core Facility, UCL Cancer Institute, University College London, London, UK
| | - Silvia Surinova
- Proteomics Research Core Facility, UCL Cancer Institute, University College London, London, UK
| | - Nicos Angelopoulos
- Proteomics Research Core Facility, UCL Cancer Institute, University College London, London, UK
| | | | | | - John A Hartley
- Cancer Research UK Drug-DNA Interactions Research Group, UCL Cancer Institute, Paul O'Gorman Building, University College London, London, WC1E 6DD, UK
| | - Clare E Futter
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK
| | - Daniel Hochhauser
- Cancer Research UK Drug-DNA Interactions Research Group, UCL Cancer Institute, Paul O'Gorman Building, University College London, London, WC1E 6DD, UK.
| |
Collapse
|
41
|
Simmet V, Eberst L, Marabelle A, Cassier PA. Immune checkpoint inhibitor-based combinations: is dose escalation mandatory for phase I trials? Ann Oncol 2019; 30:1751-1759. [PMID: 31435659 DOI: 10.1093/annonc/mdz286] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Numerous phase I trials testing immune checkpoint inhibitors (CPI)-based combinations are currently being conducted to improve response rates observed with single agents. However, methodology varies across studies, especially regarding the use of dose escalation. MATERIALS AND METHODS A literature search was conducted in Pubmed and major oncology meetings libraries for phase I trials reported between 2011 and 2018, containing at least one CPI [CLTA-4 blocking antibody or a PD(L)1 blocking antibody] plus at least one second agent (e.g. tyrosine kinase inhibitor, chemotherapy). Dose escalation schemes, target doses and recommended phase II doses (RP2D) were captured in our database for each study. Combination RP2D (combo-RP2D) was compared with target dose. RESULTS We identified 113 different studies comprising a total of 120 individual cohorts. The backbone was an anti- cytotoxic T-lymphocyte antigen 4 (CTLA-4) in 40 cohorts and an anti-PD(L)1 in 80 cohorts. Dose escalation was used for the CPI in 29 (24%) cohorts [11% for anti-PD(L)1 and 50% for anti-CTLA-4] and for the second agent in 55 cohorts (46%). For 31 s agents (26%), the combo-RP2D was significantly lower than the expected target dose. Failure to reach the target dose was explained by the type of second agent form (e.g. small molecules versus monoclonal antibodies) (P < 0.001) and the choice of trial design for the second agent by investigators. CONCLUSION Design of studies investigating new CPI-based combinations must consider the type of second agent. Dose escalation is required for combinations with small molecules but is unnecessary with vaccine/virus/dendritic therapies and monoclonal antibodies.
Collapse
Affiliation(s)
- V Simmet
- Medical Oncology Department, Léon Bérard Center, Lyon; Medical Oncology Department, Institut de Cancérologie de l'Ouest (ICO), Angers; Medical School, University of Angers, Angers.
| | - L Eberst
- Medical Oncology Department, Léon Bérard Center, Lyon; Medical School, Claude Bernard Lyon 1 University, Lyon
| | - A Marabelle
- Drug Development Department (DITEP), Paris-Saclay University; INSERM U1015, Gustave Roussy, Villejuif, France
| | - P A Cassier
- Medical Oncology Department, Léon Bérard Center, Lyon
| |
Collapse
|
42
|
Passariello M, D'Alise AM, Esposito A, Vetrei C, Froechlich G, Scarselli E, Nicosia A, De Lorenzo C. Novel Human Anti-PD-L1 mAbs Inhibit Immune-Independent Tumor Cell Growth and PD-L1 Associated Intracellular Signalling. Sci Rep 2019; 9:13125. [PMID: 31511565 PMCID: PMC6739323 DOI: 10.1038/s41598-019-49485-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 08/23/2019] [Indexed: 02/08/2023] Open
Abstract
The novel antibody-based immunotherapy in oncology exploits the activation of immune system mediated by immunomodulatory antibodies specific for immune checkpoints. Among them, the programmed death ligand-1 (PD-L1) is of particular interest as it is expressed not only on T-cells, but also on other immune cells and on a large variety of cancer cells, such as breast cancer cells, considering its high expression in both ErbB2-positive and Triple Negative Breast Cancers. We demonstrate here that PD-L1_1, a novel anti-PD-L1 T -cell stimulating antibody, inhibits PD-L1-tumor cell growth also by affecting the intracellular MAPK pathway and by activating caspase 3. Similar in vitro results were obtained for the first time here also with the clinically validated anti-PD-L1 mAb Atezolizumab and in vivo with another validated anti-mouse anti-PD-L1 mAb. Moreover, we found that two high affinity variants of PD-L1_1 inhibited tumor cell viability more efficiently than the parental PD-L1_1 by affecting the same MAPK pathways with a more potent effect. Altogether, these results shed light on the role of PD-L1 in cancer cells and suggest that PD-L1_1 and its high affinity variants could become powerful antitumor weapons to be used alone or in combination with other drugs such as the anti-ErbB2 cAb already successfully tested in in vitro combinatorial treatments.
