1
|
Moini K, Seery T, Nangia C, MacDiarmid J, Brahmbhatt H, Spilman P, Sender L, Soon-Shiong P. Recurrent pancreatic cancer treated with N-803 and PD-L1 t-haNK followed by an EGFR-targeted nanocell drug conjugate. Oncologist 2025; 30:oyae267. [PMID: 39373598 PMCID: PMC11954496 DOI: 10.1093/oncolo/oyae267] [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: 04/12/2024] [Accepted: 08/30/2024] [Indexed: 10/08/2024] Open
Abstract
Multimodal temporal therapy orchestrated to leverage immunotherapy, tumor-targeted chemotherapy, and natural killer (NK) cell therapy may provide an opportunity to induce immunogenic cell death for tumor response and increased survival in patients with recurrent cancer. The interleukin-15 (IL-15) superagonist N-803, an enhancer of NK cells, CD4 + T cells, cytotoxic CD8 + T cells, and memory T-cell activity, combined with off-the-shelf PD-L1-targeted high-affinity NK (PD-L1 t-haNK) cells represent novel immunotherapies designed to overcome an immunosuppressive tumor microenvironment (TME). The epidermal growth factor receptor-targeted antibody-nanocell conjugate E-EDV-D682 provides tumor-targeted chemotherapy in the form of its anthracycline metabolite PNU159682 (nemorubicin) cargo and is currently being studied in combination with immunomodulatory EDVs delivering the adjuvant α-galactosyl ceramide (GC). Here, we report the compassionate use treatment of this combination in a patient with recurrent, metastatic pancreatic cancer (mPC) after 3 lines of therapy. Under the initial single-patient Investigational New Drug (spIND) protocol, the patient received N-803, PD-L1 t-haNK cells, and the albumin doxorubicin conjugate aldoxorubicin for ~27 months. The patient's disease became stable on this regimen, and a transient complete response was observed by ~14 months of therapy. Due to progression, a second spIND protocol was designed whereby the patient received E-EDV-D682 plus EDV-GC for more than 24 months, which resulted in stable disease and the patient's continued survival at the time this report was written. The patient's extended survival despite the dire prognosis associated with recurrent mPC points to the merits of this temporal combination regimen in overcoming immuno-chemo resistance with enhanced immune activity required for tumor response and extended survival.
Collapse
Affiliation(s)
- Katayoun Moini
- Chan Soon-Shiong Institute for Medicine (CSSIFM), El Segundo, CA 90245, United States
| | - Tara Seery
- Chan Soon-Shiong Institute for Medicine (CSSIFM), El Segundo, CA 90245, United States
| | - Chaitali Nangia
- Chan Soon-Shiong Institute for Medicine (CSSIFM), El Segundo, CA 90245, United States
| | | | | | | | - Lennie Sender
- ImmunityBio, Inc., Culver City, CA 90232, United States
| | | |
Collapse
|
2
|
Griffiths JI, Cosgrove PA, Medina EF, Nath A, Chen J, Adler FR, Chang JT, Khan QJ, Bild AH. Cellular interactions within the immune microenvironment underpins resistance to cell cycle inhibition in breast cancers. Nat Commun 2025; 16:2132. [PMID: 40032842 PMCID: PMC11876604 DOI: 10.1038/s41467-025-56279-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 01/13/2025] [Indexed: 03/05/2025] Open
Abstract
Immune evasion by cancer cells involves reshaping the tumor microenvironment (TME) via communication with non-malignant cells. However, resistance-promoting interactions during treatment remain lesser known. Here we examine the composition, communication, and phenotypes of tumor-associated cells in serial biopsies from stage II and III high-risk estrogen receptor positive (ER+ ) breast cancers of patients receiving endocrine therapy (letrozole) as single agent or in combination with ribociclib, a CDK4/6-targeting cell cycle inhibitor. Single-cell RNA sequencing analyses on longitudinally collected samples show that in tumors overcoming the growth suppressive effects of ribociclib, first cancer cells upregulate cytokines and growth factors that stimulate immune-suppressive myeloid differentiation, resulting in reduced myeloid cell- CD8 + T-cell crosstalk via IL-15/18 signaling. Subsequently, tumors growing during treatment show diminished T-cell activation and recruitment. In vitro, ribociclib does not only inhibit cancer cell growth but also T cell proliferation and activation upon co-culturing. Exogenous IL-15 improves CDK4/6 inhibitor efficacy by augmenting T-cell proliferation and cancer cell killing by T cells. In summary, response to ribociclib in stage II and III high-risk ER + breast cancer depends on the composition, activation phenotypes and communication network of immune cells.
Collapse
Affiliation(s)
- Jason I Griffiths
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA, USA.
- Department of Mathematics, University of Utah 155 South 1400 East, Salt Lake City, UT, USA.
| | - Patrick A Cosgrove
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA, USA
| | - Eric F Medina
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA, USA
| | - Aritro Nath
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA, USA
| | - Jinfeng Chen
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA, USA
| | - Frederick R Adler
- Department of Mathematics, University of Utah 155 South 1400 East, Salt Lake City, UT, USA
- School of Biological Sciences, University of Utah 257 South 1400 East, Salt Lake City, UT, USA
| | - Jeffrey T Chang
- Department of Integrative Biology and Pharmacology, School of Medicine, School of Biomedical Informatics, UT Health Sciences Center at Houston, Houston, TX, USA
| | - Qamar J Khan
- Division of Medical Oncology, Department of Internal Medicine, The University of Kansas Medical Center, Kansas City, KS, USA
| | - Andrea H Bild
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA, USA.
| |
Collapse
|
3
|
Balkhi S, Bilato G, De Lerma Barbaro A, Orecchia P, Poggi A, Mortara L. Efficacy of Anti-Cancer Immune Responses Elicited Using Tumor-Targeted IL-2 Cytokine and Its Derivatives in Combined Preclinical Therapies. Vaccines (Basel) 2025; 13:69. [PMID: 39852848 PMCID: PMC11768832 DOI: 10.3390/vaccines13010069] [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: 11/25/2024] [Revised: 01/10/2025] [Accepted: 01/10/2025] [Indexed: 01/26/2025] Open
Abstract
Effective cancer therapies must address the tumor microenvironment (TME), a complex network of tumor cells and stromal components, including endothelial, immune, and mesenchymal cells. Durable outcomes require targeting both tumor cells and the TME while minimizing systemic toxicity. Interleukin-2 (IL-2)-based therapies have shown efficacy in cancers such as metastatic melanoma and renal cell carcinoma but are limited by severe side effects. Innovative IL-2-based immunotherapeutic approaches include immunotoxins, such as antibody-drug conjugates, immunocytokines, and antibody-cytokine fusion proteins that enhance tumor-specific delivery. These strategies activate cytotoxic CD8+ T lymphocytes and natural killer (NK) cells, eliciting a potent Th1-mediated anti-tumor response. Modified IL-2 variants with reduced Treg cell activity further improve specificity and reduce immunosuppression. Additionally, IL-2 conjugates with peptides or anti-angiogenic agents offer improved therapeutic profiles. Combining IL-2-based therapies with immune checkpoint inhibitors (ICIs), anti-angiogenic agents, or radiotherapy has demonstrated synergistic potential. Preclinical and clinical studies highlight reduced toxicity and enhanced anti-tumor efficacy, overcoming TME-driven immune suppression. These approaches mitigate the limitations of high-dose soluble IL-2 therapy, promoting immune activation and minimizing adverse effects. This review critically explores advances in IL-2-based therapies, focusing on immunotoxins, immunocytokines, and IL-2 derivatives. Emphasis is placed on their role in combination strategies, showcasing their potential to target the TME and improve clinical outcomes effectively. Also, the use of IL-2 immunocytokines in "in situ" vaccination to relieve the immunosuppression of the TME is discussed.
Collapse
Affiliation(s)
- Sahar Balkhi
- Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy; (S.B.); (G.B.); (L.M.)
| | - Giorgia Bilato
- Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy; (S.B.); (G.B.); (L.M.)
- Unit of Molecular Pathology, Biochemistry and Immunology, IRCCS MultiMedica, 20123 Milan, Italy
| | - Andrea De Lerma Barbaro
- Laboratory of Comparative Physiopathology, Department of Biotechnology and Life Sciences, University of Insubria, 20145 Varese, Italy;
| | - Paola Orecchia
- Pathology and Experimental Immunology Operative Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy;
| | - Alessandro Poggi
- SSD Oncologia Molecolare e Angiogenesi, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Lorenzo Mortara
- Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy; (S.B.); (G.B.); (L.M.)
- Unit of Molecular Pathology, Biochemistry and Immunology, IRCCS MultiMedica, 20123 Milan, Italy
| |
Collapse
|
4
|
Lv Y, Qi J, Babon JJ, Cao L, Fan G, Lang J, Zhang J, Mi P, Kobe B, Wang F. The JAK-STAT pathway: from structural biology to cytokine engineering. Signal Transduct Target Ther 2024; 9:221. [PMID: 39169031 PMCID: PMC11339341 DOI: 10.1038/s41392-024-01934-w] [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: 04/08/2024] [Revised: 06/12/2024] [Accepted: 07/16/2024] [Indexed: 08/23/2024] Open
Abstract
The Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway serves as a paradigm for signal transduction from the extracellular environment to the nucleus. It plays a pivotal role in physiological functions, such as hematopoiesis, immune balance, tissue homeostasis, and surveillance against tumors. Dysregulation of this pathway may lead to various disease conditions such as immune deficiencies, autoimmune diseases, hematologic disorders, and cancer. Due to its critical role in maintaining human health and involvement in disease, extensive studies have been conducted on this pathway, ranging from basic research to medical applications. Advances in the structural biology of this pathway have enabled us to gain insights into how the signaling cascade operates at the molecular level, laying the groundwork for therapeutic development targeting this pathway. Various strategies have been developed to restore its normal function, with promising therapeutic potential. Enhanced comprehension of these molecular mechanisms, combined with advances in protein engineering methodologies, has allowed us to engineer cytokines with tailored properties for targeted therapeutic applications, thereby enhancing their efficiency and safety. In this review, we outline the structural basis that governs key nodes in this pathway, offering a comprehensive overview of the signal transduction process. Furthermore, we explore recent advances in cytokine engineering for therapeutic development in this pathway.
Collapse
Affiliation(s)
- You Lv
- Center for Molecular Biosciences and Non-communicable Diseases Research, Xi'an University of Science and Technology, Xi'an, Shaanxi, 710054, China
- Xi'an Amazinggene Co., Ltd, Xi'an, Shaanxi, 710026, China
| | - Jianxun Qi
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100080, China
| | - Jeffrey J Babon
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Longxing Cao
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, 310024, China
| | - Guohuang Fan
- Immunophage Biotech Co., Ltd, No. 10 Lv Zhou Huan Road, Shanghai, 201112, China
| | - Jiajia Lang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Jin Zhang
- Xi'an Amazinggene Co., Ltd, Xi'an, Shaanxi, 710026, China
| | - Pengbing Mi
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Bostjan Kobe
- School of Chemistry and Molecular Biosciences, Institute for Molecular Bioscience and Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, Queensland, 4072, Australia.
| | - Faming Wang
- Center for Molecular Biosciences and Non-communicable Diseases Research, Xi'an University of Science and Technology, Xi'an, Shaanxi, 710054, China.
| |
Collapse
|
5
|
Tsiverioti CA, Gottschlich A, Trefny M, Theurich S, Anders HJ, Kroiss M, Kobold S. Beyond CAR T cells: exploring alternative cell sources for CAR-like cellular therapies. Biol Chem 2024; 405:485-515. [PMID: 38766710 DOI: 10.1515/hsz-2023-0317] [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: 10/03/2023] [Accepted: 04/18/2024] [Indexed: 05/22/2024]
Abstract
Chimeric antigen receptor (CAR)-T cell therapy has led to remarkable clinical outcomes in the treatment of hematological malignancies. However, challenges remain, such as limited infiltration into solid tumors, inadequate persistence, systemic toxicities, and manufacturing insufficiencies. The use of alternative cell sources for CAR-based therapies, such as natural killer cells (NK), macrophages (MΦ), invariant Natural Killer T (iNKT) cells, γδT cells, neutrophils, and induced pluripotent stem cells (iPSC), has emerged as a promising avenue. By harnessing these cells' inherent cytotoxic mechanisms and incorporating CAR technology, common CAR-T cell-related limitations can be effectively mitigated. We herein present an overview of the tumoricidal mechanisms, CAR designs, and manufacturing processes of CAR-NK cells, CAR-MΦ, CAR-iNKT cells, CAR-γδT cells, CAR-neutrophils, and iPSC-derived CAR-cells, outlining the advantages, limitations, and potential solutions of these therapeutic strategies.
Collapse
Affiliation(s)
| | - Adrian Gottschlich
- Division of Clinical Pharmacology, University Hospital, LMU Munich, Lindwurmstr. 2a, 80337 Munich, Germany
- Department of Medicine III, University Hospital, LMU Munich, Marchioninstr. 15, 81377 Munich, Germany
- Bavarian Cancer Research Center (BZKF), LMU Munich, Pettenkoferstr. 8a, 80336 Munich, Germany
| | - Marcel Trefny
- Division of Clinical Pharmacology, University Hospital, LMU Munich, Lindwurmstr. 2a, 80337 Munich, Germany
| | - Sebastian Theurich
- Department of Medicine III, University Hospital, LMU Munich, Marchioninstr. 15, 81377 Munich, Germany
- Bavarian Cancer Research Center (BZKF), LMU Munich, Pettenkoferstr. 8a, 80336 Munich, Germany
- 74939 German Cancer Consortium (DKTK), Partner Site Munich, A Partnership Between DKFZ and University Hospital of the LMU , Marchioninstr. 15, 81377 Munich, Germany
- Cancer and Immunometabolism Research Group, 74939 Gene Center LMU , Feodor-Lynen Str. 25, 81377 Munich, Germany
| | - Hans-Joachim Anders
- Department of Medicine IV, University Hospital, LMU Munich, Ziemssenstr. 5, 80336 Munich, Germany
| | - Matthias Kroiss
- Department of Medicine IV, University Hospital, LMU Munich, Ziemssenstr. 5, 80336 Munich, Germany
- Division of Endocrinology and Diabetes, Department of Medicine, University Hospital, University of Würzburg, Josef-Schneider-Str, 9780 Würzburg, Germany
- Comprehensive Cancer Center Mainfranken, University of Würzburg, Josef-Schneider-Str. 6, 9780 Würzburg, Germany
| | - Sebastian Kobold
- Division of Clinical Pharmacology, University Hospital, LMU Munich, Lindwurmstr. 2a, 80337 Munich, Germany
- 74939 German Cancer Consortium (DKTK), Partner Site Munich, A Partnership Between DKFZ and University Hospital of the LMU , Marchioninstr. 15, 81377 Munich, Germany
- Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München - German Research Center for Environmental Health, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
| |
Collapse
|
6
|
Boersma B, Poinot H, Pommier A. Stimulating the Antitumor Immune Response Using Immunocytokines: A Preclinical and Clinical Overview. Pharmaceutics 2024; 16:974. [PMID: 39204319 PMCID: PMC11357675 DOI: 10.3390/pharmaceutics16080974] [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: 06/03/2024] [Revised: 07/11/2024] [Accepted: 07/16/2024] [Indexed: 09/04/2024] Open
Abstract
Cytokines are immune modulators which can enhance the immune response and have been proven to be an effective class of immunotherapy. Nevertheless, the clinical use of cytokines in cancer treatment has faced several challenges associated with poor pharmacokinetic properties and the occurrence of adverse effects. Immunocytokines (ICKs) have emerged as a promising approach to overcome the pharmacological limitations observed with cytokines. ICKs are fusion proteins designed to deliver cytokines in the tumor microenvironment by taking advantage of the stability and specificity of immunoglobulin-based scaffolds. Several technological approaches have been developed. This review focuses on ICKs designed with the most impactful cytokines in the cancer field: IL-2, TNFα, IL-10, IL-12, IL-15, IL-21, IFNγ, GM-CSF, and IFNα. An overview of the pharmacological effects of the naked cytokines and ICKs tested for cancer therapy is detailed. A particular emphasis is given on the immunomodulatory effects of ICKs associated with their technological design. In conclusion, this review highlights active ways of development of ICKs. Their already promising results observed in clinical trials are likely to be improved with the advances in targeting technologies such as cytokine/linker engineering and the design of multispecific antibodies with tumor targeting and immunostimulatory functional properties.
