1
|
Levengood MR, Carosino CM, Zhang X, Lucas S, Ortiz DJ, Westendorf L, Chin AP, Martin AD, Wong A, Hengel SM, Sun H, Zeng W, Yumul R, Dominguez MM, Chen Y, Zheng JH, Karlsson CA, Trang VH, Senter PD, Gardai SJ. Preclinical Development of SGN-CD47M: Protease-Activated Antibody Technology Enables Selective Tumor Targeting of the Innate Immune Checkpoint Receptor CD47. Mol Cancer Ther 2025; 24:471-484. [PMID: 39463068 PMCID: PMC11962404 DOI: 10.1158/1535-7163.mct-24-0371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 07/26/2024] [Accepted: 09/27/2024] [Indexed: 10/29/2024]
Abstract
CD47 is a cell-surface glycoprotein that is expressed on normal human tissues and plays a key role as a marker of self. Tumor cells have co-opted CD47 overexpression to evade immune surveillance, and thus blockade of CD47 is a highly active area of clinical exploration in oncology. However, clinical development of CD47-targeted agents has been complicated by its robust expression in normal tissues and the toxicities that arise from blocking this inhibitory signal. Furthermore, pro-phagocytic signals are not uniformly expressed in tumors, and antibody blockade alone is often not sufficient to drive antitumor activity. The inclusion of an IgG1 antibody backbone into therapeutic design has been shown to not only serve as an additional pro-phagocytic signal but also exacerbate toxicities in normal tissues. Therefore, a need persists for more selective therapeutic modalities targeting CD47. To address these challenges, we developed SGN-CD47M, a humanized anti-CD47 IgG1 mAb linked to novel masking peptides through linkers designed to be cleaved by active proteases enriched in the tumor microenvironment (TME). Masking technology has the potential to increase the amount of drug that reaches the TME while concomitantly reducing systemic toxicities. We demonstrate that SGN-CD47M is well tolerated in cynomolgus monkeys and displays a 20-fold improvement in tolerability to hematologic toxicities when compared with the unmasked antibody. SGN-CD47M also displays preferential activation in the TME that leads to robust single-agent antitumor activity. For these reasons, SGN-CD47M may have enhanced antitumor activity and improved tolerability relative to existing therapies that target the CD47-signal regulatory protein α interaction.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Hao Sun
- Pfizer, Inc., Bothell, Washington
| | | | | | | | | | | | | | | | | | | |
Collapse
|
2
|
Bess SN, Igoe MJ, Muldoon TJ. The Physiological and Therapeutic Role of CD47 in Macrophage Function and Cancer. Immunol Invest 2025; 54:112-146. [PMID: 39415597 PMCID: PMC11774679 DOI: 10.1080/08820139.2024.2415409] [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: 10/19/2024]
Abstract
BACKGROUND Immunotherapy is an emerging strategy in cancer therapeutics aimed at modulating the immune system to inhibit pro-tumor pathways and increase a tumor's sensitivity to chemotherapy. Several clinically approved immunotherapy treatments, such as monoclonal antibody treatments, have been successful in solid tumors such as breast, colorectal, and pancreatic. However, an outstanding challenge of these strategies is tumor cell resistance. One target of interest for immune cell modulation is targeting macrophages that enter the tumor microenvironment. More specifically, an immune checkpoint of interest is CD47. CD47 is a transmembrane protein that inhibits phagocytic activity by acting as a "don't eat me" signal. In both mice and humans, healthy cells can express CD47, while solid malignancies like colorectal and breast cancer express it most strongly. METHODS Analysis of literature data on the physiological and functional roles of tissue-resident macrophages, along with the structure and mechanisms of action of the CD47 pathway was explored. We also explored how CD47 can influence different aspects of the tumor microenvironment (i.e. cellular metabolism and hypoxia) in addition to current clinical strategies and challenges associated with targeting CD47. RESULTS Overall, it was discovered that CD47 is overexpressed in a variety of cancer types in addition to normal tissue, making it a promising treatment regimen to enhance the capability of macrophages to phagocytose tumor cells. However, treatment efficacy is varied in pre-clinical and clinical models due to various challenges such as off-target effects. CONCLUSION This review emphasizes the diverse functionality of macrophages in normal and cancerous tissue, while also emphasizing the importance of macrophage targeting and their clinical significance.
Collapse
Affiliation(s)
- Shelby N. Bess
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR 72701
| | - Matthew J. Igoe
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR 72701
| | - Timothy J. Muldoon
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR 72701
| |
Collapse
|
3
|
Rakoczy K, Kaczor J, Sołtyk A, Szymańska N, Stecko J, Drąg-Zalesińska M, Kulbacka J. The Immune Response of Cancer Cells in Breast and Gynecologic Neoplasms. Int J Mol Sci 2024; 25:6206. [PMID: 38892394 PMCID: PMC11172873 DOI: 10.3390/ijms25116206] [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: 05/16/2024] [Revised: 05/29/2024] [Accepted: 06/03/2024] [Indexed: 06/21/2024] Open
Abstract
Cancer diseases constitute a major health problem which leads to the death of millions of people annually. They are unique among other diseases because cancer cells can perfectly adapt to the environment that they create themselves. This environment is usually highly hostile and for normal cells it would be hugely difficult to survive, however neoplastic cells not only can survive but also manage to proliferate. One of the reasons is that they can alter immunological pathways which allow them to be flexible and change their phenotype to the one needed in specific conditions. The aim of this paper is to describe some of these immunological pathways that play significant roles in gynecologic neoplasms as well as review recent research in this field. It is of high importance to possess extensive knowledge about these processes, as greater understanding leads to creating more specialized therapies which may prove highly effective in the future.
Collapse
Affiliation(s)
- Katarzyna Rakoczy
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (K.R.); (J.K.); (A.S.); (N.S.); (J.S.)
| | - Justyna Kaczor
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (K.R.); (J.K.); (A.S.); (N.S.); (J.S.)
| | - Adam Sołtyk
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (K.R.); (J.K.); (A.S.); (N.S.); (J.S.)
| | - Natalia Szymańska
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (K.R.); (J.K.); (A.S.); (N.S.); (J.S.)
| | - Jakub Stecko
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (K.R.); (J.K.); (A.S.); (N.S.); (J.S.)
| | - Małgorzata Drąg-Zalesińska
- Department of Human Morphology and Embryology, Division of Histology and Embryology, Faculty of Medicine, Wroclaw Medical University, T. Chalubińskiego 6a, 50-368 Wroclaw, Poland;
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211a, 50-556 Wroclaw, Poland
- Department of Immunology and Bioelectrochemistry, State Research Institute Centre for Innovative Medicine Santariškių g. 5, LT-08406 Vilnius, Lithuania
| |
Collapse
|
4
|
Yin N, Li X, Zhang X, Xue S, Cao Y, Niedermann G, Lu Y, Xue J. Development of pharmacological immunoregulatory anti-cancer therapeutics: current mechanistic studies and clinical opportunities. Signal Transduct Target Ther 2024; 9:126. [PMID: 38773064 PMCID: PMC11109181 DOI: 10.1038/s41392-024-01826-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 03/25/2024] [Accepted: 03/28/2024] [Indexed: 05/23/2024] Open
Abstract
Immunotherapy represented by anti-PD-(L)1 and anti-CTLA-4 inhibitors has revolutionized cancer treatment, but challenges related to resistance and toxicity still remain. Due to the advancement of immuno-oncology, an increasing number of novel immunoregulatory targets and mechanisms are being revealed, with relevant therapies promising to improve clinical immunotherapy in the foreseeable future. Therefore, comprehending the larger picture is important. In this review, we analyze and summarize the current landscape of preclinical and translational mechanistic research, drug development, and clinical trials that brought about next-generation pharmacological immunoregulatory anti-cancer agents and drug candidates beyond classical immune checkpoint inhibitors. Along with further clarification of cancer immunobiology and advances in antibody engineering, agents targeting additional inhibitory immune checkpoints, including LAG-3, TIM-3, TIGIT, CD47, and B7 family members are becoming an important part of cancer immunotherapy research and discovery, as are structurally and functionally optimized novel anti-PD-(L)1 and anti-CTLA-4 agents and agonists of co-stimulatory molecules of T cells. Exemplified by bispecific T cell engagers, newly emerging bi-specific and multi-specific antibodies targeting immunoregulatory molecules can provide considerable clinical benefits. Next-generation agents also include immune epigenetic drugs and cytokine-based therapeutics. Cell therapies, cancer vaccines, and oncolytic viruses are not covered in this review. This comprehensive review might aid in further development and the fastest possible clinical adoption of effective immuno-oncology modalities for the benefit of patients.
Collapse
Affiliation(s)
- Nanhao Yin
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center & State Key Laboratory of Biotherapy, and The National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Chengdu, 610041, Sichuan, PR China
| | - Xintong Li
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center & State Key Laboratory of Biotherapy, and The National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Chengdu, 610041, Sichuan, PR China
| | - Xuanwei Zhang
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center & State Key Laboratory of Biotherapy, and The National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Chengdu, 610041, Sichuan, PR China
| | - Shaolong Xue
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, No. 20, Section 3, South Renmin Road, Chengdu, 610041, Sichuan, PR China
| | - Yu Cao
- Department of Emergency Medicine, Laboratory of Emergency Medicine, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Chengdu, 610041, Sichuan, PR China
- Institute of Disaster Medicine & Institute of Emergency Medicine, Sichuan University, No. 17, Gaopeng Avenue, Chengdu, 610041, Sichuan, PR China
| | - Gabriele Niedermann
- Department of Radiation Oncology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK) Partner Site DKTK-Freiburg, Robert-Koch-Strasse 3, 79106, Freiburg, Germany.
| | - You Lu
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center & State Key Laboratory of Biotherapy, and The National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Chengdu, 610041, Sichuan, PR China.
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, No. 2222, Xinchuan Road, Chengdu, 610041, Sichuan, PR China.
| | - Jianxin Xue
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center & State Key Laboratory of Biotherapy, and The National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Chengdu, 610041, Sichuan, PR China.
