1
|
Duléry R, Piccinelli S, Beg MS, Jang JE, Romee R. Haploidentical hematopoietic cell transplantation as a platform for natural killer cell immunotherapy. Am J Hematol 2024; 99:2340-2350. [PMID: 39248561 DOI: 10.1002/ajh.27471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 07/19/2024] [Accepted: 08/13/2024] [Indexed: 09/10/2024]
Abstract
An innovative approach is crucially needed to manage relapse after allogeneic hematopoietic cell transplantation (HCT) in patients with advanced hematological malignancies. This review explores key aspects of haploidentical HCT with post-transplant cyclophosphamide, highlighting the potential and suitability of this platform for natural killer (NK) cell immunotherapy. NK cells, known for their unique abilities to eliminate cancer cells, can also exhibit memory-like features and enhanced cytotoxicity when activated by cytokines. By discussing promising results from clinical trials, the review delves into the recent major advances: donor-derived NK cells can be expanded ex vivo in large numbers, cytokine activation may enhance NK cell persistence and efficacy in vivo, and post-HCT NK cell infusion can improve outcomes in high-risk and/or relapsed myeloid malignancies without increasing the risk of graft-versus-host disease, severe cytokine release syndrome, or neurotoxicity. Looking ahead, cytokine-activated NK cells can be synergized with immunomodulatory agents and/or genetically engineered to enhance their tumor-targeting specificity, cytotoxicity, and persistence while preventing exhaustion. The ongoing exploration of these strategies holds promising preliminary results and could be rapidly translated into clinical applications for the benefit of the patients.
Collapse
Affiliation(s)
- Rémy Duléry
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Sara Piccinelli
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Ji Eun Jang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
- Department of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Rizwan Romee
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
2
|
French AR, Cron RQ, Cooper MA. Immunology of Cytokine Storm Syndromes: Natural Killer Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1448:145-159. [PMID: 39117813 DOI: 10.1007/978-3-031-59815-9_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
Natural killer (NK) cells are innate immune lymphocytes that rapidly produce cytokines upon activation and kill target cells. NK cells have been of particular interest in primary hemophagocytic lymphohistiocytosis (pHLH) since all of the genetic defects associated with this disorder cause diminished cytotoxic capacity of NK cells and T lymphocytes, and assays of NK cell killing are used clinically for the diagnosis of HLH. Herein, we review human NK cell biology and the significance of alterations in NK cell function in the diagnosis and pathogenesis of HLH.
Collapse
Affiliation(s)
- Anthony R French
- Department of Pediatrics, Division of Rheumatology, Washington University School of Medicine, St. Louis, MO, USA
| | - Randy Q Cron
- Department of Pediatrics, Division of Rheumatology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, USA
| | - Megan A Cooper
- Department of Pediatrics, Division of Rheumatology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
3
|
Elanany MM, Mostafa D, Hamdy NM. Remodeled tumor immune microenvironment (TIME) parade via natural killer cells reprogramming in breast cancer. Life Sci 2023; 330:121997. [PMID: 37536617 DOI: 10.1016/j.lfs.2023.121997] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/20/2023] [Accepted: 07/31/2023] [Indexed: 08/05/2023]
Abstract
Breast cancer (BC) is the main cause of cancer-related mortality among women globally. Despite substantial advances in the identification and management of primary tumors, traditional therapies including surgery, chemotherapy, and radiation cannot completely eliminate the danger of relapse and metastatic illness. Metastasis is controlled by microenvironmental and systemic mechanisms, including immunosurveillance. This led to the evolvement of immunotherapies that has gained much attention in the recent years for cancer treatment directed to the innate immune system. The long forgotten innate immune cells known as natural killer (NK) cells have emerged as novel targets for more effective therapeutics for BC. Normally, NK cells has the capacity to identify and eradicate tumor cells either directly or by releasing cytotoxic granules, chemokines and proinflammatory cytokines. Yet, NK cells are exposed to inhibitory signals by cancer cells, which causes them to become dysfunctional in the immunosuppressive tumor microenvironment (TME) in BC, supporting tumor escape and spread. Potential mechanisms of NK cell dysfunction in BC metastasis have been recently identified. Understanding these immunologic pathways driving BC metastasis will lead to improvements in the current immunotherapeutic strategies. In the current review, we highlight how BC evades immunosurveillance by rendering NK cells dysfunctional and we shed the light on novel NK cell- directed therapies.
Collapse
Affiliation(s)
- Mona M Elanany
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Ain Shams University, Abassia, 11566 Cairo, Egypt
| | - Dina Mostafa
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Ain Shams University, Abassia, 11566 Cairo, Egypt.
| | - Nadia M Hamdy
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Ain Shams University, Abassia, 11566 Cairo, Egypt.
| |
Collapse
|
4
|
Qin H, You C, Yan F, Tan K, Xu C, Zhao R, Ekpo MD, Tan S. Overcoming the challenges in translational development of natural killer cell therapeutics: An opinion paper. Front Oncol 2022; 12:1062765. [DOI: 10.3389/fonc.2022.1062765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 11/22/2022] [Indexed: 12/04/2022] Open
|
5
|
Bednarski JJ, Zimmerman C, Berrien-Elliott MM, Foltz JA, Becker-Hapak M, Neal CC, Foster M, Schappe T, McClain E, Pence PP, Desai S, Kersting-Schadek S, Wong P, Russler-Germain DA, Fisk B, Lie WR, Eisele J, Hyde S, Bhatt ST, Griffith OL, Griffith M, Petti AA, Cashen AF, Fehniger TA. Donor memory-like NK cells persist and induce remissions in pediatric patients with relapsed AML after transplant. Blood 2022; 139:1670-1683. [PMID: 34871371 PMCID: PMC8931511 DOI: 10.1182/blood.2021013972] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 11/18/2021] [Indexed: 11/20/2022] Open
Abstract
Pediatric and young adult (YA) patients with acute myeloid leukemia (AML) who relapse after allogeneic hematopoietic cell transplantation (HCT) have an extremely poor prognosis. Standard salvage chemotherapy and donor lymphocyte infusions (DLIs) have little curative potential. Previous studies showed that natural killer (NK) cells can be stimulated ex vivo with interleukin-12 (IL-12), -15, and -18 to generate memory-like (ML) NK cells with enhanced antileukemia responses. We treated 9 pediatric/YA patients with post-HCT relapsed AML with donor ML NK cells in a phase 1 trial. Patients received fludarabine, cytarabine, and filgrastim followed 2 weeks later by infusion of donor lymphocytes and ML NK cells from the original HCT donor. ML NK cells were successfully generated from haploidentical and matched-related and -unrelated donors. After infusion, donor-derived ML NK cells expanded and maintained an ML multidimensional mass cytometry phenotype for >3 months. Furthermore, ML NK cells exhibited persistent functional responses as evidenced by leukemia-triggered interferon-γ production. After DLI and ML NK cell adoptive transfer, 4 of 8 evaluable patients achieved complete remission at day 28. Two patients maintained a durable remission for >3 months, with 1 patient in remission for >2 years. No significant toxicity was experienced. This study demonstrates that, in a compatible post-HCT immune environment, donor ML NK cells robustly expand and persist with potent antileukemic activity in the absence of exogenous cytokines. ML NK cells in combination with DLI present a novel immunotherapy platform for AML that has relapsed after allogeneic HCT. This trial was registered at https://clinicaltrials.gov as #NCT03068819.
Collapse
Affiliation(s)
| | - Clare Zimmerman
- Division of Hematology and Oncology, Department of Pediatrics, and
| | - Melissa M Berrien-Elliott
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Jennifer A Foltz
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Michelle Becker-Hapak
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Carly C Neal
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Mark Foster
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Timothy Schappe
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Ethan McClain
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Patrick P Pence
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Sweta Desai
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Samantha Kersting-Schadek
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Pamela Wong
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - David A Russler-Germain
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Bryan Fisk
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | | | - Jeremy Eisele
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Stephanie Hyde
- Division of Hematology and Oncology, Department of Pediatrics, and
| | - Sima T Bhatt
- Division of Hematology and Oncology, Department of Pediatrics, and
| | - Obi L Griffith
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Malachi Griffith
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Allegra A Petti
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, MO
| | - Amanda F Cashen
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Todd A Fehniger
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
6
|
Berrien-Elliott MM, Foltz JA, Russler-Germain DA, Neal CC, Tran J, Gang M, Wong P, Fisk B, Cubitt CC, Marin ND, Zhou AY, Jacobs MT, Foster M, Schappe T, McClain E, Kersting-Schadek S, Desai S, Pence P, Becker-Hapak M, Eisele J, Mosior M, Marsala L, Griffith OL, Griffith M, Khan SM, Spencer DH, DiPersio JF, Romee R, Uy GL, Abboud CN, Ghobadi A, Westervelt P, Stockerl-Goldstein K, Schroeder MA, Wan F, Lie WR, Soon-Shiong P, Petti AA, Cashen AF, Fehniger TA. Hematopoietic cell transplantation donor-derived memory-like NK cells functionally persist after transfer into patients with leukemia. Sci Transl Med 2022; 14:eabm1375. [PMID: 35196021 PMCID: PMC9210521 DOI: 10.1126/scitranslmed.abm1375] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Natural killer (NK) cells are innate lymphoid cells that eliminate cancer cells, produce cytokines, and are being investigated as a nascent cellular immunotherapy. Impaired NK cell function, expansion, and persistence remain key challenges for optimal clinical translation. One promising strategy to overcome these challenges is cytokine-induced memory-like (ML) differentiation, whereby NK cells acquire enhanced antitumor function after stimulation with interleukin-12 (IL-12), IL-15, and IL-18. Here, reduced-intensity conditioning (RIC) for HLA-haploidentical hematopoietic cell transplantation (HCT) was augmented with same-donor ML NK cells on day +7 and 3 weeks of N-803 (IL-15 superagonist) to treat patients with relapsed/refractory acute myeloid leukemia (AML) in a clinical trial (NCT02782546). In 15 patients, donor ML NK cells were well tolerated, and 87% of patients achieved a composite complete response at day +28, which corresponded with clearing high-risk mutations, including TP53 variants. NK cells were the major blood lymphocytes for 2 months after HCT with 1104-fold expansion (over 1 to 2 weeks). Phenotypic and transcriptional analyses identified donor ML NK cells as distinct from conventional NK cells and showed that ML NK cells persisted for over 2 months. ML NK cells expressed CD16, CD57, and high granzyme B and perforin, along with a unique transcription factor profile. ML NK cells differentiated in patients had enhanced ex vivo function compared to conventional NK cells from both patients and healthy donors. Overall, same-donor ML NK cell therapy with 3 weeks of N-803 support safely augmented RIC haplo-HCT for AML.
