1
|
Walton WJ, Zhang SJ, Wilson JJ, Harvey BN, Clemens M, Gu Y. Impact of Monoclonal Antibody Aggregates on Effector Function Characterization. Antibodies (Basel) 2025; 14:31. [PMID: 40265412 PMCID: PMC12015860 DOI: 10.3390/antib14020031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 02/25/2025] [Accepted: 03/24/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND/OBJECTIVES Monoclonal antibodies have successfully been used for a variety of indications. Many therapeutic antibodies are IgG1 and elicit effector functions as part of their mechanism of action. It is well known that aggregate levels should be controlled for therapeutic antibodies. Although there are several reports describing the impact of antibody aggregates on FcγR binding, most of these have been performed with surface plasmon resonance in an avidity-based format. What is less well known is which Fcγ receptor is most impacted by antibody aggregation and how antibody aggregates impact binding to Fcγ receptors in solution-based formats and in cell-based assays. METHODS An effector-competent IgG1 (mAb1) was forcibly degraded and fractionated by size exclusion chromatography to enrich for aggregates. The fractions were examined for FcγR binding by SPR with different formats and in solution. The fractions were also analyzed with cell-based FcγR reporter assays. RESULTS All Fcγ receptors displayed increased binding to enriched mAb1 aggregates in the avidity-based SPR methods and in solution, with FcγRIIa impacted the most. When examined with an antibody-down SPR format that is not usually susceptible to avidity, FcγRIIa did not show increased binding with mAb1 aggregation. Although activity for mAb1 aggregates increased slightly in an FcγRIIa cell-based reporter assay, it decreased in the FcγRIIIa reporter assay (most likely due to differences in fucosylation from the reference standard). CONCLUSIONS Monoclonal antibody aggregation can impact FcγR binding for avidity-based binding formats. Even at low levels of antibody aggregation, FcγRII binding increases substantially.
Collapse
Affiliation(s)
- Wendy J. Walton
- Bioproduct Research & Development, Lilly Research Laboratories, Lilly Technology Center North, Indianapolis, IN 46221, USA
| | - Shousong Jason Zhang
- Bioproduct Research & Development, Lilly Research Laboratories, Lilly Technology Center North, Indianapolis, IN 46221, USA
| | - Joseph J. Wilson
- Bioproduct Research & Development, Lilly Research Laboratories, Lilly Technology Center North, Indianapolis, IN 46221, USA
| | - Briana N. Harvey
- Analytical QA, Product Research & Development, Lilly Technology Center North, Indianapolis, IN 46221, USA
| | - Matthew Clemens
- Bioproduct Research & Development, Lilly Research Laboratories, Lilly Technology Center North, Indianapolis, IN 46221, USA
| | - Yingmei Gu
- Bioproduct Research & Development, Lilly Research Laboratories, Lilly Technology Center North, Indianapolis, IN 46221, USA
| |
Collapse
|
2
|
Nakamura N, Arima N, Takakuwa T, Yoshioka S, Imada K, Fukushima K, Hotta M, Fuchida SI, Kanda J, Uoshima N, Shimura Y, Tanaka H, Ohta K, Kosugi S, Yagi H, Yoshihara S, Yamamura R, Adachi Y, Hanamoto H, Shibayama H, Hosen N, Ito T, Shimazaki C, Takaori-Kondo A, Kuroda J, Matsumura I, Hino M. Efficacy of elotuzumab for multiple myeloma deteriorates after daratumumab: a multicenter retrospective study. Ann Hematol 2024; 103:5681-5690. [PMID: 38492020 DOI: 10.1007/s00277-024-05705-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 03/10/2024] [Indexed: 03/18/2024]
Abstract
Elotuzumab-based regimens are sometimes selected for multiple myeloma treatment after daratumumab-based regimens. However, there has been insufficient discussion on the efficacy of elotuzumab after daratumumab. We used Kansai Myeloma Forum registration data in a multicenter retrospective evaluation of the efficacy of elotuzumab after daratumumab. Overall survival (OS) rate and time to next treatment (TTNT) were significantly worse in the cohort given elotuzumab after daratumumab (Dara cohort, n = 47) than in the cohort with no history of daratumumab administration before elotuzumab (No-Dara cohort, n = 80, OS: P = 0.03; TTNT: P = 0.02; best response: P < 0.01). In the Dara cohort, OS and TTNT rates were worse with sequential elotuzumab use after daratumumab than with non-sequential (OS: P = 0.02; TTNT: P = 0.03). In patients given elotuzumab < 180 days after daratumumab, OS (P = 0.08) and best response (P = 0.21) tended to be worse, and TTNT was significantly worse (P = 0.01), than in those given elotuzumab after ≥ 180 days. These findings were confirmed by subgroup analyses and multivariate analyses. Monoclonal-antibody-free treatment might be preferable after daratumumab-based regimens. If possible, elotuzumab-based regimens should be considered only ≥ 180 days after daratumumab use.
Collapse
Affiliation(s)
- Naokazu Nakamura
- Department of Hematology, Shinko Hospital, 1-4-47, Wakihamacho, Chuo-Ku, Kobe, Hyogo, 651-0072, Japan.
- Department of Hematology and Oncology Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | - Nobuyoshi Arima
- Department of Hematology, Shinko Hospital, 1-4-47, Wakihamacho, Chuo-Ku, Kobe, Hyogo, 651-0072, Japan
| | - Teruhito Takakuwa
- Department of Hematology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Satoshi Yoshioka
- Department of Hematology, Japanese Red Cross Osaka Hospital, Osaka, Japan
| | - Kazunori Imada
- Department of Hematology, Japanese Red Cross Osaka Hospital, Osaka, Japan
| | - Kentaro Fukushima
- Department of Hematology and Oncology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Masaaki Hotta
- First Department of Internal Medicine, Kansai Medical University, Hirakata, Japan
| | - Shin-Ichi Fuchida
- Department of Hematology, Japan Community Health Care Organization Kyoto Kuramaguchi Medical Center, Kyoto, Japan
| | - Junya Kanda
- Department of Hematology and Oncology Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Nobuhiko Uoshima
- Department of Hematology, Japanese Red Cross Kyoto Daini Hospital, Kyoto, Japan
| | - Yuji Shimura
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hirokazu Tanaka
- Department of Hematology and Rheumatology, Faculty of Medicine, Kindai University, Osakasayama, Japan
| | | | - Satoru Kosugi
- Department of Internal Medicine (Hematology), Toyonaka Municipal Hospital, Toyonaka, Japan
| | - Hideo Yagi
- Department of Hematology and Oncology, Nara Prefecture General Medical Center, Nara, Japan
| | - Satoshi Yoshihara
- Division of Hematology, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Japan
| | - Ryosuke Yamamura
- Department of Hematology, Osaka Saiseikai Nakatsu Hospital, Nakatsu, Japan
| | - Yoko Adachi
- Department of Internal Medicine, JCHO Kobe Central Hospital, Kobe, Japan
| | - Hitoshi Hanamoto
- Department of Hematology, Kindai University Nara Hospital, Nara, Japan
| | - Hirohiko Shibayama
- Department of Hematology, National Hospital Organization Osaka National Hospital, Osaka, Japan
| | - Naoki Hosen
- Department of Hematology and Oncology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Tomoki Ito
- First Department of Internal Medicine, Kansai Medical University, Hirakata, Japan
| | - Chihiro Shimazaki
- Department of Hematology, Japan Community Health Care Organization Kyoto Kuramaguchi Medical Center, Kyoto, Japan
| | - Akifumi Takaori-Kondo
- Department of Hematology and Oncology Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Junya Kuroda
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Itaru Matsumura
- Department of Hematology and Rheumatology, Faculty of Medicine, Kindai University, Osakasayama, Japan
| | - Masayuki Hino
- Department of Hematology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
3
|
Strati P, Champion R, Coleman M, Smith SM, Venugopal P, Martin P, Wood A, Miller K, Christian B. Acalabrutinib alone or in combination with rituximab for follicular lymphoma: An open-label study. Br J Haematol 2024; 205:2248-2253. [PMID: 39308028 PMCID: PMC11637741 DOI: 10.1111/bjh.19787] [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: 05/23/2024] [Accepted: 09/13/2024] [Indexed: 12/14/2024]
Abstract
Acalabrutinib is a selective, second-generation Bruton tyrosine kinase inhibitor. In this open-label, parallel-group study, patients with relapsed/refractory (R/R) follicular lymphoma (FL) were randomised to either acalabrutinib monotherapy or acalabrutinib plus rituximab. An additional cohort of patients with treatment-naive (TN) FL received only the acalabrutinib-rituximab combination. Acalabrutinib-rituximab was well tolerated and active in R/R and TN FL; in the TN cohort the overall response rate was 92.3% with most remissions lasting over 4 years. Acalabrutinib monotherapy was also well tolerated and active in R/R FL. These results support further study of acalabrutinib alone and in combination with rituximab in FL.
Collapse
Affiliation(s)
- Paolo Strati
- The University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | | | - Morton Coleman
- Clinical Research Alliance/Weill Cornell MedicineNew YorkNew YorkUSA
| | | | | | - Peter Martin
- Weill Cornell Medical CollegeNew York Presbyterian HospitalNew YorkNew YorkUSA
| | | | | | - Beth Christian
- The Ohio State University Comprehensive Cancer CenterColumbusOhioUSA
| |
Collapse
|
4
|
van der Horst HJ, Mutis T. Enhancing Fc-mediated effector functions of monoclonal antibodies: The example of HexaBodies. Immunol Rev 2024; 328:456-465. [PMID: 39275983 PMCID: PMC11659923 DOI: 10.1111/imr.13394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/16/2024]
Abstract
Since the approval of the CD20-targeting monoclonal antibody (mAb) rituximab for the treatment of lymphoma in 1997, mAb therapy has significantly transformed cancer treatment. With over 90 FDA-approved mAbs for the treatment of various hematological and solid cancers, modern cancer treatment relies heavily on these therapies. The overwhelming success of mAbs as cancer therapeutics is attributed to their broad applicability, high safety profile, and precise targeting of cancer-associated surface antigens. Furthermore, mAbs can induce various anti-tumor cytotoxic effector mechanisms including antibody-dependent cellular cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP), and complement-dependent cytotoxicity (CDC), all of which are mediated via their fragment crystallizable (Fc) domain. Over the past decades, these effector mechanisms have been substantially improved through Fc domain engineering. In this review, we will outline the different approaches to enhance Fc effector functions via Fc engineering of mAbs, with a specific emphasis on the so-called "HexaBody" technology, which is designed to enhance the hexamerization of mAbs on the target cell surface, thereby inducing greater complement activation, CDC, and receptor clustering. The review will summarize the development, preclinical, and clinical testing of several HexaBodies designed for the treatment of B-cell malignancies, as well as the potential use of the HexaBody technology beyond Fc-mediated effector functions.
Collapse
Affiliation(s)
- Hilma J. van der Horst
- Department of HematologyCancer Center Amsterdam, Amsterdam UMC, VU Medical CenterAmsterdamThe Netherlands
- Present address:
Department of Fundamental Oncology, Ludwig Institute for Cancer ResearchUniversity of LausanneEpalingesSwitzerland
| | - Tuna Mutis
- Department of HematologyCancer Center Amsterdam, Amsterdam UMC, VU Medical CenterAmsterdamThe Netherlands
| |
Collapse
|
5
|
Krištić J, Lauc G. The importance of IgG glycosylation-What did we learn after analyzing over 100,000 individuals. Immunol Rev 2024; 328:143-170. [PMID: 39364834 PMCID: PMC11659926 DOI: 10.1111/imr.13407] [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] [Indexed: 10/05/2024]
Abstract
All four subclasses of immunoglobulin G (IgG) antibodies have glycan structures attached to the protein part of the IgG molecules. Glycans linked to the Fc portion of IgG are found in all IgG antibodies, while about one-fifth of IgG antibodies in plasma also have glycans attached to the Fab portion of IgG. The IgG3 subclass is characterized by more complex glycosylation compared to other IgG subclasses. In this review, we discuss the significant influence that glycans exert on the structural and functional properties of IgG. We provide a comprehensive overview of how the composition of these glycans can affect IgG's effector functions by modulating its interactions with Fcγ receptors and other molecules such as the C1q component of complement, which in turn influence various immune responses triggered by IgG, including antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC). In addition, the importance of glycans for the efficacy of therapeutics like monoclonal antibodies and intravenous immunoglobulin (IVIg) therapy is discussed. Moreover, we offer insights into IgG glycosylation characteristics and roles derived from general population, disease-specific, and interventional studies. These studies indicate that IgG glycans are important biomarkers and functional effectors in health and disease.
Collapse
Affiliation(s)
| | - Gordan Lauc
- Genos Glycoscience Research LaboratoryZagrebCroatia
- Faculty of Pharmacy and BiochemistryUniversity of ZagrebZagrebCroatia
| |
Collapse
|
6
|
Gowkielewicz M, Lipka A, Zdanowski W, Waśniewski T, Majewska M, Carlberg C. Anti-Müllerian hormone: biology and role in endocrinology and cancers. Front Endocrinol (Lausanne) 2024; 15:1468364. [PMID: 39351532 PMCID: PMC11439669 DOI: 10.3389/fendo.2024.1468364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 08/29/2024] [Indexed: 10/04/2024] Open
Abstract
Anti-Müllerian hormone (AMH) is a peptide belonging to the transforming growth factor beta superfamily and acts exclusively through its receptor type 2 (AMHR2). From the 8th week of pregnancy, AMH is produced by Sertoli cells, and from the 23rd week of gestation, it is produced by granulosa cells of the ovary. AMH plays a critical role in regulating gonadotropin secretion, ovarian tissue responsiveness to pituitary hormones, and the pathogenesis of polycystic ovarian syndrome. It inhibits the transition from primordial to primary follicles and is considered the best marker of ovarian reserve. Therefore, measuring AMH concentration of the hormone is valuable in managing assisted reproductive technologies. AMH was initially discovered through its role in the degeneration of Müllerian ducts in male fetuses. However, due to its ability to inhibit the cell cycle and induce apoptosis, it has also garnered interest in oncology. For example, antibodies targeting AMHR2 are being investigated for their potential in diagnosing and treating various cancers. Additionally, AMH is present in motor neurons and functions as a protective and growth factor. Consequently, it is involved in learning and memory processes and may support the treatment of Alzheimer's disease. This review aims to provide a comprehensive overview of the biology of AMH and its role in both endocrinology and oncology.
