1
|
Farzeen Z, Khan RRM, Chaudhry AR, Pervaiz M, Saeed Z, Rasheed S, Shehzad B, Adnan A, Summer M. Dostarlimab: A promising new PD-1 inhibitor for cancer immunotherapy. J Oncol Pharm Pract 2024; 30:1411-1431. [PMID: 39056234 DOI: 10.1177/10781552241265058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/28/2024]
Abstract
OBJECTIVE Dostarlimab, a humanized monoclonal PD-1 blocking antibody, is being tested as a cancer therapy in this review. Specifically, it addresses mismatch repair failure in endometrial cancer and locally progressed rectal cancer patients. DATA SOURCES A thorough database search found Dostarlimab clinical trials and studies. Published publications and ongoing clinical trials on Dostarlimab's efficacy as a single therapy and in conjunction with other medicines across cancer types were searched. DATA SUMMARY The review recommends Dostarlimab for endometrial cancer mismatch repair failure, as supported by GARNET studies. The analysis also highlights locally advanced rectal cancer findings. In the evolving area of cancer therapy, immune checkpoint inhibitors including pembrolizumab, avelumab, atezolizumab, nivolumab, and durvalumab were discussed. CONCLUSIONS Locally advanced rectal cancer patients responded 100% to Dostarlimab. Many clinical trials, including ROSCAN, AMBER, IOLite, CITRINO, JASPER, OPAL, PRIME, PERLA, and others, are investigating Dostarlimab in combination treatment. This research sheds light on Dostarlimab's current and future possibilities, in improving cancer immunotherapy understanding.
Collapse
Affiliation(s)
- Zubaria Farzeen
- Department of Chemistry, Government College University Lahore, Lahore, Punjab, Pakistan
| | | | - Ayoub Rashid Chaudhry
- Department of Chemistry, Government College University Lahore, Lahore, Punjab, Pakistan
| | - Muhammad Pervaiz
- Department of Chemistry, Government College University Lahore, Lahore, Punjab, Pakistan
| | - Zohaib Saeed
- Department of Chemistry, Government College University Lahore, Lahore, Punjab, Pakistan
| | - Shahzad Rasheed
- Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| | - Behram Shehzad
- Department of Chemistry, Government College University Lahore, Lahore, Punjab, Pakistan
| | - Ahmad Adnan
- Department of Chemistry, Government College University Lahore, Lahore, Punjab, Pakistan
| | - Muhammad Summer
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University Lahore, Lahore, Pakistan
| |
Collapse
|
2
|
Bootz A, Reuter N, Nimmerjahn F, Britt WJ, Mach M, Winkler TH. Functional Fc receptors are crucial in antibody-mediated protection against cytomegalovirus. Eur J Immunol 2024; 54:e2451044. [PMID: 39014923 DOI: 10.1002/eji.202451044] [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: 01/31/2024] [Revised: 07/07/2024] [Accepted: 07/09/2024] [Indexed: 07/18/2024]
Abstract
Human cytomegalovirus is a medically important pathogen. Previously, using murine CMV (MCMV), we provided evidence that both neutralizing and nonneutralizing antibodies can confer protection from viral infection in vivo. In this study, we report that serum derived from infected animals had a greater protective capacity in MCMV-infected RAG-/- mice than serum from animals immunized with purified virus. The protective activity of immune serum was strictly dependent on functional Fcγ receptors (FcγR). Deletion of individual FcγRs or combined deletion of FcγRI and FcγRIV had little impact on the protection afforded by serum. Adoptive transfer of CD115-positive cells from noninfected donors demonstrated that monocytes represent important cellular mediators of the protective activity provided by immune serum. Our studies suggest that Fc-FcγR interactions and monocytic cells are critical for antibody-mediated protection against MCMV infection in vivo. These findings may provide new avenues for the development of novel strategies for more effective CMV vaccines or antiviral immunotherapies.
Collapse
Affiliation(s)
- Anna Bootz
- Institute of Clinical and Molecular Virology, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Nina Reuter
- Institute of Clinical and Molecular Virology, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Falk Nimmerjahn
- Division of Genetics, Department Biology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - William J Britt
- Departments of Pediatrics, Microbiology and Neurobiology, Children's Hospital of Alabama, School of Medicine, University of Alabama, Birmingham, Alabama, USA
| | - Michael Mach
- Institute of Clinical and Molecular Virology, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Thomas H Winkler
- Division of Genetics, Department Biology, Nikolaus-Fiebiger-Center of Molecular Medicine, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
3
|
Andreeva I, Kolb P, Rodon L, Blank N, Lorenz HM, Merkt W. Fcγ-receptor-IIIA bioactivity of circulating and synovial immune complexes in rheumatoid arthritis. RMD Open 2024; 10:e004190. [PMID: 39209371 PMCID: PMC11367361 DOI: 10.1136/rmdopen-2024-004190] [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: 02/04/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024] Open
Abstract
OBJECTIVE Previous technical limitations prevented the proof of Fcγ-receptor (FcγR)-activation by soluble immune complexes (sICs) in patients. FcγRIIIa (CD16) is a risk factor in rheumatoid arthritis (RA). We aimed at determining the presence of CD16-activating sICs in RA and control diseases. METHODS Sera from an exploratory cohort (n=50 patients with RA) and a validation cohort (n=106 patients with RA, 20 patients with psoriasis arthritis (PsA), 22 patients with systemic lupus erythematosus (SLE) and 31 healthy controls) were analysed using a new reporter cell assay. Additionally, 26 synovial fluid samples were analysed, including paired serum/synovial samples. RESULTS For the first time using a reliable and sensitive functional assay, the presence of sICs in RA sera was confirmed. sICs possess an intrinsic capacity to activate CD16 and can be found in both synovial fluid and in blood. In low experimental dilutions, circulating sICs were also detected in a subset of healthy people and in PsA. However, we report a significantly increased frequency of bioactive circulating sICs in RA. While the bioactivity of circulating sICs was low and did not correlate with clinical parameters, synovial sICs were highly bioactive and correlated with serum autoantibody levels. Receiver operator curves indicated that sICs bioactivity in synovial fluid could be used to discriminate immune complex-associated arthritis from non-associated forms. Finally, circulating sICs were more frequently found in SLE than in RA. The degree of CD16 bioactivity showed strong donor-dependent differences, especially in SLE. CONCLUSIONS RA is characterised by the presence of circulating and synovial sICs that can engage and activate CD16.