Collapse
Affiliation(s)
- Melissa M. Berrien-Elliott
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jennifer A. Foltz
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David A. Russler-Germain
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Carly C. Neal
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jennifer Tran
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Margery Gang
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Pamela Wong
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Bryan Fisk
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Celia C. Cubitt
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Nancy D. Marin
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Alice Y. Zhou
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Miriam T. Jacobs
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Mark Foster
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Timothy Schappe
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ethan McClain
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Samantha Kersting-Schadek
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sweta Desai
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Patrick Pence
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Michelle Becker-Hapak
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jeremy Eisele
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Matthew Mosior
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Lynne Marsala
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Obi L. Griffith
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Malachi Griffith
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Saad M. Khan
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David H. Spencer
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - John F. DiPersio
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Rizwan Romee
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Geoffrey L. Uy
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Camille N. Abboud
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Armin Ghobadi
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Peter Westervelt
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Keith Stockerl-Goldstein
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Mark A. Schroeder
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Fei Wan
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | - Allegra A. Petti
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Amanda F. Cashen
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Todd A. Fehniger
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
7
|
Lin HJ, Liu Y, Lofland D, Lin J. Breast Cancer Tumor Microenvironment and Molecular Aberrations Hijack Tumoricidal Immunity. Cancers (Basel) 2022; 14:cancers14020285. [PMID: 35053449 PMCID: PMC8774102 DOI: 10.3390/cancers14020285] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/27/2021] [Accepted: 12/28/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Immune therapy is designed to stimulate tumoricidal effects in a variety of solid tumors including breast carcinomas. However, the emergence of resistant clones leads to treatment failure. Understanding the molecular, cellular, and microenvironmental aberrations is crucial to uncovering underlying mechanisms and developing advanced strategies for preventing or combating these resistant malignancies. This review will summarize research findings revealing various mechanisms employed to hijack innate and adaptive immune surveillance mechanisms, develop hypoxic and tumor promoting metabolism, and foster an immune tolerance microenvironment. In addition, it will highlight potential targets for therapeutic approaches. Abstract Breast cancer is the most common malignancy among females in western countries, where women have an overall lifetime risk of >10% for developing invasive breast carcinomas. It is not a single disease but is composed of distinct subtypes associated with different clinical outcomes and is highly heterogeneous in both the molecular and clinical aspects. Although tumor initiation is largely driven by acquired genetic alterations, recent data suggest microenvironment-mediated immune evasion may play an important role in neoplastic progression. Beyond surgical resection, radiation, and chemotherapy, additional therapeutic options include hormonal deactivation, targeted-signaling pathway treatment, DNA repair inhibition, and aberrant epigenetic reversion. Yet, the fatality rate of metastatic breast cancer remains unacceptably high, largely due to treatment resistance and metastases to brain, lung, or bone marrow where tumor bed penetration of therapeutic agents is limited. Recent studies indicate the development of immune-oncological therapy could potentially eradicate this devastating malignancy. Evidence suggests tumors express immunogenic neoantigens but the immunity towards these antigens is frequently muted. Established tumors exhibit immunological tolerance. This tolerance reflects a process of immune suppression elicited by the tumor, and it represents a critical obstacle towards successful antitumor immunotherapy. In general, immune evasive mechanisms adapted by breast cancer encompasses down-regulation of antigen presentations or recognition, lack of immune effector cells, obstruction of anti-tumor immune cell maturation, accumulation of immunosuppressive cells, production of inhibitory cytokines, chemokines or ligands/receptors, and up-regulation of immune checkpoint modulators. Together with altered metabolism and hypoxic conditions, they constitute a permissive tumor microenvironment. This article intends to discern representative incidents and to provide potential innovative therapeutic regimens to reinstate tumoricidal immunity.
Collapse
Affiliation(s)
- Huey-Jen Lin
- Department of Medical & Molecular Sciences, University of Delaware, Willard Hall Education Building, 16 West Main Street, Newark, DE 19716, USA
- Correspondence: ; Tel.: +1-302-831-7576; Fax: +1-302-831-4180
| | - Yingguang Liu
- Department of Molecular and Cellular Sciences, College of Osteopathic Medicine, Liberty University, 306 Liberty View Lane, Lynchburg, VA 24502, USA;
| | - Denene Lofland
- Department of Microbiology and Immunology, Tower Campus, Drexel University College of Medicine, 50 Innovation Way, Wyomissing, PA 19610, USA;
| | - Jiayuh Lin
- Department of Biochemistry and Molecular Biology, Molecular Medicine Graduate Program, University of Maryland School of Medicine and Greenebaum Comprehensive Cancer Center, 108 N. Greene Street, Baltimore, MD 21201, USA;
| |
Collapse
|
8
|
Tarannum M, Romee R. Cytokine-induced memory-like natural killer cells for cancer immunotherapy. Stem Cell Res Ther 2021; 12:592. [PMID: 34863287 PMCID: PMC8642969 DOI: 10.1186/s13287-021-02655-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 09/25/2021] [Indexed: 12/12/2022] Open
Abstract
Natural killer cells are an important part of the innate immune system mediating robust responses to virus-infected and malignant cells without needing prior antigen priming. NK cells have always been thought to be short-lived and with no antigen specificity; however, recent data support the presence of NK cell memory including in the hapten-specific contact hypersensitivity model and in certain viral infections. The memory-like features can also be generated by short-term activation of both murine and human NK cells with cytokine combination of IL-12, IL-15 and IL-18, imparting increased longevity and enhanced anticancer functionality. Preclinical studies and very early clinical trials demonstrate safety and very promising clinical activity of these cytokine-induced memory-like (CIML) NK cells, making them an attractive cell type for developing novel adoptive cellular immunotherapy strategies. Furthermore, efforts are on to arm them with novel gene constructs for enhanced tumor targeting and function.
Collapse
Affiliation(s)
- Mubin Tarannum
- Division of Cellular Therapy and Stem Cell Transplantation, Dana Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave, Boston, MA, 02215, USA
| | - Rizwan Romee
- Division of Cellular Therapy and Stem Cell Transplantation, Dana Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave, Boston, MA, 02215, USA.
| |
Collapse
|
9
|
Becker-Hapak MK, Shrestha N, McClain E, Dee MJ, Chaturvedi P, Leclerc GM, Marsala LI, Foster M, Schappe T, Tran J, Desai S, Neal CC, Pence P, Wong P, Wagner JA, Russler-Germain DA, Zhu X, Spanoudis CM, Gallo VL, Echeverri CA, Ramirez LL, You L, Egan JO, Rhode PR, Jiao JA, Muniz GJ, Jeng EK, Prendes CA, Sullivan RP, Berrien-Elliott MM, Wong HC, Fehniger TA. A Fusion Protein Complex that Combines IL-12, IL-15, and IL-18 Signaling to Induce Memory-Like NK Cells for Cancer Immunotherapy. Cancer Immunol Res 2021; 9:1071-1087. [PMID: 34244297 PMCID: PMC8416787 DOI: 10.1158/2326-6066.cir-20-1002] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 04/14/2021] [Accepted: 07/02/2021] [Indexed: 11/16/2022]
Abstract
Natural killer (NK) cells are a promising cellular therapy for cancer, with challenges in the field including persistence, functional activity, and tumor recognition. Briefly, priming blood NK cells with recombinant human (rh)IL-12, rhIL-15, and rhIL-18 (12/15/18) results in memory-like NK cell differentiation and enhanced responses against cancer. However, the lack of available, scalable Good Manufacturing Process (GMP)-grade reagents required to advance this approach beyond early-phase clinical trials is limiting. To address this challenge, we developed a novel platform centered upon an inert tissue factor scaffold for production of heteromeric fusion protein complexes (HFPC). The first use of this platform combined IL-12, IL-15, and IL-18 receptor engagement (HCW9201), and the second adds CD16 engagement (HCW9207). This unique HFPC expression platform was scalable with equivalent protein quality characteristics in small- and GMP-scale production. HCW9201 and HCW9207 stimulated activation and proliferation signals in NK cells, but HCW9207 had decreased IL-18 receptor signaling. RNA sequencing and multidimensional mass cytometry revealed parallels between HCW9201 and 12/15/18. HCW9201 stimulation improved NK cell metabolic fitness and resulted in the DNA methylation remodeling characteristic of memory-like differentiation. HCW9201 and 12/15/18 primed similar increases in short-term and memory-like NK cell cytotoxicity and IFNγ production against leukemia targets, as well as equivalent control of leukemia in NSG mice. Thus, HFPCs represent a protein engineering approach that solves many problems associated with multisignal receptor engagement on immune cells, and HCW9201-primed NK cells can be advanced as an ideal approach for clinical GMP-grade memory-like NK cell production for cancer therapy.
Collapse
Affiliation(s)
| | | | - Ethan McClain
- Washington University School of Medicine, Division of Oncology, Saint Louis, Missouri
| | | | | | | | - Lynne I Marsala
- Washington University School of Medicine, Division of Oncology, Saint Louis, Missouri
| | - Mark Foster
- Washington University School of Medicine, Division of Oncology, Saint Louis, Missouri
| | - Timothy Schappe
- Washington University School of Medicine, Division of Oncology, Saint Louis, Missouri
| | - Jennifer Tran
- Washington University School of Medicine, Division of Oncology, Saint Louis, Missouri
| | - Sweta Desai
- Washington University School of Medicine, Division of Oncology, Saint Louis, Missouri
| | - Carly C Neal
- Washington University School of Medicine, Division of Oncology, Saint Louis, Missouri
| | - Patrick Pence
- Washington University School of Medicine, Division of Oncology, Saint Louis, Missouri
| | - Pamela Wong
- Washington University School of Medicine, Division of Oncology, Saint Louis, Missouri
| | - Julia A Wagner
- Washington University School of Medicine, Division of Oncology, Saint Louis, Missouri
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Todd A Fehniger
- Washington University School of Medicine, Division of Oncology, Saint Louis, Missouri.
| |
Collapse
|
10
|
Kerbauy LN, Marin ND, Kaplan M, Banerjee PP, Berrien-Elliott MM, Becker-Hapak M, Basar R, Foster M, Garcia Melo L, Neal CC, McClain E, Daher M, Nunez Cortes AK, Desai S, Inng Lim FW, Mendt MC, Schappe T, Li L, Shaim H, Shanley M, Ensley EL, Uprety N, Wong P, Liu E, Ang SO, Cai R, Nandivada V, Mohanty V, Miao Q, Shen Y, Baran N, Fowlkes NW, Chen K, Muniz-Feliciano L, Champlin RE, Nieto YL, Koch J, Treder M, Fischer W, Okamoto OK, Shpall EJ, Fehniger TA, Rezvani K. Combining AFM13, a Bispecific CD30/CD16 Antibody, with Cytokine-Activated Blood and Cord Blood-Derived NK Cells Facilitates CAR-like Responses Against CD30 + Malignancies. Clin Cancer Res 2021; 27:3744-3756. [PMID: 33986022 PMCID: PMC8254785 DOI: 10.1158/1078-0432.ccr-21-0164] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/15/2021] [Accepted: 04/28/2021] [Indexed: 12/17/2022]
Abstract
PURPOSE Natural killer (NK)-cell recognition and function against NK-resistant cancers remain substantial barriers to the broad application of NK-cell immunotherapy. Potential solutions include bispecific engagers that target NK-cell activity via an NK-activating receptor when simultaneously targeting a tumor-specific antigen, as well as enhancing functionality using IL12/15/18 cytokine pre-activation. EXPERIMENTAL DESIGN We assessed single-cell NK-cell responses stimulated by the tetravalent bispecific antibody AFM13 that binds CD30 on leukemia/lymphoma targets and CD16A on various types of NK cells using mass cytometry and cytotoxicity assays. The combination of AFM13 and IL12/15/18 pre-activation of blood and cord blood-derived NK cells was investigated in vitro and in vivo. RESULTS We found heterogeneity within AFM13-directed conventional blood NK cell (cNK) responses, as well as consistent AFM13-directed polyfunctional activation of mature NK cells across donors. NK-cell source also impacted the AFM13 response, with cNK cells from healthy donors exhibiting superior responses to those from patients with Hodgkin lymphoma. IL12/15/18-induced memory-like NK cells from peripheral blood exhibited enhanced killing of CD30+ lymphoma targets directed by AFM13, compared with cNK cells. Cord-blood NK cells preactivated with IL12/15/18 and ex vivo expanded with K562-based feeders also exhibited enhanced killing with AFM13 stimulation via upregulation of signaling pathways related to NK-cell effector function. AFM13-NK complex cells exhibited enhanced responses to CD30+ lymphomas in vitro and in vivo. CONCLUSIONS We identify AFM13 as a promising combination with cytokine-activated adult blood or cord-blood NK cells to treat CD30+ hematologic malignancies, warranting clinical trials with these novel combinations.
