1
|
Zhang HJ, Zhu L, Xie QH, Zhang LZ, Liu JY, Feng YYF, Chen ZK, Xia HF, Fu QY, Yu ZL, Chen G. Extracellular vesicle-packaged PD-L1 impedes macrophage-mediated antibacterial immunity in preexisting malignancy. Cell Rep 2024; 43:114903. [PMID: 39489940 DOI: 10.1016/j.celrep.2024.114903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 09/08/2024] [Accepted: 10/08/2024] [Indexed: 11/05/2024] Open
Abstract
Malignancies can compromise systemic innate immunity, but the underlying mechanisms are largely unknown. Here, we find that tumor-derived small extracellular vesicles (sEVs; TEVs) deliver PD-L1 to host macrophages, thereby impeding antibacterial immunity. Mice implanted with Rab27a-knockdown tumors are more resistant to bacterial infection than wild-type controls. Injection of TEVs into mice impairs macrophage-mediated bacterial clearance, increases systemic bacterial dissemination, and enhances sepsis score in a PD-L1-dependent manner. Mechanistically, TEV-packaged PD-L1 inhibits Bruton's tyrosine kinase/PLCγ2 signaling-mediated cytoskeleton reorganization and reactive oxygen species generation, impacting bacterial phagocytosis and killing by macrophages. Neutralizing PD-L1 markedly normalizes macrophage-mediated bacterial clearance in tumor-bearing mice. Importantly, circulating sEV PD-L1 levels in patients with tumors can predict bacterial infection susceptibility, while patients with tumors treated with αPD-1 exhibit fewer postoperative infections. These findings identify a mechanism by which cancer cells dampen host innate immunity-mediated bacterial clearance and suggest targeting TEV-packaged PD-L1 to reduce bacterial infection susceptibility in tumor-bearing conditions.
Collapse
Affiliation(s)
- He-Jing Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Lingxin Zhu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Qi-Hui Xie
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Lin-Zhou Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Jin-Yuan Liu
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yang-Ying-Fan Feng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Zhuo-Kun Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Hou-Fu Xia
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Qiu-Yun Fu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Zi-Li Yu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Gang Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China; TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, China; Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430071, China.
| |
Collapse
|
2
|
Dooling LJ, Anlaş AA, Tobin MP, Ontko NM, Marchena T, Wang M, Andrechak JC, Discher DE. Clustered macrophages cooperate to eliminate tumors via coordinated intrudopodia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.19.613918. [PMID: 39345601 PMCID: PMC11430028 DOI: 10.1101/2024.09.19.613918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Macrophages often pervade solid tumors, but their nearest neighbor organization is understudied and potentially enables key functions such as phagocytosis. Here, we observe dynamic macrophage clusters in tumors under conditions that maximize cancer cell phagocytosis and use reductionist approaches to uncover pathways to cluster formation and roles for tumor-intrusive pseudopodia, which we term 'intrudopodia'. Macrophage clusters form over hours on low- adhesion substrates after M1 polarization with interferons, including T cell-derived cytokines, and yet clusters prove fluid on timescales of minutes. Clusters also sort from M2 macrophages that disperse on the same substrates. M1 macrophages upregulate specific cell-cell adhesion receptors but suppress actomyosin contractility, and while both pathways contribute to cluster formation, decreased cortical tension was predicted to unleash pseudopodia. Macrophage neighbors in tumor spheroids indeed extend intrudopodia between adjacent cancer cell junctions - at least when phagocytosis conditions are maximized, and coordinated intrudopodia help detach and individualize cancer cells for rapid engulfment. Macrophage clusters thereby provide a cooperative advantage for phagocytosis to overcome solid tumor cohesion.
Collapse
|
3
|
Frederiksen HR, Glantz A, Vøls KK, Skov S, Tveden-Nyborg P, Freude K, Doehn U. CRISPR-Cas9 immune-evasive hESCs are rejected following transplantation into immunocompetent mice. Front Genome Ed 2024; 6:1403395. [PMID: 38863835 PMCID: PMC11165197 DOI: 10.3389/fgeed.2024.1403395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 05/07/2024] [Indexed: 06/13/2024] Open
Abstract
Although current stem cell therapies exhibit promising potential, the extended process of employing autologous cells and the necessity for donor-host matching to avert the rejection of transplanted cells significantly limit the widespread applicability of these treatments. It would be highly advantageous to generate a pluripotent universal donor stem cell line that is immune-evasive and, therefore, not restricted by the individual's immune system, enabling unlimited application within cell replacement therapies. Before such immune-evasive stem cells can be moved forward to clinical trials, in vivo testing via transplantation experiments in immune-competent animals would be a favorable approach preceding preclinical testing. By using human stem cells in immune competent animals, results will be more translatable to a clinical setting, as no parts of the immune system have been altered, although in a xenogeneic setting. In this way, immune evasiveness, cell survival, and unwanted proliferative effects can be assessed before clinical trials in humans. The current study presents the generation and characterization of three human embryonic stem cell lines (hESCs) for xenogeneic transplantation in immune-competent mice. The major histocompatibility complexes I- and II-encoding genes, B2M and CIITA, have been deleted from the hESCs using CRISPR-Cas9-targeted gene replacement strategies and knockout. B2M was knocked out by the insertion of murine CD47. Human-secreted embryonic alkaline phosphatase (hSEAP) was inserted in a safe harbor site to track cells in vivo. The edited hESCs maintained their pluripotency, karyotypic normality, and stable expression of murine CD47 and hSEAP in vitro. In vivo transplantation of hESCs into immune-competent BALB/c mice was successfully monitored by measuring hSEAP in blood samples. Nevertheless, transplantation of immune-evasive hESCs resulted in complete rejection within 11 days, with clear immune infiltration of T-cells on day 8. Our results reveal that knockout of B2M and CIITA together with species-specific expression of CD47 are insufficient to prevent rejection in an immune-competent and xenogeneic context.
Collapse
Affiliation(s)
- Henriette Reventlow Frederiksen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | - Søren Skov
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Pernille Tveden-Nyborg
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kristine Freude
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ulrik Doehn
- Cell Therapy Research, Novo Nordisk A/S, Maaloev, Denmark
| |
Collapse
|
4
|
Hayes BH, Wang M, Zhu H, Phan SH, Dooling LJ, Andrechak JC, Chang AH, Tobin MP, Ontko NM, Marchena T, Discher DE. Chromosomal instability induced in cancer can enhance macrophage-initiated immune responses that include anti-tumor IgG. eLife 2024; 12:RP88054. [PMID: 38805560 PMCID: PMC11132682 DOI: 10.7554/elife.88054] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2024] Open
Abstract
Solid tumors generally exhibit chromosome copy number variation, which is typically caused by chromosomal instability (CIN) in mitosis. The resulting aneuploidy can drive evolution and associates with poor prognosis in various cancer types as well as poor response to T-cell checkpoint blockade in melanoma. Macrophages and the SIRPα-CD47 checkpoint are understudied in such contexts. Here, CIN is induced in poorly immunogenic B16F10 mouse melanoma cells using spindle assembly checkpoint MPS1 inhibitors that generate persistent micronuclei and diverse aneuploidy while skewing macrophages toward a tumoricidal 'M1-like' phenotype based on markers and short-term anti-tumor studies. Mice bearing CIN-afflicted tumors with wild-type CD47 levels succumb similar to controls, but long-term survival is maximized by SIRPα blockade on adoptively transferred myeloid cells plus anti-tumor monoclonal IgG. Such cells are the initiating effector cells, and survivors make de novo anti-cancer IgG that not only promote phagocytosis of CD47-null cells but also suppress tumor growth. CIN does not affect the IgG response, but pairing CIN with maximal macrophage anti-cancer activity increases durable cures that possess a vaccination-like response against recurrence.