Collapse
Affiliation(s)
- Margherita Passariello
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Via Pansini 5, 80131, Napoli, Italy.,Ceinge - Biotecnologie Avanzate s.c. a.r.l., via Gaetano Salvatore 486, 80145, Naples, Italy
| | | | - Annachiara Esposito
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Via Pansini 5, 80131, Napoli, Italy.,Ceinge - Biotecnologie Avanzate s.c. a.r.l., via Gaetano Salvatore 486, 80145, Naples, Italy
| | - Cinzia Vetrei
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Via Pansini 5, 80131, Napoli, Italy.,Ceinge - Biotecnologie Avanzate s.c. a.r.l., via Gaetano Salvatore 486, 80145, Naples, Italy
| | - Guendalina Froechlich
- Ceinge - Biotecnologie Avanzate s.c. a.r.l., via Gaetano Salvatore 486, 80145, Naples, Italy.,European School of Molecular Medicine, University of Milan, Via Festa del Perdono 7, 20122, Milan, Italy
| | | | - Alfredo Nicosia
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Via Pansini 5, 80131, Napoli, Italy.,Ceinge - Biotecnologie Avanzate s.c. a.r.l., via Gaetano Salvatore 486, 80145, Naples, Italy.,Keires AG Bäumleingasse 18, CH-4051, Basel, Switzerland
| | - Claudia De Lorenzo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Via Pansini 5, 80131, Napoli, Italy. .,Ceinge - Biotecnologie Avanzate s.c. a.r.l., via Gaetano Salvatore 486, 80145, Naples, Italy.
| |
Collapse
|
43
|
Zhu G, Foletti D, Liu X, Ding S, Melton Witt J, Hasa-Moreno A, Rickert M, Holz C, Aschenbrenner L, Yang AH, Kraynov E, Evering W, Obert L, Lee C, Sai T, Mistry T, Lindquist KC, Van Blarcom T, Strop P, Chaparro-Riggers J, Liu SH. Targeting CLDN18.2 by CD3 Bispecific and ADC Modalities for the Treatments of Gastric and Pancreatic Cancer. Sci Rep 2019; 9:8420. [PMID: 31182754 PMCID: PMC6557842 DOI: 10.1038/s41598-019-44874-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 05/27/2019] [Indexed: 02/07/2023] Open
Abstract
Human CLDN18.2 is highly expressed in a significant proportion of gastric and pancreatic adenocarcinomas, while normal tissue expression is limited to the epithelium of the stomach. The restricted expression makes it a potential drug target for the treatment of gastric and pancreatic adenocarcinoma, as evidenced by efforts to target CLDN18.2 via naked antibody and CAR-T modalities. Herein we describe CLDN18.2-targeting via a CD3-bispecific and an antibody drug conjugate and the characterization of these potential therapeutic molecules in efficacy and preliminary toxicity studies. Anti-hCLDN18.2 ADC, CD3-bispecific and diabody, targeting a protein sequence conserved in rat, mouse and monkey, exhibited in vitro cytotoxicity in BxPC3/hCLDN18.2 (IC50 = 1.52, 2.03, and 0.86 nM) and KATO-III/hCLDN18.2 (IC50 = 1.60, 0.71, and 0.07 nM) respectively and inhibited tumor growth of pancreatic and gastric patient-derived xenograft tumors. In a rat exploratory toxicity study, the ADC was tolerated up to 10 mg/kg. In a preliminary assessment of tolerability, the anti-CLDN18.2 diabody (0.34 mg/kg) did not produce obvious signs of toxicity in the stomach of NSG mice 4 weeks after dosing. Taken together, our data indicate that targeting CLDN18.2 with an ADC or bispecific modality could be a valid therapeutic approach for the treatment of gastric and pancreatic cancer.