Collapse
Affiliation(s)
- Lucila N Kerbauy
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Departments of Stem Cell Transplantation and Hemotherapy/Cellular Therapy, Hospital Israelita Albert Einstein, Sao Paulo, Brazil
- Human Genome and Stem Cell Research Center, Department of Genetics and Evolutionary Biology, Biosciences Institute, University of São Paulo (USP), Sao Paulo, Brazil
| | - Nancy D Marin
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Mecit Kaplan
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Pinaki P Banerjee
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Melissa M Berrien-Elliott
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Michelle Becker-Hapak
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Rafet Basar
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mark Foster
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Luciana Garcia Melo
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Carly C Neal
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Ethan McClain
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - May Daher
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ana Karen Nunez Cortes
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sweta Desai
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Francesca Wei Inng Lim
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mayela Carolina Mendt
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Timothy Schappe
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Li Li
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hila Shaim
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mayra Shanley
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Emily L Ensley
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Nadima Uprety
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Pamela Wong
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Enli Liu
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sonny O Ang
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Rong Cai
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Vandana Nandivada
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Vakul Mohanty
- Department of Bioinformatics and Computational Biology, The University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Qi Miao
- Department of Bioinformatics and Computational Biology, The University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Yifei Shen
- Department of Bioinformatics and Computational Biology, The University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Natalia Baran
- Department of Leukemia, The University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Natalie W Fowlkes
- Department of Veterinary Medicine and Surgery, The University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Ken Chen
- Department of Bioinformatics and Computational Biology, The University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Luis Muniz-Feliciano
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Richard E Champlin
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yago L Nieto
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | | | | | - Oswaldo Keith Okamoto
- Departments of Stem Cell Transplantation and Hemotherapy/Cellular Therapy, Hospital Israelita Albert Einstein, Sao Paulo, Brazil
- Human Genome and Stem Cell Research Center, Department of Genetics and Evolutionary Biology, Biosciences Institute, University of São Paulo (USP), Sao Paulo, Brazil
| | - Elizabeth J Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Todd A Fehniger
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri.
| | - Katayoun Rezvani
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
11
|
Lim SA, Moon Y, Shin MH, Kim TJ, Chae S, Yee C, Hwang D, Park H, Lee KM. Hypoxia-Driven HIF-1α Activation Reprograms Pre-Activated NK Cells towards Highly Potent Effector Phenotypes via ERK/STAT3 Pathways. Cancers (Basel) 2021; 13:cancers13081904. [PMID: 33920906 PMCID: PMC8071270 DOI: 10.3390/cancers13081904] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/07/2021] [Accepted: 04/08/2021] [Indexed: 12/19/2022] Open
Abstract
Simple Summary In patients with advanced cancer, hypoxic stress shapes NK cells toward tumor-resistant and immunosuppressive phenotypes. Therefore, a strategy to restore NK cell function within hypoxia would be crucial for successful tumor immunotherapy. By manipulating pO2 exposure to naïve vs. pre-activated NK cells, we found that HIF-1α-dependent metabolic reprogramming of NK cells is the key to overcoming hypoxia-mediated NK cell impairment. Exposure of pre-activated NK cells to hypoxia with 1.5% pO2 initiated metabolic shift from oxidative phosphorylation to glycolysis and reduction of p21/p53-dependent apoptotic pathways, with concomitant upregulation of cell cycle-promoting genes and downregulation of cell cycle-arrest genes via HIF-1a/ERK/STAT3 activation. Furthermore, upregulation of NKp44 activating receptor in hypoxia-exposed pre-activated NK cells elevated cytotoxicity of K562, CEM, and A375 tumor cells, in both in-vitro and in-vivo tumor-clearance assays. Therefore, HIF-1α-mediated metabolic reprogramming of NK cells could reverse their impaired phenotype, generating functionally robust NK cells for adoptive therapy and clinical evaluation. Abstract NK cells are the predominant innate lymphocyte subsets specialized to kill malignant tumor cells. In patients with advanced cancer, hypoxic stress shapes NK cells toward tumor-resistant and immunosuppressive phenotypes, hence a strategy to restore NK function is critical for successful tumor immunotherapy. Here, we present evidence that pre-activation and subsequent HIF-1α-dependent metabolic shift of NK cells from oxidative phosphorylation into glycolysis are keys to overcome hypoxia-mediated impairment in NK cell survival, proliferation, and tumor cytotoxicity. Specifically, exposing NK cells to 7–9 days of normoxic culture followed by a pO2 of 1.5% hypoxia led to a highly potent effector phenotype via HIF-1α stabilization and upregulation of its target genes, BNIP3, PDK1, VEGF, PKM2, and LDHA. RNA sequencing and network analyses revealed that concomitant reduction of p21/p53 apoptotic pathways along with upregulation of cell cycle-promoting genes, CCNE1, CDC6, CDC20, and downregulation of cell cycle-arrest genes, CDKN1A, GADD45A, and MDM2 were accountable for superior expansion of NK cells via ERK/STAT3 activation. Furthermore, HIF-1α-dependent upregulation of the NKp44 receptor in hypoxia-exposed NK cells resulted in increased killing against K562, CEM, and A375 tumor targets both in-vitro and in-vivo tumor clearance assays. Therefore, hypoxic exposure on pre-activated proliferating NK cells triggered HIF-1α-dependent pathways to initiate coordinated regulation of cell cycle, apoptosis, and cytotoxicity at the global gene transcription level. Our results uncover a previously unidentified role of HIF-1α-mediated metabolic reprogramming that can reverse impaired NK effector phenotypes to generate requisite numbers of functionally robust NK cells for adoptive cellular therapy for clinical evaluation.
Collapse
Affiliation(s)
- Seon Ah Lim
- Department of Biochemistry and Molecular Biology, College of Medicine, Korea University, Seoul 02841, Korea; (S.A.L.); (M.H.S.); (T.-J.K.)
| | - Yunwon Moon
- Department of Life Science, University of Seoul, Seoul 02504, Korea;
| | - Min Hwa Shin
- Department of Biochemistry and Molecular Biology, College of Medicine, Korea University, Seoul 02841, Korea; (S.A.L.); (M.H.S.); (T.-J.K.)
| | - Tae-Jin Kim
- Department of Biochemistry and Molecular Biology, College of Medicine, Korea University, Seoul 02841, Korea; (S.A.L.); (M.H.S.); (T.-J.K.)
| | - Sehyun Chae
- Korea Brain Bank, Korea Brain Research Institute, Daegu 41068, Korea;
| | - Cassian Yee
- Departments of Melanoma Medical Oncology and Immunology, MD Anderson Cancer Center, Houston, TX 77054, USA;
| | - Daehee Hwang
- School of Biological Sciences, Seoul National University, Seoul 08826, Korea;
| | - Hyunsung Park
- Department of Life Science, University of Seoul, Seoul 02504, Korea;
- Correspondence: (H.P.); (K.-M.L.); Tel.: +82-2-6490-2670 (H.P.); +82-2-920-6251 (K-M.L.)
| | - Kyung-Mi Lee
- Department of Biochemistry and Molecular Biology, College of Medicine, Korea University, Seoul 02841, Korea; (S.A.L.); (M.H.S.); (T.-J.K.)
- Department of Biomedical Engineering, Center for Bio-Integrated Electronics, Simpson Querrey Institute, Northwestern University, Evanston, IL 60208, USA
- Correspondence: (H.P.); (K.-M.L.); Tel.: +82-2-6490-2670 (H.P.); +82-2-920-6251 (K-M.L.)
| |
Collapse
|
12
|
CAR-modified memory-like NK cells exhibit potent responses to NK-resistant lymphomas. Blood 2021; 136:2308-2318. [PMID: 32614951 DOI: 10.1182/blood.2020006619] [Citation(s) in RCA: 142] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 06/16/2020] [Indexed: 12/13/2022] Open
Abstract
Natural killer (NK) cells are a promising cellular immunotherapy for cancer. Cytokine-induced memory-like (ML) NK cells differentiate after activation with interleukin-12 (IL-12), IL-15, and IL-18, exhibit potent antitumor responses, and safely induce complete remissions in patients with leukemia. However, many cancers are not fully recognized via NK cell receptors. Chimeric antigen receptors (CARs) have been used to enhance tumor-specific recognition by effector lymphocytes. We hypothesized that ML differentiation and CAR engineering would result in complementary improvements in NK cell responses against NK-resistant cancers. To test this idea, peripheral blood ML NK cells were modified to express an anti-CD19 CAR (19-CAR-ML), which displayed significantly increased interferon γ production, degranulation, and specific killing against NK-resistant lymphoma lines and primary targets compared with nonspecific control CAR-ML NK cells or conventional CAR NK cells. The 19-CAR and ML responses were synergistic and CAR specific and required immunoreceptor tyrosine-based activation motif signaling. Furthermore, 19-CAR-ML NK cells generated from lymphoma patients exhibited improved responses against their autologous lymphomas. 19-CAR-ML NK cells controlled lymphoma burden in vivo and improved survival in human xenograft models. Thus, CAR engineering of ML NK cells enhanced responses against resistant cancers and warrants further investigation, with the potential to broaden ML NK cell recognition against a variety of NK cell-resistant tumors.
Collapse
|
13
|
Abstract
ABSTRACT Success from checkpoint blockade and adoptive cell therapy has brought a new hope in cancer immunotherapy. Adoptive cell therapy involves the isolation of immune cells, ex vivo activation and/or expansion, and reinfusion into the patients, and their effect can be dramatically increased by the incorporation of chimeric antigen receptors specific to molecules expressed on tumor cells. Chimeric antigen receptor T cells have shown exciting results in the treatment of liquid malignancies; nevertheless, they suffer from limitations including severe adverse effects such as cytokine release syndrome and neurotoxicity seen in patients as well as a potential for causing graft-versus-host disease in an allogeneic setting. It is thus imperial to explore innate immune cells including natural killer cells, macrophages, natural killer T cells, and γδ T cells. Here, we provide a broad overview of the major innate immune cells and their potential for adoptive cell therapy and chimeric antigen receptor engineering.
Collapse
|
14
|
Berrien-Elliott MM, Cashen AF, Cubitt CC, Neal CC, Wong P, Wagner JA, Foster M, Schappe T, Desai S, McClain E, Becker-Hapak M, Foltz JA, Cooper ML, Jaeger N, Srivatsan SN, Gao F, Romee R, Abboud CN, Uy GL, Westervelt P, Jacoby MA, Pusic I, Stockerl-Goldstein KE, Schroeder MA, DiPersio J, Fehniger TA. Multidimensional Analyses of Donor Memory-Like NK Cells Reveal New Associations with Response after Adoptive Immunotherapy for Leukemia. Cancer Discov 2020; 10:1854-1871. [PMID: 32826231 DOI: 10.1158/2159-8290.cd-20-0312] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 07/17/2020] [Accepted: 08/18/2020] [Indexed: 11/16/2022]
Abstract
Natural killer (NK) cells are an emerging cancer cellular therapy and potent mediators of antitumor immunity. Cytokine-induced memory-like (ML) NK cellular therapy is safe and induces remissions in patients with acute myeloid leukemia (AML). However, the dynamic changes in phenotype that occur after NK-cell transfer that affect patient outcomes remain unclear. Here, we report comprehensive multidimensional correlates from ML NK cell-treated patients with AML using mass cytometry. These data identify a unique in vivo differentiated ML NK-cell phenotype distinct from conventional NK cells. Moreover, the inhibitory receptor NKG2A is a dominant, transcriptionally induced checkpoint important for ML, but not conventional NK-cell responses to cancer. The frequency of CD8α+ donor NK cells is negatively associated with AML patient outcomes after ML NK therapy. Thus, elucidating the multidimensional dynamics of donor ML NK cells in vivo revealed critical factors important for clinical response, and new avenues to enhance NK-cell therapeutics. SIGNIFICANCE: Mass cytometry reveals an in vivo memory-like NK-cell phenotype, where NKG2A is a dominant checkpoint, and CD8α is associated with treatment failure after ML NK-cell therapy. These findings identify multiple avenues for optimizing ML NK-cell immunotherapy for cancer and define mechanisms important for ML NK-cell function.This article is highlighted in the In This Issue feature, p. 1775.