Collapse
Affiliation(s)
- Brandon H Hayes
- Physical Sciences Oncology Center at Penn, University of PennsylvaniaPhiladelphhiaUnited States
- Molecular and Cell Biophysics Lab, University of PennsylvaniaPhiladelphiaUnited States
- Bioengineering Graduate Group, University of PennsylvaniaPhiladelphiaUnited States
| | - Mai Wang
- Physical Sciences Oncology Center at Penn, University of PennsylvaniaPhiladelphhiaUnited States
- Molecular and Cell Biophysics Lab, University of PennsylvaniaPhiladelphiaUnited States
| | - Hui Zhu
- Physical Sciences Oncology Center at Penn, University of PennsylvaniaPhiladelphhiaUnited States
- Molecular and Cell Biophysics Lab, University of PennsylvaniaPhiladelphiaUnited States
| | - Steven H Phan
- Physical Sciences Oncology Center at Penn, University of PennsylvaniaPhiladelphhiaUnited States
- Molecular and Cell Biophysics Lab, University of PennsylvaniaPhiladelphiaUnited States
| | - Lawrence J Dooling
- Physical Sciences Oncology Center at Penn, University of PennsylvaniaPhiladelphhiaUnited States
- Molecular and Cell Biophysics Lab, University of PennsylvaniaPhiladelphiaUnited States
| | - Jason C Andrechak
- Physical Sciences Oncology Center at Penn, University of PennsylvaniaPhiladelphhiaUnited States
- Molecular and Cell Biophysics Lab, University of PennsylvaniaPhiladelphiaUnited States
- Bioengineering Graduate Group, University of PennsylvaniaPhiladelphiaUnited States
| | - Alexander H Chang
- Physical Sciences Oncology Center at Penn, University of PennsylvaniaPhiladelphhiaUnited States
- Molecular and Cell Biophysics Lab, University of PennsylvaniaPhiladelphiaUnited States
| | - Michael P Tobin
- Physical Sciences Oncology Center at Penn, University of PennsylvaniaPhiladelphhiaUnited States
- Molecular and Cell Biophysics Lab, University of PennsylvaniaPhiladelphiaUnited States
- Bioengineering Graduate Group, University of PennsylvaniaPhiladelphiaUnited States
| | - Nicholas M Ontko
- Physical Sciences Oncology Center at Penn, University of PennsylvaniaPhiladelphhiaUnited States
- Molecular and Cell Biophysics Lab, University of PennsylvaniaPhiladelphiaUnited States
| | - Tristan Marchena
- Physical Sciences Oncology Center at Penn, University of PennsylvaniaPhiladelphhiaUnited States
- Molecular and Cell Biophysics Lab, University of PennsylvaniaPhiladelphiaUnited States
| | - Dennis E Discher
- Physical Sciences Oncology Center at Penn, University of PennsylvaniaPhiladelphhiaUnited States
- Molecular and Cell Biophysics Lab, University of PennsylvaniaPhiladelphiaUnited States
- Bioengineering Graduate Group, University of PennsylvaniaPhiladelphiaUnited States
| |
Collapse
|
5
|
Grandclément C, Estoppey C, Dheilly E, Panagopoulou M, Monney T, Dreyfus C, Loyau J, Labanca V, Drake A, De Angelis S, Rubod A, Frei J, Caro LN, Blein S, Martini E, Chimen M, Matthes T, Kaya Z, Edwards CM, Edwards JR, Menoret E, Kervoelen C, Pellat-Deceunynck C, Moreau P, Mbow ML, Srivastava A, Dyson MR, Zhukovsky EA, Perro M, Sammicheli S. Development of ISB 1442, a CD38 and CD47 bispecific biparatopic antibody innate cell modulator for the treatment of multiple myeloma. Nat Commun 2024; 15:2054. [PMID: 38448430 PMCID: PMC10917784 DOI: 10.1038/s41467-024-46310-y] [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: 07/31/2023] [Accepted: 02/21/2024] [Indexed: 03/08/2024] Open
Abstract
Antibody engineering can tailor the design and activities of therapeutic antibodies for better efficiency or other advantageous clinical properties. Here we report the development of ISB 1442, a fully human bispecific antibody designed to re-establish synthetic immunity in CD38+ hematological malignancies. ISB 1442 consists of two anti-CD38 arms targeting two distinct epitopes that preferentially drive binding to tumor cells and enable avidity-induced blocking of proximal CD47 receptors on the same cell while preventing on-target off-tumor binding on healthy cells. The Fc portion of ISB 1442 is engineered to enhance complement dependent cytotoxicity, antibody dependent cell cytotoxicity and antibody dependent cell phagocytosis. ISB 1442 thus represents a CD47-BsAb combining biparatopic targeting of a tumor associated antigen with engineered enhancement of antibody effector function to overcome potential resistance mechanisms that hamper treatment of myeloma with monospecific anti-CD38 antibodies. ISB 1442 is currently in a Phase I clinical trial in relapsed refractory multiple myeloma.
Collapse
Affiliation(s)
| | - C Estoppey
- Ichnos Glenmark Innovation, Lausanne, CH, Switzerland
| | - E Dheilly
- Ichnos Glenmark Innovation, Lausanne, CH, Switzerland
| | | | - T Monney
- Ichnos Glenmark Innovation, Lausanne, CH, Switzerland
| | - C Dreyfus
- Ichnos Glenmark Innovation, Lausanne, CH, Switzerland
| | - J Loyau
- Ichnos Glenmark Innovation, Lausanne, CH, Switzerland
| | - V Labanca
- Ichnos Glenmark Innovation, Lausanne, CH, Switzerland
| | - A Drake
- Ichnos Glenmark Innovation, Lausanne, CH, Switzerland
| | - S De Angelis
- Ichnos Glenmark Innovation, Lausanne, CH, Switzerland
| | - A Rubod
- Ichnos Glenmark Innovation, Lausanne, CH, Switzerland
| | - J Frei
- Ichnos Glenmark Innovation, Lausanne, CH, Switzerland
| | - L N Caro
- Ichnos Glenmark Innovation, Lausanne, CH, Switzerland
| | - S Blein
- Ichnos Glenmark Innovation, Lausanne, CH, Switzerland
| | - E Martini
- Ichnos Glenmark Innovation, Lausanne, CH, Switzerland
| | - M Chimen
- Ichnos Glenmark Innovation, Lausanne, CH, Switzerland
| | - T Matthes
- Haematology Service, Department of Oncology and Clinical Pathology Service, Department of Diagnostics, University Hospital Geneva, 1211, Geneva, Switzerland
| | - Z Kaya
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Institute, University of Oxford, Oxford, UK
| | - C M Edwards
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Institute, University of Oxford, Oxford, UK
| | - J R Edwards
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Institute, University of Oxford, Oxford, UK
| | - E Menoret
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, Nantes, France
| | - C Kervoelen
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, Nantes, France
| | - C Pellat-Deceunynck
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, Nantes, France
- SIRIC ILIAD, Angers, Nantes, France
| | - P Moreau
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, Nantes, France
- SIRIC ILIAD, Angers, Nantes, France
- Service d'Hématologie Clinique, Unité d'Investigation Clinique, CHU, Nantes, France
| | - M L Mbow
- Ichnos Glenmark Innovation, Lausanne, CH, Switzerland
| | - A Srivastava
- Ichnos Glenmark Innovation, Lausanne, CH, Switzerland
| | - M R Dyson
- Ichnos Glenmark Innovation, Lausanne, CH, Switzerland
| | - E A Zhukovsky
- Ichnos Glenmark Innovation, Lausanne, CH, Switzerland
| | - M Perro
- Ichnos Glenmark Innovation, Lausanne, CH, Switzerland.
