1
|
Maas RJA, Hoogstad-van Evert JS, Hagemans IM, Brummelman J, van Ens D, de Jonge PKJD, Hooijmaijers L, Mahajan S, van der Waart AB, Hermans CKJC, de Klein J, Woestenenk R, van Herwaarden AE, Schaap NPM, Rezaeifard S, Tauriello DVF, Zusterzeel PLM, Ottevanger N, Jansen JH, Hobo W, Dolstra H. Increased peritoneal TGF-β1 is associated with ascites-induced NK-cell dysfunction and reduced survival in high-grade epithelial ovarian cancer. Front Immunol 2024; 15:1448041. [PMID: 39376560 PMCID: PMC11456434 DOI: 10.3389/fimmu.2024.1448041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 09/03/2024] [Indexed: 10/09/2024] Open
Abstract
Natural killer (NK) cell therapy represents an attractive immunotherapy approach against recurrent epithelial ovarian cancer (EOC), as EOC is sensitive to NK cell-mediated cytotoxicity. However, NK cell antitumor activity is dampened by suppressive factors in EOC patient ascites. Here, we integrated functional assays, soluble factor analysis, high-dimensional flow cytometry cellular component data and clinical parameters of advanced EOC patients to study the mechanisms of ascites-induced inhibition of NK cells. Using a suppression assay, we found that ascites from EOC patients strongly inhibits peripheral blood-derived NK cells and CD34+ progenitor-derived NK cells, albeit the latter were more resistant. Interestingly, we found that higher ascites-induced NK cell inhibition correlated with reduced progression-free and overall survival in EOC patients. Furthermore, we identified transforming growth factor (TGF)-β1 to correlate with ascites-induced NK cell dysfunction and reduced patient survival. In functional assays, we showed that proliferation and anti-tumor reactivity of CD34+ progenitor-derived NK cells are significantly affected by TGF-β1 exposure. Moreover, inhibition of TGF-β1 signaling with galunisertib partly restored NK cell functionality in some donors. For the cellular components, we showed that the secretome is associated with a different composition of CD45+ cells between ascites of EOC and benign reference samples with higher proportions of macrophages in the EOC patient samples. Furthermore, we revealed that higher TGF-β1 levels are associated with the presence of M2-like macrophages, B cell populations and T-regulatory cells in EOC patient ascites. These findings reveal that targeting TGF-β1 signaling could increase NK cell immune responses in high-grade EOC patients.
Collapse
Affiliation(s)
- Ralph J. A. Maas
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Janneke S. Hoogstad-van Evert
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
- Department of Obstetrics and Gynecology Amphia, Breda, Netherlands
| | - Iris M. Hagemans
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Jolanda Brummelman
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Diede van Ens
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Paul K. J. D. de Jonge
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Laura Hooijmaijers
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Shweta Mahajan
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Anniek B. van der Waart
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Charlotte K. J. C. Hermans
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Janne de Klein
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Rob Woestenenk
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Antonius E. van Herwaarden
- Diagnostic Laboratory – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - Somayeh Rezaeifard
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
- Department of Cell Biology, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - Petra L. M. Zusterzeel
- Department of Obstetrics and Gynecology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Nelleke Ottevanger
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Joop H. Jansen
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Willemijn Hobo
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Harry Dolstra
- Laboratory of Hematology – Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
2
|
Kaur K, Sanghu J, Memarzadeh S, Jewett A. Exploring the Potential of Natural Killer Cell-Based Immunotherapy in Targeting High-Grade Serous Ovarian Carcinomas. Vaccines (Basel) 2024; 12:677. [PMID: 38932405 PMCID: PMC11209217 DOI: 10.3390/vaccines12060677] [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: 04/19/2024] [Revised: 06/05/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
High-grade serous ovarian cancers (HGSOCs) likely consist of poorly differentiated stem-like cells (PDSLCs) and differentiated tumor cells. Conventional therapeutics are incapable of completely eradicating PDSLCs, contributing to disease progression and tumor relapse. Primary NK cells are known to effectively lyse PDSLCs, but they exhibit low or minimal cytotoxic potential against well-differentiated tumors. We have introduced and discussed the characteristics of super-charged NK (sNK) cells in this review. sNK cells, in comparison to primary NK cells, exhibit a significantly higher capability for the direct killing of both PDSLCs and well-differentiated tumors. In addition, sNK cells secrete significantly higher levels of cytokines, especially those known to induce the differentiation of tumors. In addition, we propose that a combination of sNK and chemotherapy could be one of the most effective strategies to eliminate the heterogeneous population of ovarian tumors; sNK cells can lyse both PDSLCs and well-differentiated tumors, induce the differentiation of PDSLCs, and could be used in combination with chemotherapy to target both well-differentiated and NK-induced differentiated tumors.
Collapse
Affiliation(s)
- Kawaljit Kaur
- Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, University of California School of Dentistry, 10833 Le Conte Ave, Los Angeles, CA 90095, USA;
| | - Jashan Sanghu
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA; (J.S.); (S.M.)
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Sanaz Memarzadeh
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA; (J.S.); (S.M.)
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA 90095, USA
- The Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA 90095, USA
- The VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| | - Anahid Jewett
- Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, University of California School of Dentistry, 10833 Le Conte Ave, Los Angeles, CA 90095, USA;
- The Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
3
|
Hou Y, Zhao X, Nie X. Enhancing the therapeutic efficacy of NK cells in the treatment of ovarian cancer (Review). Oncol Rep 2024; 51:50. [PMID: 38299257 PMCID: PMC10851334 DOI: 10.3892/or.2024.8709] [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: 10/31/2023] [Accepted: 01/03/2024] [Indexed: 02/02/2024] Open
Abstract
Ovarian cancer is a prevalent gynecological malignancy associated with a high mortality rate and a low 5‑year survival rate. Typically, >70% of patients present with an advanced stage of the disease, resulting in a high number of ovarian cancer‑associated deaths worldwide. Over the past decade, adoptive cellular immunotherapy has been investigated in clinical trials, and the results have led to the increased use in cancer treatment. Natural killer (NK) cells are cytotoxic lymphoid cells that recognize and lyse transformed cells, thereby impeding tumor growth. Thus, NK cells exhibit potential as a form of immunotherapy in the treatment of cancer. However, some patients with ovarian cancer treated with NK cells have experienced unsatisfactory outcomes. Therefore, further optimization of NK cells is required to increase the number of patients achieving long‑term remission. In the present review article, studies focusing on improving NK cell function were systematically summarized, and innovative strategies that augment the anticancer properties of NK cells were proposed.
Collapse
Affiliation(s)
- Yuzhu Hou
- Department of Gynecology, Qingdao Eighth People's Hospital, Qingdao, Shandong 266000, P.R. China
| | - Xiujun Zhao
- Department of Gynecology, Qingdao Eighth People's Hospital, Qingdao, Shandong 266000, P.R. China
| | - Xiaoqian Nie
- Department of Gynecology, Qingdao Eighth People's Hospital, Qingdao, Shandong 266000, P.R. China
| |
Collapse
|
4
|
van Hauten PMM, Hooijmaijers L, Vidal-Manrique M, van der Waart AB, Hobo W, Wu J, Blijlevens NMA, Jansen JH, Walcheck B, Schaap NPM, de Jonge PKJD, Dolstra H. Engineering of CD34+ progenitor-derived natural killer cells with higher-affinity CD16a for enhanced antibody-dependent cellular cytotoxicity. Cytotherapy 2024; 26:252-260. [PMID: 38127030 DOI: 10.1016/j.jcyt.2023.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 11/20/2023] [Accepted: 11/21/2023] [Indexed: 12/23/2023]
Abstract
BACKGROUND AIMS Natural killer (NK) cell transfer is a promising cellular immunotherapy for cancer. Previously, we developed a robust method to generate large NK cell numbers from CD34+ hematopoietic stem and progenitor cells (HSPCs), which exhibit strong anti-tumor activity. However, since these cells express low levels of the Fc receptor CD16a in vitro, antibody-dependent cellular cytotoxicity (ADCC) by these cells is limited. To broaden clinical applicability of our HSPC-NK cells toward less NK-sensitive malignancies, we aimed to improve ADCC through CD16a transduction. METHODS Using wildtype and S197P mutant greater-affinity (both with V158) CD16a retroviral transgenes (i.e., a cleavable and noncleavable CD16a upon stimulation), we generated CD16a HSPC-transduced NK cells, with CD34+ cells isolated from umbilical cord blood (UCB) or peripheral blood after G-CSF stem cell mobilization (MPB). CD16a expressing NK cells were enriched using flow cytometry-based cell sorting. Subsequently, phenotypic analyses and functional assays were performed to investigate natural cytotoxicity and ADCC activity. RESULTS Mean transduction efficiency was 34% for UCB-derived HSPCs and 20% for MPB-derived HSPCs, which was enriched by flow cytometry-based cell sorting to >90% for both conditions. Expression of the transgene remained stable during the entire NK expansion cell generation process. Proliferation and differentiation of HSPCs were not hampered by the transduction process, resulting in effectively differentiated CD56+ NK cells after 5 weeks. Activation of the HSPC-derived NK cells resulted in significant shedding of wildtype CD16a transcribed from the endogenous gene, but not of the noncleavable mutant CD16a protein expressed from the transduced construct. The mean increase of CD107+IFNγ+ expressing NK cells after inducing ADCC was tenfold in enriched noncleavable CD16a HSPC-NK cells. Killing capacity of CD16a-transduced NK cells was significantly improved after addition of a tumor-targeting antibody in tumor cell lines and primary B-cell leukemia and lymphoma cells compared to unmodified HSPC-NK cells. CONCLUSIONS Together, these data demonstrate that the applicability of adoptive NK cell immunotherapy may be broadened to less NK-sensitive malignancies by upregulation of CD16a expression in combination with the use of tumor-targeting monoclonal antibodies.
Collapse
Affiliation(s)
- Paulien M M van Hauten
- Laboratory of Hematology-Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Hematology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Laura Hooijmaijers
- Laboratory of Hematology-Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Marcos Vidal-Manrique
- Laboratory of Hematology-Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Anniek B van der Waart
- Laboratory of Hematology-Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Willemijn Hobo
- Laboratory of Hematology-Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jianming Wu
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Saint Paul, Minnesota, USA
| | - Nicole M A Blijlevens
- Department of Hematology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Joop H Jansen
- Laboratory of Hematology-Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Bruce Walcheck
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Saint Paul, Minnesota, USA
| | - Nicolaas P M Schaap
- Department of Hematology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Paul K J D de Jonge
- Laboratory of Hematology-Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Harry Dolstra
- Laboratory of Hematology-Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, the Netherlands.
| |
Collapse
|
5
|
Van der Meer JMR, Bulder I, Kuijk C, Kleijer M, Verheij MW, Omar SZ, Haverkate NJE, Dolstra H, Blom B, Hazenberg MD, Voermans C. Generation of human ILC3 from allogeneic and autologous CD34 + hematopoietic progenitors toward adoptive transfer. Cytotherapy 2024; 26:136-144. [PMID: 38149947 DOI: 10.1016/j.jcyt.2023.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 11/10/2023] [Accepted: 11/21/2023] [Indexed: 12/28/2023]
Abstract
Type 3 innate lymphoid cells (ILC3) are important in tissue homeostasis. In the gut, ILC3 repair damaged epithelium and suppress inflammation. In allogeneic hematopoietic cell transplantation (HCT), ILC3 protect against graft-versus-host disease (GvHD), most likely by restoring tissue damage and preventing inflammation. We hypothesize that supplementing HCT grafts with interleukin-22 (IL-22)-producing ILC3 may prevent acute GvHD. We therefore explored ex vivo generation of human IL-22-producing ILC3 from hematopoietic stem and progenitor cells (HSPC) obtained from adult, neonatal and fetal sources. We established a stroma-free system culturing human cord blood-derived CD34+ HSPC with successive cytokine mixes for 5 weeks. We analyzed the presence of phenotypically defined ILC, their viability, proliferation and IL-22 production (after stimulation) by flow cytometry and enzyme-linked immunosorbent assay (ELISA). We found that the addition of recombinant human IL-15 and the enhancer of zeste homolog 1/2 inhibitor UNC1999 promoted ILC3 generation. Similar results were demonstrated when UNC1999 was added to CD34+ HSPC derived from healthy adult granulocyte colony-stimulating factor mobilized peripheral blood and bone marrow, but not fetal liver. UNC1999 did not negatively impact IL-22 production in any of the HSPC sources. Finally, we observed that autologous HSPC mobilized from the blood of adults with hematological malignancies also developed into ILC3, albeit with a significantly lower capacity. Together, we developed a stroma-free protocol to generate large quantities of IL-22-producing ILC3 from healthy adult human HSPC that can be applied for adoptive transfer to prevent GvHD after allogeneic HCT.
Collapse
Affiliation(s)
- Jolien M R Van der Meer
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands
| | - Ingrid Bulder
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands; Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands
| | - Carlijn Kuijk
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands
| | - Marion Kleijer
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands
| | - Myrddin W Verheij
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands; Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands
| | - Said Z Omar
- Department of Experimental Immunology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, The Netherlands
| | - Nienke J E Haverkate
- Department of Experimental Immunology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, The Netherlands
| | - Harry Dolstra
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Bianca Blom
- Department of Experimental Immunology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, The Netherlands
| | - Mette D Hazenberg
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands; Department of Experimental Immunology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, The Netherlands; Cancer Center Amsterdam, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands; Department of Hematology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Carlijn Voermans
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands; Department of Hematology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
6
|
de Jonge PKJD, van Hauten PMM, Janssen LD, de Goede AL, Berrien-Elliott MM, van der Meer JMR, Mousset CM, Roeven MWH, Foster M, Blijlevens N, Hobo W, Fehniger TA, Jansen JH, Schaap NPM, Dolstra H. Good manufacturing practice production of CD34 + progenitor-derived NK cells for adoptive immunotherapy in acute myeloid leukemia. Cancer Immunol Immunother 2023; 72:3323-3335. [PMID: 37477653 PMCID: PMC10491545 DOI: 10.1007/s00262-023-03492-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 06/30/2023] [Indexed: 07/22/2023]
Abstract
Allogeneic natural killer (NK) cell-based immunotherapy is a promising, well-tolerated adjuvant therapeutic approach for acute myeloid leukemia (AML). For reproducible NK cell immunotherapy, a homogenous, pure and scalable NK cell product is preferred. Therefore, we developed a good manufacturing practice (GMP)-compliant, cytokine-based ex vivo manufacturing process for generating NK cells from CD34+ hematopoietic stem and progenitor cells (HSPC). This manufacturing process combines amongst others IL15 and IL12 and the aryl hydrocarbon receptor antagonist StemRegenin-1 (SR1) to generate a consistent and active NK cell product that fits the requirements for NK cell immunotherapy well. The cell culture protocol was first optimized to generate NK cells with required expansion and differentiation capacity in GMP-compliant closed system cell culture bags. In addition, phenotype, antitumor potency, proliferative and metabolic capacity were evaluated to characterize the HSPC-NK product. Subsequently, seven batches were manufactured for qualification of the process. All seven runs demonstrated consistent results for proliferation, differentiation and antitumor potency, and preliminary specifications for the investigational medicinal product for early clinical phase trials were set. This GMP-compliant manufacturing process for HSPC-NK cells (named RNK001 cells) is used to produce NK cell batches applied in the clinical trial 'Infusion of ex vivo-generated allogeneic natural killer cells in combination with subcutaneous IL2 in patients with acute myeloid leukemia' approved by the Dutch Ethics Committee (EudraCT 2019-001929-27).