Collapse
Affiliation(s)
- Melissa M Berrien-Elliott
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri.
| | - Amanda F Cashen
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri.,Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri
| | - Celia C Cubitt
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Carly C Neal
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Pamela Wong
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Julia A Wagner
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Mark Foster
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Timothy Schappe
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Sweta Desai
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Ethan McClain
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Michelle Becker-Hapak
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Jennifer A Foltz
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Matthew L Cooper
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Natalia Jaeger
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | | | - Feng Gao
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri
| | - Rizwan Romee
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Camille N Abboud
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri.,Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri
| | - Geoffrey L Uy
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri.,Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri
| | - Peter Westervelt
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri.,Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri
| | - Meagan A Jacoby
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri.,Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri
| | - Iskra Pusic
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri.,Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri
| | - Keith E Stockerl-Goldstein
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri.,Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri
| | - Mark A Schroeder
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri.,Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri
| | - John DiPersio
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri.,Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri
| | - Todd A Fehniger
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri. .,Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
15
|
Olguín JE, Medina-Andrade I, Rodríguez T, Rodríguez-Sosa M, Terrazas LI. Relevance of Regulatory T Cells during Colorectal Cancer Development. Cancers (Basel) 2020; 12:E1888. [PMID: 32674255 PMCID: PMC7409056 DOI: 10.3390/cancers12071888] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 06/21/2020] [Accepted: 07/01/2020] [Indexed: 12/14/2022] Open
Abstract
In recent years, there has been a significant increase in the study of own and foreign human factors favoring the development of different types of cancer, including genetic and environmental ones. However, the fact that the immune response plays a fundamental role in the development of immunity and susceptibility to colorectal cancer (CRC) is much stronger. Among the many cell populations of the immune system that participate in restricting or favoring CRC development, regulatory T cells (Treg) play a major role in orchestrating immunomodulation during CRC. In this review, we established concrete evidence supporting the fact that Treg cells have an important role in the promotion of tumor development during CRC, mediating an increasing suppressive capacity which controls the effector immune response, and generating protection for tumors. Furthermore, Treg cells go through a process called "phenotypic plasticity", where they co-express transcription factors that promote an inflammatory profile. We reunited evidence that describes the interaction between the different effector populations of the immune response and its modulation by Treg cells adapted to the tumor microenvironment, including the mechanisms used by Treg cells to suppress the protective immune response, as well as the different subpopulations of Treg cells participating in tumor progression, generating susceptibility during CRC development. Finally, we discussed whether Treg cells might or might not be a therapeutic target for an effective reduction in the morbidity and mortality caused by CRC.
Collapse
Affiliation(s)
- Jonadab E. Olguín
- Laboratorio Nacional en Salud, Diagnóstico Molecular y Efecto Ambiental en Enfermedades Crónico-Degenerativas, Facultad de Estudios Superiores (FES) Iztacala, Universidad Nacional Autónoma de México (UNAM), Av. De los Barrios # 1, Tlalnepantla 54090, Mexico; (J.E.O.); (I.M.-A.); (T.R.); (M.R.-S.)
- Unidad de Biomedicina, FES Iztacala, UNAM, Av. De los Barrios # 1, Tlalnepantla 54090, Mexico
| | - Itzel Medina-Andrade
- Laboratorio Nacional en Salud, Diagnóstico Molecular y Efecto Ambiental en Enfermedades Crónico-Degenerativas, Facultad de Estudios Superiores (FES) Iztacala, Universidad Nacional Autónoma de México (UNAM), Av. De los Barrios # 1, Tlalnepantla 54090, Mexico; (J.E.O.); (I.M.-A.); (T.R.); (M.R.-S.)
- Unidad de Biomedicina, FES Iztacala, UNAM, Av. De los Barrios # 1, Tlalnepantla 54090, Mexico
| | - Tonathiu Rodríguez
- Laboratorio Nacional en Salud, Diagnóstico Molecular y Efecto Ambiental en Enfermedades Crónico-Degenerativas, Facultad de Estudios Superiores (FES) Iztacala, Universidad Nacional Autónoma de México (UNAM), Av. De los Barrios # 1, Tlalnepantla 54090, Mexico; (J.E.O.); (I.M.-A.); (T.R.); (M.R.-S.)
| | - Miriam Rodríguez-Sosa
- Laboratorio Nacional en Salud, Diagnóstico Molecular y Efecto Ambiental en Enfermedades Crónico-Degenerativas, Facultad de Estudios Superiores (FES) Iztacala, Universidad Nacional Autónoma de México (UNAM), Av. De los Barrios # 1, Tlalnepantla 54090, Mexico; (J.E.O.); (I.M.-A.); (T.R.); (M.R.-S.)
| | - Luis I. Terrazas
- Laboratorio Nacional en Salud, Diagnóstico Molecular y Efecto Ambiental en Enfermedades Crónico-Degenerativas, Facultad de Estudios Superiores (FES) Iztacala, Universidad Nacional Autónoma de México (UNAM), Av. De los Barrios # 1, Tlalnepantla 54090, Mexico; (J.E.O.); (I.M.-A.); (T.R.); (M.R.-S.)
- Unidad de Biomedicina, FES Iztacala, UNAM, Av. De los Barrios # 1, Tlalnepantla 54090, Mexico
| |
Collapse
|
16
|
Fantini M, David JM, Annunziata CM, Morelli MP, Arlen PM, Tsang KY. The Monoclonal Antibody NEO-201 Enhances Natural Killer Cell Cytotoxicity Against Tumor Cells Through Blockade of the Inhibitory CEACAM5/CEACAM1 Immune Checkpoint Pathway. Cancer Biother Radiopharm 2020; 35:190-198. [PMID: 31928422 DOI: 10.1089/cbr.2019.3141] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Background: Natural killer (NK) cells are essential to innate immunity and participate in cancer immune surveillance. Heterophilic interactions between carcinoembryonic antigen (CEA) on tumor cells and carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) on NK cells inhibit NK cell cytotoxicity against tumor cells. NEO-201 is a humanized IgG1 monoclonal antibody that recognizes members of CEACAM family, expressed specifically on a variety of human carcinoma cell lines and tumor tissues. This investigation was designed to determine whether the binding of NEO-201 with CEACAM5 on tumor cells can block the CEACAM5/CEACAM1 interaction to restore antitumor cytotoxicity of NK cells. Materials and Methods: In vitro functional assays, using various human tumor cell lines as target cells and NK-92 cells as effectors, were conducted to assess the ability of NEO-201 to block the interaction between CEACAM5 on tumor cells and CEACAM1 on NK cells to enhance the in vitro killing of tumor cells by NK-92. NK-92 cells were used as a model of direct NK killing of tumor cells because they lack antibody-dependent cellular cytotoxicity activity. Results: Expression profiling revealed that various human carcinoma cell lines expressed different levels of CEACAM5+ and NEO-201+ cells. Addition of NEO-201 significantly enhanced NK-92 cell cytotoxicity against highly CEACAM5+/NEO-201+ expressing tumor cells, suggesting that its activity is correlated with the level of CEACAM5+/NEO-201+ expression. Conclusions: These findings demonstrate that NEO-201 can block the interaction between CEACAM5 on tumor cells and CEACAM1 on NK cells to reverse CEACAM1-dependent inhibition of NK cytotoxicity.
Collapse
Affiliation(s)
| | | | - Christina M Annunziata
- Women's Malignancy Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Maria Pia Morelli
- Women's Malignancy Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | | | | |
Collapse
|
17
|
Koo BW, Lim DJ, Oh AY, Na HS. Retrospective Comparison between the Effects of Propofol and Inhalation Anesthetics on Postoperative Recurrence of Early- and Intermediate-Stage Hepatocellular Carcinoma. Med Princ Pract 2020; 29:422-428. [PMID: 32074612 PMCID: PMC7511682 DOI: 10.1159/000506637] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 02/17/2020] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE Previous studies have reported that propofol has antitumor, anti-inflammatory, and antioxidant effects in addition to its anesthetic properties. To confirm this, a retrospective investigation was conducted to determine whether different anesthetic agents, particularly propofol and inhalation anesthetics, have an effect on the recurrence of hepatocellular carcinoma (HCC) in patients who were diagnosed with primary HCC and underwent laparoscopic hepatectomy. SUBJECTS AND METHODS Patients with Barcelona Clinic Liver Cancer stages 0, A, and B HCC, who underwent laparoscopic hepatic resection, were enrolled in this study. Post-operative HCC recurrence, which was determined from postoperative liver CT, was evaluated 24 months postoperatively with respect to the main anesthetic agents. The characteristics of HCC and other patient-related or surgery-related variables were evaluated together. RESULTS AND CONCLUSION During the 24-month period after hepatic resection, less HCC patients in the propofol group than in the inhalation group recurred (p = 0.046). The mean time to recurrence was 20.8 months (95% CI, 19.7-22.0) and 19.1 months (95% CI, 17.8-20.4) in the propofol group and the inhalation group, respectively. In addition, multivariable Cox proportional regression analysis revealed that the propofol group showed significantly decreased recurrence versus the inhalation group (hazard ratio, 0.57; 95% CI, 0.47-0.69; p = 0.029). When propofol was used as the main general anesthetic agent for laparoscopic hepatic resection, the postoperative 2-year recurrence rate decreased in early- and intermediate-stage HCC.