| | - S Sammicheli
- Ichnos Glenmark Innovation, Lausanne, CH, Switzerland.
| |
Collapse
|
6
|
Hayes BH, Wang M, Zhu H, Phan SH, Dooling LJ, Andrechak JC, Chang AH, Tobin MP, Ontko NM, Marchena T, Discher DE. Chromosomal instability can favor macrophage-mediated immune response and induce a broad, vaccination-like anti-tumor IgG response. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.04.02.535275. [PMID: 37066426 PMCID: PMC10103980 DOI: 10.1101/2023.04.02.535275] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/22/2023]
Abstract
Chromosomal instability (CIN), a state in which cells undergo mitotic aberrations that generate chromosome copy number variations, generates aneuploidy and is thought to drive cancer evolution. Although associated with poor prognosis and reduced immune response, CIN generates aneuploidy-induced stresses that could be exploited for immunotherapies. In such contexts, macrophages and the CD47-SIRPα checkpoint are understudied. Here, CIN is induced pharmacologically induced in poorly immunogenic B16F10 mouse melanoma cells, generating persistent micronuclei and diverse aneuploidy while skewing macrophages towards an anti-cancer M1-like phenotype, based on RNA-sequencing profiling, surface marker expression and short-term antitumor studies. These results further translate to in vivo efficacy: Mice bearing CIN-afflicted tumors with wild-type CD47 levels survive only slightly longer relative to chromosomally stable controls, but long-term survival is maximized when combining macrophage-stimulating anti-tumor IgG opsonization and some form of disruption of the CD47-SIRPα checkpoint. Survivors make multi-epitope, de novo anti-cancer IgG that promote macrophage-mediated phagocytosis of CD47 knockout B16F10 cells and suppress tumoroids in vitro and growth of tumors in vivo . CIN does not greatly affect the level of the IgG response compared to previous studies but does significantly increase survival. These results highlight an unexpected therapeutic benefit from CIN when paired with maximal macrophage anti-cancer activity: an anti-cancer vaccination-like antibody response that can lead to more durable cures and further potentiate cell-mediated acquired immunity.
Collapse
|
7
|
Arias CF, Valente-Leal N, Bertocchini F, Marques S, Acosta FJ, Fernandez-Arias C. A new role for erythropoietin in the homeostasis of red blood cells. Commun Biol 2024; 7:58. [PMID: 38191841 PMCID: PMC10774343 DOI: 10.1038/s42003-023-05758-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 12/29/2023] [Indexed: 01/10/2024] Open
Abstract
The regulation of red blood cell (RBC) homeostasis is widely assumed to rely on the control of cell production by erythropoietin (EPO) and the destruction of cells at a fixed, species-specific age. In this work, we show that such a regulatory mechanism would be a poor homeostatic solution to satisfy the changing needs of the body. Effective homeostatic control would require RBC lifespan to be variable and tightly regulated. We suggest that EPO may control RBC lifespan by determining CD47 expression in newly formed RBCs and SIRP-α expression in sinusoidal macrophages. EPO could also regulate the initiation and intensity of anti-RBC autoimmune responses that curtail RBC lifespan in some circumstances. These mechanisms would continuously modulate the rate of RBC destruction depending on oxygen availability. The control of RBC lifespan by EPO and autoimmunity emerges as a key mechanism in the homeostasis of RBCs.
Collapse
Affiliation(s)
- Clemente F Arias
- Centro de Investigaciones Biológicas (CSIC), Madrid, Spain.
- Grupo Interdisciplinar de Sistemas Complejos (GISC), Madrid, Spain.
| | - Nuno Valente-Leal
- Instituto de Medicina Molecular, Universidade de Lisboa, Lisboa, Portugal
| | | | - Sofia Marques
- Instituto de Medicina Molecular, Universidade de Lisboa, Lisboa, Portugal
| | - Francisco J Acosta
- Departamento de Ecología, Universidad Complutense de Madrid, Madrid, Spain
| | - Cristina Fernandez-Arias
- Instituto de Medicina Molecular, Universidade de Lisboa, Lisboa, Portugal.
- Departamento de Immunología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain.
| |
Collapse
|
8
|
Osorio JC, Smith P, Knorr DA, Ravetch JV. The antitumor activities of anti-CD47 antibodies require Fc-FcγR interactions. Cancer Cell 2023; 41:2051-2065.e6. [PMID: 37977147 PMCID: PMC10842210 DOI: 10.1016/j.ccell.2023.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 09/01/2023] [Accepted: 10/26/2023] [Indexed: 11/19/2023]
Abstract
While anti-CD47 antibodies hold promise for cancer immunotherapy, early-phase clinical trials have shown limited clinical benefit, suggesting that CD47 blockade alone might be insufficient for effective tumor control. Here, we investigate the contributions of the Fc domain of anti-CD47 antibodies required for optimal in vivo antitumor activity across multiple species-matched models, providing insights into the mechanisms behind the efficacy of this emerging class of therapeutic antibodies. Using a mouse model humanized for CD47, SIRPα, and FcγRs, we demonstrate that local administration of Fc-engineered anti-CD47 antibodies with enhanced binding to activating FcγRs promotes tumor infiltration of macrophages and antigen-specific T cells, while depleting regulatory T cells. These effects result in improved long-term systemic antitumor immunity and minimal on-target off-tumor toxicity. Our results highlight the importance of Fc optimization in the development of effective anti-CD47 therapies and provide an attractive strategy to enhance the activity of this promising immunotherapy.
Collapse
Affiliation(s)
- Juan C Osorio
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Laboratory of Molecular Genetics and Immunology, Rockefeller University, New York, NY 10065, USA.
| | - Patrick Smith
- Laboratory of Molecular Genetics and Immunology, Rockefeller University, New York, NY 10065, USA
| | - David A Knorr
- Laboratory of Molecular Genetics and Immunology, Rockefeller University, New York, NY 10065, USA; Regeneron, Inc., Tarrytown, NY, USA
| | - Jeffrey V Ravetch
- Laboratory of Molecular Genetics and Immunology, Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
9
|
Aizaz M, Khan A, Khan F, Khan M, Musad Saleh EA, Nisar M, Baran N. The cross-talk between macrophages and tumor cells as a target for cancer treatment. Front Oncol 2023; 13:1259034. [PMID: 38033495 PMCID: PMC10682792 DOI: 10.3389/fonc.2023.1259034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 10/17/2023] [Indexed: 12/02/2023] Open
Abstract
Macrophages represent an important component of the innate immune system. Under physiological conditions, macrophages, which are essential phagocytes, maintain a proinflammatory response and repair damaged tissue. However, these processes are often impaired upon tumorigenesis, in which tumor-associated macrophages (TAMs) protect and support the growth, proliferation, and invasion of tumor cells and promote suppression of antitumor immunity. TAM abundance is closely associated with poor outcome of cancer, with impediment of chemotherapy effectiveness and ultimately a dismal therapy response and inferior overall survival. Thus, cross-talk between cancer cells and TAMs is an important target for immune checkpoint therapies and metabolic interventions, spurring interest in it as a therapeutic vulnerability for both hematological cancers and solid tumors. Furthermore, targeting of this cross-talk has emerged as a promising strategy for cancer treatment with the antibody against CD47 protein, a critical macrophage checkpoint recognized as the "don't eat me" signal, as well as other metabolism-focused strategies. Therapies targeting CD47 constitute an important milestone in the advancement of anticancer research and have had promising effects on not only phagocytosis activation but also innate and adaptive immune system activation, effectively counteracting tumor cells' evasion of therapy as shown in the context of myeloid cancers. Targeting of CD47 signaling is only one of several possibilities to reverse the immunosuppressive and tumor-protective tumor environment with the aim of enhancing the antitumor response. Several preclinical studies identified signaling pathways that regulate the recruitment, polarization, or metabolism of TAMs. In this review, we summarize the current understanding of the role of macrophages in cancer progression and the mechanisms by which they communicate with tumor cells. Additionally, we dissect various therapeutic strategies developed to target macrophage-tumor cell cross-talk, including modulation of macrophage polarization, blockade of signaling pathways, and disruption of physical interactions between leukemia cells and macrophages. Finally, we highlight the challenges associated with tumor hypoxia and acidosis as barriers to effective cancer therapy and discuss opportunities for future research in this field.