Collapse
Affiliation(s)
- Ivana Andreeva
- Department of Hematology, Oncology and Rheumatology, Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Philipp Kolb
- Institute of Virology, Medical Center—University of Freiburg, Freiburg, Germany
- Albert-Ludwigs-Universitat Freiburg Medizinische Fakultat, Freiburg, Germany
| | - Lea Rodon
- Department of Hematology, Oncology and Rheumatology, Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Norbert Blank
- Department of Hematology, Oncology and Rheumatology, Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Hanns-Martin Lorenz
- Department of Hematology, Oncology and Rheumatology, Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Wolfgang Merkt
- Department of Hematology, Oncology and Rheumatology, Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany
- Hiller Forschungszentrum, Department of Rheumatology, University Hospital of Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
4
|
Millan AJ, Allain V, Nayak I, Aguilar OA, Arakawa-Hoyt JS, Ureno G, Rothrock AG, Shemesh A, Eyquem J, Das J, Lanier LL. Spleen Tyrosine Kinase (SYK) negatively regulates ITAM-mediated human NK cell signaling and CD19-CAR NK cell efficacy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.09.602676. [PMID: 39026749 PMCID: PMC11257556 DOI: 10.1101/2024.07.09.602676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
NK cells express activating receptors that signal through ITAM-bearing adapter proteins. The phosphorylation of each ITAM creates binding sites for SYK and ZAP70 protein tyrosine kinases to propagate downstream signaling including the induction ofCa 2 + influx. While all immature and mature human NK cells co-express SYK and ZAP70, clonally driven memory or adaptive NK cells can methylate SYK genes and signaling is mediated exclusively using ZAP70. Here, we examined the role of SYK and ZAP70 in a clonal human NK cell line KHYG1 by CRISPR-based deletion using a combination of experiments and mechanistic computational modeling. Elimination of SYK resulted in more robustCa + + influx after cross-linking of the CD16 and NKp30 receptors and enhanced phosphorylation of downstream proteins, whereas ZAP70 deletion diminished these responses. By contrast, ZAP70 depletion increased proliferation of the NK cells. As immature T cells express both SYK and ZAP70 but mature T cells often express only ZAP70, we transduced the human Jurkat cell line with SYK and found that expression of SYK increased proliferation but diminished TCR-inducedCa 2 + flux and activation. We performed transcriptional analysis of the matched sets of variant Jurkat and KHYG1 cells and observed profound alterations caused by SYK expression. As depletion of SYK in NK cells increased their activation, primary human NK cells were transduced with a CD19-targeting CAR and were CRISPR edited to ablate SYK or ZAP70. Deletion of SYK resulted in more robust cytotoxic activity and cytokine production, providing a new therapeutic strategy of NK cell engineering for cancer immunotherapy.
Collapse
Affiliation(s)
- Alberto J. Millan
- Department of Microbiology and Immunology and the Parker Institute for Cancer Immunotherapy, University of California-San Francisco, San Francisco, CA, USA
| | - Vincent Allain
- Department of Medicine, University of California-San Francisco, San Francisco, CA, USA
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Université Paris Cité, INSERM UMR976, Hôpital Saint-Louis, Paris, France
| | - Indrani Nayak
- Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children’s Hospital, Biomedical Sciences Graduate Program, Department of Pediatrics, Pelotonia Institute for Immuno-Oncology, College of Medicine, The Ohio State University, Columbus OH
| | - Oscar A. Aguilar
- Department of Microbiology and Immunology and the Parker Institute for Cancer Immunotherapy, University of California-San Francisco, San Francisco, CA, USA
| | - Janice S. Arakawa-Hoyt
- Department of Microbiology and Immunology and the Parker Institute for Cancer Immunotherapy, University of California-San Francisco, San Francisco, CA, USA
| | - Gabriella Ureno
- Department of Microbiology and Immunology and the Parker Institute for Cancer Immunotherapy, University of California-San Francisco, San Francisco, CA, USA
| | - Allison Grace Rothrock
- Department of Medicine, University of California-San Francisco, San Francisco, CA, USA
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
| | - Avishai Shemesh
- Department of Microbiology and Immunology and the Parker Institute for Cancer Immunotherapy, University of California-San Francisco, San Francisco, CA, USA
- Department of Medicine, University of California-San Francisco, San Francisco, CA, USA
| | - Justin Eyquem
- Department of Medicine, University of California-San Francisco, San Francisco, CA, USA
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
