1
|
Boonchalermvichian C, Yan H, Gupta B, Rubin A, Baker J, Negrin RS. invariant Natural Killer T cell therapy as a novel therapeutic approach in hematological malignancies. FRONTIERS IN TRANSPLANTATION 2024; 3:1353803. [PMID: 38993780 PMCID: PMC11235242 DOI: 10.3389/frtra.2024.1353803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 03/04/2024] [Indexed: 07/13/2024]
Abstract
Invariant Natural Killer T cell therapy is an emerging platform of immunotherapy for cancer treatment. This unique cell population is a promising candidate for cell therapy for cancer treatment because of its inherent cytotoxicity against CD1d positive cancers as well as its ability to induce host CD8 T cell cross priming. Substantial evidence supports that iNKT cells can modulate myelomonocytic populations in the tumor microenvironment to ameliorate immune dysregulation to antagonize tumor progression. iNKT cells can also protect from graft-versus-host disease (GVHD) through several mechanisms, including the expansion of regulatory T cells (Treg). Ultimately, iNKT cell-based therapy can retain antitumor activity while providing protection against GVHD simultaneously. Therefore, these biological properties render iNKT cells as a promising "off-the-shelf" therapy for diverse hematological malignancies and possible solid tumors. Further the introduction of a chimeric antigen recetor (CAR) can further target iNKT cells and enhance function. We foresee that improved vector design and other strategies such as combinatorial treatments with small molecules or immune checkpoint inhibitors could improve CAR iNKT in vivo persistence, functionality and leverage anti-tumor activity along with the abatement of iNKT cell dysfunction or exhaustion.
Collapse
|
2
|
Mooney AH, Draper SL, Burn OK, Anderson RJ, Compton BJ, Tang C, Farrand KJ, Di Lucia P, Ravà M, Fumagalli V, Giustini L, Bono E, Godfrey DI, Heath WR, Yuan W, Chisari FV, Guidotti LG, Iannacone M, Sidney J, Sette A, Gulab SA, Painter GF, Hermans IF. Preclinical evaluation of therapeutic vaccines for chronic hepatitis B that stimulate antiviral activities of T cells and NKT cells. JHEP Rep 2024; 6:101038. [PMID: 38694959 PMCID: PMC11061331 DOI: 10.1016/j.jhepr.2024.101038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 02/01/2024] [Accepted: 02/06/2024] [Indexed: 05/04/2024] Open
Abstract
Background & Aims Liver diseases resulting from chronic HBV infection are a significant cause of morbidity and mortality. Vaccines that elicit T-cell responses capable of controlling the virus represent a treatment strategy with potential for long-term effects. Here, we evaluated vaccines that induce the activity of type I natural killer T (NKT) cells to limit viral replication and license stimulation of conventional antiviral T-cells. Methods Vaccines were prepared by conjugating peptide epitopes to an NKT-cell agonist to promote co-delivery to antigen-presenting cells, encouraging NKT-cell licensing and stimulation of T cells. Activity of the conjugate vaccines was assessed in transgenic mice expressing the complete HBV genome, administered intravenously to maximise access to NKT cell-rich tissues. Results The vaccines induced only limited antiviral activity in unmanipulated transgenic hosts, likely attributable to NKT-cell activation as T-cell tolerance to viral antigens is strong. However, in a model of chronic hepatitis B involving transfer of naive HBcAg-specific CD8+ T cells into the transgenic mice, which typically results in specific T-cell dysfunction without virus control, vaccines containing the targeted HBcAg epitope induced prolonged antiviral activity because of qualitatively improved T-cell stimulation. In a step towards a clinical product, vaccines were prepared using synthetic long peptides covering clusters of known HLA-binding epitopes and shown to be immunogenic in HLA transgenic mice. Predictions based on HLA distribution suggest a product containing three selected SLP-based vaccines could give >90 % worldwide coverage, with an average of 3.38 epitopes targeted per individual. Conclusions The novel vaccines described show promise for further clinical development as a treatment for chronic hepatitis B. Impact and Implications Although there are effective prophylactic vaccines for HBV infection, it is estimated that 350-400 million people worldwide have chronic hepatitis B, putting these individuals at significant risk of life-threatening liver diseases. Therapeutic vaccination aimed at activating or boosting HBV-specific T-cell responses holds potential as a strategy for treating chronic infection, but has so far met with limited success. Here, we show that a glycolipid-peptide conjugate vaccine designed to coordinate activity of type I NKT cells alongside conventional antiviral T cells has antiviral activity in a mouse model of chronic infection. It is anticipated that a product based on a combination of three such conjugates, each prepared using long peptides covering clusters of known HLA-binding epitopes, could be developed further as a treatment for chronic hepatitis B with broad global HLA coverage.
Collapse
Affiliation(s)
- Anna H. Mooney
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Sarah L. Draper
- Ferrier Research Institute, Victoria University of Wellington, Wellington, New Zealand
| | - Olivia K. Burn
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Regan J. Anderson
- Ferrier Research Institute, Victoria University of Wellington, Wellington, New Zealand
| | - Benjamin J. Compton
- Ferrier Research Institute, Victoria University of Wellington, Wellington, New Zealand
| | - Chingwen Tang
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | | | - Pietro Di Lucia
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Micol Ravà
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Valeria Fumagalli
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Leonardo Giustini
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Elisa Bono
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Dale I. Godfrey
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Victoria, Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Australia
| | - William R. Heath
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Victoria, Australia
| | - Weiming Yuan
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Francis V. Chisari
- Department of Immunology & Microbial Sciences, The Scripps Research Institute, La Jolla, CA, USA
| | - Luca G. Guidotti
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Matteo Iannacone
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - John Sidney
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Shivali A. Gulab
- Ferrier Research Institute, Victoria University of Wellington, Wellington, New Zealand
- Avalia Immunotherapies Limited, Wellington, New Zealand
| | - Gavin F. Painter
- Ferrier Research Institute, Victoria University of Wellington, Wellington, New Zealand
| | - Ian F. Hermans
- Malaghan Institute of Medical Research, Wellington, New Zealand
| |
Collapse
|
3
|
Hammond TC, Purbhoo MA, Kadel S, Ritz J, Nikiforow S, Daley H, Shaw K, van Besien K, Gomez-Arteaga A, Stevens D, Ortuzar W, Michelet X, Smith R, Moskowitz D, Masakayan R, Yigit B, Boi S, Soh KT, Chamberland J, Song X, Qin Y, Mishchenko I, Kirby M, Nasonenko V, Buffa A, Buell JS, Chand D, van Dijk M, Stebbing J, Exley MA. A phase 1/2 clinical trial of invariant natural killer T cell therapy in moderate-severe acute respiratory distress syndrome. Nat Commun 2024; 15:974. [PMID: 38321023 PMCID: PMC10847411 DOI: 10.1038/s41467-024-44905-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 01/09/2024] [Indexed: 02/08/2024] Open
Abstract
Invariant natural killer T (iNKT) cells, a unique T cell population, lend themselves for use as adoptive therapy due to diverse roles in orchestrating immune responses. Originally developed for use in cancer, agenT-797 is a donor-unrestricted allogeneic ex vivo expanded iNKT cell therapy. We conducted an open-label study in virally induced acute respiratory distress syndrome (ARDS) caused by the severe acute respiratory syndrome-2 virus (trial registration NCT04582201). Here we show that agenT-797 rescues exhausted T cells and rapidly activates both innate and adaptive immunity. In 21 ventilated patients including 5 individuals receiving veno-venous extracorporeal membrane oxygenation (VV-ECMO), there are no dose-limiting toxicities. We observe an anti-inflammatory systemic cytokine response and infused iNKT cells are persistent during follow-up, inducing only transient donor-specific antibodies. Clinical signals of associated survival and prevention of secondary infections are evident. Cellular therapy using off-the-shelf iNKT cells is safe, can be rapidly scaled and is associated with an anti-inflammatory response. The safety and therapeutic potential of iNKT cells across diseases including infections and cancer, warrants randomized-controlled trials.
Collapse
Affiliation(s)
- Terese C Hammond
- Pulmonary Critical Care Sleep Medicine, Providence Saint John's Health Center, Santa Monica, CA, USA
- David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | | | | | - Jerome Ritz
- Dana Farber Cancer Institute, Boston, MA, USA
| | | | | | - Kit Shaw
- Dana Farber Cancer Institute, Boston, MA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Yu Qin
- MiNK Therapeutics, Lexington, MA, USA
- Agenus, Lexington, MA, USA
| | | | | | | | - Alexa Buffa
- MiNK Therapeutics, Lexington, MA, USA
- Agenus, Lexington, MA, USA
| | | | | | | | | | | |
Collapse
|
4
|
Lee MS, Webb TJ. Novel lipid antigens for NKT cells in cancer. Front Immunol 2023; 14:1173375. [PMID: 37908366 PMCID: PMC10613688 DOI: 10.3389/fimmu.2023.1173375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 10/02/2023] [Indexed: 11/02/2023] Open
Abstract
Cancer immunotherapy aims to unleash the power of the immune system against tumors without the side effects of traditional chemotherapy. Immunotherapeutic methods vary widely, but all follow the same basic principle: overcome the barriers utilized by cancers to avoid immune destruction. These approaches often revolve around classical T cells, such as with CAR T cells and neoantigen vaccines; however, the utility of the innate-like iNKT cell in cancer immunotherapy has gained significant recognition. iNKT cells parallel classic T cell recognition of peptide antigens presented on MHC through their recognition of lipid antigens presented on the MHC I-like molecule CD1d. Altered metabolism and a lipogenic phenotype are essential properties of tumor cells, representing a unique feature that may be exploited by iNKT cells. In this review, we will cover properties of iNKT cells, CD1d, and lipid antigen presentation. Next, we will discuss the cancer lipidome and how it may be exploited by iNKT cells through a window of opportunity. Finally, we will review, in detail, novel lipid antigens for iNKT cells in cancer.