Collapse
Affiliation(s)
- Muhammad Aizaz
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Aakif Khan
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Faisal Khan
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Maria Khan
- Center of Biotechnology and Microbiology, University of Peshawar, Peshawar, Pakistan
| | - Ebraheem Abdu Musad Saleh
- Department of Chemistry, College of Arts & Science, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| | - Maryum Nisar
- School of Interdisciplinary Engineering & Sciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Natalia Baran
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
10
|
Dooling LJ, Andrechak JC, Hayes BH, Kadu S, Zhang W, Pan R, Vashisth M, Irianto J, Alvey CM, Ma L, Discher DE. Cooperative phagocytosis of solid tumours by macrophages triggers durable anti-tumour responses. Nat Biomed Eng 2023; 7:1081-1096. [PMID: 37095318 PMCID: PMC10791169 DOI: 10.1038/s41551-023-01031-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 03/27/2023] [Indexed: 04/26/2023]
Abstract
In solid tumours, the abundance of macrophages is typically associated with a poor prognosis. However, macrophage clusters in tumour-cell nests have been associated with survival in some tumour types. Here, by using tumour organoids comprising macrophages and cancer cells opsonized via a monoclonal antibody, we show that highly ordered clusters of macrophages cooperatively phagocytose cancer cells to suppress tumour growth. In mice with poorly immunogenic tumours, the systemic delivery of macrophages with signal-regulatory protein alpha (SIRPα) genetically knocked out or else with blockade of the CD47-SIRPα macrophage checkpoint was combined with the monoclonal antibody and subsequently triggered the production of endogenous tumour-opsonizing immunoglobulin G, substantially increased the survival of the animals and helped confer durable protection from tumour re-challenge and metastasis. Maximizing phagocytic potency by increasing macrophage numbers, by tumour-cell opsonization and by disrupting the phagocytic checkpoint CD47-SIRPα may lead to durable anti-tumour responses in solid cancers.
Collapse
Affiliation(s)
- Lawrence J Dooling
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA
- Physical Sciences-Oncology Center at Penn, University of Pennsylvania, Philadelphia, PA, USA
| | - Jason C Andrechak
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA
- Physical Sciences-Oncology Center at Penn, University of Pennsylvania, Philadelphia, PA, USA
- Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, PA, USA
| | - Brandon H Hayes
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA
- Physical Sciences-Oncology Center at Penn, University of Pennsylvania, Philadelphia, PA, USA
- Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, PA, USA
| | - Siddhant Kadu
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA
- Physical Sciences-Oncology Center at Penn, University of Pennsylvania, Philadelphia, PA, USA
| | - William Zhang
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA
- Physical Sciences-Oncology Center at Penn, University of Pennsylvania, Philadelphia, PA, USA
- Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, PA, USA
| | - Ruby Pan
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA
- Physical Sciences-Oncology Center at Penn, University of Pennsylvania, Philadelphia, PA, USA
| | - Manasvita Vashisth
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA
- Physical Sciences-Oncology Center at Penn, University of Pennsylvania, Philadelphia, PA, USA
| | - Jerome Irianto
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL, USA
| | - Cory M Alvey
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA
- Physical Sciences-Oncology Center at Penn, University of Pennsylvania, Philadelphia, PA, USA
| | - Leyuan Ma
- Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, PA, USA
- Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dennis E Discher
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA.
- Physical Sciences-Oncology Center at Penn, University of Pennsylvania, Philadelphia, PA, USA.
- Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
11
|
Hayes BH, Zhu H, Andrechak JC, Dooling LJ, Discher DE. Titrating CD47 by mismatch CRISPR-interference reveals incomplete repression can eliminate IgG-opsonized tumors but limits induction of antitumor IgG. PNAS NEXUS 2023; 2:pgad243. [PMID: 37593202 PMCID: PMC10427748 DOI: 10.1093/pnasnexus/pgad243] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 06/22/2023] [Accepted: 07/19/2023] [Indexed: 08/19/2023]
Abstract
Phagocytic elimination of solid tumors by innate immune cells seems attractive for immunotherapy, particularly because of the possibilities for acquired immunity. However, the approach remains challenging, with blockade of the macrophage checkpoint CD47 working in immunodeficient mice and against highly immunogenic tumors but not in the clinic where tumors are poorly immunogenic. Even when mouse tumors of poorly immunogenic B16F10 melanoma are opsonized to drive engulfment with a suitable monoclonal antibody (mAb), anti-CD47 blockade remains insufficient. Using both in vitro immuno-tumoroids and in vivo mouse models, we show with CRISPR interference (CRISPRi) that a relatively uniform minimum repression of CD47 by 80% is needed for phagocytosis to dominate net growth when combined with an otherwise ineffective mAb (anti-Tyrp1). Heterogeneity enriches for CD47-high cells, but mice that eliminate tumors generate prophagocytic IgGs that increase in titer with CD47 repression and with tumor accumulation of macrophages, although deeper repression does not improve survival. Given well-known limitations of antibody permeation into solid tumors, our studies clarify benchmarks for CD47 disruption that should be more clinically feasible and safer but just as effective as complete ablation. Additionally, safe but ineffective opsonization in human melanoma trials suggests that combinations with deep repression of CD47 could prove effective and initiate durable immunity.
Collapse
Affiliation(s)
- Brandon H Hayes
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104, USA
- Physical Sciences Oncology Center at Penn, University of Pennsylvania, Philadelphia, PA 19104, USA
- Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hui Zhu
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104, USA
- Physical Sciences Oncology Center at Penn, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jason C Andrechak
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104, USA
- Physical Sciences Oncology Center at Penn, University of Pennsylvania, Philadelphia, PA 19104, USA
- Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lawrence J Dooling
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104, USA
- Physical Sciences Oncology Center at Penn, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Dennis E Discher
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104, USA
- Physical Sciences Oncology Center at Penn, University of Pennsylvania, Philadelphia, PA 19104, USA
- Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
12
|
Osorio JC, Smith P, Knorr DA, Ravetch JV. The Antitumor Activities of Anti-CD47 Antibodies Require Fc-FcγR interactions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.29.547082. [PMID: 37455857 PMCID: PMC10347539 DOI: 10.1101/2023.06.29.547082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/18/2023]
Abstract
While anti-CD47 antibodies hold promise for cancer immunotherapy, early phase clinical trials have shown limited signs of clinical benefit, suggesting that blockade of CD47 alone may not be sufficient for effective tumor control. Here, we investigate the contributions of the Fc domain of anti-CD47 antibodies required for optimal in vivo antitumor activity across multiple species-matched models, providing new insights into the mechanisms underlying the efficacy of this emerging class of therapeutic antibodies. Using a novel mouse model humanized for CD47, SIRPα and FcγRs, we demonstrate that local administration of an Fc-engineered anti-CD47 antibody with enhanced binding to activating FcγRs modulates myeloid and T-cell subsets in the tumor microenvironment, resulting in improved long-term systemic antitumor immunity and minimal on-target off-tumor toxicity. Our results highlight the importance of Fc optimization in the development of effective anti-CD47 therapies and provide a novel approach for enhancing the antitumor activity of this promising immunotherapy.