Collapse
Affiliation(s)
- P K J D de Jonge
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Geert Grooteplein 8, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - P M M van Hauten
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Geert Grooteplein 8, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
- Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - L D Janssen
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Geert Grooteplein 8, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - A L de Goede
- Department of Pharmacy, Radboud University Medical Center, Nijmegen, The Netherlands
| | - M M Berrien-Elliott
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, MO, USA
| | - J M R van der Meer
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Geert Grooteplein 8, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - C M Mousset
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Geert Grooteplein 8, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - M W H Roeven
- Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - M Foster
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, MO, USA
| | - N Blijlevens
- Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - W Hobo
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Geert Grooteplein 8, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - T A Fehniger
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, MO, USA
| | - J H Jansen
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Geert Grooteplein 8, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - N P M Schaap
- Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - H Dolstra
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Geert Grooteplein 8, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.
| |
Collapse
|
7
|
Carannante V, Wiklund M, Önfelt B. In vitro models to study natural killer cell dynamics in the tumor microenvironment. Front Immunol 2023; 14:1135148. [PMID: 37457703 PMCID: PMC10338882 DOI: 10.3389/fimmu.2023.1135148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 06/05/2023] [Indexed: 07/18/2023] Open
Abstract
Immunotherapy is revolutionizing cancer therapy. The rapid development of new immunotherapeutic strategies to treat solid tumors is posing new challenges for preclinical research, demanding novel in vitro methods to test treatments. Such methods should meet specific requirements, such as enabling the evaluation of immune cell responses like cytotoxicity or cytokine release, and infiltration into the tumor microenvironment using cancer models representative of the original disease. They should allow high-throughput and high-content analysis, to evaluate the efficacy of treatments and understand immune-evasion processes to facilitate development of new therapeutic targets. Ideally, they should be suitable for personalized immunotherapy testing, providing information for patient stratification. Consequently, the application of in vitro 3-dimensional (3D) cell culture models, such as tumor spheroids and organoids, is rapidly expanding in the immunotherapeutic field, coupled with the development of novel imaging-based techniques and -omic analysis. In this paper, we review the recent advances in the development of in vitro 3D platforms applied to natural killer (NK) cell-based cancer immunotherapy studies, highlighting the benefits and limitations of the current methods, and discuss new concepts and future directions of the field.
Collapse
Affiliation(s)
- Valentina Carannante
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Martin Wiklund
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Björn Önfelt
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
- Center for Infectious Medicine, Department of Medicine Huddinge, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
8
|
Parodi M, Astigiano S, Carrega P, Pietra G, Vitale C, Damele L, Grottoli M, Guevara Lopez MDLL, Ferracini R, Bertolini G, Roato I, Vitale M, Orecchia P. Murine models to study human NK cells in human solid tumors. Front Immunol 2023; 14:1209237. [PMID: 37388731 PMCID: PMC10301748 DOI: 10.3389/fimmu.2023.1209237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 06/02/2023] [Indexed: 07/01/2023] Open
Abstract
Since the first studies, the mouse models have provided crucial support for the most important discoveries on NK cells, on their development, function, and circulation within normal and tumor tissues. Murine tumor models were initially set to study murine NK cells, then, ever more sophisticated human-in-mice models have been developed to investigate the behavior of human NK cells and minimize the interferences from the murine environment. This review presents an overview of the models that have been used along time to study NK cells, focusing on the most popular NOG and NSG models, which work as recipients for the preparation of human-in-mice tumor models, the study of transferred human NK cells, and the evaluation of various enhancers of human NK cell function, including cytokines and chimeric molecules. Finally, an overview of the next generation humanized mice is also provided along with a discussion on how traditional and innovative in-vivo and in-vitro approaches could be integrated to optimize effective pre-clinical studies.
Collapse
Affiliation(s)
- Monica Parodi
- Unità Operativa UO Patologia e Immunologia Sperimentale, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Simonetta Astigiano
- Animal Facility, IRCCS Ospedale Policlinico San Martino Genova, Genova, Italy
| | - Paolo Carrega
- Laboratory of Immunology and Biotherapy, Department of Human Pathology, University of Messina, Messina, Italy
| | - Gabriella Pietra
- Unità Operativa UO Patologia e Immunologia Sperimentale, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Dipartimento di Medicina Sperimentale, Università di Genova, Genova, Italy
| | - Chiara Vitale
- Unità Operativa UO Patologia e Immunologia Sperimentale, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Dipartimento di Medicina Sperimentale, Università di Genova, Genova, Italy
| | - Laura Damele
- Unità Operativa UO Patologia e Immunologia Sperimentale, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Melania Grottoli
- Dipartimento di Medicina Sperimentale, Università di Genova, Genova, Italy
| | | | - Riccardo Ferracini
- Department of Surgical Sciences, Bone and Dental Bioengineering Laboratory, C.I.R Dental School, University of Turin, Turin, Italy
- Department of Surgical Sciences (DISC), University of Genoa, Genoa, Italy
| | - Giulia Bertolini
- “Epigenomics and Biomarkers of Solid Tumors”, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Ilaria Roato
- Department of Surgical Sciences, Bone and Dental Bioengineering Laboratory, C.I.R Dental School, University of Turin, Turin, Italy
| | - Massimo Vitale
- Unità Operativa UO Patologia e Immunologia Sperimentale, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Paola Orecchia
- Unità Operativa UO Patologia e Immunologia Sperimentale, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| |
Collapse
|
9
|
D’Silva SZ, Singh M, Pinto AS. NK cell defects: implication in acute myeloid leukemia. Front Immunol 2023; 14:1112059. [PMID: 37228595 PMCID: PMC10203541 DOI: 10.3389/fimmu.2023.1112059] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 04/25/2023] [Indexed: 05/27/2023] Open
Abstract
Acute Myeloid Leukemia (AML) is a complex disease with rapid progression and poor/unsatisfactory outcomes. In the past few years, the focus has been on developing newer therapies for AML; however, relapse remains a significant problem. Natural Killer cells have strong anti-tumor potential against AML. This NK-mediated cytotoxicity is often restricted by cellular defects caused by disease-associated mechanisms, which can lead to disease progression. A stark feature of AML is the low/no expression of the cognate HLA ligands for the activating KIR receptors, due to which these tumor cells evade NK-mediated lysis. Recently, different Natural Killer cell therapies have been implicated in treating AML, such as the adoptive NK cell transfer, Chimeric antigen receptor-modified NK (CAR-NK) cell therapy, antibodies, cytokine, and drug treatment. However, the data available is scarce, and the outcomes vary between different transplant settings and different types of leukemia. Moreover, remission achieved by some of these therapies is only for a short time. In this mini-review, we will discuss the role of NK cell defects in AML progression, particularly the expression of different cell surface markers, the available NK cell therapies, and the results from various preclinical and clinical trials.
Collapse
Affiliation(s)
- Selma Z. D’Silva
- Transplant Immunology and Immunogenetics Lab, Advanced Centre for Treatment, Education and Research in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| | - Meenakshi Singh
- Transplant Immunology and Immunogenetics Lab, Advanced Centre for Treatment, Education and Research in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Andrea S. Pinto
- Transplant Immunology and Immunogenetics Lab, Advanced Centre for Treatment, Education and Research in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| |
Collapse
|
10
|
Kutle I, Polten R, Hachenberg J, Klapdor R, Morgan M, Schambach A. Tumor Organoid and Spheroid Models for Cervical Cancer. Cancers (Basel) 2023; 15:cancers15092518. [PMID: 37173984 PMCID: PMC10177622 DOI: 10.3390/cancers15092518] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
Cervical cancer is one of the most common malignant diseases in women worldwide. Despite the global introduction of a preventive vaccine against the leading cause of cervical cancer, human papillomavirus (HPV) infection, the incidence of this malignant disease is still very high, especially in economically challenged areas. New advances in cancer therapy, especially the rapid development and application of different immunotherapy strategies, have shown promising pre-clinical and clinical results. However, mortality from advanced stages of cervical cancer remains a significant concern. Precise and thorough evaluation of potential novel anti-cancer therapies in pre-clinical phases is indispensable for efficient development of new, more successful treatment options for cancer patients. Recently, 3D tumor models have become the gold standard in pre-clinical cancer research due to their capacity to better mimic the architecture and microenvironment of tumor tissue as compared to standard two-dimensional (2D) cell cultures. This review will focus on the application of spheroids and patient-derived organoids (PDOs) as tumor models to develop novel therapies against cervical cancer, with an emphasis on the immunotherapies that specifically target cancer cells and modulate the tumor microenvironment (TME).
Collapse
Affiliation(s)
- Ivana Kutle
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Robert Polten
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Jens Hachenberg
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
- Department of Obstetrics and Gynecology, Hannover Medical School, 30625 Hannover, Germany
| | - Rüdiger Klapdor
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
- Department of Obstetrics and Gynecology, Hannover Medical School, 30625 Hannover, Germany
| | - Michael Morgan
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
11
|
Liu Y, Wu W, Cai C, Zhang H, Shen H, Han Y. Patient-derived xenograft models in cancer therapy: technologies and applications. Signal Transduct Target Ther 2023; 8:160. [PMID: 37045827 PMCID: PMC10097874 DOI: 10.1038/s41392-023-01419-2] [Citation(s) in RCA: 87] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 03/21/2023] [Indexed: 04/14/2023] Open
Abstract
Patient-derived xenograft (PDX) models, in which tumor tissues from patients are implanted into immunocompromised or humanized mice, have shown superiority in recapitulating the characteristics of cancer, such as the spatial structure of cancer and the intratumor heterogeneity of cancer. Moreover, PDX models retain the genomic features of patients across different stages, subtypes, and diversified treatment backgrounds. Optimized PDX engraftment procedures and modern technologies such as multi-omics and deep learning have enabled a more comprehensive depiction of the PDX molecular landscape and boosted the utilization of PDX models. These irreplaceable advantages make PDX models an ideal choice in cancer treatment studies, such as preclinical trials of novel drugs, validating novel drug combinations, screening drug-sensitive patients, and exploring drug resistance mechanisms. In this review, we gave an overview of the history of PDX models and the process of PDX model establishment. Subsequently, the review presents the strengths and weaknesses of PDX models and highlights the integration of novel technologies in PDX model research. Finally, we delineated the broad application of PDX models in chemotherapy, targeted therapy, immunotherapy, and other novel therapies.
Collapse
Affiliation(s)
- Yihan Liu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Wantao Wu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Changjing Cai
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Hao Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Hong Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China.
| | - Ying Han
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China.
| |
Collapse
|
12
|
Li Y, Xie S, Chen M, Li H, Wang Y, Fan Y, An K, Wu Y, Xiao W. Development of an antibody-ligand fusion protein scFvCD16A -sc4-1BBL in Komagataella phaffii with stimulatory activity for Natural Killer cells. Microb Cell Fact 2023; 22:67. [PMID: 37041591 PMCID: PMC10091686 DOI: 10.1186/s12934-023-02082-6] [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: 12/12/2022] [Accepted: 04/04/2023] [Indexed: 04/13/2023] Open
Abstract
BACKGROUND Natural killer (NK) cell-based immunotherapies have demonstrated substantial potential for the treatment of hematologic malignancies. However, its application is limited due to the difficulty in the production of a large number of NK cells in vitro and the insufficient therapeutic efficacy against solid tumors in vivo. Engineered antibodies or fusion proteins targeting activating receptors and costimulatory molecules of NK cells have been developed to encounter these problems. They are mostly produced in mammalian cells with high cost and long processing times. Yeast systems, such as Komagataella phaffii, present a convenient manipulation of microbial systems with the key advantages of improved folding machinery and low cost. RESULTS In this study, we designed an antibody fusion protein scFvCD16A-sc4-1BBL, composed of the single chain variant fragment (scFv) of anti-CD16A antibody and the three extracellular domains (ECDs) of human 4-1BBL in a single-chain format (sc) with the GS linker, aiming to boost NK cell proliferation and activation. This protein complex was produced in the K. phaffii X33 system and purified by affinity chromatography and size exclusion chromatography. The scFvCD16A-sc4-1BBL complex showed comparable binding abilities to its two targets human CD16A and 4-1BB as its two parental moieties (scFvCD16A and monomer ECD (mn)4-1BBL). scFvCD16A-sc4-1BBL specifically stimulated the expansion of peripheral blood mononuclear cell (PBMC)-derived NK cells in vitro. Furthermore, in the ovarian cancer xenograft mouse model, adoptive NK cell infusion combined with intraperitoneal (i.p) injection of scFvCD16A-sc4-1BBL further reduced the tumor burden and prolonged the survival time of mice. CONCLUSION Our studies demonstrate the feasibility of the expression of the antibody fusion protein scFvCD16A-sc4-1BBL in K. phaffii with favourable properties. scFvCD16A-sc4-1BBL stimulates PBMC-derived NK cell expansion in vitro and improves the antitumor activity of adoptively transferred NK cells in a murine model of ovarian cancer and may serve as a synergistic drug for NK immunotherapy in future research and applications.