| | - Jayajit Das
- Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children’s Hospital, Biomedical Sciences Graduate Program, Department of Pediatrics, Pelotonia Institute for Immuno-Oncology, College of Medicine, The Ohio State University, Columbus OH
| | - Lewis L. Lanier
- Department of Microbiology and Immunology and the Parker Institute for Cancer Immunotherapy, University of California-San Francisco, San Francisco, CA, USA
| |
Collapse
|
5
|
Dick JK, Hicks D, Krishna VD, Sangala JA, Zandstra BT, Baehr C, Verbeek JS, Cragg MS, Cheeran MCJ, Pravetoni M, Hart GT. ACE2 decoy Fc-fusions and bi-specific killer engager (BiKEs) require Fc engagement for in vivo efficacy against SARS-CoV-2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.20.599956. [PMID: 38948747 PMCID: PMC11212978 DOI: 10.1101/2024.06.20.599956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
SARS-CoV-2 virus has continued to evolve over time necessitating the adaptation of vaccines to maintain efficacy. Monoclonal antibodies (mAbs) against SARS-CoV-2 were a key line of defense for unvaccinated or immunocompromised individuals. However, these mAbs are now ineffective against current SARS-CoV-2 variants. Here, we tested three aspects of αSARS-CoV-2 therapeutics. First, we tested whether Fc engagement is necessary for in vivo clearance of SARS-CoV-2. Secondly, we tested bi-specific killer engagers (BiKEs) that simultaneously engage SARS-CoV-2 and a specific Fc receptor. Benefits of these engagers include the ease of manufacturing, stability, more cell-specific targeting, and high affinity binding to Fc receptors. Using both mAbs and BiKEs, we found that both neutralization and Fc receptor engagement were necessary for effective SARS-CoV-2 clearance. Thirdly, due to ACE2 being necessary for viral entry, ACE2 will maintain binding to SARS-CoV-2 despite viral evolution. Therefore, we used an ACE2 decoy Fc-fusion or BiKE, instead of an anti-SARS-CoV-2 antibody sequence, as a potential therapeutic that would withstand viral evolution. We found that the ACE2 decoy approach also required Fc receptor engagement and, unlike traditional neutralizing antibodies against specific variants, enabled the clearance of two distinct SARS-CoV-2 variants. These data show the importance of Fc engagement for mAbs, the utility of BiKEs as therapies for infectious disease, and the in vivo effectiveness of the ACE2 decoy approach. With further studies, we predict combining neutralization, the cellular response, and this ACE2 decoy approach will benefit individuals with ineffective antibody levels. Abbreviations ACE2, scFv, mAb, BiKE, COVID-19, Fc, CD16, CD32b, CD64, d.p.i. Key points With equal dosing, both neutralization and Fc engagement are necessary for the optimal efficacy of in vivo antibodies and bi-specific killer engagers (BiKEs) against SARS-CoV-2. BiKEs can clear SARS-CoV-2 virus and protect against severe infection in the hACE2-K18 mouse model. ACE2 decoys as part of Fc-fusions or BiKEs provide in vivo clearance of two disparate SARS-CoV-2 variants.
Collapse
|
6
|
Yang J, Wang L, Byrnes JR, Kirkemo LL, Driks H, Belair CD, Aguilar OA, Lanier LL, Wells JA, Fong L, Blelloch R. PVRL2 Suppresses Antitumor Immunity through PVRIG- and TIGIT-independent Pathways. Cancer Immunol Res 2024; 12:575-591. [PMID: 38588410 PMCID: PMC11063765 DOI: 10.1158/2326-6066.cir-23-0722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 01/04/2024] [Accepted: 03/07/2024] [Indexed: 04/10/2024]
Abstract
Poliovirus receptor-related 2 (PVRL2, also known as nectin-2 or CD112) is believed to act as an immune checkpoint protein in cancer; however, most insight into its role is inferred from studies on its known receptor, poliovirus receptor (PVR)-related immunoglobulin domain protein (PVRIG, also known as CD112R). Here, we study PVRL2 itself. PVRL2 levels were found to be high in tumor cells and tumor-derived exosomes. Deletion of PVRL2 in multiple syngeneic mouse models of cancer showed a dramatic reduction in tumor growth that was immune dependent. This effect was even greater than that seen with deletion of PD-L1. PVRL2 was shown to function by suppressing CD8+ T and natural killer cells in the tumor microenvironment. The loss of PVRL2 suppressed tumor growth even in the absence of PVRIG. In contrast, PVRIG loss showed no additive effect in the absence of PVRL2. T-cell immunoreceptor with Ig and ITIM domains (TIGIT) blockade combined with PVRL2 deletion resulted in a near complete block in tumor growth. This effect was not recapitulated by the combined deletion of PVRL2 with its paralog, PVR, which is the ligand for TIGIT. These data uncover PVRL2 as a distinct inhibitor of the antitumor immune response with functions beyond that of its known receptor PVRIG. Moreover, the data provide a strong rationale for combinatorial targeting of PVRL2 and TIGIT for cancer immunotherapy.