Collapse
Affiliation(s)
- Marek Gowkielewicz
- Department of Gynecology and Obstetrics, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Aleksandra Lipka
- Institute of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Wojciech Zdanowski
- Department of Gynecology and Obstetrics, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Tomasz Waśniewski
- Department of Gynecology and Obstetrics, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Marta Majewska
- Department of Human Physiology and Pathophysiology, School of Medicine, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Carsten Carlberg
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
- School of Medicine, Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
7
|
Gerassy-Vainberg S, Starosvetsky E, Gaujoux R, Blatt A, Maimon N, Gorelik Y, Pressman S, Alpert A, Bar-Yoseph H, Dubovik T, Perets B, Katz A, Milman N, Segev M, Chowers Y, Shen-Orr SS. A personalized network framework reveals predictive axis of anti-TNF response across diseases. Cell Rep Med 2024; 5:101300. [PMID: 38118442 PMCID: PMC10829759 DOI: 10.1016/j.xcrm.2023.101300] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 08/20/2023] [Accepted: 10/31/2023] [Indexed: 12/22/2023]
Abstract
Personalized treatment of complex diseases has been mostly predicated on biomarker identification of one drug-disease combination at a time. Here, we use a computational approach termed Disruption Networks to generate a data type, contextualized by cell-centered individual-level networks, that captures biology otherwise overlooked when performing standard statistics. This data type extends beyond the "feature level space", to the "relations space", by quantifying individual-level breaking or rewiring of cross-feature relations. Applying Disruption Networks to dissect high-dimensional blood data, we discover and validate that the RAC1-PAK1 axis is predictive of anti-TNF response in inflammatory bowel disease. Intermediate monocytes, which correlate with the inflammatory state, play a key role in the RAC1-PAK1 responses, supporting their modulation as a therapeutic target. This axis also predicts response in rheumatoid arthritis, validated in three public cohorts. Our findings support blood-based drug response diagnostics across immune-mediated diseases, implicating common mechanisms of non-response.
Collapse
Affiliation(s)
- Shiran Gerassy-Vainberg
- Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 32000, Israel; Department of Gastroenterology, Rambam Health Care Campus, Haifa 3109601, Israel
| | - Elina Starosvetsky
- Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | - Renaud Gaujoux
- Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 32000, Israel; CytoReason, Tel Aviv 67012, Israel
| | - Alexandra Blatt
- Department of Gastroenterology, Rambam Health Care Campus, Haifa 3109601, Israel
| | - Naama Maimon
- Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 32000, Israel; Department of Gastroenterology, Rambam Health Care Campus, Haifa 3109601, Israel
| | - Yuri Gorelik
- Department of Gastroenterology, Rambam Health Care Campus, Haifa 3109601, Israel
| | - Sigal Pressman
- Department of Gastroenterology, Rambam Health Care Campus, Haifa 3109601, Israel; Clinical Research Institute, Rambam Health Care Campus, Haifa 3109601, Israel
| | - Ayelet Alpert
- Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | - Haggai Bar-Yoseph
- Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 32000, Israel; Department of Gastroenterology, Rambam Health Care Campus, Haifa 3109601, Israel
| | - Tania Dubovik
- Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | - Benny Perets
- Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | | | - Neta Milman
- Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | - Meital Segev
- Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | - Yehuda Chowers
- Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 32000, Israel; Department of Gastroenterology, Rambam Health Care Campus, Haifa 3109601, Israel; Clinical Research Institute, Rambam Health Care Campus, Haifa 3109601, Israel.
| | - Shai S Shen-Orr
- Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 32000, Israel.
| |
Collapse
|
8
|
Bousso P, Grandjean CL. Immunomodulation under the lens of real-time in vivo imaging. Eur J Immunol 2023; 53:e2249921. [PMID: 37051691 DOI: 10.1002/eji.202249921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 11/16/2022] [Accepted: 04/11/2023] [Indexed: 04/14/2023]
Abstract
Modulation of cells and molecules of the immune system not only represents a major opportunity to treat a variety of diseases including infections, cancer, autoimmune, and inflammatory disorders but could also help understand the intricacies of immune responses. A detailed mechanistic understanding of how a specific immune intervention may provide clinical benefit is essential for the rational design of efficient immunomodulators. Visualizing the impact of immunomodulation in real-time and in vivo has emerged as an important approach to achieve this goal. In this review, we aim to illustrate how multiphoton intravital imaging has helped clarify the mode of action of immunomodulatory strategies such as antibodies or cell therapies. We also discuss how optogenetics combined with imaging will further help manipulate and precisely understand immunomodulatory pathways. Combined with other single-cell technologies, in vivo dynamic imaging has therefore a major potential for guiding preclinical development of immunomodulatory drugs.
Collapse
Affiliation(s)
- Philippe Bousso
- Dynamics of Immune Responses Unit, Institut Pasteur, INSERM U1223, Université de Paris Cité, Paris, France
| | - Capucine L Grandjean
- Dynamics of Immune Responses Unit, Institut Pasteur, INSERM U1223, Université de Paris Cité, Paris, France
| |
Collapse
|
9
|
Fagnano E, Pendharkar S, Colton M, Jones PN, Sallan MC, Klymenko T, Braun A, Klein C, Honeychurch J, Cheadle EJ, Illidge TM. Stromal cell inhibition of anti-CD20 antibody mediated killing of B-cell malignancies. Front Cell Dev Biol 2023; 11:1270398. [PMID: 38020903 PMCID: PMC10646167 DOI: 10.3389/fcell.2023.1270398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 10/12/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction: The glycoengineered type II anti-CD20 monoclonal antibody obinutuzumab has been licensed for treatment in follicular non-Hodgkin lymphoma and B-CLL following clinical trials demonstrating superior outcomes to standard of care treatment. However, ultimately many patients still relapse, highlighting the need to understand the mechanisms behind treatment failure to improve patient care. Resistance to chemotherapy is often caused by the ability of malignant B-cells to migrate to the bone marrow and home into the stromal layer. Therefore, this study aimed to investigate whether stromal cells were also able to inhibit type II anti-CD20 antibody mechanisms of action, contributing to resistance to therapy. Methods: A stromal-tumor co-culture was established in vitro between Raji or Daudi B-cell tumor cells and M210B4 stromal cells in 24 well plates. Results: Contact with stromal cells was able to protect tumor cells from obinutuzumab mediated programmed cell death (PCD), antibody dependent cellular phagocytosis and antibody dependent cellular cytotoxicity. Furthermore, such protection required direct contact between stroma and tumor cells. Stromal cells appeared to interfere with obinutuzumab mediated B-cell homotypic adhesion through inhibiting and reversing actin remodelling, potentially as a result of stromal-tumor cell contact leading to downregulation of CD20 on the surface of tumor cells. Further evidence for the potential role of CD20 downregulation comes through the reduction in surface CD20 expression and inhibition of obinutuzumab mediated PCD when tumor cells are treated with Ibrutinib in the presence of stromal cells. The proteomic analysis of tumor cells after contact with stromal cells led to the identification of a number of altered pathways including those involved in cell adhesion and the actin cytoskeleton and remodeling. Discussion: This work demonstrates that contact between tumor cells and stromal cells leads to inhibition of Obinutuzumab effector functions and has important implications for future therapies to improve outcomes to anti-CD20 antibodies. A deeper understanding of how anti-CD20 antibodies interact with stromal cells could prove a useful tool to define better strategies to target the micro-environment and ultimately improve patient outcomes in B-cell malignancies.
Collapse
Affiliation(s)
- Ester Fagnano
- Targeted Therapy Group, Division of Cancer Sciences, The University of Manchester, Manchester, United Kingdom
| | - Swati Pendharkar
- Targeted Therapy Group, Division of Cancer Sciences, The University of Manchester, Manchester, United Kingdom
| | - Madyson Colton
- Targeted Therapy Group, Division of Cancer Sciences, The University of Manchester, Manchester, United Kingdom
| | - Philip N. Jones
- Targeted Therapy Group, Division of Cancer Sciences, The University of Manchester, Manchester, United Kingdom
| | - Marta Crespi Sallan
- Centre for Haemato-Oncology, John Vane Science Centre, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Tetyana Klymenko
- Department of Biosciences and Chemistry, Sheffield Hallam University, Sheffield, United Kingdom
| | - Andrejs Braun
- Centre for Haemato-Oncology, John Vane Science Centre, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Christian Klein
- Roche Innovation Center Zurch, Roche Glycart AG, Schlieren, Switzerland
| | - Jamie Honeychurch
- Targeted Therapy Group, Division of Cancer Sciences, The University of Manchester, Manchester, United Kingdom
| | - Eleanor J. Cheadle
- Targeted Therapy Group, Division of Cancer Sciences, The University of Manchester, Manchester, United Kingdom
| | - Timothy M. Illidge
- Targeted Therapy Group, Division of Cancer Sciences, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
10
|
Fox JM, Roy V, Gunn BM, Bolton GR, Fremont DH, Alter G, Diamond MS, Boesch AW. Enhancing the therapeutic activity of hyperimmune IgG against chikungunya virus using FcγRIIIa affinity chromatography. Front Immunol 2023; 14:1153108. [PMID: 37251375 PMCID: PMC10213286 DOI: 10.3389/fimmu.2023.1153108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 04/24/2023] [Indexed: 05/31/2023] Open
Abstract
Introduction Chikungunya virus (CHIKV) is a re-emerging mosquito transmitted alphavirus of global concern. Neutralizing antibodies and antibody Fc-effector functions have been shown to reduce CHIKV disease and infection in animals. However, the ability to improve the therapeutic activity of CHIKV-specific polyclonal IgG by enhancing Fc-effector functions through modulation of IgG subclass and glycoforms remains unknown. Here, we evaluated the protective efficacy of CHIKV-immune IgG enriched for binding to Fc-gamma receptor IIIa (FcγRIIIa) to select for IgG with enhanced Fc effector functions. Methods Total IgG was isolated from CHIKV-immune convalescent donors with and without additional purification by FcγRIIIa affinity chromatography. The enriched IgG was characterized in biophysical and biological assays and assessed for therapeutic efficacy during CHIKV infection in mice. Results FcγRIIIa-column purification enriched for afucosylated IgG glycoforms. In vitro characterization showed the enriched CHIKV-immune IgG had enhanced human FcγRIIIa and mouse FcγRIV affinity and FcγR-mediated effector function without reducing virus neutralization in cellular assays. When administered as post-exposure therapy in mice, CHIKV-immune IgG enriched in afucosylated glycoforms promoted reduction in viral load. Discussion Our study provides evidence that, in mice, increasing Fc engagement of FcγRs on effector cells, by leveraging FcγRIIIa-affinity chromatography, enhanced the antiviral activity of CHIKV-immune IgG and reveals a path to produce more effective therapeutics against these and potentially other emerging viruses.
Collapse
Affiliation(s)
- Julie M. Fox
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Vicky Roy
- Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT), and Harvard University, Cambridge, MA, United States
| | - Bronwyn M. Gunn
- Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT), and Harvard University, Cambridge, MA, United States
| | | | - Daved H. Fremont
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO, United States
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, MO, United States
| | - Galit Alter
- Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT), and Harvard University, Cambridge, MA, United States
- Moderna, Inc., Cambridge, MA, United States
| | - Michael S. Diamond
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO, United States
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, MO, United States
| | | |
Collapse
|
11
|
Advances in antibody-based therapy in oncology. NATURE CANCER 2023; 4:165-180. [PMID: 36806801 DOI: 10.1038/s43018-023-00516-z] [Citation(s) in RCA: 89] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 01/10/2023] [Indexed: 02/22/2023]
Abstract
Monoclonal antibodies are a growing class of targeted cancer therapeutics, characterized by exquisite specificity, long serum half-life, high affinity and immune effector functions. In this review, we outline key advances in the field with a particular focus on recent and emerging classes of engineered antibody therapeutic candidates, discuss molecular structure and mechanisms of action and provide updates on clinical development and practice.
Collapse
|
12
|
Whalen KA, Rakhra K, Mehta NK, Steinle A, Michaelson JS, Baeuerle PA. Engaging natural killer cells for cancer therapy via NKG2D, CD16A and other receptors. MAbs 2023; 15:2208697. [PMID: 37165468 PMCID: PMC10173799 DOI: 10.1080/19420862.2023.2208697] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 04/26/2023] [Indexed: 05/12/2023] Open
Abstract
The field of immuno-oncology has revolutionized cancer patient care and improved survival and quality of life for patients. Much of the focus in the field has been on exploiting the power of the adaptive immune response through therapeutic targeting of T cells. While these approaches have markedly advanced the field, some challenges remain, and the clinical benefit of T cell therapies does not extend to all patients or tumor indications. Alternative strategies, such as engaging the innate immune system, have become an intense area of focus in the field. In particular, the engagement of natural killer (NK) cells as potent effectors of the innate immune response has emerged as a promising modality in immunotherapy. Here, we review therapeutic approaches for selective engagement of NK cells for cancer therapy, with a particular focus on targeting the key activating receptors NK Group 2D (NKG2D) and cluster of differentiation 16A (CD16A).
Collapse
Affiliation(s)
- Kerry A. Whalen
- Preclinical and Early Development, Cullinan Oncology, Inc, Cambridge, MA, USA
| | - Kavya Rakhra
- Preclinical and Early Development, Cullinan Oncology, Inc, Cambridge, MA, USA
| | - Naveen K. Mehta
- Preclinical and Early Development, Cullinan Oncology, Inc, Cambridge, MA, USA
| | - Alexander Steinle
- Institute for Molecular Medicine, Goethe-University Frankfurt, Frankfurt am Main, Germany
- Preclinical and Early Development, Frankfurt Cancer Institute, Frankfurt am Main, Germany
| | | | - Patrick A. Baeuerle
- Preclinical and Early Development, Cullinan Oncology, Inc, Cambridge, MA, USA
- Institute for Immunology, Ludwig Maximilians University, Munich, Germany
| |
Collapse
|
13
|
Mariottini A, Muraro PA, Lünemann JD. Antibody-mediated cell depletion therapies in multiple sclerosis. Front Immunol 2022; 13:953649. [PMID: 36172350 PMCID: PMC9511140 DOI: 10.3389/fimmu.2022.953649] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/29/2022] [Indexed: 11/30/2022] Open
Abstract
Development of disease-modifying therapies including monoclonal antibody (mAb)-based therapeutics for the treatment of multiple sclerosis (MS) has been extremely successful over the past decades. Most of the mAb-based therapies approved for MS deplete immune cell subsets and act through activation of cellular Fc-gamma receptors expressed by cytotoxic lymphocytes and phagocytes, resulting in antibody-dependent cellular cytotoxicity or by initiation of complement-mediated cytotoxicity. The therapeutic goal is to eliminate pathogenic immune cell components and to potentially foster the reconstitution of a new and healthy immune system. Ab-mediated immune cell depletion therapies include the CD52-targeting mAb alemtuzumab, CD20-specific therapeutics, and new Ab-based treatments which are currently being developed and tested in clinical trials. Here, we review recent developments in effector mechanisms and clinical applications of Ab-based cell depletion therapies, compare their immunological and clinical effects with the prototypic immune reconstitution treatment strategy, autologous hematopoietic stem cell transplantation, and discuss their potential to restore immunological tolerance and to achieve durable remission in people with MS.