Collapse
Affiliation(s)
- Bart Boersma
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland;
- School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland
| | - Hélène Poinot
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland;
- Translational Research Centre in Oncohaematology, University of Geneva, 1211 Geneva, Switzerland
| | - Aurélien Pommier
- UMR1240 Imagerie Moléculaire et Stratégies Théranostiques INSERM, Université Clermont Auvergne, BP 184, F-63005 Clermont-Ferrand, France
| |
Collapse
|
7
|
Yi M, Li T, Niu M, Zhang H, Wu Y, Wu K, Dai Z. Targeting cytokine and chemokine signaling pathways for cancer therapy. Signal Transduct Target Ther 2024; 9:176. [PMID: 39034318 PMCID: PMC11275440 DOI: 10.1038/s41392-024-01868-3] [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: 02/28/2024] [Revised: 04/30/2024] [Accepted: 05/11/2024] [Indexed: 07/23/2024] Open
Abstract
Cytokines are critical in regulating immune responses and cellular behavior, playing dual roles in both normal physiology and the pathology of diseases such as cancer. These molecules, including interleukins, interferons, tumor necrosis factors, chemokines, and growth factors like TGF-β, VEGF, and EGF, can promote or inhibit tumor growth, influence the tumor microenvironment, and impact the efficacy of cancer treatments. Recent advances in targeting these pathways have shown promising therapeutic potential, offering new strategies to modulate the immune system, inhibit tumor progression, and overcome resistance to conventional therapies. In this review, we summarized the current understanding and therapeutic implications of targeting cytokine and chemokine signaling pathways in cancer. By exploring the roles of these molecules in tumor biology and the immune response, we highlighted the development of novel therapeutic agents aimed at modulating these pathways to combat cancer. The review elaborated on the dual nature of cytokines as both promoters and suppressors of tumorigenesis, depending on the context, and discussed the challenges and opportunities this presents for therapeutic intervention. We also examined the latest advancements in targeted therapies, including monoclonal antibodies, bispecific antibodies, receptor inhibitors, fusion proteins, engineered cytokine variants, and their impact on tumor growth, metastasis, and the tumor microenvironment. Additionally, we evaluated the potential of combining these targeted therapies with other treatment modalities to overcome resistance and improve patient outcomes. Besides, we also focused on the ongoing research and clinical trials that are pivotal in advancing our understanding and application of cytokine- and chemokine-targeted therapies for cancer patients.
Collapse
Affiliation(s)
- Ming Yi
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310000, People's Republic of China
| | - Tianye Li
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310000, People's Republic of China
| | - Mengke Niu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Haoxiang Zhang
- Department of Hepatopancreatobiliary Surgery, Fujian Provincial Hospital, Fuzhou, 350001, People's Republic of China
| | - Yuze Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.
| | - Zhijun Dai
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310000, People's Republic of China.
| |
Collapse
|
8
|
Hall G. Interleukin-15 in kidney disease and therapeutics. Curr Opin Nephrol Hypertens 2024; 33:174-180. [PMID: 38164877 PMCID: PMC10893218 DOI: 10.1097/mnh.0000000000000964] [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: 01/03/2024]
Abstract
PURPOSE OF REVIEW Interleukin 15 (IL-15) is a member of the IL-2 family of common gamma chain receptor cytokines with well described anti-inflammatory, pro-survival and pro-proliferative signaling properties. The cytoprotective role of IL-15 in the kidney is now coming into focus with recent reports of its beneficial actions in various forms of kidney disease. This review will summarize what is currently known about IL-15 signaling in the kidney and highlight recent evidence of its beneficial effects on kidney physiology. RECENT FINDINGS IL-15 and its heterotrimeric receptor are expressed throughout the kidney. Like all IL-2 family cytokines, IL-15 can activate signaling through the Janus Kinase (JAK)/Signal transducer of activated T-cells (STAT), phosphoinositol-3 kinase (PI-3K)/AKT and mitogen activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) pathways and recent evidence suggests that STAT5B is an essential transcriptional mediator of prosurvival signaling in glomerular visceral epithelial cells (i.e. podocytes). IL-15 has also been shown to suppress pro-apoptotic signaling in models of acute kidney injury and pro-fibrotic signaling in models of chronic kidney disease. SUMMARY The cytoprotective properties of IL-15 suggest that it may have potential as a nonimmunosuppresive therapeutic for kidney disease. A novel class of IL-15 immunotherapies has emerged for the treatment cancer and some have demonstrated efficacy in clinical trials. These well tolerated IL-15 agonists could possibly be repurposed for the treatment of kidney disease and warrant further exploration.
Collapse
Affiliation(s)
- Gentzon Hall
- Division of Nephrology, Duke University, Durham, North Carolina, USA
| |
Collapse
|
9
|
Lee H, Park SH, Shin EC. IL-15 in T-Cell Responses and Immunopathogenesis. Immune Netw 2024; 24:e11. [PMID: 38455459 PMCID: PMC10917573 DOI: 10.4110/in.2024.24.e11] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 02/01/2024] [Accepted: 02/01/2024] [Indexed: 03/09/2024] Open
Abstract
IL-15 belongs to the common gamma chain cytokine family and has pleiotropic immunological functions. IL-15 is a homeostatic cytokine essential for the development and maintenance of NK cells and memory CD8+ T cells. In addition, IL-15 plays a critical role in the activation, effector functions, tissue residency, and senescence of CD8+ T cells. IL-15 also activates virtual memory T cells, mucosal-associated invariant T cells and γδ T cells. Recently, IL-15 has been highlighted as a major trigger of TCR-independent activation of T cells. This mechanism is involved in T cell-mediated immunopathogenesis in diverse diseases, including viral infections and chronic inflammatory diseases. Deeper understanding of IL-15-mediated T-cell responses and their underlying mechanisms could optimize therapeutic strategies to ameliorate host injury by T cell-mediated immunopathogenesis. This review highlights recent advancements in comprehending the role of IL-15 in relation to T cell responses and immunopathogenesis under various host conditions.
Collapse
Affiliation(s)
- Hoyoung Lee
- The Center for Viral Immunology, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon 34126, Korea
| | - Su-Hyung Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Eui-Cheol Shin
- The Center for Viral Immunology, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon 34126, Korea
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| |
Collapse
|
10
|
Schober R, Brandus B, Laeremans T, Iserentant G, Rolin C, Dessilly G, Zimmer J, Moutschen M, Aerts JL, Dervillez X, Seguin-Devaux C. Multimeric immunotherapeutic complexes activating natural killer cells towards HIV-1 cure. J Transl Med 2023; 21:791. [PMID: 37936122 PMCID: PMC10631209 DOI: 10.1186/s12967-023-04669-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/27/2023] [Indexed: 11/09/2023] Open
Abstract
BACKGROUND Combination antiretroviral therapy (cART) has dramatically extended the life expectancy of people living with HIV-1 and improved their quality of life. There is nevertheless no cure for HIV-1 infection since HIV-1 persists in viral reservoirs of latently infected CD4+ T cells. cART does not eradicate HIV-1 reservoirs or restore cytotoxic natural killer (NK) cells which are dramatically reduced by HIV-1 infection, and express the checkpoint inhibitors NKG2A or KIR2DL upregulated after HIV-1 infection. Cytotoxic NK cells expressing the homing receptor CXCR5 were recently described as key subsets controlling viral replication. METHODS We designed and evaluated the potency of "Natural killer activating Multimeric immunotherapeutic compleXes", called as NaMiX, combining multimers of the IL-15/IL-15Rα complex with an anti-NKG2A or an anti-KIR single-chain fragment variable (scFv) to kill HIV-1 infected CD4+ T cells. The oligomerization domain of the C4 binding protein was used to associate the IL-15/IL-15Rα complex to the scFv of each checkpoint inhibitor as well as to multimerize each entity into a heptamer (α form) or a dimer (β form). Each α or β form was compared in different in vitro models using one-way ANOVA and post-hoc Tukey's tests before evaluation in humanized NSG tg-huIL-15 mice having functional NK cells. RESULTS All NaMiX significantly enhanced the cytolytic activity of NK and CD8+ T cells against Raji tumour cells and HIV-1+ ACH-2 cells by increasing degranulation, release of granzyme B, perforin and IFN-γ. Targeting NKG2A had a stronger effect than targeting KIR2DL due to higher expression of NKG2A on NK cells. In viral inhibition assays, NaMiX initially increased viral replication of CD4+ T cells which was subsequently inhibited by cytotoxic NK cells. Importantly, anti-NKG2A NaMiX enhanced activation, cytotoxicity, IFN-γ production and CXCR5 expression of NK cells from HIV-1 positive individuals. In humanized NSG tg-huIL-15 mice, we confirmed enhanced activation, degranulation, cytotoxicity of NK cells, and killing of HIV-1 infected cells from mice injected with the anti-NKG2A.α NaMiX, as compared to control mice, as well as decreased total HIV-1 DNA in the lung. CONCLUSIONS NK cell-mediated killing of HIV-1 infected cells by NaMiX represents a promising approach to support HIV-1 cure strategies.
Collapse
Affiliation(s)
- Rafaëla Schober
- Department of Infection and Immunity, Luxembourg Institute of Health, 29, Rue Henri Koch, L-4354, Esch-Sur-Alzette, Luxembourg
| | - Bianca Brandus
- Department of Infection and Immunity, Luxembourg Institute of Health, 29, Rue Henri Koch, L-4354, Esch-Sur-Alzette, Luxembourg
| | - Thessa Laeremans
- Neuro-Aging and Viro-Immunotherapy (NAVI) Research Group, Faculty of Pharmacy and Medicine, Vrije Universiteit Brussel, 1090, Brussels, Belgium
| | - Gilles Iserentant
- Department of Infection and Immunity, Luxembourg Institute of Health, 29, Rue Henri Koch, L-4354, Esch-Sur-Alzette, Luxembourg
| | - Camille Rolin
- Department of Infection and Immunity, Luxembourg Institute of Health, 29, Rue Henri Koch, L-4354, Esch-Sur-Alzette, Luxembourg
| | - Géraldine Dessilly
- AIDS Reference Laboratory, Catholic University of Louvain, Ottignies-Louvain-la-Neuve, Belgium
| | - Jacques Zimmer
- Department of Infection and Immunity, Luxembourg Institute of Health, 29, Rue Henri Koch, L-4354, Esch-Sur-Alzette, Luxembourg
| | - Michel Moutschen
- Department of Infectious Diseases, University of Liège, CHU de Liège, Liège, Belgium
| | - Joeri L Aerts
- Neuro-Aging and Viro-Immunotherapy (NAVI) Research Group, Faculty of Pharmacy and Medicine, Vrije Universiteit Brussel, 1090, Brussels, Belgium
| | - Xavier Dervillez
- Department of Infection and Immunity, Luxembourg Institute of Health, 29, Rue Henri Koch, L-4354, Esch-Sur-Alzette, Luxembourg
| | - Carole Seguin-Devaux
- Department of Infection and Immunity, Luxembourg Institute of Health, 29, Rue Henri Koch, L-4354, Esch-Sur-Alzette, Luxembourg.
| |
Collapse
|
11
|
Fukushima H, Furusawa A, Kato T, Wakiyama H, Takao S, Okuyama S, Choyke PL, Kobayashi H. Intratumoral IL15 Improves Efficacy of Near-Infrared Photoimmunotherapy. Mol Cancer Ther 2023; 22:1215-1227. [PMID: 37461129 PMCID: PMC10592297 DOI: 10.1158/1535-7163.mct-23-0210] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/09/2023] [Accepted: 07/14/2023] [Indexed: 10/03/2023]
Abstract
IL15 is a potent inducer of differentiation and proliferation of CD8+ T and natural killer (NK) cells, making it a promising candidate for cancer immunotherapy. However, limited efficacy of systemic monotherapy utilizing intravenous IL15 suggests the needs for alternative routes of administration or combination treatment with other therapies. Near-infrared photoimmunotherapy (NIR-PIT) is a highly selective anticancer treatment that elicits a massive release of tumor antigens and immunogenic signals. Here, we investigated whether intratumoral IL15 can enhance the effectiveness of cancer cell-targeted NIR-PIT using syngeneic murine tumor models. Intratumoral injection of IL15 was more effective than intraperitoneal IL15 in vivo in suppressing tumor growth and inducing intratumoral immune responses. When the efficacy of CD44-targeted NIR-PIT was compared in vivo between IL15-secreting MC38 (hIL15-MC38) and parental MC38 tumors, the hIL15-MC38/NIR-PIT group showed the best tumor growth inhibition and survival. In addition, the hIL15-MC38/NIR-PIT group showed significant dendritic cell maturation and significant increases in the number and Granzyme B expression of tumor-infiltrating CD8+ T, NK, and natural killer T cells compared with the treated parental line. Furthermore, intratumoral IL15 injection combined with CD44-targeted NIR-PIT showed significant tumor control in MC38 and Pan02-luc tumor models. In bilateral tumor models, CD44-targeted NIR-PIT in hIL15-MC38 tumors significantly suppressed the growth of untreated MC38 tumors, suggesting abscopal effects. Mice that achieved complete response after the combination therapy completely rejected later tumor rechallenge. In conclusion, local IL15 administration synergistically improves the efficacy of cancer cell-targeted NIR-PIT probably by inducing stronger anticancer immunity, indicating its potential as an anticancer treatment strategy.