Collapse
Affiliation(s)
- Yangyang Li
- Department of Oncology of the First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Institute of Immunology, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Engineering Technology Research Center of Biotechnology Drugs Anhui, University of Science and Technology of China, Hefei, 230027, Anhui, China
| | - Siqi Xie
- Department of Oncology of the First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Institute of Immunology, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Engineering Technology Research Center of Biotechnology Drugs Anhui, University of Science and Technology of China, Hefei, 230027, Anhui, China
| | - Minhua Chen
- Department of Oncology of the First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Institute of Immunology, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Engineering Technology Research Center of Biotechnology Drugs Anhui, University of Science and Technology of China, Hefei, 230027, Anhui, China
| | - Hao Li
- Department of Oncology of the First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Institute of Immunology, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Engineering Technology Research Center of Biotechnology Drugs Anhui, University of Science and Technology of China, Hefei, 230027, Anhui, China
| | - Yehai Wang
- Department of Oncology of the First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Institute of Immunology, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Engineering Technology Research Center of Biotechnology Drugs Anhui, University of Science and Technology of China, Hefei, 230027, Anhui, China
| | - Yan Fan
- Department of Oncology of the First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Institute of Immunology, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Engineering Technology Research Center of Biotechnology Drugs Anhui, University of Science and Technology of China, Hefei, 230027, Anhui, China
| | - Kang An
- Department of Oncology of the First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Institute of Immunology, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Engineering Technology Research Center of Biotechnology Drugs Anhui, University of Science and Technology of China, Hefei, 230027, Anhui, China
| | - Yu Wu
- Department of Oncology of the First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Institute of Immunology, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Engineering Technology Research Center of Biotechnology Drugs Anhui, University of Science and Technology of China, Hefei, 230027, Anhui, China
| | - Weihua Xiao
- Department of Oncology of the First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China.
- Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, Anhui, China.
- Institute of Immunology, University of Science and Technology of China, Hefei, 230027, Anhui, China.
- Engineering Technology Research Center of Biotechnology Drugs Anhui, University of Science and Technology of China, Hefei, 230027, Anhui, China.
| |
Collapse
|
13
|
Piccinelli S, Romee R, Shapiro RM. The natural killer cell immunotherapy platform: an overview of the landscape of clinical trials in liquid and solid tumors. Semin Hematol 2023; 60:42-51. [PMID: 37080710 DOI: 10.1053/j.seminhematol.2023.02.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 02/23/2023] [Indexed: 03/07/2023]
Abstract
The translation of natural killer (NK) cells to the treatment of malignant disease has made significant progress in the last few decades. With a variety of available sources and improvements in both in vitro and in vivo NK cell expansion, the NK cell immunotherapy platform has come into its own. The enormous effort continues to further optimize this platform, including ways to enhance NK cell persistence, trafficking to the tumor microenvironment, and cytotoxicity. As this effort bears fruit, it is translated into a plethora of clinical trials in patients with advanced malignancies. The adoptive transfer of NK cells, either as a standalone therapy or in combination with other immunotherapies, has been applied for the treatment of both liquid and solid tumors, with numerous early-phase trials showing promising results. This review aims to summarize the key advantages of NK cell immunotherapy, highlight several of the current approaches being taken for its optimization, and give an overview of the landscape of clinical trials translating this platform into clinic.
Collapse
|
14
|
Steinkamp MP, Lagutina I, Brayer KJ, Schultz F, Burke D, Pankratz VS, Adams SF, Hudson LG, Ness SA, Wandinger-Ness A. Humanized Patient-derived Xenograft Models of Disseminated Ovarian Cancer Recapitulate Key Aspects of the Tumor Immune Environment within the Peritoneal Cavity. CANCER RESEARCH COMMUNICATIONS 2023; 3:309-324. [PMID: 36860657 PMCID: PMC9973420 DOI: 10.1158/2767-9764.crc-22-0300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 11/23/2022] [Accepted: 02/01/2023] [Indexed: 06/18/2023]
Abstract
The importance of the immune microenvironment in ovarian cancer progression, metastasis, and response to therapies has become increasingly clear, especially with the new emphasis on immunotherapies. To leverage the power of patient-derived xenograft (PDX) models within a humanized immune microenvironment, three ovarian cancer PDXs were grown in humanized NBSGW (huNBSGW) mice engrafted with human CD34+ cord blood-derived hematopoietic stem cells. Analysis of cytokine levels in the ascites fluid and identification of infiltrating immune cells in the tumors demonstrated that these humanized PDX (huPDX) established an immune tumor microenvironment similar to what has been reported for patients with ovarian cancer. The lack of human myeloid cell differentiation has been a major setback for humanized mouse models, but our analysis shows that PDX engraftment increases the human myeloid population in the peripheral blood. Analysis of cytokines within the ascites fluid of huPDX revealed high levels of human M-CSF, a key myeloid differentiation factor as well as other elevated cytokines that have previously been identified in ovarian cancer patient ascites fluid including those involved in immune cell differentiation and recruitment. Human tumor-associated macrophages and tumor-infiltrating lymphocytes were detected within the tumors of humanized mice, demonstrating immune cell recruitment to tumors. Comparison of the three huPDX revealed certain differences in cytokine signatures and in the extent of immune cell recruitment. Our studies show that huNBSGW PDX models reconstitute important aspects of the ovarian cancer immune tumor microenvironment, which may recommend these models for preclinical therapeutic trials. Significance huPDX models are ideal preclinical models for testing novel therapies. They reflect the genetic heterogeneity of the patient population, enhance human myeloid differentiation, and recruit immune cells to the tumor microenvironment.
Collapse
Affiliation(s)
- Mara P. Steinkamp
- Department of Pathology, University of New Mexico School of Medicine, Albuquerque, New Mexico
- Comprehensive Cancer Center, University of New Mexico, Albuquerque, New Mexico
| | - Irina Lagutina
- Comprehensive Cancer Center, University of New Mexico, Albuquerque, New Mexico
| | - Kathryn J. Brayer
- Analytical and Translational Genomics Shared Resource, Comprehensive Cancer Center, University of New Mexico, Albuquerque, New Mexico
| | - Fred Schultz
- Department of Pathology, University of New Mexico School of Medicine, Albuquerque, New Mexico
| | - Danielle Burke
- Department of Pathology, University of New Mexico School of Medicine, Albuquerque, New Mexico
| | - Vernon S. Pankratz
- Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, New Mexico
- Biostatistics Shared Resource, Comprehensive Cancer Center, University of New Mexico, Albuquerque, New Mexico
| | - Sarah F. Adams
- Comprehensive Cancer Center, University of New Mexico, Albuquerque, New Mexico
- Department of Obstetrics and Gynecology, University of New Mexico School of Medicine, Albuquerque, New Mexico
| | - Laurie G. Hudson
- Comprehensive Cancer Center, University of New Mexico, Albuquerque, New Mexico
- Department of Pharmaceutical Sciences, University of New Mexico School of Medicine, Albuquerque, New Mexico
| | - Scott A. Ness
- Comprehensive Cancer Center, University of New Mexico, Albuquerque, New Mexico
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, New Mexico
| | - Angela Wandinger-Ness
- Department of Pathology, University of New Mexico School of Medicine, Albuquerque, New Mexico
- Comprehensive Cancer Center, University of New Mexico, Albuquerque, New Mexico
| |
Collapse
|
15
|
Subedi N, Verhagen LP, de Jonge P, Van Eyndhoven LC, van Turnhout MC, Koomen V, Baudry J, Eyer K, Dolstra H, Tel J. Single‐Cell Profiling Reveals Functional Heterogeneity and Serial Killing in Human Peripheral and Ex Vivo‐Generated CD34+ Progenitor‐Derived Natural Killer Cells. Adv Biol (Weinh) 2022; 7:e2200207. [PMID: 36517083 DOI: 10.1002/adbi.202200207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 11/25/2022] [Indexed: 12/23/2022]
Abstract
Increasing evidence suggests that natural killer (NK) cells are composed of distinct functional subsets. This multifunctional role has made them an attractive choice for anticancer immunotherapy. A functional NK cell repertoire is generated through cellular education, resulting in a heterogeneous NK cell population with distinct capabilities responding to different stimuli. The application of a high-throughput droplet-based microfluidic platform allows monitoring of NK cell-target cell interactions at the single-cell level and in real-time. A variable response of single NK cells toward different target cells is observed, and a distinct population of NK cells (serial killers) capable of inducing multiple target lysis is identified. By assessing the cytotoxic dynamics, it is shown that single umbilical cord blood-derived CD34+ hematopoietic progenitor (HPC)-NK cells display superior antitumor cytotoxicity. With an integrated analysis of cytotoxicity and cytokine secretion, it is shown that target cell interactions augment cytotoxic as well as secretory behavior of NK cells. By providing an integrated assessment of NK cell functions by microfluidics, this study paves the way to further functionally characterize NK cells ultimately aimed to improve cancer immunotherapy.
Collapse
Affiliation(s)
- Nikita Subedi
- Laboratory of Immunoengineering Department of Biomedical Engineering Eindhoven University of Technology Groene Loper 5 Eindhoven 5600 MB The Netherlands
- Institute for Complex Molecular Systems Eindhoven University of Technology Groene Loper 5 Eindhoven 5600 MB The Netherlands
| | - Liesbeth Petronella Verhagen
- Laboratory of Immunoengineering Department of Biomedical Engineering Eindhoven University of Technology Groene Loper 5 Eindhoven 5600 MB The Netherlands
- Institute for Complex Molecular Systems Eindhoven University of Technology Groene Loper 5 Eindhoven 5600 MB The Netherlands
| | - Paul de Jonge
- Department of Laboratory Medicine – Laboratory of Hematology Radboud Institute of Molecular Life Sciences Radboud University Medical Center Nijmegen 6525 GA The Netherlands
| | - Laura C. Van Eyndhoven
- Laboratory of Immunoengineering Department of Biomedical Engineering Eindhoven University of Technology Groene Loper 5 Eindhoven 5600 MB The Netherlands
- Institute for Complex Molecular Systems Eindhoven University of Technology Groene Loper 5 Eindhoven 5600 MB The Netherlands
| | - Mark C. van Turnhout
- Soft Tissue Engineering and Mechanobiology Department of Biomedical Engineering Eindhoven University of Technology Groene Loper 5 Eindhoven 5600 MB The Netherlands
| | - Vera Koomen
- Laboratory of Immunoengineering Department of Biomedical Engineering Eindhoven University of Technology Groene Loper 5 Eindhoven 5600 MB The Netherlands
| | - Jean Baudry
- Laboratoire Colloïdes et Matériaux Divisés (LCMD) ESPCI Paris PSL Research University CNRS UMR8231 Chimie Biologie Innovation Paris 75005 France
| | - Klaus Eyer
- Laboratoire Colloïdes et Matériaux Divisés (LCMD) ESPCI Paris PSL Research University CNRS UMR8231 Chimie Biologie Innovation Paris 75005 France
- Laboratory for Functional Immune Repertoire Analysis Institute of Pharmaceutical Sciences D‐CHAB, ETH, Zürich Zurich 8093 Switzerland
| | - Harry Dolstra
- Department of Laboratory Medicine – Laboratory of Hematology Radboud Institute of Molecular Life Sciences Radboud University Medical Center Nijmegen 6525 GA The Netherlands
| | - Jurjen Tel
- Laboratory of Immunoengineering Department of Biomedical Engineering Eindhoven University of Technology Groene Loper 5 Eindhoven 5600 MB The Netherlands
- Institute for Complex Molecular Systems Eindhoven University of Technology Groene Loper 5 Eindhoven 5600 MB The Netherlands
| |
Collapse
|
16
|
Abstract
Natural killer (NK) cells comprise a unique population of innate lymphoid cells endowed with intrinsic abilities to identify and eliminate virally infected cells and tumour cells. Possessing multiple cytotoxicity mechanisms and the ability to modulate the immune response through cytokine production, NK cells play a pivotal role in anticancer immunity. This role was elucidated nearly two decades ago, when NK cells, used as immunotherapeutic agents, showed safety and efficacy in the treatment of patients with advanced-stage leukaemia. In recent years, following the paradigm-shifting successes of chimeric antigen receptor (CAR)-engineered adoptive T cell therapy and the advancement in technologies that can turn cells into powerful antitumour weapons, the interest in NK cells as a candidate for immunotherapy has grown exponentially. Strategies for the development of NK cell-based therapies focus on enhancing NK cell potency and persistence through co-stimulatory signalling, checkpoint inhibition and cytokine armouring, and aim to redirect NK cell specificity to the tumour through expression of CAR or the use of engager molecules. In the clinic, the first generation of NK cell therapies have delivered promising results, showing encouraging efficacy and remarkable safety, thus driving great enthusiasm for continued innovation. In this Review, we describe the various approaches to augment NK cell cytotoxicity and longevity, evaluate challenges and opportunities, and reflect on how lessons learned from the clinic will guide the design of next-generation NK cell products that will address the unique complexities of each cancer.
Collapse
Affiliation(s)
- Tamara J Laskowski
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Alexander Biederstädt
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
- Department of Medicine III: Hematology and Oncology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Katayoun Rezvani
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
17
|
Cord Blood-Derived Natural Killer Cell Exploitation in Immunotherapy Protocols: More Than a Promise? Cancers (Basel) 2022; 14:cancers14184439. [PMID: 36139598 PMCID: PMC9496735 DOI: 10.3390/cancers14184439] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/25/2022] [Accepted: 09/09/2022] [Indexed: 11/30/2022] Open
Abstract
Simple Summary NK cell anti-tumor activity against hematological malignancies is well-established and many studies support their role in the control of solid tumor growth and metastasis generation. However, tumor microenvironment may affect NK cell function. Ongoing studies are aimed to design novel immunotherapeutic protocols to combine NK cell-based immunotherapy with other therapeutic strategies to improve the anti-tumor NK cell response. In this context, UCB is one of the main sources of both mature NK cells and of CD34+ HSPC that can generate NK cells, both in-vivo and in-vitro. UCB-derived NK cells represent a valuable tool to perform in-vitro and preclinical analyses and are already used in several clinical settings, particularly against hematological malignancies. The present review describes the characteristics of different types of UCB-derived NK cells and the in-vitro models to expand them, both for research and clinical purposes in the context of cancer immunotherapy. Abstract In the last 20 years, Natural Killer (NK) cell-based immunotherapy has become a promising approach to target various types of cancer. Indeed, NK cells play a pivotal role in the first-line defense against tumors through major histocompatibility complex-independent immunosurveillance. Their role in the control of leukemia relapse has been clearly established and, moreover, the presence of NK cells in the tumor microenvironment (TME) generally correlates with good prognosis. However, it has also been observed that, often, NK cells poorly infiltrate the tumor tissue, and, in TME, their functions may be compromised by immunosuppressive factors that contribute to the failure of anti-cancer immune response. Currently, studies are focused on the design of effective strategies to expand NK cells and enhance their cytotoxic activity, exploiting different cell sources, such as peripheral blood (PB), umbilical cord blood (UCB) and NK cell lines. Among them, UCB represents an important source of mature NK cells and CD34+ Hematopoietic Stem and Progenitor Cells (HSPCs), as precursors of NK cells. In this review, we summarize the UCB-derived NK cell activity in the tumor context, review the different in-vitro models to expand NK cells from UCB, and discuss the importance of their exploitation in anti-tumor immunotherapy protocols.