Collapse
Affiliation(s)
- Bon-Wook Koo
- Department of Anesthesiology and Pain Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Dae-Jin Lim
- Department of Anesthesiology and Pain Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Ah-Young Oh
- Department of Anesthesiology and Pain Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Hyo-Seok Na
- Department of Anesthesiology and Pain Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea,
| |
Collapse
|
18
|
Decaup E, Rossi C, Gravelle P, Laurent C, Bordenave J, Tosolini M, Tourette A, Perrial E, Dumontet C, Poupot M, Klein C, Savina A, Fournié JJ, Bezombes C. A Tridimensional Model for NK Cell-Mediated ADCC of Follicular Lymphoma. Front Immunol 2019; 10:1943. [PMID: 31475004 PMCID: PMC6702952 DOI: 10.3389/fimmu.2019.01943] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 08/01/2019] [Indexed: 12/22/2022] Open
Abstract
Follicular lymphoma (FL) is the second most frequent subtype of B non-Hodgkin's lymphomas (NHL) for which the treatment is based on the use of anti-CD20 mAbs. NK cells play a crucial role in their mechanism of action and the number of these cells mediating antibody-dependent cell cycotoxicity (ADCC) in the peripheral blood of FL patients predict the outcome. However, their presence in FL biopsies, their activation and their role have been poorly investigated. Moreover, in vitro studies have not deciphered the exact signaling cascades triggered by NK cells in presence of anti-CD20 mAbs on both effector and target cells in a relevant FL model. We performed in silico analyses and ex vivo functional assays to determine the presence and the activation status of NK cells in FL biopsies. We modelized ADCC phenomenon by developing a co-culture model composed by 3D-cultured FL cells and NK cells. Thus, we investigated the biological effect of anti-CD20 mAbs by fluorescent microscopy and the phosphorylation status of survival pathways by cell bar coding phosphoflow in target cells. In parallel, we measured the status of activation of downstream FcγRIIIa signaling pathways in effector cells and their activation (CD69, perforin, granzyme B, IFNγ) by flow cytometry. We determined by in vivo experiments the effects of anti-CD20 mAbs in presence of NK cells in SCID-Beige engrafted FL mice. Here, we show that functional NK cells infiltrate FL biopsies, and that their presence tends to correlate with the survival of FL patients. Using our 3D co-culture model, we show that rituximab and GA101 are able to promote degranulation, CD69 expression, IFNγ production and activate FcγRIIIa signaling cascade in NK cells, and inhibit survival pathways and induce apoptosis in FL cells. The effect of GA101 seems to be more pronounced as observed in vivo in a xenograft FL model. This study strongly supports the role of NK cells in FL and highlights the application of the 3D co-culture model for in vitro validation.
Collapse
Affiliation(s)
- Emilie Decaup
- Centre de Recherches en Cancérologie de Toulouse (CRCT), UMR1037 INSERM, Université Toulouse III: Paul-Sabatier, ERL5294 CNRS, Université de Toulouse, Toulouse, France.,Laboratoire d'Excellence TOUCAN, Toulouse, France.,Programme Hospitalo-Universitaire en Cancérologie CAPTOR, Toulouse, France
| | - Cédric Rossi
- Centre de Recherches en Cancérologie de Toulouse (CRCT), UMR1037 INSERM, Université Toulouse III: Paul-Sabatier, ERL5294 CNRS, Université de Toulouse, Toulouse, France.,Laboratoire d'Excellence TOUCAN, Toulouse, France.,Programme Hospitalo-Universitaire en Cancérologie CAPTOR, Toulouse, France.,CHU Dijon, Hématologie Clinique, Hôpital François Mitterand, Dijon, France
| | - Pauline Gravelle
- Centre de Recherches en Cancérologie de Toulouse (CRCT), UMR1037 INSERM, Université Toulouse III: Paul-Sabatier, ERL5294 CNRS, Université de Toulouse, Toulouse, France.,Laboratoire d'Excellence TOUCAN, Toulouse, France.,Programme Hospitalo-Universitaire en Cancérologie CAPTOR, Toulouse, France.,Department of Pathology, Institut Universitaire du Cancer de Toulouse, Toulouse, France
| | - Camille Laurent
- Centre de Recherches en Cancérologie de Toulouse (CRCT), UMR1037 INSERM, Université Toulouse III: Paul-Sabatier, ERL5294 CNRS, Université de Toulouse, Toulouse, France.,Laboratoire d'Excellence TOUCAN, Toulouse, France.,Programme Hospitalo-Universitaire en Cancérologie CAPTOR, Toulouse, France.,Department of Pathology, Institut Universitaire du Cancer de Toulouse, Toulouse, France
| | - Julie Bordenave
- Centre de Recherches en Cancérologie de Toulouse (CRCT), UMR1037 INSERM, Université Toulouse III: Paul-Sabatier, ERL5294 CNRS, Université de Toulouse, Toulouse, France.,Laboratoire d'Excellence TOUCAN, Toulouse, France.,Programme Hospitalo-Universitaire en Cancérologie CAPTOR, Toulouse, France
| | - Marie Tosolini
- Pôle Technologique du Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
| | - Anne Tourette
- INSERM1052/CNRS5286/Université Claude Bernard, Lyon, France
| | | | | | - Mary Poupot
- Centre de Recherches en Cancérologie de Toulouse (CRCT), UMR1037 INSERM, Université Toulouse III: Paul-Sabatier, ERL5294 CNRS, Université de Toulouse, Toulouse, France.,Laboratoire d'Excellence TOUCAN, Toulouse, France.,Programme Hospitalo-Universitaire en Cancérologie CAPTOR, Toulouse, France
| | - Christian Klein
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland
| | | | - Jean-Jacques Fournié
- Centre de Recherches en Cancérologie de Toulouse (CRCT), UMR1037 INSERM, Université Toulouse III: Paul-Sabatier, ERL5294 CNRS, Université de Toulouse, Toulouse, France.,Laboratoire d'Excellence TOUCAN, Toulouse, France.,Programme Hospitalo-Universitaire en Cancérologie CAPTOR, Toulouse, France
| | - Christine Bezombes
- Centre de Recherches en Cancérologie de Toulouse (CRCT), UMR1037 INSERM, Université Toulouse III: Paul-Sabatier, ERL5294 CNRS, Université de Toulouse, Toulouse, France.,Laboratoire d'Excellence TOUCAN, Toulouse, France.,Programme Hospitalo-Universitaire en Cancérologie CAPTOR, Toulouse, France
| |
Collapse
|
19
|
Klener P, Etrych T, Klener P. Biological Therapy of Hematologic Malignancies: Toward a Chemotherapy- free Era. Curr Med Chem 2019; 26:1002-1018. [PMID: 28990505 DOI: 10.2174/0929867324666171006144725] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 09/07/2017] [Accepted: 09/15/2017] [Indexed: 12/20/2022]
Abstract
Less than 70 years ago, the vast majority of hematologic malignancies were untreatable diseases with fatal prognoses. The development of modern chemotherapy agents, which had begun after the Second World War, was markedly accelerated by the discovery of the structure of DNA and its role in cancer biology and tumor cell division. The path travelled from the first temporary remissions observed in children with acute lymphoblastic leukemia treated with single-agent antimetabolites until the first cures achieved by multi-agent chemotherapy regimens was incredibly short. Despite great successes, however, conventional genotoxic cytostatics suffered from an inherently narrow therapeutic index and extensive toxicity, which in many instances limited their clinical utilization. In the last decade of the 20th century, increasing knowledge on the biology of certain malignancies resulted in the conception and development of first molecularly targeted agents designed to inhibit specific druggable molecules involved in the survival of cancer cells. Advances in technology and genetic engineering enabled the production of structurally complex anticancer macromolecules called biologicals, including therapeutic monoclonal antibodies, antibody-drug conjugates and antibody fragments. The development of drug delivery systems (DDSs), in which conventional drugs were attached to various types of carriers including nanoparticles, liposomes or biodegradable polymers, represented an alternative approach to the development of new anticancer agents. Despite the fact that the antitumor activity of drugs attached to DDSs was not fundamentally different, the improved pharmacokinetic profiles, decreased toxic side effects and significantly increased therapeutic indexes resulted in their enhanced antitumor efficacy compared to conventional (unbound) drugs. Approval of the first immune checkpoint inhibitor for the treatment of cancer in 2011 initiated the era of cancer immunotherapy. Checkpoint inhibitors, bispecific T-cell engagers, adoptive T-cell approaches and cancer vaccines have joined the platform so far, represented mainly by recombinant cytokines, therapeutic monoclonal antibodies and immunomodulatory agents. In specific clinical indications, conventional drugs have already been supplanted by multi-agent, chemotherapy-free regimens comprising diverse immunotherapy and/or targeted agents. The very distinct mechanisms of the anticancer activity of new immunotherapy approaches not only call for novel response criteria, but might also change fundamental treatment paradigms of certain types of hematologic malignancies in the near future.
Collapse
Affiliation(s)
- Pavel Klener
- First Medical Department- Dept. of Hematology, First Faculty of Medicine and General University Hospital, Charles University, Czech Republic.,Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Czech Republic
| | - Tomas Etrych
- Department of biomedical polymers, Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Heyrovského náměstí 2, 162 06 Prague, Czech Republic
| | - Pavel Klener
- First Medical Department- Dept. of Hematology, First Faculty of Medicine and General University Hospital, Charles University, Czech Republic
| |
Collapse
|
20
|
Ellwanger K, Reusch U, Fucek I, Wingert S, Ross T, Müller T, Schniegler-Mattox U, Haneke T, Rajkovic E, Koch J, Treder M, Tesar M. Redirected optimized cell killing (ROCK®): A highly versatile multispecific fit-for-purpose antibody platform for engaging innate immunity. MAbs 2019; 11:899-918. [PMID: 31172847 PMCID: PMC6601565 DOI: 10.1080/19420862.2019.1616506] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Redirection of immune cells to efficiently eliminate tumor cells holds great promise. Natural killer cells (NK), macrophages, or T cells are specifically engaged with target cells expressing markers after infection or neoplastic transformation, resulting in their activation and subsequent killing of those targets. Multiple strategies to redirect immunity have been developed in the past two decades, but they have technical hurdles or cause undesirable side-effects, as exemplified by the T cell-based chimeric antigen receptor approaches (CAR-T therapies) or bispecific T cell engager platforms. Our first-in-class bispecific antibody redirecting innate immune cells to tumors (AFM13, a CD30/CD16A-specific innate immune cell engager) has shown signs of clinical efficacy in CD30-positive lymphomas and the potential to be safely administered, indicating a wider therapeutic window compared to T cell engaging therapies. AFM13 is the most advanced candidate from our fit-for-purpose redirected optimized cell killing (ROCK®) antibody platform, which comprises a plethora of CD16A-binding innate immune cell engagers with unique properties. Here, we discuss aspects of this modular platform, including the advantages of innate immune cell engagement over classical monoclonal antibodies and other engager concepts. We also present details on its potential to engineer a fit-for-purpose innate immune cell engager format that can be equipped with unique CD16A domains, modules that influence pharmacokinetic properties and molecular architectures that influence the activation of immune effectors, as well as tumor targeting. The ROCK® platform is aimed at the activation of innate immunity for the effective lysis of tumor cells and holds the promise of overcoming limitations of other approaches that redirect immune cells by widening the therapeutic window.
Collapse
Affiliation(s)
| | - Uwe Reusch
- a Affimed GmbH, Research Department , Heidelberg , Germany
| | - Ivica Fucek
- a Affimed GmbH, Research Department , Heidelberg , Germany
| | | | - Thorsten Ross
- a Affimed GmbH, Research Department , Heidelberg , Germany
| | - Thomas Müller
- a Affimed GmbH, Research Department , Heidelberg , Germany
| | | | - Torsten Haneke
- a Affimed GmbH, Research Department , Heidelberg , Germany
| | - Erich Rajkovic
- a Affimed GmbH, Research Department , Heidelberg , Germany
| | - Joachim Koch
- a Affimed GmbH, Research Department , Heidelberg , Germany
| | - Martin Treder
- a Affimed GmbH, Research Department , Heidelberg , Germany
| | - Michael Tesar
- a Affimed GmbH, Research Department , Heidelberg , Germany
| |
Collapse
|
21
|
Singh S, Chakrabarti R. Consequences of EMT-Driven Changes in the Immune Microenvironment of Breast Cancer and Therapeutic Response of Cancer Cells. J Clin Med 2019; 8:jcm8050642. [PMID: 31075939 PMCID: PMC6572359 DOI: 10.3390/jcm8050642] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 04/30/2019] [Accepted: 05/04/2019] [Indexed: 02/06/2023] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) is a process through which epithelial cells lose their epithelial characteristics and cell–cell contact, thus increasing their invasive potential. In addition to its well-known roles in embryonic development, wound healing, and regeneration, EMT plays an important role in tumor progression and metastatic invasion. In breast cancer, EMT both increases the migratory capacity and invasive potential of tumor cells, and initiates protumorigenic alterations in the tumor microenvironment (TME). In particular, recent evidence has linked increased expression of EMT markers such as TWIST1 and MMPs in breast tumors with increased immune infiltration in the TME. These immune cells then provide cues that promote immune evasion by tumor cells, which is associated with enhanced tumor progression and metastasis. In the current review, we will summarize the current knowledge of the role of EMT in the biology of different subtypes of breast cancer. We will further explore the correlation between genetic switches leading to EMT and EMT-induced alterations within the TME that drive tumor growth and metastasis, as well as their possible effect on therapeutic response in breast cancer.