Collapse
Affiliation(s)
- Jiuling Yang
- Department of Urology, University of California San Francisco, San Francisco, California
| | - Li Wang
- Department of Urology, University of California San Francisco, San Francisco, California
| | - James R. Byrnes
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California
| | - Lisa L. Kirkemo
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California
| | - Hannah Driks
- Department of Urology, University of California San Francisco, San Francisco, California
| | - Cassandra D. Belair
- Department of Urology, University of California San Francisco, San Francisco, California
| | - Oscar A. Aguilar
- Department of Microbiology and Immunology, University of California, San Francisco, and Parker Institute for Cancer Immunotherapy, San Francisco, California
| | - Lewis L. Lanier
- Department of Microbiology and Immunology, University of California, San Francisco, and Parker Institute for Cancer Immunotherapy, San Francisco, California
| | - James A. Wells
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California
| | - Lawrence Fong
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Robert Blelloch
- Department of Urology, University of California San Francisco, San Francisco, California
| |
Collapse
|
7
|
Aguilar OA, Fong LK, Lanier LL. ITAM-based receptors in natural killer cells. Immunol Rev 2024; 323:40-53. [PMID: 38411263 PMCID: PMC11102329 DOI: 10.1111/imr.13313] [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: 10/31/2023] [Accepted: 02/15/2024] [Indexed: 02/28/2024]
Abstract
The ability of cells of the immune system to acquire features such as increased longevity and enhanced secondary responses was long thought to be restricted to cells of the adaptive immune system. Natural killer (NK) cells have challenged this notion by demonstrating that they can also gain adaptive features. This has been observed in both humans and mice during infection with cytomegalovirus (CMV). The generation of adaptive NK cells requires antigen-specific recognition of virally infected cells through stimulatory NK receptors. These receptors lack the ability to signal on their own and rather rely on adaptor molecules that contain ITAMs for driving signals. Here, we highlight our understanding of how these receptors influence the production of adaptive NK cells and propose areas in the field that merit further investigation.
Collapse
Affiliation(s)
- Oscar A. Aguilar
- Dept. of Microbiology and Immunology, University of California - San Francisco, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy, University of California - San Francisco, San Francisco, CA, USA
| | - Lam-Kiu Fong
- Dept. of Pharmaceutical Chemistry, University of California – San Francisco, San Francisco, CA
| | - Lewis L. Lanier
- Dept. of Microbiology and Immunology, University of California - San Francisco, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy, University of California - San Francisco, San Francisco, CA, USA
| |
Collapse
|
8
|
Besla R, Penuel E, Del Rosario G, Cosino E, Myrta S, Dillon M, Lazar GA, Nickles D, Spiess C, Yu SF, Polson AG. T cell-Dependent Bispecific Therapy Enhances Innate Immune Activation and Antibody-Mediated Killing. Cancer Immunol Res 2024; 12:60-71. [PMID: 37902604 DOI: 10.1158/2326-6066.cir-23-0072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 05/15/2023] [Accepted: 10/26/2023] [Indexed: 10/31/2023]
Abstract
T cell-retargeting therapies have transformed the therapeutic landscape for hematologic diseases. T cell-dependent bispecific antibodies (TDB) function as conditional agonists that induce a polyclonal T-cell response, resulting in target cell destruction and cytokine release. The relationship between this response and its effects on surrounding innate immune populations has not been fully explored. Here we show that treatment with mosunetuzumab in patients results in natural killer (NK) cell activation in the peripheral blood. We modeled this phenomenon in vitro and found that TDB-mediated killing activated NK cells, increasing NK function and antibody-dependent cellular cytotoxicity (ADCC), and enhanced the capability of macrophages to perform antibody-dependent cellular phagocytosis (ADCP). This enhancement was triggered by cytokines released through TDB treatment, with IL2 and IFNγ being major drivers for increased ADCC and ADCP, respectively. Surprisingly, cytolytic ability could be further augmented through neutralization of IL10 for NK cells and TNFα for macrophages. Finally, we showed that TDB treatment enhanced the efficacy of Fc-driven killing to an orthogonal solid tumor target in vivo. These results provide rationale for novel antibody therapy combinations that take advantage of both adaptive and innate immune responses.
Collapse
Affiliation(s)
- Rickvinder Besla
- Genentech Research and Early Development, Genentech Inc., South San Francisco, California
| | - Elicia Penuel
- Genentech Research and Early Development, Genentech Inc., South San Francisco, California
| | - Geoff Del Rosario
- Genentech Research and Early Development, Genentech Inc., South San Francisco, California
| | - Ely Cosino
- Genentech Research and Early Development, Genentech Inc., South San Francisco, California
| | | | - Mike Dillon
- Genentech Research and Early Development, Genentech Inc., South San Francisco, California
| | - Greg A Lazar
- Genentech Research and Early Development, Genentech Inc., South San Francisco, California
| | - Dorothee Nickles
- Genentech Research and Early Development, Genentech Inc., South San Francisco, California
| | - Christoph Spiess
- Genentech Research and Early Development, Genentech Inc., South San Francisco, California
| | - Shang-Fan Yu
- Genentech Research and Early Development, Genentech Inc., South San Francisco, California
| | - Andrew G Polson
- Genentech Research and Early Development, Genentech Inc., South San Francisco, California
| |
Collapse
|
9
|
Aguilar OA, Qualls AE, Gonzalez-Hinojosa MDR, Obeidalla S, Kerchberger VE, Tsao T, Singer JP, Looney MR, Raymond W, Hays SR, Golden JA, Kukreja J, Shaver CM, Ware LB, Christie J, Diamond JM, Lanier LL, Greenland JR, Calabrese DR. MICB Genomic Variant Is Associated with NKG2D-mediated Acute Lung Injury and Death. Am J Respir Crit Care Med 2024; 209:70-82. [PMID: 37878820 PMCID: PMC10870895 DOI: 10.1164/rccm.202303-0472oc] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 10/17/2023] [Indexed: 10/27/2023] Open
Abstract
Rationale: Acute lung injury (ALI) carries a high risk of mortality but has no established pharmacologic therapy. We previously found that experimental ALI occurs through natural killer (NK) cell NKG2D receptor activation and that the cognate human ligand, MICB, was associated with ALI after transplantation. Objectives: To investigate the association of a common missense variant, MICBG406A, with ALI. Methods: We assessed MICBG406A genotypes within two multicenter observational study cohorts at risk for ALI: primary graft dysfunction (N = 619) and acute respiratory distress syndrome (N = 1,376). Variant protein functional effects were determined in cultured and ex vivo human samples. Measurements and Main Results: Recipients of MICBG406A-homozygous allografts had an 11.1% absolute risk reduction (95% confidence interval [CI], 3.2-19.4%) for severe primary graft dysfunction after lung transplantation and reduced risk for allograft failure (hazard ratio, 0.36; 95% CI, 0.13-0.98). In participants with sepsis, we observed 39% reduced odds of moderately or severely impaired oxygenation among MICBG406A-homozygous individuals (95% CI, 0.43-0.86). BAL NK cells were less frequent and less mature in participants with MICBG406A. Expression of missense variant protein MICBD136N in cultured cells resulted in reduced surface MICB and reduced NKG2D ligation relative to wild-type MICB. Coculture of variant MICBD136N cells with NK cells resulted in less NKG2D activation and less susceptibility to NK cell killing relative to the wild-type cells. Conclusions: These data support a role for MICB signaling through the NKG2D receptor in mediating ALI, suggesting a novel therapeutic approach.