Collapse
Affiliation(s)
- Michael S. Lee
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Tonya J. Webb
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
5
|
Ahmed K, Jha S. Oncoviruses: How do they hijack their host and current treatment regimes. Biochim Biophys Acta Rev Cancer 2023; 1878:188960. [PMID: 37507056 DOI: 10.1016/j.bbcan.2023.188960] [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: 03/20/2023] [Revised: 07/05/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023]
Abstract
Viruses have the ability to modulate the cellular machinery of their host to ensure their survival. While humans encounter numerous viruses daily, only a select few can lead to disease progression. Some of these viruses can amplify cancer-related traits, particularly when coupled with factors like immunosuppression and co-carcinogens. The global burden of cancer development resulting from viral infections is approximately 12%, and it arises as an unfortunate consequence of persistent infections that cause chronic inflammation, genomic instability from viral genome integration, and dysregulation of tumor suppressor genes and host oncogenes involved in normal cell growth. This review provides an in-depth discussion of oncoviruses and their strategies for hijacking the host's cellular machinery to induce cancer. It delves into how viral oncogenes drive tumorigenesis by targeting key cell signaling pathways. Additionally, the review discusses current therapeutic approaches that have been approved or are undergoing clinical trials to combat malignancies induced by oncoviruses. Understanding the intricate interactions between viruses and host cells can lead to the development of more effective treatments for virus-induced cancers.
Collapse
Affiliation(s)
- Kainat Ahmed
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA
| | - Sudhakar Jha
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA.
| |
Collapse
|
6
|
Wang J, Cheng X, Jin Y, Xia B, Qin R, Zhang W, Hu H, Mao X, Zhou L, Yan J, Zhang X, Xu J. Safety and Clinical Response to Combined Immunotherapy with Autologous iNKT Cells and PD-1 +CD8 + T Cells in Patients Failing First-line Chemotherapy in Stage IV Pancreatic Cancer. CANCER RESEARCH COMMUNICATIONS 2023; 3:991-1003. [PMID: 37377605 PMCID: PMC10246506 DOI: 10.1158/2767-9764.crc-23-0137] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 04/11/2023] [Accepted: 05/15/2023] [Indexed: 06/29/2023]
Abstract
Purpose A phase I clinical trial was conducted to assess the safety and feasibility of invariant natural killer T (iNKT) cells combined with PD-1+CD8+ T cells in patients with advanced pancreatic cancer and failing the first-line chemotherapy. Patients and Methods Fifteen eligible patients were enrolled, of whom 9 received at least three cycles of treatment each. In total, 59 courses were administered. Results Fever was the most common adverse event, peaking at about 2-4 hours after cell infusion and reverting within 24 hours without treatment in all patients. Influenza-like reactions such as headache, myalgia, and arthralgia were also observed in 4, 4, and 3 of the patients, respectively. In addition, vomiting and dizziness were prevalent, while abdominal pain, chest pain, rash, and stuffy nose were rare adverse events, each reported in 1 patient. Side effects above grade 2 were not observed. Two patients achieved partial regression, while 1 patient experienced disease progression assessed 4 weeks after the third course. Three patients are still alive at the time of writing and have progression-free survival longer than 12 months. The overall survival time has been extended to over 12 months in 6 of the 9 patients. No constant changes of CD4+ T, B, and NK cells were recorded except for elevated CD8+ T cells after the first course. Conclusions The combination of autologous iNKT cells and PD-1+CD8+ T cells was a safe therapeutic strategy against advanced pancreatic cancer. The patients exhibited a potentially promising prolonged survival time. Further study appears warranted to evaluate the efficacy of these combined cell infusions in pancreatic cancer. Trial registration This trial was included in the clinical trial which was registered in ClinicalTrials.gov (ID:NCT03093688) on March 15, 2017. Significance There is an unmet need for novel, more effective, and tolerable therapies for pancreatic cancer. Here we present a phase I clinical trial employing iNKT cells combined with PD-1+CD8+ T cells in 9 patients with advanced pancreatic cancer and failing the first-line chemotherapy. The combined immunotherapy was shown to be feasible in the enrolled patients with limited side effects and optimistic clinical responses, which could bring opportunity of therapeutic advancement.
Collapse
Affiliation(s)
- Jing Wang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, P.R. China
| | - Xiaobo Cheng
- Clinical Research Center, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, P.R. China
| | - Yanling Jin
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, P.R. China
| | - Bili Xia
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, P.R. China
| | - Ran Qin
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, P.R. China
| | - Wei Zhang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, P.R. China
| | - Huiliang Hu
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, P.R. China
| | - Xiaoting Mao
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, P.R. China
| | - Liting Zhou
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, P.R. China
| | - Jia Yan
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, P.R. China
| | - Xiaoyan Zhang
- Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, P.R. China
- Clinical Center for Biotherapy, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, P.R. China
- Clinical Center for Biotherapy, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Jianqing Xu
- Shanghai Public Health Clinical Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, P.R. China
- Clinical Center for Biotherapy, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, P.R. China
- Clinical Center for Biotherapy, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| |
Collapse
|
7
|
Luo Y, Luo X, Zhang T, Li S, Liu S, Ma Y, Wang Z, Jin X, Liu J, Wang X. Anti-Tumor Secondary Metabolites Originating from Fungi in the South China Sea's Mangrove Ecosystem. BIOENGINEERING (BASEL, SWITZERLAND) 2022; 9:bioengineering9120776. [PMID: 36550982 PMCID: PMC9774444 DOI: 10.3390/bioengineering9120776] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/14/2022] [Accepted: 11/28/2022] [Indexed: 12/13/2022]
Abstract
A mangrove is a unique ecosystem with abundant resources, in which fungi are an indispensable microbial part. Numerous mangrove fungi-derived secondary metabolites are considerable sources of novel bioactive substances, such as polyketides, terpenoids, alkaloids, peptides, etc., which arouse people's interest in the search for potential natural anti-tumor drugs. This review includes a total of 44 research publications that described 110 secondary metabolites that were all shown to be anti-tumor from 39 mangrove fungal strains belonging to 18 genera that were acquired from the South China Sea between 2016 and 2022. To identify more potential medications for clinical tumor therapy, their sources, unique structures, and cytotoxicity qualities were compiled. This review could serve as a crucial resource for the research status of mangrove fungal-derived natural products deserving of further development.
Collapse
Affiliation(s)
- Yuyou Luo
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Xiongming Luo
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Tong Zhang
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Siyuan Li
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Shuping Liu
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yuxin Ma
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Zongming Wang
- Pituitary Tumor Center, Department of Neurosurgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Xiaobao Jin
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jing Liu
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Correspondence: (J.L.); (X.W.); Tel.: +86-134-2412-4716 (J.L.); +86-20-39352189 (X.W.)
| | - Xin Wang
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Correspondence: (J.L.); (X.W.); Tel.: +86-134-2412-4716 (J.L.); +86-20-39352189 (X.W.)
| |
Collapse
|
8
|
Varghese B, Lynch L, Vriend LE, Draganov D, Clark JM, Kissick HT, Varghese S, Sanda MG, Dranoff G, Arredouani MS, Balk SP, Exley MA. Invariant NKT cell-augmented GM-CSF-secreting tumor vaccine is effective in advanced prostate cancer model. Cancer Immunol Immunother 2022; 71:2943-2955. [PMID: 35523889 PMCID: PMC10992623 DOI: 10.1007/s00262-022-03210-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 04/08/2022] [Indexed: 10/18/2022]
Abstract
Invariant natural killer T cells (iNKT cells) express a semi-invariant T cell receptor that recognizes certain glycolipids (including α-galactosylceramide, αGC) bound to CD1d, and can induce potent antitumor responses. Here, we assessed whether αGC could enhance the efficacy of a GM-CSF-producing tumor cell vaccine in the transgenic SV40 T antigen-driven TRAMP prostate cancer model. In healthy mice, we initially found that optimal T cell responses were obtained with αGC-pulsed TRAMP-C2 cells secreting GM-CSF and milk fat globule epidermal growth factor protein-8 (MFG-E8) with an RGD to RGE mutation (GM-CSF/RGE TRAMP-C2), combined with systemic low dose IL-12. In a therapeutic model, transgenic TRAMP mice were then castrated at ~ 20 weeks, followed by treatment with the combination vaccine. Untreated mice succumbed to tumor by ~ 40 weeks, but survival was markedly prolonged by vaccine treatment, with most mice surviving past 80 weeks. Prostates in the treated mice were heavily infiltrated with T cells and iNKT cells, which both secreted IFNγ in response to tumor cells. The vaccine was not effective if the αGC, IL-12, or GM-CSF secretion was eliminated. Finally, immunized mice were fully resistant to challenge with TRAMP-C2 cells. Together these findings support further development of therapeutic vaccines that exploit iNKT cell activation.
Collapse
Affiliation(s)
- Bindu Varghese
- Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA, 02215, USA
- Sana Biotechnology Inc., Boston, MA, USA
| | - Lydia Lynch
- Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA, 02215, USA
- Brigham and Women's Hospital, 75 Francis St., NRB 6, Boston, MA, 02115, USA
| | - Lianne E Vriend
- Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA, 02215, USA
| | - Dobrin Draganov
- Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Sanofi Inc., San Diego, CA, USA
| | - Justice M Clark
- Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA, 02215, USA
| | - Haydn T Kissick
- Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA, 02215, USA
- Emory University, Atlanta, GA, USA
| | - Sharlin Varghese
- Medical Center School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA
| | - Martin G Sanda
- Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA, 02215, USA
- Emory University, Atlanta, GA, USA
| | - Glenn Dranoff
- Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Novartis Biomedical Institutes of Research, Cambridge, MA, USA
| | - M Simo Arredouani
- Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA, 02215, USA
- Intellia Inc., Cambridge, MA, USA
| | - Steven P Balk
- Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA, 02215, USA.
| | - Mark A Exley
- Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA, 02215, USA.