Collapse
Affiliation(s)
- Hiroshi Fukushima
- Molecular Imaging Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Aki Furusawa
- Molecular Imaging Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Takuya Kato
- Molecular Imaging Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Hiroaki Wakiyama
- Molecular Imaging Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Seiichiro Takao
- Molecular Imaging Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Shuhei Okuyama
- Molecular Imaging Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Peter L Choyke
- Molecular Imaging Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | | |
Collapse
|
12
|
Leidner R, Conlon K, McNeel DG, Wang-Gillam A, Gupta S, Wesolowski R, Chaudhari M, Hassounah N, Lee JB, Ho Lee L, O'Keeffe JA, Lewis N, Pavlakis GN, Thompson JA. First-in-human phase I/Ib study of NIZ985, a recombinant heterodimer of IL-15 and IL-15Rα, as a single agent and in combination with spartalizumab in patients with advanced and metastatic solid tumors. J Immunother Cancer 2023; 11:e007725. [PMID: 37907221 PMCID: PMC10619015 DOI: 10.1136/jitc-2023-007725] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/04/2023] [Indexed: 11/02/2023] Open
Abstract
BACKGROUND Preclinically, interleukin-15 (IL-15) monotherapy promotes antitumor immune responses, which are enhanced when IL-15 is used in combination with immune checkpoint inhibitors (ICIs). This first-in-human study investigated NIZ985, a recombinant heterodimer comprising physiologically active IL-15 and IL-15 receptor α, as monotherapy and in combination with spartalizumab, an anti-programmed cell death protein-1 (anti-PD-1) monoclonal antibody, in patients with advanced solid tumors. METHODS This phase I/Ib study had two dose-escalation arms: single-agent NIZ985 administered subcutaneously thrice weekly (TIW, 2 weeks on/2 weeks off) or once weekly (QW, 3 weeks on/1 week off), and NIZ985 TIW or QW administered subcutaneously plus spartalizumab (400 mg intravenously every 4 weeks (Q4W)). The dose-expansion phase investigated NIZ985 1 µg/kg TIW/spartalizumab 400 mg Q4W in patients with anti-PD-1-sensitive or anti-PD-1-resistant tumor types stratified according to approved indications. The primary objectives were the safety, tolerability, and the maximum tolerated doses (MTDs) and/or recommended dose for expansion (RDE) of NIZ985 for the dose-expansion phase. RESULTS As of February 17, 2020, 83 patients (median age: 63 years; range: 28-85) were treated in dose escalation (N=47; single-agent NIZ985: n=27; NIZ985/spartalizumab n=20) and dose expansion (N=36). No dose-limiting toxicities occurred nor was the MTD identified. The most common treatment-related adverse event (TRAE) was injection site reaction (primarily grades 1-2; single-agent NIZ985: 85% (23/27)); NIZ985/spartalizumab: 89% [50/56]). The most common grade 3-4 TRAE was decreased lymphocyte count (single-agent NIZ985: 7% [2/27]; NIZ985/spartalizumab: 5% [3/56]). The best overall response was stable disease in the single-agent arm (30% (8/27)) and partial response in the NIZ985/spartalizumab arm (5% [3/56]; melanoma, pancreatic cancer, gastric cancer). In dose expansion, the disease control rate was 45% (5/11) in the anti-PD-1-sensitive and 20% (5/25) in the anti-PD-1-resistant tumor type cohorts. Pharmacokinetic parameters were similar across arms. The transient increase in CD8+ T cell and natural killer cell proliferation and induction of several cytokines occurred in response to the single-agent and combination treatments. CONCLUSIONS NIZ985 was well tolerated in the single-agent and NIZ985/spartalizumab regimens. The RDE was established at 1 µg/kg TIW. Antitumor activity of the combination was observed against tumor types known to have a poor response to ICIs. TRIAL REGISTRATION NUMBER NCT02452268.
Collapse
Affiliation(s)
- Rom Leidner
- EACRI, Providence Cancer Institute, Portland, Oregon, USA
| | - Kevin Conlon
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Douglas G McNeel
- Department of Medicine, Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin, USA
| | - Andrea Wang-Gillam
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Sumati Gupta
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - Robert Wesolowski
- Division of Medical Oncology, James Cancer Hospital and the Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA
| | | | - Nadia Hassounah
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, USA
| | - Jong Bong Lee
- Novartis Pharmaceuticals Corporation, East Hanover, New Jersey, USA
| | - Lang Ho Lee
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, USA
| | - Jessica A O'Keeffe
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, USA
| | - Nancy Lewis
- Novartis Pharmaceuticals Corporation, East Hanover, New Jersey, USA
| | - George N Pavlakis
- Center for Cancer Research, National Cancer Institute, Frederick, Maryland, USA
| | | |
Collapse
|
13
|
Dickerson LK, Carter JA, Kohli K, Pillarisetty VG. Emerging interleukin targets in the tumour microenvironment: implications for the treatment of gastrointestinal tumours. Gut 2023; 72:1592-1606. [PMID: 37258094 DOI: 10.1136/gutjnl-2023-329650] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 05/15/2023] [Indexed: 06/02/2023]
Abstract
The effectiveness of antitumour immunity is dependent on intricate cytokine networks. Interleukins (ILs) are important mediators of complex interactions within the tumour microenvironment, including regulation of tumour-infiltrating lymphocyte proliferation, differentiation, migration and activation. Our evolving and increasingly nuanced understanding of the cell type-specific and heterogeneous effects of IL signalling has presented unique opportunities to fine-tune elaborate IL networks and engineer new targeted immunotherapeutics. In this review, we provide a primer for clinicians on the challenges and potential of IL-based treatment. We specifically detail the roles of IL-2, IL-10, IL-12 and IL-15 in shaping the tumour-immune landscape of gastrointestinal malignancies, paying particular attention to promising preclinical findings, early-stage clinical research and innovative therapeutic approaches that may properly place ILs to the forefront of immunotherapy regimens.
Collapse
Affiliation(s)
| | - Jason A Carter
- Hepatopancreatobiliary Surgery, University of Washington, Seattle, Washington, USA
| | - Karan Kohli
- Hepatopancreatobiliary Surgery, University of Washington, Seattle, Washington, USA
- Flatiron Bio, Palo Alto, California, USA
| | - Venu G Pillarisetty
- Hepatopancreatobiliary Surgery, University of Washington, Seattle, Washington, USA
| |
Collapse
|
14
|
Cody JW, Ellis-Connell AL, O’Connor SL, Pienaar E. Mathematical modeling indicates that regulatory inhibition of CD8+ T cell cytotoxicity can limit efficacy of IL-15 immunotherapy in cases of high pre-treatment SIV viral load. PLoS Comput Biol 2023; 19:e1011425. [PMID: 37616311 PMCID: PMC10482305 DOI: 10.1371/journal.pcbi.1011425] [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: 01/10/2023] [Revised: 09/06/2023] [Accepted: 08/10/2023] [Indexed: 08/26/2023] Open
Abstract
Immunotherapeutic cytokines can activate immune cells against cancers and chronic infections. N-803 is an IL-15 superagonist that expands CD8+ T cells and increases their cytotoxicity. N-803 also temporarily reduced viral load in a limited subset of non-human primates infected with simian immunodeficiency virus (SIV), a model of HIV. However, viral suppression has not been observed in all SIV cohorts and may depend on pre-treatment viral load and the corresponding effects on CD8+ T cells. Starting from an existing mechanistic mathematical model of N-803 immunotherapy of SIV, we develop a model that includes activation of SIV-specific and non-SIV-specific CD8+ T cells by antigen, inflammation, and N-803. Also included is a regulatory counter-response that inhibits CD8+ T cell proliferation and function, representing the effects of immune checkpoint molecules and immunosuppressive cells. We simultaneously calibrate the model to two separate SIV cohorts. The first cohort had low viral loads prior to treatment (≈3-4 log viral RNA copy equivalents (CEQ)/mL), and N-803 treatment transiently suppressed viral load. The second had higher pre-treatment viral loads (≈5-7 log CEQ/mL) and saw no consistent virus suppression with N-803. The mathematical model can replicate the viral and CD8+ T cell dynamics of both cohorts based on different pre-treatment viral loads and different levels of regulatory inhibition of CD8+ T cells due to those viral loads (i.e. initial conditions of model). Our predictions are validated by additional data from these and other SIV cohorts. While both cohorts had high numbers of activated SIV-specific CD8+ T cells in simulations, viral suppression was precluded in the high viral load cohort due to elevated inhibition of cytotoxicity. Thus, we mathematically demonstrate how the pre-treatment viral load can influence immunotherapeutic efficacy, highlighting the in vivo conditions and combination therapies that could maximize efficacy and improve treatment outcomes.
Collapse
Affiliation(s)
- Jonathan W. Cody
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, United States of America
| | - Amy L. Ellis-Connell
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Shelby L. O’Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Elsje Pienaar
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, United States of America
- Regenstrief Center for Healthcare Engineering, Purdue University, West Lafayette, Indiana, United States of America
| |
Collapse
|
15
|
Merino A, Maakaron J, Bachanova V. Advances in NK cell therapy for hematologic malignancies: NK source, persistence and tumor targeting. Blood Rev 2023; 60:101073. [PMID: 36959057 PMCID: PMC10979648 DOI: 10.1016/j.blre.2023.101073] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 03/07/2023] [Accepted: 03/09/2023] [Indexed: 03/14/2023]
Abstract
Natural Killer (NK) cells yield promise in therapy of hematologic malignancies. The clinical experience with adoptively transferred allogeneic NK cells over past two decades has revealed safety and minimal risk of CRS or ICANS. Unlike T cells which have to be genetically altered to avoid graft vs host disease (GVHD), HLA mismatched NK cells can be infused without GVHD risk. This makes them ideal for the development of off-the-shelf products. In this review we focus on NK biology relevant to the cancer therapy, the trajectory of NK therapeutics for leukemia, lymphoma, and myeloma; and advantages of the NK cell platform. We will also discuss novel methods to enhance NK cell targeting, persistence, and function in the tumor microenvironment. The future of NK cell therapy depends on novel strategies to realize these qualities.
Collapse
Affiliation(s)
- Aimee Merino
- Division of Hematology, Oncology, and Transplantation, University of Minnesota, 420 Delaware St, Minneapolis, MN, United States of America
| | - Joseph Maakaron
- Division of Hematology, Oncology, and Transplantation, University of Minnesota, 420 Delaware St, Minneapolis, MN, United States of America
| | - Veronika Bachanova
- Division of Hematology, Oncology, and Transplantation, University of Minnesota, 420 Delaware St, Minneapolis, MN, United States of America.
| |
Collapse
|
16
|
Wang YA, Ranti D, Bieber C, Galsky M, Bhardwaj N, Sfakianos JP, Horowitz A. NK Cell-Targeted Immunotherapies in Bladder Cancer: Beyond Checkpoint Inhibitors. Bladder Cancer 2023; 9:125-139. [PMID: 38993289 PMCID: PMC11181717 DOI: 10.3233/blc-220109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 05/15/2023] [Indexed: 07/13/2024]
Abstract
BACKGROUND For decades, immunotherapies have been integral for the treatment and management of bladder cancer, with immune checkpoint inhibitors (ICIs) transforming patient care in recent years. However, response rates are poor to T cell-targeted ICIs such as programmed cell death protein 1 (PD-1) and programmed cell death-ligand 1 (PD-L1) blocking antibodies, framing a critical need for complementary immunotherapies. Promising strategies involve harnessing the activation potential of natural killer (NK) cells. They quickly exert their antitumor activity via signaling through germline-encoded activating receptors and are rapidly sensitized to new tissue microenvironments via their regulation by polymorphic HLA class I, KIR and NKG2A receptors. OBJECTIVE In this review, we examined the roles of currently available NK-targeted antitumor treatment strategies such as engineered viral vectors, small-molecule IMiDs, NK agonist antibodies, interleukins, and chimeric antigen receptor (CAR) NK cells, and their potential for improving the efficacy of immunotherapy in the treatment of bladder cancer. METHODS Through review of current literature, we summarized our knowledge of NK cells in solid tumors and hematologic malignancies as their roles pertain to novel immunotherapies already being applied to the treatment of bladder cancer or that offer rationale for considering as potential novel immunotherapeutic strategies. RESULTS NK cells play a critical role in shaping the tumor microenvironment (TME) that can be exploited to improve T cell-targeted immunotherapies. CONCLUSIONS Emerging evidence suggests that NK cells are a prime target for improving antitumor functions in immunotherapies for the treatment of bladder cancer. Further research into profiling NK cells in settings of immunotherapies for bladder cancer could help identify patients who might maximally benefit from NK cell-targeted immunotherapies and the various approaches for exploiting their antitumor properties.
Collapse
Affiliation(s)
- Yuanshuo A Wang
- The Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daniel Ranti
- The Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christine Bieber
- The Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthew Galsky
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Hematology and Medical Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nina Bhardwaj
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Hematology and Medical Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John P Sfakianos
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Amir Horowitz
- The Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
17
|
Lui G, Minnar CM, Soon-Shiong P, Schlom J, Gameiro SR. Exploiting an Interleukin-15 Heterodimeric Agonist (N803) for Effective Immunotherapy of Solid Malignancies. Cells 2023; 12:1611. [PMID: 37371081 DOI: 10.3390/cells12121611] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/13/2023] [Accepted: 05/15/2023] [Indexed: 06/29/2023] Open
Abstract
Identifying effective immunotherapies for solid tumors remains challenging despite the significant clinical responses observed in subsets of patients treated with immune checkpoint inhibitors. Interleukin-15 (IL-15) is a promising cytokine for the treatment of cancer as it stimulates NK and CD8+ lymphocytes. However, unfavorable pharmacokinetics and safety concerns render recombinant IL-15 (rIL-15) a less attractive modality. These shortcomings were addressed by the clinical development of heterodimeric IL-15 agonists, including N803. In preclinical tumor models, N803 elicited significant Th1 immune activation and tumor suppressive effects, primarily mediated by NK and CD8+ T lymphocytes. In addition, multiple clinical studies have demonstrated N803 to be safe for the treatment of cancer patients. The combination of N803 with the immune checkpoint inhibitor nivolumab demonstrated encouraging clinical responses in nivolumab-naïve and nivolumab-refractory patients with non-small cell lung cancer. In a recent Phase II/III clinical study, most Bacillus Calmette-Guerin (BCG)-refractory bladder cancer patients treated with N803 plus BCG experienced durable complete responses. Currently, N803 is being evaluated preclinically and clinically in combination with various agents, including chemotherapeutics, immune checkpoint inhibitors, vaccines, and other immuno-oncology agents. This report will review the mechanism(s) of action of N803 and how it relates to the preclinical and clinical studies of N803.
Collapse
Affiliation(s)
- Grace Lui
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Christine M Minnar
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - Jeffrey Schlom
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sofia R Gameiro
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
18
|
Dehghani T, Shahrjerdi A, Kahrizi MS, Soleimani E, Ravandeh S, Merza MS, Rahnama N, Ebrahimzadeh F, Bakhshesh M. Targeting programmed cell death protein 1 (PD-1) for treatment of non-small-cell lung carcinoma (NSCLC); the recent advances. Pathol Res Pract 2023; 246:154470. [PMID: 37150133 DOI: 10.1016/j.prp.2023.154470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/17/2023] [Accepted: 04/18/2023] [Indexed: 05/09/2023]
Abstract
The immune system uses various immune checkpoint axes to adjust responses, support homeostasis, and deter self-reactivity and autoimmunity. Nevertheless, non-small-cell lung carcinoma (NSCLC) can use protective mechanisms to facilitate immune evasion, which leads to potentiated cancer survival and proliferation. In this light, many blocking anti-bodies have been developed to negatively regulate checkpoint molecules, in particular, programmed cell death protein 1 (PD-1) / PD-ligand 1 (L1), and bypass these immune suppressive mechanisms. Meanwhile, anti-PD-1 anti-bodies such as nivolumab, pembrolizumab, cemiplimab, and sintilimab have shown excellent competence in successfully inspiring immune responses versus NSCLC. Accordingly, the United States Food and Drug Administration (FDA) has recently approved nivolumab (alone or in combination with ipilimumab) and pembrolizumab (alone or in combination with chemotherapy) as first-line treatment for advanced NSCLC patients. However, PD-1 blockade monotherapy remains inefficient in more than 60% of NSCLC patients, and many patients don't respond or acquire resistance to this modality. Also, toxicities related to anti-PD-1 anti-body have been progressively identified in clinical trials and oncology practice. Herein, we will outline the clinical benefits of PD-1 blockade therapy alone or in combination with other treatments (e.g., chemotherapy, radiotherapy, anti-angiogenic therapy) in NSCLC patients. Moreover, we will take a glimpse into the recently identified predictive biomarkers to determine patients most likely to suffer serious adverse events to decrease untoward toxicity risk and diminish treatment costs.