Collapse
Affiliation(s)
- Snahlata Singh
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Rumela Chakrabarti
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
22
|
Fantini M, David JM, Wong HC, Annunziata CM, Arlen PM, Tsang KY. An IL-15 Superagonist, ALT-803, Enhances Antibody-Dependent Cell-Mediated Cytotoxicity Elicited by the Monoclonal Antibody NEO-201 Against Human Carcinoma Cells. Cancer Biother Radiopharm 2019; 34:147-159. [PMID: 30601063 PMCID: PMC6482908 DOI: 10.1089/cbr.2018.2628] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND A major mechanism of action for therapeutic antibodies is antibody-dependent cell-mediated cytotoxicity (ADCC). ALT-803 is an interleukin-15 superagonist complex that enhances ADCC against human carcinoma cells in vitro and exerts an antitumor activity in murine, rat, and human carcinomas in vivo. The authors investigated the ability of ALT-803 to modulate ADCC mediated by the humanized IgG1 monoclonal antibody (mAb) NEO-201 against human carcinoma cells. MATERIALS AND METHODS ALT-803 modulating activity on ADCC mediated by NEO-201 was evaluated on several NEO-201 ligand-expressing human carcinoma cells. Purified human natural killer (NK) cells from multiple healthy donors were treated with ALT-803 before their use as effectors in ADCC assay. Modulation of NK cell phenotype and cytotoxic function by exposure to ALT-803 was evaluated by flow cytometry and gene expression analysis. RESULTS ALT-803 significantly enhanced ADCC mediated by NEO-201. ALT-803 also upregulated NK activating receptors, antiapoptotic factors, and factors involved in the NK cytotoxicity, as well as downregulated gene expression of NK inhibiting receptors. CONCLUSIONS These findings indicate that ALT-803 can enhance ADCC activity mediated by NEO-201, by modulating NK activation and cytotoxicity, suggesting a possible clinical use of ALT-803 in combination with NEO-201 for the treatment of human carcinomas.
Collapse
Affiliation(s)
| | | | | | - Christina M. Annunziata
- Women's Malignancy Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | | | - Kwong Y. Tsang
- Precision Biologics, Inc., Rockville, Maryland
- Address correspondence to: Kwong Y. Tsang; Precision Biologics, Inc.; 9600 Medical Center Drive, Suite 300, Rockville, MD 20850
| |
Collapse
|
23
|
Jacobs B, Pfefferle A, Clement D, Berg-Larsen A, Saetersmoen ML, Lorenz S, Wiiger MT, Goodridge JP, Malmberg KJ. Induction of the BIM Short Splice Variant Sensitizes Proliferating NK Cells to IL-15 Withdrawal. THE JOURNAL OF IMMUNOLOGY 2018; 202:736-746. [PMID: 30578306 DOI: 10.4049/jimmunol.1801146] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 11/20/2018] [Indexed: 01/26/2023]
Abstract
Adoptive transfer of allogeneic NK cells holds great promise for cancer immunotherapy. There is a variety of protocols to expand NK cells in vitro, most of which are based on stimulation with cytokines alone or in combination with feeder cells. Although IL-15 is essential for NK cell homeostasis in vivo, it is commonly used at supraphysiological levels to induce NK cell proliferation in vitro. As a result, adoptive transfer of such IL-15-addicted NK cells is associated with cellular stress because of sudden cytokine withdrawal. In this article, we describe a dose-dependent addiction to IL-15 during in vitro expansion of human NK cells, leading to caspase-3 activation and profound cell death upon IL-15 withdrawal. NK cell addiction to IL-15 was tightly linked to the BCL-2/BIM ratio, which rapidly dropped during IL-15 withdrawal. Furthermore, we observed a proliferation-dependent induction of BIM short, a highly proapoptotic splice variant of BIM in IL-15-activated NK cells. These findings shed new light on the molecular mechanisms involved in NK cell apoptosis following cytokine withdrawal and may guide future NK cell priming strategies in a cell therapy setting.
Collapse
Affiliation(s)
- Benedikt Jacobs
- K.G. Jebsen Centre for Cancer Immunotherapy, Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway.,Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, 0310 Oslo, Norway
| | - Aline Pfefferle
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, 14186 Stockholm, Sweden
| | - Dennis Clement
- K.G. Jebsen Centre for Cancer Immunotherapy, Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway.,Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, 0310 Oslo, Norway
| | - Axel Berg-Larsen
- K.G. Jebsen Centre for Cancer Immunotherapy, Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway.,Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, 0310 Oslo, Norway
| | - Michelle L Saetersmoen
- K.G. Jebsen Centre for Cancer Immunotherapy, Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway.,Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, 0310 Oslo, Norway
| | - Susanne Lorenz
- Department of Tumor Biology, Norwegian Radium Hospital, Oslo University Hospital, 0310 Oslo, Norway; and.,Genomics Core Facility, Department of Core Facilities, Norwegian Radium Hospital, Oslo University Hospital, 0310 Oslo, Norway
| | - Merete Thune Wiiger
- K.G. Jebsen Centre for Cancer Immunotherapy, Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway.,Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, 0310 Oslo, Norway
| | - Jodie P Goodridge
- K.G. Jebsen Centre for Cancer Immunotherapy, Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway.,Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, 0310 Oslo, Norway
| | - Karl-Johan Malmberg
- K.G. Jebsen Centre for Cancer Immunotherapy, Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway; .,Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, 0310 Oslo, Norway.,Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, 14186 Stockholm, Sweden
| |
Collapse
|
24
|
Ishikawa T, Okayama T, Sakamoto N, Ideno M, Oka K, Enoki T, Mineno J, Yoshida N, Katada K, Kamada K, Uchiyama K, Handa O, Takagi T, Konishi H, Kokura S, Uno K, Naito Y, Itoh Y. Phase I clinical trial of adoptive transfer of expanded natural killer cells in combination with IgG1 antibody in patients with gastric or colorectal cancer. Int J Cancer 2018; 142:2599-2609. [PMID: 29388200 DOI: 10.1002/ijc.31285] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 01/06/2018] [Accepted: 01/18/2018] [Indexed: 12/16/2022]
Abstract
Natural killer (NK) cells exhibit strong cytotoxic activity against tumor cells without prior sensitization, and have the potential to exert antibody-dependent cellular cytotoxicity (ADCC). In this clinical trial, we examined the safety and efficacy of the use of NK cells, generated using a novel expansion system, in combination with IgG1 antibodies for the treatment of advanced gastric or colorectal cancers. Treatment consisted of trastuzumab- or cetuximab-based chemotherapy, plus adoptive NK cell therapy. For administration of expanded NK cells, dose escalation with a sequential 3 + 3 design was performed in three steps, at doses of 0.5 × 109 , 1.0 × 109 , and 2.0 × 109 cells/injection (N = 9). After 3 days of IgG1 antibody administration, patients were infused with expanded NK cells three times at triweekly intervals. NK cell populations expanded with our system were confirmed as being enriched in NK cells (median 92.9%) with high expression of NKG2D (97.6%) and CD16 (69.6%). The combination therapy was very well tolerated with no severe adverse events. Among six evaluable patients, four presented stable disease (SD) and two presented progressive disease. Of the four SD patients, three showed an overall decrease in tumor size after combination therapy. Immune monitoring suggested that combination therapy enhanced whole blood IFN-γ production and reduced peripheral regulatory T cells (Tregs). In conclusion, this phase I trial provides evidence of good tolerability, induction of Th1 immune responses, and preliminary anti-tumor activity for this combination therapy, in patients with advanced gastric and colorectal cancer that have received previous therapy.
Collapse
Affiliation(s)
- Takeshi Ishikawa
- Department of Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tetsuya Okayama
- Department of Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Naoyuki Sakamoto
- Department of Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | | | - Kaname Oka
- Department of Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | | | | | - Naohisa Yoshida
- Department of Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kazuhiro Katada
- Department of Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kazuhiro Kamada
- Department of Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kazuhiko Uchiyama
- Department of Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Osamu Handa
- Department of Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tomohisa Takagi
- Department of Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hideyuki Konishi
- Department of Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Satoshi Kokura
- Department of Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kazuko Uno
- Division of Basic Research, Louis Pasteur Center for Medical Research, Kyoto, Japan
| | - Yuji Naito
- Department of Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yoshito Itoh
- Department of Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
25
|
Romee R, Cooley S, Berrien-Elliott MM, Westervelt P, Verneris MR, Wagner JE, Weisdorf DJ, Blazar BR, Ustun C, DeFor TE, Vivek S, Peck L, DiPersio JF, Cashen AF, Kyllo R, Musiek A, Schaffer A, Anadkat MJ, Rosman I, Miller D, Egan JO, Jeng EK, Rock A, Wong HC, Fehniger TA, Miller JS. First-in-human phase 1 clinical study of the IL-15 superagonist complex ALT-803 to treat relapse after transplantation. Blood 2018; 131:2515-2527. [PMID: 29463563 PMCID: PMC5992862 DOI: 10.1182/blood-2017-12-823757] [Citation(s) in RCA: 301] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 02/13/2018] [Indexed: 01/15/2023] Open
Abstract
New therapies for patients with hematologic malignancies who relapse after allogeneic hematopoietic cell transplantation (allo-HCT) are needed. Interleukin 15 (IL-15) is a cytokine that stimulates CD8+ T-cell and natural killer (NK) cell antitumor responses, and we hypothesized this cytokine may augment antileukemia/antilymphoma immunity in vivo. To test this, we performed a first-in-human multicenter phase 1 trial of the IL-15 superagonist complex ALT-803 in patients who relapsed >60 days after allo-HCT. ALT-803 was administered to 33 patients via the IV or subcutaneous (SQ) routes once weekly for 4 doses (dose levels of 1, 3, 6, and 10 μg/kg). ALT-803 was well tolerated, and no dose-limiting toxicities or treatment-emergent graft-versus-host disease requiring systemic therapy was observed in this clinical setting. Adverse events following IV administration included constitutional symptoms temporally related to increased serum IL-6 and interferon-γ. To mitigate these effects, the SQ route was tested. SQ delivery resulted in self-limited injection site rashes infiltrated with lymphocytes without acute constitutional symptoms. Pharmacokinetic analysis revealed prolonged (>96 hour) serum concentrations following SQ, but not IV, injection. ALT-803 stimulated the activation, proliferation, and expansion of NK cells and CD8+ T cells without increasing regulatory T cells. Responses were observed in 19% of evaluable patients, including 1 complete remission lasting 7 months. Thus, ALT-803 is a safe, well-tolerated agent that significantly increased NK and CD8+ T cell numbers and function. This immunostimulatory IL-15 superagonist warrants further investigation to augment antitumor immunity alone and combined with other immunotherapies. This trial was registered at www.clinicaltrials.gov as #NCT01885897.