Collapse
Affiliation(s)
- Oscar A. Aguilar
- Department Microbiology and Immunology
- Parker Institute for Cancer Immunotherapy
| | | | | | | | | | | | | | | | | | | | | | - Jasleen Kukreja
- Department of Surgery, University of California San Francisco, San Francisco, California
| | | | - Lorraine B. Ware
- Department Medicine and
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jason Christie
- Department Medicine and
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania; and
| | | | - Lewis L. Lanier
- Department Microbiology and Immunology
- Parker Institute for Cancer Immunotherapy
| | - John R. Greenland
- Department Medicine
- San Francisco Veterans Affairs Medical Center, San Francisco, California
| | - Daniel R. Calabrese
- Department Medicine
- San Francisco Veterans Affairs Medical Center, San Francisco, California
| |
Collapse
|
10
|
Galvez-Cancino F, Simpson AP, Costoya C, Matos I, Qian D, Peggs KS, Litchfield K, Quezada SA. Fcγ receptors and immunomodulatory antibodies in cancer. Nat Rev Cancer 2024; 24:51-71. [PMID: 38062252 DOI: 10.1038/s41568-023-00637-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/10/2023] [Indexed: 12/24/2023]
Abstract
The discovery of both cytotoxic T lymphocyte-associated antigen 4 (CTLA4) and programmed cell death protein 1 (PD1) as negative regulators of antitumour immunity led to the development of numerous immunomodulatory antibodies as cancer treatments. Preclinical studies have demonstrated that the efficacy of immunoglobulin G (IgG)-based therapies depends not only on their ability to block or engage their targets but also on the antibody's constant region (Fc) and its interactions with Fcγ receptors (FcγRs). Fc-FcγR interactions are essential for the activity of tumour-targeting antibodies, such as rituximab, trastuzumab and cetuximab, where the killing of tumour cells occurs at least in part due to these mechanisms. However, our understanding of these interactions in the context of immunomodulatory antibodies designed to boost antitumour immunity remains less explored. In this Review, we discuss our current understanding of the contribution of FcγRs to the in vivo activity of immunomodulatory antibodies and the challenges of translating results from preclinical models into the clinic. In addition, we review the impact of genetic variability of human FcγRs on the activity of therapeutic antibodies and how antibody engineering is being utilized to develop the next generation of cancer immunotherapies.
Collapse
Affiliation(s)
- Felipe Galvez-Cancino
- Cancer Immunology Unit, Research Department of Haematology, University College London Cancer Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Alexander P Simpson
- Cancer Immunology Unit, Research Department of Haematology, University College London Cancer Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Cristobal Costoya
- Cancer Immunology Unit, Research Department of Haematology, University College London Cancer Institute, London, UK
| | - Ignacio Matos
- Cancer Immunology Unit, Research Department of Haematology, University College London Cancer Institute, London, UK
| | - Danwen Qian
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Tumour Immunogenomics and Immunosurveillance Laboratory, University College London Cancer Institute, London, UK
| | - Karl S Peggs
- Cancer Immunology Unit, Research Department of Haematology, University College London Cancer Institute, London, UK
| | - Kevin Litchfield
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Tumour Immunogenomics and Immunosurveillance Laboratory, University College London Cancer Institute, London, UK
| | - Sergio A Quezada
- Cancer Immunology Unit, Research Department of Haematology, University College London Cancer Institute, London, UK.
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK.
| |
Collapse
|
11
|
Balzasch BM, Cerwenka A. Microenvironmental signals shaping NK-cell reactivity in cancer. Eur J Immunol 2023; 53:e2250103. [PMID: 37194594 DOI: 10.1002/eji.202250103] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/05/2023] [Accepted: 05/09/2023] [Indexed: 05/18/2023]
Abstract
Since the postulation of the "missing-self" concept, much progress has been made in defining requirements for NK-cell activation. Unlike T lymphocytes that process signals from receptors in a hierarchic manner dominated by the T-cell receptors, NK cells integrate receptor signals more "democratically." Signals originate not only the downstream of cell-surface receptors triggered by membrane-bound ligands or cytokines, but are also mediated by specialized microenvironmental sensors that perceive the cellular surrounding by detecting metabolites or the availability of oxygen. Thus, NK-cell effector functions are driven in an organ and disease-dependent manner. Here, we review the latest findings on how NK-cell reactivity in cancer is determined by the reception and integration of complex signals. Finally, we discuss how this knowledge can be exploited to guide novel combinatorial approaches for NK-cell-based anticancer therapies.