- Brigham and Women's Hospital, 75 Francis St., NRB 6, Boston, MA, 02115, USA.
- Imvax Inc., Philadelphia, PA, USA.
- University of Manchester, Manchester, UK.
- MiNK Therapeutics Inc., New York, NY, USA.
| |
Collapse
|
9
|
Li H, Mao H, Chen C, Xu Y, Meng S, Sun T, Zong C. Efficient synthesis of α-galactosylceramide and its C-6 modified analogs. Front Chem 2022; 10:1039731. [PMID: 36505742 PMCID: PMC9732566 DOI: 10.3389/fchem.2022.1039731] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 11/07/2022] [Indexed: 11/27/2022] Open
Abstract
The synthesis of α-galactosylceramide (KRN7000) and its C-6 modified analogs remains a challenge due to the difficult α-1,2-cis-glycosidic bond. A non-participating benzyl (Bn) protecting group has been commonly used to favor the α-glycosylation product. Here, we report the α-selective glycosylation by using a bulky 4,6-O-di-tert-butylsilylene (DTBS) galactosyl donor, regardless of the 2-benzoyl (Bz) participating group. Compared with Bn, Bz groups can be selectively removed in basic conditions without impacting the C-6 azide modification. The azide has the potential for clicking with alkyne or being easily transformed to other functional groups.
Collapse
Affiliation(s)
- Huiting Li
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, College of Marine Science, Hainan University, Haikou, China
| | - Hongzhao Mao
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, College of Marine Science, Hainan University, Haikou, China
| | | | - Ying Xu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, College of Marine Science, Hainan University, Haikou, China
| | - Shuai Meng
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, College of Marine Science, Hainan University, Haikou, China
| | - Tiantian Sun
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, College of Marine Science, Hainan University, Haikou, China
| | - Chengli Zong
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, College of Marine Science, Hainan University, Haikou, China,*Correspondence: Chengli Zong,
| |
Collapse
|
10
|
Potential role of Marine Bioactive Compounds targeting signaling pathways in cancer: A review. Eur J Pharmacol 2022; 936:175330. [DOI: 10.1016/j.ejphar.2022.175330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/12/2022] [Accepted: 10/13/2022] [Indexed: 11/23/2022]
|
11
|
Chu MJ, Li M, Ma H, Li PL, Li GQ. Secondary metabolites from marine sponges of the genus Agelas: a comprehensive update insight on structural diversity and bioactivity. RSC Adv 2022; 12:7789-7820. [PMID: 35424773 PMCID: PMC8982468 DOI: 10.1039/d1ra08765g] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 03/03/2022] [Indexed: 01/07/2023] Open
Abstract
As one of the most common marine sponges in tropical and subtropical oceans, the sponges of the genus Agelas, have emerged as unique and yet under-investigated pools for discovery of natural products with fabulous molecular diversity and myriad interesting biological activities. The present review highlights the chemical structure and biological activity of 355 compounds that have been isolated and characterized from the members of Agelas sponges, over the period of about five decades (from 1971 to November 2021). For a better understanding, these numerous compounds are firstly classified and presented according to their carbon skeleton as well as their biosynthetic origins. Relevant summaries focusing on the source organism and the associated bioactivity of these compounds belonging to different chemical classes are also provided. This review highlights sponges of the genus Agelas as exciting source for discovery of intriguing natural compounds.
Collapse
Affiliation(s)
- Mei-Jun Chu
- College of Veterinary Medicine, Qingdao Agricultural University Qingdao 266109 China
| | - Meng Li
- Department of Pharmacy, Qingdao Central Hospital Qingdao 266042 China
| | - He Ma
- College of Veterinary Medicine, Qingdao Agricultural University Qingdao 266109 China
| | - Ping-Lin Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China Qingdao 266003 China
- Laboratory of Marine Drugs and Biological Products, National Laboratory for Marine Science and Technology Qingdao 266235 China
| | - Guo-Qiang Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China Qingdao 266003 China
- Laboratory of Marine Drugs and Biological Products, National Laboratory for Marine Science and Technology Qingdao 266235 China
| |
Collapse
|
12
|
Cheng X, Wang J, Qiu C, Jin Y, Xia B, Qin R, Hu H, Yan J, Zhang X, Xu J. Feasibility of iNKT cell and PD-1+CD8+ T cell-based immunotherapy in patients with lung adenocarcinoma: Preliminary results of a phase I/II clinical trial. Clin Immunol 2022; 238:108992. [DOI: 10.1016/j.clim.2022.108992] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 02/16/2022] [Accepted: 03/26/2022] [Indexed: 01/08/2023]
|
13
|
Liu Y, Wang G, Chai D, Dang Y, Zheng J, Li H. iNKT: A new avenue for CAR-based cancer immunotherapy. Transl Oncol 2022; 17:101342. [PMID: 35063813 PMCID: PMC8784340 DOI: 10.1016/j.tranon.2022.101342] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 01/07/2022] [Indexed: 01/16/2023] Open
Abstract
Chimeric antigen receptor (CAR) T cell is a T lymphocyte-based immunotherapy, which achieves great successes in treating blood malignancies and provides new hope to cue advanced cancer patients. Invariant natural killer T (iNKT) cells are a kind of special T lymphocytes characterized by expressing invariant TCR of Vα24Vβ11 to recognize CD1d-presented glycolipid antigens, which bridge innate and adaptive immune responses. iNKT cells themselves show strong anti-tumor effect in tumor models via CD1d-mediated killing of CD1d-positive tumor cells and immunosuppressive TAMs and MDSCs, and are closely related to the prognosis of cancer patients. iNKT cells are not restricted to polymorphic human leukocyte antigen (HLA) and can prevent Graft versus Host Disease (GvHD), which makes it to be an ideal CAR vector for allogeneic therapy. Although CAR-iNKT was developed and verified by several different teams and attracts more and more attentions, many obstacles are still needed to be resolved before obtaining CAR-iNKT therapeutics. In this review, we summarized the current status of clinical application of iNKT cells and the latest achievements of CAR-iNKT cells, which provides new insight in CAR-iNKT development and usages.
Collapse
Affiliation(s)
- Yilin Liu
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China
| | - Gang Wang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China
| | - Dafei Chai
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China
| | - Yuanyuan Dang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China
| | - Junnian Zheng
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China.
| | - Huizhong Li
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China.
| |
Collapse
|
14
|
Gao Y, Guo J, Bao X, Xiong F, Ma Y, Tan B, Yu L, Zhao Y, Lu J. Adoptive Transfer of Autologous Invariant Natural Killer T Cells as Immunotherapy for Advanced Hepatocellular Carcinoma: A Phase I Clinical Trial. Oncologist 2021; 26:e1919-e1930. [PMID: 34255901 PMCID: PMC8571770 DOI: 10.1002/onco.13899] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 07/08/2021] [Indexed: 12/17/2022] Open
Abstract
Lessons Learned Administration of autologous invariant natural killer T (iNKT) cells was safe and well‐tolerated in patients with hepatocellular carcinoma (Barcelona Clinic Liver Cancer stage B/C). Expanded iNKT cells produced T‐helper 1–like responses with possible antitumor activity. No severe adverse events were observed in any of the enrolled patients, including one patient who received 1010 in vitro–expanded autologous iNKT cells as a single infusion.
Background Invariant natural killer T cells co‐express T‐cell antigen receptor and natural killer (NK) cell receptors. Invariant natural killer T (iNKT) cells exhibit antitumor activity, but their numbers and functions are impaired in patients with hepatocellular carcinoma (HCC). The adoptive transfer of iNKT cells might treat advanced HCC. Methods This phase I study (NCT03175679) enrolled 10 patients with HCC (Barcelona Clinic Liver Cancer [BCLC] stage B/C) at Beijing YouAn Hospital (April 2017 to May 2018). iNKT cells isolated from peripheral blood mononuclear cells (PBMCs) were expanded and alpha‐galactosylceramide (α‐GalCer)–pulsed. Dosage escalated from 3 × 107 to 6 × 107 to 9 × 107 cells/m2 (3+3 design). An exploratory dose trial (1 × 1010 cells/m2) was conducted in one patient. Results Expanded iNKT cells produced greater quantities of T‐helper 1 (Th1) cytokines (e.g., interferon‐gamma, perforin, and granzyme B) but less interleukin‐4 than nonexpanded iNKT cells. Circulating numbers of iNKT cells and activated NK cells were increased after iNKT cell infusion. Most treatment‐related adverse events were grade 1–2, and three grade 3 adverse events were reported; all resolved without treatment. Four patients were progression‐free at 5.5, 6, 7, and 11 months after therapy, and one patient was alive and without tumor recurrence at the last follow‐up. Five patients died at 1.5 to 11 months after treatment. Conclusion Autologous iNKT cell treatment is safe and well‐tolerated. Expanded iNKT cells produce Th1‐like responses with possible antitumor activity. The antitumor effects of iNKT cell infusion in patients with advanced HCC merit further investigation.