Collapse
Affiliation(s)
- Rizwan Romee
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | | | - Melissa M Berrien-Elliott
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Peter Westervelt
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | | | | | | | | | | | | | - Sithara Vivek
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | - Lindsey Peck
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - John F DiPersio
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Amanda F Cashen
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Rachel Kyllo
- Division of Dermatology, Department of Medicine, and
| | - Amy Musiek
- Division of Dermatology, Department of Medicine, and
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | | | - Milan J Anadkat
- Division of Dermatology, Department of Medicine, and
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Ilana Rosman
- Division of Dermatology, Department of Medicine, and
| | - Daniel Miller
- Department of Dermatology, University of Minnesota, Minneapolis, MN; and
| | - Jack O Egan
- Altor BioScience, a Nantworks company, Miramar, FL
| | - Emily K Jeng
- Altor BioScience, a Nantworks company, Miramar, FL
| | - Amy Rock
- Altor BioScience, a Nantworks company, Miramar, FL
| | - Hing C Wong
- Altor BioScience, a Nantworks company, Miramar, FL
| | - Todd A Fehniger
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | | |
Collapse
|
26
|
Ooi SL, McMullen D, Golombick T, Nut D, Pak SC. Evidence-Based Review of BioBran/MGN-3 Arabinoxylan Compound as a Complementary Therapy for Conventional Cancer Treatment. Integr Cancer Ther 2018; 17:165-178. [PMID: 29037071 PMCID: PMC6041933 DOI: 10.1177/1534735417735379] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Revised: 08/06/2017] [Accepted: 09/12/2017] [Indexed: 12/18/2022] Open
Abstract
INTRODUCTION Conventional cancer treatment, including surgery, chemotherapy, and radiotherapy, may not be sufficient to eradicate all malignant cells and prevent recurrence. Intensive treatment often leads to a depressed immune system, drug resistance, and toxicity, hampering the treatment outcomes. BioBran/MGN-3 Arabinoxylan is a standardized arabinoxylan concentrate which has been proposed as a plant-based immunomodulator that can restore the tumor-induced disturbance of the natural immune system, including natural killer cell activity to fight cancer, complementing conventional therapies. OBJECTIVES To comprehensively review the available evidence on the effects and efficacies of MGN-3 as a complementary therapy for conventional cancer treatment. METHODS Systematic search of journal databases and gray literature for primary studies reporting the effects of MGN-3 on cancer and cancer treatment. RESULTS Thirty full-text articles and 2 conference abstracts were included in this review. MGN-3 has been shown to possess immunomodulating anticancer effects and can work synergistically with chemotherapeutic agents, in vitro. In murine models, MGN-3 has been shown to act against carcinogenic agents, and inhibit tumor growth, either by itself or in combination with other anticancer compounds. Fourteen successful MGN-3 treated clinical cases were found. Eleven clinical studies, including 5 nonrandomized, pre-post intervention studies and 6 randomized controlled trials (RCTs) were located. Reported effects include enhanced immunoprofile, reduced side effects, improved treatment outcomes; one RCT established significantly increased survival rates. There are no reports on adverse events on MGN-3. Most of the clinical trials are small studies with short duration. CONCLUSION There is sufficient evidence suggesting MGN-3 to be an effective immunomodulator that can complement conventional cancer treatment. However, more well-designed RCTs on MGN-3 are needed to strengthen the evidence base.
Collapse
Affiliation(s)
- Soo Liang Ooi
- Centre of Complementary & Alternative Medicine, Singapore
| | - Debbie McMullen
- Charles Sturt University, Bathurst, New South Wales, Australia
| | | | - Dipl Nut
- St George Hospital, Sydney, New South Wales, Australia
| | - Sok Cheon Pak
- Charles Sturt University, Bathurst, New South Wales, Australia
| |
Collapse
|
27
|
Fehniger TA, Miller JS, Stuart RK, Cooley S, Salhotra A, Curtsinger J, Westervelt P, DiPersio JF, Hillman TM, Silver N, Szarek M, Gorelik L, Lowdell MW, Rowinsky E. A Phase 1 Trial of CNDO-109-Activated Natural Killer Cells in Patients with High-Risk Acute Myeloid Leukemia. Biol Blood Marrow Transplant 2018; 24:1581-1589. [PMID: 29597002 DOI: 10.1016/j.bbmt.2018.03.019] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 03/22/2018] [Indexed: 12/24/2022]
Abstract
Natural killer (NK) cells are an emerging immunotherapy approach to acute myeloid leukemia (AML); however, the optimal approach to activate NK cells before adoptive transfer remains unclear. Human NK cells that are primed with the CTV-1 leukemia cell line lysate CNDO-109 exhibit enhanced cytotoxicity against NK cell-resistant cell lines. To translate this finding to the clinic, CNDO-109-activated NK cells (CNDO-109-NK cells) isolated from related HLA-haploidentical donors were evaluated in a phase 1 dose-escalation trial at doses of 3 × 105 (n = 3), 1 × 106 (n = 3), and 3 × 106 (n = 6) cells/kg in patients with AML in first complete remission (CR1) at high risk for recurrence. Before CNDO-109-NK cell administration, patients were treated with lymphodepleting fludarabine/cyclophosphamide. CNDO-109-NK cells were well tolerated, and no dose-limiting toxicities were observed at the highest tested dose. The median relapse-free survival (RFS) by dose level was 105 (3 × 105), 156 (1 × 106), and 337 (3 × 106) days. Two patients remained relapse-free in post-trial follow-up, with RFS durations exceeding 42.5 months. Donor NK cell microchimerism was detected on day 7 in 10 of 12 patients, with 3 patients having evidence of donor cells on day 14 or later. This trial establishes that CNDO-109-NK cells generated from related HLA haploidentical donors, cryopreserved, and then safely administered to AML patients with transient persistence without exogenous cytokine support. Three durable complete remissions of 32.6 to 47.6+ months were observed, suggesting additional clinical investigation of CNDO-109-NK cells for patients with myeloid malignancies, alone or in combination with additional immunotherapy strategies, is warranted.
Collapse
Affiliation(s)
- Todd A Fehniger
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri.
| | - Jeffrey S Miller
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Robert K Stuart
- Department of Hematology and Oncology, Medical University of South Carolina, Charleston, South Carolina
| | - Sarah Cooley
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Amandeep Salhotra
- Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute of City of Hope, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Julie Curtsinger
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Peter Westervelt
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - John F DiPersio
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | | | | | - Michael Szarek
- Department of Epidemiology and Biostatistics, SUNY Downstate School of Public Health, Brooklyn New York
| | | | - Mark W Lowdell
- Department of Hematology, Royal Free Hospital, UCL Medical School, London, United Kingdom
| | | |
Collapse
|
28
|
Targeting autophagy inhibits melanoma growth by enhancing NK cells infiltration in a CCL5-dependent manner. Proc Natl Acad Sci U S A 2017; 114:E9271-E9279. [PMID: 29078276 PMCID: PMC5676879 DOI: 10.1073/pnas.1703921114] [Citation(s) in RCA: 172] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The failure in achieving a durable clinical immune response against cancer cells depends on the ability of cancer cells to establish a microenvironment that prevent cytotoxic immune cells to infiltrate tumors and kill cancer cells. Therefore, the key approach to achieving successful antitumor immune response is to harness strategies allowing the reorientation of immune cells to the tumor. Herein we reveal that inhibiting autophagy induces a massive infiltration of natural killer immune cells into the tumor bed, and a subsequent dramatic decrease in the tumor volume of melanomas. These results highlight the role of targeting autophagy in breaking the immunosuppressive tumor microenvironment barrier, thus allowing the infiltration of natural killer cells into the tumor to kill cancer cells. While blocking tumor growth by targeting autophagy is well established, its role on the infiltration of natural killer (NK) cells into tumors remains unknown. Here, we investigate the impact of targeting autophagy gene Beclin1 (BECN1) on the infiltration of NK cells into melanomas. We show that, in addition to inhibiting tumor growth, targeting BECN1 increased the infiltration of functional NK cells into melanoma tumors. We provide evidence that driving NK cells to the tumor bed relied on the ability of autophagy-defective tumors to transcriptionally overexpress the chemokine gene CCL5. Such infiltration and tumor regression were abrogated by silencing CCL5 in BECN1-defective tumors. Mechanistically, we show that the up-regulated expression of CCL5 occurred through the activation of its transcription factor c-Jun by a mechanism involving the impairment of phosphatase PP2A catalytic activity and the subsequent activation of JNK. Similar to BECN1, targeting other autophagy genes, such as ATG5, p62/SQSTM1, or inhibiting autophagy pharmacologically by chloroquine, also induced the expression of CCL5 in melanoma cells. Clinically, a positive correlation between CCL5 and NK cell marker NKp46 expression was found in melanoma patients, and a high expression level of CCL5 was correlated with a significant improvement of melanoma patients’ survival. We believe that this study highlights the impact of targeting autophagy on the tumor infiltration by NK cells and its benefit as a novel therapeutic approach to improve NK-based immunotherapy.
Collapse
|
29
|
Wagner JA, Rosario M, Romee R, Berrien-Elliott MM, Schneider SE, Leong JW, Sullivan RP, Jewell BA, Becker-Hapak M, Schappe T, Abdel-Latif S, Ireland AR, Jaishankar D, King JA, Vij R, Clement D, Goodridge J, Malmberg KJ, Wong HC, Fehniger TA. CD56bright NK cells exhibit potent antitumor responses following IL-15 priming. J Clin Invest 2017; 127:4042-4058. [PMID: 28972539 DOI: 10.1172/jci90387] [Citation(s) in RCA: 216] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 08/15/2017] [Indexed: 12/12/2022] Open
Abstract
NK cells, lymphocytes of the innate immune system, are important for defense against infectious pathogens and cancer. Classically, the CD56dim NK cell subset is thought to mediate antitumor responses, whereas the CD56bright subset is involved in immunomodulation. Here, we challenge this paradigm by demonstrating that brief priming with IL-15 markedly enhanced the antitumor response of CD56bright NK cells. Priming improved multiple CD56bright cell functions: degranulation, cytotoxicity, and cytokine production. Primed CD56bright cells from leukemia patients demonstrated enhanced responses to autologous blasts in vitro, and primed CD56bright cells controlled leukemia cells in vivo in a murine xenograft model. Primed CD56bright cells from multiple myeloma (MM) patients displayed superior responses to autologous myeloma targets, and furthermore, CD56bright NK cells from MM patients primed with the IL-15 receptor agonist ALT-803 in vivo displayed enhanced ex vivo functional responses to MM targets. Effector mechanisms contributing to IL-15-based priming included improved cytotoxic protein expression, target cell conjugation, and LFA-1-, CD2-, and NKG2D-dependent activation of NK cells. Finally, IL-15 robustly stimulated the PI3K/Akt/mTOR and MEK/ERK pathways in CD56bright compared with CD56dim NK cells, and blockade of these pathways attenuated antitumor responses. These findings identify CD56bright NK cells as potent antitumor effectors that warrant further investigation as a cancer immunotherapy.