Collapse
Affiliation(s)
- Tannaz Dehghani
- Department of Internal Medicine, Lorestan University of Medical Sciences, Lorestan, Iran
| | - Alireza Shahrjerdi
- National Institute for Genetic Engineering and Biotechnology (NIGEB), P.O. Box: 14965/161, Tehran, Iran
| | | | - Elnaz Soleimani
- Departmant of Genetic, Babol University of Medical Science, Babol, Iran
| | | | - Muna S Merza
- Prosthetic Dental Techniques Department, Al-Mustaqbal university College, Babylon 51001, Iraq
| | - Negin Rahnama
- Department of Internal Medicine and Health Services, Semnan University of Medical Sciences, Semnan, Iran
| | - Farnoosh Ebrahimzadeh
- Department of Internal Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Morteza Bakhshesh
- Molecular and Medicine Research Center, Khomein University of Medical Sciences, Khomein, Iran.
| |
Collapse
|
19
|
Fousek K, Horn LA, Qin H, Dahut M, Iida M, Yacubovich D, Hamilton DH, Thomas A, Schlom J, Palena C. An Interleukin-15 Superagonist Enables Antitumor Efficacy of Natural Killer Cells Against All Molecular Variants of SCLC. J Thorac Oncol 2023; 18:350-368. [PMID: 36410696 PMCID: PMC9974560 DOI: 10.1016/j.jtho.2022.11.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 11/23/2022]
Abstract
INTRODUCTION SCLC is a highly aggressive tumor with a 5-year survival rate of less than 6%. A heterogeneous disease, SCLC is classified into four subtypes that include tumors with neuroendocrine and non-neuroendocrine features. Immune checkpoint blockade has been recently added for the frontline treatment of SCLC; however, this therapy has only led to modest clinical improvements. The lack of clinical benefit in a cancer type known to have a high tumor mutational burden has been attributed to poor T-cell infiltration and low expression of MHC-class I in most SCLC tumors. In an attempt to devise a more effective immunotherapeutic regimen, this study investigated an alternate approach on the basis of the use of the clinical-stage interleukin-15 superagonist, N-803. METHODS Preclinical models of SCLC spanning all molecular subtypes were used to evaluate the susceptibility of SCLC to natural killer (NK)-mediated lysis in vitro, including NK cells activated by N-803. Antitumor activity of N-803 was evaluated in vivo with a xenograft model of SCLC. RESULTS In vitro and in vivo data revealed differences in susceptibility of SCLC subtypes to lysis by NK cells and that NK cells activated by N-803 effectively lyse SCLC tumor cells across all variant subtypes, regardless of their expression of MHC-class I. CONCLUSIONS These findings highlight the potential of a novel immune-based intervention using a cytokine-based therapeutic option for the treatment of SCLC. We hypothesize that N-803 may provide benefit to most patients with SCLC, including those with immunologically cold tumors lacking MHC expression.
Collapse
Affiliation(s)
- Kristen Fousek
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Lucas A. Horn
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Haiyan Qin
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Madeline Dahut
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Masafumi Iida
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Dan Yacubovich
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Duane H. Hamilton
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Anish Thomas
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Jeffrey Schlom
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Claudia Palena
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
20
|
Stravokefalou V, Stellas D, Karaliota S, Nagy BA, Valentin A, Bergamaschi C, Dimas K, Pavlakis GN. Heterodimeric IL-15 (hetIL-15) reduces circulating tumor cells and metastasis formation improving chemotherapy and surgery in 4T1 mouse model of TNBC. Front Immunol 2023; 13:1014802. [PMID: 36713398 PMCID: PMC9880212 DOI: 10.3389/fimmu.2022.1014802] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 12/23/2022] [Indexed: 01/15/2023] Open
Abstract
Immunotherapy has emerged as a viable approach in cancer therapy, with cytokines being of great interest. Interleukin IL-15 (IL-15), a cytokine that supports cytotoxic immune cells, has been successfully tested as an anti-cancer and anti-metastatic agent, but combinations with conventional chemotherapy and surgery protocols have not been extensively studied. We have produced heterodimeric IL-15 (hetIL-15), which has shown anti-tumor efficacy in several murine cancer models and is being evaluated in clinical trials for metastatic cancers. In this study, we examined the therapeutic effects of hetIL-15 in combination with chemotherapy and surgery in the 4T1 mouse model of metastatic triple negative breast cancer (TNBC). hetIL-15 monotherapy exhibited potent anti-metastatic effects by diminishing the number of circulating tumor cells (CTCs) and by controlling tumor cells colonization of the lungs. hetIL-15 treatment in combination with doxorubicin resulted in enhanced anti-metastatic activity and extended animal survival. Systemic immune phenotype analysis showed that the chemoimmunotherapeutic regimen shifted the tumor-induced imbalance of polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) in favor of cytotoxic effector cells, by simultaneously decreasing PMN-MDSCs and increasing the frequency and activation of effector (CD8+T and NK) cells. Tumor resection supported by neoadjuvant and adjuvant administration of hetIL-15, either alone or in combination with doxorubicin, resulted in the cure of approximately half of the treated animals and the development of anti-4T1 tumor immunity. Our findings demonstrate a significant anti-metastatic potential of hetIL-15 in combination with chemotherapy and surgery and suggest exploring the use of this regimen for the treatment of TNBC.
Collapse
Affiliation(s)
- Vasiliki Stravokefalou
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States,Department of Pharmacology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Dimitris Stellas
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States,Department of Chemical Biology, National Hellenic Research Foundation, Athens, Greece
| | - Sevasti Karaliota
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States,Basic Science Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, MD, United States
| | - Bethany A. Nagy
- Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, MD, United States
| | - Antonio Valentin
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States
| | - Cristina Bergamaschi
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States
| | - Konstantinos Dimas
- Department of Pharmacology, Faculty of Medicine, University of Thessaly, Larissa, Greece,*Correspondence: Konstantinos Dimas, ; George N. Pavlakis,
| | - George N. Pavlakis
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States,*Correspondence: Konstantinos Dimas, ; George N. Pavlakis,
| |
Collapse
|
21
|
Kowalczyk JT, Fabian KP, Padget MR, Lopez DC, Hoke ATK, Allen CT, Hermsen M, London, NR, Hodge JW. Exploiting the immunogenic potential of standard of care radiation or cisplatin therapy in preclinical models of HPV-associated malignancies. J Immunother Cancer 2022; 10:jitc-2022-005752. [PMID: 36564129 PMCID: PMC9791467 DOI: 10.1136/jitc-2022-005752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/03/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND While radiation and chemotherapy are primarily purposed for their cytotoxic effects, a growing body of preclinical and clinical evidence demonstrates an immunogenic potential for these standard therapies. Accordingly, we sought to characterize the immunogenic potential of radiation and cisplatin in human tumor models of HPV-associated malignancies. These studies may inform rational combination immuno-oncology (IO) strategies to be employed in the clinic on the backbone of standard of care, and in so doing exploit the immunogenic potential of standard of care to improve durable responses in HPV-associated malignancies. METHODS Retroviral transduction with HPV16 E7 established a novel HPV-associated sinonasal squamous cell carcinoma (SNSCC) cell line. Three established HPV16-positive cell lines were also studied (cervical carcinoma and head and neck squamous cell carcinoma). Following determination of sensitivities to standard therapies using MTT assays, flow cytometry was used to characterize induction of immunogenic cell stress following sublethal exposure to radiation or cisplatin, and the functional consequence of this induction was determined using impedance-based real time cell analysis cytotoxicity assays employing HPV16 E7-specific cytotoxic lymphocytes (CTLs) with or without N803 (IL-15/IL-15-Rα superagonist) or exogenous death receptor ligands. In vitro observations were translated using an in vivo xenograft NSG mouse model of human cervical carcinoma evaluating cisplatin in combination with CTL adoptive cell transfer. RESULTS We showed that subpopulations surviving clinically relevant doses of radiation or cisplatin therapy were more susceptible to CTL-mediated lysis in four of four tumor models of HPV-associated malignancies, serving as a model for HPV therapeutic vaccine or T-cell receptor adoptive cell transfer. This increased killing was further amplified by IL-15 agonism employing N803. We further characterized that radiation or cisplatin induced immunogenic cell stress in three of three cell lines, and consequently demonstrated that upregulated surface expression of Fas and TRAIL-R2 death receptors at least in part mediated enhanced CTL-mediated lysis. In vivo, cisplatin-induced immunogenic cell stress synergistically potentiated CTL-mediated tumor control in a human model of HPV-associated malignancy. CONCLUSION Standard of care radiation or cisplatin therapy induced immunogenic cell stress in preclinical models of HPV-associated malignancies, presenting an opportunity poised for exploitation by employing IO strategies in combination with standard of care.
Collapse
Affiliation(s)
- Joshua T Kowalczyk
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Kellsye P Fabian
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Michelle R Padget
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Diana C Lopez
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Austin TK Hoke
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Clint T Allen
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Mario Hermsen
- Department Head and Neck Cancer, Centro de Investigación Biomédica en Red, Madrid, Spain
| | - Nyall R London,
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA,Sinonasal and Skull Base Tumor Program, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland, USA,Department of Otolaryngology - Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - James W Hodge
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
22
|
Fabian KP, Kowalczyk JT, Reynolds ST, Hodge JW. Dying of Stress: Chemotherapy, Radiotherapy, and Small-Molecule Inhibitors in Immunogenic Cell Death and Immunogenic Modulation. Cells 2022; 11:cells11233826. [PMID: 36497086 PMCID: PMC9737874 DOI: 10.3390/cells11233826] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 11/11/2022] [Accepted: 11/26/2022] [Indexed: 12/03/2022] Open
Abstract
Innovative strategies to re-establish the immune-mediated destruction of malignant cells is paramount to the success of anti-cancer therapy. Accumulating evidence suggests that radiotherapy and select chemotherapeutic drugs and small molecule inhibitors induce immunogenic cell stress on tumors that results in improved immune recognition and targeting of the malignant cells. Through immunogenic cell death, which entails the release of antigens and danger signals, and immunogenic modulation, wherein the phenotype of stressed cells is altered to become more susceptible to immune attack, radiotherapies, chemotherapies, and small-molecule inhibitors exert immune-mediated anti-tumor responses. In this review, we discuss the mechanisms of immunogenic cell death and immunogenic modulation and their relevance in the anti-tumor activity of radiotherapies, chemotherapies, and small-molecule inhibitors. Our aim is to feature the immunological aspects of conventional and targeted cancer therapies and highlight how these therapies may be compatible with emerging immunotherapy approaches.
Collapse
|
23
|
Pre-Existing HSV-1 Immunity Enhances Anticancer Efficacy of a Novel Immune-Stimulating Oncolytic Virus. Viruses 2022; 14:v14112327. [PMID: 36366425 PMCID: PMC9693100 DOI: 10.3390/v14112327] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 10/03/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022] Open
Abstract
Oncolytic viruses (OVs) can specifically replicate in the host and cause cancer cell lysis while inducing an antitumor immune response. The aim of this study is to investigate the impact of either pre-existing immunity against herpes simplex virus type-1 (HSV-1) or multicycle treatment with OVs on anticancer efficacy of VG161, an HSV-1 OV in phase 2 clinical trial. VG161 efficacy was tested in CT26 mouse models by comparing the efficacy and immune response in naïve mice or in mice that were immunized with VG161. Moreover, VG161 efficacy in HLA-matched CD34+ humanized intrahepatic cholangiocarcinoma (ICC) patient-derived xenograft (PDX) models was also tested in multicycle treatment and was compared to standard chemotherapy for this type of cancer (gemcitabine). The HSV-1-immunized mice significantly inhibited tumor growth in VG161-treated mice compared to control naïve treated mice. RNA expression profiling and ELISPOT analyses indicated changes in the tumor's immune profile in the immunized and treated group compared to naïve and treated mice, as well as enhanced T cell function depicted by higher numbers of tumor specific lymphocytes, which was enhanced by immunization. In the ICC PDX model, repeated treatment of VG161 with 2 or 3 cycles seemed to increase the anticancer efficacy of VG161. In conclusion, the anticancer efficacy of VG161 can be enhanced by pre-immunization with HSV-1 and multicycle administration when the virus is given intratumorally, indicating that pre-existing antiviral immunity might enhance OV-induced antitumor immunity. Our results suggest potential clinical benefits of HSV-1-based OV therapy in HSV-1-seropositive patients and multicycle administration of VG161 for long-term maintenance treatment.
Collapse
|
24
|
Harnessing natural killer cells for cancer immunotherapy: dispatching the first responders. Nat Rev Drug Discov 2022; 21:559-577. [PMID: 35314852 PMCID: PMC10019065 DOI: 10.1038/s41573-022-00413-7] [Citation(s) in RCA: 113] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/24/2022] [Indexed: 02/07/2023]
Abstract
Natural killer (NK) cells have crucial roles in the innate immunosurveillance of cancer and viral infections. They are 'first responders' that can spontaneously recognize abnormal cells in the body, rapidly eliminate them through focused cytotoxicity mechanisms and potently produce pro-inflammatory cytokines and chemokines that recruit and activate other immune cells to initiate an adaptive response. From the initial discovery of the diverse cell surface receptors on NK cells to the characterization of regulatory events that control their function, our understanding of the basic biology of NK cells has improved dramatically in the past three decades. This advanced knowledge has revealed increased mechanistic complexity, which has opened the doors to the development of a plethora of exciting new therapeutics that can effectively manipulate and target NK cell functional responses, particularly in cancer patients. Here, we summarize the basic mechanisms that regulate NK cell biology, review a wide variety of drugs, cytokines and antibodies currently being developed and used to stimulate NK cell responses, and outline evolving NK cell adoptive transfer approaches to treat cancer.
Collapse
|
25
|
Chen X, Jiang L, Liu X. Natural killer cells: the next wave in cancer immunotherapy. Front Immunol 2022; 13:954804. [PMID: 35967421 PMCID: PMC9364606 DOI: 10.3389/fimmu.2022.954804] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 06/30/2022] [Indexed: 12/05/2022] Open
Abstract
Immunotherapies focusing on rejuvenating T cell activities, like PD-1/PD-L1 and CTLA-4 blockade, have unprecedentedly revolutionized the landscape of cancer treatment. Yet a previously underexplored component of the immune system - natural killer (NK) cell, is coming to the forefront of immunotherapeutic attempts. In this review, we discuss the contributions of NK cells in the success of current immunotherapies, provide an overview of the current preclinical and clinical strategies at harnessing NK cells for cancer treatment, and highlight that NK cell-mediated therapies emerge as a major target in the next wave of cancer immunotherapy.
Collapse
Affiliation(s)
- Xin Chen
- Department of Biology, BeiGene (Beijing) Co., Ltd., Beijing, China
| | | | | |
Collapse
|
26
|
Kim D, Park JH, Kim TY, Kim DG, Byun JH, Kim HS. Enhanced half-life and antitumor activity of Interleukin-15 through genetic fusion of a serum albumin-specific protein binder. Int J Pharm 2022; 625:122059. [PMID: 35905933 DOI: 10.1016/j.ijpharm.2022.122059] [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: 04/12/2022] [Revised: 07/07/2022] [Accepted: 07/24/2022] [Indexed: 11/26/2022]
Abstract
Human interleukin-15 (hIL-15) has attracted a considerable attention as a promising cancer immunotherapeutic due to its function to directly stimulate the proliferation and cytotoxic activity of NK and T cells. Nevertheless, a relatively short half-life of hIL-15 requires repeated administration and higher doses, causing serious side effects. Here, we demonstrate an enhanced blood half-life and biological activity of hIL-15 through genetic fusion of a human serum albumin-specific protein binder (rHSA). The fusion construct (rHSA-IL15) was observed to maintain respective binding activities for both hIL-15 receptor α and human serum albumin. The rHSA-IL15 led to a significant increase in the secretion of Granzyme B and INF-γ by immune cells compare to free hIL-15, expanding the population of activated T cell subset such as CD4 + T and CD8+ T cells. The terminal half-life of the rHSA-IL15 was prolonged by around a 40-fold in transgenic mice expressing human serum albumin, compared to free hIL-15. The rHSA-IL15 resulted in distinct anti-tumor activities in xenograft SCC (squamous cell carcinoma) mouse and allograft melanoma mouse models through activation of NK and CD8+ T cells. The rHSA-IL15 is expected to be used in cancer immunotherapy, assisting in the development of other cytokines as immunotherapeutic agents with greater efficacy.