Collapse
Affiliation(s)
- Alice Mariottini
- Department of Brain Sciences, Imperial College London, London, United Kingdom
- Department of Neurosciences, Drug and Child Health, University of Florence, Florence, Italy
| | - Paolo A. Muraro
- Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Jan D. Lünemann
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
- *Correspondence: Jan D. Lünemann,
| |
Collapse
|
14
|
Harnessing IgG Fc glycosylation for clinical benefit. Curr Opin Immunol 2022; 77:102231. [PMID: 35797920 PMCID: PMC9870045 DOI: 10.1016/j.coi.2022.102231] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 06/01/2022] [Accepted: 06/02/2022] [Indexed: 01/26/2023]
Abstract
The effector activity of IgG antibodies is regulated at several levels, including IgG subclass, modifications of the Fc glycan, and the distribution of Type I and II Fcγ receptors (FcγR) on effector cells. Here, we explore how Fc glycosylation, particularly sialylation and fucosylation, tunes cellular responses to immune complexes. We review the current understanding of the pathways and mechanisms underlying this biology, address FcγR in antigen presentation, and discuss aspects of the clinical understanding of Fc glycans in therapies and disease.
Collapse
|
15
|
Golay J, Andrea AE, Cattaneo I. Role of Fc Core Fucosylation in the Effector Function of IgG1 Antibodies. Front Immunol 2022; 13:929895. [PMID: 35844552 PMCID: PMC9279668 DOI: 10.3389/fimmu.2022.929895] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 06/03/2022] [Indexed: 11/13/2022] Open
Abstract
The presence of fucose on IgG1 Asn-297 N-linked glycan is the modification of the human IgG1 Fc structure with the most significant impact on FcɣRIII affinity. It also significantly enhances the efficacy of antibody dependent cellular cytotoxicity (ADCC) by natural killer (NK) cells in vitro, induced by IgG1 therapeutic monoclonal antibodies (mAbs). The effect of afucosylation on ADCC or antibody dependent phagocytosis (ADCP) mediated by macrophages or polymorphonuclear neutrophils (PMN) is less clear. Evidence for enhanced efficacy of afucosylated therapeutic mAbs in vivo has also been reported. This has led to the development of several therapeutic antibodies with low Fc core fucose to treat cancer and inflammatory diseases, seven of which have already been approved for clinical use. More recently, the regulation of IgG Fc core fucosylation has been shown to take place naturally during the B-cell immune response: A decrease in α-1,6 fucose has been observed in polyclonal, antigen-specific IgG1 antibodies which are generated during alloimmunization of pregnant women by fetal erythrocyte or platelet antigens and following infection by some enveloped viruses and parasites. Low IgG1 Fc core fucose on antigen-specific polyclonal IgG1 has been linked to disease severity in several cases, such as SARS-CoV 2 and Dengue virus infection and during alloimmunization, highlighting the in vivo significance of this phenomenon. This review aims to summarize the current knowledge about human IgG1 Fc core fucosylation and its regulation and function in vivo, in the context of both therapeutic antibodies and the natural immune response. The parallels in these two areas are informative about the mechanisms and in vivo effects of Fc core fucosylation, and may allow to further exploit the desired properties of this modification in different clinical contexts.
Collapse
Affiliation(s)
- Josée Golay
- Center of Cellular Therapy "G. Lanzani", Division of Hematology, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
- *Correspondence: Josée Golay,
| | - Alain E. Andrea
- Laboratoire de Biochimie et Thérapies Moléculaires, Faculté de Pharmacie, Université Saint Joseph de Beyrouth, Beirut, Lebanon
| | - Irene Cattaneo
- Center of Cellular Therapy "G. Lanzani", Division of Hematology, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| |
Collapse
|
16
|
Tumor Microenvironment and Immunotherapy-Based Approaches in Mantle Cell Lymphoma. Cancers (Basel) 2022; 14:cancers14133229. [PMID: 35804999 PMCID: PMC9265015 DOI: 10.3390/cancers14133229] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 06/23/2022] [Accepted: 06/26/2022] [Indexed: 01/27/2023] Open
Abstract
Mantle cell lymphoma (MCL) is an aggressive B-cell non-Hodgkin lymphoma (NHL) characterized by the translocation t(11;14) (q13;q32) and a poor response to rituximab–anthracycline-based chemotherapy. High-dose cytarabine-based regimens offer a durable response, but an important number of MCL patients are not eligible for intensive treatment and are ideal candidates for novel targeted therapies (such as BTK, proteasome or BCL2 inhibitors, Immunomodulatory Drugs (IMiDs), bispecific antibodies, or CAR-T cell therapy). On the bench side, several studies aiming to integrate the tumor within its ecosystem highlighted a critical role of the tumor microenvironment (TME) in the expansion and resistance of MCL. This led to important insights into the role of the TME in the management of MCL, including potential targets and biomarkers. Indeed, targeted agents often have a combined mechanism of action on the tumor B cell but also on the tumor microenvironment. The aim of this review is to briefly describe the current knowledge on the biology of the TME in MCL and expose the results of the different therapeutic strategies integrating the TME in this disease.
Collapse
|
17
|
Delidakis G, Kim JE, George K, Georgiou G. Improving Antibody Therapeutics by Manipulating the Fc Domain: Immunological and Structural Considerations. Annu Rev Biomed Eng 2022; 24:249-274. [PMID: 35363537 PMCID: PMC9648538 DOI: 10.1146/annurev-bioeng-082721-024500] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Interactions between the crystallizable fragment (Fc) domain of antibodies and a plethora of cellular Fc receptors (FcRs) or soluble proteins form a critical link between humoral and innate immunity. In particular, the immunoglobulin G Fc domain is critical for the clearance of target cells by processes that include (a) cytotoxicity, phagocytosis, or complement lysis; (b) modulation of inflammation; (c) antigen presentation; (d) antibody-mediated receptor clustering; and (e) cytokine release. More than 30 Fc-engineered antibodies aimed primarily at tailoring these effects for optimal therapeutic outcomes are in clinical evaluation or have already been approved. Nonetheless, our understanding of how FcR engagement impacts various immune cell phenotypes is still largely incomplete. Recent insights into FcR biology coupled with advances in Fc:FcR structural analysis, Fc engineering, and mouse models that recapitulate human biology are helping to fill in existing knowledge gaps. These advances will provide a blueprint on how to fine-tune the Fc domain to achieve optimal therapeutic efficacy. Expected final online publication date for the Annual Review of Biomedical Engineering, Volume 24 is June 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- George Delidakis
- Department of Chemical Engineering, University of Texas at Austin, Austin, Texas, USA;
| | - Jin Eyun Kim
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas, USA
| | - Katia George
- Department of Molecular Biosciences, University of Texas at Austin, Austin, Texas, USA
| | - George Georgiou
- Department of Chemical Engineering, University of Texas at Austin, Austin, Texas, USA; .,Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas, USA.,Department of Molecular Biosciences, University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
18
|
Effects of a Fully Humanized Type II Anti-CD20 Monoclonal Antibody on Peripheral and CNS B Cells in a Transgenic Mouse Model of Multiple Sclerosis. Int J Mol Sci 2022; 23:ijms23063172. [PMID: 35328594 PMCID: PMC8949956 DOI: 10.3390/ijms23063172] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 03/09/2022] [Indexed: 11/17/2022] Open
Abstract
Successful therapy with anti-CD20 monoclonal antibodies (mAbs) has reinforced the key role of B cells in the immunopathology of multiple sclerosis (MS). This study aimed to determine the effects of a novel class of anti-CD20 mAbs on vascular and extravascular central nervous system (CNS)-infiltrating B cells in experimental autoimmune encephalomyelitis (EAE), an animal model of MS. Male hCD20xhIgR3 mice and wild-type C57BL/6 (B6) mice were immunized with human myelin oligodendrocyte glycoprotein (MOG)1–125 to induce EAE. While hCD20xhIgR3 mice were injected intravenously with an anti-human CD20 mAb (5 mg/kg) (rituximab (a type I anti-CD20 mAb) or obinutuzumab (a type II anti-CD20 mAb), B6 mice received the anti-mouse CD20 antibody 18B12. Neither mAb affected clinical disease or serum antibody levels. Obinutuzumab and rituximab had an impact on splenic and CNS-infiltrated B cells with slightly differential depletion efficacy. Additionally, obinutuzumab had beneficial effects on spinal cord myelination. B cell depletion rates in the 18B12/B6 model were comparable with those observed in obinutuzumab-treated hCD20xhIgR3 mice. Our results demonstrate the usefulness of anti-CD20 mAbs for the modulation of B cell-driven peripheral immune response and CNS pathology, with type II antibodies potentially being superior to type I in the depletion of tissue-infiltrating B cells.
Collapse
|
19
|
Musolino A, Gradishar WJ, Rugo HS, Nordstrom JL, Rock EP, Arnaldez F, Pegram MD. Role of Fcγ receptors in HER2-targeted breast cancer therapy. J Immunother Cancer 2022; 10:jitc-2021-003171. [PMID: 34992090 PMCID: PMC8739678 DOI: 10.1136/jitc-2021-003171] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2021] [Indexed: 01/03/2023] Open
Abstract
Several therapeutic monoclonal antibodies (mAbs), including those targeting epidermal growth factor receptor, human epidermal growth factor receptor 2 (HER2), and CD20, mediate fragment crystallizable gamma receptor (FcγR)–dependent activities as part of their mechanism of action. These activities include induction of antibody-dependent cellular cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP), which are innate immune mechanisms of cancer cell elimination. FcγRs are distinguished by their affinity for the Fc fragment, cell distribution, and type of immune response they induce. Activating FcγRIIIa (CD16A) on natural killer cells plays a crucial role in mediating ADCC, and activating FcγRIIa (CD32A) and FcγRIIIa on macrophages are important for mediating ADCP. Polymorphisms in FcγRIIIa and FcγRIIa generate variants that bind to the Fc portion of antibodies with different affinities. This results in differential FcγR-mediated activities associated with differential therapeutic outcomes across multiple clinical settings, from early stage to metastatic disease, in patients with HER2+ breast cancer treated with the anti-HER2 mAb trastuzumab. Trastuzumab has, nonetheless, revolutionized HER2+ breast cancer treatment, and several HER2-directed mAbs have been developed using Fc glyco-engineering or Fc protein-engineering to enhance FcγR-mediated functions. An example of an approved anti-HER2 Fc-engineered chimeric mAb is margetuximab, which targets the same epitope as trastuzumab, but features five amino acid substitutions in the IgG 1 Fc domain that were deliberately introduced to increase binding to activating FcγRIIIa and decrease binding to inhibitory FcγRIIb (CD32B). Margetuximab enhances Fc-dependent ADCC in vitro more potently than the combination of pertuzumab (another approved mAb directed against an alternate HER2 epitope) and trastuzumab. Margetuximab administration also enhances HER2-specific B cell and T cell–mediated responses ex vivo in samples from patients treated with prior lines of HER2 antibody-based therapies. Stemming from these observations, a worthwhile future goal in the treatment of HER2+ breast cancer is to promote combinatorial approaches that better eradicate HER2+ cancer cells via enhanced immunological mechanisms.
Collapse
Affiliation(s)
- Antonino Musolino
- Department of Medicine and Surgery, University Hospital of Parma, Medical Oncology and Breast Unit, Parma, Italy
| | - William J Gradishar
- Division of Hematology/Oncology, Northwestern University, Chicago, Illinois, USA
| | - Hope S Rugo
- Helen Diller Family Comprehensive Cancer Center, Breast Oncology and Clinical Trials Education, University of California San Francisco, San Francisco, California, USA
| | | | | | | | - Mark D Pegram
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
20
|
Abstract
Antibody-based therapeutics are powerful tools to treat disease. While their mechanism of action (MOA) always involves binding to a specific target via the antibody-binding fragment (Fab) region of the antibody, the induction of immune-mediated effector functions through the fragment crystallizable (Fc) region is a vital aspect of antibody therapeutics targeting tumor cells. Cross-linking of the Fc gamma receptors (FcγRs) via cell-bound antibodies activate immune effector cells, leading to antibody-dependent cellular cytotoxicity via natural killer (NK) cells. Linking of FcγRs on macrophages triggers the process of antibody-dependent cellular phagocytosis (ADCP), where antibody-opsonized target cells are internalized in phagosomes and degraded through the process of phagosome maturation and acidification. ADCP activity can be challenging to measure accurately due to the difficulty in differentiating target cells that are bound to a macrophage versus those that are internalized within phagosomes. In this chapter, we describe a protocol that measures ADCP activity by labeling target cells with a pH-sensitive dye that fluoresces brightly in mature phagosomes. The ADCP activity of therapeutics is then measured via flow cytometry. This assay is capable of detecting glycosylation differences arising from manufacturing processes and is suitable for evaluation of ADCP activity of monoclonal antibodies (mAb) to support in vitro biological characterization of drug candidates and lead candidate selection for desirable effector functions.
Collapse
|
21
|
Nawab DH. Vaccinal antibodies: Fc antibody engineering to improve the antiviral antibody response and induce vaccine-like effects. Hum Vaccin Immunother 2021; 17:5532-5545. [PMID: 34844516 PMCID: PMC8903937 DOI: 10.1080/21645515.2021.1985891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 09/21/2021] [Indexed: 10/19/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic highlights the urgent clinical need for efficient virus therapies and vaccines. Although the functional importance of antibodies is indisputable in viral infections, there are still significant unmet needs that require vast improvements in antibody-based therapeutics. The IgG Fc domain can be engineered to produce antibodies with tailored and potent responses that will meet these clinical demands. Engaging Fc receptors (FcRs) to perform effector functions as cytotoxicity, phagocytosis, complement activation, intracellular neutralization and controlling antibody persistence. Furthermore, it produces vaccine-like effects by activating signals to stimulate T-cell responses, have proven to be required for protection, as neutralization alone does not off the full protection capacity of antibodies. This review highlights antiviral Fc functions and FcRs' contributions in linking innate and adaptive immunity against viral threats. Moreover, it provides the latest Fc engineering strategies to improve the safety and efficacy of human antiviral antibodies and vaccines.