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, No. 2222, Xinchuan Road, Chengdu, 610041, Sichuan, PR China.
| |
Collapse
|
5
|
Pereira MP, Herrity E, Kim DDH. TP53-mutated acute myeloid leukemia and myelodysplastic syndrome: biology, treatment challenges, and upcoming approaches. Ann Hematol 2024; 103:1049-1067. [PMID: 37770618 DOI: 10.1007/s00277-023-05462-5] [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: 05/11/2023] [Accepted: 09/17/2023] [Indexed: 09/30/2023]
Abstract
Improved understanding of TP53 biology and the clinicopathological features of TP53-mutated myeloid neoplasms has led to the recognition of TP53-mutated acute myeloid leukemia/myelodysplastic syndrome (TP53m AML/MDS) as a unique entity, characterized by dismal outcomes following conventional therapies. Several clinical trials have investigated combinations of emerging therapies for these patients with the poorest molecular prognosis among myeloid neoplasms. Although some emerging therapies have shown improvement in overall response rates, this has not translated into better overall survival, hence the notion that p53 remains an elusive target. New therapeutic strategies, including novel targeted therapies, immune checkpoint inhibitors, and monoclonal antibodies, represent a shift away from cytotoxic and hypomethylating-based therapies, towards approaches combining non-immune and novel immune therapeutic strategies. The triple combination of azacitidine and venetoclax with either magrolimab or eprenetapopt have demonstrated safety in early trials, with phase III trials currently underway, and promising interim clinical results. This review compiles background on TP53 biology, available and emerging therapies along with their mechanisms of action for the TP53m disease entity, current treatment challenges, and recently published data and status of ongoing clinical trials for TP53m AML/MDS.
Collapse
Affiliation(s)
- Mariana Pinto Pereira
- Hans Messner Allogeneic Blood and Marrow Transplantation Program, Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, M5G2M9, Toronto, ON, Canada
| | - Elizabeth Herrity
- Hans Messner Allogeneic Blood and Marrow Transplantation Program, Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, M5G2M9, Toronto, ON, Canada
| | - Dennis D H Kim
- Hans Messner Allogeneic Blood and Marrow Transplantation Program, Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, M5G2M9, Toronto, ON, Canada.
- Leukemia Program, Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada.
- Department of Hematology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
6
|
Wang T, Wang SQ, Du YX, Sun DD, Liu C, Liu S, Sun YY, Wang HL, Zhang CS, Liu HL, Jin L, Chen XP. Gentulizumab, a novel anti-CD47 antibody with potent antitumor activity and demonstrates a favorable safety profile. J Transl Med 2024; 22:220. [PMID: 38429732 PMCID: PMC10905820 DOI: 10.1186/s12967-023-04710-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 11/08/2023] [Indexed: 03/03/2024] Open
Abstract
BACKGROUND Targeting CD47/SIRPα axis has emerged as a promising strategy in cancer immunotherapy. Despite the encouraging clinical efficacy observed in hematologic malignancies through CD47-SIRPα blockade, there are safety concerns related to the binding of anti-CD47 antibodies to CD47 on the membrane of peripheral blood cells. METHODS In order to enhance the selectivity and therapeutic efficacy of the antibody, we developed a humanized anti-CD47 monoclonal antibody called Gentulizumab (GenSci059). The binding capacity of GenSci059 to CD47 was evaluated using flow cytometry and surface plasmon resonance (SPR) methods, the inhibitory effect of GenSci059 on the CD47-SIRPα interaction was evaluated through competitive ELISA assays. The anti-tumor activity of GenSci059 was assessed using in vitro macrophage models and in vivo patient-derived xenograft (PDX) models. To evaluate the safety profile of GenSci059, binding assays were conducted using blood cells. Additionally, we investigated the underlying mechanisms contributing to the weaker binding of GenSci059 to erythrocytes. Finally, toxicity studies were performed in non-human primates to assess the potential risks associated with GenSci059. RESULTS GenSci059 displayed strong binding to CD47 in both human and monkey, and effectively inhibited the CD47-SIRPα interaction. With doses ranging from 5 to 20 mg/kg, GenSci059 demonstrated potent inhibition of the growth of subcutaneous tumor with the inhibition rates ranged from 30.3% to complete regression. Combination of GenSci059 with 2.5 mg/kg Rituximab at a dose of 2.5 mg/kg showed enhanced tumor inhibition compared to monotherapy, exhibiting synergistic effects. GenSci059 exhibited minimal binding to hRBCs compared to Hu5F9-G4. The binding of GenSci059 to CD47 depended on the cyclization of N-terminal pyroglutamic acid and the spatial conformation of CD47, but was not affected by its glycosylation modifications. A maximum tolerated dose (MTD) of 450 mg/kg was observed for GenSci059, and no significant adverse effects were observed in repeated dosages up to 10 + 300 mg/kg, indicating a favorable safety profile. CONCLUSION GenSci059 selectively binds to CD47, effectively blocks the CD47/SIRPα axis signaling pathway and enhances the phagocytosis effects of macrophages toward tumor cells. This monoclonal antibody demonstrates potent antitumor activity and exhibits a favorable safety profile, positioning it as a promising and effective therapeutic option for cancer.
Collapse
Affiliation(s)
- Tao Wang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
- GeneScience Pharmaceuticals Co., Ltd, Changchun, 130012, Jilin, People's Republic of China
| | - Si-Qin Wang
- GeneScience Pharmaceuticals Co., Ltd, Changchun, 130012, Jilin, People's Republic of China
| | - Yin-Xiao Du
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Dan-Dan Sun
- GeneScience Pharmaceuticals Co., Ltd, Changchun, 130012, Jilin, People's Republic of China
| | - Chang Liu
- GeneScience Pharmaceuticals Co., Ltd, Changchun, 130012, Jilin, People's Republic of China
| | - Shuang Liu
- GeneScience Pharmaceuticals Co., Ltd, Changchun, 130012, Jilin, People's Republic of China
| | - Ying-Ying Sun
- GeneScience Pharmaceuticals Co., Ltd, Changchun, 130012, Jilin, People's Republic of China
| | - Hai-Long Wang
- GeneScience Pharmaceuticals Co., Ltd, Changchun, 130012, Jilin, People's Republic of China
| | - Chun-Sheng Zhang
- GeneScience Pharmaceuticals Co., Ltd, Changchun, 130012, Jilin, People's Republic of China
| | - Hai-Long Liu
- GeneScience Pharmaceuticals Co., Ltd, Changchun, 130012, Jilin, People's Republic of China
| | - Lei Jin
- GeneScience Pharmaceuticals Co., Ltd, Changchun, 130012, Jilin, People's Republic of China.
| | - Xiao-Ping Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.
| |
Collapse
|
7
|
Zhao P, Xie L, Yu L, Wang P. Targeting CD47-SIRPα axis for Hodgkin and non-Hodgkin lymphoma immunotherapy. Genes Dis 2024; 11:205-217. [PMID: 37588232 PMCID: PMC10425755 DOI: 10.1016/j.gendis.2022.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 11/22/2022] [Accepted: 12/05/2022] [Indexed: 01/12/2023] Open
Abstract
The interaction between cluster of differentiation 47 (CD47) and signal regulatory protein α (SIRPα) protects healthy cells from macrophage attack, which is crucial for maintaining immune homeostasis. Overexpression of CD47 occurs widely across various tumor cell types and transmits the "don't eat me" signal to macrophages to avoid phagocytosis through binding to SIRPα. Blockade of the CD47-SIRPα axis is therefore a promising approach for cancer treatment. Lymphoma is the most common hematological malignancy and is an area of unmet clinical need. This review mainly described the current strategies targeting the CD47-SIRPα axis, including antibodies, SIRPα Fc fusion proteins, small molecule inhibitors, and peptides both in preclinical studies and clinical trials with Hodgkin lymphoma and non-Hodgkin lymphoma.
Collapse
Affiliation(s)
- Pengcheng Zhao
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong 255000, China
| | - Longyan Xie
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Lei Yu
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Ping Wang
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong 255000, China
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| |
Collapse
|
8
|
Zhu D, Hadjivassiliou H, Jennings C, Mikolon D, Ammirante M, Acharya S, Lloyd J, Abbasian M, Narla RK, Piccotti JR, Stamp K, Cho H, Hariharan K. CC-96673 (BMS-986358), an affinity-tuned anti-CD47 and CD20 bispecific antibody with fully functional fc, selectively targets and depletes non-Hodgkin's lymphoma. MAbs 2024; 16:2310248. [PMID: 38349008 PMCID: PMC10865928 DOI: 10.1080/19420862.2024.2310248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 01/22/2024] [Indexed: 02/15/2024] Open
Abstract
Cluster of differentiation 47 (CD47) is a transmembrane protein highly expressed in tumor cells that interacts with signal regulatory protein alpha (SIRPα) and triggers a "don't eat me" signal to the macrophage, inhibiting phagocytosis and enabling tumor escape from immunosurveillance. The CD47-SIRPα axis has become an important target for cancer immunotherapy. To date, the advancement of CD47-targeted modalities is hindered by the ubiquitous expression of the target, often leading to rapid drug elimination and hematologic toxicity including anemia. To overcome those challenges a bispecific approach was taken. CC-96673, a humanized IgG1 bispecific antibody co-targeting CD47 and CD20, is designed to bind CD20 with high affinity and CD47 with optimally lowered affinity. As a result of the detuned CD47 affinity, CC-96673 selectively binds to CD20-expressing cells, blocking the interaction of CD47 with SIRPα. This increased selectivity of CC-96673 over monospecific anti-CD47 approaches allows for the use of wild-type IgG1 Fc, which engages activating crystallizable fragment gamma receptors (FcγRs) to fully potentiate macrophages to engulf and destroy CD20+ cells, while sparing CD47+CD20- normal cells. The combined targeting of anti-CD20 and anti-CD47 results in enhanced anti- tumor activity compared to anti-CD20 targeting antibodies alone. Furthermore, preclinical studies have demonstrated that CC-96673 exhibits acceptable pharmacokinetic properties with a favorable toxicity profile in non-human primates. Collectively, these findings define CC-96673 as a promising CD47 × CD20 bispecific antibody that selectively destroys CD20+ cancer cells via enhanced phagocytosis and other effector functions.