Collapse
Affiliation(s)
- Yao Gao
- Hepatology and Cancer Biotherapy Ward, Beijing YouAn Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Jia Guo
- Hepatology and Cancer Biotherapy Ward, Beijing YouAn Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Xuli Bao
- Hepatology and Cancer Biotherapy Ward, Beijing YouAn Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Fang Xiong
- Hepatology and Cancer Biotherapy Ward, Beijing YouAn Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Yanpin Ma
- Hepatology and Cancer Biotherapy Ward, Beijing YouAn Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Bingqin Tan
- Hepatology and Cancer Biotherapy Ward, Beijing YouAn Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Lele Yu
- Hepatology and Cancer Biotherapy Ward, Beijing YouAn Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Yong Zhao
- The State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Jun Lu
- Hepatology and Cancer Biotherapy Ward, Beijing YouAn Hospital, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
15
|
Exploiting CD1-restricted T cells for clinical benefit. Mol Immunol 2021; 132:126-131. [PMID: 33582549 DOI: 10.1016/j.molimm.2020.12.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 12/07/2020] [Indexed: 01/11/2023]
Abstract
CD1-restricted T cells were first described over 30 years ago along with the cloning of the CD1 family. Around the same time, invariant Natural Killer cells (iNKT) were identified based on invariant TCR-alpha chains with additional expression of natural killer (NK) cell markers. About 5 years later, iNKT were shown to react with CD1d. Since then, iNKT have been shown to be a major population of CD1d-restricted T cells in humans and many animals. Like NK cells, iNKT are innate lymphocytes with rapid and wide-ranging effector potential. These activities include cytotoxicity and an unusually broad and high-level cytokine production. The development of highly-specific methods of isolating, stimulating, expanding or depleting these relatively rare cells and controlling their potent activities has stimulated considerable interest in therapeutic targeting of iNKT cells. Potential applications include cancers, inflammatory, infectious and autoimmune among other diseases. To date, most trials have targeted various cancers, there are 2 published trials in viral hepatitis and one in sickle cell lung disease. Uniform safety, evidence of immunologic activity and increasingly clinical efficacy have been seen. Approaches to targeting iNKT cells in clinical development include highly specific natural glycolipid ligands presented by CD1d and chemical analogues thereof and monoclonal antibody-based targeting of iNKT cells. In the case of iNKT cell-based therapies, novel approaches include arming them with Chimeric Antigen Receptors (CARs) and recombinant TCRs (rTCR), gene editing and allogeneic use. Controlling the iTCR:CD1d molecular interaction and consequences is a unique and promising therapeutic technology.
Collapse
|
16
|
Application of marine natural products in drug research. Bioorg Med Chem 2021; 35:116058. [PMID: 33588288 DOI: 10.1016/j.bmc.2021.116058] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/31/2021] [Accepted: 02/02/2021] [Indexed: 12/14/2022]
Abstract
New diseases are emerging as the environment changes, so drug manufacturers are always on the lookout for new resources to develop effective and safe drugs. In recent years, many bioactive substances have been produced in the marine environment, which represents an alternative resource for new drugs used to combat major diseases such as cancer or inflammation. Many marine-derived medicinal substances are in preclinical or early stage of clinical development, and some marine drugs have been put on the market, such as ET743 (Yondelis®). This review presents the sources, activities, mechanisms of action and syntheses of bioactive substances based on marine natural products in clinical trials and on the market, which is helpful to understand the progress of drug research by application of marine natural products.
Collapse
|
17
|
Li W, Guillaume J, Baqi Y, Wachsmann I, Gieselmann V, Van Calenbergh S, Müller CE. Synthesis and structure-activity relationships of cerebroside analogues as substrates of cerebroside sulphotransferase and discovery of a competitive inhibitor. J Enzyme Inhib Med Chem 2020; 35:1503-1512. [PMID: 32657203 PMCID: PMC7470129 DOI: 10.1080/14756366.2020.1791841] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 06/09/2020] [Accepted: 06/29/2020] [Indexed: 01/24/2023] Open
Abstract
Metachromatic leukodystrophy (MLD) is a rare genetic disease characterised by a dysfunction of the enzyme arylsulphatase A leading to the lysosomal accumulation of cerebroside sulphate (sulphatide) causing subsequent demyelination in patients. The enzyme galactosylceramide (cerebroside) sulphotransferase (CST) catalyses the transfer of a sulphate group from 3'-phosphoadenosine-5'-phosphosulphate (PAPS) to cerebrosides producing sulphatides. Substrate reduction therapy for arylsulphatase A by inhibition of CST was proposed as a promising therapeutic approach. To identify competitive CST inhibitors, we synthesised and investigated analogues of the substrate galactosylceramide with variations at the anomeric position, the acyl substituent and the carbohydrate moiety, and investigated their structure-activity relationships. While most of the compounds behaved as substrates, α-galactosylceramide 16 was identified as the first competitive CST inhibitor. Compound 16 can serve as a new lead structure for the development of drugs for the treatment of this devastating disease, MLD, for which small molecule therapeutics are currently not available.
Collapse
Affiliation(s)
- Wenjin Li
- Department of Pharmaceutical & Medicinal Chemistry, PharmaCenter Bonn, Pharmaceutical Institute, University of Bonn, Bonn, Germany
| | | | - Younis Baqi
- Department of Chemistry, College of Science, Sultan Qaboos University, Muscat, Oman
| | - Isabell Wachsmann
- Institut für Biochemie und Molekularbiologie, University of Bonn, Bonn, Germany
| | - Volkmar Gieselmann
- Institut für Biochemie und Molekularbiologie, University of Bonn, Bonn, Germany
| | | | - Christa E. Müller
- Department of Pharmaceutical & Medicinal Chemistry, PharmaCenter Bonn, Pharmaceutical Institute, University of Bonn, Bonn, Germany
| |
Collapse
|
18
|
Exley MA, Gensollen T, Blumberg RS. A nano-engager for iNKT cells in cancer. NATURE CANCER 2020; 1:1032-1034. [PMID: 35122067 DOI: 10.1038/s43018-020-00138-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Affiliation(s)
- Mark A Exley
- Department of Medicine, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA.
- University of Manchester, Manchester, UK.
- Imvax, Philadelphia, PA, USA.
- Agentus, Lexington, MA, USA.
| | - Thomas Gensollen
- Department of Medicine, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Richard S Blumberg
- Department of Medicine, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
19
|
Martins LS, Duarte EL, Lamy MT, Rozenfeld JHK. Supramolecular organization of α-galactosylceramide in pure dispersions and in cationic DODAB bilayers. Chem Phys Lipids 2020; 232:104963. [PMID: 32882224 DOI: 10.1016/j.chemphyslip.2020.104963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 08/01/2020] [Accepted: 08/20/2020] [Indexed: 11/30/2022]
Abstract
α-galactosylceramide (α-GalCer; KRN7000) strongly stimulates NKT cells. The structures of α-GalCer assemblies and of cationic DODAB bilayers containing α-GalCer were investigated by differential scanning calorimetry (DSC) and electron spin resonance (ESR) spectroscopy. Assemblies of α-GalCer have a very tightly packed gel phase, causing spin labels to cluster and display spin exchange interactions. An endothermic phase transition is observed by DSC, leading to a fluid phase. This phase transition peak disappears upon mixing with DODAB, showing that up to 9 mol% α-GalCer is miscible with the cationic lipid. ESR spectra show that α-GalCer decreases DODAB gel phase packing, resulting in a decrease of gel-fluid transition temperature and cooperativity in DSC thermograms of mixed bilayers. In contrast, α-GalCer increases the rigidity of the fluid phase. These effects are probably due to the conformation of the rigid amide bond that connects the phytosphingosine base of α-GalCer to its long and saturated acyl chain. Possibly, α-GalCer adopts a V-shaped conformation because of the perpendicular orientation of the amide bond towards the axes of the hydrocarbon chains. Apparently, the effect of the amide bond configuration is a key structural feature for the interaction between ceramide-based glycolipids and DODAB molecules, since we have previously reported a similar decrease of gel phase packing and increase in fluid phase rigidity for DODAB bilayers containing C24:1β-glucosylceramide. Since the structure of delivery systems is critical to the biological activity of α-GalCer, this work certainly contributes to the planning and development of novel immunotherapeutic tools.
Collapse
Affiliation(s)
- Letícia S Martins
- Departamento de Biofísica, Escola Paulista de Medicina, Universidade Federal de São Paulo, R. Botucatu 862, 04023-062, São Paulo, SP, Brazil
| | - Evandro L Duarte
- Instituto de Física, Universidade de São Paulo, Rua do Matão 1371, 05508090, São Paulo, SP, Brazil
| | - M Teresa Lamy
- Instituto de Física, Universidade de São Paulo, Rua do Matão 1371, 05508090, São Paulo, SP, Brazil
| | - Julio H K Rozenfeld
- Departamento de Biofísica, Escola Paulista de Medicina, Universidade Federal de São Paulo, R. Botucatu 862, 04023-062, São Paulo, SP, Brazil.
| |
Collapse
|
20
|
Sabry M, Lowdell MW. Killers at the crossroads: The use of innate immune cells in adoptive cellular therapy of cancer. Stem Cells Transl Med 2020; 9:974-984. [PMID: 32416056 PMCID: PMC7445022 DOI: 10.1002/sctm.19-0423] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 04/01/2020] [Accepted: 04/18/2020] [Indexed: 12/16/2022] Open
Abstract
Adoptive cell therapy (ACT) is an approach to cancer treatment that involves the use of antitumor immune cells to target residual disease in patients after completion of chemo/radiotherapy. ACT has several advantages compared with other approaches in cancer immunotherapy, including the ability to specifically expand effector cells in vitro before selection for adoptive transfer, as well as the opportunity for host manipulation in order to enhance the ability of transferred cells to recognize and kill established tumors. One of the main challenges to the success of ACT in cancer clinical trials is the identification and generation of antitumor effector cells with high avidity for tumor recognition. Natural killer (NK) cells, cytokine‐induced killers and natural killer T cells are key innate or innate‐like effector cells in cancer immunosurveillance that act at the interface between innate and adaptive immunity, to have a greater influence over immune responses to cancer. In this review, we discuss recent studies that highlight their potential in cancer therapy and summarize clinical trials using these effector immune cells in adoptive cellular therapy for the treatment of cancer.