Collapse
Affiliation(s)
- Juan C Osorio
- Laboratory of Molecular Genetics and Immunology, Rockefeller University, New York, NY, 10065, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Patrick Smith
- Laboratory of Molecular Genetics and Immunology, Rockefeller University, New York, NY, 10065, USA
| | - David A Knorr
- Laboratory of Molecular Genetics and Immunology, Rockefeller University, New York, NY, 10065, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Jeffrey V Ravetch
- Laboratory of Molecular Genetics and Immunology, Rockefeller University, New York, NY, 10065, USA
| |
Collapse
|
13
|
Arnold D, Takatori SC. Bio-enabled Engineering of Multifunctional "Living" Surfaces. ACS NANO 2023; 17:11077-11086. [PMID: 37294942 PMCID: PMC10311588 DOI: 10.1021/acsnano.3c03138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 06/06/2023] [Indexed: 06/11/2023]
Abstract
Through the magic of "active matter"─matter that converts chemical energy into mechanical work to drive emergent properties─biology solves a myriad of seemingly enormous physical challenges. Using active matter surfaces, for example, our lungs clear an astronomically large number of particulate contaminants that accompany each of the 10,000 L of air we respire per day, thus ensuring that the lungs' gas exchange surfaces remain functional. In this Perspective, we describe our efforts to engineer artificial active surfaces that mimic active matter surfaces in biology. Specifically, we seek to assemble the basic active matter components─mechanical motor, driven constituent, and energy source─to design surfaces that support the continuous operation of molecular sensing, recognition, and exchange. The successful realization of this technology would generate multifunctional, "living" surfaces that combine the dynamic programmability of active matter and the molecular specificity of biological surfaces and apply them to applications in biosensors, chemical diagnostics, and other surface transport and catalytic processes. We describe our recent efforts in bio-enabled engineering of living surfaces through the design of molecular probes to understand and integrate native biological membranes into synthetic materials.
Collapse
Affiliation(s)
- Daniel
P. Arnold
- Department of Chemical Engineering, University of California, Santa
Barbara, California 93106, United States
| | - Sho C. Takatori
- Department of Chemical Engineering, University of California, Santa
Barbara, California 93106, United States
| |
Collapse
|
14
|
Xu S, Wang C, Yang L, Wu J, Li M, Xiao P, Xu Z, Xu Y, Wang K. Targeting immune checkpoints on tumor-associated macrophages in tumor immunotherapy. Front Immunol 2023; 14:1199631. [PMID: 37313405 PMCID: PMC10258331 DOI: 10.3389/fimmu.2023.1199631] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 05/16/2023] [Indexed: 06/15/2023] Open
Abstract
Unprecedented breakthroughs have been made in cancer immunotherapy in recent years. Particularly immune checkpoint inhibitors have fostered hope for patients with cancer. However, immunotherapy still exhibits certain limitations, such as a low response rate, limited efficacy in certain populations, and adverse events in certain tumors. Therefore, exploring strategies that can improve clinical response rates in patients is crucial. Tumor-associated macrophages (TAMs) are the predominant immune cells that infiltrate the tumor microenvironment and express a variety of immune checkpoints that impact immune functions. Mounting evidence indicates that immune checkpoints in TAMs are closely associated with the prognosis of patients with tumors receiving immunotherapy. This review centers on the regulatory mechanisms governing immune checkpoint expression in macrophages and strategies aimed at improving immune checkpoint therapies. Our review provides insights into potential therapeutic targets to improve the efficacy of immune checkpoint blockade and key clues to developing novel tumor immunotherapies.
Collapse
Affiliation(s)
- Shumin Xu
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Chenyang Wang
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Lingge Yang
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Jiaji Wu
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Mengshu Li
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Peng Xiao
- School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhiyong Xu
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Yun Xu
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Kai Wang
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| |
Collapse
|
15
|
Arnold DP, Xu Y, Takatori SC. Antibody binding reports spatial heterogeneities in cell membrane organization. Nat Commun 2023; 14:2884. [PMID: 37208326 DOI: 10.1038/s41467-023-38525-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 05/05/2023] [Indexed: 05/21/2023] Open
Abstract
The spatial organization of cell membrane glycoproteins and glycolipids is critical for mediating the binding of ligands, receptors, and macromolecules on the plasma membrane. However, we currently do not have the methods to quantify the spatial heterogeneities of macromolecular crowding on live cell surfaces. In this work, we combine experiment and simulation to report crowding heterogeneities on reconstituted membranes and live cell membranes with nanometer spatial resolution. By quantifying the effective binding affinity of IgG monoclonal antibodies to engineered antigen sensors, we discover sharp gradients in crowding within a few nanometers of the crowded membrane surface. Our measurements on human cancer cells support the hypothesis that raft-like membrane domains exclude bulky membrane proteins and glycoproteins. Our facile and high-throughput method to quantify spatial crowding heterogeneities on live cell membranes may facilitate monoclonal antibody design and provide a mechanistic understanding of plasma membrane biophysical organization.
Collapse
Affiliation(s)
- Daniel P Arnold
- Department of Chemical Engineering, University of California, Santa Barbara, Santa Barbara, CA, 93106, USA
| | - Yaxin Xu
- Department of Chemical Engineering, University of California, Santa Barbara, Santa Barbara, CA, 93106, USA
| | - Sho C Takatori
- Department of Chemical Engineering, University of California, Santa Barbara, Santa Barbara, CA, 93106, USA.
| |
Collapse
|
16
|
Hao Y, Zhou X, Li Y, Li B, Cheng L. The CD47-SIRPα axis is a promising target for cancer immunotherapies. Int Immunopharmacol 2023; 120:110255. [PMID: 37187126 DOI: 10.1016/j.intimp.2023.110255] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/22/2023] [Accepted: 04/27/2023] [Indexed: 05/17/2023]
Abstract
Cluster of differentiation 47(CD47) is a transmembrane protein that is ubiquitously found on the surface of many cells in the body and uniquely overexpressed by both solid and hematologic malignant cells. CD47 interacts with signal-regulatory protein α (SIRPα), to trigger a "don't eat me" signal and thereby achieve cancer immune escape by inhibiting macrophage-mediated phagocytosis. Thus, blocking the CD47-SIRPα phagocytosis checkpoint, for release of the innate immune system, is a current research focus. Indeed, targeting the CD47-SIRPα axis as a cancer immunotherapy has shown promising efficacies in pre-clinical outcomes. Here, we first reviewed the origin, structure, and function of the CD47-SIRPα axis. Then, we reviewed its role as a target for cancer immunotherapies, as well as the factors regulating CD47-SIRPα axis-based immunotherapies. We specifically focused on the mechanism and progress of CD47-SIRPα axis-based immunotherapies and their combination with other treatment strategies. Finally, we discussed the challenges and directions for future research and identified potential CD47-SIRPα axis-based therapies that are suitable for clinical application.