Collapse
|
18
|
Preclinical In Vitro and In Vivo Models for Adoptive Cell Therapy of Cancer. Cancer J 2022; 28:257-262. [PMID: 35880934 DOI: 10.1097/ppo.0000000000000609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
ABSTRACT Adoptive cellular therapies are making major strides in the treatment of cancer, both for hematologic and solid tumors. These cellular products include chimeric antigen receptor T cells and T-cell receptor-modified T cells, tumor-infiltrating lymphocytes, marrow-infiltrating T cells, natural killer cells as well as macrophage-based therapeutics. Advancement in genomics, computational biology, immunology, and cell therapy manufacturing has facilitated advancement of adoptive T cell therapies into the clinic, whereas clinical efficacy has driven Food and Drug Administration approvals. The growth of adoptive cellular therapy has, in turn, led to innovation in the preclinical models available, from ex vivo cell-based models to in vivo xenograft models of treatment. This review focuses on the development and application of in vitro models and in vivo models (cell line xenograft, humanized mice, and patient-derived xenograft models) that directly evaluate these human cellular products.
Collapse
|
19
|
Vitale C, Marzagalli M, Scaglione S, Dondero A, Bottino C, Castriconi R. Tumor Microenvironment and Hydrogel-Based 3D Cancer Models for In Vitro Testing Immunotherapies. Cancers (Basel) 2022; 14:1013. [PMID: 35205760 PMCID: PMC8870468 DOI: 10.3390/cancers14041013] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/12/2022] [Accepted: 02/14/2022] [Indexed: 02/05/2023] Open
Abstract
In recent years, immunotherapy has emerged as a promising novel therapeutic strategy for cancer treatment. In a relevant percentage of patients, however, clinical benefits are lower than expected, pushing researchers to deeply analyze the immune responses against tumors and find more reliable and efficient tools to predict the individual response to therapy. Novel tissue engineering strategies can be adopted to realize in vitro fully humanized matrix-based models, as a compromise between standard two-dimensional (2D) cell cultures and animal tests, which are costly and hardly usable in personalized medicine. In this review, we describe the main mechanisms allowing cancer cells to escape the immune surveillance, which may play a significant role in the failure of immunotherapies. In particular, we discuss the role of the tumor microenvironment (TME) in the establishment of a milieu that greatly favors cancer malignant progression and impact on the interactions with immune cells. Then, we present an overview of the recent in vitro engineered preclinical three-dimensional (3D) models that have been adopted to resemble the interplays between cancer and immune cells and for testing current therapies and immunotherapeutic approaches. Specifically, we focus on 3D hydrogel-based tools based on different types of polymers, discussing the suitability of each of them in reproducing the TME key features based on their intrinsic or tunable characteristics. Finally, we introduce the possibility to combine the 3D models with technological fluid dynamics platforms, reproducing the dynamic complex interactions between tumor cells and immune effectors migrated in situ via the systemic circulation, pointing out the challenges that still have to be overcome for setting more predictive preclinical assays.
Collapse
Affiliation(s)
- Chiara Vitale
- Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy; (C.V.); (A.D.); (R.C.)
| | | | - Silvia Scaglione
- React4life SRL, 16121 Genova, Italy; (M.M.); (S.S.)
- National Research Council of Italy, Institute of Electronics, Information Engineering and Telecommunications (IEIIT), 16149 Genova, Italy
| | - Alessandra Dondero
- Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy; (C.V.); (A.D.); (R.C.)
| | - Cristina Bottino
- Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy; (C.V.); (A.D.); (R.C.)
- IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
| | - Roberta Castriconi
- Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy; (C.V.); (A.D.); (R.C.)
| |
Collapse
|
20
|
Hombach A, Barden M, Hannappel L, Chmielewski M, Rappl G, Sachinidis A, Abken H. IL12 integrated into the CAR exodomain converts CD8 + T cells to poly-functional NK-like cells with superior killing of antigen-loss tumors. Mol Ther 2022; 30:593-605. [PMID: 34678512 PMCID: PMC8821972 DOI: 10.1016/j.ymthe.2021.10.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 08/30/2021] [Accepted: 10/10/2021] [Indexed: 02/04/2023] Open
Abstract
Chimeric antigen receptor (CAR)-redirected T cell therapy often fails to control tumors in the long term due to selecting cancer cells that downregulated or lost CAR targeted antigen. To reprogram the functional capacities specifically of engineered CAR T cells, we inserted IL12 into the extracellular moiety of a CD28-ζ CAR; both the CAR endodomain and IL12 were functionally active, as indicated by antigen-redirected effector functions and STAT4 phosphorylation, respectively. The IL12-CAR reprogrammed CD8+ T cells toward a so far not recognized natural killer (NK) cell-like signature and a CD94+CD56+CD62Lhigh phenotype closely similar, but not identical, to NK and cytokine induced killer (CIK) cells. In contrast to conventional CAR T cells, IL12-CAR T cells acquired antigen-independent, human leukocyte antigen E (HLA-E) restricted cytotoxic capacities eliminating antigen-negative cancer cells in addition to eliminating cancer cells with CAR cognate antigen. Simultaneous signaling through both the CAR endodomain and IL12 were required for inducing maximal NK-like cytotoxicity; adding IL12 to conventional CAR T cells was not sufficient. Antigen-negative tumors were attacked by IL12-CAR T cells, but not by conventional CAR T cells. Overall, we present a prototype of a new family of CARs that augments tumor recognition and elimination through expanded functional capacities by an appropriate cytokine integrated into the CAR exodomain.
Collapse
Affiliation(s)
- Andreas Hombach
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany,Department I Internal Medicine, University Hospital Cologne, 50931 Cologne, Germany
| | - Markus Barden
- RCI, Regensburg Center for Interventional Immunology, Department Genetic Immunotherapy, and University Hospital Regensburg, 93053 Regensburg, Germany
| | - Lisa Hannappel
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Markus Chmielewski
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany,Department I Internal Medicine, University Hospital Cologne, 50931 Cologne, Germany
| | - Gunter Rappl
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Agapios Sachinidis
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany,University of Cologne, Faculty of Medicine and Center for Physiology, University Hospital Cologne, 50931 Cologne, Germany
| | - Hinrich Abken
- RCI, Regensburg Center for Interventional Immunology, Department Genetic Immunotherapy, and University Hospital Regensburg, 93053 Regensburg, Germany,Corresponding author: Hinrich Abken, RCI, Regensburg Center for Interventional Immunology, Department Genetic Immunotherapy, and University Hospital Regensburg, 93053 Regensburg, Germany.
| |
Collapse
|
21
|
Ullmo I, Koksal N, Ang HYK, Brady HJM. In Vitro Development of Mouse and Human NK Cells from Hematopoietic Progenitor Cells. Methods Mol Biol 2022; 2463:31-45. [PMID: 35344165 DOI: 10.1007/978-1-0716-2160-8_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Natural killer (NK) cells are lymphocytes that play an important role at clearing virally infected or cancer cells. Their potential and role in cancer immunotherapy have generated great interest, given the promising results of NK cell adoptive transfer clinical trials. The remaining challenge to bring emerging NK cell immunotherapies to the clinic is to enhance the production of large numbers of functionally competent NK cells ex vivo. Here, we describe two in vitro NK cell development assays using hematopoietic progenitor cells (HPCs), one for human NK cells and one for mouse NK cells. These protocols describe two robust methods that can be utilized for investigation of NK cell development and function.
Collapse
Affiliation(s)
- Ines Ullmo
- Department of Life Sciences, Imperial College London, London, UK
| | - Nahide Koksal
- Department of Life Sciences, Imperial College London, London, UK
| | - Heather Y K Ang
- Department of Life Sciences, Imperial College London, London, UK
| | - Hugh J M Brady
- Department of Life Sciences, Imperial College London, London, UK.
| |
Collapse
|
22
|
Chen M, Li Y, Wu Y, Xie S, Ma J, Yue J, Lv R, Tian Z, Fang F, Xiao W. Anti-Tumor Activity of Expanded PBMC-Derived NK Cells by Feeder-Free Protocol in Ovarian Cancer. Cancers (Basel) 2021; 13:5866. [PMID: 34831019 PMCID: PMC8616155 DOI: 10.3390/cancers13225866] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 11/22/2022] Open
Abstract
Natural killer (NK) cells have shown great therapeutic potential against a wide range of cancers due to their pan-specific target recognition. Numerous reports indicate that NK cell immunotherapy is an effective therapeutic approach for treating hematological malignancies, but shows limited effects against solid tumors. In this study, several models of ovarian cancer (OC) were used to test the anti-cancer effects of NK cells derived from human peripheral blood mononuclear cells and expanded using a feeder cell-free expansion system (eNKs). The results show that eNKs exhibit potent inhibitory activity on tumor growth in different ovarian cancer xenograft mice (i.e., solid tumors, abdominal metastatic tumors, and ascites), importantly, in a dose-dependent manner. Moreover, adoptive transfer of eNKs resulted in significant reduction in ascites formation in OC peritoneal tumor models, and especially in reducing intraperitoneal ascites. We found that eNKs could migrate to the tumor site, retain their activity, and proliferate to maintain high cell counts in cutaneous xenograft mice. In addition, when increased the infusion with a high dose of 12 × 107 cells/mouse, Graft-versus-host disease could be induced by eNK. These data show that eNK cell immunotherapy could be a promising treatment strategy for ovarian cancers, including solid tumors and ascites.
Collapse
Affiliation(s)
- Minhua Chen
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; (M.C.); (Y.L.); (Y.W.); (S.X.); (J.M.); (J.Y.); (Z.T.)
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
- Institute of Immunology, University of Science and Technology of China, Hefei 230027, China
- Engineering Technology Research Center of Biotechnology Drugs Anhui, University of Science and Technology of China, Hefei 230027, China
| | - Yutong Li
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; (M.C.); (Y.L.); (Y.W.); (S.X.); (J.M.); (J.Y.); (Z.T.)
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
- Institute of Immunology, University of Science and Technology of China, Hefei 230027, China
- Engineering Technology Research Center of Biotechnology Drugs Anhui, University of Science and Technology of China, Hefei 230027, China
| | - Yu Wu
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; (M.C.); (Y.L.); (Y.W.); (S.X.); (J.M.); (J.Y.); (Z.T.)
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
- Institute of Immunology, University of Science and Technology of China, Hefei 230027, China
- Engineering Technology Research Center of Biotechnology Drugs Anhui, University of Science and Technology of China, Hefei 230027, China
| | - Siqi Xie
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; (M.C.); (Y.L.); (Y.W.); (S.X.); (J.M.); (J.Y.); (Z.T.)
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
- Institute of Immunology, University of Science and Technology of China, Hefei 230027, China
- Engineering Technology Research Center of Biotechnology Drugs Anhui, University of Science and Technology of China, Hefei 230027, China
| | - Jie Ma
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; (M.C.); (Y.L.); (Y.W.); (S.X.); (J.M.); (J.Y.); (Z.T.)
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
- Institute of Immunology, University of Science and Technology of China, Hefei 230027, China
- Engineering Technology Research Center of Biotechnology Drugs Anhui, University of Science and Technology of China, Hefei 230027, China
| | - Jingjing Yue
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; (M.C.); (Y.L.); (Y.W.); (S.X.); (J.M.); (J.Y.); (Z.T.)
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
- Institute of Immunology, University of Science and Technology of China, Hefei 230027, China
- Engineering Technology Research Center of Biotechnology Drugs Anhui, University of Science and Technology of China, Hefei 230027, China
| | - Rong Lv
- Blood Transfusion Laboratory, Anhui Blood Center, Hefei 230031, China;
| | - Zhigang Tian
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; (M.C.); (Y.L.); (Y.W.); (S.X.); (J.M.); (J.Y.); (Z.T.)
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
- Institute of Immunology, University of Science and Technology of China, Hefei 230027, China
- Engineering Technology Research Center of Biotechnology Drugs Anhui, University of Science and Technology of China, Hefei 230027, China
| | - Fang Fang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; (M.C.); (Y.L.); (Y.W.); (S.X.); (J.M.); (J.Y.); (Z.T.)
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
- Institute of Immunology, University of Science and Technology of China, Hefei 230027, China
- Engineering Technology Research Center of Biotechnology Drugs Anhui, University of Science and Technology of China, Hefei 230027, China
| | - Weihua Xiao
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; (M.C.); (Y.L.); (Y.W.); (S.X.); (J.M.); (J.Y.); (Z.T.)
- Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
- Institute of Immunology, University of Science and Technology of China, Hefei 230027, China
- Engineering Technology Research Center of Biotechnology Drugs Anhui, University of Science and Technology of China, Hefei 230027, China
| |
Collapse
|
23
|
Van der Meer JMR, de Jonge PKJD, van der Waart AB, Geerlings AC, Moonen JP, Brummelman J, de Klein J, Vermeulen MC, Maas RJA, Schaap NPM, Hoogstad-van Evert JS, Ottevanger PB, Jansen JH, Hobo W, Dolstra H. CD34 + progenitor-derived NK cell and gemcitabine combination therapy increases killing of ovarian cancer cells in NOD/SCID/IL2Rg null mice. Oncoimmunology 2021; 10:1981049. [PMID: 34616589 PMCID: PMC8489932 DOI: 10.1080/2162402x.2021.1981049] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Combining natural killer (NK) cell adoptive transfer with tumor-sensitizing chemotherapy is an attractive approach against recurrent ovarian cancer (OC), as OC is sensitive to NK cell-mediated immunity. Previously, we showed that CD34+ hematopoietic progenitor cell (HPC)-derived NK cells can kill OC cells in vitro and inhibit OC tumor growth in mice. Here, we investigated the potential of HPC-NK cell therapy combined with chemotherapeutic gemcitabine (used in recurrent OC patients) against OC. We examined the phenotypical, functional, and cytotoxic effects of gemcitabine on HPC-NK cells and/or OC cells in vitro and in OC-bearing mice. To this end, we treated OC cells and/or HPC-NK cells with or without gemcitabine and analyzed the phenotype, cytokine production, and anti-tumor reactivity. We found that gemcitabine did not affect the phenotype and functionality of HPC-NK cells, while on OC cells expression of NK cell activating ligands and death receptors was upregulated. Although gemcitabine pre-treatment of OC cells did not improve the functionality of HPC-NK cells, importantly, HPC-NK cells and gemcitabine additively killed OC cells in vitro. Similarly, combined HPC-NK cell and gemcitabine treatment additively decreased tumor growth in OC-bearing mice. Collectively, our results indicate that combination therapy of HPC-NK cells and gemcitabine results in augmented OC killing in vitro and in vivo. This provides a rationale for exploring this therapeutic strategy in patients with recurrent OC.
Collapse
Affiliation(s)
- Jolien M R Van der Meer
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center/Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Paul K J D de Jonge
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center/Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Anniek B van der Waart
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center/Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Alexander C Geerlings
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center/Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Jurgen P Moonen
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center/Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Jolanda Brummelman
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center/Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Janne de Klein
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center/Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Malou C Vermeulen
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center/Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Ralph J A Maas
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center/Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Nicolaas P M Schaap
- Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Janneke S Hoogstad-van Evert
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center/Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands.,Department of Obstetrics and Gynecology, Amphia Hospital, The Netherlands
| | - Petronella B Ottevanger
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Joop H Jansen
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center/Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Willemijn Hobo
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center/Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Harry Dolstra
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center/Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| |
Collapse
|
24
|
Stem cells-derived natural killer cells for cancer immunotherapy: current protocols, feasibility, and benefits of ex vivo generated natural killer cells in treatment of advanced solid tumors. Cancer Immunol Immunother 2021; 70:3369-3395. [PMID: 34218295 DOI: 10.1007/s00262-021-02975-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 05/26/2021] [Indexed: 12/13/2022]
Abstract
Nowadays, natural killer (NK) cell-based immunotherapy provides a practical therapeutic strategy for patients with advanced solid tumors (STs). This approach is adaptively conducted by the autologous and identical NK cells after in vitro expansion and overnight activation. However, the NK cell-based cancer immunotherapy has been faced with some fundamental and technical limitations. Moreover, the desirable outcomes of the NK cell therapy may not be achieved due to the complex tumor microenvironment by inhibition of intra-tumoral polarization and cytotoxicity of implanted NK cells. Currently, stem cells (SCs) technology provides a powerful opportunity to generate more effective and universal sources of the NK cells. Till now, several strategies have been developed to differentiate types of the pluripotent and adult SCs into the mature NK cells, with both feeder layer-dependent and/or feeder laye-free strategies. Higher cytokine production and intra-tumoral polarization capabilities as well as stronger anti-tumor properties are the main features of these SCs-derived NK cells. The present review article focuses on the principal barriers through the conventional NK cell immunotherapies for patients with advanced STs. It also provides a comprehensive resource of protocols regarding the generation of SCs-derived NK cells in an ex vivo condition.
Collapse
|
25
|
Islam R, Pupovac A, Evtimov V, Boyd N, Shu R, Boyd R, Trounson A. Enhancing a Natural Killer: Modification of NK Cells for Cancer Immunotherapy. Cells 2021; 10:cells10051058. [PMID: 33946954 PMCID: PMC8146003 DOI: 10.3390/cells10051058] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/27/2021] [Accepted: 04/27/2021] [Indexed: 12/14/2022] Open
Abstract
Natural killer (NK) cells are potent innate immune system effector lymphocytes armed with multiple mechanisms for killing cancer cells. Given the dynamic roles of NK cells in tumor surveillance, they are fast becoming a next-generation tool for adoptive immunotherapy. Many strategies are being employed to increase their number and improve their ability to overcome cancer resistance and the immunosuppressive tumor microenvironment. These include the use of cytokines and synthetic compounds to bolster propagation and killing capacity, targeting immune-function checkpoints, addition of chimeric antigen receptors (CARs) to provide cancer specificity and genetic ablation of inhibitory molecules. The next generation of NK cell products will ideally be readily available as an “off-the-shelf” product and stem cell derived to enable potentially unlimited supply. However, several considerations regarding NK cell source, genetic modification and scale up first need addressing. Understanding NK cell biology and interaction within specific tumor contexts will help identify necessary NK cell modifications and relevant choice of NK cell source. Further enhancement of manufacturing processes will allow for off-the-shelf NK cell immunotherapies to become key components of multifaceted therapeutic strategies for cancer.
Collapse
Affiliation(s)
- Rasa Islam
- Cartherics Pty Ltd., Clayton 3168, Australia; (R.I.); (A.P.); (V.E.); (N.B.); (R.S.); (R.B.)
- Department of Obstetrics and Gynaecology, Monash University, Clayton 3168, Australia
| | - Aleta Pupovac
- Cartherics Pty Ltd., Clayton 3168, Australia; (R.I.); (A.P.); (V.E.); (N.B.); (R.S.); (R.B.)
| | - Vera Evtimov
- Cartherics Pty Ltd., Clayton 3168, Australia; (R.I.); (A.P.); (V.E.); (N.B.); (R.S.); (R.B.)
| | - Nicholas Boyd
- Cartherics Pty Ltd., Clayton 3168, Australia; (R.I.); (A.P.); (V.E.); (N.B.); (R.S.); (R.B.)
| | - Runzhe Shu
- Cartherics Pty Ltd., Clayton 3168, Australia; (R.I.); (A.P.); (V.E.); (N.B.); (R.S.); (R.B.)
| | - Richard Boyd
- Cartherics Pty Ltd., Clayton 3168, Australia; (R.I.); (A.P.); (V.E.); (N.B.); (R.S.); (R.B.)
| | - Alan Trounson
- Cartherics Pty Ltd., Clayton 3168, Australia; (R.I.); (A.P.); (V.E.); (N.B.); (R.S.); (R.B.)
- Department of Obstetrics and Gynaecology, Monash University, Clayton 3168, Australia
- Correspondence:
| |
Collapse
|
26
|
Horowitz NB, Mohammad I, Moreno-Nieves UY, Koliesnik I, Tran Q, Sunwoo JB. Humanized Mouse Models for the Advancement of Innate Lymphoid Cell-Based Cancer Immunotherapies. Front Immunol 2021; 12:648580. [PMID: 33968039 PMCID: PMC8100438 DOI: 10.3389/fimmu.2021.648580] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 03/11/2021] [Indexed: 12/12/2022] Open
Abstract
Innate lymphoid cells (ILCs) are a branch of the immune system that consists of diverse circulating and tissue-resident cells, which carry out functions including homeostasis and antitumor immunity. The development and behavior of human natural killer (NK) cells and other ILCs in the context of cancer is still incompletely understood. Since NK cells and Group 1 and 2 ILCs are known to be important for mediating antitumor immune responses, a clearer understanding of these processes is critical for improving cancer treatments and understanding tumor immunology as a whole. Unfortunately, there are some major differences in ILC differentiation and effector function pathways between humans and mice. To this end, mice bearing patient-derived xenografts or human cell line-derived tumors alongside human genes or human immune cells represent an excellent tool for studying these pathways in vivo. Recent advancements in humanized mice enable unparalleled insights into complex tumor-ILC interactions. In this review, we discuss ILC behavior in the context of cancer, the humanized mouse models that are most commonly employed in cancer research and their optimization for studying ILCs, current approaches to manipulating human ILCs for antitumor activity, and the relative utility of various mouse models for the development and assessment of these ILC-related immunotherapies.
Collapse
Affiliation(s)
- Nina B Horowitz
- Department of Otolaryngology-Head and Neck Surgery, Stanford Cancer Institute and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States.,Department of Bioengineering, Stanford University School of Medicine and School of Engineering, Stanford, CA, United States
| | - Imran Mohammad
- Department of Otolaryngology-Head and Neck Surgery, Stanford Cancer Institute and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Uriel Y Moreno-Nieves
- Department of Otolaryngology-Head and Neck Surgery, Stanford Cancer Institute and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Ievgen Koliesnik
- Department of Otolaryngology-Head and Neck Surgery, Stanford Cancer Institute and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Quan Tran
- Department of Otolaryngology-Head and Neck Surgery, Stanford Cancer Institute and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - John B Sunwoo
- Department of Otolaryngology-Head and Neck Surgery, Stanford Cancer Institute and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
27
|
Van der Meer JMR, Maas RJA, Guldevall K, Klarenaar K, de Jonge PKJD, Evert JSHV, van der Waart AB, Cany J, Safrit JT, Lee JH, Wagena E, Friedl P, Önfelt B, Massuger LF, Schaap NPM, Jansen JH, Hobo W, Dolstra H. IL-15 superagonist N-803 improves IFNγ production and killing of leukemia and ovarian cancer cells by CD34 + progenitor-derived NK cells. Cancer Immunol Immunother 2020; 70:1305-1321. [PMID: 33140189 PMCID: PMC8053152 DOI: 10.1007/s00262-020-02749-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 10/14/2020] [Indexed: 12/17/2022]
Abstract
Allogeneic natural killer (NK) cell transfer is a potential immunotherapy to eliminate and control cancer. A promising source are CD34 + hematopoietic progenitor cells (HPCs), since large numbers of cytotoxic NK cells can be generated. Effective boosting of NK cell function can be achieved by interleukin (IL)-15. However, its in vivo half-life is short and potent trans-presentation by IL-15 receptor α (IL-15Rα) is absent. Therefore, ImmunityBio developed IL-15 superagonist N-803, which combines IL-15 with an activating mutation, an IL-15Rα sushi domain for trans-presentation, and IgG1-Fc for increased half-life. Here, we investigated whether and how N-803 improves HPC-NK cell functionality in leukemia and ovarian cancer (OC) models in vitro and in vivo in OC-bearing immunodeficient mice. We used flow cytometry-based assays, enzyme-linked immunosorbent assay, microscopy-based serial killing assays, and bioluminescence imaging, for in vitro and in vivo experiments. N-803 increased HPC-NK cell proliferation and interferon (IFN)γ production. On leukemia cells, co-culture with HPC-NK cells and N-803 increased ICAM-1 expression. Furthermore, N-803 improved HPC-NK cell-mediated (serial) leukemia killing. Treating OC spheroids with HPC-NK cells and N-803 increased IFNγ-induced CXCL10 secretion, and target killing after prolonged exposure. In immunodeficient mice bearing human OC, N-803 supported HPC-NK cell persistence in combination with total human immunoglobulins to prevent Fc-mediated HPC-NK cell depletion. Moreover, this combination treatment decreased tumor growth. In conclusion, N-803 is a promising IL-15-based compound that boosts HPC-NK cell expansion and functionality in vitro and in vivo. Adding N-803 to HPC-NK cell therapy could improve cancer immunotherapy.
Collapse
Affiliation(s)
- J M R Van der Meer
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 8, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - R J A Maas
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 8, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - K Guldevall
- Department of Applied Physics, Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - K Klarenaar
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 8, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - P K J D de Jonge
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 8, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - J S Hoogstad-van Evert
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 8, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
- Department of Obstetrics and Gynecology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - A B van der Waart
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 8, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - J Cany
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 8, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | | | - J H Lee
- ImmunityBio, Culver City, CA, USA
| | - E Wagena
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - P Friedl
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
- David H. Koch Center for Applied Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Cancer Genomics Center, Utrecht, The Netherlands
| | - B Önfelt
- Department of Applied Physics, Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - L F Massuger
- Department of Obstetrics and Gynecology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - N P M Schaap
- Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - J H Jansen
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 8, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - W Hobo
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 8, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - H Dolstra
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Geert Grooteplein Zuid 8, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.
| |
Collapse
|
28
|
Saito Y, Shultz LD, Ishikawa F. Understanding Normal and Malignant Human Hematopoiesis Using Next-Generation Humanized Mice. Trends Immunol 2020; 41:706-720. [PMID: 32631635 DOI: 10.1016/j.it.2020.06.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 06/12/2020] [Accepted: 06/14/2020] [Indexed: 12/11/2022]
Abstract
Rodent models for human diseases contribute significantly to understanding human physiology and pathophysiology. However, given the accelerating pace of drug development, there is a crucial need for in vivo preclinical models of human biology and pathology. The humanized mouse is one tool to bridge the gap between traditional animal models and the clinic. The development of immunodeficient mouse strains with high-level engraftment of normal and diseased human immune/hematopoietic cells has made in vivo functional characterization possible. As a patient-derived xenograft (PDX) model, humanized mice functionally correlate putative mechanisms with in vivo behavior and help to reveal pathogenic mechanisms. Combined with single-cell genomics, humanized mice can facilitate functional precision medicine such as risk stratification and individually optimized therapeutic approaches.