Collapse
Affiliation(s)
- May Sabry
- Department of HaematologyUniversity College LondonLondonUK
| | | |
Collapse
|
21
|
Lee C, Hong SN, Kim YH. A glycolipid adjuvant, 7DW8-5, provides a protective effect against colonic inflammation in mice by the recruitment of CD1d-restricted natural killer T cells. Intest Res 2020; 18:402-411. [PMID: 32248672 PMCID: PMC7609397 DOI: 10.5217/ir.2019.00132] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 01/08/2020] [Indexed: 01/31/2023] Open
Abstract
Background/Aims The modulation of CD1d-restricted natural killer T (NKT) cells by glycolipids has been considered as a potential therapy against immunologic diseases, including inflammatory bowel disease. A recently identified a glycolipid analog, 7DW8-5, which is derived from α-galactosylceramide (α-GalCer), is as much as 100-fold more active at stimulating both human and mice NKT cells when compared to α-GalCer. We explored the effects of 7DW8-5 in mouse models of acute and chronic colitis. Methods We investigated the effects of 7DW8-5 on intestinal inflammation by assessing the effects of 7dW8-5 on a murine dextran sulfate sodium (DSS)-induced acute colitis model and a chronic colitis-associated tumor model. Results The acute DSS-induced colitis model showed a dose-dependent response to 7DW8-5, as mice administered 7DW8-5 showed a significant improvement in DSS-induced colitis based on their disease activity index, histologic analysis, and serum C-reactive protein levels, when compared to mice administered vehicle alone. However, DSS-induced colitis in CD1d-KO mice showed no response to 7DW8-5. A fluorescence-activating cell sorting analysis revealed an increase in NKT cells in colonic tissues of 7DW8-5-treated mice. RNA-seq and real-time quantitative polymerase chain reaction showed a significant increase in the expression of interleukin (IL)-4, IL-13, and interferon-gamma in 7DW8-5-treated mice. In addition, 7DW8-5 treatment reduced colitis-associated tumor development in an azoxymethane/DSS mouse model. Conclusions 7DW8-5 activates NKT cells through CD1d and provides a protective effect against intestinal inflammation in mice. Therefore, 7DW8-5 may be a promising therapeutic agent for treatment of inflammatory bowel disease.
Collapse
Affiliation(s)
- Chansu Lee
- Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, Korea
| | - Sung Noh Hong
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Young-Ho Kim
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
22
|
Florean C, Dicato M, Diederich M. Immune-modulating and anti-inflammatory marine compounds against cancer. Semin Cancer Biol 2020; 80:58-72. [PMID: 32070764 DOI: 10.1016/j.semcancer.2020.02.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 02/10/2020] [Accepted: 02/10/2020] [Indexed: 02/07/2023]
Abstract
The recent advances in cancer immunotherapy confirm the crucial role of the immune system in cancer progression and treatment. Chronic inflammation and reduced immune surveillance are both features of the tumor microenvironment. Strategies aimed at reverting pro-tumor inflammation and stimulating the antitumor immune components are being actively searched, and the anticancer effects of many candidate drugs have been linked to their ability to modulate the immune system. Marine organisms constitute a rich reservoir of new bioactive molecules; some of them have already been exploited for pharmaceutical use, whereas many others are undergoing clinical or preclinical investigations for the treatment of different diseases, including cancer. In this review, we will discuss the immune-modulatory properties of marine compounds for their potential use in cancer prevention and treatment and as possible tools in the context of cancer immunotherapy.
Collapse
Affiliation(s)
- Cristina Florean
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg, L-2540 Luxembourg
| | - Mario Dicato
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg, L-2540 Luxembourg
| | - Marc Diederich
- College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea.
| |
Collapse
|
23
|
3,4-Dideoxy-3,3,4,4-tetrafluoro- and 4-OH epimeric 3-deoxy-3,3-difluoro-α-GalCer analogues: Synthesis and biological evaluation on human iNKT cells stimulation. Eur J Med Chem 2019; 178:195-213. [PMID: 31185411 DOI: 10.1016/j.ejmech.2019.05.069] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 05/23/2019] [Accepted: 05/24/2019] [Indexed: 02/07/2023]
Abstract
iNKT cells recognize CD1d/α-galactosylceramide (α-GalCer) complexes via their invariant TCR receptor and stimulate the immune response. Many α-GalCer analogues have been investigated to interrogate this interaction. Following our previous work related to the modification of the hydrogen bond network between α-GalCer and CD1d, we have now focused our attention on the synthesis of 3-deoxy-3,3-difluoro- and 3,4-dideoxy-3,3,4,4-tetrafluoro-α-GalCer analogues, and studied their ability to stimulate human iNKT cells. In each case, deoxygenation at the indicated positions was accompanied by difluoro introduction in order to evaluate the resulting electronic effect on the stability of the ternary CD1d/Galcer/TCR complex which has been rationalized by modeling study. With deoxy-difluorination at the 3-position, the two epimeric 4-OH analogues were investigated to establish their capacity to compensate for the lack of the hydrogen bond donating group at the 3-position. The 3,4-dideoxytetrafluoro analogue was of interest to highlight the amide NH-bond hydrogen bond properties.
Collapse
|
24
|
Nano spray dryer for vectorizing α-galactosylceramide in polymeric nanoparticles: A single step process to enhance invariant Natural Killer T lymphocyte responses. Int J Pharm 2019; 565:123-132. [DOI: 10.1016/j.ijpharm.2019.05.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 05/03/2019] [Accepted: 05/06/2019] [Indexed: 12/31/2022]
|
25
|
Patel S, Burga RA, Powell AB, Chorvinsky EA, Hoq N, McCormack SE, Van Pelt SN, Hanley PJ, Cruz CRY. Beyond CAR T Cells: Other Cell-Based Immunotherapeutic Strategies Against Cancer. Front Oncol 2019; 9:196. [PMID: 31024832 PMCID: PMC6467966 DOI: 10.3389/fonc.2019.00196] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 03/07/2019] [Indexed: 12/13/2022] Open
Abstract
Background: Chimeric antigen receptor (CAR)-modified T cells have successfully harnessed T cell immunity against malignancies, but they are by no means the only cell therapies in development for cancer. Main Text Summary: Systemic immunity is thought to play a key role in combatting neoplastic disease; in this vein, genetic modifications meant to explore other components of T cell immunity are being evaluated. In addition, other immune cells—from both the innate and adaptive compartments—are in various stages of clinical application. In this review, we focus on these non-CAR T cell immunotherapeutic approaches for malignancy. The first section describes engineering T cells to express non-CAR constructs, and the second section describes other gene-modified cells used to target malignancy. Conclusions: CAR T cell therapies have demonstrated the clinical benefits of harnessing our body's own defenses to combat tumor cells. Similar research is being conducted on lesser known modifications and gene-modified immune cells, which we highlight in this review.
Collapse
Affiliation(s)
- Shabnum Patel
- GW Cancer Center, The George Washington University, Washington, DC, United States
| | - Rachel A Burga
- GW Cancer Center, The George Washington University, Washington, DC, United States
| | - Allison B Powell
- GW Cancer Center, The George Washington University, Washington, DC, United States
| | - Elizabeth A Chorvinsky
- Center for Cancer and Immunology Research, Children's National Health System, Washington, DC, United States
| | - Nia Hoq
- GW Cancer Center, The George Washington University, Washington, DC, United States
| | - Sarah E McCormack
- GW Cancer Center, The George Washington University, Washington, DC, United States
| | - Stacey N Van Pelt
- GW Cancer Center, The George Washington University, Washington, DC, United States
| | - Patrick J Hanley
- Center for Cancer and Immunology Research, Children's National Health System, Washington, DC, United States
| | - Conrad Russell Y Cruz
- GW Cancer Center, The George Washington University, Washington, DC, United States.,Center for Cancer and Immunology Research, Children's National Health System, Washington, DC, United States
| |
Collapse
|
26
|
Escribà-Garcia L, Alvarez-Fernández C, Caballero AC, Schaub R, Sierra J, Briones J. The novel agonistic iNKT-cell antibody NKT14m induces a therapeutic antitumor response against B-cell lymphoma. Oncoimmunology 2019; 8:e1546543. [PMID: 30713807 DOI: 10.1080/2162402x.2018.1546543] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 10/24/2018] [Accepted: 11/02/2018] [Indexed: 02/02/2023] Open
Abstract
Invariant natural killer T (iNKT) cells are a small population of T lymphocytes that expresses an invariant T cell receptor with a unique specificity for glycolipid antigens. Their activation using the glycolipid α-galactosylceramide (α-GalCer) triggers innate and adaptive immune responses. The use of α-GalCer in preclinical models as a single antitumor treatment showed moderate effect, but its efficacy in cancer patients was less effective. In addition, this glycolipid induces long-term iNKT-cell anergy precluding the possibility of retreatment. Recently, the first murine iNKT-cell agonistic antibody, NKT14m, has been developed. Here, we analyzed, for the first time, the antitumor efficacy of NKT14m in a B-cell lymphoma model. In a therapeutic setting, a single dose of NKT14m had a moderate antitumor efficacy that was associated with an increase of IFN-γ producing iNKT cells even after a second dose of the NKT14m antibody. Importantly, the combination of a single dose of NKT14m with cyclophosphamide had a potent antitumor efficacy and long-lasting immunity in vivo. Our findings provide the first evidence of the in vivo antitumor efficacy of NKT14m antibody, showing that, either alone or in combination with chemotherapy, induces an effective antitumor response. These results open new opportunities for iNKT-cell mediated immunotherapy to treat B-cell lymphoma.