Collapse
Affiliation(s)
- Dasom Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Jin-Ho Park
- Department of Oral and Maxillofacial Surgery, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Jinju 52727, Korea; Department of Convergence Medical Science, Gyeongsang National University, Jinju 52727, Korea
| | - Tae-Yoon Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea; Present address: Beckmann Research Institute, City of Hope, Duarte, CA, USA
| | - Dong-Gun Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - June-Ho Byun
- Department of Oral and Maxillofacial Surgery, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Jinju 52727, Korea; Department of Convergence Medical Science, Gyeongsang National University, Jinju 52727, Korea.
| | - Hak-Sung Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea.
| |
Collapse
|
27
|
Interleukin 15 in Cell-Based Cancer Immunotherapy. Int J Mol Sci 2022; 23:ijms23137311. [PMID: 35806311 PMCID: PMC9266896 DOI: 10.3390/ijms23137311] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 06/24/2022] [Accepted: 06/25/2022] [Indexed: 02/01/2023] Open
Abstract
Cell-based cancer immunotherapy, such as chimeric antigen receptor (CAR) engineered T and natural killer (NK) cell therapies, has become a revolutionary new pillar in cancer treatment. Interleukin 15 (IL-15), a potent immunostimulatory cytokine that potentiates T and NK cell immune responses, has demonstrated the reliability and potency to potentially improve the therapeutic efficacy of current cell therapy. Structurally similar to interleukin 2 (IL-2), IL-15 supports the persistence of CD8+ memory T cells while inhibiting IL-2-induced T cell death that better maintains long-term anti-tumor immunity. In this review, we describe the biology of IL-15, studies on administrating IL-15 and/or its derivatives as immunotherapeutic agents, and IL-15-armored immune cells in adoptive cell therapy. We also discuss the advantages and challenges of incorporating IL-15 in cell-based immunotherapy and provide directions for future investigation.
Collapse
|
28
|
Della Chiesa M, Setti C, Giordano C, Obino V, Greppi M, Pesce S, Marcenaro E, Rutigliani M, Provinciali N, Paleari L, DeCensi A, Sivori S, Carlomagno S. NK Cell-Based Immunotherapy in Colorectal Cancer. Vaccines (Basel) 2022; 10:1033. [PMID: 35891197 PMCID: PMC9323201 DOI: 10.3390/vaccines10071033] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 06/22/2022] [Accepted: 06/25/2022] [Indexed: 02/01/2023] Open
Abstract
Human Natural Killer (NK) cells are all round players in immunity thanks to their powerful and immediate response against transformed cells and the ability to modulate the subsequent adaptive immune response. The potential of immunotherapies based on NK cell involvement has been initially revealed in the hematological setting but has inspired the design of different immune tools to also be applied against solid tumors, including colorectal cancer (CRC). Indeed, despite cancer prevention screening plans, surgery, and chemotherapy strategies, CRC is one of the most widespread cancers and with the highest mortality rate. Therefore, further efficient and complementary immune-based therapies are in urgent need. In this review, we gathered the most recent advances in NK cell-based immunotherapies aimed at fighting CRC, in particular, the use of monoclonal antibodies targeting tumor-associated antigens (TAAs), immune checkpoint blockade, and adoptive NK cell therapy, including NK cells modified with chimeric antigen receptor (CAR-NK).
Collapse
Affiliation(s)
- Mariella Della Chiesa
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy; (C.S.); (C.G.); (V.O.); (M.G.); (S.P.); (E.M.); (S.S.)
| | - Chiara Setti
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy; (C.S.); (C.G.); (V.O.); (M.G.); (S.P.); (E.M.); (S.S.)
| | - Chiara Giordano
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy; (C.S.); (C.G.); (V.O.); (M.G.); (S.P.); (E.M.); (S.S.)
| | - Valentina Obino
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy; (C.S.); (C.G.); (V.O.); (M.G.); (S.P.); (E.M.); (S.S.)
| | - Marco Greppi
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy; (C.S.); (C.G.); (V.O.); (M.G.); (S.P.); (E.M.); (S.S.)
| | - Silvia Pesce
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy; (C.S.); (C.G.); (V.O.); (M.G.); (S.P.); (E.M.); (S.S.)
| | - Emanuela Marcenaro
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy; (C.S.); (C.G.); (V.O.); (M.G.); (S.P.); (E.M.); (S.S.)
| | | | | | - Laura Paleari
- A.Li.Sa., Liguria Region Health Authority, 16121 Genoa, Italy;
| | - Andrea DeCensi
- Medical Oncology, Galliera Hospital, 16128 Genoa, Italy; (N.P.); (A.D.)
| | - Simona Sivori
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy; (C.S.); (C.G.); (V.O.); (M.G.); (S.P.); (E.M.); (S.S.)
| | - Simona Carlomagno
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy; (C.S.); (C.G.); (V.O.); (M.G.); (S.P.); (E.M.); (S.S.)
| |
Collapse
|
29
|
Zhang W, Zhang Q, Yang N, Shi Q, Su H, Lin T, He Z, Wang W, Guo H, Shen P. Crosstalk between IL-15Rα + tumor-associated macrophages and breast cancer cells reduces CD8 + T cell recruitment. Cancer Commun (Lond) 2022; 42:536-557. [PMID: 35615815 PMCID: PMC9198341 DOI: 10.1002/cac2.12311] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 02/07/2022] [Accepted: 05/10/2022] [Indexed: 12/23/2022] Open
Abstract
Background Interleukin‐15 (IL‐15) is a promising immunotherapeutic agent owing to its powerful immune‐activating effects. However, the clinical benefits of these treatments are limited. Crosstalk between tumor cells and immune cells plays an important role in immune escape and immunotherapy drug resistance. Herein, this study aimed to obtain in‐depth understanding of crosstalk in the tumor microenvironment for providing potential therapeutic strategies to prevent tumor progression. Methods T‐cell killing assays and co‐culture models were developed to determine the role of crosstalk between macrophages and tumor cells in breast cancer resistant to IL‐15. Western blotting, histological analysis, CRISPR‐Cas9 knockout, multi‐parameter flow cytometry, and tumor cell‐macrophage co‐injection mouse models were developed to examine the mechanism by which IL‐15Rα+ tumor‐associated macrophages (TAMs) regulate breast cancer cell resistance to IL‐15. Results We found that macrophages contributed to the resistance of tumor cells to IL‐15, and tumor cells induced macrophages to express high levels of the α subunit of the IL‐15 receptor (IL‐15Rα). Further investigation showed that IL‐15Rα+ TAMs reduced the protein levels of chemokine CX3C chemokine ligand 1 (CX3CL1) in tumor cells to inhibit the recruitment of CD8+ T cells by releasing the IL‐15/IL‐15Rα complex (IL‐15Rc). Administration of an IL‐15Rc blocking peptide markedly suppressed breast tumor growth and overcame the resistance of cancer cells to anti‐ programmed cell death protein 1 (PD‐1) antibody immunotherapy. Interestingly, Granulocyte‐macrophage colony‐stimulating factor (GMCSF) induced γ chain (γc) expression to promote tumor cell‐macrophage crosstalk, which facilitated tumor resistance to IL‐15. Additionally, we observed that the non‐transcriptional regulatory function of hypoxia inducible factor‐1alpha (HIF‐1α) was essential for IL‐15Rc to regulate CX3CL1 expression in tumor cells. Conclusions The IL‐15Rc‐HIF‐1α‐CX3CL1 signaling pathway serves as a crosstalk between macrophages and tumor cells in the tumor microenvironment of breast cancer. Targeting this pathway may provide a potential therapeutic strategy for enhancing the efficacy of cancer immunotherapy.
Collapse
Affiliation(s)
- Wenlong Zhang
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, P. R. China.,Department of Urology, Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, Nanjing, Jiangsu, 210008, P. R. China
| | - Qing Zhang
- Department of Urology, Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, Nanjing, Jiangsu, 210008, P. R. China
| | - Nanfei Yang
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, P. R. China
| | - Qian Shi
- Department of Cellular and Integrative Physiology, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, 78229-3904, USA
| | - Huifang Su
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, P. R. China
| | - Tingsheng Lin
- Department of Urology, Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, Nanjing, Jiangsu, 210008, P. R. China
| | - Zhonglei He
- Charles Institute of Dermatology, School of Medicine, University College Dublin, Dublin, Eircode D04 V1W8, Ireland
| | - Wenxin Wang
- Charles Institute of Dermatology, School of Medicine, University College Dublin, Dublin, Eircode D04 V1W8, Ireland
| | - Hongqian Guo
- Department of Urology, Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, Nanjing, Jiangsu, 210008, P. R. China
| | - Pingping Shen
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, P. R. China.,Shenzhen Research Institute of Nanjing University, Shenzhen, 518000, China
| |
Collapse
|
30
|
Sajid M, Liu L, Sun C. The Dynamic Role of NK Cells in Liver Cancers: Role in HCC and HBV Associated HCC and Its Therapeutic Implications. Front Immunol 2022; 13:887186. [PMID: 35669776 PMCID: PMC9165341 DOI: 10.3389/fimmu.2022.887186] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 04/12/2022] [Indexed: 11/21/2022] Open
Abstract
Hepatocellular carcinoma (HCC) remains an important complication of chronic liver disease, especially when cirrhosis occurs. Existing treatment strategies include surgery, loco-regional techniques, and chemotherapy. Natural killer cells are distinctive cytotoxic lymphocytes that play a vital role in fighting tumors and infections. As an important constituent of the innate immune system against cancer, phenotypic and functional deviations of NK cells have been demonstrated in HCC patients who also exhibit perturbation of the NK-activating receptor/ligand axis. The rate of recurrence of tumor-infiltrating and circulating NK cells are positively associated with survival benefits in HCC and have prognostic significance, suggesting that NK cell dysfunction is closely related to HCC progression. NK cells are the first-line effector cells of viral hepatitis and play a significant role by directly clearing virus-infected cells or by activating antigen-specific T cells by producing IFN-γ. In addition, chimeric antigen receptor (CAR) engineered NK cells suggest an exclusive opportunity to produce CAR-NKs with several specificities with fewer side effects. In the present review, we comprehensively discuss the innate immune landscape of the liver, particularly NK cells, and the impact of tumor immune microenvironment (TIME) on the function of NK cells and the biological function of HCC. Furthermore, the role of NK cells in HCC and HBV-induced HCC has also been comprehensively elaborated. We also elaborate on available NK cell-based immunotherapeutic approaches in HCC treatment and summarize current advancements in the treatment of HCC. This review will facilitate researchers to understand the importance of the innate immune landscape of NK cells and lead to devising innovative immunotherapeutic strategies for the systematic treatment of HCC.
Collapse
Affiliation(s)
- Muhammad Sajid
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Heifei, China
- Transplant and Immunology Laboratory, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Lianxin Liu
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Heifei, China
| | - Cheng Sun
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Heifei, China
- Transplant and Immunology Laboratory, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Immunology, University of Science and Technology of China, Hefei, China
| |
Collapse
|
31
|
Advances of research of Fc-fusion protein that activate NK cells for tumor immunotherapy. Int Immunopharmacol 2022; 109:108783. [PMID: 35561479 DOI: 10.1016/j.intimp.2022.108783] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 04/02/2022] [Accepted: 04/14/2022] [Indexed: 12/21/2022]
Abstract
The rapid development of bioengineering technology has introduced Fc-fusion proteins, representing a novel kind of recombinant protein, as promising biopharmaceutical products in tumor therapy. Numerous related anti-tumor Fc-fusion proteins have been investigated and are in different stages of development. Fc-fusion proteins are constructed by fusing the Fc-region of the antibody with functional proteins or peptides. They retain the bioactivity of the latter and partial properties of the former. This structural and functional advantage makes Fc-fusion proteins an effective tool in tumor immunotherapy, especially for the recruitment and activation of natural killer (NK) cells, which play a critical role in tumor immunotherapy. Even though tumor cells have developed mechanisms to circumvent the cytotoxic effect of NK cells or induce defective NK cells, Fc-fusion proteins have been proven to effectively activate NK cells to kill tumor cells in different ways, such as antibody-dependent cell-mediated cytotoxicity (ADCC), activate NK cells in different ways in order to promote killing of tumor cells. In this review, we focus on NK cell-based immunity for cancers and current research progress of the Fc-fusion proteins for anti-tumor therapy by activating NK cells.
Collapse
|
32
|
Goldenson BH, Hor P, Kaufman DS. iPSC-Derived Natural Killer Cell Therapies - Expansion and Targeting. Front Immunol 2022; 13:841107. [PMID: 35185932 PMCID: PMC8851389 DOI: 10.3389/fimmu.2022.841107] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 01/13/2022] [Indexed: 12/15/2022] Open
Abstract
Treatment of cancer with allogeneic natural killer (NK) cell therapies has seen rapid development, especially use against hematologic malignancies. Clinical trials of NK cell-based adoptive transfer to treat relapsed or refractory malignancies have used peripheral blood, umbilical cord blood and pluripotent stem cell-derived NK cells, with each approach undergoing continued clinical development. Improving the potency of these therapies relies on genetic modifications to improve tumor targeting and to enhance expansion and persistence of the NK cells. Induced pluripotent stem cell (iPSC)-derived NK cells allow for routine targeted introduction of genetic modifications and expansion of the resulting NK cells derived from a clonal starting cell population. In this review, we discuss and summarize recent important advances in the development of new iPSC-derived NK cell therapies, with a focus on improved targeting of cancer. We then discuss improvements in methods to expand iPSC-derived NK cells and how persistence of iPSC-NK cells can be enhanced. Finally, we describe how these advances may combine in future NK cell-based therapy products for the treatment of both hematologic malignancies and solid tumors.
Collapse
Affiliation(s)
- Benjamin H Goldenson
- Department of Medicine, Division of Regenerative Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Pooja Hor
- Department of Medicine, Division of Regenerative Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Dan S Kaufman
- Department of Medicine, Division of Regenerative Medicine, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
33
|
Rubinstein MP, Williams C, Mart C, Beall J, MacPherson L, Azar J, Swiderska-Syn M, Manca P, Gibney BC, Robinson MD, Krieg C, Hill EG, Taha SA, Rock AD, Lee JH, Soon-Shiong P, Wrangle J. Phase I Trial Characterizing the Pharmacokinetic Profile of N-803, a Chimeric IL-15 Superagonist, in Healthy Volunteers. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:1362-1370. [PMID: 35228263 DOI: 10.4049/jimmunol.2100066] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 01/03/2022] [Indexed: 12/24/2022]
Abstract
The oncotherapeutic promise of IL-15, a potent immunostimulant, is limited by a short serum t 1/2 The fusion protein N-803 is a chimeric IL-15 superagonist that has a >20-fold longer in vivo t 1/2 versus IL-15. This phase 1 study characterized the pharmacokinetic (PK) profile and safety of N-803 after s.c. administration to healthy human volunteers. Volunteers received two doses of N-803, and after each dose, PK and safety were assessed for 9 d. The primary endpoint was the N-803 PK profile, the secondary endpoint was safety, and immune cell levels and immunogenicity were measures of interest. Serum N-803 concentrations peaked 4 h after administration and declined with a t 1/2 of ∼20 h. N-803 did not cause treatment-emergent serious adverse events (AEs) or grade ≥3 AEs. Injection site reactions, chills, and pyrexia were the most common AEs. Administration of N-803 was well tolerated and accompanied by proliferation of NK cells and CD8+ T cells and sustained increases in the number of NK cells. Our results suggest that N-803 administration can potentiate antitumor immunity.