Collapse
Affiliation(s)
- Dhuha H Nawab
- Pharmacy Department, Ministry of Health, Saudi Arabia
| |
Collapse
|
22
|
Follicular lymphoma and macrophages: impact of approved and novel therapies. Blood Adv 2021; 5:4303-4312. [PMID: 34570196 PMCID: PMC8945644 DOI: 10.1182/bloodadvances.2021005722] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 08/08/2021] [Indexed: 12/20/2022] Open
Abstract
The survival and proliferation of follicular lymphoma (FL) cells are strongly dependent on macrophages, because their presence is necessary for the propagation of FL cells in vitro. To this regard, as also shown for the majority of solid tumors, a high tissue content of tumor-associated macrophages (TAMs), particularly if showing a protumoral phenotype (also called M2), is strongly associated with a poor outcome among patients with FL treated with chemotherapy. The introduction of rituximab, an anti-CD20 antibody that can be used by TAMs to facilitate antibody-dependent cellular cytotoxicity and antibody-dependent cellular phagocytosis, has challenged this paradigm. In the rituximab era, clinical studies have yielded conflicting results in FL, showing variable outcomes based on the type of regimen used. This highlighted, for the first time, that the impact of TAMs on the prognosis of patients with FL may depend on the administered treatment, emphasizing the need to better understand how currently available therapies affect macrophage function in FL. We summarize the impact of approved and novel therapies for FL, including radiation therapy, chemotherapy, anti-CD20 monoclonal antibodies, lenalidomide, and targeted agents, on the biology of TAMs and describe their effects on macrophage phagocytosis, polarization, and function. Although novel agents targeting the CD47/SIRPα axis are being developed and show promising activity in FL, a deeper understanding of macrophage biology and their complex pathways will help to develop novel and safer therapeutic strategies for patients with this type of lymphoma.
Collapse
|
23
|
The selection of variable regions affects effector mechanisms of IgA antibodies against CD20. Blood Adv 2021; 5:3807-3820. [PMID: 34525171 DOI: 10.1182/bloodadvances.2021004598] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 07/05/2021] [Indexed: 11/20/2022] Open
Abstract
Blockade of the CD47-SIRPα axis improves lymphoma cell killing by myeloid effector cells, which is an important effector mechanism for CD20 antibodies in vivo. The approved CD20 antibodies rituximab, ofatumumab, and obinutuzumab are of human immunoglobulin G1 (IgG1) isotype. We investigated the impact of the variable regions of these 3 CD20 antibodies when expressed as human IgA2 isotype variants. All 3 IgA2 antibodies mediated antibody-dependent cellular phagocytosis (ADCP) by macrophages and antibody-dependent cellular cytotoxicity (ADCC) by polymorphonuclear cells. Both effector mechanisms were significantly enhanced in the presence of a CD47-blocking antibody or by glutaminyl cyclase inhibition to interfere with CD47-SIRPα interactions. Interestingly, an IgA2 variant of obinutuzumab (OBI-IgA2) was consistently more potent than an IgA2 variant of rituximab (RTX-IgA2) or an IgA2 variant of ofatumumab (OFA-IgA2) in triggering ADCC. Furthermore, we observed more effective direct tumor cell killing by OBI-IgA2 compared with RTX-IgA2 and OFA-IgA2, which was caspase independent and required a functional cytoskeleton. IgA2 variants of all 3 antibodies triggered complement-dependent cytotoxicity, with OBI-IgA2 being less effective than RTX-IgA2 and OFA-IgA2. When we investigated the therapeutic efficacy of the CD20 IgA2 antibodies in different in vivo models, OBI-IgA2 was therapeutically more effective than RTX-IgA2 or OFA-IgA2. In vivo efficacy required the presence of a functional IgA receptor on effector cells and was independent of complement activation or direct lymphoma cell killing. These data characterize the functional activities of human IgA2 antibodies against CD20, which were affected by the selection of the respective variable regions. OBI-IgA2 proved particularly effective in vitro and in vivo, which may be relevant in the context of CD47-SIRPα blockade.
Collapse
|
24
|
Liu C, Yu C, Yang Y, Huang J, Yu X, Duan M, Wang L, Wang J. Development of a novel reporter gene assay to evaluate antibody-dependent cellular phagocytosis for anti-CD20 therapeutic antibodies. Int Immunopharmacol 2021; 100:108112. [PMID: 34521023 DOI: 10.1016/j.intimp.2021.108112] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/25/2021] [Accepted: 08/27/2021] [Indexed: 10/20/2022]
Abstract
More than 100 monoclonal antibodies (mAbs) have been approved by FDA. The mechanism of action (MoA) involves in neutralization of a specific target via the Fab region and Fc effector functions through Fc region, while the latter include complement-dependent cytotoxicity (CDC), antibody-dependent cell-mediated cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP). ADCP has been recognized one of the most important MoAs, especially for anti-cancer mAbs in recent years. However, traditional bioassays measuring ADCP always introduced primary macrophages and flow cytometry, which are difficult to handle and highly variable. In this study, we engineered a monoclonal Jurkat/NFAT/CD32a-FcεRIγ effector cell line that stably expresses CD32a-FcεRIγ chimeric receptor and NFAT-controlled luciferase. The corresponding mAb could bind with the membrane antigens on the target cells with its Fab fragment and CD32a-FcεRIγ on the effector cells with its Fc fragment, leading to the crosslinking of CD32a-FcεRIγ and the resultant expression of subsequent NFAT-controlled luciferase, which represents the bioactivity of ADCP based on the MoA of the mAb. With rituximab as the model mAb, Raji cells as the target cells, and Jurkat/NFAT/CD32a-FcεRIγ cells as the effector cells, we adopted the strategy of Design of Experiment (DoE) to optimize the bioassay. Then we fully validated the established bioassay according to ICH-Q2(R1), which proved the good assay performance characteristics of the bioassay, including specificity, accuracy, precision, linearity, stability and robustness. This RGA can be applied to evaluate the -ADCP bioactivity for anti-CD20 mAbs in lot release, stability testing as well as biosimilar comparability. The engineered cells may also potentially be used to evaluate the ADCP bioactivity of mAbs with other targets.
Collapse
Affiliation(s)
- Chunyu Liu
- Division of Monoclonal Antibody Products, National Institu-tes for Food and Drug Control, Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, Beijing 102629, China
| | - Chuanfei Yu
- Division of Monoclonal Antibody Products, National Institu-tes for Food and Drug Control, Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, Beijing 102629, China
| | - Yalan Yang
- Division of Monoclonal Antibody Products, National Institu-tes for Food and Drug Control, Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, Beijing 102629, China
| | - Jing Huang
- Division of Monoclonal Antibody Products, National Institu-tes for Food and Drug Control, Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, Beijing 102629, China
| | - Xiaojuan Yu
- Division of Monoclonal Antibody Products, National Institu-tes for Food and Drug Control, Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, Beijing 102629, China
| | - Maoqin Duan
- Division of Monoclonal Antibody Products, National Institu-tes for Food and Drug Control, Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, Beijing 102629, China
| | - Lang Wang
- Division of Monoclonal Antibody Products, National Institu-tes for Food and Drug Control, Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, Beijing 102629, China.
| | - Junzhi Wang
- Division of Monoclonal Antibody Products, National Institu-tes for Food and Drug Control, Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, Beijing 102629, China
| |
Collapse
|
25
|
Mao C, Near R, Zhong X, Gao W. Cross-species higher sensitivities of FcγRIIIA/FcγRIV to afucosylated IgG for enhanced ADCC. Antib Ther 2021; 4:159-170. [PMID: 34485821 PMCID: PMC8408537 DOI: 10.1093/abt/tbab016] [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: 06/30/2021] [Revised: 07/26/2021] [Accepted: 08/13/2021] [Indexed: 11/27/2022] Open
Abstract
Background Expressing afucosylated human IgG1 antibodies with Chinese hamster ovary (CHO) cells deficient of α-(1,6)-fucosyltransferase (FUT8) is being more and more accepted as a routine method to enhance antibody-dependent cellular cytotoxicity (ADCC) of therapeutic antibodies, especially for anti-cancer regimens. However, in pre-clinical studies relying on disease models other than mice and primates, e.g., those underrepresented species for infectious diseases, it is less clear whether such afucosylated antibodies can demonstrate enhanced therapeutic index. This is because the orthologues of human FcγRIIIA or mouse FcγRIV from those species have not been well characterized. Methods We set up a luciferase-based ADCC assay with Jurkat reporter cells expressing FcγRIIIA/FcγRIV from human, mouse, rat, hamster, guinea pig, ferret, rabbit, cat, dog, pig and monkey, and also produced human, mouse, hamster, rabbit and pig IgG from wild type and Fut8−/− CHO cells or hybridomas. Results We confirmed that enhanced stimulation through FcγRIIIA/FcγRIV by afucosylated IgG, as compared with wild type IgG, is a cross-species phenomenon. Conclusions Thus, efficacy and toxicology studies of the next generation afucosylated therapeutic IgG and Fc fusion proteins in these underrepresented animal models should be expected to generate translatable data for treating human diseases, leading to the expanded applications of this new class of glycoengineered biologics.
Collapse
Affiliation(s)
| | - Richard Near
- Department of Medicine, Boston University Medical Center, Boston, MA 02118, USA
| | - Xuemei Zhong
- Department of Medicine, Boston University Medical Center, Boston, MA 02118, USA
| | - Wenda Gao
- Antagen Pharmaceuticals, Inc., Canton, MA 02021, USA
| |
Collapse
|
26
|
Rodgers RJ, Abbott JA, Walters KA, Ledger WL. Translational Physiology of Anti-Müllerian Hormone: Clinical Applications in Female Fertility Preservation and Cancer Treatment. Front Endocrinol (Lausanne) 2021; 12:689532. [PMID: 34557157 PMCID: PMC8454407 DOI: 10.3389/fendo.2021.689532] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 08/09/2021] [Indexed: 12/21/2022] Open
Abstract
Background Whilst the ability of AMH to induce the regression of the Müllerian ducts in the male fetus is well appreciated, AMH has additional biological actions in relation to steroid biosynthesis and ovarian follicle dynamics. An understanding of the physiology of AMH illuminates the potential therapeutic utility of AMH to protect the ovarian reserve during chemotherapy and in the treatment of female malignancies. The translation of the biological actions of AMH into clinical applications is an emerging focus of research, with promising preliminary results. Objective and Rationale Studies indicate AMH restrains primordial follicle development, thus administration of AMH during chemotherapy may protect the ovarian reserve by preventing the mass activation of primordial follicles. As AMH induces regression of tissues expressing the AMH receptor (AMHRII), administration of AMH may inhibit growth of malignancies expressing AMHR II. This review evaluates the biological actions of AMH in females and appraises human clinical applications. Search Methods A comprehensive search of the Medline and EMBASE databases seeking articles related to the physiological functions and therapeutic applications of AMH was conducted in July 2021. The search was limited to studies published in English. Outcomes AMH regulates primordial follicle recruitment and moderates sex steroid production through the inhibition of transcription of enzymes in the steroid biosynthetic pathway, primarily aromatase and 17α-hydroxylase/17,20-lyase. Preliminary data indicates that administration of AMH to mice during chemotherapy conveys a degree of protection to the ovarian reserve. Administration of AMH at the time of ovarian tissue grafting has the potential to restrain uncontrolled primordial follicle growth during revascularization. Numerous studies demonstrate AMH induced regression of AMHR II expressing malignancies. As this action occurs via a different mechanism to traditional chemotherapeutic agents, AMH has the capacity to inhibit proliferation of chemo-resistant ovarian cancer cells and cancer stem cells. Wider Implications To date, AMH has not been administered to humans. Data identified in this review suggests administration of AMH would be safe and well tolerated. Administration of AMH during chemotherapy may provide a synchronistic benefit to women with an AMHR II expressing malignancy, protecting the ovarian reserve whilst the cancer is treated by dual mechanisms.
Collapse
Affiliation(s)
- Rachael Jean Rodgers
- School of Women’s and Children’s Health, University of New South Wales, Sydney, NSW, Australia
| | | | | | | |
Collapse
|
27
|
Elter A, Yanakieva D, Fiebig D, Hallstein K, Becker S, Betz U, Kolmar H. Protease-Activation of Fc-Masked Therapeutic Antibodies to Alleviate Off-Tumor Cytotoxicity. Front Immunol 2021; 12:715719. [PMID: 34413859 PMCID: PMC8369199 DOI: 10.3389/fimmu.2021.715719] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 07/19/2021] [Indexed: 11/18/2022] Open
Abstract
The interaction of the Fc region of therapeutic antibodies and antibody-drug conjugates with Fcγ receptors (FcγRs) can lead to unpredictable and severe side effects. Over the last decades several strategies have been developed to overcome this drawback, including extensive Fc- and glycoengineering and antibody isotype switching. However, these approaches result in permanently Fc-silenced antibody derivates which partially or completely lack antibody-mediated effector functions. Nevertheless, for a majority of antibody-based drugs, Fc-mediated effector functions, like antibody-dependent cell-mediated cytotoxicity (ADCC), antibody-dependent cell-mediated phagocytosis (ADCP) as well as complement-dependent cytotoxicity (CDC), represent the most substantial modes of action. We argued that a new strategy combining the beneficial properties of Fc-silencing and controlled activation of effector functions can pave the way to potent antibody therapeutics, reducing the FcγRs-mediated off-target toxicity. We present a novel Fc-tamed antibody format, where the FcγR-binding sites of antibodies are blocked by anti-isotypic masking units, hindering the association of FcγR and complement component 1 (c1q) to the Fc domain. The masking units were genetically fused to trastuzumab, including a protease-addressable peptide-liker. Our Fc-tamed antibodies demonstrated completely abolished interaction to soluble high-affinity Fcγ-Receptor I and c1q. In reporter cell-based ADCC assays, our Fc-tamed antibodies exhibited a 2,700 to 7,100-fold reduction in activation, compared to trastuzumab. Upon demasking by a tumor-associated protease, the Fc-activated antibodies demonstrated restored FcγR-binding, c1q-binding and the ability to induce potent ADCC activation. Furthermore, cell killing assays using donor-derived NK cells were performed to validate the functionality of the Fc-tamed antibody variants. To our knowledge, this approach represents the first non-permanently Fc-silenced antibody, which can be re-activated by a tumor-associated protease, eventually extending the field of novel antibody formats.