Collapse
Affiliation(s)
- Bianca M Balzasch
- Department of Immunobiochemistry, Mannheim Institute for Innate Immunosciences (MI3), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Adelheid Cerwenka
- Department of Immunobiochemistry, Mannheim Institute for Innate Immunosciences (MI3), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
12
|
Dahlvang JD, Dick JK, Sangala JA, Kennedy PR, Pomeroy EJ, Snyder KM, Moushon JM, Thefaine CE, Wu J, Hamilton SE, Felices M, Miller JS, Walcheck B, Webber BR, Moriarity BS, Hart GT. Ablation of SYK Kinase from Expanded Primary Human NK Cells via CRISPR/Cas9 Enhances Cytotoxicity and Cytokine Production. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1108-1122. [PMID: 36881874 PMCID: PMC10073313 DOI: 10.4049/jimmunol.2200488] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 02/07/2023] [Indexed: 03/09/2023]
Abstract
CMV infection alters NK cell phenotype and function toward a more memory-like immune state. These cells, termed adaptive NK cells, typically express CD57 and NKG2C but lack expression of the FcRγ-chain (gene: FCER1G, FcRγ), PLZF, and SYK. Functionally, adaptive NK cells display enhanced Ab-dependent cellular cytotoxicity (ADCC) and cytokine production. However, the mechanism behind this enhanced function is unknown. To understand what drives enhanced ADCC and cytokine production in adaptive NK cells, we optimized a CRISPR/Cas9 system to ablate genes from primary human NK cells. We ablated genes that encode molecules in the ADCC pathway, such as FcRγ, CD3ζ, SYK, SHP-1, ZAP70, and the transcription factor PLZF, and tested subsequent ADCC and cytokine production. We found that ablating the FcRγ-chain caused a modest increase in TNF-α production. Ablation of PLZF did not enhance ADCC or cytokine production. Importantly, SYK kinase ablation significantly enhanced cytotoxicity, cytokine production, and target cell conjugation, whereas ZAP70 kinase ablation diminished function. Ablating the phosphatase SHP-1 enhanced cytotoxicity but reduced cytokine production. These results indicate that the enhanced cytotoxicity and cytokine production of CMV-induced adaptive NK cells is more likely due to the loss of SYK than the lack of FcRγ or PLZF. We found the lack of SYK expression could improve target cell conjugation through enhanced CD2 expression or limit SHP-1-mediated inhibition of CD16A signaling, leading to enhanced cytotoxicity and cytokine production.
Collapse
Affiliation(s)
- James D. Dahlvang
- Department of Medicine, Division of Infectious Disease and International Medicine, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jenna K. Dick
- Department of Medicine, Division of Infectious Disease and International Medicine, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jules A. Sangala
- Department of Medicine, Division of Infectious Disease and International Medicine, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Philippa R. Kennedy
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Medicine, Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, MN 55455, USA
| | - Emily J. Pomeroy
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Kristin M. Snyder
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Veterinary and Biological Sciences, University of Minnesota, St. Paul, MN 55108, USA
| | - Juliette M. Moushon
- Department of Medicine, Division of Infectious Disease and International Medicine, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Claire E. Thefaine
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jianming Wu
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Veterinary and Biological Sciences, University of Minnesota, St. Paul, MN 55108, USA
| | - Sara E. Hamilton
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Martin Felices
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Medicine, Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jeffrey S. Miller
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Medicine, Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, MN 55455, USA
| | - Bruce Walcheck
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Veterinary and Biological Sciences, University of Minnesota, St. Paul, MN 55108, USA
| | - Beau R. Webber
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Branden S. Moriarity
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Geoffrey T. Hart
- Department of Medicine, Division of Infectious Disease and International Medicine, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Lead contact
| |
Collapse
|
13
|
Wong JKM, Dolcetti R, Rhee H, Simpson F, Souza-Fonseca-Guimaraes F. Weaponizing natural killer cells for solid cancer immunotherapy. Trends Cancer 2023; 9:111-121. [PMID: 36379852 DOI: 10.1016/j.trecan.2022.10.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/16/2022] [Accepted: 10/19/2022] [Indexed: 11/13/2022]
Abstract
Enhancing natural killer (NK) cell-based innate immunity has become a promising strategy for immunotherapy against hard-to-cure solid cancers. Monoclonal antibody (mAb) therapy has been used to activate NK-cell-mediated antibody-dependent cellular cytotoxicity (ADCC) towards solid cancers. Cancer cells, however, can subvert immunosurveillance using multiple immunosuppressive mechanisms, which may hamper NK cell ADCC. Mechanisms to safely enhance ADCC by NK cells, such as utilizing temporary inhibition of receptor endocytosis to increase antibody presentation from target to effector cells can now be used to enhance NK-cell-mediated ADCC against solid tumors. This review summarizes and discusses the recent advances in the field and highlights current and potential future use of immunotherapies to maximize the therapeutic efficacy of innate anticancer immunity.