Collapse
Affiliation(s)
- Dan Zhu
- Department of Discovery Biotherapeutics, Bristol Myers Squibb, San Diego, CA, USA
| | | | - Catherine Jennings
- Department of Discovery Biotherapeutics, Bristol Myers Squibb, San Diego, CA, USA
| | - David Mikolon
- Department of Discovery Biotherapeutics, Bristol Myers Squibb, San Diego, CA, USA
| | - Massimo Ammirante
- Oncogenesis Thematic Research Center, Bristol Myers Squibb, San Diego, CA, USA
| | - Sharmistha Acharya
- Department of Discovery Biotherapeutics, Bristol Myers Squibb, San Diego, CA, USA
| | - Jon Lloyd
- Department of Discovery Biotherapeutics, Bristol Myers Squibb, San Diego, CA, USA
| | - Mahan Abbasian
- Department of Discovery Biotherapeutics, Bristol Myers Squibb, San Diego, CA, USA
| | - Rama Krishna Narla
- Oncogenesis Thematic Research Center, Bristol Myers Squibb, San Diego, CA, USA
| | - Joseph R. Piccotti
- Department of Nonclinical Development, Bristol Myers Squibb, San Diego, CA, USA
| | - Katie Stamp
- Department of Nonclinical Development, Bristol Myers Squibb, San Diego, CA, USA
| | - Ho Cho
- Department of Discovery Biotherapeutics, Bristol Myers Squibb, San Diego, CA, USA
| | - Kandasamy Hariharan
- Department of Discovery Biotherapeutics, Bristol Myers Squibb, San Diego, CA, USA
| |
Collapse
|
9
|
Mustafa B, Fetse J, Kandel S, Lin CY, Adhikary P, Mamani UF, Liu Y, Ibrahim MN, Alahmari M, Cheng K. Discovery of Anti-CD47 Peptides as Innate Immune Checkpoint Inhibitors. ADVANCED THERAPEUTICS 2023; 6:2300114. [PMID: 38655206 PMCID: PMC11034909 DOI: 10.1002/adtp.202300114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Indexed: 04/26/2024]
Abstract
Cancer immunotherapy targeting adaptive immune cells has been attracting considerable interest due to its great success in treating multiple cancers. Recently, there is also increasing interest in agents that can stimulate innate immune cell activities. Immune checkpoint inhibitors targeting innate immune cells can block inhibitory interactions ('don't eat me' signals) between tumor cells and phagocytes. CD47 is a transmembrane protein overexpressed in various cancers and acts as a potent 'do not eat me' signal that contributes to the immune evasion of cancer cells. Anti-CD47 peptides that can bind to CD47 and block CD47/SIRPα interaction were discovered using a novel phage display biopanning strategy. Anti-CD47 peptides enhanced the macrophage-mediated phagocytosis of NCI-H82 tumor cells in vitro. Unlike anti-CD47 antibodies, these peptides do not induce the agglutination of RBCs. Moreover, anti-CD47 peptides exhibit high specificity for MC-38 cancer cells expressing CD47. CMP-22 peptide showed the ability to increase the antitumor activity of doxorubicin and extends the survival of CT26 tumor-bearing mice. The discovered anti-CD47 peptides can be considered potential candidates for cancer immunotherapy by blocking the CD47/SIRPα interaction, especially in combination with chemotherapy, to elicit synergistic effects.
Collapse
Affiliation(s)
- Bahaa Mustafa
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - John Fetse
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - Sashi Kandel
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - Chien-Yu Lin
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - Pratik Adhikary
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - Umar-Farouk Mamani
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - Yanli Liu
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - Mohammed Nurudeen Ibrahim
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - Mohammed Alahmari
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - Kun Cheng
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| |
Collapse
|
10
|
Li M, Wang M, Wen Y, Zhang H, Zhao G, Gao Q. Signaling pathways in macrophages: molecular mechanisms and therapeutic targets. MedComm (Beijing) 2023; 4:e349. [PMID: 37706196 PMCID: PMC10495745 DOI: 10.1002/mco2.349] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 07/24/2023] [Accepted: 07/27/2023] [Indexed: 09/15/2023] Open
Abstract
Macrophages play diverse roles in development, homeostasis, and immunity. Accordingly, the dysfunction of macrophages is involved in the occurrence and progression of various diseases, such as coronavirus disease 2019 and atherosclerosis. The protective or pathogenic effect that macrophages exert in different conditions largely depends on their functional plasticity, which is regulated via signal transduction such as Janus kinase-signal transducer and activator of transcription, Wnt and Notch pathways, stimulated by environmental cues. Over the past few decades, the molecular mechanisms of signaling pathways in macrophages have been gradually elucidated, providing more alternative therapeutic targets for diseases treatment. Here, we provide an overview of the basic physiology of macrophages and expound the regulatory pathways within them. We also address the crucial role macrophages play in the pathogenesis of diseases, including autoimmune, neurodegenerative, metabolic, infectious diseases, and cancer, with a focus on advances in macrophage-targeted strategies exploring modulation of components and regulators of signaling pathways. Last, we discuss the challenges and possible solutions of macrophage-targeted therapy in clinical applications. We hope that this comprehensive review will provide directions for further research on therapeutic strategies targeting macrophage signaling pathways, which are promising to improve the efficacy of disease treatment.
Collapse
Affiliation(s)
- Ming Li
- Department of Gynecological OncologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- National Clinical Research Center for Obstetrics and GynecologyCancer Biology Research Center (Key Laboratory of the Ministry of Education)Tongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Mengjie Wang
- Department of Gynecological OncologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- National Clinical Research Center for Obstetrics and GynecologyCancer Biology Research Center (Key Laboratory of the Ministry of Education)Tongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yuanjia Wen
- Department of Gynecological OncologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- National Clinical Research Center for Obstetrics and GynecologyCancer Biology Research Center (Key Laboratory of the Ministry of Education)Tongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Hongfei Zhang
- Department of Gynecological OncologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- National Clinical Research Center for Obstetrics and GynecologyCancer Biology Research Center (Key Laboratory of the Ministry of Education)Tongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Guang‐Nian Zhao
- Department of Gynecological OncologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- National Clinical Research Center for Obstetrics and GynecologyCancer Biology Research Center (Key Laboratory of the Ministry of Education)Tongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Qinglei Gao
- Department of Gynecological OncologyTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- National Clinical Research Center for Obstetrics and GynecologyCancer Biology Research Center (Key Laboratory of the Ministry of Education)Tongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
11
|
Zeller T, Münnich IA, Windisch R, Hilger P, Schewe DM, Humpe A, Kellner C. Perspectives of targeting LILRB1 in innate and adaptive immune checkpoint therapy of cancer. Front Immunol 2023; 14:1240275. [PMID: 37781391 PMCID: PMC10533923 DOI: 10.3389/fimmu.2023.1240275] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 08/08/2023] [Indexed: 10/03/2023] Open
Abstract
Immune checkpoint blockade is a compelling approach in tumor immunotherapy. Blocking inhibitory pathways in T cells has demonstrated clinical efficacy in different types of cancer and may hold potential to also stimulate innate immune responses. A novel emerging potential target for immune checkpoint therapy is leukocyte immunoglobulin-like receptor subfamily B member 1 (LILRB1). LILRB1 belongs to the superfamily of leukocyte immunoglobulin-like receptors and exerts inhibitory functions. The receptor is expressed by a variety of immune cells including macrophages as well as certain cytotoxic lymphocytes and contributes to the regulation of different immune responses by interaction with classical as well as non-classical human leukocyte antigen (HLA) class I molecules. LILRB1 has gained increasing attention as it has been demonstrated to function as a phagocytosis checkpoint on macrophages by recognizing HLA class I, which represents a 'Don't Eat Me!' signal that impairs phagocytic uptake of cancer cells, similar to CD47. The specific blockade of the HLA class I:LILRB1 axis may provide an option to promote phagocytosis by macrophages and also to enhance cytotoxic functions of T cells and natural killer (NK) cells. Currently, LILRB1 specific antibodies are in different stages of pre-clinical and clinical development. In this review, we introduce LILRB1 and highlight the features that make this immune checkpoint a promising target for cancer immunotherapy.
Collapse
Affiliation(s)
- Tobias Zeller
- 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
| | - Denis M. Schewe
- Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, 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
| |
Collapse
|
12
|
Li SY, Guo YL, Tian JW, Zhang HJ, Li RF, Gong P, Yu ZL. Anti-Tumor Strategies by Harnessing the Phagocytosis of Macrophages. Cancers (Basel) 2023; 15:2717. [PMID: 37345054 DOI: 10.3390/cancers15102717] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 05/07/2023] [Accepted: 05/09/2023] [Indexed: 06/23/2023] Open
Abstract
Macrophages are essential for the human body in both physiological and pathological conditions, engulfing undesirable substances and participating in several processes, such as organism growth, immune regulation, and maintenance of homeostasis. Macrophages play an important role in anti-bacterial and anti-tumoral responses. Aberrance in the phagocytosis of macrophages may lead to the development of several diseases, including tumors. Tumor cells can evade the phagocytosis of macrophages, and "educate" macrophages to become pro-tumoral, resulting in the reduced phagocytosis of macrophages. Hence, harnessing the phagocytosis of macrophages is an important approach to bolster the efficacy of anti-tumor treatment. In this review, we elucidated the underlying phagocytosis mechanisms, such as the equilibrium among phagocytic signals, receptors and their respective signaling pathways, macrophage activation, as well as mitochondrial fission. We also reviewed the recent progress in the area of application strategies on the basis of the phagocytosis mechanism, including strategies targeting the phagocytic signals, antibody-dependent cellular phagocytosis (ADCP), and macrophage activators. We also covered recent studies of Chimeric Antigen Receptor Macrophage (CAR-M)-based anti-tumor therapy. Furthermore, we summarized the shortcomings and future applications of each strategy and look into their prospects with the hope of providing future research directions for developing the application of macrophage phagocytosis-promoting therapy.
Collapse
Affiliation(s)
- Si-Yuan Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Yong-Lin Guo
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Jia-Wen Tian
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - He-Jing Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Rui-Fang Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Ping Gong
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
- Department of Anesthesiology, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Zi-Li Yu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| |
Collapse
|
13
|
揭 晓, 孔 阳, 周 光. [Latest Findings on the Role of CD47 in Tumor Immune Evasion and Related Targeted Therapies]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2023; 54:455-461. [PMID: 37248568 PMCID: PMC10475431 DOI: 10.12182/20230560101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Indexed: 05/31/2023]
Abstract
CD47 is an immunoglobulin that is overexpressed on the surface of a variety of cancer cells. CD47 forms a signaling complex with signal regulatory protein alpha (SIRPα), prompting the escape of cancer cells from macrophage-mediated phagocytosis. In recent years, CD47 has been shown to be highly expressed in many types of solid tumors and is associated with poor prognosis in patients. More and more studies have shown that inhibition of the CD47-SIRPα signaling pathway can promote adaptive immune responses and enhance the phagocytosis of tumor cells by macrophages. Humanized anti-CD47 IgG4 monoclonal antibody has been studied in clinical trials for the treatment of a variety of advanced solid tumors and lymphomas, demonstrating a sound safety profile and achieving partial remission in some patients. In this review we discuss the structure and function of CD47 and the mechanism of CD47 regulation in tumors, summarize the research progress in therapeutic antibody drugs targeting CD47 and a bottleneck in research that targeted drugs are more prone to result in serious adverse effects, and evaluated the potential of the applying CD47-SIRPα signaling pathway in anti-cancer therapy.