Collapse
Affiliation(s)
- Guoyun Zhu
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA.
| | - Davide Foletti
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA.,23 and Me, 349 Oyster Point Blvd, South San Francisco, CA, 94080, USA
| | - Xiaohui Liu
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA
| | - Sheng Ding
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA.,Gilead Sciences, 333 Lakeside Drive, Foster City, CA, 94404, USA
| | - Jody Melton Witt
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA.,Grifols Diagnostic Solutions, 6455 Christie Ave B-334C, Emeryville, CA, 94608, USA
| | - Adela Hasa-Moreno
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA.,Kodiak Sciences Inc., 2631 Hanover St, Palo Alto, CA, 94304, USA
| | - Mathias Rickert
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA.,Applied Molecular Transport, 1 Tower Place, Suite 850, South San Francisco, CA, 94080, USA
| | - Charles Holz
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA.,Grifols Diagnostic Solutions, 6455 Christie Ave B-334C, Emeryville, CA, 94608, USA
| | - Laura Aschenbrenner
- Drug Safety Research and Development, Pfizer Worldwide Research and Development, 10646 Science Center Dr., San Diego, CA, 92121, USA.,Covance Inc. Early Phase Development Solutions, 3301 Kinsman Blvd, Madison, WI, 53704, USA
| | - Amy H Yang
- Drug Safety Research and Development, Pfizer Worldwide Research and Development, 10646 Science Center Dr., San Diego, CA, 92121, USA
| | - Eugenia Kraynov
- BioMedicine Design, Pfizer Worldwide Research and Development, 10646 Science Center Dr., San Diego, CA, 92121, USA
| | - Winston Evering
- Drug Safety Research and Development, Pfizer Worldwide Research and Development, 10646 Science Center Dr., San Diego, CA, 92121, USA
| | - Leslie Obert
- Drug Safety Research and Development, Pfizer Worldwide Research and Development, 280 Shennecossett Rd, Groton, CT, 06340, USA.,GSK, 1250 South Collegeville Road, Collegeville, PA, 19426, USA
| | - Chenyu Lee
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA.,Alector, 151 Oyster Point Blvd #300, South San Francisco, CA, 94080, USA
| | - Tao Sai
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA
| | - Tina Mistry
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA
| | - Kevin C Lindquist
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA
| | - Thomas Van Blarcom
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA.,Allogene Therapeutics, 210 E. Grand Avenue, South San Francisco, CA, 94080, USA
| | - Pavel Strop
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA.,Bristol-Myers Squibb, 700 Bay Rd suite A, Redwood City, CA, 94063, USA
| | - Javier Chaparro-Riggers
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA
| | - Shu-Hui Liu
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA, 94080, USA.,Multitude Therapeutics, Abmart, 3698 Haven Avenue Suite A, Redwood City, CA, 94063, USA
| |
Collapse
|
44
|
Zhou C, Zhang J. Immunotherapy-based combination strategies for treatment of gastrointestinal cancers: current status and future prospects. Front Med 2019; 13:12-23. [PMID: 30796606 DOI: 10.1007/s11684-019-0685-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Accepted: 12/27/2018] [Indexed: 12/29/2022]
Abstract
Strategies in comprehensive therapy for gastrointestinal (GI) cancer have been optimized in the last decades to improve patients' outcomes. However, treatment options remain limited for late-stage or refractory diseases. The efficacy of immune checkpoint inhibitors (ICIs) for treatment of refractory GI cancer has been confirmed by randomized clinical trials. In 2017, pembrolizumab was approved by the US Food and Drug Administration as the first agent for treatment of metastatic solid tumors with mismatch repair deficiency, especially for colorectal cancer. Given the different mechanisms, oncologists have focused on determining whether ICIs-based combination strategies could achieve higher efficacy than conventional therapy alone in late-stage or even front-line treatment of GI cancer. This review discusses the current status of combining immune checkpoint inhibitors with molecular targeted therapy, chemotherapy, or radiotherapy in GI cancer in terms of mechanisms, safety, and efficacy to provide basis for future research.