Collapse
Affiliation(s)
- Joshua K M Wong
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Riccardo Dolcetti
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia; Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria 3010, Australia; Department of Microbiology and Immunology, The University of Melbourne, Victoria 3010, Australia
| | - Handoo Rhee
- Princess Alexandra Hospital and Queen Elizabeth Jubilee II Hospital, Woolloongabba, QLD 4102, Australia; The School of Medicine, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Fiona Simpson
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | | |
Collapse
|
14
|
Aguilar OA, Gonzalez-Hinojosa MD, Arakawa-Hoyt JS, Millan AJ, Gotthardt D, Nabekura T, Lanier LL. The CD16 and CD32b Fc-gamma receptors regulate antibody-mediated responses in mouse natural killer cells. J Leukoc Biol 2023; 113:27-40. [PMID: 36822164 PMCID: PMC10197019 DOI: 10.1093/jleuko/qiac003] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Indexed: 01/12/2023] Open
Abstract
Natural killer (NK) cells are innate lymphocytes capable of mediating immune responses without prior sensitization. NK cells express Fc-gamma receptors (FcγRs) that engage the Fc region of IgG. Studies investigating the role of FcγRs on mouse NK cells have been limited due to lack specific reagents. In this study, we characterize the expression and biological consequences of activating mouse NK cells through their FcγRs. We demonstrate that most NK cells express the activating CD16 receptor, and a subset of NK cells also expresses the inhibitory CD32b receptor. Critically, these FcγRs are functional on mouse NK cells and can modulate antibody-mediated responses. We also characterized mice with conditional knockout alleles of Fcgr3 (CD16) or Fcgr2b (CD32b) in the NK and innate lymphoid cell (ILC) lineage. NK cells in these mice did not reveal any developmental defects and were responsive to cross-linking activating NK receptors, cytokine stimulation, and killing of YAC-1 targets. Importantly, CD16-deficient NK cells failed to induce antibody-directed cellular cytotoxicity of antibody-coated B-cell lymphomas in in vitro assays. In addition, we demonstrate the important role of CD16 on NK cells using an in vivo model of cancer immunotherapy using anti-CD20 antibody treatment of B-cell lymphomas.
Collapse
Affiliation(s)
- Oscar A. Aguilar
- Department of Microbiology and Immunology, University of California - San Francisco and Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Maria D.R. Gonzalez-Hinojosa
- Department of Microbiology and Immunology, University of California - San Francisco and Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Janice S. Arakawa-Hoyt
- Department of Microbiology and Immunology, University of California - San Francisco and Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Alberto J. Millan
- Department of Microbiology and Immunology, University of California - San Francisco and Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Dagmar Gotthardt
- Department of Microbiology and Immunology, University of California - San Francisco and Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
- Present Address: Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Tsukasa Nabekura
- Department of Microbiology and Immunology, University of California - San Francisco and Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Ibaraki, Japan
| | - Lewis L. Lanier
- Department of Microbiology and Immunology, University of California - San Francisco and Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| |
Collapse
|
15
|
Dangi T, Sanchez S, Class J, Richner M, Visvabharathy L, Chung YR, Bentley K, Stanton RJ, Koralnik IJ, Richner JM, Penaloza-MacMaster P. Improved control of SARS-CoV-2 by treatment with a nucleocapsid-specific monoclonal antibody. J Clin Invest 2022; 132:162282. [PMID: 36219482 PMCID: PMC9711872 DOI: 10.1172/jci162282] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 10/05/2022] [Indexed: 11/17/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein is the main antigen in all approved COVID-19 vaccines and is also the only target for monoclonal antibody (mAb) therapies. Immune responses to other viral antigens are generated after SARS-CoV-2 infection, but their contribution to the antiviral response remains unclear. Here, we interrogated whether nucleocapsid-specific antibodies can improve protection against SARS-CoV-2. We first immunized mice with a nucleocapsid-based vaccine and then transferred sera from these mice into naive mice, followed by challenge with SARS-CoV-2. We show that mice that received nucleocapsid-specific sera or a nucleocapsid-specific mAb exhibited enhanced control of SARS-CoV-2. Nucleocapsid-specific antibodies elicited NK-mediated, antibody-dependent cellular cytotoxicity (ADCC) against infected cells. To our knowledge, these findings provide the first demonstration in the coronavirus literature that antibody responses specific to the nucleocapsid protein can improve viral clearance, providing a rationale for the clinical evaluation of nucleocapsid-based mAb therapies to treat COVID-19.
Collapse
Affiliation(s)
- Tanushree Dangi
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Sarah Sanchez
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Jacob Class
- Department of Microbiology and Immunology, University of Illinois College of Medicine at Chicago, Chicago, Illinois, USA
| | - Michelle Richner
- Department of Microbiology and Immunology, University of Illinois College of Medicine at Chicago, Chicago, Illinois, USA
| | - Lavanya Visvabharathy
- Ken and Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Young Rock Chung
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Kirsten Bentley
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Richard J Stanton
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Igor J Koralnik
- Ken and Ruth Davee Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Justin M Richner
- Department of Microbiology and Immunology, University of Illinois College of Medicine at Chicago, Chicago, Illinois, USA
| | - Pablo Penaloza-MacMaster
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
16
|
Shemesh A, Su Y, Calabrese DR, Chen D, Arakawa-Hoyt J, Roybal KT, Heath JR, Greenland JR, Lanier LL. Diminished cell proliferation promotes natural killer cell adaptive-like phenotype by limiting FcεRIγ expression. J Exp Med 2022; 219:e20220551. [PMID: 36066491 PMCID: PMC9448639 DOI: 10.1084/jem.20220551] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 06/14/2022] [Accepted: 08/05/2022] [Indexed: 12/20/2022] Open
Abstract
Human adaptive-like natural killer (NK) cells express low levels of FcεRIγ (FcRγ-/low) and are reported to accumulate during COVID-19 infection; however, the mechanism underlying and regulating FcRγ expression in NK cells has yet to be fully defined. We observed lower FcRγ protein expression in NK cell subsets from lung transplant patients during rapamycin treatment, suggesting a link with reduced mTOR activity. Further, FcRγ-/low NK cell subsets from healthy donors displayed reduced mTOR activity. We discovered that FcRγ upregulation is dependent on cell proliferation progression mediated by IL-2, IL-15, or IL-12, is sensitive to mTOR suppression, and is inhibited by TGFβ or IFNα. Accordingly, the accumulation of adaptive-like FcRγ-/low NK cells in COVID-19 patients corresponded to increased TGFβ and IFNα levels and disease severity. Our results show that an adaptive-like NK cell phenotype is induced by diminished cell proliferation and has an early prognostic value for increased TGFβ and IFNα levels in COVID-19 infection associated with disease severity.