Collapse
Affiliation(s)
- 晓亮 揭
- 国家癌症中心/中国医学科学院肿瘤医院 分子肿瘤学国家重点实验室 (北京 100021)State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences, Beijing 100021, China
| | - 阳阳 孔
- 国家癌症中心/中国医学科学院肿瘤医院 分子肿瘤学国家重点实验室 (北京 100021)State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences, Beijing 100021, China
| | - 光飚 周
- 国家癌症中心/中国医学科学院肿瘤医院 分子肿瘤学国家重点实验室 (北京 100021)State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences, Beijing 100021, China
| |
Collapse
|
14
|
Luo X, Shen Y, Huang W, Bao Y, Mo J, Yao L, Yuan L. Blocking CD47-SIRPα Signal Axis as Promising Immunotherapy in Ovarian Cancer. Cancer Control 2023; 30:10732748231159706. [PMID: 36826231 PMCID: PMC9969460 DOI: 10.1177/10732748231159706] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023] Open
Abstract
Among the three primary gynecological malignancies, ovarian cancer has the lowest incidence but the worst prognosis. Because of the poor prognosis of ovarian cancer patients treated with existing treatments, immunotherapy is emerging as a potentially ideal alternative to surgery, chemotherapy, and targeted therapy. Among immunotherapies, immune checkpoint inhibitors have been the most thoroughly studied, and many drugs have been successfully used in the clinic. CD47, a novel immune checkpoint, provides insights into ovarian cancer immunotherapy. This review highlights the mechanisms of tumor immune evasion via CD47-mediated inhibition of phagocytosis and provides a comprehensive insight into the progress of the relevant targeted agents in ovarian cancer.
Collapse
Affiliation(s)
- Xukai Luo
- Department of Gynecologic Oncology, Obstetrics and Gynecology Hospital of
Fudan University, Shanghai, China
| | - Yini Shen
- Department of Gynecologic Oncology, Obstetrics and Gynecology Hospital of
Fudan University, Shanghai, China
| | - Wu Huang
- Department of Gynecologic Oncology, Obstetrics and Gynecology Hospital of
Fudan University, Shanghai, China
| | - Yiting Bao
- Department of Gynecologic Oncology, Obstetrics and Gynecology Hospital of
Fudan University, Shanghai, China
| | - Jiahang Mo
- Department of Gynecologic Oncology, Obstetrics and Gynecology Hospital of
Fudan University, Shanghai, China
| | - Liangqing Yao
- Department of Gynecologic Oncology, Obstetrics and Gynecology Hospital of
Fudan University, Shanghai, China
| | - Lei Yuan
- Department of Gynecologic Oncology, Obstetrics and Gynecology Hospital of
Fudan University, Shanghai, China,Lei Yuan, MD, Obstetrics and Gynecology
Hospital, Fudan University, 419 Fangxie Road, Huangpu District, Shanghai 200011,
China.
| |
Collapse
|
15
|
Chang MR, Rusanov DA, Arakelyan J, Alshehri M, Asaturova AV, Kireeva GS, Babak MV, Ang WH. Targeting emerging cancer hallmarks by transition metal complexes: Cancer stem cells and tumor microbiome. Part I. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2022.214923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
|
16
|
De Re V, Repetto O, Mussolin L, Brisotto G, Elia C, Lopci E, d’Amore ESG, Burnelli R, Mascarin M. Promising drugs and treatment options for pediatric and adolescent patients with Hodgkin lymphoma. Front Cell Dev Biol 2022; 10:965803. [PMID: 36506094 PMCID: PMC9729954 DOI: 10.3389/fcell.2022.965803] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 11/07/2022] [Indexed: 11/25/2022] Open
Abstract
Currently-available therapies for newly-diagnosed pediatric and adolescent patients with Hodgkin lymphoma result in >95% survival at 5 years. Long-term survivors may suffer from long-term treatment-related side effects, however, so the past 20 years have seen clinical trials for children and adolescents with HL gradually abandon the regimens used in adults in an effort to improve this situation. Narrower-field radiotherapy can reduce long-term toxicity while maintaining good tumor control. Various risk-adapted chemo-radiotherapy strategies have been used. Early assessment of tumor response with interim positron emission tomography and/or measuring metabolic tumor volume has been used both to limit RT in patients with favorable characteristics and to adopt more aggressive therapies in patients with a poor response. Most classical Hodgkin's lymphoma relapses occur within 3 years of initial treatment, while relapses occurring 5 years or more after diagnosis are rare. As the outcome for patients with relapsed/refractory classical Hodgkin lymphoma remains unsatisfactory, new drugs have been proposed for its prevention or treatment. This review summarizes the important advances made in recent years in the management of pediatric and adolescent with classical Hodgkin lymphoma, and the novel targeted treatments for relapsed and refractory classical Hodgkin lymphoma.
Collapse
Affiliation(s)
- Valli De Re
- Immunopatologia e Biomarcatori Oncologici, Dipartimento di Ricerca e Diagnostica Avanzata dei Tumori, CRO Aviano, National Cancer Institute, Istituto di Ricovero e Cura a Carattere Scientifico, IRCCS, Aviano, Italy,*Correspondence: Valli De Re, ; Maurizio Mascarin,
| | - Ombretta Repetto
- Immunopatologia e Biomarcatori Oncologici, Dipartimento di Ricerca e Diagnostica Avanzata dei Tumori, CRO Aviano, National Cancer Institute, Istituto di Ricovero e Cura a Carattere Scientifico, IRCCS, Aviano, Italy
| | - Lara Mussolin
- Pediatric Hemato-Oncology Unit, Department of Women’s and Children’s Health, University of Padua, Padua, Italy
| | - Giulia Brisotto
- Immunopatologia e Biomarcatori Oncologici, Dipartimento di Ricerca e Diagnostica Avanzata dei Tumori, CRO Aviano, National Cancer Institute, Istituto di Ricovero e Cura a Carattere Scientifico, IRCCS, Aviano, Italy
| | - Caterina Elia
- AYA Oncology and Pediatric Radiotherapy Unit, Centro di Riferimento Oncologico IRCCS, Aviano, Italy
| | - Egesta Lopci
- Nuclear Medicine, IRCCS—Humanitas Research Hospital, Rozzano, MI, Italy
| | | | - Roberta Burnelli
- Pediatric Hematology-Oncology Unit, Azienda Ospedaliera Universitaria, Ospedale Sant’Anna, Ferrara, Italy
| | - Maurizio Mascarin
- AYA Oncology and Pediatric Radiotherapy Unit, Centro di Riferimento Oncologico IRCCS, Aviano, Italy,*Correspondence: Valli De Re, ; Maurizio Mascarin,
| |
Collapse
|
17
|
Qu T, Zhong T, Pang X, Huang Z, Jin C, Wang ZM, Li B, Xia Y. Ligufalimab, a novel anti-CD47 antibody with no hemagglutination demonstrates both monotherapy and combo antitumor activity. J Immunother Cancer 2022; 10:e005517. [PMID: 36450383 PMCID: PMC9717234 DOI: 10.1136/jitc-2022-005517] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/08/2022] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND CD47 is a widely expressed transmembrane glycoprotein that delivers an antiphagocytic signal on macrophages through its interaction with SIRPα. CD47 is highly expressed in cancer cells and its overexpression is correlated with poor prognosis. CD47 blocking antibodies are actively being developed worldwide for cancer therapy, and the most challenging concern is associated with hematotoxicity. Ligufalimab (AK117) is a novel humanized IgG4 anti-CD47 antibody without hemagglutination effect. Blockade of CD47-SIRPα pathway by AK117 leads to a promising therapeutic strategy for cancer treatment with unique safety features. METHODS AK117 was discovered through a screening hierarchy excluding hemagglutination. AK117 was characterized by detecting CD47-SIRPα blocking potential. Its effect on human red blood cells was examined and the mechanism of its binding with erythrocytes was studied. The abilities of AK117 and its combination with various opsonizing antibodies to promote macrophage-dependent phagocytosis of multiple human tumor cells were determined using fluorescence microscopy and flow cytometry. In vivo, the antitumor efficacy of AK117 monotherapy and combination with AK112 (an anti-PD-1/VEGF-A bispecific antibody) was assessed in a variety of xenograft models. Toxicologic studies were evaluated in non-human primates. RESULTS AK117 bound to CD47 with high affinity and blocked the CD47-SIRPα interaction. AK117 did not induce hemagglutination and showed significantly lower degree of erythrophagocytosis compared with Hu5F9-G4, and this mechanism of hemagglutination resistance might be related to the binding conformation. AK117 enhanced macrophage-mediated phagocytosis in both hematologic cancer and solid tumor cell lines as a single agent or in combination with cetuximab and rituximab in vitro, respectively. The antitumor effects of AK117 as a single agent or in combination with AK112 were also encouraging in various xenograft models. In non-human primates, AK117 showed less hematotoxicity compared with Hu5F9-G4. CONCLUSIONS AK117 eliminated hemagglutination and also enabled to maintain full effectiveness of CD47 blockade on tumor cells, which resulted in excellent antitumor efficacy and favorable safety profile of AK117. A series of clinical trials of AK117 as a therapeutic agent in combination with various agents such as AK112 are in progress for the treatment of multiple hematologic malignancies and solid tumors.