Collapse
Affiliation(s)
- Chenfei Zhou
- Department of Oncology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jun Zhang
- Department of Oncology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
45
|
Insel PA, Sriram K, Wiley SZ, Wilderman A, Katakia T, McCann T, Yokouchi H, Zhang L, Corriden R, Liu D, Feigin ME, French RP, Lowy AM, Murray F. GPCRomics: GPCR Expression in Cancer Cells and Tumors Identifies New, Potential Biomarkers and Therapeutic Targets. Front Pharmacol 2018; 9:431. [PMID: 29872392 PMCID: PMC5972277 DOI: 10.3389/fphar.2018.00431] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 04/12/2018] [Indexed: 12/16/2022] Open
Abstract
G protein-coupled receptors (GPCRs), the largest family of targets for approved drugs, are rarely targeted for cancer treatment, except for certain endocrine and hormone-responsive tumors. Limited knowledge regarding GPCR expression in cancer cells likely has contributed to this lack of use of GPCR-targeted drugs as cancer therapeutics. We thus undertook GPCRomic studies to define the expression of endoGPCRs (which respond to endogenous molecules such as hormones, neurotransmitters and metabolites) in multiple types of cancer cells. Using TaqMan qPCR arrays to quantify the mRNA expression of ∼340 such GPCRs, we found that human chronic lymphocytic leukemia (CLL) cells/stromal cells associated with CLL, breast cancer cell lines, colon cancer cell lines, pancreatic ductal adenocarcinoma (PDAC) cells, cancer associated fibroblasts (CAFs), and PDAC tumors express 50 to >100 GPCRs, including many orphan GPCRs (which lack known physiologic agonists). Limited prior data exist regarding the expression or function of most of the highly expressed GPCRs in these cancer cells and tumors. Independent results from public cancer gene expression databases confirm the expression of such GPCRs. We propose that highly expressed GPCRs in cancer cells (for example, GPRC5A in PDAC and colon cancer cells and GPR68 in PDAC CAFs) may contribute to the malignant phenotype, serve as biomarkers and/or may be novel therapeutic targets for the treatment of cancer.
Collapse
Affiliation(s)
- Paul A. Insel
- Department of Pharmacology, University of California, San Diego, San Diego, CA, United States
- Department of Medicine, University of California, San Diego, San Diego, CA, United States
| | - Krishna Sriram
- Department of Pharmacology, University of California, San Diego, San Diego, CA, United States
| | - Shu Z. Wiley
- Department of Pharmacology, University of California, San Diego, San Diego, CA, United States
| | - Andrea Wilderman
- Department of Pharmacology, University of California, San Diego, San Diego, CA, United States
| | - Trishna Katakia
- Department of Pharmacology, University of California, San Diego, San Diego, CA, United States
| | - Thalia McCann
- Department of Pharmacology, University of California, San Diego, San Diego, CA, United States
| | - Hiroshi Yokouchi
- Department of Pharmacology, University of California, San Diego, San Diego, CA, United States
| | - Lingzhi Zhang
- Department of Pharmacology, University of California, San Diego, San Diego, CA, United States
| | - Ross Corriden
- Department of Pharmacology, University of California, San Diego, San Diego, CA, United States
| | - Dongling Liu
- Department of Pharmacology, University of California, San Diego, San Diego, CA, United States
| | - Michael E. Feigin
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Randall P. French
- Department of Surgery, University of California, San Diego, San Diego, CA, United States
- Moores Cancer Center, University of California, San Diego, San Diego, CA, United States
| | - Andrew M. Lowy
- Department of Surgery, University of California, San Diego, San Diego, CA, United States
- Moores Cancer Center, University of California, San Diego, San Diego, CA, United States
| | - Fiona Murray
- Department of Pharmacology, University of California, San Diego, San Diego, CA, United States