Collapse
Affiliation(s)
- Yoriko Saito
- RIKEN Center for Integrative Medical Sciences, Yokohama City, Kanagawa, 230-0045, Japan
| | | | - Fumihiko Ishikawa
- RIKEN Center for Integrative Medical Sciences, Yokohama City, Kanagawa, 230-0045, Japan.
| |
Collapse
|
29
|
van den Brand D, van Lith SAM, de Jong JM, Gorris MAJ, Palacio-Castañeda V, Couwenbergh ST, Goldman MRG, Ebisch I, Massuger LF, Leenders WPJ, Brock R, Verdurmen WPR. EpCAM-Binding DARPins for Targeted Photodynamic Therapy of Ovarian Cancer. Cancers (Basel) 2020; 12:E1762. [PMID: 32630661 PMCID: PMC7409335 DOI: 10.3390/cancers12071762] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 06/30/2020] [Indexed: 12/11/2022] Open
Abstract
Ovarian cancer is the most lethal gynecological malignancy due to late detection associated with dissemination throughout the abdominal cavity. Targeted photodynamic therapy (tPDT) aimed at epithelial cell adhesion molecule (EpCAM), overexpressed in over 90% of ovarian cancer metastatic lesions, is a promising novel therapeutic modality. Here, we tested the specificity and activity of conjugates of EpCAM-directed designed ankyrin repeat proteins (DARPins) with the photosensitizer IRDye 700DX in in vitro and in vivo ovarian cancer models. EpCAM-binding DARPins (Ec1: Kd = 68 pM; Ac2: Kd = 130 nM) and a control DARPin were site-specifically functionalized with fluorophores or IRDye 700DX. Conjugation of anti-EpCAM DARPins with fluorophores maintained EpCAM-specific binding in cell lines and patient-derived ovarian cancer explants. Penetration of DARPin Ec1 into tumor spheroids was slower than that of Ac2, indicative of a binding site barrier effect for Ec1. DARPin-IRDye 700DX conjugates killed EpCAM-expressing cells in a highly specific and illumination-dependent fashion in 2D and 3D cultures. Furthermore, they effectively homed to EpCAM-expressing subcutaneous OV90 xenografts in mice. In conclusion, the high activity and specificity observed in preclinical ovarian cancer models, combined with a high specificity in patient material, warrant a further investigation of EpCAM-targeted PDT for ovarian cancer.
Collapse
Affiliation(s)
- Dirk van den Brand
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands; (D.v.d.B.); (J.M.d.J.); (V.P.-C.); (S.T.C.); (M.R.G.G.); (W.P.J.L.); (R.B.)
- Department of Obstetrics and Gynaecology, Radboud University Medical Center, Geert Grooteplein 10, 6525 GA Nijmegen, The Netherlands;
| | - Sanne A. M. van Lith
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Geert Grooteplein 10, 6525 GA Nijmegen, The Netherlands;
| | - Jelske M. de Jong
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands; (D.v.d.B.); (J.M.d.J.); (V.P.-C.); (S.T.C.); (M.R.G.G.); (W.P.J.L.); (R.B.)
| | - Mark A. J. Gorris
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Geert Grooteplein 10, 6525 GA Nijmegen, The Netherlands;
| | - Valentina Palacio-Castañeda
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands; (D.v.d.B.); (J.M.d.J.); (V.P.-C.); (S.T.C.); (M.R.G.G.); (W.P.J.L.); (R.B.)
| | - Stijn T. Couwenbergh
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands; (D.v.d.B.); (J.M.d.J.); (V.P.-C.); (S.T.C.); (M.R.G.G.); (W.P.J.L.); (R.B.)
| | - Mark R. G. Goldman
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands; (D.v.d.B.); (J.M.d.J.); (V.P.-C.); (S.T.C.); (M.R.G.G.); (W.P.J.L.); (R.B.)
| | - Inge Ebisch
- Department of Obstetrics and Gynaecology, Canisius Wilhelmina Hospital, Weg door Jonkerbos 100, 6532 SZ Nijmegen, The Netherlands;
| | - Leon F. Massuger
- Department of Obstetrics and Gynaecology, Radboud University Medical Center, Geert Grooteplein 10, 6525 GA Nijmegen, The Netherlands;
| | - William P. J. Leenders
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands; (D.v.d.B.); (J.M.d.J.); (V.P.-C.); (S.T.C.); (M.R.G.G.); (W.P.J.L.); (R.B.)
| | - Roland Brock
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands; (D.v.d.B.); (J.M.d.J.); (V.P.-C.); (S.T.C.); (M.R.G.G.); (W.P.J.L.); (R.B.)
| | - Wouter P. R. Verdurmen
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands; (D.v.d.B.); (J.M.d.J.); (V.P.-C.); (S.T.C.); (M.R.G.G.); (W.P.J.L.); (R.B.)
| |
Collapse
|
30
|
Martinez A, Delord JP, Ayyoub M, Devaud C. Preclinical and Clinical Immunotherapeutic Strategies in Epithelial Ovarian Cancer. Cancers (Basel) 2020; 12:E1761. [PMID: 32630708 PMCID: PMC7409311 DOI: 10.3390/cancers12071761] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 06/24/2020] [Accepted: 06/26/2020] [Indexed: 12/25/2022] Open
Abstract
In the past 20 years, the immune system has increasingly been recognized as a major player in tumor cell control, leading to considerable advances in cancer treatment. While promising with regards to melanoma, renal cancer and non-small cell lung cancer, immunotherapy provides, for the time being, limited success in other cancers, including ovarian cancer, potentially due to insufficient immunogenicity or to a particularly immunosuppressive microenvironment. In this review, we provide a global description of the immune context of ovarian cancer, in particular epithelial ovarian cancer (EOC). We describe the adaptive and innate components involved in the EOC immune response, including infiltrating tumor-specific T lymphocytes, B lymphocytes, and natural killer and myeloid cells. In addition, we highlight the rationale behind the use of EOC preclinical mouse models to assess resistance to immunotherapy, and we summarize the main preclinical studies that yielded anti-EOC immunotherapeutic strategies. Finally, we focus on major published or ongoing immunotherapy clinical trials concerning EOC.
Collapse
Affiliation(s)
- Alejandra Martinez
- Cancer Research Center of Toulouse (CRCT), Institut National de la Santé Et de la Recherche Médicale (INSERM) Unité 1037, 31037 Toulouse, France; (A.M.); (J.-P.D.); (M.A.)
- Department of Surgery, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse (IUCT), 31037 Toulouse, France
| | - Jean-Pierre Delord
- Cancer Research Center of Toulouse (CRCT), Institut National de la Santé Et de la Recherche Médicale (INSERM) Unité 1037, 31037 Toulouse, France; (A.M.); (J.-P.D.); (M.A.)
- Department of Medical Oncology, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse, 31037 Toulouse, France
- Université Toulouse III Paul Sabatier, 31037 Toulouse, France
| | - Maha Ayyoub
- Cancer Research Center of Toulouse (CRCT), Institut National de la Santé Et de la Recherche Médicale (INSERM) Unité 1037, 31037 Toulouse, France; (A.M.); (J.-P.D.); (M.A.)
- Université Toulouse III Paul Sabatier, 31037 Toulouse, France
- Immune Monitoring Core Facility, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse, 31037 Toulouse, France
| | - Christel Devaud
- Cancer Research Center of Toulouse (CRCT), Institut National de la Santé Et de la Recherche Médicale (INSERM) Unité 1037, 31037 Toulouse, France; (A.M.); (J.-P.D.); (M.A.)
- Immune Monitoring Core Facility, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse, 31037 Toulouse, France
| |
Collapse
|
31
|
Hoogstad-van Evert JS, Bekkers R, Ottevanger N, Jansen JH, Massuger L, Dolstra H. Harnessing natural killer cells for the treatment of ovarian cancer. Gynecol Oncol 2020; 157:810-816. [PMID: 32268953 DOI: 10.1016/j.ygyno.2020.03.020] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 03/15/2020] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Adoptive cellular immunotherapy could be an interesting new treatment option for ovarian carcinoma (OC), as research has demonstrated that OC is an immunogenic disease. In particular, natural killer (NK) cells have attracted attention due to their ability to kill tumor cells without prior sensitization. The therapeutic value of allogeneic NK cells has been first observed in hematological cancers and is increasingly being explored in solid tumors. METHODS To substantiate the rationale for NK cell therapy in OC we performed a literature search in the Pubmed database and in the international trial register clinicaltrials.gov with attention for the effect of OC on NK cell function, the effect of current treatment on NK cell biology and the evidence on the therapeutic value of NK cell therapy against OC. RESULTS In six clinical trials only 31 OC patients have been reported that received NK cell adoptive transfer. The majority of patients reached stable disease after NK cell therapy, with a mild pattern of side effects. In patients who received repeated infusions, more complete responses are described. All reported studies investigated the intravenous infusion of NK cells. Whereas the studies that are currently recruiting, investigate intraperitoneal infusion of allogeneic NK cells. CONCLUSION In this review the pre-clinical evidence and current trials on NK cell immunotherapy in OC patients are summarized. Furthermore, challenges that have to be overcome for NK cell adoptive therapy to have a significant impact on disease outcome are discussed.
Collapse
Affiliation(s)
- Janneke S Hoogstad-van Evert
- Department of Obstetrics and Gynecology, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Obstetrics and Gynecology, Amphia Hospital, Breda, the Netherlands.
| | - Ruud Bekkers
- Department of Obstetrics and Gynecology, Catharina Ziekenhuis, Eindhoven, the Netherlands; GROW school for oncology and developmental biology, Maastricht University Medical Centre, the Netherlands
| | - Nelleke Ottevanger
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Joop H Jansen
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Leon Massuger
- Department of Obstetrics and Gynecology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Harry Dolstra
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
32
|
Curran M, Mairesse M, Matas-Céspedes A, Bareham B, Pellegrini G, Liaunardy A, Powell E, Sargeant R, Cuomo E, Stebbings R, Betts CJ, Saeb-Parsy K. Recent Advancements and Applications of Human Immune System Mice in Preclinical Immuno-Oncology. Toxicol Pathol 2019; 48:302-316. [PMID: 31847725 DOI: 10.1177/0192623319886304] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Significant advances in immunotherapies have resulted in the increasing need of predictive preclinical models to improve immunotherapeutic drug development, treatment combination, and to prevent or minimize toxicity in clinical trials. Immunodeficient mice reconstituted with human immune system (HIS), termed humanized mice or HIS mice, permit detailed analysis of human immune biology, development, and function. Although this model constitutes a great translational model, some aspects need to be improved as the incomplete engraftment of immune cells, graft versus host disease and the lack of human cytokines and growth factors. In this review, we discuss current HIS platforms, their pathology, and recent advances in their development to improve the quality of human immune cell reconstitution. We also highlight new technologies that can be used to better understand these models and how improved characterization is needed for their application in immuno-oncology safety, efficacy, and new modalities therapy development.
Collapse
Affiliation(s)
- Michelle Curran
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom.,Department of Surgery, University of Cambridge and NIHR Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Maelle Mairesse
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Alba Matas-Céspedes
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom.,Department of Surgery, University of Cambridge and NIHR Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Bethany Bareham
- Department of Surgery, University of Cambridge and NIHR Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Giovanni Pellegrini
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Ardi Liaunardy
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Edward Powell
- Department of Surgery, University of Cambridge and NIHR Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Rebecca Sargeant
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Emanuela Cuomo
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Richard Stebbings
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Catherine J Betts
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Kourosh Saeb-Parsy
- Department of Surgery, University of Cambridge and NIHR Cambridge Biomedical Campus, Cambridge, United Kingdom
| |
Collapse
|
33
|
Herrera L, Santos S, Vesga MA, Anguita J, Martin-Ruiz I, Carrascosa T, Juan M, Eguizabal C. Adult peripheral blood and umbilical cord blood NK cells are good sources for effective CAR therapy against CD19 positive leukemic cells. Sci Rep 2019; 9:18729. [PMID: 31822751 PMCID: PMC6904575 DOI: 10.1038/s41598-019-55239-y] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 11/25/2019] [Indexed: 12/13/2022] Open
Abstract
Among hematological cancers, Acute Lymphoblastic Leukemia (ALL) and Chronic Lymphocytic Leukemia (CLL) are the most common leukemia in children and elderly people respectively. Some patients do not respond to chemotherapy treatments and it is necessary to complement it with immunotherapy-based treatments such as chimeric antigen receptor (CAR) therapy, which is one of the newest and more effective treatments against these cancers and B-cell lymphoma. Although complete remission results are promising, CAR T cell therapy presents still some risks for the patients, including cytokine release syndrome (CRS) and neurotoxicity. We proposed a different immune cell source for CAR therapy that might prevent these side effects while efficiently targeting malignant cells. NK cells from different sources are a promising vehicle for CAR therapy, as they do not cause graft versus host disease (GvHD) in allogenic therapies and they are prompt to attack cancer cells without prior sensitization. We studied the efficacy of NK cells from adult peripheral blood (AB) and umbilical cord blood (CB) against different target cells in order to determine the best source for CAR therapy. AB CAR-NK cells are slightly better at killing CD19 presenting target cells and CB NK cells are easier to stimulate and they have more stable number from donor to donor. We conclude that CAR-NK cells from both sources have their advantages to be an alternative and safer candidate for CAR therapy.
Collapse
Affiliation(s)
- L Herrera
- Cell Therapy, Stem Cells and Tissues Group, Basque Centre for Blood Transfusion and Human Tissues, Galdakao, Spain.,Biocruces Bizkaia Health Research Institute, Barkaldo, Spain
| | - S Santos
- Cell Therapy, Stem Cells and Tissues Group, Basque Centre for Blood Transfusion and Human Tissues, Galdakao, Spain.,Biocruces Bizkaia Health Research Institute, Barkaldo, Spain
| | - M A Vesga
- Cell Therapy, Stem Cells and Tissues Group, Basque Centre for Blood Transfusion and Human Tissues, Galdakao, Spain.,Biocruces Bizkaia Health Research Institute, Barkaldo, Spain
| | - J Anguita
- Macrophage and Tick Vaccine Laboratory, CIC bioGUNE, Derio, Biscay, Spain.,Ikerbasque, Basque Foundation for Science, Bilbao, Biscay, Spain
| | - I Martin-Ruiz
- Macrophage and Tick Vaccine Laboratory, CIC bioGUNE, Derio, Biscay, Spain
| | - T Carrascosa
- Servicio de Hematología, Hospital Galdakao-Usansolo, Galdakao, Spain.,Biocruces Bizkaia Health Research Institute, Barkaldo, Spain
| | - M Juan
- Servei d´Immunologia, Hospital Clínic de Barcelona, Hospital Sant Joan de Déu, Institut d'Investigacions Biomèdiques August Pi i Sunyer Hospital, Universitat de Barcelona, Barcelona, Spain
| | - C Eguizabal
- Cell Therapy, Stem Cells and Tissues Group, Basque Centre for Blood Transfusion and Human Tissues, Galdakao, Spain. .,Biocruces Bizkaia Health Research Institute, Barkaldo, Spain.
| |
Collapse
|
34
|
Alisjahbana A, Mohammad I, Gao Y, Evren E, Ringqvist E, Willinger T. Human macrophages and innate lymphoid cells: Tissue-resident innate immunity in humanized mice. Biochem Pharmacol 2019; 174:113672. [PMID: 31634458 DOI: 10.1016/j.bcp.2019.113672] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 10/15/2019] [Indexed: 12/17/2022]
Abstract
Macrophages and innate lymphoid cells (ILCs) are tissue-resident cells that play important roles in organ homeostasis and tissue immunity. Their intricate relationship with the organs they reside in allows them to quickly respond to perturbations of organ homeostasis and environmental challenges, such as infection and tissue injury. Macrophages and ILCs have been extensively studied in mice, yet important species-specific differences exist regarding innate immunity between humans and mice. Complementary to ex-vivo studies with human cells, humanized mice (i.e. mice with a human immune system) offer the opportunity to study human macrophages and ILCs in vivo within their surrounding tissue microenvironments. In this review, we will discuss how humanized mice have helped gain new knowledge about the basic biology of these cells, as well as their function in infectious and malignant conditions. Furthermore, we will highlight active areas of investigation related to human macrophages and ILCs, such as their cellular heterogeneity, ontogeny, tissue residency, and plasticity. In the near future, we expect more fundamental discoveries in these areas through the combined use of improved humanized mouse models together with state-of-the-art technologies, such as single-cell RNA-sequencing and CRISPR/Cas9 genome editing.