Collapse
Affiliation(s)
- Mark P Rubinstein
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University James Comprehensive Cancer Center, Columbus, OH
- Department of Internal Medicine, The Ohio State University, Columbus, OH
| | - Cameron Williams
- Department of Surgery, Medical University of South Carolina, Charleston, SC
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC
| | - Caroline Mart
- Department of Surgery, Medical University of South Carolina, Charleston, SC
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC
| | - Jonathan Beall
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC
| | - Linda MacPherson
- Department of Surgery, Medical University of South Carolina, Charleston, SC
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC
| | - Joseph Azar
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University James Comprehensive Cancer Center, Columbus, OH
- Department of Internal Medicine, The Ohio State University, Columbus, OH
| | - Marzena Swiderska-Syn
- Department of Surgery, Medical University of South Carolina, Charleston, SC
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC
| | - Paolo Manca
- Department of Medical Oncology, IRCCS Foundation - National Cancer Institute, Milan, Italy
| | - Barry C Gibney
- Department of Surgery, Medical University of South Carolina, Charleston, SC
| | - Mark D Robinson
- Department of Molecular Life Sciences and SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich, Switzerland
| | - Carsten Krieg
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC
| | - Elizabeth G Hill
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC
| | | | | | | | | | - John Wrangle
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC;
- Division of Hematology and Oncology, Department of Medicine, Medical University of South Carolina, Charleston, SC
| |
Collapse
|
34
|
Mylod E, Lysaght J, Conroy MJ. Natural killer cell therapy: A new frontier for obesity-associated cancer. Cancer Lett 2022; 535:215620. [PMID: 35283210 DOI: 10.1016/j.canlet.2022.215620] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 02/16/2022] [Accepted: 03/03/2022] [Indexed: 02/09/2023]
Abstract
Natural killer (NK) cell infiltration of solid tumours is associated with better outcomes, placing augmentation of NK cell abundance in tumours as an attractive immunotherapeutic approach. The unique ability of NK cells to target cancer cells without antigen specificity increases their versatility and applicability as an immunotherapeutic tool. However, successful utilisation of NK cell-based therapies in solid tumours is still at an early stage. Obesity has become a global health epidemic, and the prevalence of obesity-associated cancers has significantly increased. Obesity-associated malignancies provide a unique challenge for the successful application of cell-based immunotherapies including NK cell-based therapies because significant numbers of NK and T cells are recruited to the visceral adipose tissue at the expense of successful tumour infiltration and eradication. As such, immunotherapy efficacy has been disappointing for obesity-associated malignancies such as oesophageal and gastric adenocarcinoma. Therefore, immunotherapies for obesity-associated cancers warrant our further attention. Indeed, it is becoming ever more obvious that more innovative approaches are needed to re-invigorate anti-tumour immunity and overcome immune exclusion in such tumours. In this review, we briefly summarise the dysfunctionality of NK cells in obesity-associated cancer. We outline the NK cell-based immunotherapeutic approaches which hold promise as effective treatments in this disease space, including CAR-NK cells. Furthermore, we suggest future avenues which possess the potential to transform immunotherapy and specifically NK cell therapy efficacy for obesity-associated cancer.
Collapse
Affiliation(s)
- Eimear Mylod
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity Translational Medicine Institute and Trinity St. James's Cancer Institute, St. James's Hospital, Trinity College Dublin, Dublin, 8, Ireland
| | - Joanne Lysaght
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity Translational Medicine Institute and Trinity St. James's Cancer Institute, St. James's Hospital, Trinity College Dublin, Dublin, 8, Ireland
| | - Melissa J Conroy
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity Translational Medicine Institute and Trinity St. James's Cancer Institute, St. James's Hospital, Trinity College Dublin, Dublin, 8, Ireland; Cancer Immunology Research Group, Department of Physiology, School of Medicine, Trinity College Dublin, Dublin, 2, Ireland.
| |
Collapse
|
35
|
Wolfarth AA, Dhar S, Goon JB, Ezeanya UI, Ferrando-Martínez S, Lee BH. Advancements of Common Gamma-Chain Family Cytokines in Cancer Immunotherapy. Immune Netw 2022; 22:e5. [PMID: 35291658 PMCID: PMC8901704 DOI: 10.4110/in.2022.22.e5] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 02/15/2022] [Accepted: 02/15/2022] [Indexed: 12/01/2022] Open
Affiliation(s)
| | - Swati Dhar
- NeoImmuneTech, Inc., Rockville, MD 20850, USA
| | | | | | | | | |
Collapse
|
36
|
Wang G, Yang Y, Yi D, Yuan L, Yin PH, Ke X, Jun-Jie W, Tao MF. Eudragit S100 prepared pH-responsive liposomes-loaded betulinic acid against colorectal cancer in vitro and in vivo. J Liposome Res 2021; 32:250-264. [PMID: 34895013 DOI: 10.1080/08982104.2021.1999974] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
This study aimed to develop polymer Eudragit S100 for preparing pH-responsive liposomes-loaded betulinic acid (pH-BA-LP) to improve the therapeutic index of chemotherapy for colorectal cancer. BA-loaded liposomes were coated with Eudragit S100 by a thin film dispersion and easily scalable pH-driven method. The prepared liposomes were evaluated for size, surface morphology, entrapment efficiency, stability, in vitro drug release, and antitumor activity. In particular, pH-BA-LP showed advantages such as lower size (<100 nm), encapsulation efficiency of 90%, high stability, and stably cumulative release. By detecting the antitumor effects of pH-BA-LP in vivo, it showed that the tumor proliferation and cell migration were significantly inhibited in colorectal cancer. The pH-BA-LP also inhibited tumor growth via the regulation of Akt/TLR-mediated signalling and significantly down-regulated the expression of NFAT1 and NFAT4 proteins. It was found that pH-BA-LP can increase NK cells and CD3+ cells in tumor tissues, and the proportion of CD8+ cells in CD3+ cells was also increased, which proved that pH-BA-LP can play an antitumor effect by enhancing the autoimmunity level in tumor-bearing mice. The positive infiltration rates of CD8 and CD68 were increased and CD163 was relatively decreased by using pH-BA-LP, which proved that pH-BA-LP can regulate the immune infiltration levels in tumor-bearing mice. Therefore, the present work provides an effective method to prepare pH-responsive polymer-coated liposomes for colonic delivery with biologically active compounds.
Collapse
Affiliation(s)
- Gang Wang
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Jiangsu University, Shanghai, China
| | - Yu Yang
- Jiangsu University School of Pharmacy, Zhenjiang City, China
| | - Du Yi
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Jiangsu University, Shanghai, China
| | - Lu Yuan
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Jiangsu University, Shanghai, China
| | - Pei-Hao Yin
- Interventional Cancer Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xu Ke
- Interventional Cancer Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wang Jun-Jie
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Jiangsu University, Shanghai, China
| | - Min-Fang Tao
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Jiangsu University, Shanghai, China
| |
Collapse
|
37
|
Wang G, Yu Y, Li ZM, Zhu ZM, Wang ZJ, Tao MF. Triterpenoids of Rhus chinensis Supressed Colorectal Cancer Progress by Enhancing Antitumor Immunity and CD8 + T Cells Tumor Infiltration. Nutr Cancer 2021; 74:2550-2564. [PMID: 34866510 DOI: 10.1080/01635581.2021.2009523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The absence of CD8+ T cells in the tumor center has become a major obstacle in the immunotherapy of colorectal cancer. Therefore, new therapeutic strategies are urgently needed to promote the accumulation of CD8+ T cells in the tumor center. Previous studies have shown that triterpenoid of Rhus chinensis (TER) is involved in the proliferation and apoptosis of colorectal cancer cells, and can regulate their immune activity, but its mechanism needs to be further elucidated. In this study, the antitumor effect and adaptive immune response of TER on tumor-bearing mice were evaluated and compared with 5-fluorouracil. The results showed that TER could significantly inhibit tumor growth and prolong the survival time of tumor-bearing mice. The In Vivo studies have shown that TER can not only enhance antitumor immunity and promote the accumulation of CD8 + T cells to tumor sites, but also inhibit tumor progression by regulating the expression of PD-1 and PD-L1 and significantly reducing the mortality of mice. Our study demonstrated for the first time that TER has oncolytic effect, and recruited adaptive immune cells to enhance the efficacy of anti-PD-1/PD-L1 in colorectal cancer, which provides a potential therapeutic target for combined immunotherapy of colorectal cancer.
Collapse
Affiliation(s)
- Gang Wang
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Jiangsu University, Shanghai, China
| | - Yang Yu
- Department of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Zi-Meng Li
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Jiangsu University, Shanghai, China
| | - Zhi-Min Zhu
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Jiangsu University, Shanghai, China
| | - Zhi-Jie Wang
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Jiangsu University, Shanghai, China
| | - Min-Fang Tao
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Jiangsu University, Shanghai, China
| |
Collapse
|
38
|
Hoke AT, Padget MR, Fabian KP, Nandal A, Gallia GL, Bilusic M, Soon-Shiong P, Hodge JW, London NR. Combinatorial Natural Killer Cell-based Immunotherapy Approaches Selectively Target Chordoma Cancer Stem Cells. CANCER RESEARCH COMMUNICATIONS 2021; 1:127-139. [PMID: 35765577 PMCID: PMC9236084 DOI: 10.1158/2767-9764.crc-21-0020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Chordoma is a rare tumor derived from notochord remnants that has a propensity to recur and metastasize despite conventional multimodal treatment. Cancer stem cells (CSC) are implicated in chordoma's resistant and recurrent behavior; thus strategies that target CSCs are of particular interest. Using in vitro cytotoxicity models, we demonstrated that anti-programmed death-ligand 1 (N-601) and anti-epidermal growth factor receptor (cetuximab) antibodies enhanced lysis of chordoma cells by healthy donor and chordoma patient NK cells through antibody-dependent cellular cytotoxicity (ADCC). Treatment of NK cells with an IL-15 superagonist complex (N-803) increased their cytotoxicity against chordoma cells, which was further enhanced by treatment with N-601 and/or cetuximab. PD-L1-targeted chimeric antigen receptor NK cells (PD-L1 t-haNKs) were also effective against chordoma cells. CSCs were preferentially vulnerable to NK cell killing in the presence of N-601 and N-803. Flow cytometric analysis of a chordoma CSC population showed that CSCs expressed significantly more NK activating ligand B7-H6 and PD-L1 than non-CSCs, thus explaining a potential mechanism of selective targeting. These data suggest that chordoma may be effectively targeted by combinatorial NK cell-mediated immunotherapeutic approaches and that the efficacy of these approaches in chordoma and other CSC-driven tumor types should be investigated further in clinical studies.
Collapse
Affiliation(s)
- Austin T.K. Hoke
- Sinonasal and Skull Base Tumor Program, National Institutes on Deafness and Other Communication Disorders, NIH, Bethesda, Maryland
- University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, North Carolina
| | - Michelle R. Padget
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Kellsye P. Fabian
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Anjali Nandal
- Sinonasal and Skull Base Tumor Program, National Institutes on Deafness and Other Communication Disorders, NIH, Bethesda, Maryland
| | - Gary L. Gallia
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Marijo Bilusic
- Genitourinary Malignancies Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | | | - James W. Hodge
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Nyall R. London
- Sinonasal and Skull Base Tumor Program, National Institutes on Deafness and Other Communication Disorders, NIH, Bethesda, Maryland
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Corresponding Author: Nyall R. London Jr., Sinonasal and Skull Base Tumor Program, National Institute on Deafness and Other Communications Disorders (NIDCD), NIH, 10 Center Drive Room #7N256, Bethesda, MD USA 20892-2320. Phone: 301-402-4216; E-mail:
| |
Collapse
|
39
|
Conlon K, Watson DC, Waldmann TA, Valentin A, Bergamaschi C, Felber BK, Peer CJ, Figg WD, Potter EL, Roederer M, McNeel DG, Thompson JA, Gupta S, Leidner R, Wang-Gillam A, Parikh NS, Long D, Kurtulus S, Ho Lee L, Chowdhury NR, Bender F, Pavlakis GN. Phase I study of single agent NIZ985, a recombinant heterodimeric IL-15 agonist, in adult patients with metastatic or unresectable solid tumors. J Immunother Cancer 2021; 9:jitc-2021-003388. [PMID: 34799399 PMCID: PMC8606766 DOI: 10.1136/jitc-2021-003388] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/22/2021] [Indexed: 11/30/2022] Open
Abstract
Background NIZ985 is a recombinant heterodimer of physiologically active interleukin (IL-)15 and IL-15 receptor alpha. In preclinical models, NIZ985 promotes cytotoxic lymphocyte proliferation, killing function, and organ/tumor infiltration, with resultant anticancer effects. In this first-in-human study, we assessed the safety, pharmacokinetics, and immune effects of NIZ985 in patients with metastatic or unresectable solid tumors. Methods Single agent NIZ985 dose escalation data are reported from a phase I dose escalation/expansion study of NIZ985 as monotherapy. Adult patients (N=14) received 0.25, 0.5, 1, 2 or 4 µg/kg subcutaneous NIZ985 three times weekly (TIW) for the first 2 weeks of each 28-day cycle, in an accelerated 3+3 dose escalation trial design. IL-15 and endogenous cytokines were monitored by ELISA and multiplexed electrochemiluminescent assays. Multiparameter flow cytometry assessed the frequency, phenotype and proliferation of peripheral blood mononuclear cells. Preliminary antitumor activity was assessed by overall response rate (Response Evaluation Criteria in Solid Tumors V.1.1). Results As of March 2, 2020, median treatment duration was 7.5 weeks (range 1.1–77.1). Thirteen patients had discontinued and one (uveal melanoma) remains on treatment with stable disease. Best clinical response was stable disease (3 of 14 patients; 21%). The most frequent adverse events (AEs) were circular erythematous injection site reactions (100%), chills (71%), fatigue (57%), and fever (50%). Treatment-related grade 3/4 AEs occurred in six participants (43%); treatment-related serious AEs (SAEs) in three (21%). The per-protocol maximum tolerated dose was not reached. Pharmacokinetic accumulation of serum IL-15 in the first week was followed by significantly lower levels in week 2, likely due to more rapid cytokine consumption by an expanding lymphocyte pool. NIZ985 treatment was associated with increases in several cytokines, including interferon (IFN)-γ, IL-18, C-X-C motif chemokine ligand 10, and tumor necrosis factor-β, plus significant induction of cytotoxic lymphocyte proliferation (including natural killer and CD8+ T cells), increased CD16+ monocytes, and increased CD163+ macrophages at injection sites. Conclusions Subcutaneous NIZ985 TIW was generally well tolerated in patients with advanced cancer and produced immune activation paralleling preclinical observations, with induction of IFN-γ and proliferation of cytotoxic lymphocytes. Due to delayed SAEs at the two highest dose levels, administration is being changed to once-weekly in a revised protocol, as monotherapy and combined with checkpoint inhibitor spartalizumab. These alterations are expected to maximize the potential of NIZ985 as a novel immunotherapy. Trial registration number NCT02452268.