Collapse
Affiliation(s)
- Adrian Elter
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
| | - Desislava Yanakieva
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany.,Protein Engineering and Antibody Technologies, Merck Healthcare KGaA, Darmstadt, Germany
| | - David Fiebig
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
| | - Kerstin Hallstein
- Protein Engineering and Antibody Technologies, Merck Healthcare KGaA, Darmstadt, Germany
| | - Stefan Becker
- Protein Engineering and Antibody Technologies, Merck Healthcare KGaA, Darmstadt, Germany
| | - Ulrich Betz
- Protein Engineering and Antibody Technologies, Merck Healthcare KGaA, Darmstadt, Germany
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
| |
Collapse
|
28
|
Tikhonov A, Smoldovskaya O, Feyzkhanova G, Kushlinskii N, Rubina A. Glycan-specific antibodies as potential cancer biomarkers: a focus on microarray applications. Clin Chem Lab Med 2021; 58:1611-1622. [PMID: 32324152 DOI: 10.1515/cclm-2019-1161] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Accepted: 03/10/2020] [Indexed: 02/06/2023]
Abstract
Glycosylation is one of the most common posttranslational modifications of proteins and lipids. In the case of tumors, cell transformation accompanied by aberrant glycosylation results in the expression of tumor-associated glycans that promote tumor invasion. As part of the innate immunity, anti-glycan antibodies recognize tumor-associated glycans, and these antibodies can be present in the bloodstream in the early stages of cancer. Recently, anti-glycan antibody profiles have been of interest in various cancer studies. Novel advantages in the field of analytical techniques have simplified the analysis of anti-glycan antibodies and made it easier to have more comprehensive knowledge about their functions. One of the robust approaches for studying anti-glycan antibodies engages in microarray technology. The analysis of glycan microarrays can provide more expanded information to simultaneously specify or suggest the role of antibodies to a wide variety of glycans in the progression of different diseases, therefore making it possible to identify new biomarkers for diagnosing cancer and/or the state of the disease. Thus, in this review, we discuss antibodies to various glycans, their application for diagnosing cancer and one of the most promising tools for the investigation of these molecules, microarrays.
Collapse
Affiliation(s)
- Aleksei Tikhonov
- Laboratory of Biological Microchips, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Olga Smoldovskaya
- Laboratory of Biological Microchips, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Guzel Feyzkhanova
- Laboratory of Biological Microchips, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Nikolay Kushlinskii
- Laboratory of Clinical Biochemistry, Federal State Budgetary Institution «N.N. Blokhin National Medical Research Center of Oncology» оf the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Alla Rubina
- Laboratory of Biological Microchips, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
29
|
Prat M, Salon M, Allain T, Dubreuil O, Noël G, Preisser L, Jean B, Cassard L, Lemée F, Tabah-Fish I, Pipy B, Jeannin P, Prost JF, Barret JM, Coste A. Murlentamab, a Low Fucosylated Anti-Müllerian Hormone Type II Receptor (AMHRII) Antibody, Exhibits Anti-Tumor Activity through Tumor-Associated Macrophage Reprogrammation and T Cell Activation. Cancers (Basel) 2021; 13:cancers13081845. [PMID: 33924378 PMCID: PMC8070390 DOI: 10.3390/cancers13081845] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/19/2021] [Accepted: 04/07/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary AMHRII, the anti-Müllerian hormone receptor, is selectively expressed in normal sexual organs in healthy adults but is also re-expressed in ovarian, colorectal and lung cancers. In this context, we developed murlentamab, a humanized glyco-engineered anti-AMHRII monoclonal antibody, currently in clinical trial. Preliminary data suggest that murlentamab anti-tumor activity involves immune response activation. Thus, in vitro experiments were performed to precisely characterize the murlentamab effect on the human immune system. We show that murlentamab treatment is associated with evidences of innate and adaptive immune cell activation in cancer patient samples. Moreover, we demonstrate that the murlentamab opsonization of AMHRII-expressing ovarian tumor cells promotes a polarization switch of both naïve and tumor-associated macrophages towards an anti-tumor M1-like phenotype. Our work also supports that, through macrophage reeducation, murlentamab activates an anti-tumor adaptive immune response. Finally, the combination of murlentamab with pembrolizumab confirmed novel clinical perspectives of murlentamab association with checkpoint inhibitors and other immuno-modulators. Abstract AMHRII, the anti-Müllerian hormone receptor, is selectively expressed in normal sexual organs but is also re-expressed in gynecologic cancers. Hence, we developed murlentamab, a humanized glyco-engineered anti-AMHRII monoclonal antibody currently in clinical trial. Low-fucosylated antibodies are known to increase the antibody-dependent cell-mediated cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP) potency of effector cells, but some preliminary results suggest a more global murlentamab-dependent activation of the immune system. In this context, we demonstrate here that the murlentamab opsonization of AMHRII-expressing ovarian tumor cells, in the presence of unstimulated- or tumor-associated macrophage (TAM)-like macrophages, significantly promotes macrophage-mediated ADCC and shifts the whole microenvironment towards a pro-inflammatory and anti-tumoral status, thus triggering anti-tumor activity. We also report that murlentamab orients both unstimulated- and TAM-like macrophages to an M1-like phenotype characterized by a strong expression of co-stimulation markers, pro-inflammatory cytokines and chemokines, favoring T cell recruitment and activation. Moreover, we show that murlentamab treatment shifts CD4+ Th1/Th2 balance towards a Th1 response and activates CD8+ T cells. Altogether, these results suggest that murlentamab, through naïve macrophage orientation and TAM reprogrammation, stimulates the anti-tumor adaptive immune response. Those mechanisms might contribute to the sustained clinical benefit observed in advanced cancer patients treated with murlentamab. Finally, the enhanced murlentamab activity in combination with pembrolizumab opens new therapeutic perspectives.
Collapse
Affiliation(s)
- Mélissa Prat
- UMR 152 Pharma Dev, Université de Toulouse, IRD, UPS, 31062 Toulouse, France; (M.P.); (M.S.); (T.A.); (B.P.)
| | - Marie Salon
- UMR 152 Pharma Dev, Université de Toulouse, IRD, UPS, 31062 Toulouse, France; (M.P.); (M.S.); (T.A.); (B.P.)
- RESTORE Research Center, Université de Toulouse, INSERM, CNRS, EFS, UPS, 31100 Toulouse, France
| | - Thibault Allain
- UMR 152 Pharma Dev, Université de Toulouse, IRD, UPS, 31062 Toulouse, France; (M.P.); (M.S.); (T.A.); (B.P.)
| | - Olivier Dubreuil
- GamaMabs Pharma, 31106 Toulouse, France; (O.D.); (B.J.); (F.L.); (J.-F.P.); (J.-M.B.)
| | - Grégory Noël
- Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium;
| | - Laurence Preisser
- Univ Angers, Université de Nantes, CHU Angers, Inserm, CRCINA, SFR ICAT, 49000 Angers, France; (L.P.); (P.J.)
| | - Bérangère Jean
- GamaMabs Pharma, 31106 Toulouse, France; (O.D.); (B.J.); (F.L.); (J.-F.P.); (J.-M.B.)
| | - Lydie Cassard
- Laboratory of Immunomonitoring in Oncology, Gustave Roussy, 94905 Villejuif, France;
| | - Fanny Lemée
- GamaMabs Pharma, 31106 Toulouse, France; (O.D.); (B.J.); (F.L.); (J.-F.P.); (J.-M.B.)
| | - Isabelle Tabah-Fish
- GamaMabs Pharma, 31106 Toulouse, France; (O.D.); (B.J.); (F.L.); (J.-F.P.); (J.-M.B.)
| | - Bernard Pipy
- UMR 152 Pharma Dev, Université de Toulouse, IRD, UPS, 31062 Toulouse, France; (M.P.); (M.S.); (T.A.); (B.P.)
- RESTORE Research Center, Université de Toulouse, INSERM, CNRS, EFS, UPS, 31100 Toulouse, France
| | - Pascale Jeannin
- Univ Angers, Université de Nantes, CHU Angers, Inserm, CRCINA, SFR ICAT, 49000 Angers, France; (L.P.); (P.J.)
| | - Jean-François Prost
- GamaMabs Pharma, 31106 Toulouse, France; (O.D.); (B.J.); (F.L.); (J.-F.P.); (J.-M.B.)
| | - Jean-Marc Barret
- GamaMabs Pharma, 31106 Toulouse, France; (O.D.); (B.J.); (F.L.); (J.-F.P.); (J.-M.B.)
| | - Agnès Coste
- UMR 152 Pharma Dev, Université de Toulouse, IRD, UPS, 31062 Toulouse, France; (M.P.); (M.S.); (T.A.); (B.P.)
- RESTORE Research Center, Université de Toulouse, INSERM, CNRS, EFS, UPS, 31100 Toulouse, France
- Correspondence: ; Tel.: +33-534-609-501
| |
Collapse
|
30
|
Damelang T, Aitken EH, Hasang W, Lopez E, Killian M, Unger HW, Salanti A, Shub A, McCarthy E, Kedzierska K, Lappas M, Kent SJ, Rogerson SJ, Chung AW. Antibody mediated activation of natural killer cells in malaria exposed pregnant women. Sci Rep 2021; 11:4130. [PMID: 33602987 PMCID: PMC7893158 DOI: 10.1038/s41598-021-83093-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 01/25/2021] [Indexed: 12/21/2022] Open
Abstract
Immune effector responses against Plasmodium falciparum include antibody-mediated activation of innate immune cells, which can induce Fc effector functions, including antibody-dependent cellular cytotoxicity, and the secretion of cytokines and chemokines. These effector functions are regulated by the composition of immunoglobulin G (IgG) Fc N-linked glycans. However, a role for antibody-mediated natural killer (NK) cells activation or Fc N-linked glycans in pregnant women with malaria has not yet been established. Herein, we studied the capacity of IgG antibodies from pregnant women, with placental malaria or non-placental malaria, to induce NK cell activation in response to placental malaria-associated antigens DBL2 and DBL3. Antibody-mediated NK cell activation was observed in pregnant women with malaria, but no differences were associated with susceptibility to placental malaria. Elevated anti-inflammatory glycosylation patterns of IgG antibodies were observed in pregnant women with or without malaria infection, which were not seen in healthy non-pregnant controls. This suggests that pregnancy-associated anti-inflammatory Fc N-linked glycans may dampen the antibody-mediated activation of NK cells in pregnant women with malaria infection. Overall, although anti-inflammatory glycans and antibody-dependent NK cell activation were detected in pregnant women with malaria, a definitive role for these antibody features in protecting against placental malaria remains to be proven.
Collapse
Affiliation(s)
- Timon Damelang
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Elizabeth H Aitken
- Department of Medicine, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Wina Hasang
- Department of Medicine, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Ester Lopez
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Martin Killian
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
- Department of Internal Medicine, Centre Hospitalier Universitaire de Saint-Etienne, Saint-Etienne, France
- Groupe sur l'Immunité des Muqueuses et Agents Pathogènes, Université Jean Monnet Saint-Etienne, Saint-Etienne, France
| | - Holger W Unger
- Liverpool School of Tropical Medicine, Liverpool, UK
- Department of Obstetrics and Gynaecology, Royal Darwin Hospital, Darwin, NT, Australia
- Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia
| | - Ali Salanti
- Department for Immunology and Microbiology, Centre for Medical Parasitology, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Disease, Copenhagen University Hospital, Copenhagen, Denmark
| | - Alexis Shub
- Department of Obstetrics and Gynaecology, University of Melbourne, Melbourne, VIC, Australia
| | - Elizabeth McCarthy
- Department of Obstetrics and Gynaecology, University of Melbourne, Melbourne, VIC, Australia
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Martha Lappas
- Department of Obstetrics and Gynaecology, University of Melbourne, Melbourne, VIC, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
- Infectious Diseases Department, Alfred Health, Melbourne Sexual Health Centre, Monash University, Melbourne, VIC, Australia
| | - Stephen J Rogerson
- Department of Medicine, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Amy W Chung
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
31
|
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: 6.0] [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
|
32
|
Grandjean CL, Garcia Z, Lemaître F, Bréart B, Bousso P. Imaging the mechanisms of anti-CD20 therapy in vivo uncovers spatiotemporal bottlenecks in antibody-dependent phagocytosis. SCIENCE ADVANCES 2021; 7:7/8/eabd6167. [PMID: 33608271 PMCID: PMC7895428 DOI: 10.1126/sciadv.abd6167] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 12/31/2020] [Indexed: 05/16/2023]
Abstract
Anti-CD20 antibody (mAb) represents an effective strategy for the treatment of B cell malignancies, possibly involving complement activity, antibody-dependent cellular cytotoxicity and phagocytosis (ADP). While ADP by Kupffer cells deplete circulating tumors, mechanisms targeting non-circulating tumors remain unclear. Using intravital imaging in a model of B cell lymphoma, we establish here the dominance and limitations of ADP in the bone marrow (BM). We found that tumor cells were stably residing in the BM with little evidence for recirculation. To elucidate the mechanism of depletion, we designed a dual fluorescent reporter to visualize phagocytosis and apoptosis. ADP by BM-associated macrophages was the primary mode of tumor elimination but was no longer active after one hour, resulting in partial depletion. Moreover, macrophages were present at low density in tumor-rich regions, targeting only neighboring tumors. Overcoming spatiotemporal bottlenecks in tumor-targeting Ab therapy thus represents a critical path towards the design of optimized therapies.
Collapse
Affiliation(s)
- Capucine L Grandjean
- Dynamics of Immune Responses Unit, Equipe Labellisée Ligue Contre le Cancer, Institut Pasteur, 75015 Paris, France.
- INSERM U1223, 75015 Paris, France
| | - Zacarias Garcia
- Dynamics of Immune Responses Unit, Equipe Labellisée Ligue Contre le Cancer, Institut Pasteur, 75015 Paris, France
- INSERM U1223, 75015 Paris, France
| | - Fabrice Lemaître
- Dynamics of Immune Responses Unit, Equipe Labellisée Ligue Contre le Cancer, Institut Pasteur, 75015 Paris, France
- INSERM U1223, 75015 Paris, France
| | - Béatrice Bréart
- Dynamics of Immune Responses Unit, Equipe Labellisée Ligue Contre le Cancer, Institut Pasteur, 75015 Paris, France
- INSERM U1223, 75015 Paris, France
| | - Philippe Bousso
- Dynamics of Immune Responses Unit, Equipe Labellisée Ligue Contre le Cancer, Institut Pasteur, 75015 Paris, France.