Collapse
Affiliation(s)
- Tailong Qu
- Research and Development Department, Akeso Biopharma Inc, Zhongshan, Guangdong, China
| | - Tingting Zhong
- Research and Development Department, Akeso Biopharma Inc, Zhongshan, Guangdong, China
| | - Xinghua Pang
- Research and Development Department, Akeso Biopharma Inc, Zhongshan, Guangdong, China
| | - Zhaoliang Huang
- Research and Development Department, Akeso Biopharma Inc, Zhongshan, Guangdong, China
| | - Chunshan Jin
- Research and Development Department, Akeso Biopharma Inc, Zhongshan, Guangdong, China
| | - Zhongmin Maxwell Wang
- Procurement and Sourcing Department and Clinical Operation Department, Akeso Biopharma Inc, Zhongshan, Guangdong, China
| | - Baiyong Li
- Research and Development Department, Akeso Biopharma Inc, Zhongshan, Guangdong, China
| | - Yu Xia
- President Office, Akeso Biopharma Inc, Zhongshan, Guangdong, China
| |
Collapse
|
18
|
Targeting CXCR4 and CD47 Receptors: An Overview of New and Old Molecules for a Biological Personalized Anticancer Therapy. Int J Mol Sci 2022; 23:ijms232012499. [PMID: 36293358 PMCID: PMC9604048 DOI: 10.3390/ijms232012499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/14/2022] [Accepted: 10/14/2022] [Indexed: 11/26/2022] Open
Abstract
Biological therapy, with its multifaceted applications, has revolutionized the treatment of tumors, mainly due to its ability to exclusively target cancer cells and reduce the adverse effects on normal tissues. This review focuses on the therapies targeting the CXCR4 and CD47 receptors. We surveyed the results of early clinical trials testing compounds classified as nonpeptides, small peptides, CXCR4 antagonists or specific antibodies whose activity reduces or completely blocks the intracellular signaling pathways and cell proliferation. We then examined antibodies and fusion proteins against CD47, the receptor that acts as a “do not eat me” signal to phagocytes escaping immune surveillance. Despite these molecules being tested in early clinical trials, some drawbacks are emerging that impair their use in practice. Finally, we examined the ImmunoGenic Surrender mechanism that involves crosstalk and co-internalization of CXCR4 and CD47 upon engagement of CXCR4 by ligands or other molecules. The favorable effect of such compounds is dual as CD47 surface reduction impact on the immune response adds to the block of CXCR4 proliferative potential. These results suggest that a combination of different therapeutic approaches has more beneficial effects on patients’ survival and may pave the way for new accomplishments in personalized anticancer therapy.
Collapse
|
19
|
Yue Y, Cao Y, Wang F, Zhang N, Qi Z, Mao X, Guo S, Li F, Guo Y, Lin Y, Dong W, Huang Y, Gu W. Bortezomib-resistant multiple myeloma patient-derived xenograft is sensitive to anti-CD47 therapy. Leuk Res 2022; 122:106949. [PMID: 36113267 DOI: 10.1016/j.leukres.2022.106949] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 08/28/2022] [Accepted: 09/02/2022] [Indexed: 12/17/2022]
Abstract
Multiple myeloma (MM) remains an incurable hematologic malignancy due to its frequent drug resistance and relapse. Cluster of Differentiation 47 (CD47) is reported to be highly expressed on MM cells, suggesting that the blockade of CD47 signaling pathway could be a potential therapeutic candidate for MM. In this study, we developed a bortezomib-resistant myeloma patient-derived xenograft (PDX) from an extramedullary pleural effusion myeloma patient sample. Notably, anti-CD47 antibody treatments significantly inhibited tumor growth not only in MM cell line-derived models, including MM.1S and NCI-H929, but also in the bortezomib-resistant MM PDX model. Flow cytometric data showed that anti-CD47 therapy promoted the polarization of tumor-associated macrophages from an M2- to an M1-like phenotype. In addition, anti-CD47 therapy decreased the expression of pro-angiogenic factors, increased the expression of anti-angiogenic factors, and improved tumor vascular function, suggesting that anti-CD47 therapy induces tumor vascular normalization. Taken together, these data show that anti-CD47 antibody therapy reconditions the tumor immune microenvironment and inhibits the tumor growth of bortezomib-resistant myeloma PDX. Our findings suggest that CD47 is a potential new target to treat bortezomib-resistant MM.
Collapse
Affiliation(s)
- Yanhua Yue
- Department of Hematology, The First People's Hospital of Changzhou, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, PR China; Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, Jiangsu Province, PR China
| | - Yang Cao
- Department of Hematology, The First People's Hospital of Changzhou, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, PR China
| | - Fei Wang
- Department of Hematology, The First People's Hospital of Changzhou, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, PR China
| | - Naidong Zhang
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, Jiangsu Province, PR China
| | - Ziwei Qi
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, Jiangsu Province, PR China
| | - Xunyuan Mao
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, Jiangsu Province, PR China
| | - Shuxin Guo
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, Jiangsu Province, PR China
| | - Feng Li
- Department of Hematology, The First People's Hospital of Changzhou, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, PR China
| | - Yanting Guo
- Department of Hematology, The First People's Hospital of Changzhou, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, PR China
| | - Yan Lin
- Department of Hematology, The First People's Hospital of Changzhou, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, PR China
| | - Weimin Dong
- Department of Hematology, The First People's Hospital of Changzhou, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, PR China
| | - Yuhui Huang
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, Jiangsu Province, PR China.
| | - Weiying Gu
- Department of Hematology, The First People's Hospital of Changzhou, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, PR China.
| |
Collapse
|
20
|
Bian HT, Shen YW, Zhou YD, Nagle DG, Guan YY, Zhang WD, Luan X. CD47: Beyond an immune checkpoint in cancer treatment. Biochim Biophys Acta Rev Cancer 2022; 1877:188771. [PMID: 35931392 DOI: 10.1016/j.bbcan.2022.188771] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 06/23/2022] [Accepted: 07/29/2022] [Indexed: 11/29/2022]
Abstract
The transmembrane protein, CD47, is recognized as an important innate immune checkpoint, and CD47-targeted drugs have been in development with the aim of inhibiting the interaction between CD47 and the regulatory glycoprotein SIRPα, for antitumor immunotherapy. Further, CD47 mediates other essential functions such as cell proliferation, caspase-independent cell death (CICD), angiogenesis and other integrin-activation-dependent cell phenotypic responses when bound to thrombospondin-1 (TSP-1) or other ligands. Mounting strategies that target CD47 have been developed in pre-clinical and clinical trials, including antibodies, small molecules, siRNAs, and peptides, and some of them have shown great promise in cancer treatment. Herein, the authors endeavor to provide a retrospective of ligand-mediated CD47 regulatory mechanisms, their roles in controlling antitumor intercellular and intracellular signal transduction, and an overview of CD47-targetd drug design.
Collapse
Affiliation(s)
- Hui-Ting Bian
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yi-Wen Shen
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yu-Dong Zhou
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Department of Chemistry and Biochemistry, College of Liberal Arts, University of Mississippi, University, MS, 38677-1848, USA
| | - Dale G Nagle
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Department of BioMolecular Sciences and Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University, MS 38677-1848, USA
| | - Ying-Yun Guan
- Department of Pharmacy, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China.
| | - Wei-Dong Zhang
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Xin Luan
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
21
|
Lv M, Liu Y, Liu W, Xing Y, Zhang S. Immunotherapy for Pediatric Acute Lymphoblastic Leukemia: Recent Advances and Future Perspectives. Front Immunol 2022; 13:921894. [PMID: 35769486 PMCID: PMC9234114 DOI: 10.3389/fimmu.2022.921894] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 05/19/2022] [Indexed: 11/13/2022] Open
Abstract
Pediatric acute lymphoblastic leukemia (ALL) is the most common subtype of childhood leukemia, which is characterized by the abnormal proliferation and accumulation of immature lymphoid cell in the bone marrow. Although the long-term survival rate for pediatric ALL has made significant progress over years with the development of contemporary therapeutic regimens, patients are still suffered from relapse, leading to an unsatisfactory outcome. Since the immune system played an important role in the progression and relapse of ALL, immunotherapy including bispecific T-cell engagers and chimeric antigen receptor T cells has been demonstrated to be capable of enhancing the immune response in pediatric patients with refractory or relapsed B-cell ALL, and improving the cure rate of the disease and patients’ quality of life, thus receiving the authorization for market. Nevertheless, the resistance and toxicities associated with the current immunotherapy remains a huge challenge. Novel therapeutic options to overcome the above disadvantages should be further explored. In this review, we will thoroughly discuss the emerging immunotherapeutics for the treatment of pediatric ALL, as well as side-effects and new development.
Collapse
Affiliation(s)
- Meng Lv
- Department of Pharmacy, Children’s Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Yan Liu
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wei Liu
- Department of Hematology Oncology, Children’s Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Yabing Xing
- Department of Pharmacy, Children’s Hospital Affiliated to Zhengzhou University, Zhengzhou, China
- *Correspondence: Yabing Xing, ; Shengnan Zhang,
| | - Shengnan Zhang
- Department of Pharmacy, Children’s Hospital Affiliated to Zhengzhou University, Zhengzhou, China
- *Correspondence: Yabing Xing, ; Shengnan Zhang,
| |
Collapse
|
22
|
Shallis RM, Bewersdorf JP, Stahl MF, Halene S, Zeidan AM. Are We Moving the Needle for Patients with TP53-Mutated Acute Myeloid Leukemia? Cancers (Basel) 2022; 14:2434. [PMID: 35626039 PMCID: PMC9140008 DOI: 10.3390/cancers14102434] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/06/2022] [Accepted: 05/11/2022] [Indexed: 12/12/2022] Open
Abstract
The currently available therapeutic options for patients with TP53-mutated acute myeloid leukemia (AML) are insufficient, as they translate to a median overall of only 6-9 months, and less than 10% of patients undergoing the most aggressive treatments, such as intensive induction therapy and allogeneic hematopoietic stem cell transplantation, will be cured. The lack of clear differences in outcomes with different treatments precludes the designation of a standard of care. Recently, there has been growing attention on this critical area of need by way of better understanding the biology of TP53 alterations and the disparities in outcomes among patients in this molecular subgroup, reflected in the development and testing of agents with novel mechanisms of action. Promising preclinical and efficacy data exist for therapies that are directed at the p53 protein rendered dysfunctional via mutation or that inhibit the CD47/SIRPα axis or other immune checkpoints such as TIM-3. In this review, we discuss recently attractive and emerging therapeutic agents, their preclinical rationale and the available clinical data as a monotherapy or in combination with the currently accepted backbones in frontline and relapsed/refractory settings for patients with TP53-mutated AML.