Collapse
Affiliation(s)
- Kevin Conlon
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Dionysios C Watson
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland, USA.,University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Thomas A Waldmann
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Antonio Valentin
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland, USA
| | - Cristina Bergamaschi
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, USA
| | - Barbara K Felber
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, USA
| | - Cody J Peer
- Clinical Pharmacology Program, Center for Cancer Research, NCI, Bethesda, Maryland, USA
| | - William D Figg
- Clinical Pharmacology Program, Center for Cancer Research, NCI, Bethesda, Maryland, USA
| | - E Lake Potter
- Vaccine Research Center, NIAID, Bethesda, Maryland, USA
| | | | - Douglas G McNeel
- Carbone Cancer Center, University of Wisconsin Madison, Madison, Wisconsin, USA
| | | | - Sumati Gupta
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - Rom Leidner
- Earle A Chiles Research Institute, Providence Cancer Institute, Portland, Oregon, USA
| | - Andrea Wang-Gillam
- Division of Oncology, Department of Medicine, Washington University in Saint Louis, St Louis, Missouri, USA
| | - Nehal S Parikh
- Novartis Institutes for BioMedical Research Inc, Cambridge, Massachusetts, USA
| | - Debby Long
- Novartis Institutes for BioMedical Research Inc, Cambridge, Massachusetts, USA
| | - Sema Kurtulus
- Novartis Institutes for BioMedical Research Inc, Cambridge, Massachusetts, USA
| | - Lang Ho Lee
- Novartis Institutes for BioMedical Research Inc, Cambridge, Massachusetts, USA
| | | | - Florent Bender
- Novartis Institutes for BioMedical Research Inc, Cambridge, Massachusetts, USA
| | - George N Pavlakis
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland, USA
| |
Collapse
|
40
|
Fiore PF, Di Matteo S, Tumino N, Mariotti FR, Pietra G, Ottonello S, Negrini S, Bottazzi B, Moretta L, Mortier E, Azzarone B. Interleukin-15 and cancer: some solved and many unsolved questions. J Immunother Cancer 2021; 8:jitc-2020-001428. [PMID: 33203664 PMCID: PMC7674108 DOI: 10.1136/jitc-2020-001428] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/22/2020] [Indexed: 12/29/2022] Open
Abstract
Soluble interleukin (IL)-15 exists under two forms: as monomer (sIL-15) or as heterodimeric complex in association with sIL-15Rα (sIL-15/IL-15Rα). Both forms have been successfully tested in experimental tumor murine models and are currently undergoing investigation in phase I/II clinical trials. Despite more than 20 years research on IL-15, some controversial issues remain to be addressed. A first point concerns the detection of the sIL-15/IL-15Rα in plasma of healthy donors or patients with cancer and its biological significance. The second and third unsolved question regards the protumorigenic role of the IL-15/IL-15Rα complex in human cancer and the detrimental immunological consequences associated to prolonged exposure of natural killer (NK) cells to both forms of soluble IL-15, respectively. Data suggest that in vivo prolonged or repeated exposure to monomeric sIL-15 or the soluble complex may lead to NK hypo-responsiveness through the expansion of the CD8+/CD44+ T cell subset that would suppress NK cell functions. In vitro experiments indicate that soluble complex and monomeric IL-15 may cause NK hyporesponsiveness through a direct effect caused by their prolonged stimulation, suggesting that this mechanism could also be effective in vivo. Therefore, a better knowledge of IL-15 and a more appropriate use of both its soluble forms, in terms of concentrations and time of exposure, are essential in order to improve their therapeutic use. In cancer, the overproduction of sIL-15/IL-15Rα could represent a novel mechanism of immune escape. The soluble complex may act as a decoy cytokine unable to efficiently foster NK cells, or could induce NK hyporesponsiveness through an excessive and prolonged stimulation depending on the type of IL-15Rα isoforms associated. All these unsolved questions are not merely limited to the knowledge of IL-15 pathophysiology, but are crucial also for the therapeutic use of this cytokine. Therefore, in this review, we will discuss key unanswered issues on the heterogeneity and biological significance of IL-15 isoforms, analyzing both their cancer-related biological functions and their therapeutic implications.
Collapse
Affiliation(s)
| | - Sabina Di Matteo
- Immunology Area, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Nicola Tumino
- Immunology Area, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | - Gabriella Pietra
- Immuology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,Department of Experimental Medicine (DiMES), University of Genoa, Genoa, Italy
| | - Selene Ottonello
- Department of Experimental Medicine (DiMES), University of Genoa, Genoa, Italy.,Center of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| | - Simone Negrini
- Clinical Immunology Unit, Department of Internal Medicine, University of Genoa and Ospedale Policlinico San Martino, Genova, Italy
| | - Barbara Bottazzi
- Department of Immunology and Inflammation, Humanitas Clinical and Research Institute, Milan, Italy
| | - Lorenzo Moretta
- Immunology Area, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Erwan Mortier
- University of Nantes, CNRS, Inserm, CRCINA, University of Nantes, Nantes, France .,Immunotherapy, Graft, Oncology, LabEx IGO, Nantes, France
| | - Bruno Azzarone
- Immunology Area, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| |
Collapse
|
41
|
Hicks KC, Chariou PL, Ozawa Y, Minnar CM, Knudson KM, Meyer TJ, Bian J, Cam M, Schlom J, Gameiro SR. Tumour-targeted interleukin-12 and entinostat combination therapy improves cancer survival by reprogramming the tumour immune cell landscape. Nat Commun 2021; 12:5151. [PMID: 34446712 PMCID: PMC8390765 DOI: 10.1038/s41467-021-25393-x] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 07/30/2021] [Indexed: 01/01/2023] Open
Abstract
Poorly inflamed carcinomas do not respond well to immune checkpoint blockade. Converting the tumour microenvironment into a functionally inflamed immune hub would extend the clinical benefit of immune therapy to a larger proportion of cancer patients. Here we show, by using comprehensive single-cell transcriptome, proteome, and immune cell analysis, that Entinostat, a class I histone deacetylase inhibitor, facilitates accumulation of the necrosis-targeted recombinant murine immune-cytokine, NHS-rmIL12, in experimental mouse colon carcinomas and poorly immunogenic breast tumours. This combination therapy reprograms the tumour innate and adaptive immune milieu to an inflamed landscape, where the concerted action of highly functional CD8+ T cells and activated neutrophils drive macrophage M1-like polarization, leading to complete tumour eradication in 41.7%-100% of cases. Biomarker signature of favourable overall survival in multiple human tumor types shows close resemblance to the immune pattern generated by Entinostat/NHS-rmIL12 combination therapy. Collectively, these findings provide a rationale for combining NHS-IL12 with Entinostat in the clinical setting.
Collapse
Affiliation(s)
- Kristin C Hicks
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Paul L Chariou
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yohei Ozawa
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Christine M Minnar
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Karin M Knudson
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Thomas J Meyer
- CCR Collaborative Bioinformatics Resource (CCBR), Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jing Bian
- CCR Collaborative Bioinformatics Resource (CCBR), Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Margaret Cam
- CCR Collaborative Bioinformatics Resource (CCBR), Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Sofia R Gameiro
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
42
|
Do-Thi VA, Lee H, Jeong HJ, Lee JO, Kim YS. Protective and Therapeutic Effects of an IL-15:IL-15Rα-Secreting Cell-Based Cancer Vaccine Using a Baculovirus System. Cancers (Basel) 2021; 13:cancers13164039. [PMID: 34439192 PMCID: PMC8394727 DOI: 10.3390/cancers13164039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/22/2021] [Accepted: 08/04/2021] [Indexed: 12/14/2022] Open
Abstract
This study reports the use of the BacMam system to deliver and express self-assembling IL-15 and IL-15Rα genes to murine B16F10 melanoma and CT26 colon cancer cells. BacMam-based IL-15 and IL-15Rα were well-expressed and assembled to form the biologically functional IL-15:IL-15Rα complex. Immunization with this IL-15:IL-15Rα cancer vaccine delayed tumor growth in mice by inducing effector memory CD4+ and CD8+ cells and effector NK cells which are tumor-infiltrating. It caused strong antitumor immune responses of CD8+ effector cells in a tumor-antigen specific manner both in vitro and in vivo and significantly attenuated Treg cells which a control virus-infected cancer vaccine could induce. Post-treatment with this cancer vaccine after a live cancer cell injection also prominently delayed the growth of the tumor. Collectively, we demonstrate a vaccine platform consisting of BacMam virus-infected B16F10 or CT26 cancer cells that secrete IL-15:IL-15Rα. This study is the first demonstration of a functionally competent soluble IL-15:IL-15Rα complex-related cancer vaccine using a baculovirus system and advocates that the BacMam system can be used as a secure and rapid method of producing a protective and therapeutic cancer vaccine.
Collapse
Affiliation(s)
- Van Anh Do-Thi
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Korea; (V.A.D.-T.); (H.J.J.)
| | - Hayyoung Lee
- Institute of Biotechnology, Chungnam National University, Daejeon 34134, Korea;
| | - Hye Jin Jeong
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Korea; (V.A.D.-T.); (H.J.J.)
| | - Jie-Oh Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Korea; (V.A.D.-T.); (H.J.J.)
- Correspondence: (J.-O.L.); (Y.S.K.)
| | - Young Sang Kim
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon 34134, Korea
- Correspondence: (J.-O.L.); (Y.S.K.)
| |
Collapse
|
43
|
Robinson TO, Hegde SM, Chang AJ, Gangadharan A, Rivas S, Madakamutil L, Zalevsky J, Miyazaki T, Schluns KS. NKTR-255, a polymer-conjugated IL-15 with unique mechanisms of action on T and natural killer cells. J Clin Invest 2021; 131:e144365. [PMID: 34375310 DOI: 10.1172/jci144365] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 08/06/2021] [Indexed: 11/17/2022] Open
Abstract
NKTR-255 is a novel polyethylene glycol (PEG)-conjugate of recombinant human IL-15 (rhIL-15) being examined as a potential cancer immunotherapeutic. Since IL-15 responses can be mediated by trans- or cis-presentation via IL-15Rα or soluble IL-15/IL-15Rα complexes, we investigated the role of IL-15Rα in driving NKTR-255 responses using defined naïve and memory ovalbumin-specific CD8 T cells (OT-I) CD8 T and NK cells in mice. NKTR-255 induced a 2.5 and 2.0-fold expansion of CD8 T and NK cells, respectively in WT mice. In adoptive transfer studies, proliferation of naïve and memory Wt OT-I T cells in response to NKTR-255 was not impaired in IL-15Rα-/- mice, suggesting trans-presentation was not utilized by NKTR-255. Interestingly, naïve IL-15Rα-/- OT-I cells had deficient responses to NKTR-255 while memory IL-15Rα-/- OT-I cell responses were partially impaired, suggesting that naive CD8 T cells are more dependent on cis-presentation of NKTR-255 than memory CD8 T cells. In bone marrow chimeras studies, IL-15Rα-/- and WT NK cells present in WT recipients had similar responses to NKTR-255, suggesting that cis-presentation is not utilized by NK cells. NKTR-255 could form soluble complexes with IL-15Rα; binding to murine IL-15Rα generated superagonists that preferentially stimulated NK cells showing that conversion to IL-15Rβ agonist biases the response towards NK cells. These findings highlight the ability of NKTR-255 to utilize IL-15Rα for cis-presentation and act as an IL-15Rαβ agonist on CD8 T cells.
Collapse
Affiliation(s)
- Tanya O Robinson
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Shweta M Hegde
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Allison J Chang
- Chemical and Biomedical Engineering, Rice University, Houston, United States of America
| | - Achintyan Gangadharan
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Sarai Rivas
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Loui Madakamutil
- Research Biology: Pharmacology, Nektar Therapeutics, San Francisco, United States of America
| | - Jonathan Zalevsky
- Research and Development, Nektar Therapeutics, San Francisco, United States of America
| | - Takahiro Miyazaki
- Research Biology: Pharmacology, Nektar Therapeutics, San Francisco, United States of America
| | - Kimberly S Schluns
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, United States of America
| |
Collapse
|
44
|
Differential Response of MDA-MB-231 and MCF-7 Breast Cancer Cells to In Vitro Inhibition with CTLA-4 and PD-1 through Cancer-Immune Cells Modified Interactions. Cells 2021; 10:cells10082044. [PMID: 34440813 PMCID: PMC8392578 DOI: 10.3390/cells10082044] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 07/22/2021] [Accepted: 08/03/2021] [Indexed: 12/25/2022] Open
Abstract
Drugs targeting immune checkpoint molecules have been found effective in melanoma, lung cancer, and other malignancies treatment. Recent studies on breast cancer demonstrated the significance of inhibitory anti-CTLA-4 and anti-PD-1 in the regulation of disease progression. However, seemingly the same types of breast cancer do not always respond unambiguously to immunotherapy. Thus, here we set out to analyze the in vitro effects of inhibiting CTLA-4 and PD-1 on interactions between co-cultured lymphocytes and two selected breast adenocarcinoma cell lines. Breast cancer cells were co-cultured with lymphocytes to evaluate the effects of CTLA-4 and PD-1 inhibition. Proliferation, cell cycle, and viability assessment were measured in cancer cells. IFN-gamma, IL-10, perforin, granzyme B production, and CTLA-4 and PD-1 expression were analyzed in lymphocytes. We found that administration of anti-CTLA-4 improved the anti-cancer activity of T cells with reduced proliferation and viability of MDA-MB-231. Lack of response was observed in the context of MCF-7. In addition, differential expression of checkpoint proteins was found between studied cancer cells lines. Inhibition of molecules was followed by IL-10 and IFN-gamma decrease in lymphocytes co-cultured with MDA-MB-231, not demonstrated in reference to MCF-7. Furthermore, CTLA-4 blockage was associated with reduction of CTLA-4+ and PD-1+ lymphocytes in MDA-MB-231, with a significant increase in MCF-7, reduced by anti-PD-1. Altogether, our study revealed that anti-CTLA-4 and anti-PD-1 treatment can improve lymphocytes effects on breast cancer cells. Favorable effects seemed to be related to breast cancer cells features as differential responses were reported. Novel blocking antibodies strategies should be tested for more effective cancer inhibition.