Collapse
Affiliation(s)
- Yu Hao
- State Key Laboratory of Oral Diseases & West China Hospital of Stomatology & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610041, China; Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xinxuan Zhou
- State Key Laboratory of Oral Diseases & West China Hospital of Stomatology & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610041, China
| | - Yiling Li
- State Key Laboratory of Oral Diseases & West China Hospital of Stomatology & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610041, China; Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Bolei Li
- State Key Laboratory of Oral Diseases & West China Hospital of Stomatology & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610041, China; Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Lei Cheng
- State Key Laboratory of Oral Diseases & West China Hospital of Stomatology & National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu 610041, China; Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
17
|
Ye ZH, Yu WB, Huang MY, Chen J, Lu JJ. Building on the backbone of CD47-based therapy in cancer: Combination strategies, mechanisms, and future perspectives. Acta Pharm Sin B 2023; 13:1467-1487. [PMID: 37139405 PMCID: PMC10149906 DOI: 10.1016/j.apsb.2022.12.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 10/10/2022] [Accepted: 11/18/2022] [Indexed: 12/27/2022] Open
Abstract
Described as a "don't eat me" signal, CD47 becomes a vital immune checkpoint in cancer. Its interaction with signal regulatory protein alpha (SIRPα) prevents macrophage phagocytosis. In recent years, a growing body of evidences have unveiled that CD47-based combination therapy exhibits a superior anti-cancer effect. Latest clinical trials about CD47 have adopted the regimen of collaborating with other therapies or developing CD47-directed bispecific antibodies, indicating the combination strategy as a general trend of the future. In this review, clinical and preclinical cases about the current combination strategies targeting CD47 are collected, their underlying mechanisms of action are discussed, and ideas from future perspectives are shared.
Collapse
Affiliation(s)
- Zi-Han Ye
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Wei-Bang Yu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Mu-Yang Huang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Jun Chen
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Jin-Jian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macao 999078, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Macao 999078, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, University of Macau, Macao 999078, China
| |
Collapse
|
18
|
Ghantous L, Volman Y, Hefez R, Wald O, Stern E, Friehmann T, Chajut A, Bremer E, Elhalel MD, Rachmilewitz J. The DNA damage response pathway regulates the expression of the immune checkpoint CD47. Commun Biol 2023; 6:245. [PMID: 36882648 PMCID: PMC9992352 DOI: 10.1038/s42003-023-04615-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 02/21/2023] [Indexed: 03/09/2023] Open
Abstract
CD47 is a cell surface ligand expressed on all nucleated cells. It is a unique immune checkpoint protein acting as "don't eat me" signal to prevent phagocytosis and is constitutively overexpressed in many tumors. However, the underlying mechanism(s) for CD47 overexpression is not clear. Here, we show that irradiation (IR) as well as various other genotoxic agents induce elevated expression of CD47. This upregulation correlates with the extent of residual double-strand breaks (DSBs) as determined by γH2AX staining. Interestingly, cells lacking mre-11, a component of the MRE11-RAD50-NBS1 (MRN) complex that plays a central role in DSB repair, or cells treated with the mre-11 inhibitor, mirin, fail to elevate the expression of CD47 upon DNA damage. On the other hand, both p53 and NF-κB pathways or cell-cycle arrest do not play a role in CD47 upregualtion upon DNA damage. We further show that CD47 expression is upregulated in livers harvested from mice treated with the DNA-damage inducing agent Diethylnitrosamine (DEN) and in cisplatin-treated mesothelioma tumors. Hence, our results indicate that CD47 is upregulated following DNA damage in a mre-11-dependent manner. Chronic DNA damage response in cancer cells might contribute to constitutive elevated expression of CD47 and promote immune evasion.
Collapse
Affiliation(s)
- Lucy Ghantous
- Goldyne Savad Institute of Gene Therapy, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Nephrology and Hypertension, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yael Volman
- Goldyne Savad Institute of Gene Therapy, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ruth Hefez
- Goldyne Savad Institute of Gene Therapy, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ori Wald
- Goldyne Savad Institute of Gene Therapy, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Cardiothoracic Surgery, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Esther Stern
- Goldyne Savad Institute of Gene Therapy, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Tomer Friehmann
- Goldyne Savad Institute of Gene Therapy, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | | | - Edwin Bremer
- Department of Hematology, University Medical Center Groningen, Groningen, The Netherlands
| | - Michal Dranitzki Elhalel
- Department of Nephrology and Hypertension, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.
| | - Jacob Rachmilewitz
- Goldyne Savad Institute of Gene Therapy, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
19
|
Li Y, Zhou H, Liu P, Lv D, Shi Y, Tang B, Xu J, Zhong T, Xu W, Zhang J, Zhou J, Ying K, Zhao Y, Sun Y, Jiang Z, Cheng H, Zhang X, Ke Y. SHP2 deneddylation mediates tumor immunosuppression in colon cancer via the CD47/SIRPα axis. J Clin Invest 2023; 133:162870. [PMID: 36626230 PMCID: PMC9927946 DOI: 10.1172/jci162870] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 01/05/2023] [Indexed: 01/11/2023] Open
Abstract
SIPRα on macrophages binds with CD47 to resist proengulfment signals, but how the downstream signal of SIPRα controls tumor-infiltrating macrophages (TIMs) is still poorly clarified. Here, we report that the CD47/signal regulatory protein α (SIRPα) axis requires the deneddylation of tyrosine phosphatase SHP2. Mechanistically, Src homology region 2-containing protein tyrosine phosphatase 2 (SHP2) was constitutively neddylated on K358 and K364 sites; thus, its autoinhibited conformation was maintained. In response to CD47-liganded SIRPα, SHP2 was deneddylated by sentrin-specific protease 8 (SENP8), which led to the dephosphorylation of relevant substrates at the phagocytic cup and subsequent inhibition of macrophage phagocytosis. Furthermore, neddylation inactivated myeloid-SHP2 and greatly boosted the efficacy of colorectal cancer (CRC) immunotherapy. Importantly, we observed that supplementation with SHP2 allosteric inhibitors sensitized immune treatment-resistant CRC to immunotherapy. Our results emphasize that the CRC subtype that is unresponsive to immunotherapy relies on SIRPαhiSHP2hiNEDD8lo TIMs and highlight the need to further explore the strategy of SHP2 targeting in CRC therapy.