Collapse
Affiliation(s)
- Rory M. Shallis
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine and Yale Cancer Center, New Haven, CT 06520, USA; (R.M.S.); (S.H.)
| | - Jan P. Bewersdorf
- Division of Hematologic Malignancies, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - Maximilian F. Stahl
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA;
| | - Stephanie Halene
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine and Yale Cancer Center, New Haven, CT 06520, USA; (R.M.S.); (S.H.)
| | - Amer M. Zeidan
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine and Yale Cancer Center, New Haven, CT 06520, USA; (R.M.S.); (S.H.)
| |
Collapse
|
23
|
Chauchet X, Cons L, Chatel L, Daubeuf B, Didelot G, Moine V, Chollet D, Malinge P, Pontini G, Masternak K, Ferlin W, Buatois V, Shang L. CD47xCD19 bispecific antibody triggers recruitment and activation of innate immune effector cells in a B-cell lymphoma xenograft model. Exp Hematol Oncol 2022; 11:26. [PMID: 35538512 PMCID: PMC9088114 DOI: 10.1186/s40164-022-00279-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 04/15/2022] [Indexed: 12/31/2022] Open
Abstract
Background CD47/SIRPα axis is recognized as an innate immune checkpoint and emerging clinical data validate the interest of interrupting this pathway in cancer, particularly in hematological malignancies. In preclinical models, CD47/SIRPα blocking agents have been shown to mobilize phagocytic cells and trigger adaptive immune responses to eliminate tumors. Here, we describe the mechanisms afforded by a CD47xCD19 bispecific antibody (NI-1701) at controlling tumor growth in a mouse xenograft B-cell lymphoma model. Methods The contribution of immune effector cell subsets behind the antitumor activity of NI-1701 was investigated using flow cytometry, transcriptomic analysis, and in vivo immune-cell depletion experiments. Results We showed that NI-1701 treatment transformed the tumor microenvironment (TME) into a more anti-tumorigenic state with increased NK cells, monocytes, dendritic cells (DC) and MHCIIhi tumor-associated macrophages (TAMs) and decreased granulocytic myeloid-derived suppressor cells. Notably, molecular analysis of isolated tumor-infiltrating leukocytes following NI-1701 administration revealed an upregulation of genes linked to immune activation, including IFNγ and IL-12b. Moreover, TAM-mediated phagocytosis of lymphoma tumor cells was enhanced in the TME in the presence of NI-1701, highlighting the role of macrophages in tumor control. In vivo cell depletion experiments demonstrated that both macrophages and NK cells contribute to the antitumor activity. In addition, NI-1701 enhanced dendritic cell-mediated phagocytosis of tumor cells in vitro, resulting in an increased cross-priming of tumor-specific CD8 T cells. Conclusions The study described the mechanisms afforded by the CD47xCD19 bispecific antibody, NI-1701, at controlling tumor growth in lymphoma mouse model. NI-1701 is currently being evaluated in a Phase I clinical trial for the treatment of refractory or relapsed B-cell lymphoma (NCT04806035). Supplementary Information The online version contains supplementary material available at 10.1186/s40164-022-00279-w.
Collapse
Affiliation(s)
- Xavier Chauchet
- Light Chain Bioscience/Novimmune S.A, 15 Chemin du Pré-Fleuri, 1228, Plan-les-Ouates, Switzerland.
| | - Laura Cons
- Light Chain Bioscience/Novimmune S.A, 15 Chemin du Pré-Fleuri, 1228, Plan-les-Ouates, Switzerland
| | - Laurence Chatel
- Light Chain Bioscience/Novimmune S.A, 15 Chemin du Pré-Fleuri, 1228, Plan-les-Ouates, Switzerland
| | - Bruno Daubeuf
- Light Chain Bioscience/Novimmune S.A, 15 Chemin du Pré-Fleuri, 1228, Plan-les-Ouates, Switzerland
| | - Gérard Didelot
- Light Chain Bioscience/Novimmune S.A, 15 Chemin du Pré-Fleuri, 1228, Plan-les-Ouates, Switzerland
| | - Valéry Moine
- Light Chain Bioscience/Novimmune S.A, 15 Chemin du Pré-Fleuri, 1228, Plan-les-Ouates, Switzerland
| | - Didier Chollet
- iGE3 Genomics Platform, CMU-University of Geneva, Geneva, Switzerland
| | - Pauline Malinge
- Light Chain Bioscience/Novimmune S.A, 15 Chemin du Pré-Fleuri, 1228, Plan-les-Ouates, Switzerland
| | - Guillemette Pontini
- Light Chain Bioscience/Novimmune S.A, 15 Chemin du Pré-Fleuri, 1228, Plan-les-Ouates, Switzerland
| | - Krzysztof Masternak
- Light Chain Bioscience/Novimmune S.A, 15 Chemin du Pré-Fleuri, 1228, Plan-les-Ouates, Switzerland
| | - Walter Ferlin
- Light Chain Bioscience/Novimmune S.A, 15 Chemin du Pré-Fleuri, 1228, Plan-les-Ouates, Switzerland
| | - Vanessa Buatois
- Light Chain Bioscience/Novimmune S.A, 15 Chemin du Pré-Fleuri, 1228, Plan-les-Ouates, Switzerland
| | - Limin Shang
- Light Chain Bioscience/Novimmune S.A, 15 Chemin du Pré-Fleuri, 1228, Plan-les-Ouates, Switzerland
| |
Collapse
|
24
|
Abstract
CD47 is a "don't eat me" signal to phagocytes that is overexpressed on many tumor cells as a potential mechanism for immune surveillance evasion. CD47 and its interaction with signal-regulating protein alpha (SIRPα) on phagocytes is therefore a promising cancer target. Therapeutic antibodies and fusion proteins that block CD47 or SIRPα have been developed and have shown activity in preclinical models of hematologic and solid tumors. Anemia is a common adverse event associated with anti-CD47 treatment, but mitigation strategies-including use of a low 'priming' dose-have substantially reduced this risk in clinical studies. While efficacy in single-agent clinical studies is lacking, findings from studies of CD47-SIRPα blockade in combination with agents that increase 'eat me' signals or with antitumor antibodies are promising. Magrolimab, an anti-CD47 antibody, is the furthest along in clinical development among agents in this class. Magrolimab combination therapy in phase Ib/II studies has been well tolerated with encouraging response rates in hematologic and solid malignancies. Similar combination therapy studies with other anti-CD47-SIRPα agents are beginning to report. Based on these early clinical successes, many trials have been initiated in hematologic and solid tumors testing combinations of CD47-SIRPα blockade with standard therapies. The results of these studies will help determine the role of this novel approach in clinical practice and are eagerly awaited.
Collapse
Affiliation(s)
- R. Maute
- Gilead Sciences, Inc., Foster City, USA
| | - J. Xu
- Gilead Sciences, Inc., Foster City, USA
| | - I.L. Weissman
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, USA
| |
Collapse
|
25
|
Moradinasab S, Pourbagheri-Sigaroodi A, Ghaffari SH, Bashash D. Targeting macrophage-mediated tumor cell phagocytosis: An overview of phagocytosis checkpoints blockade, nanomedicine intervention, and engineered CAR-macrophage therapy. Int Immunopharmacol 2021; 103:108499. [PMID: 34972068 DOI: 10.1016/j.intimp.2021.108499] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/15/2021] [Accepted: 12/22/2021] [Indexed: 11/05/2022]
Abstract
Immunotherapy has been developing at an unprecedented speed with promising therapeutic outcomes in the wide spectrum of cancers. Up until now, most immunotherapies have focused on adaptive immunity; however, investigating the potential of macrophage phagocytosis and consequent adaptive immune cross-priming has led to a growing interest in exploiting macrophages in cancer therapy. In light of the positive evidence from preclinical studies and early clinical data, targeting macrophage phagocytosis has become a promising therapeutic strategy. Here, we review therapies based on harnessing and amplifying macrophage phagocytosis, such as blocking phagocytosis checkpoints and exploiting nanoparticles as efficient approaches in elevating macrophages-mediated phagocytosis. The present study introduces CAR-macrophage as the state-of-the-art modality serving as the bridge between the innate and adaptive immune system to mount a superior anti-tumor response in the treatment of cancer. We also take a look at the recent reports of therapies based on CAR-engineered macrophages with the hope of providing a future research direction for expanding the application of CAR-macrophage therapy.
Collapse
Affiliation(s)
- Susan Moradinasab
- Iranian Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Atieh Pourbagheri-Sigaroodi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed H Ghaffari
- Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
26
|
Billerhart M, Schönhofer M, Schueffl H, Polzer W, Pichler J, Decker S, Taschauer A, Maier J, Anton M, Eckmann S, Blaschek M, Heffeter P, Sami H, Ogris M. CD47-targeted cancer immunogene therapy: Secreted SIRPα-Fc fusion protein eradicates tumors by macrophage and NK cell activation. MOLECULAR THERAPY-ONCOLYTICS 2021; 23:192-204. [PMID: 34729396 PMCID: PMC8526499 DOI: 10.1016/j.omto.2021.09.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 09/22/2021] [Indexed: 12/15/2022]
Abstract
CD47 protects healthy cells from macrophage attack by binding to signal regulatory protein α (SIRPα), while its upregulation in cancer prevents immune clearance. Systemic treatment with CD47 antibodies requires a weakened Fc-mediated effector function or lower CD47-binding affinity to prevent side effects. Our approach combines “the best of both worlds,” i.e., maximized CD47 binding and full Fc-mediated immune activity, by exploiting gene therapy for paracrine release. We developed a plasmid vector encoding for the secreted fusion protein sCV1-hIgG1, comprising highly efficient CD47-blocking moiety CV1 and Fc domain of human immunoglobulin G1 (IgG1) with maximized immune activation. sCV1-hIgG1 exhibited a potent bystander effect, blocking CD47 on all cells via fusion protein secreted from only a fraction of cells or when transferring transfection supernatant to untransfected cells. The CpG-free plasmid ensured sustained secretion of sCV1-hIgG1. In orthotopic human triple-negative breast cancer in CB17-severe combined immunodeficiency (SCID) mice, ex vivo transfection significantly delayed tumor growth and eradicated one-third of tumors. In intratumoral transfection experiments, CD47 blockage and increased migration of macrophages into the tumor were observed within 17 h of a single injection. Natural killer (NK) cell-mediated lysis of sCV1-hIgG1-expressing cells was demonstrated in vitro. Taken together, this approach also opens the opportunity to block, in principle, any immune checkpoints.