- INSERM U1223, 75015 Paris, France
| |
Collapse
|
33
|
Narimatsu Y, Büll C, Chen YH, Wandall HH, Yang Z, Clausen H. Genetic glycoengineering in mammalian cells. J Biol Chem 2021; 296:100448. [PMID: 33617880 PMCID: PMC8042171 DOI: 10.1016/j.jbc.2021.100448] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 02/17/2021] [Accepted: 02/18/2021] [Indexed: 02/06/2023] Open
Abstract
Advances in nuclease-based gene-editing technologies have enabled precise, stable, and systematic genetic engineering of glycosylation capacities in mammalian cells, opening up a plethora of opportunities for studying the glycome and exploiting glycans in biomedicine. Glycoengineering using chemical, enzymatic, and genetic approaches has a long history, and precise gene editing provides a nearly unlimited playground for stable engineering of glycosylation in mammalian cells to explore and dissect the glycome and its many biological functions. Genetic engineering of glycosylation in cells also brings studies of the glycome to the single cell level and opens up wider use and integration of data in traditional omics workflows in cell biology. The last few years have seen new applications of glycoengineering in mammalian cells with perspectives for wider use in basic and applied glycosciences, and these have already led to discoveries of functions of glycans and improved designs of glycoprotein therapeutics. Here, we review the current state of the art of genetic glycoengineering in mammalian cells and highlight emerging opportunities.
Collapse
Affiliation(s)
- Yoshiki Narimatsu
- Department of Cellular and Molecular Medicine, Faculty of Health Sciences, Copenhagen Center for Glycomics, University of Copenhagen, Copenhagen, Denmark; GlycoDisplay ApS, Copenhagen, Denmark.
| | - Christian Büll
- Department of Cellular and Molecular Medicine, Faculty of Health Sciences, Copenhagen Center for Glycomics, University of Copenhagen, Copenhagen, Denmark.
| | | | - Hans H Wandall
- Department of Cellular and Molecular Medicine, Faculty of Health Sciences, Copenhagen Center for Glycomics, University of Copenhagen, Copenhagen, Denmark
| | - Zhang Yang
- Department of Cellular and Molecular Medicine, Faculty of Health Sciences, Copenhagen Center for Glycomics, University of Copenhagen, Copenhagen, Denmark; GlycoDisplay ApS, Copenhagen, Denmark
| | - Henrik Clausen
- Department of Cellular and Molecular Medicine, Faculty of Health Sciences, Copenhagen Center for Glycomics, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
34
|
Mimura Y, Saldova R, Mimura-Kimura Y, Rudd PM, Jefferis R. Importance and Monitoring of Therapeutic Immunoglobulin G Glycosylation. EXPERIENTIA SUPPLEMENTUM (2012) 2021; 112:481-517. [PMID: 34687020 DOI: 10.1007/978-3-030-76912-3_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The complex diantennary-type oligosaccharides at Asn297 residues of the IgG heavy chains have a profound impact on the safety and efficacy of therapeutic IgG monoclonal antibodies (mAbs). Fc glycosylation of a mAb is an established critical quality attribute (CQA), and its oligosaccharide profile is required to be thoroughly characterized by state-of-the-art analytical methods. The Fc oligosaccharides are highly heterogeneous, and the differentially glycosylated species (glycoforms) of IgG express unique biological activities. Glycoengineering is a promising approach for the production of selected mAb glycoforms with improved effector functions, and non- and low-fucosylated mAbs exhibiting enhanced antibody-dependent cellular cytotoxicity activity have been approved or are under clinical evaluation for treatment of cancers, autoimmune/chronic inflammatory diseases, and infection. Recently, the chemoenzymatic glycoengineering method that allows for the transfer of structurally defined oligosaccharides to Asn-linked GlcNAc residues with glycosynthase has been developed for remodeling of IgG-Fc oligosaccharides with high efficiency and flexibility. Additionally, various glycoengineering methods have been developed that utilize the Fc oligosaccharides of IgG as reaction handles to conjugate cytotoxic agents by "click chemistry", providing new routes to the design of antibody-drug conjugates (ADCs) with tightly controlled drug-antibody ratios (DARs) and homogeneity. This review focuses on current understanding of the biological relevance of individual IgG glycoforms and advances in the development of next-generation antibody therapeutics with improved efficacy and safety through glycoengineering.
Collapse
Affiliation(s)
- Yusuke Mimura
- Department of Clinical Research, National Hospital Organization Yamaguchi Ube Medical Center, Ube, Japan.
| | - Radka Saldova
- NIBRT GlycoScience Group, National Institute for Bioprocessing Research and Training, Mount Merrion, Blackrock, Dublin, Ireland
- UCD School of Medicine, College of Health and Agricultural Science, University College Dublin, Belfield, Dublin, Ireland
| | - Yuka Mimura-Kimura
- Department of Clinical Research, National Hospital Organization Yamaguchi Ube Medical Center, Ube, Japan
| | - Pauline M Rudd
- NIBRT GlycoScience Group, National Institute for Bioprocessing Research and Training, Mount Merrion, Blackrock, Dublin, Ireland
- Bioprocessing Technology Institute, Agency for Science, Technology and Research, Centros, Singapore
| | - Roy Jefferis
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
35
|
Enhancing antibody-dependent cellular phagocytosis by Re-education of tumor-associated macrophages with resiquimod-encapsulated liposomes. Biomaterials 2020; 268:120601. [PMID: 33338932 DOI: 10.1016/j.biomaterials.2020.120601] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 11/13/2020] [Accepted: 12/08/2020] [Indexed: 02/08/2023]
Abstract
Tumor-associated macrophages (TAMs) exist in nearly all tumors, and form a major part of the tumor microenvironment. TAMs are divided into two groups: tumor-suppressing M1 type and tumor-promoting M2 type. Most TAMs are educated by the tumor cells to become M2 type, which support tumor growth and make immunotherapy ineffective. Antibody-dependent cellular phagocytosis (ADCP) is an important mechanism for antibody cancer therapy, and this mechanism is dependent on TAMs. In this study, we found that the M1 type macrophages elicit a more efficient ADCP response than the M2 type, which was confirmed by three tumor cell lines, Raji, A431, and SKBR3, along with their corresponding therapeutic antibody Rituximab, anti-EGFR mouse monoclonal antibody (clone 528), and Trastuzumab, respectively. Resiquimod (R848), an immune system activating agent, has been shown to stimulate the M1 type macrophages, and re-educate the TAMs from M2 type to M1 type. By treating TAMs with R848, the ADCP response increased significantly in vitro and in in vivo mouse xenograft models. R848 encapsulated liposomes (R848-LPs) not only accumulated efficiently in the tumor tissues, but also distributed in the TAMs. Synergizing the R848-LPs with the anti-EGFR mouse monoclonal antibody (clone 528) significantly inhibited WiDr-tumor growth in vivo. Our study also revealed that the TAM-targeted delivery of R848 is able to re-educate the TAMs to M1 type, enhance the ADCP effect of the antibodies, and hence, enhance the anti-tumor effect of the therapeutic antibodies.
Collapse
|
36
|
Liu R, Oldham RJ, Teal E, Beers SA, Cragg MS. Fc-Engineering for Modulated Effector Functions-Improving Antibodies for Cancer Treatment. Antibodies (Basel) 2020; 9:E64. [PMID: 33212886 PMCID: PMC7709126 DOI: 10.3390/antib9040064] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 09/28/2020] [Accepted: 11/04/2020] [Indexed: 12/30/2022] Open
Abstract
The majority of monoclonal antibody (mAb) therapeutics possess the ability to engage innate immune effectors through interactions mediated by their fragment crystallizable (Fc) domain. By delivering Fc-Fc gamma receptor (FcγR) and Fc-C1q interactions, mAb are able to link exquisite specificity to powerful cellular and complement-mediated effector functions. Fc interactions can also facilitate enhanced target clustering to evoke potent receptor signaling. These observations have driven decades-long research to delineate the properties within the Fc that elicit these various activities, identifying key amino acid residues and elucidating the important role of glycosylation. They have also fostered a growing interest in Fc-engineering whereby this knowledge is exploited to modulate Fc effector function to suit specific mechanisms of action and therapeutic purposes. In this review, we document the insight that has been generated through the study of the Fc domain; revealing the underpinning structure-function relationships and how the Fc has been engineered to produce an increasing number of antibodies that are appearing in the clinic with augmented abilities to treat cancer.
Collapse
Affiliation(s)
- Rena Liu
- GlaxoSmithKline Research and Development, Stevenage SG1 2NY, UK;
| | - Robert J. Oldham
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO171BJ, UK; (R.J.O.); (E.T.); (M.S.C.)
- Cancer Research UK Centre, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO171BJ, UK
| | - Emma Teal
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO171BJ, UK; (R.J.O.); (E.T.); (M.S.C.)
- Cancer Research UK Centre, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO171BJ, UK
| | - Stephen A. Beers
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO171BJ, UK; (R.J.O.); (E.T.); (M.S.C.)
- Cancer Research UK Centre, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO171BJ, UK
| | - Mark S. Cragg
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO171BJ, UK; (R.J.O.); (E.T.); (M.S.C.)
- Cancer Research UK Centre, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO171BJ, UK
| |
Collapse
|
37
|
Klein C, Jamois C, Nielsen T. Anti-CD20 treatment for B-cell malignancies: current status and future directions. Expert Opin Biol Ther 2020; 21:161-181. [PMID: 32933335 DOI: 10.1080/14712598.2020.1822318] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
INTRODUCTION The introduction of anti-CD20 monoclonal antibody therapy with rituximab in the 1990s greatly improved outcomes for patients with B-cell malignancies. Disease resistance or relapse after successful initial therapy and declining efficacy of subsequent rounds of treatment were the basis for the development of alternative anti-CD20-based antibody therapies. AREAS COVERED The novel anti-CD20 antibodies of atumumab, ublituximab, and obinutuzumab were developed to be differentiated via structural and mechanistic features over rituximab. We provide an overview of preclinical and clinical data, and demonstrate ways in which the pharmacodynamic properties of these novel agents translate into clinical benefit for patients. EXPERT OPINION Of the novel anti-CD20 antibodies, only obinutuzumab has shown consistently improved efficacy over rituximab in randomized pivotal trials in indolent non-Hodgkin lymphoma and chronic lymphocytic leukemia. The Phase 3 GALLIUM trial demonstrated significant improvements in progression-free survival with obinutuzumab-based immunochemotherapy over rituximab-based immunochemotherapy. Novel combinations of obinutuzumab, including with chemotherapy-free options are being explored, such as with the newly approved combinations of obinutuzumab with venetoclax, ibrutinib, or acalabrutinib. The biggest unmet need remains in the treatment of diffuse large B-cell lymphoma; emerging options in this field include the use of CAR-T cells and T-cell bispecific antibodies.
Collapse
Affiliation(s)
- Christian Klein
- Roche Pharma Research & Early Development, Roche Innovation Center Zurich , Schlieren, Switzerland
| | - Candice Jamois
- Clinical Pharmacology, Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Center Basel , Basel, Switzerland
| | - Tina Nielsen
- Product Development Oncology, F. Hoffmann-La Roche Ltd , Basel, Switzerland
| |
Collapse
|
38
|
van der Horst HJ, Nijhof IS, Mutis T, Chamuleau MED. Fc-Engineered Antibodies with Enhanced Fc-Effector Function for the Treatment of B-Cell Malignancies. Cancers (Basel) 2020; 12:E3041. [PMID: 33086644 PMCID: PMC7603375 DOI: 10.3390/cancers12103041] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/12/2020] [Accepted: 10/14/2020] [Indexed: 11/17/2022] Open
Abstract
Monoclonal antibody (mAb) therapy has rapidly changed the field of cancer therapy. In 1997, the CD20-targeting mAb rituximab was the first mAb to be approved by the U.S. Food and Drug Administration (FDA) for treatment of cancer. Within two decades, dozens of mAbs entered the clinic for treatment of several hematological cancers and solid tumors, and numerous more are under clinical investigation. The success of mAbs as cancer therapeutics lies in their ability to induce various cytotoxic machineries against specific targets. These cytotoxic machineries include antibody-dependent cellular cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP), and complement-dependent cytotoxicity (CDC), which are all mediated via the fragment crystallizable (Fc) domain of mAbs. In this review article, we will outline the novel approaches of engineering these Fc domains of mAbs to enhance their Fc-effector function and thereby their anti-tumor potency, with specific focus to summarize their (pre-) clinical status for the treatment of B-cell malignancies, including chronic lymphocytic leukemia (CLL), B-cell non-Hodgkin lymphoma (B-NHL), and multiple myeloma (MM).
Collapse
Affiliation(s)
- Hilma J. van der Horst
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, VU Medical Center, 1081 HV Amsterdam, The Netherlands; (I.S.N.); (T.M.); (M.E.D.C.)
| | | | | | | |
Collapse
|
39
|
Paul A, Frenkel-Pinter M, Escobar Alvarez D, Milordini G, Gazit E, Zacco E, Segal D. Tryptophan-galactosylamine conjugates inhibit and disaggregate amyloid fibrils of Aβ42 and hIAPP peptides while reducing their toxicity. Commun Biol 2020; 3:484. [PMID: 32879439 PMCID: PMC7468108 DOI: 10.1038/s42003-020-01216-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 07/31/2020] [Indexed: 12/14/2022] Open
Abstract
Self-assembly of proteins into amyloid fibrils is a hallmark of various diseases, including Alzheimer's disease (AD) and Type-2 diabetes Mellitus (T2DM). Aggregation of specific peptides, like Aβ42 in AD and hIAPP in T2DM, causes cellular dysfunction resulting in the respective pathology. While these amyloidogenic proteins lack sequence homology, they all contain aromatic amino acids in their hydrophobic core that play a major role in their self-assembly. Targeting these aromatic residues by small molecules may be an attractive approach for inhibiting amyloid aggregation. Here, various biochemical and biophysical techniques revealed that a panel of tryptophan-galactosylamine conjugates significantly inhibit fibril formation of Aβ42 and hIAPP, and disassemble their pre-formed fibrils in a dose-dependent manner. They are also not toxic to mammalian cells and can reduce the cytotoxicity induced by Aβ42 and hIAPP aggregates. These tryptophan-galactosylamine conjugates can therefore serve as a scaffold for the development of therapeutics towards AD and T2DM.