Collapse
Affiliation(s)
- Laura Escribà-Garcia
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Laboratory of Experimental Hematology-IIB, Institut Recerca Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Josep Carreras Leukaemia Research Institute, Barcelona, Spain
| | - Carmen Alvarez-Fernández
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Laboratory of Experimental Hematology-IIB, Institut Recerca Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Josep Carreras Leukaemia Research Institute, Barcelona, Spain
| | - Ana Carolina Caballero
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Laboratory of Experimental Hematology-IIB, Institut Recerca Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Josep Carreras Leukaemia Research Institute, Barcelona, Spain
| | | | - Jorge Sierra
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Josep Carreras Leukaemia Research Institute, Barcelona, Spain.,Autonomous University, Barcelona, Spain
| | - Javier Briones
- Hematology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Laboratory of Experimental Hematology-IIB, Institut Recerca Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Josep Carreras Leukaemia Research Institute, Barcelona, Spain.,Autonomous University, Barcelona, Spain
| |
Collapse
|
27
|
Chen L, Gu J, Qian Y, Li M, Qian Y, Xu M, Li J, Wen Y, Xia L, Li J, Xia Q, Kong X, Wu H. Deletion of C-C Motif Chemokine Ligand 5 Worsens Invariant Natural Killer T-Cell-Mediated Hepatitis via Compensatory Up-regulation of CXCR2-Related Chemokine Activity. Cell Mol Gastroenterol Hepatol 2019; 7:623-639. [PMID: 30630119 PMCID: PMC6434496 DOI: 10.1016/j.jcmgh.2018.12.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 12/29/2018] [Accepted: 12/31/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Chemokine-mediated immune cell recruitment plays pivotal roles in liver inflammation. C-C motif chemokine ligand 5 (CCL5) has been shown to be responsible for the recruitment of monocytes/macrophages and has been implicated in various liver diseases, including nonalcoholic fatty liver disease, fibrosis, and hepatocellular carcinoma. Previous studies have also shown that inhibition of CCL5 appears to be a promising therapeutic approach for several chronic liver diseases. However, whether blocking CCL5 could benefit immune cell-mediated hepatitis remains largely elusive. METHODS By adopting a specific agonist, alpha-galactosylceramide (α-Galcer), of invariant natural killer T cells (iNKTs), we investigated the function and mechanism of CCL5 in the iNKT induced murine hepatitis model. RESULTS We found significantly increased CCL5 expression in α-Galcer-induced hepatitis murine model. Such an increase in CCL5 is mainly enriched in non-parenchymal cells such as macrophages and iNKTs but not in hepatocytes. Surprisingly, CCL5 blockage by genetic deletion of Ccl5 does not affect the α-Galcer-induced iNKT activation but greatly worsens α-Galcer-induced liver injury accompanied by an increased hepatic neutrophil infiltration. Mechanistically, we demonstrated that greater neutrophil accumulation in the liver is responsible for the enhanced liver injury in Ccl5-/- mice. Such an increased hepatic neutrophil infiltration is mainly caused by an enhanced CXCL1-CXCR2 signal in Ccl5-/- mice. Therapeutically, either antibody-mediated neutrophil depletion or a CXCR2 antagonist, SB225002, mediated CXCR2 signaling blockage significantly ameliorated α-Galcer-induced liver injury in Ccl5-/- mice. CONCLUSIONS Our present study demonstrates that (1) α-Galcer-induced murine hepatitis could greatly induce CCL5 production in macrophages and iNKT cells; (2) loss of CCL5 could enhance CXCL1 expression in hepatocytes and activate CXCL1-CXCR2 axis in neutrophils to augment their hepatic infiltration; and (3) neutrophil depletion or blockage of CXCL1-CXCR2 axis greatly improves α-Galcer-induced liver injury in Ccl5-/- mice. This study suggests that clinical utilization of CCL5 blockage may compensatorily induce the activation of other chemokine pathways to enhance neutrophil recruitment and liver injury in hepatitis.
Collapse
Affiliation(s)
- Lili Chen
- Shanghai Key Laboratory for Molecular Imaging, Collaborative Research Center, Shanghai University of Medicine and Health Sciences, Shanghai, China; Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jinyang Gu
- Department of Transplantation, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yihan Qian
- School of Pharmacy, Fudan University, Shanghai, China
| | - Meng Li
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yongbing Qian
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Min Xu
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jichang Li
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yankai Wen
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lei Xia
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jiaxin Li
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qiang Xia
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoni Kong
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Hailong Wu
- Shanghai Key Laboratory for Molecular Imaging, Collaborative Research Center, Shanghai University of Medicine and Health Sciences, Shanghai, China.
| |
Collapse
|
28
|
Dorta-Estremera S, Chin RL, Sierra G, Nicholas C, Yanamandra AV, Nookala SMK, Yang G, Singh S, Curran MA, Sastry KJ. Mucosal HPV E6/E7 Peptide Vaccination in Combination with Immune Checkpoint Modulation Induces Regression of HPV + Oral Cancers. Cancer Res 2018; 78:5327-5339. [PMID: 30054333 DOI: 10.1158/0008-5472.can-18-0892] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 06/15/2018] [Accepted: 07/17/2018] [Indexed: 12/22/2022]
Abstract
High-risk human papillomavirus (HPV)-associated squamous cell carcinomas of the oropharynx (SCCOP) are among the fastest growing cancers. After standard-of-care treatment, however, patients with HPV+ SCCOP have better overall and disease-specific survival than patients with HPV- SCCOP, suggesting the importance of HPV-specific immunity. We reasoned that therapeutic vaccination targeting the HPV-16 E6 and E7 oncogenes could elicit high-affinity, high-frequency tumor antigen-specific T-cell responses, which could then be augmented and shielded from suppression in the tumor microenvironment by immune checkpoint modulation. In this study, we used a preclinical syngeneic mouse model of oral cancer comprised of mouse tonsil-derived epithelial cells stably expressing HPV-16 E6 and E7 genes along with H-ras oncogene (mEER) to identify combinations of vaccination and checkpoint antibodies capable of promoting tumor regression. Intranasal HPV E6/E7 peptide vaccination and single checkpoint antibodies failed to elicit responses in more than half of animals; however, 4-1BB agonist antibody along with either CD40 agonist antibody or CTLA-4 blockade eliminated the majority of established mEER tumors. The combination of intranasal HPV peptide vaccine and α4-1BB and αCTLA-4 antibodies produced curative efficacy and a better safety profile against orally implanted mEER tumors. Correlates of protective immunity included enhanced intratumoral levels of CD8 T cells relative to immunosuppressive regulatory T cells and myeloid-derived suppressor cells. Overall, our results demonstrate combination vaccine-immunotherapy modalities as novel treatment options for HPV+ SCCOP.Significance: Combinations of vaccine and checkpoint modulation are effective and safe treatment options for HPV+ oral cancers. Cancer Res; 78(18); 5327-39. ©2018 AACR.
Collapse
Affiliation(s)
| | - Renee L Chin
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gloria Sierra
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences at Houston, Houston, Texas
| | - Courtney Nicholas
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ananta V Yanamandra
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sita M K Nookala
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Guojun Yang
- The University of Texas MD Anderson Cancer Center, Oncology Research for Biologics and Immunotherapy Translation, Houston, Texas, Texas
| | - Shail Singh
- Department of Melanoma and Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, Texas
| | - Michael A Curran
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences at Houston, Houston, Texas
| | - K Jagannadha Sastry
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas. .,The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences at Houston, Houston, Texas
| |
Collapse
|
29
|
Wolf BJ, Choi JE, Exley MA. Novel Approaches to Exploiting Invariant NKT Cells in Cancer Immunotherapy. Front Immunol 2018; 9:384. [PMID: 29559971 PMCID: PMC5845557 DOI: 10.3389/fimmu.2018.00384] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 02/12/2018] [Indexed: 12/24/2022] Open
Abstract
iNKT cells are a subset of innate-like T cells that utilize an invariant TCR alpha chain complexed with a limited repertoire of TCR beta chains to recognize specific lipid antigens presented by CD1d molecules. Because iNKT cells have an invariant TCR, they can be easily identified and targeted in both humans and mice via standard reagents, making this a population of T cells that has been well characterized. iNKT cells are some of the first cells to respond during an infection. By making different types of cytokines in response to different infection stimuli, iNKT cells help determine what kind of immune response then develops. It has been shown that iNKT cells are some of the first cells to respond during infection with a pathogen and the type of cytokines that iNKT cells make help determine the type of immune response that develops in various situations. Indeed, along with immunity to pathogens, pre-clinical mouse studies have clearly demonstrated that iNKT cells play a critical role in tumor immunosurveillance. They can mediate anti-tumor immunity by direct recognition of tumor cells that express CD1d, and/or via targeting CD1d found on cells within the tumor microenvironment. Multiple groups are now working on manipulating iNKT cells for clinical benefit within the context of cancer and have demonstrated that targeting iNKT cells can have a therapeutic benefit in patients. In this review, we briefly introduce iNKT cells, then discuss preclinical data on roles of iNKT cells and clinical trials that have targeted iNKT cells in cancer patients. We finally discuss how future trials could be modified to further increase the efficacy of iNKT cell therapies, in particular CAR-iNKT and rTCR-iNKT cells.