Collapse
Affiliation(s)
- Magdalena Billerhart
- University of Vienna, Faculty of Life Sciences, Department of Pharmaceutical Sciences, Laboratory of MacroMolecular Cancer Therapeutics (MMCT), Althanstrasse 14, 1090 Vienna, Austria
| | - Monika Schönhofer
- University of Vienna, Faculty of Life Sciences, Department of Pharmaceutical Sciences, Laboratory of MacroMolecular Cancer Therapeutics (MMCT), Althanstrasse 14, 1090 Vienna, Austria
| | - Hemma Schueffl
- Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
| | - Wolfram Polzer
- University of Vienna, Faculty of Life Sciences, Department of Pharmaceutical Sciences, Laboratory of MacroMolecular Cancer Therapeutics (MMCT), Althanstrasse 14, 1090 Vienna, Austria
| | - Julia Pichler
- University of Vienna, Faculty of Life Sciences, Department of Pharmaceutical Sciences, Laboratory of MacroMolecular Cancer Therapeutics (MMCT), Althanstrasse 14, 1090 Vienna, Austria
| | - Simon Decker
- University of Vienna, Faculty of Life Sciences, Department of Pharmaceutical Sciences, Laboratory of MacroMolecular Cancer Therapeutics (MMCT), Althanstrasse 14, 1090 Vienna, Austria
| | - Alexander Taschauer
- University of Vienna, Faculty of Life Sciences, Department of Pharmaceutical Sciences, Laboratory of MacroMolecular Cancer Therapeutics (MMCT), Althanstrasse 14, 1090 Vienna, Austria
| | - Julia Maier
- University of Vienna, Faculty of Life Sciences, Department of Pharmaceutical Sciences, Laboratory of MacroMolecular Cancer Therapeutics (MMCT), Althanstrasse 14, 1090 Vienna, Austria
| | - Martina Anton
- Institutes of Molecular Immunology and Experimental Oncology, Klinikum rechts der Isar, Technische Universität München, Ismaninger Straße 22, 81675 Munich, Germany
| | - Sebastian Eckmann
- University of Vienna, Faculty of Life Sciences, Department of Pharmaceutical Sciences, Laboratory of MacroMolecular Cancer Therapeutics (MMCT), Althanstrasse 14, 1090 Vienna, Austria
| | - Manuel Blaschek
- University of Vienna, Faculty of Life Sciences, Department of Pharmaceutical Sciences, Laboratory of MacroMolecular Cancer Therapeutics (MMCT), Althanstrasse 14, 1090 Vienna, Austria
| | - Petra Heffeter
- Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
| | - Haider Sami
- University of Vienna, Faculty of Life Sciences, Department of Pharmaceutical Sciences, Laboratory of MacroMolecular Cancer Therapeutics (MMCT), Althanstrasse 14, 1090 Vienna, Austria
| | - Manfred Ogris
- University of Vienna, Faculty of Life Sciences, Department of Pharmaceutical Sciences, Laboratory of MacroMolecular Cancer Therapeutics (MMCT), Althanstrasse 14, 1090 Vienna, Austria
| |
Collapse
|
27
|
Zhao C, Lv H, Tao S, Zhang T, Xu N, Zhu L. Exosomes: Promising nanocarrier for cancer therapy. NANO SELECT 2021. [DOI: 10.1002/nano.202100142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Chen‐Chen Zhao
- College of Life Sciences and Health Wuhan University of Science and Technology Wuhan Hubei Province China
| | - Hui‐Zhong Lv
- College of Life Sciences and Health Wuhan University of Science and Technology Wuhan Hubei Province China
| | - Su‐Wan Tao
- College of Life Sciences and Health Wuhan University of Science and Technology Wuhan Hubei Province China
| | - Tong‐Cun Zhang
- College of Life Sciences and Health Wuhan University of Science and Technology Wuhan Hubei Province China
| | - Na Xu
- College of Life Sciences and Health Wuhan University of Science and Technology Wuhan Hubei Province China
| | - Lian Zhu
- College of Life Sciences and Health Wuhan University of Science and Technology Wuhan Hubei Province China
- School of Chemical and Environmental Engineering Wuhan Polytechnic University Wuhan Hubei China
| |
Collapse
|
28
|
Jiang Z, Sun H, Yu J, Tian W, Song Y. Targeting CD47 for cancer immunotherapy. J Hematol Oncol 2021; 14:180. [PMID: 34717705 PMCID: PMC8557524 DOI: 10.1186/s13045-021-01197-w] [Citation(s) in RCA: 201] [Impact Index Per Article: 50.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 10/19/2021] [Indexed: 02/04/2023] Open
Abstract
Much progress has been made in targeting CD47 for cancer immunotherapy in solid tumors (ST) and hematological malignancies. We summarized the CD47-related clinical research and analyzed the research trend both in the USA and in China. As of August 28, 2021, there are a total 23 related therapeutic agents with 46 clinical trials in the NCT registry platform. Among these trials, 29 are in ST, 14 in hematological malignancies and 3 in both solid tumor and hematological malignancy. The ST include gastric cancer, head and neck squamous cell carcinoma and leiomyosarcoma, while the hematological malignancies include non-Hodgkin's lymphoma, acute myeloid leukemia, myelodysplastic syndrome, multiple myeloma and chronic myeloid leukemia. Majority of the CD47-related clinical trials are at the early phases, such as 31 at phase I, 14 at phase II and 1 at phase III in the USA and 9, 6, 1, in China, respectively. The targets and spectrums of mechanism of action include 26 with mono-specific and 20 with bi-specific targets in the USA and 13 with mono-specific and 3 with bi-specific targets in China. The new generation CD47 antibodies have demonstrated promising results, and it is highly hopeful that some candidate agents will emerge and make into clinical application to meet the urgent needs of patients.
Collapse
Affiliation(s)
- Zhongxing Jiang
- Department of Hematology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Hao Sun
- Department of Radiation Therapy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Jifeng Yu
- Department of Hematology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China. .,Henan International Joint Laboratory of Nuclear Protein Gene Regulation, Henan University College of Medicine, Kaifeng, 475004, Henan, China.
| | - Wenzhi Tian
- ImmuneOnco Biopharmaceuticals (Shanghai) Co., Ltd., Shanghai, 201203, China.
| | - Yongping Song
- Department of Hematology, the Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, 450008, Henan, China.
| |
Collapse
|
29
|
Jia X, Yan B, Tian X, Liu Q, Jin J, Shi J, Hou Y. CD47/SIRPα pathway mediates cancer immune escape and immunotherapy. Int J Biol Sci 2021; 17:3281-3287. [PMID: 34512146 PMCID: PMC8416724 DOI: 10.7150/ijbs.60782] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 07/14/2021] [Indexed: 12/25/2022] Open
Abstract
The adaptive immune checkpoints such as PD-1(programmed death-1)/PD-L1 (programmed death-ligand 1) play an important role in cancer immunotherapy, whereas increasing evidence suggests that cancer cell evades immune surveillance by innate immune checkpoints such as SIRPα (signal-regulatory protein α)/CD47 (cluster of differentiation 47). In multiple types of cancer cells and solid tumor tissues, highly expressed CD47 protein level has been observed, which is triggered by some transcription factors including NFκB, Myc, and HIF. As a transmembrane protein, the binding of CD47 to SIRPα ligand on phagocytes results in phagocytosis resistance and cancer cell immune escape. In contrast, CD47-SIRPα interaction blockade enhances cancer cell clearance by phagocytes such as macrophages and dendritic cells (DCs) to activate an innate immune response, whereas this process could promote antigen cross-presentation by antigen present cells (APCs) leading to T cell priming, consequently, activates an adaptive antitumor immune response. In this review, we discussed the current SIRPα-CD47 axis-mediated cancer cell immune escape and immunotherapy, which could provide an effective antitumor strategy by the innate and adaptive immune response.
Collapse
Affiliation(s)
- Xiao Jia
- Department of Oncology, the Affiliated Wujin Hospital, Jiangsu University, Changzhou, Jiangsu Province, 213017, the People's Republic of China.,School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu Province, 212013, the People's Republic of China
| | - Bingjun Yan
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu Province, 212013, the People's Republic of China
| | - Xiaoqing Tian
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu Province, 212013, the People's Republic of China
| | - Qian Liu
- Department of Oncology, the Affiliated Wujin Hospital, Jiangsu University, Changzhou, Jiangsu Province, 213017, the People's Republic of China.,Department of Oncology, the Wujin Clinical College of Xuzhou Medical University, Jiangsu Province, 212017, the People's Republic of China
| | - Jianhua Jin
- Department of Oncology, the Affiliated Wujin Hospital, Jiangsu University, Changzhou, Jiangsu Province, 213017, the People's Republic of China.,Department of Oncology, the Wujin Clinical College of Xuzhou Medical University, Jiangsu Province, 212017, the People's Republic of China
| | - Juanjuan Shi
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu Province, 212013, the People's Republic of China
| | - Yongzhong Hou
- Department of Oncology, the Affiliated Wujin Hospital, Jiangsu University, Changzhou, Jiangsu Province, 213017, the People's Republic of China.,School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu Province, 212013, the People's Republic of China
| |
Collapse
|
30
|
Sun J, Chen Y, Lubben B, Adebayo O, Muz B, Azab AK. CD47-targeting antibodies as a novel therapeutic strategy in hematologic malignancies. Leuk Res Rep 2021; 16:100268. [PMID: 34584838 PMCID: PMC8455363 DOI: 10.1016/j.lrr.2021.100268] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 09/13/2021] [Indexed: 01/08/2023] Open
Abstract
CD47 is a surface glycoprotein expressed by host cells to impede phagocytosis upon binding to macrophage SIRPα, thereby represents an immune checkpoint known as the "don't-eat-me" signal. However, accumulating evidence shows that solid and hematologic tumor cells overexpress CD47 to escape immune surveillance. Thus, targeting the CD47-SIRPa axis by limiting the activity of this checkpoint has emerged as a key area of research. In this review, we will provide an update on the landscape of CD47-targeting antibodies for hematological malignancies, including monoclonal and bi-specific antibodies, with a special emphasis on agents in clinical trials and novel approaches to overcome toxicity.