Collapse
|
45
|
Cody JW, Ellis-Connell AL, O’Connor SL, Pienaar E. Mathematical modeling of N-803 treatment in SIV-infected non-human primates. PLoS Comput Biol 2021; 17:e1009204. [PMID: 34319980 PMCID: PMC8351941 DOI: 10.1371/journal.pcbi.1009204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 08/09/2021] [Accepted: 06/21/2021] [Indexed: 12/01/2022] Open
Abstract
Immunomodulatory drugs could contribute to a functional cure for Human Immunodeficiency Virus (HIV). Interleukin-15 (IL-15) promotes expansion and activation of CD8+ T cell and natural killer (NK) cell populations. In one study, an IL-15 superagonist, N-803, suppressed Simian Immunodeficiency Virus (SIV) in non-human primates (NHPs) who had received prior SIV vaccination. However, viral suppression attenuated with continued N-803 treatment, partially returning after long treatment interruption. While there is evidence of concurrent drug tolerance, immune regulation, and viral escape, the relative contributions of these mechanisms to the observed viral dynamics have not been quantified. Here, we utilize mathematical models of N-803 treatment in SIV-infected macaques to estimate contributions of these three key mechanisms to treatment outcomes: 1) drug tolerance, 2) immune regulation, and 3) viral escape. We calibrated our model to viral and lymphocyte responses from the above-mentioned NHP study. Our models track CD8+ T cell and NK cell populations with N-803-dependent proliferation and activation, as well as viral dynamics in response to these immune cell populations. We compared mathematical models with different combinations of the three key mechanisms based on Akaike Information Criterion and important qualitative features of the NHP data. Two minimal models were capable of reproducing the observed SIV response to N-803. In both models, immune regulation strongly reduced cytotoxic cell activation to enable viral rebound. Either long-term drug tolerance or viral escape (or some combination thereof) could account for changes to viral dynamics across long breaks in N-803 treatment. Theoretical explorations with the models showed that less-frequent N-803 dosing and concurrent immune regulation blockade (e.g. PD-L1 inhibition) may improve N-803 efficacy. However, N-803 may need to be combined with other immune therapies to countermand viral escape from the CD8+ T cell response. Our mechanistic model will inform such therapy design and guide future studies. Immune therapy may be a critical component in the functional cure for Human Immunodeficiency Virus (HIV). N-803 is an immunotherapeutic drug that activates antigen-specific CD8+ T cells of the immune system. These CD8+ T cells eliminate HIV-infected cells in order to limit the spread of infection in the body. In one study, N-803 reduced plasma viremia in macaques that were infected with Simian Immunodeficiency Virus, an analog of HIV. Here, we used mathematical models to analyze the data from this study to better understand the effects of N-803 therapy on the immune system. Our models indicated that inhibitory signals may be reversing the stimulatory effect of N-803. Results also suggested the possibilities that tolerance to N-803 could build up within the CD8+ T cells themselves and that the treatment may be selecting for virus strains that are not targeted by CD8+ T cells. Our models predict that N-803 therapy may be made more effective if the time between doses is increased or if inhibitory signals are blocked by an additional drug. Also, N-803 may need to be combined with other immune therapies to target virus that would otherwise evade CD8+ T cells.
Collapse
Affiliation(s)
- Jonathan W. Cody
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, United States of America
| | - Amy L. Ellis-Connell
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Shelby L. O’Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Elsje Pienaar
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, United States of America
- * E-mail:
| |
Collapse
|
46
|
Wolfson B, Padget MR, Schlom J, Hodge JW. Exploiting off-target effects of estrogen deprivation to sensitize estrogen receptor negative breast cancer to immune killing. J Immunother Cancer 2021; 9:jitc-2020-002258. [PMID: 34244306 PMCID: PMC8268928 DOI: 10.1136/jitc-2020-002258] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2021] [Indexed: 12/17/2022] Open
Abstract
Background There are highly effective treatment strategies for estrogen receptor (ER)+, progesterone receptor (PR)+, and HER2+ breast cancers; however, there are limited targeted therapeutic strategies for the 10%–15% of women who are diagnosed with triple-negative breast cancer. Here, we hypothesize that ER targeting drugs induce phenotypic changes to sensitize breast tumor cells to immune-mediated killing regardless of their ER status. Methods Real-time cell analysis, flow cytometry, qRT-PCR, western blotting, and multiplexed RNA profiling were performed to characterize ER+ and ER− breast cancer cells and to interrogate the phenotypic effects of ER targeting drugs. Sensitization of breast cancer cells to immune cell killing by the tamoxifen metabolite 4-hydroxytamoxifen (4-OHT) and fulvestrant was determined through in vitro health-donor natural killer cell 111IN-release killing assays. A syngeneic tumor study was performed to validate these findings in vivo. Results Pretreatment with tamoxifen metabolite 4-OHT or fulvestrant resulted in increased natural killer (NK)–mediated cell lysis of both ER+ and ER− breast cancer cells. Through multiplexed RNA profiling analysis of 4-OHT-treated ER+ and ER− cells, we identified increased activation of apoptotic and death receptor signaling pathways and identified G protein-coupled receptor for estrogen (GPR30) engagement as a putative mechanism for immunogenic modulation. Using the specific GPR30 agonist G-1, we demonstrate that targeted activation of GPR30 signaling resulted in increased NK cell killing. Furthermore, we show that knockdown of GPR30 inhibited 4-OHT and fulvestrant mediated increases to NK cell killing, demonstrating this is dependent on GPR30 expression. Moreover, we demonstrate that this mechanism remains active in a 4-OHT-resistant MCF7 cell line, showing that even in patient populations with ER+ tumors that are resistant to the cytotoxic effects of tamoxifen, 4-OHT treatment sensitizes them to immune-mediated killing. Moreover, we find that fulvestrant pretreatment of tumor cells synergizes with the IL-15 superagonist N-803 treatment of NK cells and sensitizes tumor cells to killing by programmed death-ligand 1 (PD-L1) targeting high-affinity natural killer (t-haNK) cells. Finally, we demonstrate that the combination of fulvestrant and N-803 is effective in triple-negative breast cancer in vivo. Conclusion Together, these findings demonstrate a novel effect of ER targeting drugs on the interaction of ER+ and, surprisingly, ER− tumors cells with the immune system. This study is the first to demonstrate the potential use of ER targeting drugs as immunomodulatory agents in an ER agnostic manner and may inform novel immunotherapy strategies in breast cancer.
Collapse
Affiliation(s)
- Benjamin Wolfson
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Michelle R Padget
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - James W Hodge
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| |
Collapse
|
47
|
Le DT, Huynh TR, Burt B, Van Buren G, Abeynaike SA, Zalfa C, Nikzad R, Kheradmand F, Tyner JJ, Paust S. Natural killer cells and cytotoxic T lymphocytes are required to clear solid tumor in a patient-derived xenograft. JCI Insight 2021; 6:e140116. [PMID: 34081628 PMCID: PMC8410059 DOI: 10.1172/jci.insight.140116] [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] [Indexed: 12/31/2022] Open
Abstract
Existing patient-derived xenograft (PDX) mouse models of solid tumors lack a fully tumor donor-matched, syngeneic, and functional immune system. We developed a model that overcomes these limitations by engrafting lymphopenic recipient mice with a fresh, undisrupted piece of solid tumor, whereby tumor-infiltrating lymphocytes (TILs) persisted in the recipient mice for several weeks. Successful tumor engraftment was achieved in 83% to 89% of TIL-PDX mice, and these were seen to harbor exhausted immuno-effector as well as functional immunoregulatory cells persisting for at least 6 months postengraftment. Combined treatment with interleukin-15 stimulation and immune checkpoint inhibition resulted in complete or partial tumor response in this model. Further, depletion of cytotoxic T lymphocytes and/or natural killer cells before combined immunotherapy revealed that both cell types were required for maximal tumor regression. Our TIL-PDX model provides a valuable resource for powerful mechanistic and therapeutic studies in solid tumors.
Collapse
Affiliation(s)
- Duy Tri Le
- Department of Pediatrics, Texas Children's Hospital, Houston, Texas, USA
| | - Tridu R Huynh
- Scripps Research Translational Institute, La Jolla, California, USA.,Division of Internal Medicine, Scripps Clinic/Scripps Green Hospital, La Jolla, California, USA.,Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA
| | - Bryan Burt
- Division of General Thoracic Surgery and
| | - George Van Buren
- Division of Surgical Oncology, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas, USA.,Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - Shawn A Abeynaike
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA
| | - Cristina Zalfa
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA
| | - Rana Nikzad
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA
| | - Farrah Kheradmand
- Margaret M. and Albert B. Alkek Department of Medicine, Baylor College of Medicine and Michael E. DeBakey VA Medical Center, US Department of Veterans Affairs, Houston, Texas, USA
| | - John J Tyner
- Division of Cardiovascular/Thoracic Surgery, Scripps Clinic, La Jolla, California, USA
| | - Silke Paust
- Department of Pediatrics, Texas Children's Hospital, Houston, Texas, USA.,Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA
| |
Collapse
|
48
|
Miazek-Zapala N, Slusarczyk A, Kusowska A, Zapala P, Kubacz M, Winiarska M, Bobrowicz M. The "Magic Bullet" Is Here? Cell-Based Immunotherapies for Hematological Malignancies in the Twilight of the Chemotherapy Era. Cells 2021; 10:1511. [PMID: 34203935 PMCID: PMC8232692 DOI: 10.3390/cells10061511] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 06/09/2021] [Accepted: 06/11/2021] [Indexed: 12/14/2022] Open
Abstract
Despite the introduction of a plethora of different anti-neoplastic approaches including standard chemotherapy, molecularly targeted small-molecule inhibitors, monoclonal antibodies, and finally hematopoietic stem cell transplantation (HSCT), there is still a need for novel therapeutic options with the potential to cure hematological malignancies. Although nowadays HSCT already offers a curative effect, its implementation is largely limited by the age and frailty of the patient. Moreover, its efficacy in combating the malignancy with graft-versus-tumor effect frequently coexists with undesirable graft-versus-host disease (GvHD). Therefore, it seems that cell-based adoptive immunotherapies may constitute optimal strategies to be successfully incorporated into the standard therapeutic protocols. Thus, modern cell-based immunotherapy may finally represent the long-awaited "magic bullet" against cancer. However, enhancing the safety and efficacy of this treatment regimen still presents many challenges. In this review, we summarize the up-to-date state of the art concerning the use of CAR-T cells and NK-cell-based immunotherapies in hemato-oncology, identify possible obstacles, and delineate further perspectives.
Collapse
Affiliation(s)
- Nina Miazek-Zapala
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland; (N.M.-Z.); (A.S.); (A.K.); (M.K.); (M.W.)
- Institute of Physiology and Pathophysiology of Hearing, World Hearing Center, 05-830 Nadarzyn, Poland
| | - Aleksander Slusarczyk
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland; (N.M.-Z.); (A.S.); (A.K.); (M.K.); (M.W.)
- Department of General, Oncological and Functional Urology, Medical University of Warsaw, 02-005 Warsaw, Poland;
| | - Aleksandra Kusowska
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland; (N.M.-Z.); (A.S.); (A.K.); (M.K.); (M.W.)
| | - Piotr Zapala
- Department of General, Oncological and Functional Urology, Medical University of Warsaw, 02-005 Warsaw, Poland;
| | - Matylda Kubacz
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland; (N.M.-Z.); (A.S.); (A.K.); (M.K.); (M.W.)
| | - Magdalena Winiarska
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland; (N.M.-Z.); (A.S.); (A.K.); (M.K.); (M.W.)
| | - Malgorzata Bobrowicz
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland; (N.M.-Z.); (A.S.); (A.K.); (M.K.); (M.W.)
| |
Collapse
|
49
|
Abstract
PURPOSE OF REVIEW Patients with Lynch syndrome have a high probability of developing colorectal and other carcinomas. This review provides a comprehensive assessment of the immunologic aspects of Lynch syndrome pathogenesis and provides an overview of potential immune interventions for patients with Lynch syndrome polyps and Lynch syndrome-associated carcinomas. RECENT FINDINGS Immunogenic properties of the majority of Lynch syndrome polyps and associated cancers include microsatellite instability leading to a high mutational burden and the development of novel frameshift peptides, i.e., neoantigens. In addition, patients with Lynch syndrome develop T cell responses in the periphery and in the tumor microenvironment (TME) to tumor-associated antigens, and a proinflammatory cytokine TME has also been identified. However, Lynch syndrome lesions also possess immunosuppressive entities such as alterations in MHC class I antigen presentation, TGFβ receptor mutations, regulatory T cells, and upregulation of PD-L1 on tumor-associated lymphocytes. The rich immune microenvironment of Lynch syndrome polyps and associated carcinomas provides an opportunity to employ the spectrum of immune-mediating agents now available to induce and enhance host immune responses and/or to also reduce immunosuppressive entities. These agents can be employed in the so-called prevention trials for the treatment of patients with Lynch syndrome polyps and for trials in patients with Lynch syndrome-associated cancers.
Collapse
Affiliation(s)
- Danielle M Pastor
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- NIH Hematology Oncology Fellowship Program, National Institutes of Health, Bethesda, MD, USA
| | - Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
50
|
Fabian KP, Padget MR, Donahue RN, Solocinski K, Robbins Y, Allen CT, Lee JH, Rabizadeh S, Soon-Shiong P, Schlom J, Hodge JW. PD-L1 targeting high-affinity NK (t-haNK) cells induce direct antitumor effects and target suppressive MDSC populations. J Immunother Cancer 2021; 8:jitc-2019-000450. [PMID: 32439799 PMCID: PMC7247398 DOI: 10.1136/jitc-2019-000450] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2020] [Indexed: 12/12/2022] Open
Abstract
Background Although immune checkpoint inhibitors have revolutionized cancer treatment, clinical benefit with this class of agents has been limited to a subset of patients. Hence, more effective means to target tumor cells that express immune checkpoint molecules should be developed. For the first time, we report a novel natural killer (NK) cell line, programmed death-ligand 1 (PD-L1) targeting high-affinity natural killer (t-haNK), which was derived from NK-92 and was engineered to express high-affinity CD16, endoplasmic reticulum-retained interleukin (IL)-2, and a PD-L1-specific chimeric antigen receptor (CAR). We show that PD-L1 t-haNK cells also retained the expression of native NK receptors and carried a high content of granzyme and perforin granules. Methods NanoString, flow cytometry, and immunofluorescence analyses were performed to characterize the phenotype of irradiated PD-L1 t-haNK cells. In vitro PD-L1 t-haNK cell activity against cancer cell lines and human peripheral blood mononuclear cells (PBMCs) was determined via flow-based and 111In-release killing assays. The antitumor effect of PD-L1 t-haNK cells in vivo was investigated using MDA-MB-231, H460, and HTB1 xenograft models in NOD-scid IL2Rgammanull (NSG) mice. Additionally, the antitumor effect of PD-L1 t-haNK cells, in combination with anti-PD-1 and N-803, an IL-15 superagonist, was evaluated using mouse oral cancer 1 syngeneic model in C57BL/6 mice. Results We show that PD-L1 t-haNK cells expressed PD-L1-targeting CAR and CD16, retained the expression of native NK receptors, and carried a high content of granzyme and perforin granules. In vitro, we demonstrate the ability of irradiated PD-L1 t-haNK cells to lyse 20 of the 20 human cancer cell lines tested, including triple negative breast cancer (TNBC) and lung, urogenital, and gastric cancer cells. The cytotoxicity of PD-L1 t-haNK cells was correlated to the PD-L1 expression of the tumor targets and can be improved by pretreating the targets with interferon (IFN)-γ. In vivo, irradiated PD-L1 t-haNK cells inhibited the growth of engrafted TNBC and lung and bladder tumors in NSG mice. The combination of PD-L1 t-haNK cells with N-803 and anti-PD-1 antibody resulted in superior tumor growth control of engrafted oral cavity squamous carcinoma tumors in C57BL/6 mice. In addition, when cocultured with human PBMCs, PD-L1 t-haNK cells preferentially lysed the myeloid-derived suppressor cell population but not other immune cell types. Conclusion These studies demonstrate the antitumor efficacy of PD-L1 t-haNK cells and provide a rationale for the potential use of these cells in clinical studies.
Collapse
Affiliation(s)
- Kellsye P Fabian
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Michelle R Padget
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Renee N Donahue
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Kristen Solocinski
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Yvette Robbins
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Clint T Allen
- Section on Translational Tumor Immunology, National Institute on Deafness and Other Communication Disorders, Bethesda, Maryland, USA
| | - John H Lee
- ImmunityBio, Santa Cruz, California, USA
| | - Shahrooz Rabizadeh
- NantOmics, Culver City, California, USA.,ImmunityBio, Culver City, California, USA
| | - Patrick Soon-Shiong
- NantOmics, Culver City, California, USA.,ImmunityBio, Culver City, California, USA
| | - Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - James W Hodge
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|