Collapse
Affiliation(s)
- Yiqing Li
- Department of Pathology and Pathophysiology, and Department of Respiratory Medicine at Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hui Zhou
- Department of Pathology and Pathophysiology, and Department of Respiratory Medicine at Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Pan Liu
- Zhejiang Cancer Hospital, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Dandan Lv
- Department of Respiratory Medicine at Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yichun Shi
- Department of Pathology and Pathophysiology, and Department of Respiratory Medicine at Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Bufu Tang
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research at The Lishui Hospital, Zhejiang University School of Medicine, Lishui, China
| | - Jiaqi Xu
- Department of Pathology at Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangou, China
| | - Tingting Zhong
- Department of Pathology at Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangou, China
| | - Wangting Xu
- Department of Respiratory Medicine at The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jie Zhang
- Department of Urology at Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianying Zhou
- Department of Respiratory Medicine at The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kejing Ying
- Department of Respiratory Medicine at Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yongchao Zhao
- Cancer Institute of The Second Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Yi Sun
- Cancer Institute of The Second Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhinong Jiang
- Department of Pathology at Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangou, China
| | - Hongqiang Cheng
- Department of Pathology and Pathophysiology, and Department of Cardiology at Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xue Zhang
- Department of Pathology and Pathophysiology, and Department of Respiratory Medicine at Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuehai Ke
- Department of Pathology and Pathophysiology, and Department of Respiratory Medicine at Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
20
|
Semiz HS, Küçük Ü, Kısa E, Keskinkılıç M, Süyün DE, Arayıcı ME, Atağ E, Karaoglu A. CD47 (don't eat me signal) expression levels and its relationship with clinicopathologic features in early-stage prostate carcinoma. Prostate 2022; 82:1564-1571. [PMID: 36031743 DOI: 10.1002/pros.24432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 07/11/2022] [Accepted: 08/10/2022] [Indexed: 11/11/2022]
Abstract
BACKGROUND Prostate cancer is a cancer with poor host immune response and could be defined as a non-T-cell inflamed tumor. Therefore, immunotherapy treatments could not be included in the treatment of prostate cancer until recently. Inadequate antitumoral response is one of the main reasons why tumor cells multiply rapidly and cause lethal results. It was shown that CD47 molecule, which is secreted at high levels by leukemia cells, reduces macrophage-mediated phagocytosis and thus facilitates escape from the antitumoral immune response. The aim of this study was to show don't eat me signaling in prostate carcinoma tissues and its relationship with macrophage polarization. MATERIALS AND METHODS A total of 263 patients with a diagnosis of prostatic adenocarcinoma after radical prostatectomy between 2015 and 2020 at our institute were included in the study. CD47, CD68, and CD163 expression levels were examined immunohistochemically (IHC) in these tissues. The relationship of these expression levels with unfavorable prognostic factors and survival for prostate carcinoma was investigated. RESULTS In this study, all the operated prostate carcinoma cases had CD47 expression in tumor tissue, but only 52.5% had a high level of expression. Of 263 prostate cancer tissues, 135 (51.3%) showed high expression of CD68 protein and 189 (71.9%) showed high expression of CD163 protein. There was a statistically strong relationship between CD47, CD68, and CD163. CONCLUSIONS The CD47 molecule is basically a molecule that inhibits macrophage activation. CD68 is mostly used for macrophage classification, while CD163 is used for tumor-associated macrophage classification. Unlike others, we IHC examined CD47, CD68, and CD163 expressions in the surgical materials of patients who were operated for prostate carcinoma. In addition, we concluded that strong CD47 expression was closely associated with strong CD68 and CD163 expression in all tumor samples. However, a significant relationship between these expression levels and survival could not be demonstrated.
Collapse
Affiliation(s)
- Hüseyin S Semiz
- Department of Medical Oncology, Institute of Oncology, Dokuz Eylul University, Izmir, Turkey
| | - Ülkü Küçük
- Department of Pathology, Tepecik Education and Research Hospital, Health Science University, Izmir, Turkey
| | - Erdem Kısa
- Department of Urology, Tepecik Education and Research Hospital, Health Science University, Izmir, Turkey
| | - Merve Keskinkılıç
- Department of Internal Medicine, Division of Medical Oncology, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - Dilara Ecem Süyün
- Department of Internal Medicine, Tepecik Education and Research Hospital, Izmir, Turkey
| | - Mehmet Emin Arayıcı
- Department of Preventive Oncology, Institute of Health Sciences, Dokuz Eylul University, Izmir, Turkey
| | - Elif Atağ
- Department of Medical Oncology, Istanbul Haydarpasa Numune Education and Research Hospital, Health Science University, Istanbul, Turkey
| | - Aziz Karaoglu
- Department of Medical Oncology, Institute of Oncology, Dokuz Eylul University, Izmir, Turkey
| |
Collapse
|
21
|
Zeller T, Lutz S, Münnich IA, Windisch R, Hilger P, Herold T, Tahiri N, Banck JC, Weigert O, Moosmann A, von Bergwelt-Baildon M, Flamann C, Bruns H, Wichmann C, Baumann N, Valerius T, Schewe DM, Peipp M, Rösner T, Humpe A, Kellner C. Dual checkpoint blockade of CD47 and LILRB1 enhances CD20 antibody-dependent phagocytosis of lymphoma cells by macrophages. Front Immunol 2022; 13:929339. [PMID: 36389667 PMCID: PMC9647079 DOI: 10.3389/fimmu.2022.929339] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 10/12/2022] [Indexed: 11/28/2022] Open
Abstract
Antibody-dependent cellular phagocytosis (ADCP) by macrophages, an important effector function of tumor targeting antibodies, is hampered by ‘Don´t Eat Me!’ signals such as CD47 expressed by cancer cells. Yet, human leukocyte antigen (HLA) class I expression may also impair ADCP by engaging leukocyte immunoglobulin-like receptor subfamily B (LILRB) member 1 (LILRB1) or LILRB2. Analysis of different lymphoma cell lines revealed that the ratio of CD20 to HLA class I cell surface molecules determined the sensitivity to ADCP by the combination of rituximab and an Fc-silent variant of the CD47 antibody magrolimab (CD47-IgGσ). To boost ADCP, Fc-silent antibodies against LILRB1 and LILRB2 were generated (LILRB1-IgGσ and LILRB2-IgGσ, respectively). While LILRB2-IgGσ was not effective, LILRB1-IgGσ significantly enhanced ADCP of lymphoma cell lines when combined with both rituximab and CD47-IgGσ. LILRB1-IgGσ promoted serial engulfment of lymphoma cells and potentiated ADCP by non-polarized M0 as well as polarized M1 and M2 macrophages, but required CD47 co-blockade and the presence of the CD20 antibody. Importantly, complementing rituximab and CD47-IgGσ, LILRB1-IgGσ increased ADCP of chronic lymphocytic leukemia (CLL) or lymphoma cells isolated from patients. Thus, dual checkpoint blockade of CD47 and LILRB1 may be promising to improve antibody therapy of CLL and lymphomas through enhancing ADCP by macrophages.
Collapse
Affiliation(s)
- Tobias Zeller
- Division of Transfusion Medicine, Cell Therapeutics and Haemostaseology, University Hospital, LMU Munich, Munich, Germany
| | - Sebastian Lutz
- Division of Transfusion Medicine, Cell Therapeutics and Haemostaseology, University Hospital, LMU Munich, Munich, Germany
| | - Ira A. Münnich
- Division of Transfusion Medicine, Cell Therapeutics and Haemostaseology, University Hospital, LMU Munich, Munich, Germany
| | - Roland Windisch
- Division of Transfusion Medicine, Cell Therapeutics and Haemostaseology, University Hospital, LMU Munich, Munich, Germany
| | - Patricia Hilger
- Division of Transfusion Medicine, Cell Therapeutics and Haemostaseology, University Hospital, LMU Munich, Munich, Germany
| | - Tobias Herold
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Natyra Tahiri
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
| | - Jan C. Banck
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
| | - Oliver Weigert
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Andreas Moosmann
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
- DZIF – German Center for Infection Research, Munich, Germany
- Helmholtz Zentrum München, Munich, Germany
| | - Michael von Bergwelt-Baildon
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Cindy Flamann
- Department of Internal Medicine 5, University Hospital Erlangen, Erlangen, Germany
| | - Heiko Bruns
- Department of Internal Medicine 5, University Hospital Erlangen, Erlangen, Germany
| | - Christian Wichmann
- Division of Transfusion Medicine, Cell Therapeutics and Haemostaseology, University Hospital, LMU Munich, Munich, Germany