Collapse
Affiliation(s)
- Jennifer Sun
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St. Louis School of Medicine, 4511 Forest Park Ave, St. Louis, MO 63108, USA
- Department of Biomedical Engineering, Washington University in St. Louis McKelvy School of Engineering, St. Louis, MO, USA
| | - Yixuan Chen
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St. Louis School of Medicine, 4511 Forest Park Ave, St. Louis, MO 63108, USA
| | - Berit Lubben
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St. Louis School of Medicine, 4511 Forest Park Ave, St. Louis, MO 63108, USA
| | - Ola Adebayo
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St. Louis School of Medicine, 4511 Forest Park Ave, St. Louis, MO 63108, USA
| | - Barbara Muz
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St. Louis School of Medicine, 4511 Forest Park Ave, St. Louis, MO 63108, USA
| | - Abdel Kareem Azab
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St. Louis School of Medicine, 4511 Forest Park Ave, St. Louis, MO 63108, USA
- Department of Biomedical Engineering, Washington University in St. Louis McKelvy School of Engineering, St. Louis, MO, USA
| |
Collapse
|
31
|
Modulation of CD47-SIRPα innate immune checkpoint axis with Fc-function detuned anti-CD47 therapeutic antibody. Cancer Immunol Immunother 2021; 71:473-489. [PMID: 34247273 DOI: 10.1007/s00262-021-03010-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 07/04/2021] [Indexed: 01/09/2023]
Abstract
Cluster of differentiation 47 (CD47) is a transmembrane protein ubiquitously expressed on human cells but overexpressed on many different tumor cells. The interaction of CD47 with signal-regulatory protein alpha (SIRPα) triggers a "don't eat me" signal to the macrophage, inhibiting phagocytosis. Thus, overexpression of CD47 enables tumor cells to escape from immune surveillance via the blockade of phagocytic mechanisms. We report here the development and characterization of CC-90002, a humanized anti-CD47 antibody. CC-90002 is unique among previously reported anti-CD47 bivalent antibodies that it does not promote hemagglutination while maintaining high-affinity binding to CD47 and inhibition of the CD47-SIRPα interaction. Studies in a panel of hematological cancer cell lines showed concentration-dependent CC-90002-mediated phagocytosis in acute lymphoblastic leukemia, acute myeloid leukemia (AML), lenalidomide-resistant multiple myeloma (MM) cell lines and AML cells from patients. In vivo studies with MM cell line-derived xenograft models established in immunodeficient mice demonstrated significant dose-dependent antitumor activity of CC-90002. Treatment with CC-90002 significantly prolonged survival in an HL-60-disseminated AML model. Mechanistic studies confirmed the binding of CC-90002 to tumor cells and concomitant recruitment of F4-80 positive macrophages into the tumor and an increase in expression of select chemokines and cytokines of murine origin. Furthermore, the role of macrophages in the CC-90002-mediated antitumor activity was demonstrated by transient depletion of macrophages with liposome-clodronate treatment. In non-human primates, CC-90002 displayed acceptable pharmacokinetic properties and a favorable toxicity profile. These data demonstrate the potential activity of CC-90002 across hematological malignancies and provided basis for clinical studies CC-90002-ST-001 (NCT02367196) and CC-90002-AML-001 (NCT02641002).
Collapse
|
32
|
Lentz RW, Colton MD, Mitra SS, Messersmith WA. Innate Immune Checkpoint Inhibitors: The Next Breakthrough in Medical Oncology? Mol Cancer Ther 2021; 20:961-974. [PMID: 33850005 PMCID: PMC9028741 DOI: 10.1158/1535-7163.mct-21-0041] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/01/2021] [Accepted: 03/29/2021] [Indexed: 11/16/2022]
Abstract
While immunotherapy has revolutionized the treatment of many types of advanced cancer, most patients still do not derive benefit. The currently available immune checkpoint inhibitors target the adaptive immune system, generating a T-cell antitumor response. However, an antitumor immune response depends on a complex interplay of both innate and adaptive immune cells. The innate immune system is a promising new target, and innate immune checkpoint inhibitors can disrupt inhibitory interactions ("don't eat me" signals) between tumor and both phagocytes and natural killer cells. The checkpoint inhibitor may also provide a stimulatory interaction ("eat me" signal), or this can be achieved through use of combination therapy. This generates antitumor effector functions including phagocytosis, natural cytotoxicity, antibody-dependent effects, and synergistic activation of the adaptive immune system via antigen presentation. This is a rapidly expanding area of drug development, either alone or in combination (with anticancer antibodies or adaptive immune checkpoint inhibitors). Here, we comprehensively review the mechanism of action and up-to-date solid tumor clinical trial data of the drugs targeting phagocytosis checkpoints (SIRPα/CD47, LILRB1/MHC-I, and LILRB2/MHC-I) and natural killer-cell checkpoints (TIGIT/CD112 + CD155, PVRIG/CD112, KIRs/MHC-I, and NKG2A-CD94/HLA-E). Innate immune checkpoint inhibitors could once again revolutionize immune-based cancer therapies.
Collapse
Affiliation(s)
- Robert W Lentz
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Meryl D Colton
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Siddhartha S Mitra
- Division of Hematology, Oncology, and Bone Marrow Transplant, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
| | - Wells A Messersmith
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado.
| |
Collapse
|
33
|
Chen S, Lai SWT, Brown CE, Feng M. Harnessing and Enhancing Macrophage Phagocytosis for Cancer Therapy. Front Immunol 2021; 12:635173. [PMID: 33790906 PMCID: PMC8006289 DOI: 10.3389/fimmu.2021.635173] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 01/18/2021] [Indexed: 12/14/2022] Open
Abstract
Cancer immunotherapy has revolutionized the paradigm for the clinical management of cancer. While FDA-approved cancer immunotherapies thus far mainly exploit the adaptive immunity for therapeutic efficacy, there is a growing appreciation for the importance of innate immunity in tumor cell surveillance and eradication. The past decade has witnessed macrophages being thrust into the spotlight as critical effectors of an innate anti-tumor response. Promising evidence from preclinical and clinical studies have established targeting macrophage phagocytosis as an effective therapeutic strategy, either alone or in combination with other therapeutic moieties. Here, we review the recent translational advances in harnessing macrophage phagocytosis as a pivotal therapeutic effort in cancer treatment. In addition, this review emphasizes phagocytosis checkpoint blockade and the use of nanoparticles as effective strategies to potentiate macrophages for phagocytosis. We also highlight chimeric antigen receptor macrophages as a next-generation therapeutic modality linking the closely intertwined innate and adaptive immunity to induce efficacious anti-tumor immune responses.
Collapse
Affiliation(s)
- Siqi Chen
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Seigmund W. T. Lai
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Christine E. Brown
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, United States
| | - Mingye Feng
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| |
Collapse
|
34
|
Leclair P, Lim CJ. CD47 (Cluster of differentiation 47): an anti-phagocytic receptor with a multitude of signaling functions. Anim Cells Syst (Seoul) 2020; 24:243-252. [PMID: 33224442 PMCID: PMC7654641 DOI: 10.1080/19768354.2020.1818618] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
CD47 is a tumor-associated antigen best known for its ability to bind counter-receptors on the surface of professional phagocytes as an immune-evasion strategy. Recently, CD47 has been shown to play a role as a signaling receptor, involving a number of cell physiological processes. This review provides a comprehensive survey of the signaling pathways triggered by CD47 ligand-mediated cell death in tumor cells. Such an understanding should lead to improvement of CD47-targeted anti-tumor therapeutics able to both neutralize the anti-phagocytic role and trigger autonomous tumor cell death.
Collapse
Affiliation(s)
- Pascal Leclair
- Department of Pediatrics, University of British Columbia, and, Michael Cuccione Childhood Cancer Research Program, B.C. Children's Hospital Research Institute, Vancouver, Canada
| | - Chinten James Lim
- Department of Pediatrics, University of British Columbia, and, Michael Cuccione Childhood Cancer Research Program, B.C. Children's Hospital Research Institute, Vancouver, Canada
| |
Collapse
|
35
|
Cham LB, Adomati T, Li F, Ali M, Lang KS. CD47 as a Potential Target to Therapy for Infectious Diseases. Antibodies (Basel) 2020; 9:antib9030044. [PMID: 32882841 PMCID: PMC7551396 DOI: 10.3390/antib9030044] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/12/2020] [Accepted: 07/27/2020] [Indexed: 12/12/2022] Open
Abstract
The integrin associated protein (CD47) is a widely and moderately expressed glycoprotein in all healthy cells. Cancer cells are known to induce increased CD47 expression. Similar to cancer cells, all immune cells can upregulate their CD47 surface expression during infection. The CD47-SIRPa interaction induces an inhibitory effect on macrophages and dendritic cells (dendritic cells) while CD47-thrombospondin-signaling inhibits T cells. Therefore, the disruption of the CD47 interaction can mediate several biologic functions. Upon the blockade and knockout of CD47 reveals an immunosuppressive effect of CD47 during LCMV, influenza virus, HIV-1, mycobacterium tuberculosis, plasmodium and other bacterial pneumonia infections. In our recent study we shows that the blockade of CD47 using the anti-CD47 antibody increases the activation and effector function of macrophages, dendritic cells and T cells during viral infection. By enhancing both innate and adaptive immunity, CD47 blocking antibody promotes antiviral effect. Due to its broad mode of action, the immune-stimulatory effect derived from this antibody could be applicable in nonresolving and (re)emerging infections. The anti-CD47 antibody is currently under clinical trial for the treatment of cancer and could also have amenable therapeutic potential against infectious diseases. This review highlights the immunotherapeutic targeted role of CD47 in the infectious disease realm.
Collapse
|
36
|
Kaur S, Cicalese KV, Banerjee R, Roberts DD. Preclinical and Clinical Development of Therapeutic Antibodies Targeting Functions of CD47 in the Tumor Microenvironment. Antib Ther 2020; 3:179-192. [PMID: 33244513 PMCID: PMC7687918 DOI: 10.1093/abt/tbaa017] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/22/2020] [Accepted: 08/05/2020] [Indexed: 02/07/2023] Open
Abstract
CD47 is a ubiquitously expressed cell surface glycoprotein that functions as a signaling receptor for thrombospondin-1 and as the counter-receptor for signal regulatory protein-α (SIRPα). Engaging SIRPα on macrophages inhibits phagocytosis, and CD47 thereby serves as a physiological marker of self. However, elevated CD47 expression on some cancer cells also protects tumors from innate immune surveillance and limits adaptive antitumor immunity via inhibitory SIRPα signaling in antigen presenting cells. CD47 also mediates inhibitory thrombospondin-1 signaling in vascular cells, T cells, and NK cells, and blocking inhibitory CD47 signaling on cytotoxic T cells directly increases tumor cell killing. Therefore, CD47 functions as an innate and adaptive immune checkpoint. These findings have led to the development of antibodies and other therapeutic approaches to block CD47 functions in the tumor microenvironment. Preclinical studies in mice demonstrated that blocking CD47 can limit the growth of hematologic malignancies and solid tumors and enhance the efficacy of conventional chemotherapy, radiation therapy, and some targeted cancer therapies. Humanized CD47 antibodies are showing promise in early clinical trials, but side effects related to enhanced phagocytic clearance of circulating blood cells remain a concern. Approaches to circumvent these include antibody preloading strategies, development of antibodies that recognize tumor-specific epitopes of CD47, SIRPα antibodies, and bivalent antibodies that restrict CD47 blockade to specific tumor cells. Preclinical and clinical development of antibodies and related biologics that inhibit CD47/SIRPα signaling are reviewed, including strategies to combine these agents with various conventional and targeted therapeutics to improve patient outcome for various cancers.
Collapse
Affiliation(s)
- Sukhbir Kaur
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kyle V Cicalese
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Rajdeep Banerjee
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - David D Roberts
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|