Collapse
Affiliation(s)
| | - Jiyoung Elizabeth Choi
- Agenus Inc., Lexington, MA, United States.,Brigham & Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Mark A Exley
- Agenus Inc., Lexington, MA, United States.,Brigham & Women's Hospital, Harvard Medical School, Boston, MA, United States.,University of Manchester, Manchester, United Kingdom
| |
Collapse
|
30
|
Okazaki S, Iwasaki T, Yuba E, Watarai S. Evaluation of pH-sensitive fusogenic polymer-modified liposomes co-loaded with antigen and α-galactosylceramide as an anti-tumor vaccine. J Vet Med Sci 2017; 80:197-204. [PMID: 29311431 PMCID: PMC5836753 DOI: 10.1292/jvms.17-0491] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
pH-Sensitive fusogenic polymer-modified (pH-sensitive) liposomes co-loaded with tumor
model antigen, ovalbumin (OVA), and adjuvant, α-galactosylceramide (α-GalCer) were
fabricated and administered subcutaneously into mice. The ability of pH-sensitive
liposomes containing OVA and α-GalCer to stimulate cellular and humoral immune responses
in vivo was compared with OVA-encapsulating pH-sensitive liposomes as
well as with OVA alone. After immunization, significant OVA-specific antibodies were
detected in the serum. When sera were analyzed for isotype distribution, antigen-specific
IgG1 antibody responses were noted in mice immunized with OVA alone, whereas immunization
with OVA-containing pH-sensitive liposomes and with pH-sensitive liposomes containing OVA
and α-GalCer resulted in the induction of OVA-specific IgG1 and IgG2b antibody responses.
Moreover, more substantial production of IFN-γ and IL-4 was demonstrated in spleen cells
from mice immunized with pH-sensitive liposomes having OVA and α-GalCer than
OVA-containing pH-sensitive liposomes in vitro. Spleen cells from the
immunized mice showed strong cytotoxic activity against E.G7-OVA tumor cells. In addition,
prophylactic vaccination efficacy against tumor formation was evaluated. In all mice
immunized with pH-sensitive liposomes having OVA and α-GalCer, immunization provided
substantial protection from tumor formation. The therapeutic efficacy of pH-sensitive
liposomes containing OVA and α-GalCer against already established E.G7-OVA tumors was also
investigated. Tumor growth was reduced significantly in all mice treated with pH-sensitive
liposomes having OVA and α-GalCer. The provided evidence on the advantage of antigen and
α-GalCer co-encapsulation into pH-sensitive liposomes should be considered in the design
of future cancer vaccines for prophylactic and therapeutic purposes.
Collapse
Affiliation(s)
- Seiji Okazaki
- Division of Veterinary Science, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Izumisano, Osaka 598-8531, Japan
| | - Tadashi Iwasaki
- Division of Veterinary Science, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Izumisano, Osaka 598-8531, Japan
| | - Eiji Yuba
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, Sakai, Osaka 599-8531, Japan
| | - Shinobu Watarai
- Division of Veterinary Science, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Izumisano, Osaka 598-8531, Japan
| |
Collapse
|
31
|
Fallon EA, Chun TT, Young WA, Gray C, Ayala A, Heffernan DS. Program Cell Death Receptor-1-Mediated Invariant Natural Killer T-Cell Control of Peritoneal Macrophage Modulates Survival in Neonatal Sepsis. Front Immunol 2017; 8:1469. [PMID: 29209308 PMCID: PMC5701916 DOI: 10.3389/fimmu.2017.01469] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 10/19/2017] [Indexed: 02/02/2023] Open
Abstract
We have shown that invariant natural killer T (iNKT) cells mediate sepsis-induced end-organ changes and immune responses, including macrophage bacterial phagocytosis, a finding regulated by the check point protein program cell death receptor-1 (PD-1). Furthermore, PD-1 mediates mortality in both adult and neonatal murine sepsis as well as in surgical patients. Given our previous findings, we hypothesize that iNKT cells will also modulate neonatal sepsis survival, and that this effect is regulated in part through PD-1. We utilized a polymicrobial intra-peritoneal cecal slurry (CS) sepsis model in wild type (WT), iNKT−/− or PD-1−/− 5–7 day old neonatal pups. Typically, tissues were harvested at 24 h for various bioassays/histology and, in some cases, survival was assessed for up to 7 days. Interestingly, similar to what we recently reported for PD-1−/− mice following CS, iNKT−/−-deficient animals exhibit a markedly improved survival vs. WT. Histologically, minor alterations in liver architectural, which were noted in WT pups, were attenuated in both iNKT−/− and PD-1−/− pups. Following CS, PECAM-1 expression was unchanged in the WT pups but increased in both iNKT−/− and PD-1−/− pups. In WT, following CS the emergence of a Ly6Clow subpopulation was noted among the influxed peritoneal macrophage population. Conversely, within iNKT−/− pups, there were fewer peritoneal macrophages and a greater percentage of Ly6Chigh macrophages. We show not only a key role for iNKT cells in affecting end-organ damage as well as alterations in phagocytes phenotypes in neonatal sepsis but that this iNKT cell mediated effect is driven by the central checkpoint protein PD-1.
Collapse
Affiliation(s)
- Eleanor A Fallon
- Division of Surgical Research, Department of Surgery, Brown University and Rhode Island Hospital, Providence, RI, United States
| | - Tristen T Chun
- Division of Surgical Research, Department of Surgery, Brown University and Rhode Island Hospital, Providence, RI, United States
| | - Whitney A Young
- Division of Surgical Research, Department of Surgery, Brown University and Rhode Island Hospital, Providence, RI, United States
| | - Chyna Gray
- Division of Surgical Research, Department of Surgery, Brown University and Rhode Island Hospital, Providence, RI, United States
| | - Alfred Ayala
- Division of Surgical Research, Department of Surgery, Brown University and Rhode Island Hospital, Providence, RI, United States
| | - Daithi S Heffernan
- Division of Surgical Research, Department of Surgery, Brown University and Rhode Island Hospital, Providence, RI, United States
| |
Collapse
|
32
|
Teyton L. New Directions for Natural Killer T Cells in the Immunotherapy of Cancer. Front Immunol 2017; 8:1480. [PMID: 29209309 PMCID: PMC5701619 DOI: 10.3389/fimmu.2017.01480] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 10/23/2017] [Indexed: 01/07/2023] Open
Abstract
Natural killer T (NKT) cells have been placed at the interface between innate and adaptive immunity by a long series of experiments that convincingly showed that beyond cytokine secretion and NK cell recruitment, NKT cells were coordinating dendritic cell and B cell maturation through direct membrane contacts and initiate productive responses. As such, NKT cells are the cellular adjuvant of many immune reactions and have functions that go much beyond what their name encapsulates. In addition, the initial discovery of the ligands of NKT cells is deeply linked to cancer biology and therapy. However, for a host of reasons, animal models in which agonists of NKT cells were used did not translate well to human cancers. A systematic reassessment of NKT cells role in tumorigenesis, especially spontaneous one, is now accessible using single cell analysis technologies both in mouse and man, and should be taken advantage of. Similarly, the migration, localization, phenotype of NKT cells following induced expansion after injection of an agonist can be examined at the single cell level. This technological revolution will help evaluate where and how NKT cells can be used in cancer.
Collapse
Affiliation(s)
- Luc Teyton
- Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, CA, United States
| |
Collapse
|
33
|
Lam PY, Nissen MD, Mattarollo SR. Invariant Natural Killer T Cells in Immune Regulation of Blood Cancers: Harnessing Their Potential in Immunotherapies. Front Immunol 2017; 8:1355. [PMID: 29109728 PMCID: PMC5660073 DOI: 10.3389/fimmu.2017.01355] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 10/03/2017] [Indexed: 01/03/2023] Open
Abstract
Invariant natural killer T (iNKT) cells are a unique innate T lymphocyte population that possess cytolytic properties and profound immunoregulatory activities. iNKT cells play an important role in the immune surveillance of blood cancers. They predominantly recognize glycolipid antigens presented on CD1d, but their activation and cytolytic activities are not confined to CD1d expressing cells. iNKT cell stimulation and subsequent production of immunomodulatory cytokines serve to enhance the overall antitumor immune response. Crucially, the activation of iNKT cells in cancer often precedes the activation and priming of other immune effector cells, such as NK cells and T cells, thereby influencing the generation and outcome of the antitumor immune response. Blood cancers can evade or dampen iNKT cell responses by downregulating expression of recognition receptors or by actively suppressing or diverting iNKT cell functions. This review will discuss literature on iNKT cell activity and associated dysregulation in blood cancers as well as highlight some of the strategies designed to harness and enhance iNKT cell functions against blood cancers.