Collapse
Affiliation(s)
- Julia A Wagner
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Maximillian Rosario
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Rizwan Romee
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Melissa M Berrien-Elliott
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Stephanie E Schneider
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jeffrey W Leong
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Ryan P Sullivan
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Brea A Jewell
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Michelle Becker-Hapak
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Timothy Schappe
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Sara Abdel-Latif
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Aaron R Ireland
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Devika Jaishankar
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Justin A King
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Ravi Vij
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Dennis Clement
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Radiumhospitalet, Oslo, Norway.,The KG Jebsen Centre for Cancer Immunotherapy, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Jodie Goodridge
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Radiumhospitalet, Oslo, Norway
| | - Karl-Johan Malmberg
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Radiumhospitalet, Oslo, Norway.,The KG Jebsen Centre for Cancer Immunotherapy, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Centre for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | | | - Todd A Fehniger
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
30
|
Lowry LE, Zehring WA. Potentiation of Natural Killer Cells for Cancer Immunotherapy: A Review of Literature. Front Immunol 2017; 8:1061. [PMID: 28919894 PMCID: PMC5585139 DOI: 10.3389/fimmu.2017.01061] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 08/15/2017] [Indexed: 11/14/2022] Open
Abstract
It is widely acknowledged that the human immune system plays a crucial role in preventing the formation and progression of innumerable types of cancer (1). The mechanisms by which this occurs are numerous, including contributions from both the innate and adaptive immune systems. As such, immunotherapy has long been believed to be an auspicious solution in the treatment of malignancy (2). Recent research has highlighted the promise of natural killer (NK) cells as a more directed immunotherapy approach. This paper will focus on the methods of potentiation of NK cells for their use in cancer therapy.
Collapse
Affiliation(s)
- Lacy E Lowry
- Department of Basic Science, Geisinger Commonwealth School of Medicine, Scranton, PA, United States
| | - William A Zehring
- Department of Biochemistry, Geisinger Commonwealth School of Medicine, Scranton, PA, United States
| |
Collapse
|
31
|
Koch J, Tesar M. Recombinant Antibodies to Arm Cytotoxic Lymphocytes in Cancer Immunotherapy. Transfus Med Hemother 2017; 44:337-350. [PMID: 29070979 PMCID: PMC5649249 DOI: 10.1159/000479981] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 08/01/2017] [Indexed: 12/18/2022] Open
Abstract
Immunotherapy has the potential to support and expand the body's own armamentarium of immune effector functions, which have been circumvented during malignant transformation and establishment of cancer and is presently considered to be the most promising treatment option for cancer patients. Recombinant antibody technologies have led to a multitude of novel antibody formats, which are in clinical development and hold great promise for future therapies. Among these formats, bispecific antibodies are extremely versatile due to their high efficacy to recruit and activate anti-tumoral immune effector cells, their excellent safety profile, and the opportunity for use in combination with cellular therapies. This review article summarizes the latest developments in cancer immunotherapy using immuno-engagers for recruiting T cells and NK cells to the tumor site. In addition to antibody formats, malignant cell targets, and immune cell targets, opportunities for combination therapies, including check point inhibitors, cytokines and adoptive transfer of immune cells, will be summarized and discussed.
Collapse
Affiliation(s)
- Joachim Koch
- Affimed GmbH, Technologiepark, Heidelberg, Germany
| | | |
Collapse
|
32
|
Kim JS, Shin BR, Lee HK, Lee JH, Kim KH, Choi JE, Ji AY, Hong JT, Kim Y, Han SB. Cd226-/- natural killer cells fail to establish stable contacts with cancer cells and show impaired control of tumor metastasis in vivo. Oncoimmunology 2017; 6:e1338994. [PMID: 28920003 DOI: 10.1080/2162402x.2017.1338994] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 05/30/2017] [Accepted: 06/02/2017] [Indexed: 12/21/2022] Open
Abstract
CD226 is an activating receptor expressed on natural killer (NK) cells, CD8+ T cells, and other immune cells. Upon binding to its ligands expressed on target cells, CD226 activates intracellular signaling that triggers cytokine production and degranulation in NK cells. However, the role of CD226 in contact dynamics between NK and cancer cells has remained unclear. Our time-lapse images showed that individual wild-type CD226+ NK cells contacted B16F10 melanoma cells for 23.7 min, but Cd226-/- NK cells only for 12.8 min, although both NK cell subsets showed equal contact frequency over 4 h. On the surface of B16F10 cells, CD226+ cells stayed at the same site with oscillating movement (named stable contact), while Cd226-/- NK cells moved around at a velocity of 4 μm/min (named unstable contact). Consequently, Cd226-/- NK cells did not kill B16F10 cells in vitro and did not inhibit their metastasis into the lung in vivo. Taken together, our data demonstrate that CD226 enables prolonged stable interaction between NK and cancer cells, which is needed for efficient killing of cancer cells.
Collapse
Affiliation(s)
- Ji Sung Kim
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Bo Ram Shin
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Hong Kyung Lee
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Jae Hee Lee
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Ki Hun Kim
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Jeong Eun Choi
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - A Young Ji
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Jin Tae Hong
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Youngsoo Kim
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Sang-Bae Han
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| |
Collapse
|
33
|
Rakhmilevich AL, Felder M, Lever L, Slowinski J, Rasmussen K, Hoefges A, Van De Voort TJ, Loibner H, Korman AJ, Gillies SD, Sondel PM. Effective Combination of Innate and Adaptive Immunotherapeutic Approaches in a Mouse Melanoma Model. THE JOURNAL OF IMMUNOLOGY 2017; 198:1575-1584. [PMID: 28062694 DOI: 10.4049/jimmunol.1601255] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 12/02/2016] [Indexed: 01/06/2023]
Abstract
Most cancer immunotherapies include activation of either innate or adaptive immune responses. We hypothesized that the combined activation of both innate and adaptive immunity will result in better antitumor efficacy. We have previously shown the synergy of an agonistic anti-CD40 mAb (anti-CD40) and CpG-oligodeoxynucleotides in activating macrophages to induce tumor cell killing in mice. Separately, we have shown that a direct intratumoral injection of immunocytokine (IC), an anti-GD2 Ab linked to IL-2, can activate T and NK cells resulting in antitumor effects. We hypothesized that activation of macrophages with anti-CD40/CpG, and NK cells with IC, would cause innate tumor destruction, leading to increased presentation of tumor Ags and adaptive T cell activation; the latter could be further augmented by anti-CTLA-4 Ab to achieve tumor eradication and immunological memory. Using the mouse GD2+ B78 melanoma model, we show that anti-CD40/CpG treatment led to upregulation of T cell activation markers in draining lymph nodes. Anti-CD40/CpG + IC/anti-CTLA-4 synergistically induced regression of advanced s.c. tumors, resulting in cure of some mice and development of immunological memory against B78 and wild type B16 tumors. Although the antitumor effect of anti-CD40/CpG did not require T cells, the antitumor effect of IC/anti-CTLA-4 was dependent on T cells. The combined treatment with anti-CD40/CpG + IC/anti-CTLA-4 reduced T regulatory cells in the tumors and was effective against distant solid tumors and lung metastases. We suggest that a combination of anti-CD40/CpG and IC/anti-CTLA-4 should be developed for clinical testing as a potentially effective novel immunotherapy strategy.
Collapse
Affiliation(s)
- Alexander L Rakhmilevich
- Department of Human Oncology, University of Wisconsin, Madison, WI 53705; .,Paul P. Carbone Comprehensive Cancer Center, Madison, WI 53705
| | - Mildred Felder
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, WI 53705
| | - Lauren Lever
- Department of Human Oncology, University of Wisconsin, Madison, WI 53705
| | - Jacob Slowinski
- Department of Human Oncology, University of Wisconsin, Madison, WI 53705
| | - Kayla Rasmussen
- Department of Human Oncology, University of Wisconsin, Madison, WI 53705
| | - Anna Hoefges
- Department of Human Oncology, University of Wisconsin, Madison, WI 53705
| | | | | | - Alan J Korman
- Bristol-Myers Squibb Company, Redwood City, CA 94063
| | | | - Paul M Sondel
- Department of Human Oncology, University of Wisconsin, Madison, WI 53705.,Paul P. Carbone Comprehensive Cancer Center, Madison, WI 53705.,Department of Pediatrics, University of Wisconsin, Madison, WI 53705
| |
Collapse
|
34
|
Fehniger TA, Cooper MA. Harnessing NK Cell Memory for Cancer Immunotherapy. Trends Immunol 2016; 37:877-888. [PMID: 27773685 DOI: 10.1016/j.it.2016.09.005] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 09/24/2016] [Accepted: 09/26/2016] [Indexed: 12/16/2022]
Abstract
Due to their ability to kill cancer cells and produce proinflammatory cytokines, natural killer (NK) cells have long been of clinical interest for their antitumor properties. The recent discovery of NK cell memory demonstrates that NK cell functions, and potentially antitumor responses, can be enhanced long term. Following nonspecific activation with the cytokines IL-12, IL-15, and IL-18 or in response to antigens or cytomegalovirus (CMV), human and mouse NK cells exhibit stable, enhanced functional responses with phenotypic and molecular changes. Here we review mechanisms driving the differentiation of NK cell memory-like properties, evidence for antitumor activity, and the challenges and opportunities in harnessing memory-like NK cells for cancer immunotherapy.
Collapse
Affiliation(s)
- Todd A Fehniger
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Megan A Cooper
- Department of Pediatrics, Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
35
|
Maghazachi AA, Sand KL, Al-Jaderi Z. Glatiramer Acetate, Dimethyl Fumarate, and Monomethyl Fumarate Upregulate the Expression of CCR10 on the Surface of Natural Killer Cells and Enhance Their Chemotaxis and Cytotoxicity. Front Immunol 2016; 7:437. [PMID: 27807435 PMCID: PMC5069502 DOI: 10.3389/fimmu.2016.00437] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Accepted: 10/04/2016] [Indexed: 02/06/2023] Open
Abstract
In vitro harnessing of immune cells is the most important advance in the field of cancer immunotherapy. Results shown in the current paper may be used to harness natural killer (NK) cells in vitro. It is observed that drugs used to treat multiple sclerosis such as glatiramer acetate, dimethyl fumarate, and monomethyl fumarate upregulate the expression of chemokines receptor 10 (CCR10) on the surface of human IL-2-activated NK cells. This is corroborated with increased chemotaxis of these cells toward the concentration gradients of the ligands for CCR10, namely CCL27 and CCL28. It is also demonstrated that these three drugs enhance NK cell cytotoxicity against tumor target cells, an activity that is abrogated by prior incubation of the cells with anti-CCR10 antibody. Because CCL27 and CCL28 are secreted by selective tumor types such as malignant melanoma, squamous cell carcinomas, and colorectal cancer, respectively, it is hypothesized that activated NK cells may be harnessed in vitro with any of these drugs before utilizing them as a therapeutic modality for cancer.
Collapse
Affiliation(s)
- Azzam A. Maghazachi
- Department of Clinical Sciences, College of Medicine, and the Sharjah Institute for Medical Research (SIMR), University of Sharjah, Sharjah, United Arab Emirates
| | | | - Zaidoon Al-Jaderi
- Department of Clinical Sciences, College of Medicine, and the Sharjah Institute for Medical Research (SIMR), University of Sharjah, Sharjah, United Arab Emirates
- University of Oslo, Oslo, Norway
| |
Collapse
|
36
|
Abstract
Natural killer cells are a diverse group of innate lymphocytes that are specialized to rapidly respond to cancerous or virus-infected cells. NK cell function is controlled by the integration of signals from activating and inhibitory receptors expressed at the cell surface. Variegated expression patterns of these activating and inhibitory receptors at the single cell level leads to a highly diverse NK cell repertoire. Here I review the factors that influence NK cell repertoire diversity and its functional consequences for our ability to fight viruses.
Collapse
Affiliation(s)
- Catherine A. Blish
- Department of Medicine, Division of Infectious Diseases and Geographic Medicine
- Immunology Program, Stanford University School of Medicine, Stanford, California
| |
Collapse
|