- Department of Medicine, University of California, San Diego, San Diego, CA, United States
| |
Collapse
|
46
|
Plant-Derived Anticancer Agents: Lessons from the Pharmacology of Geniposide and Its Aglycone, Genipin. Biomedicines 2018; 6:biomedicines6020039. [PMID: 29587429 PMCID: PMC6027249 DOI: 10.3390/biomedicines6020039] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 03/21/2018] [Accepted: 03/22/2018] [Indexed: 12/24/2022] Open
Abstract
For centuries, plants have been exploited by mankind as sources of numerous cancer chemotherapeutic agents. Good examples of anticancer compounds of clinical significance today include the taxanes (e.g., taxol), vincristine, vinblastine, and the podophyllotoxin analogues that all trace their origin to higher plants. While all these drugs, along with the various other available therapeutic options, brought some relief in cancer management, a real breakthrough or cure has not yet been achieved. This critical review is a reflection on the lessons learnt from decades of research on the iridoid glycoside geniposide and its aglycone, genipin, which are currently used as gold standard reference compounds in cancer studies. Their effects on tumour development (carcinogenesis), cancer cell survival, and death, with particular emphasis on their mechanisms of actions, are discussed. Particular attention is also given to mechanisms related to the dual pro-oxidant and antioxidant effects of these compounds, the mitochondrial mechanism of cancer cell killing through reactive oxygen species (ROS), including that generated through the uncoupling protein-2 (UCP-2), the inflammatory mechanism, and cell cycle regulation. The implications of various studies for the evaluation of glycosidic and aglycone forms of natural products in vitro and in vivo through pharmacokinetic scrutiny are also addressed.
Collapse
|
47
|
Santamaria S, Delgado M, Kremer L, Garcia-Sanz JA. Will a mAb-Based Immunotherapy Directed against Cancer Stem Cells Be Feasible? Front Immunol 2017; 8:1509. [PMID: 29170667 PMCID: PMC5684111 DOI: 10.3389/fimmu.2017.01509] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 10/25/2017] [Indexed: 12/12/2022] Open
Abstract
The cancer stem cell (CSC) hypothesis suggests that within a tumor, there is a small subpopulation of cells with stem cell properties responsible for tumor maintenance and metastasis generation. This hypothesis also implies that new antitumor drugs, rather than targeting the bulk of the tumor mass, would be more effective if they directly targeted the CSC subpopulation. The CSCs from several types of tumors have been identified with mAbs recognizing surface antigens in these cells; however, antigens specifically or exclusively expressed in the CSC population have not yet been identified. Thus, questioning the possibility of using therapeutic antibodies directed against the CSCs. Here, we review the possibilities of using antibodies directly targeting the CSCs as therapeutic agents in the form of naked antibodies, antibodies conjugated to nanoparticles, or antibody cocktails.
Collapse
Affiliation(s)
- Silvia Santamaria
- Cancer Genetics and Cancer Stem Cell Laboratory, Centro de Investigaciones Biologicas, Department of Cellular and Molecular Medicine, Spanish National Research Council (CSIC), Madrid, Spain
| | - Marisa Delgado
- Cancer Genetics and Cancer Stem Cell Laboratory, Centro de Investigaciones Biologicas, Department of Cellular and Molecular Medicine, Spanish National Research Council (CSIC), Madrid, Spain
| | - Leonor Kremer
- Centro Nacional de Biotecnologia, Department of Immunology and Oncology, Spanish National Research Council (CSIC), Madrid, Spain
| | - Jose A Garcia-Sanz
- Cancer Genetics and Cancer Stem Cell Laboratory, Centro de Investigaciones Biologicas, Department of Cellular and Molecular Medicine, Spanish National Research Council (CSIC), Madrid, Spain
| |
Collapse
|