Collapse
Affiliation(s)
- Arlisa Alisjahbana
- Center for Infectious Medicine, Karolinska Institutet, Alfred Nobels allé 8, 141 52 Stockholm, Sweden
| | - Imran Mohammad
- Center for Infectious Medicine, Karolinska Institutet, Alfred Nobels allé 8, 141 52 Stockholm, Sweden
| | - Yu Gao
- Center for Infectious Medicine, Karolinska Institutet, Alfred Nobels allé 8, 141 52 Stockholm, Sweden
| | - Elza Evren
- Center for Infectious Medicine, Karolinska Institutet, Alfred Nobels allé 8, 141 52 Stockholm, Sweden
| | - Emma Ringqvist
- Center for Infectious Medicine, Karolinska Institutet, Alfred Nobels allé 8, 141 52 Stockholm, Sweden
| | - Tim Willinger
- Center for Infectious Medicine, Karolinska Institutet, Alfred Nobels allé 8, 141 52 Stockholm, Sweden.
| |
Collapse
|
35
|
Peptide-mediated delivery of therapeutic mRNA in ovarian cancer. Eur J Pharm Biopharm 2019; 141:180-190. [PMID: 31103743 DOI: 10.1016/j.ejpb.2019.05.014] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 05/13/2019] [Accepted: 05/15/2019] [Indexed: 12/27/2022]
Abstract
Ovarian cancer is the most lethal gynecological malignancy in the developed world. In spite of intensive research, the mortality has hardly decreased over the past twenty years. This necessitates the exploration of novel therapeutic modalities. Transient protein expression through delivery of mRNA is emerging as a highly promising option. In contrast to gene therapy there is no risk of integration into the genome. Here, we explore the expression of mRNA in models of ovarian cancer of increasing complexity. The cell-penetrating peptide (CPP) PepFect 14 (PF14) was used to formulate CPP-mRNA nanoparticles. Efficient expression of a reporter protein was achieved in two-dimensional tissue cultures and in three-dimensional cancer cell spheroids. PF14 nanoparticles greatly outperformed a lipid-based transfection agent in vivo, leading to expression in various cell types of tumor associated tissue. Protein expression was restricted to the peritoneal cavity. Messenger RNA expression across different cell types was confirmed in primary ovarian cancer explants. As ovarian cancer is confined to the peritoneal cavity in most cases, the results create the basis for applications in which the tumor microenvironment is transiently modified through protein expression.
Collapse
|
36
|
Patel S, Burga RA, Powell AB, Chorvinsky EA, Hoq N, McCormack SE, Van Pelt SN, Hanley PJ, Cruz CRY. Beyond CAR T Cells: Other Cell-Based Immunotherapeutic Strategies Against Cancer. Front Oncol 2019; 9:196. [PMID: 31024832 PMCID: PMC6467966 DOI: 10.3389/fonc.2019.00196] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 03/07/2019] [Indexed: 12/13/2022] Open
Abstract
Background: Chimeric antigen receptor (CAR)-modified T cells have successfully harnessed T cell immunity against malignancies, but they are by no means the only cell therapies in development for cancer. Main Text Summary: Systemic immunity is thought to play a key role in combatting neoplastic disease; in this vein, genetic modifications meant to explore other components of T cell immunity are being evaluated. In addition, other immune cells—from both the innate and adaptive compartments—are in various stages of clinical application. In this review, we focus on these non-CAR T cell immunotherapeutic approaches for malignancy. The first section describes engineering T cells to express non-CAR constructs, and the second section describes other gene-modified cells used to target malignancy. Conclusions: CAR T cell therapies have demonstrated the clinical benefits of harnessing our body's own defenses to combat tumor cells. Similar research is being conducted on lesser known modifications and gene-modified immune cells, which we highlight in this review.
Collapse
Affiliation(s)
- Shabnum Patel
- GW Cancer Center, The George Washington University, Washington, DC, United States
| | - Rachel A Burga
- GW Cancer Center, The George Washington University, Washington, DC, United States
| | - Allison B Powell
- GW Cancer Center, The George Washington University, Washington, DC, United States
| | - Elizabeth A Chorvinsky
- Center for Cancer and Immunology Research, Children's National Health System, Washington, DC, United States
| | - Nia Hoq
- GW Cancer Center, The George Washington University, Washington, DC, United States
| | - Sarah E McCormack
- GW Cancer Center, The George Washington University, Washington, DC, United States
| | - Stacey N Van Pelt
- GW Cancer Center, The George Washington University, Washington, DC, United States
| | - Patrick J Hanley
- Center for Cancer and Immunology Research, Children's National Health System, Washington, DC, United States
| | - Conrad Russell Y Cruz
- GW Cancer Center, The George Washington University, Washington, DC, United States.,Center for Cancer and Immunology Research, Children's National Health System, Washington, DC, United States
| |
Collapse
|
37
|
Courau T, Bonnereau J, Chicoteau J, Bottois H, Remark R, Assante Miranda L, Toubert A, Blery M, Aparicio T, Allez M, Le Bourhis L. Cocultures of human colorectal tumor spheroids with immune cells reveal the therapeutic potential of MICA/B and NKG2A targeting for cancer treatment. J Immunother Cancer 2019; 7:74. [PMID: 30871626 PMCID: PMC6417026 DOI: 10.1186/s40425-019-0553-9] [Citation(s) in RCA: 161] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 02/28/2019] [Indexed: 12/11/2022] Open
Abstract
Background Immunotherapies still fail to benefit colorectal cancer (CRC) patients. Relevant functional assays aimed at studying these failures and the efficacy of cancer immunotherapy in human are scarce. 3D tumor cultures, called tumor organoids or spheroids, represent interesting models to study cancer treatments and could help to challenge these issues. Methods We analyzed heterotypic cocultures of human colon tumor-derived spheroids with immune cells to assess the infiltration, activation and function of T and NK cells toward human colorectal tumors in vitro. Results We showed that allogeneic T and NK cells rapidly infiltrated cell line-derived spheroids, inducing immune-mediated tumor cell apoptosis and spheroid destruction. NKG2D, a key activator of cytotoxic responses, was engaged on infiltrating cells. We thus assessed the therapeutic potential of an antibody targeting the specific ligands of NKG2D, MICA and MICB, in this system. Anti-MICA/B enhanced immune-dependent destruction of tumor spheroid by driving an increased NK cells infiltration and activation. Interestingly, tumor cells reacted to immune infiltration by upregulating HLA-E, ligand of the inhibitory receptor NKG2A expressed by CD8 and NK cells. NKG2A was increased after anti-MICA/B treatment and, accordingly, combination of anti-MICA/B and anti-NKG2A was synergistic. These observations were ultimately confirmed in a clinical relevant model of coculture between CRC patients-derived spheroids and autologous tumor-infiltrating lymphocytes. Conclusions Altogether, we show that tumor spheroids represent a relevant tool to study tumor-lymphocyte interactions on human tissues and revealed the antitumor potential of immunomodulatory antibodies targeting MICA/B and NKG2A. Electronic supplementary material The online version of this article (10.1186/s40425-019-0553-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tristan Courau
- INSERM U1160, Institut de Recherche Saint-Louis, Saint Louis Hospital, Paris, France
| | - Julie Bonnereau
- INSERM U1160, Institut de Recherche Saint-Louis, Saint Louis Hospital, Paris, France.,Paris-Diderot University, Sorbonne Paris Cité, Paris, France
| | - Justine Chicoteau
- INSERM U1160, Institut de Recherche Saint-Louis, Saint Louis Hospital, Paris, France.,Gastroenterology and Digestive Oncology Department, Saint Louis Hospital, AP-HP, Paris, France
| | - Hugo Bottois
- INSERM U1160, Institut de Recherche Saint-Louis, Saint Louis Hospital, Paris, France.,Paris-Diderot University, Sorbonne Paris Cité, Paris, France
| | | | | | - Antoine Toubert
- INSERM U1160, Institut de Recherche Saint-Louis, Saint Louis Hospital, Paris, France.,Paris-Diderot University, Sorbonne Paris Cité, Paris, France
| | | | - Thomas Aparicio
- INSERM U1160, Institut de Recherche Saint-Louis, Saint Louis Hospital, Paris, France.,Paris-Diderot University, Sorbonne Paris Cité, Paris, France.,Gastroenterology and Digestive Oncology Department, Saint Louis Hospital, AP-HP, Paris, France
| | - Matthieu Allez
- INSERM U1160, Institut de Recherche Saint-Louis, Saint Louis Hospital, Paris, France.,Paris-Diderot University, Sorbonne Paris Cité, Paris, France.,Gastroenterology and Digestive Oncology Department, Saint Louis Hospital, AP-HP, Paris, France
| | - Lionel Le Bourhis
- INSERM U1160, Institut de Recherche Saint-Louis, Saint Louis Hospital, Paris, France.
| |
Collapse
|
38
|
Nayyar G, Chu Y, Cairo MS. Overcoming Resistance to Natural Killer Cell Based Immunotherapies for Solid Tumors. Front Oncol 2019; 9:51. [PMID: 30805309 PMCID: PMC6378304 DOI: 10.3389/fonc.2019.00051] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 01/18/2019] [Indexed: 12/22/2022] Open
Abstract
Despite advances in the diagnostic and therapeutic modalities, the prognosis of several solid tumor malignancies remains poor. Different factors associated with solid tumors including a varied genetic signature, complex molecular signaling pathways, defective cross talk between the tumor cells and immune cells, hypoxic and immunosuppressive effects of tumor microenvironment result in a treatment resistant and metastatic phenotype. Over the past several years, immunotherapy has emerged as an attractive therapeutic option against multiple malignancies. The unique ability of natural killer (NK) cells to target cancer cells without antigen specificity makes them an ideal candidate for use against solid tumors. However, the outcomes of adoptive NK cell infusions into patients with solid tumors have been disappointing. Extensive studies have been done to investigate different strategies to improve the NK cell function, trafficking and tumor targeting. Use of cytokines and cytokine analogs has been well described and utilized to enhance the proliferation, stimulation and persistence of NK cells. Other techniques like blocking the human leukocyte antigen-killer cell receptors (KIR) interactions with anti-KIR monoclonal antibodies, preventing CD16 receptor shedding, increasing the expression of activating NK cell receptors like NKG2D, and use of immunocytokines and immune checkpoint inhibitors can enhance NK cell mediated cytotoxicity. Using genetically modified NK cells with chimeric antigen receptors and bispecific and trispecific NK cell engagers, NK cells can be effectively redirected to the tumor cells improving their cytotoxic potential. In this review, we have described these strategies and highlighted the need to further optimize these strategies to improve the clinical outcome of NK cell based immunotherapy against solid tumors.
Collapse
Affiliation(s)
- Gaurav Nayyar
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States
| | - Yaya Chu
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States
| | - Mitchell S Cairo
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States.,Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY, United States.,Department of Microbiology & Immunology, New York Medical College, Valhalla, NY, United States.,Department of Medicine, New York Medical College, Valhalla, NY, United States.,Department of Pathology, New York Medical College, Valhalla, NY, United States
| |
Collapse
|
39
|
Hoogstad-van Evert J, Bekkers R, Ottevanger N, Schaap N, Hobo W, Jansen JH, Massuger L, Dolstra H. Intraperitoneal infusion of ex vivo-cultured allogeneic NK cells in recurrent ovarian carcinoma patients (a phase I study). Medicine (Baltimore) 2019; 98:e14290. [PMID: 30702598 PMCID: PMC6380776 DOI: 10.1097/md.0000000000014290] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
INTRODUCTION Recurrent ovarian carcinoma has dismal prognosis, but control of disease and prolonged survival are possible in some patients. The estimated 5-year survival is 46% for all stages of ovarian cancer, and only 28% for metastasized disease. Notably, the majority of women with ovarian cancer are diagnosed with stage III or IV disease with a high recurrence rate. As most women with relapsed or metastatic cancer will die of progressive disease, there is an urgent need for novel therapeutic strategies. The primary aim of our study is to evaluate safety and toxicity of intraperitoneal infusion of ex vivo-expanded natural killer cells (NK), generated from CD34+ umbilical cord blood (UCB) progenitor cells, with and without a preceding non-myeloablative immunosuppressive conditioning regimen in patients suffering from recurrent ovarian cancer. The secondary objectives are to compare the in vivo lifespan, expansion, and biological activity of intraperitoneally infused NK cell products with or without preparative chemotherapy, as well as evaluate effects on disease load. METHODS In this phase I safety trial, 12 patients who are suffering from recurrent ovarian cancer, detected by a significant rise in serum level of CA-125 on two successive time points, will be included. Prior to UCB-NK cell infusion, a laparoscopy is performed to place a catheter in the peritoneal cavity. The first cohort of three patients will receive a single intraperitoneal infusion of 1.5-3×10 UCB-NK cells, generated ex vivo from CD34+ hematopoietic progenitor cells obtained from an allogeneic UCB unit, without a preparative chemotherapy regimen. The second group of three patients will be treated with a similar dose of UCB-NK cells following a preparative four days non-myeloablative immunosuppressive conditioning regimen with cyclophosphamide and fludarabine (Cy/Flu). If no severe toxicity is seen in these 6 patients, an extension cohort of 6 patients will be included to answer the secondary objectives. DISCUSSION This study investigates the safety of a promising new cellular therapy in a group of patients with a poor prognosis. Demonstration of safety and in vivo expansion capacity of allogeneic UCB-NK cells in the absence of Cy/Flu pretreatment will provide rationale for UCB-NK cell infusion after regular second-line chemotherapy.