Collapse
Affiliation(s)
- Ashim Paul
- Department of Molecular Microbiology and Biotechnology, School of Molecular Cell Biology and Biotechnology, Tel Aviv University, Ramat Aviv, Tel Aviv, 6997801, Israel
| | - Moran Frenkel-Pinter
- Department of Molecular Microbiology and Biotechnology, School of Molecular Cell Biology and Biotechnology, Tel Aviv University, Ramat Aviv, Tel Aviv, 6997801, Israel
| | - Daniela Escobar Alvarez
- Department of Molecular Microbiology and Biotechnology, School of Molecular Cell Biology and Biotechnology, Tel Aviv University, Ramat Aviv, Tel Aviv, 6997801, Israel
| | - Giulia Milordini
- The Maurice Wohl Clinical Neuroscience Institute, King's College London, Brixton, London, SE5 9RT, UK
| | - Ehud Gazit
- Department of Molecular Microbiology and Biotechnology, School of Molecular Cell Biology and Biotechnology, Tel Aviv University, Ramat Aviv, Tel Aviv, 6997801, Israel
| | - Elsa Zacco
- The Maurice Wohl Clinical Neuroscience Institute, King's College London, Brixton, London, SE5 9RT, UK.
- RNA Central Lab, Center for Human Technologies, Istituto Italiano di Tecnologia, 16152, Genova, Italy.
| | - Daniel Segal
- Department of Molecular Microbiology and Biotechnology, School of Molecular Cell Biology and Biotechnology, Tel Aviv University, Ramat Aviv, Tel Aviv, 6997801, Israel.
- Sagol Interdisciplinary School of Neuroscience, Tel Aviv University, Ramat Aviv, Tel Aviv, 6997801, Israel.
| |
Collapse
|
40
|
Chu Y, Awasthi A, Lee S, Edani D, Yin C, Hochberg J, Shah T, Chung TH, Ayello J, van de Ven C, Klein C, Lee D, Cairo MS. Obinutuzumab (GA101) vs. rituximab significantly enhances cell death, antibody-dependent cytotoxicity and improves overall survival against CD20+ primary mediastinal B-cell lymphoma (PMBL) in a xenograft NOD-scid IL2Rgnull (NSG) mouse model: a potential targeted agent in the treatment of PMBL. Oncotarget 2020; 11:3035-3047. [PMID: 32850008 PMCID: PMC7429176 DOI: 10.18632/oncotarget.27691] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 07/14/2020] [Indexed: 12/21/2022] Open
Abstract
Primary mediastinal large B-cell lymphoma (PMBL), a distinct mature B-cell lymphoma, expresses CD20 and has recently been successfully treated with the combination of a type I anti-CD20 monoclonal antibody, rituximab, with multiple combination chemotherapy regimens. Obinutuzumab is a glycoengineered type II anti-CD20 monoclonal antibody (mAb), recognizing a unique CD20 extracellular membrane epitope with enhanced antibody dependent cellular cytotoxicity (ADCC) vs rituximab. We hypothesize that obinutuzumab vs rituximab will significantly enhance in-vitro and in-vivo cytotoxicity against PMBL. PMBL cells were treated with equal dose of obinutuzumab and rituximab for 24 hours (1–100 μg/ml). ADCC were performed with ex-vivo expanded natural killer cells at 10:1 E: T ratio. Mice were xenografted with intravenous injections of luciferase expressing Karpas1106P cells and treated every 7 days for 8 weeks. Tumor burden was monitored by IVIS spectrum system. Compared with rituximab, obinutuzumab significantly inhibited PMBL cell proliferation (p = 0.01), promoted apoptosis (p = 0.05) and enhanced ADCC (p = 0.0002) against PMBL. Similarly, in PMBL xenografted NOD scid gamma mice, obinutuzumab significantly enhanced survival than rituximab when treated with equal doses (p = 0.05). Taken together our results suggest that obinutuzumab significantly enhanced natural killer cytotoxicity, reduced PMBL proliferation and prolonged the overall survival in humanized PMBL xenografted NOD scid gamma mice.
Collapse
Affiliation(s)
- Yaya Chu
- Department of Pediatrics, New York Medical College, Valhalla, NY, USA.,Co-first authors
| | - Aradhana Awasthi
- Department of Pediatrics, New York Medical College, Valhalla, NY, USA.,Co-first authors
| | - Sanghoon Lee
- Department of Pediatrics, New York Medical College, Valhalla, NY, USA.,Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY, USA
| | - Dina Edani
- Department of Pediatrics, New York Medical College, Valhalla, NY, USA
| | - Changhong Yin
- Department of Pediatrics, New York Medical College, Valhalla, NY, USA
| | - Jessica Hochberg
- Department of Pediatrics, New York Medical College, Valhalla, NY, USA
| | - Tishi Shah
- Department of Pediatrics, New York Medical College, Valhalla, NY, USA
| | - Tae-Hoon Chung
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Janet Ayello
- Department of Pediatrics, New York Medical College, Valhalla, NY, USA
| | | | - Christian Klein
- Roche Pharmaceutical Research & Early Development, Roche Innovation Center, Zurich, Switzerland
| | - Dean Lee
- Department of Pediatrics, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Mitchell S Cairo
- Department of Pediatrics, New York Medical College, Valhalla, NY, USA.,Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY, USA.,Department of Microbiology & Immunology, New York Medical College, Valhalla, NY, USA.,Department of Medicine, New York Medical College, Valhalla, NY, USA.,Department of Pathology, New York Medical College, Valhalla, NY, USA
| |
Collapse
|
41
|
Freitas Monteiro M, Papaserafeim M, Réal A, Puga Yung GL, Seebach JD. Anti-CD20 rituximab IgG1, IgG3, and IgG4 but not IgG2 subclass trigger Ca 2+ mobilization and cytotoxicity in human NK cells. J Leukoc Biol 2020; 108:1409-1423. [PMID: 32620047 DOI: 10.1002/jlb.5ma0620-039r] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 06/11/2020] [Accepted: 06/16/2020] [Indexed: 12/12/2022] Open
Abstract
NK cell-mediated Ab-dependent cellular cytotoxicity (ADCC) is increasingly recognized to play an important role in cancer immunotherapy, transplant rejection, and autoimmunity. However, several aspects of the molecular interactions of IgG subclasses with the Fc-gamma receptor IIIA (FcγRIIIA)/CD16a expressed on NK cells remain unknown. The aim of the current study was to further analyze the role of IgG subclasses and FCGR3A V158F single nucleotide polymorphism (SNP) on Ca2+ signaling and NK cell-mediated ADCC against Daudi target cells in vitro. NK cells were isolated from donors with different FCGR3A SNP. The affinity of rituximab IgG subclasses to CD20 expressed on Daudi cells showed similar dissociation constant as tested by flow cytometry. Induction of Ca2+ signaling, degranulation, intracellular cytokine production, and ADCC was demonstrated for IgG1 and IgG3, to a lesser degree also for IgG4, but not for IgG2. Compared to NK cells carrying the low-affinity (FF) variant for the FCGR3A V158F SNP, binding of IgG1 and IgG3 to NK cells carrying the high-affinity (VV) and VF SNP variants was two- to threefold higher. Variations of FCGR3A SNP among the eight tested donors (1 VV, 3FF, and 4VF) revealed no significant differences of Ca2+ signaling and degranulation; however, ADCC was somewhat weaker in donors with the low-affinity FF variation. In conclusion, this is the first study correlating Ca2+ signaling and NK cell-mediated ADCC triggered by the four IgG subclasses with the FCGR3A V158F SNP. Our findings indicate important differences in the interactions of IgG subclasses with FcγRIIIA/CD16a but no major impact of FCGR3A SNP and may therefore help to better correlate the functional properties of particular engineered therapeutic antibodies in vitro with individual differences of their clinical efficacy.
Collapse
Affiliation(s)
- Marta Freitas Monteiro
- Division of Immunology and Allergology, Department of Medicine, University Hospitals and Medical Faculty, Geneva, Switzerland
| | - Maria Papaserafeim
- Division of Immunology and Allergology, Department of Medicine, University Hospitals and Medical Faculty, Geneva, Switzerland
| | - Aline Réal
- Division of Immunology and Allergology, Department of Medicine, University Hospitals and Medical Faculty, Geneva, Switzerland
| | - Gisella L Puga Yung
- Division of Immunology and Allergology, Department of Medicine, University Hospitals and Medical Faculty, Geneva, Switzerland
| | - Jörg D Seebach
- Division of Immunology and Allergology, Department of Medicine, University Hospitals and Medical Faculty, Geneva, Switzerland
| |
Collapse
|
42
|
CyBorD-DARA is potent initial induction for MM and enhances ADCP: initial results of the 16-BCNI-001/CTRIAL-IE 16-02 study. Blood Adv 2020; 3:1815-1825. [PMID: 31201169 DOI: 10.1182/bloodadvances.2019000010] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 05/09/2019] [Indexed: 12/12/2022] Open
Abstract
Daratumumab (DARA) has shown impressive activity in combination with other agents for the treatment of multiple myeloma (MM). We conducted a phase 1b study to assess the safety and preliminary efficacy, as well as potential mechanisms of action, of DARA (16 mg/kg) in combination with a weekly schedule of subcutaneous bortezomib (1.3-1.5 mg/m2), cyclophosphamide (150-300 mg/m2), and dexamethasone (40 mg) (CyBorD DARA) as initial induction before autologous stem cell transplantation (ASCT). Eligible patients were ≤70 years of age with untreated MM requiring treatment and who lacked significant comorbidities. A total of 18 patients were enrolled. Their median age was 56 years (range, 32-66 years), and all patients had Eastern Cooperative Oncology Group performance status ≤1. The International Staging System stages were I, II, and III in 78%, 17%, and 6% of patients, respectively; 28% of patients had high-risk genetic features. There was no dose-limiting toxicity, and the incidence of grade 3 or 4 infection or neutropenia was <10%. On an intention-to-treat basis, 94% achieved ≥very good partial response with ≥complete response in 44% of patients. Among 14 of 15 patients who underwent ASCT and were evaluable for response, all 14 achieved at least very good partial response, with 8 (57%) of 14 achieving complete response. After ASCT, 10 (83%) of 12 patients in whom minimal residual disease analysis was possible were negative at a sensitivity of 10-5 (56% on intention-to-treat/whole study population) according to next-generation sequencing. Flow cytometry analysis of patient samples indicated CyBorD DARA induced activation of macrophage-mediated antibody-dependent cellular phagocytosis. This trial was registered at www.clinicaltrials.gov as #NCT02955810.
Collapse
|
43
|
Kim JO, Kim HN, Kim KH, Baek EJ, Park JY, Ha K, Heo DR, Seo MD, Park SG. Development and characterization of a fully human antibody targeting SCF/c-kit signaling. Int J Biol Macromol 2020; 159:66-78. [PMID: 32437800 DOI: 10.1016/j.ijbiomac.2020.05.045] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 05/06/2020] [Accepted: 05/06/2020] [Indexed: 12/23/2022]
Abstract
CD117/c-kit, a tyrosine kinase receptor, plays a critical role in hematopoiesis, pigmentation, and fertility. The overexpression and activation of c-kit are thought to promote tumor growth and have been reported in various cancers, including leukemia, glioblastoma and mastocytosis. To disrupt the SCF/c-kit signaling axis in cancer, we generated a c-kit antagonist human antibody (NN2101) that binds to domain 2/3 of c-kit. This completely blocked the SCF-mediated phosphorylation of c-kit and inhibited TF-1 cell proliferation, erythroleukemia. In addition, the examination of binding affinity using surface plasmon resonance (SPR) assay showed that NN2101 can bind to c-kit of monkeys (KD = 2.92 × 10-10 M), rats (KD = 1.68 × 10-6 M), mice (KD = 11.5 × 10-9 M), and humans (KD = 2.83 × 10-12 M). We showed that NN2101 does not cause antibody-dependent cell-mediated cytotoxicity and complement-dependent cytotoxicity. The immunogenicity of NN2101 was similar to that of bevacizumab. Furthermore, the crystal structure of NN2101 Fab was determined and the structure of NN2101 Fab:c-kit complex was modeled. Structural information, as well as mutagenesis results, revealed that NN2101 can bind to the SCF-binding regions of c-kit. Collectively, we generated a c-kit neutralizing human antibody (NN2101) for the treatment of erythroleukemia and characterized its biophysical properties. NN2101 can potentially be used as a therapeutic antibody to treat different cancers.