| | - Niklas Baumann
- Division of Stem Cell Transplantation and Immunotherapy, Department of Internal Medicine II, Christian Albrechts University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Thomas Valerius
- Division of Stem Cell Transplantation and Immunotherapy, Department of Internal Medicine II, Christian Albrechts University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Denis M. Schewe
- Department of Pediatrics, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Matthias Peipp
- Division of Antibody-Based Immunotherapy, Department of Internal Medicine II, Christian Albrechts University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Thies Rösner
- Division of Stem Cell Transplantation and Immunotherapy, Department of Internal Medicine II, Christian Albrechts University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Andreas Humpe
- Division of Transfusion Medicine, Cell Therapeutics and Haemostaseology, University Hospital, LMU Munich, Munich, Germany
| | - Christian Kellner
- Division of Transfusion Medicine, Cell Therapeutics and Haemostaseology, University Hospital, LMU Munich, Munich, Germany
- *Correspondence: Christian Kellner,
| |
Collapse
|
22
|
Baumann N, Arndt C, Petersen J, Lustig M, Rösner T, Klausz K, Kellner C, Bultmann M, Bastian L, Vogiatzi F, Leusen JHW, Burger R, Schewe DM, Peipp M, Valerius T. Myeloid checkpoint blockade improves killing of T-acute lymphoblastic leukemia cells by an IgA2 variant of daratumumab. Front Immunol 2022; 13:949140. [PMID: 36052078 PMCID: PMC9427194 DOI: 10.3389/fimmu.2022.949140] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 07/26/2022] [Indexed: 11/13/2022] Open
Abstract
Antibody-based immunotherapy is increasingly employed to treat acute lymphoblastic leukemia (ALL) patients. Many T-ALL cells express CD38 on their surface, which can be targeted by the CD38 antibody daratumumab (DARA), approved for the treatment of multiple myeloma. Tumor cell killing by myeloid cells is relevant for the efficacy of many therapeutic antibodies and can be more efficacious with human IgA than with IgG antibodies. This is demonstrated here by investigating antibody-dependent cellular phagocytosis (ADCP) by macrophages and antibody-dependent cell-mediated cytotoxicity (ADCC) by polymorphonuclear (PMN) cells using DARA (human IgG1) and an IgA2 isotype switch variant (DARA-IgA2) against T-ALL cell lines and primary patient-derived tumor cells. ADCP and ADCC are negatively regulated by interactions between CD47 on tumor cells and signal regulatory protein alpha (SIRPα) on effector cells. In order to investigate the impact of this myeloid checkpoint on T-ALL cell killing, CD47 and glutaminyl-peptide cyclotransferase like (QPCTL) knock-out T-ALL cells were employed. QPTCL is an enzymatic posttranslational modifier of CD47 activity, which can be targeted by small molecule inhibitors. Additionally, we used an IgG2σ variant of the CD47 blocking antibody magrolimab, which is in advanced clinical development. Moreover, treatment of T-ALL cells with all-trans retinoic acid (ATRA) increased CD38 expression leading to further enhanced ADCP and ADCC, particularly when DARA-IgA2 was applied. These studies demonstrate that myeloid checkpoint blockade in combination with IgA2 variants of CD38 antibodies deserves further evaluation for T-ALL immunotherapy.
Collapse
Affiliation(s)
- Niklas Baumann
- Division of Stem Cell Transplantation and Immunotherapy, Department of Medicine II, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Christian Arndt
- Division of Stem Cell Transplantation and Immunotherapy, Department of Medicine II, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Judith Petersen
- Division of Stem Cell Transplantation and Immunotherapy, Department of Medicine II, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Marta Lustig
- Division of Stem Cell Transplantation and Immunotherapy, Department of Medicine II, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Thies Rösner
- Division of Stem Cell Transplantation and Immunotherapy, Department of Medicine II, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Katja Klausz
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Christian- Albrechts-University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Christian Kellner
- Division of Transfusion Medicine, Cell Therapeutics and Haemostaseology, University Hospital, LMU Munich, Munich, Germany
| | - Miriam Bultmann
- Department of Medicine II, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Lorenz Bastian
- Department of Medicine II, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Fotini Vogiatzi
- Pediatric Hematology/Oncology, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Jeanette H. W. Leusen
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Renate Burger
- Division of Stem Cell Transplantation and Immunotherapy, Department of Medicine II, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Denis M. Schewe
- Children’s Hospital, University Medical Center Magdeburg, Magdeburg, Germany
| | - Matthias Peipp
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Christian- Albrechts-University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Thomas Valerius
- Division of Stem Cell Transplantation and Immunotherapy, Department of Medicine II, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein, Kiel, Germany
- *Correspondence: Thomas Valerius,
| |
Collapse
|
23
|
Combining daratumumab with CD47 blockade prolongs survival in preclinical models of pediatric T-ALL. Blood 2022; 140:45-57. [PMID: 35452517 DOI: 10.1182/blood.2021014485] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 04/18/2022] [Indexed: 11/20/2022] Open
Abstract
Acute lymphoblastic leukemia (ALL) is the most common malignant disease affecting children. Although therapy strategies improved, T-cell acute lymphoblastic leukemia (T-ALL) relapse is associated with chemoresistance and a poor prognosis. One strategy to overcome this obstacle is the application of monoclonal antibodies. Here, we show that leukemic cells from T-ALL patients express surface CD38 and CD47, both attractive targets for antibody therapy. We therefore investigated the commercially available CD38 antibody daratumumab (Dara) in combination with a proprietary modified CD47 antibody (Hu5F9-IgG2σ) in vitro and in vivo. Compared to single treatments, this combination significantly increased in vitro antibody-dependent cellular phagocytosis (ADCP) in T-ALL cell lines as well as in random de novo and in relapsed/refractory T-ALL patient derived xenograft (PDX) samples. Similarly, enhanced ADCP was observed when combining Dara with pharmacological inhibition of CD47 interactions using a glutaminyl cyclase inhibitor. Phase II-like preclinical in vivo trials using T-ALL PDX samples in experimental minimal residual disease like (MRD-like) and overt leukemia models revealed a high anti-leukemic efficacy of CD47 blockade alone. However, T-ALL xenograft mice subjected to chemotherapy first (post-chemo MRD) and subsequently co-treated with Dara and Hu5F9-IgG2σ displayed significantly reduced bone marrow infiltration as compared to single treatments. In relapsed and highly refractory T-ALL PDX combined treatment with Dara and Hu5F9-IgG2σ was required to substantially prolong survival as compared to single treatments. These findings suggest that combining CD47 blockade with Dara is a promising therapy for T-ALL, especially for relapsed/refractory disease harbouring a dismal prognosis in patients.
Collapse
|
24
|
Nguyen J, Pettmann J, Kruger P, Dushek O. Quantitative contributions of TNF receptor superfamily members to CD8 + T-cell responses. Mol Syst Biol 2021; 17:e10560. [PMID: 34806839 PMCID: PMC8607805 DOI: 10.15252/msb.202110560] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 11/09/2022] Open
Abstract
T-cell responses to infections and cancers are regulated by co-signalling receptors grouped into the binary categories of co-stimulation or co-inhibition. The co-stimulation TNF receptor superfamily (TNFRSF) members 4-1BB, CD27, GITR and OX40 have similar signalling mechanisms raising the question of whether they have similar impacts on T-cell responses. Here, we screened for the quantitative impact of these TNFRSFs on primary human CD8+ T-cell cytokine production. Although both 4-1BB and CD27 increased production, only 4-1BB was able to prolong the duration over which cytokine was produced, and both had only modest effects on antigen sensitivity. An operational model explained these different phenotypes using shared signalling based on the surface expression of 4-1BB being regulated through signalling feedback. The model predicted and experiments confirmed that CD27 co-stimulation increases 4-1BB expression and subsequent 4-1BB co-stimulation. GITR and OX40 displayed only minor effects on their own but, like 4-1BB, CD27 could enhance GITR expression and subsequent GITR co-stimulation. Thus, different co-stimulation receptors can have different quantitative effects allowing for synergy and fine-tuning of T-cell responses.
Collapse
Affiliation(s)
- John Nguyen
- SirWilliam Dunn School of PathologyUniversity of OxfordOxfordUK
| | | | - Philipp Kruger
- SirWilliam Dunn School of PathologyUniversity of OxfordOxfordUK
| | - Omer Dushek
- SirWilliam Dunn School of PathologyUniversity of OxfordOxfordUK
| |
Collapse
|