Collapse
Affiliation(s)
| | | | | | | | - Willemijn Hobo
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud Institute of Molecular Life Sciences, Nijmegen, The Netherlands
| | - Joop H. Jansen
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud Institute of Molecular Life Sciences, Nijmegen, The Netherlands
| | | | - Harry Dolstra
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud Institute of Molecular Life Sciences, Nijmegen, The Netherlands
| |
Collapse
|
40
|
Ayuso JM, Truttschel R, Gong MM, Humayun M, Virumbrales-Munoz M, Vitek R, Felder M, Gillies SD, Sondel P, Wisinski KB, Patankar M, Beebe DJ, Skala MC. Evaluating natural killer cell cytotoxicity against solid tumors using a microfluidic model. Oncoimmunology 2018; 8:1553477. [PMID: 30723584 DOI: 10.1080/2162402x.2018.1553477] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 11/06/2018] [Accepted: 11/14/2018] [Indexed: 12/12/2022] Open
Abstract
Immunotherapies against solid tumors face additional challenges compared with hematological cancers. In solid tumors, immune cells and antibodies need to extravasate from vasculature, find the tumor, and migrate through a dense mass of cells. These multiple steps pose significant obstacles for solid tumor immunotherapy and their study has remained difficult using classic in vitro models based on Petri dishes. In this work, a microfluidic model has been developed to study natural killer cell response. The model includes a 3D breast cancer spheroid in a 3D extracellular matrix, and two flanking lumens lined with endothelial cells, replicating key structures and components during the immune response. Natural Killer cells and antibodies targeting the tumor cells were either embedded in the matrix or perfused through the lateral blood vessels. Antibodies that were perfused through the lateral lumens extravasated out of the blood vessels and rapidly diffused through the matrix. However, tumor cell-cell junctions hindered antibody penetration within the spheroid. On the other hand, natural killer cells were able to detect the presence of the tumor spheroid several hundreds of microns away and penetrate the spheroid faster than the antibodies. Once inside the spheroid, natural killer cells were able to destroy tumor cells at the spheroid periphery and, importantly, also at the innermost layers. Finally, the combination of antibody-cytokine conjugates and natural killer cells led to an enhanced cytotoxicity located mostly at the spheroid periphery. Overall, these results demonstrate the utility of the model for informing immunotherapy of solid tumors.
Collapse
Affiliation(s)
- Jose M Ayuso
- Morgridge Institute for Research, Madison, WI, USA.,Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA.,The University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| | - Regan Truttschel
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
| | - Max M Gong
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA.,The University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| | - Mouhita Humayun
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA.,The University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| | - Maria Virumbrales-Munoz
- Morgridge Institute for Research, Madison, WI, USA.,Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA.,The University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI, USA.,Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, USA.,Provenance Biopharmaceuticals Corp., Carlisle, MA USA.,Department of Pathology & Laboratory Medicine, University of Wisconsin, Madison, WI, USA
| | - Ross Vitek
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA.,The University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| | - Mildred Felder
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, USA
| | | | - Paul Sondel
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, USA
| | - Kari B Wisinski
- The University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| | - Manish Patankar
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, USA
| | - David J Beebe
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA.,The University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI, USA.,Department of Pathology & Laboratory Medicine, University of Wisconsin, Madison, WI, USA
| | - Melissa C Skala
- Morgridge Institute for Research, Madison, WI, USA.,Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA.,The University of Wisconsin Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
41
|
Humanized Mice for the Study of Immuno-Oncology. Trends Immunol 2018; 39:748-763. [DOI: 10.1016/j.it.2018.07.001] [Citation(s) in RCA: 137] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 07/05/2018] [Accepted: 07/05/2018] [Indexed: 01/28/2023]
|
42
|
Wefers C, Schreibelt G, Massuger LFAG, de Vries IJM, Torensma R. Immune Curbing of Cancer Stem Cells by CTLs Directed to NANOG. Front Immunol 2018; 9:1412. [PMID: 29971070 PMCID: PMC6018198 DOI: 10.3389/fimmu.2018.01412] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 06/06/2018] [Indexed: 12/15/2022] Open
Abstract
Cancer stem cells (CSCs) have been identified as the source of tumor growth and disease recurrence. Eradication of CSCs is thus essential to achieve durable responses, but CSCs are resistant to current anti-tumor therapies. Novel therapeutic approaches that specifically target CSCs will, therefore, be crucial to improve patient outcome. Immunotherapies, which boost the body's own immune system to eliminate cancerous cells, could be an alternative approach to target CSCs. Vaccines of dendritic cells (DCs) loaded with tumor antigens can evoke highly specific anti-tumor T cell responses. Importantly, DC vaccination also promotes immunological memory formation, paving the way for long-term cancer control. Here, we propose a DC vaccination that specifically targets CSCs. DCs loaded with NANOG peptides, a protein required for maintaining stem cell properties, could evoke a potent anti-tumor immune response against CSCs. We hypothesize that the resulting immunological memory will also control newly formed CSCs, thereby preventing disease recurrence.
Collapse
Affiliation(s)
- Christina Wefers
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, Netherlands
- Department of Obstetrics and Gynecology, Radboudumc, Nijmegen, Netherlands
| | - Gerty Schreibelt
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, Netherlands
| | | | - I. Jolanda M. de Vries
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, Netherlands
| | - Ruurd Torensma
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, Netherlands
| |
Collapse
|
43
|
Sherman H, Gitschier HJ, Rossi AE. A Novel Three-Dimensional Immune Oncology Model for High-Throughput Testing of Tumoricidal Activity. Front Immunol 2018; 9:857. [PMID: 29740450 PMCID: PMC5924962 DOI: 10.3389/fimmu.2018.00857] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 04/06/2018] [Indexed: 12/22/2022] Open
Abstract
The latest advancements in oncology research are focused on autologous immune cell therapy. However, the effectiveness of this type of immunotherapy for cancer remediation is not equivalent for all patients or cancer types. This suggests the need for better preclinical screening models that more closely recapitulate in vivo tumor biology. The established method for investigating tumoricidal activity of immunotherapies has been study of two-dimensional (2D) monolayer cultures of immortalized cancer cell lines or primary tumor cells in standard tissue culture vessels. Indeed, a proven means to examine immune cell migration and invasion are 2D chemotaxis assays in permeabilized supports or Boyden chambers. Nevertheless, the more in vivo-like three-dimensional (3D) multicellular tumor spheroids are quickly becoming the favored model to examine immune cell invasion and tumor cell cytotoxicity. Accordingly, we have developed a 3D immune oncology model by combining 96-well permeable support systems and 96-well low-attachment microplates. The use of the permeable support system enables assessment of immune cell migration, which was tested in this study as chemotactic response of natural killer NK-92MI cells to human stromal-cell derived factor-1 (SDF-1α). Immune invasion was assessed by measuring NK-92MI infiltration into lung carcinoma A549 cell spheroids that were formed in low-attachment microplates. The novel pairing of the permeable support system with low-attachment microplates permitted simultaneous investigation of immune cell homing, immune invasion of tumor spheroids, and spheroid cytotoxicity. In effect, the system represents a more comprehensive and in vivo-like immune oncology model that can be utilized for high-throughput study of tumoricidal activity.
Collapse
Affiliation(s)
- Hilary Sherman
- Life Sciences Division, Corning Incorporated, Kennebunk, ME, United States
| | - Hannah J Gitschier
- Life Sciences Division, Corning Incorporated, Kennebunk, ME, United States
| | - Ann E Rossi
- Life Sciences Division, Corning Incorporated, Kennebunk, ME, United States
| |
Collapse
|
44
|
Penetration in 3D tumor spheroids and explants: Adding a further dimension to the structure-activity relationship of cell-penetrating peptides. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2018; 1860:1342-1349. [PMID: 29550289 DOI: 10.1016/j.bbamem.2018.03.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 02/23/2018] [Accepted: 03/08/2018] [Indexed: 12/30/2022]
Abstract
Drug delivery into tumors and metastases is a major challenge in the eradication of cancers such as epithelial ovarian carcinoma. Cationic cell-penetrating peptides (CPPs) are a promising group of delivery vehicles to mediate cellular entry of molecules that otherwise poorly enter cells. However, little is known about their penetration behavior in tissues. Here, we investigated penetration of cationic CPPs in 3D ovarian cancer spheroids and patient-derived 3D tumor explants. Penetration kinetics and distribution after long-term incubation were imaged by confocal microscopy. In addition, spheroids and tumor explants were dissociated and cell-associated fluorescence determined by flow cytometry. CPPs with high uptake activity showed enhanced sequestration in the periphery of the spheroid, whereas less active CPPs were able to penetrate deeper into the tissue. CPPs consisting of d-amino acids were advantageous over l-amino acid CPPs as they showed less but long lasting cellular uptake activity, which benefitted penetration and retention over time. In primary tumor cultures, in contrast to nonaarginine, the amphipathic CPP penetratin was strongly sequestered by cell debris and matrix components pointing towards arginine-rich CPPs as a preferred choice. Overall, the data show that testing in 3D models leads to a different choice of the preferred peptide in comparison to a standard 2D cell culture.
Collapse
|
45
|
Uppendahl LD, Dahl CM, Miller JS, Felices M, Geller MA. Natural Killer Cell-Based Immunotherapy in Gynecologic Malignancy: A Review. Front Immunol 2018; 8:1825. [PMID: 29354116 PMCID: PMC5760535 DOI: 10.3389/fimmu.2017.01825] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 12/04/2017] [Indexed: 12/20/2022] Open
Abstract
Harnessing the immune system has proven an effective therapy in treating malignancies. Since the discovery of natural killer (NK) cells, strategies aimed to manipulate and augment their effector function against cancer have been the subject of intense research. Recent progress in the immunobiology of NK cells has led to the development of promising therapeutic approaches. In this review, we will focus on the recent advances in NK cell immunobiology and the clinical application of NK cell immunotherapy in ovarian, cervical, and uterine cancer.
Collapse
Affiliation(s)
- Locke D Uppendahl
- Department of Obstetrics, Gynecology and Women's Health, Division of Gynecologic Oncology, University of Minnesota School of Medicine, Minneapolis, MN, United States
| | - Carly M Dahl
- University of Minnesota School of Medicine, Minneapolis, MN, United States
| | - Jeffrey S Miller
- Department of Medicine, Division of Hematology, Oncology, and Transplantation, University of Minnesota School of Medicine, Minneapolis, MN, United States
| | - Martin Felices
- Department of Medicine, Division of Hematology, Oncology, and Transplantation, University of Minnesota School of Medicine, Minneapolis, MN, United States
| | - Melissa A Geller
- Department of Obstetrics, Gynecology and Women's Health, Division of Gynecologic Oncology, University of Minnesota School of Medicine, Minneapolis, MN, United States
| |
Collapse
|
46
|
Leveraging Epigenetics to Enhance the Cellular Response to Chemotherapies and Improve Tumor Immunogenicity. Adv Cancer Res 2018; 138:1-39. [PMID: 29551125 DOI: 10.1016/bs.acr.2018.02.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cancer chemotherapeutic drugs have greatly advanced our ability to successfully treat a variety of human malignancies. The different forms of stress produced by these agents in cancer cells result in both cell autonomous and cell nonautonomous effects. Desirable cell autonomous effects include reduced proliferative potential, cellular senescence, and cell death. More recently recognized cell nonautonomous effects, usually in the form of stimulating an antitumor immune response, have significant roles in therapeutic efficiency for a select number of chemotherapies. Unfortunately, the success of these therapeutics is not universal as not all tumors respond to treatment, and those that do respond will frequently relapse into therapy-resistant disease. Numerous strategies have been developed to sensitize tumors toward chemotherapies as a means to either improve initial responses, or serve as a secondary treatment strategy for therapy-resistant disease. Recently, targeting epigenetic regulators has emerged as a viable method of sensitizing tumors to the effects of chemotherapies, many of which are cytotoxic. In this review, we summarize these strategies and propose a path for future progress.
Collapse
|
47
|
Decitabine enhances targeting of AML cells by CD34 + progenitor-derived NK cells in NOD/SCID/IL2Rg null mice. Blood 2017; 131:202-214. [PMID: 29138222 DOI: 10.1182/blood-2017-06-790204] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 11/06/2017] [Indexed: 12/29/2022] Open
Abstract
Combining natural killer (NK) cell adoptive transfer with hypomethylating agents (HMAs) is an attractive therapeutic approach for patients with acute myeloid leukemia (AML). However, data regarding the impact of HMAs on NK cell functionality are mostly derived from in vitro studies with high nonclinical relevant drug concentrations. In the present study, we report a comparative study of azacitidine (AZA) and decitabine (DAC) in combination with allogeneic NK cells generated from CD34+ hematopoietic stem and progenitor cells (HSPC-NK cells) in in vitro and in vivo AML models. In vitro, low-dose HMAs did not impair viability of HSPC-NK cells. Furthermore, low-dose DAC preserved HSPC-NK killing, proliferation, and interferon gamma production capacity, whereas AZA diminished their proliferation and reactivity. Importantly, we showed HMAs and HSPC-NK cells could potently work together to target AML cell lines and patient AML blasts. In vivo, both agents exerted a significant delay in AML progression in NOD/SCID/IL2Rgnull mice, but the persistence of adoptively transferred HSPC-NK cells was not affected. Infused NK cells showed sustained expression of most activating receptors, upregulated NKp44 expression, and remarkable killer cell immunoglobulin-like receptor acquisition. Most importantly, only DAC potentiated HSPC-NK cell anti-leukemic activity in vivo. Besides upregulation of NKG2D- and DNAM-1-activating ligands on AML cells, DAC enhanced messenger RNA expression of inflammatory cytokines, perforin, and TRAIL by HSPC-NK cells. In addition, treatment resulted in increased numbers of HSPC-NK cells in the bone marrow compartment, suggesting that DAC could positively modulate NK cell activity, trafficking, and tumor targeting. These data provide a rationale to explore combination therapy of adoptive HSPC-NK cells and DAC in patients with AML.
Collapse
|