Collapse
Affiliation(s)
- Jin-Ock Kim
- College of Pharmacy and Research Institute of Pharmaceutical Science and Technology (RIPST), Ajou University, 206 World Cup-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do 16499, Republic of Korea
| | - Ha-Neul Kim
- College of Pharmacy and Research Institute of Pharmaceutical Science and Technology (RIPST), Ajou University, 206 World Cup-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do 16499, Republic of Korea; Department of Molecular Science and Technology, Ajou University, 206 World Cup-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do 16499, Republic of Korea
| | - Kwang-Hyeok Kim
- College of Pharmacy and Research Institute of Pharmaceutical Science and Technology (RIPST), Ajou University, 206 World Cup-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do 16499, Republic of Korea
| | - Eun Ji Baek
- College of Pharmacy and Research Institute of Pharmaceutical Science and Technology (RIPST), Ajou University, 206 World Cup-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do 16499, Republic of Korea
| | - Jeong-Yang Park
- College of Pharmacy and Research Institute of Pharmaceutical Science and Technology (RIPST), Ajou University, 206 World Cup-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do 16499, Republic of Korea; Department of Molecular Science and Technology, Ajou University, 206 World Cup-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do 16499, Republic of Korea
| | - Kyungsoo Ha
- New Drug Development Center, Osong Medical Innovation Foundation, Osong 28160, Republic of Korea
| | - Deok Rim Heo
- New Drug Development Center, Osong Medical Innovation Foundation, Osong 28160, Republic of Korea; College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong 28160, Republic of Korea
| | - Min-Duk Seo
- College of Pharmacy and Research Institute of Pharmaceutical Science and Technology (RIPST), Ajou University, 206 World Cup-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do 16499, Republic of Korea; Department of Molecular Science and Technology, Ajou University, 206 World Cup-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do 16499, Republic of Korea; Novelty Nobility, 227 Unjung-ro, Seongnam-si, Gyeonggi-do 13477, Republic of Korea.
| | - Sang Gyu Park
- College of Pharmacy and Research Institute of Pharmaceutical Science and Technology (RIPST), Ajou University, 206 World Cup-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do 16499, Republic of Korea; Novelty Nobility, 227 Unjung-ro, Seongnam-si, Gyeonggi-do 13477, Republic of Korea.
| |
Collapse
|
44
|
Inoue S, Horiuchi Y, Setoyama Y, Takeuchi Y, Beck Y, Murakami T, Odaka A. Immune Checkpoint Inhibition Followed by Tumor Infiltration of Dendritic Cells in Murine Neuro-2a Neuroblastoma. J Surg Res 2020; 253:201-213. [PMID: 32380346 DOI: 10.1016/j.jss.2020.03.059] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 03/04/2020] [Accepted: 03/26/2020] [Indexed: 12/31/2022]
Abstract
BACKGROUND Most tumors responding to immunotherapy with monoclonal antibodies targeting programmed cell death protein1 (PD1) and programmed death ligand-1 (PD-L1) show surface expression of PD-L1. Neuroblastoma has been reported to show low PD-L1 surface expression. METHODS The effect of immune checkpoint inhibitor on mouse neuroblastoma was investigated, and host immune cells were analyzed in the tumor microenvironment. Expression of co-stimulatory molecules by Neuro-2a mouse neuroblastoma cells was analyzed using flow cytometer. Neuro-2a cells were inoculated subcutaneously into A/J mice, followed by intraperitoneal injection of antibodies targeting PD-1 and PD-L1. Mice were sacrificed for the measurement of tumor weights on day 14 following tumor inoculation, and tumor-infiltrating cells were analyzed using a flow cytometer. RESULTS Dim expression of PD-L1 was observed on the cell surface of cultured Neuro-2a cells. Growth of subcutaneous tumors was significantly suppressed, and PD-L1-expressing tumor cells were depleted by the antibody treatment. We confirmed that Neuro-2a cells opsonized by the anti-PD-L1 antibody were phagocytosed in the in vitro setting. In the treated tumor microenvironments, CD8α+ lymphocyte and CD11c+ MHC II+ cells were significantly accumulated in comparison with the control group. These CD11c+ MHC II+ cells expressed CD80, CD86, CD14, and CD40, but not CD205, PD-L1, or CTLA4. PD-1 expression was detected dimly. Immune suppressive effects of CD11b+Gr-1+ myeloid-derived suppressor cells by the administration of anti-PD-1 and PD-L1 antibodies were not observed in spleen, regional lymph nodes, or tumor microenvironment. CONCLUSIONS Our findings raise the possibility that co-administration of anti-PD-1 and anti-PD-L1 antibodies have a synergistic effect on inhibition of tumor growth and could be an effective therapy against neuroblastoma with dim expression of PD-L1.
Collapse
Affiliation(s)
- Seiichiro Inoue
- Department of Hepato-Biliary-Pancreatic and Pediatric Surgery, Saitama Medical Center, Saitama Medical University, Kawagoe, Saitama, Japan.
| | - Yutaka Horiuchi
- Department of Microbiology, Faculty of Medicine, Saitama Medical University, Iruma-gun, Saitama, Japan
| | - Yumiko Setoyama
- Department of Biomedical Sciences, Saitama Medical Center, Saitama Medical University, Kawagoe, Saitama, Japan
| | - Yuta Takeuchi
- Department of Hepato-Biliary-Pancreatic and Pediatric Surgery, Saitama Medical Center, Saitama Medical University, Kawagoe, Saitama, Japan
| | - Yoshifumi Beck
- Department of Hepato-Biliary-Pancreatic and Pediatric Surgery, Saitama Medical Center, Saitama Medical University, Kawagoe, Saitama, Japan
| | - Takashi Murakami
- Department of Microbiology, Faculty of Medicine, Saitama Medical University, Iruma-gun, Saitama, Japan
| | - Akio Odaka
- Department of Hepato-Biliary-Pancreatic and Pediatric Surgery, Saitama Medical Center, Saitama Medical University, Kawagoe, Saitama, Japan
| |
Collapse
|
45
|
Disis ML, Corulli LR, Gad EA, Koehnlein MR, Cecil DL, Senter PD, Gardai SJ, Okeley NM. Therapeutic and Prophylactic Antitumor Activity of an Oral Inhibitor of Fucosylation in Spontaneous Mammary Cancers. Mol Cancer Ther 2020; 19:1102-1109. [PMID: 32165557 DOI: 10.1158/1535-7163.mct-19-0500] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 09/20/2019] [Accepted: 02/28/2020] [Indexed: 11/16/2022]
Abstract
2-fluorofucose (2FF) inhibits protein and cellular fucosylation. Afucosylation of IgG antibodies enhances antibody-dependent cell-mediated cytotoxicity by modulating antibody affinity for FcγRIIIa, which can impact secondary T-cell activation. Immune responses toward most common solid tumors are dominated by a humoral immune response rather than the presence of tumor-infiltrating cytotoxic T cells. IgG antibodies directed against numerous tumor-associated proteins are found in the sera of both patients with breast cancer and transgenic mice bearing mammary cancer. We questioned whether 2FF would have antitumor activity in two genetically distinct transgenic models; TgMMTV-neu (luminal B) and C3(1)-Tag (basal) mammary cancer. 2FF treatment significantly improved overall survival. The TgMMTV-neu doubled survival time compared with controls [P < 0.0001; HR, 7.04; 95% confidence interval (CI), 3.31-15.0], and survival was significantly improved in C3(1)-Tag (P = 0.0013; HR, 3.36; 95% CI, 1.58-7.14). 2FF treated mice, not controls, developed delayed-type hypersensitivity and T-cell responses specific for syngeneic tumor lysates (P < 0.0001). Serum IgG from 2FF-treated mice enhanced tumor lysis more efficiently than control sera (P = 0.004). Administration of 2FF for prophylaxis, at two different doses, significantly delayed tumor onset in both TgMMTV-neu; 20 mmol/L (P = 0.0004; HR, 3.55; 95% CI, 1.60-7.88) and 50 mmol/L (P = 0.0002; HR: 3.89; 95% CI, 1.71-8.86) and C3(1)-Tag; 20 mmol/L (P = 0.0020; HR, 2.51; 95% CI, 1.22-5.18), and 50 mmol/L (P = 0.0012; HR, 3.36; 95% CI, 1.57-7.18). Mammary cancer was prevented in 33% of TgMMTV-neu and 26% of C3(1)-Tag. 2FF has potent antitumor effects in mammary cancer models. The agent shows preclinical efficacy for both cancer treatment and prevention.
Collapse
Affiliation(s)
- Mary L Disis
- UW Medicine Cancer Vaccine Institute, University of Washington, Seattle, Washington.
| | - Lauren R Corulli
- UW Medicine Cancer Vaccine Institute, University of Washington, Seattle, Washington
| | - Ekram A Gad
- UW Medicine Cancer Vaccine Institute, University of Washington, Seattle, Washington
| | - Marlese R Koehnlein
- UW Medicine Cancer Vaccine Institute, University of Washington, Seattle, Washington
| | - Denise L Cecil
- UW Medicine Cancer Vaccine Institute, University of Washington, Seattle, Washington
| | | | | | | |
Collapse
|
46
|
Roberts JT, Patel KR, Barb AW. Site-specific N-glycan Analysis of Antibody-binding Fc γ Receptors from Primary Human Monocytes. Mol Cell Proteomics 2020; 19:362-374. [PMID: 31888963 PMCID: PMC7000114 DOI: 10.1074/mcp.ra119.001733] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 12/10/2019] [Indexed: 12/29/2022] Open
Abstract
FcγRIIIa (CD16a) and FcγRIIa (CD32a) on monocytes are essential for proper effector functions including antibody dependent cellular cytotoxicity (ADCC) and phagocytosis (ADCP). Indeed, therapeutic monoclonal antibodies (mAbs) that bind FcγRs with greater affinity exhibit greater efficacy. Furthermore, post-translational modification impacts antibody binding affinity, most notably the composition of the asparagine(N)-linked glycan at N162 of CD16a. CD16a is widely recognized as the key receptor for the monocyte response, however the post-translational modifications of CD16a from endogenous monocytes are not described. Here we isolated monocytes from individual donors and characterized the composition of CD16a and CD32a N-glycans from all modified sites. The composition of CD16a N-glycans varied by glycosylation site and donor. CD16a displayed primarily complex-type biantennary N-glycans at N162, however some individuals expressed CD16a V158 with ∼20% hybrid and oligomannose types which increased affinity for IgG1 Fc according to surface plasmon resonance binding analyses. The CD16a N45-glycans contain markedly less processing than other sites with >75% hybrid and oligomannose forms. N38 and N74 of CD16a both contain highly processed complex-type N-glycans with N-acetyllactosamine repeats and complex-type biantennary N-glycans dominate at N169. The composition of CD16a N-glycans isolated from monocytes included a higher proportion of oligomannose-type N-glycans at N45 and less sialylation plus greater branch fucosylation than we observed in a recent analysis of NK cell CD16a. The additional analysis of CD32a from monocytes revealed different features than observed for CD16a including the presence of a predominantly biantennary complex-type N-glycans with two sialic acids at both sites (N64 and N145).
Collapse
Affiliation(s)
- Jacob T Roberts
- Roy J. Carver Department of Biochemistry, Biophysics & Molecular Biology, Iowa State University, Ames Iowa 50011
| | - Kashyap R Patel
- Roy J. Carver Department of Biochemistry, Biophysics & Molecular Biology, Iowa State University, Ames Iowa 50011
| | - Adam W Barb
- Roy J. Carver Department of Biochemistry, Biophysics & Molecular Biology, Iowa State University, Ames Iowa 50011
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia 30602
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia 30602
| |
Collapse
|
47
|
Mechanisms of Resistance to Anti-CD38 Daratumumab in Multiple Myeloma. Cells 2020; 9:cells9010167. [PMID: 31936617 PMCID: PMC7017193 DOI: 10.3390/cells9010167] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 12/31/2019] [Accepted: 01/03/2020] [Indexed: 02/06/2023] Open
Abstract
Daratumumab (Dara) is the first-in-class human-specific anti-CD38 mAb approved for the treatment of multiple myeloma (MM). Although recent data have demonstrated very promising results in clinical practice and trials, some patients do not achieve a partial response, and ultimately all patients undergo progression. Dara exerts anti-MM activity via antibody-dependent cell-mediated cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP), complement-dependent cytotoxicity (CDC), and immunomodulatory effects. Deregulation of these pleiotropic mechanisms may cause development of Dara resistance. Knowledge of this resistance may improve the therapeutic management of MM patients.
Collapse
|
48
|
Flow cytometry-based assessment of direct-targeting anti-cancer antibody immune effector functions. Methods Enzymol 2020; 632:431-456. [PMID: 32000909 PMCID: PMC7000137 DOI: 10.1016/bs.mie.2019.07.026] [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] [Indexed: 02/04/2023]
Abstract
Monoclonal antibody-based therapies are increasingly being used to treat cancer. Some mediate their therapeutic effects through modifying the function of immune cells globally, while others bind directly to tumor cells and can recruit immune effector cells through their Fc regions. As new direct-binding agents are developed, having the ability to test their Fc-mediated functions in a high-throughput manner is important for selecting antibodies with immune effector properties. Here, using monoclonal anti-CD20 antibody (rituximab) as an example and the CD20+ Raji cell line as tumor target, we describe flow cytometry-based assays for determining an antibody's capacity for mediating antibody-dependent cellular cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP) and complement-dependent cytotoxicity (CDC). These assays are sensitive, reliable, affordable and avoid the use of radioactivity.
Collapse
|
49
|
Luan C, Chen B. Clinical application of obinutuzumab for treating chronic lymphocytic leukemia. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:2899-2909. [PMID: 31692500 PMCID: PMC6707935 DOI: 10.2147/dddt.s212500] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 07/14/2019] [Indexed: 12/02/2022]
Abstract
Alkylators and nucleoside analogs were the main drugs for treatingchronic lymphoblastic leukemia (CLL), which have been replaced by monoclonal antibodies, such as rituximab in the past 10 years for refractory or relapsed CLL. The first-line immunochemotherapy regimen, rituximab combined with nucleoside analogs, significantly increased CLL patients’ first-reaction rate and improved progression-free survival. Despite the long-lasting remissions by the use of chemoimmunotherapy, most CLL patients will relapse eventually. The obinutuzumab (GA101), an updated CD20 antibody, that is thought to achieve a more durable response with unique molecular and functional characteristics. Obinutuzumab is a humanized, monoclonal type II CD20 antibody modified by glycoengineering. The glycoengineered Fc portion enhances the binding affinity to the FcγRIII receptor on immune effector cells, resulting in increased antibody-dependent cellular cytotoxicity and phagocytosis. In addition, the type II antibody binding characteristics of obinutuzumab to CD20 lead to an efficient induction of direct non-apoptotic cell death. This review summarizes the results of clinical studies using obinutuzumab and looks forward to its further application in treating CLL clinically.
Collapse
Affiliation(s)
- Chunyan Luan
- Department of Hematology and Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu Province 210009, People's Republic of China
| | - Baoan Chen
- Department of Hematology and Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu Province 210009, People's Republic of China
| |
Collapse
|
50
|
NK Cells in the Treatment of Hematological Malignancies. J Clin Med 2019; 8:jcm8101557. [PMID: 31569769 PMCID: PMC6832953 DOI: 10.3390/jcm8101557] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 09/18/2019] [Accepted: 09/25/2019] [Indexed: 02/06/2023] Open
Abstract
Natural killer (NK) cells have the innate ability to kill cancer cells, however, tumor cells may acquire the capability of evading the immune response, thereby leading to malignancies. Restoring or potentiation of this natural antitumor activity of NK cells has become a relevant therapeutic approach in cancer and, particularly, in hematological cancers. The use of tumor-specific antibodies that promote antibody-dependent cell-mediated cytotoxicity (ADCC) through the ligation of CD16 receptor on NK cells has become standard for many hematologic malignancies. Hematopoietic stem cell transplantation is another key therapeutic strategy that harnesses the alloreactivity of NK cells against cancer cells. This strategy may be refined by adoptive transfer of NK cells that may be previously expanded, activated, or redirected (chimeric antigen receptor (CAR)-NK cells) against cancer cells. The antitumor activity of NK cells can also be boosted by cytokines or immunostimulatory drugs such as lenalidomide or pomalidomide. Finally, targeting immunosubversive mechanisms developed by hematological cancers and, in particular, using antibodies that block NK cell inhibitory receptors and checkpoint proteins are novel promising therapeutic approaches in these malignant diseases.
Collapse
|