Collapse
Affiliation(s)
- Avishai Shemesh
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA
- Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, CA
| | - Yapeng Su
- Institute for Systems Biology, Seattle, WA
| | - Daniel R. Calabrese
- Department of Medicine, University of California, San Francisco, CA
- Medical Service, Veterans Affairs Health Care System, San Francisco, CA
| | - Daniel Chen
- Institute for Systems Biology, Seattle, WA
- Department of Microbiology, University of Washington, Seattle, WA
- Department of Informatics, University of Washington, Seattle, WA
| | - Janice Arakawa-Hoyt
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA
| | - Kole T. Roybal
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA
- Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, CA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA
- Chan Zuckerberg Biohub, San Francisco, CA
- Gladstone University of California, San Francisco Institute for Genetic Immunology, San Francisco, CA
- University of California, San Francisco Cell Design Institute, San Francisco, CA
| | - James R. Heath
- Institute for Systems Biology, Seattle, WA
- Department of Bioengineering, University of Washington, Seattle, WA
| | - John R. Greenland
- Department of Medicine, University of California, San Francisco, CA
- Medical Service, Veterans Affairs Health Care System, San Francisco, CA
| | - Lewis L. Lanier
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA
- Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
17
|
Demaria O, Gauthier L, Vetizou M, Blanchard Alvarez A, Vagne C, Habif G, Batista L, Baron W, Belaïd N, Girard-Madoux M, Cesari C, Caratini M, Bosco F, Benac O, Lopez J, Fenis A, Galluso J, Trichard S, Carrette B, Carrette F, Maguer A, Jaubert S, Sansaloni A, Letay-Drouet R, Kosthowa C, Lovera N, Dujardin A, Chanuc F, Le Van M, Bokobza S, Jarmuzynski N, Fos C, Gourdin N, Remark R, Lechevallier E, Fakhry N, Salas S, Deville JL, Le Grand R, Bonnafous C, Vollmy L, Represa A, Carpentier S, Rossi B, Morel A, Cornen S, Perrot I, Morel Y, Vivier E. Antitumor immunity induced by antibody-based natural killer cell engager therapeutics armed with not-alpha IL-2 variant. Cell Rep Med 2022; 3:100783. [PMID: 36260981 PMCID: PMC9589122 DOI: 10.1016/j.xcrm.2022.100783] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 07/29/2022] [Accepted: 09/21/2022] [Indexed: 11/05/2022]
Abstract
Harnessing innate immunity is emerging as a promising therapeutic approach in cancer. We report here the design of tetraspecific molecules engaging natural killer (NK) cell-activating receptors NKp46 and CD16a, the β-chain of the interleukin-2 receptor (IL-2R), and a tumor-associated antigen (TAA). In vitro, these tetraspecific antibody-based natural killer cell engager therapeutics (ANKETs) induce a preferential activation and proliferation of NK cells, and the binding to the targeted TAA triggers NK cell cytotoxicity and cytokine and chemokine production. In vivo, tetraspecific ANKETs induce NK cell proliferation and their accumulation at the tumor bed, as well as the control of local and disseminated tumors. Treatment of non-human primates with CD20-directed tetraspecific ANKET leads to CD20+ circulating B cell depletion, with minimal systemic cytokine release and no sign of toxicity. Tetraspecific ANKETs, thus, constitute a technological platform for harnessing NK cells as next-generation cancer immunotherapies. Tetraspecific ANKETs constitute a technological platform to harness NK cells in cancer Tetraspecific ANKETs target NKp46, CD16a, IL-2Rβ, and a tumor antigen Tetraspecific ANKETs stimulate NK cell proliferation, activation, and antitumor functions In vivo, tetraspecific ANKETs promote NK cell tumor accumulation and antitumor activity
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Eric Lechevallier
- Assistance Publique des Hôpitaux de Marseille, Chirurgie Urologique et Transplantation Rénale, Hôpital de la Conception, Marseille, France
| | - Nicolas Fakhry
- Assistance Publique des Hôpitaux de Marseille, ORL et Chirurgie Cervico-Faciale, Hôpital de la Conception, Marseille, France
| | - Sébastien Salas
- Assistance Publique des Hôpitaux de Marseille, Service d'Oncologie Médicale et de Soins Palliatifs, CHU Timone Adulte, Marseille, France
| | - Jean-Laurent Deville
- Assistance Publique des Hôpitaux de Marseille, Oncologie Médicale, Hôpital de la Timone, Marseille, France
| | - Roger Le Grand
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | | | | | | | | | | | | | | | | | | | - Eric Vivier
- Innate Pharma, Marseille, France,Aix Marseille University, CNRS, INSERM, CIML, Marseille, France,Assistance Publique des Hôpitaux de Marseille, Hôpital de la Timone, Marseille-Immunopôle, Marseille, France,Corresponding author
| |
Collapse
|