Collapse
Affiliation(s)
- Pui Yeng Lam
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| | - Michael D. Nissen
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| | - Stephen R. Mattarollo
- The University of Queensland Diamantina Institute, The University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| |
Collapse
|
34
|
Macho-Fernandez E, Chekkat N, Ehret C, Thomann JS, De Giorgi M, Spanedda MV, Bourel-Bonnet L, Betbeder D, Heurtault B, Faveeuw C, Fournel S, Frisch B, Trottein F. Solubilization of α-galactosylceramide in aqueous medium: Impact on Natural Killer T cell activation and antitumor responses. Int J Pharm 2017; 530:354-363. [DOI: 10.1016/j.ijpharm.2017.07.054] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 07/17/2017] [Accepted: 07/18/2017] [Indexed: 12/21/2022]
|
35
|
Ghinnagow R, De Meester J, Cruz LJ, Aspord C, Corgnac S, Macho-Fernandez E, Soulard D, Fontaine J, Chaperot L, Charles J, Soncin F, Mami-Chouaib F, Plumas J, Faveeuw C, Trottein F. Co-delivery of the NKT agonist α-galactosylceramide and tumor antigens to cross-priming dendritic cells breaks tolerance to self-antigens and promotes antitumor responses. Oncoimmunology 2017; 6:e1339855. [PMID: 28932640 DOI: 10.1080/2162402x.2017.1339855] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 06/01/2017] [Accepted: 06/01/2017] [Indexed: 10/19/2022] Open
Abstract
Vaccines designed to abrogate the tolerance of tumor self-antigens and amplify cytotoxic CD8+ T cells (CTLs) have promise for the treatment of cancer. Type I natural killer (NKT) cells have attracted considerable interest in the cancer therapy field. In the current study, we have exploited the unique ability of NKT cells to serve as T-helper cells to license dendritic cells (DCs) for cross priming with the aim to generate efficient CTL antitumor responses. To this end, we designed a nanoparticle-based vaccine to target cross-priming DCs via the Clec9a endocytic pathway. Our results showed for the first time that simultaneous co-delivery of the NKT agonist α-galactosylceramide and tumor self-antigens (Trp2 and gp100) to CD8α+ DCs promotes strong antitumor responses in prophylactic and therapeutic settings (advanced solid tumor model in the mouse). We attributed the vaccine's therapeutic effects to NKT cells (but not to T-helper lymphocytes) and CD8+ T cells. Efficacy was correlated with an elevated ratio between tumor antigen-specific CD8+ T cells and regulatory CD4+ T lymphocytes within the tumor. The nanoparticle-based vaccine actively targeted human CLEC9A-expressing BDCA3+ DCs - the equivalent of murine cross-priming CD8α+ DCs - and induced a strong expansion of effector memory tumor self-antigen (Melan -A)-specific CD8+ T cells from peripheral blood mononuclear cells sourced from healthy donors and melanoma patients. Together, our result shed light on novel therapeutic approaches for controlling tumor development.
Collapse
Affiliation(s)
- Reem Ghinnagow
- Univ. Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, F-59000 Lille, France.,Centre National de la Recherche Scientifique, UMR 8204, F-59000 Lille, France.,Institut National de la Santé et de la Recherche Médicale U1019, F-59000 Lille, France.,Centre Hospitalier Universitaire de Lille, F-59000 Lille, France.,Institut Pasteur de Lille, F-59000 Lille, France
| | - Julie De Meester
- Univ. Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, F-59000 Lille, France.,Centre National de la Recherche Scientifique, UMR 8204, F-59000 Lille, France.,Institut National de la Santé et de la Recherche Médicale U1019, F-59000 Lille, France.,Centre Hospitalier Universitaire de Lille, F-59000 Lille, France.,Institut Pasteur de Lille, F-59000 Lille, France
| | - Luis Javier Cruz
- Translational Nanobiomaterials and Imaging, Department of radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Caroline Aspord
- Institute for Advanced Biosciences (IAB), Team Immunobiology and Immunotherapy in Chronic Diseases, INSERM U1209, CNRS UMR5309, Université Grenoble Alpes, Etablissement Français du Sang-Rhone-Alpes, F-38700 Grenoble, France
| | - Stéphanie Corgnac
- INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy, Fac. de médecine - Univ. Paris-Sud, Université Paris-Saclay, 94805, Villejuif, France
| | - Elodie Macho-Fernandez
- Univ. Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, F-59000 Lille, France.,Centre National de la Recherche Scientifique, UMR 8204, F-59000 Lille, France.,Institut National de la Santé et de la Recherche Médicale U1019, F-59000 Lille, France.,Centre Hospitalier Universitaire de Lille, F-59000 Lille, France.,Institut Pasteur de Lille, F-59000 Lille, France
| | - Daphnée Soulard
- Univ. Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, F-59000 Lille, France.,Centre National de la Recherche Scientifique, UMR 8204, F-59000 Lille, France.,Institut National de la Santé et de la Recherche Médicale U1019, F-59000 Lille, France.,Centre Hospitalier Universitaire de Lille, F-59000 Lille, France.,Institut Pasteur de Lille, F-59000 Lille, France
| | - Josette Fontaine
- Univ. Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, F-59000 Lille, France.,Centre National de la Recherche Scientifique, UMR 8204, F-59000 Lille, France.,Institut National de la Santé et de la Recherche Médicale U1019, F-59000 Lille, France.,Centre Hospitalier Universitaire de Lille, F-59000 Lille, France.,Institut Pasteur de Lille, F-59000 Lille, France
| | - Laurence Chaperot
- Institute for Advanced Biosciences (IAB), Team Immunobiology and Immunotherapy in Chronic Diseases, INSERM U1209, CNRS UMR5309, Université Grenoble Alpes, Etablissement Français du Sang-Rhone-Alpes, F-38700 Grenoble, France
| | - Julie Charles
- Institute for Advanced Biosciences (IAB), Team Immunobiology and Immunotherapy in Chronic Diseases, INSERM U1209, CNRS UMR5309, Université Grenoble Alpes, Etablissement Français du Sang-Rhone-Alpes, F-38700 Grenoble, France.,Dermatology department, Grenoble Alpes University Hospital, F-38700 Grenoble, France
| | - Fabrice Soncin
- Centre Hospitalier Universitaire de Lille, F-59000 Lille, France.,Institut Pasteur de Lille, F-59000 Lille, France.,Centre National de la Recherche Scientifique, UMR 8161, F-59000 Lille, France
| | - Fathia Mami-Chouaib
- INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy, Fac. de médecine - Univ. Paris-Sud, Université Paris-Saclay, 94805, Villejuif, France
| | - Joel Plumas
- Institute for Advanced Biosciences (IAB), Team Immunobiology and Immunotherapy in Chronic Diseases, INSERM U1209, CNRS UMR5309, Université Grenoble Alpes, Etablissement Français du Sang-Rhone-Alpes, F-38700 Grenoble, France
| | - Christelle Faveeuw
- Univ. Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, F-59000 Lille, France.,Centre National de la Recherche Scientifique, UMR 8204, F-59000 Lille, France.,Institut National de la Santé et de la Recherche Médicale U1019, F-59000 Lille, France.,Centre Hospitalier Universitaire de Lille, F-59000 Lille, France.,Institut Pasteur de Lille, F-59000 Lille, France
| | - François Trottein
- Univ. Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, F-59000 Lille, France.,Centre National de la Recherche Scientifique, UMR 8204, F-59000 Lille, France.,Institut National de la Santé et de la Recherche Médicale U1019, F-59000 Lille, France.,Centre Hospitalier Universitaire de Lille, F-59000 Lille, France.,Institut Pasteur de Lille, F-59000 Lille, France
| |
Collapse
|
36
|
Ghinnagow R, Cruz LJ, Macho-Fernandez E, Faveeuw C, Trottein F. Enhancement of Adjuvant Functions of Natural Killer T Cells Using Nanovector Delivery Systems: Application in Anticancer Immune Therapy. Front Immunol 2017; 8:879. [PMID: 28798749 PMCID: PMC5529346 DOI: 10.3389/fimmu.2017.00879] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 07/10/2017] [Indexed: 11/13/2022] Open
Abstract
Type I natural killer T (NKT) cells have gained considerable interest in anticancer immune therapy over the last decade. This “innate-like” T lymphocyte subset has the unique ability to recognize foreign and self-derived glycolipid antigens in association with the CD1d molecule expressed by antigen-presenting cells. An important property of these cells is to bridge innate and acquired immune responses. The adjuvant function of NKT cells might be exploited in the clinics. In this review, we discuss the approaches currently being used to target NKT cells for cancer therapy. In particular, we highlight ongoing strategies utilizing NKT cell-based nanovaccines to optimize immune therapy.
Collapse
Affiliation(s)
- Reem Ghinnagow
- Univ. Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Lille, France.,Centre National de la Recherche Scientifique, UMR 8204, Lille, France.,Institut National de la Santé et de la Recherche Médicale U1019, Lille, France.,Hospitalier Universitaire de Lille, Lille, France.,Institut Pasteur de Lille, Lille, France
| | - Luis Javier Cruz
- Translational Nanobiomaterials and Imaging, Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| | - Elodie Macho-Fernandez
- Univ. Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Lille, France.,Centre National de la Recherche Scientifique, UMR 8204, Lille, France.,Institut National de la Santé et de la Recherche Médicale U1019, Lille, France.,Hospitalier Universitaire de Lille, Lille, France.,Institut Pasteur de Lille, Lille, France
| | - Christelle Faveeuw
- Univ. Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Lille, France.,Centre National de la Recherche Scientifique, UMR 8204, Lille, France.,Institut National de la Santé et de la Recherche Médicale U1019, Lille, France.,Hospitalier Universitaire de Lille, Lille, France.,Institut Pasteur de Lille, Lille, France
| | - François Trottein
- Univ. Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Lille, France.,Centre National de la Recherche Scientifique, UMR 8204, Lille, France.,Institut National de la Santé et de la Recherche Médicale U1019, Lille, France.,Hospitalier Universitaire de Lille, Lille, France.,Institut Pasteur de Lille, Lille, France
| |
Collapse
|
37
|
Dendritic cells combined with tumor cells and α-galactosylceramide induce a potent, therapeutic and NK-cell dependent antitumor immunity in B cell lymphoma. J Transl Med 2017; 15:115. [PMID: 28549432 PMCID: PMC5446707 DOI: 10.1186/s12967-017-1219-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 05/18/2017] [Indexed: 02/06/2023] Open
Abstract
Background Invariant natural killer T (iNKT) cells are a small population of lymphocytes with unique specificity for glycolipid antigens presented by non-polymorphic CD1d receptor on dendritic cells (DCs). iNKT cells play a central role in tumor immunology since they are implicated in the coordination of innate and adaptive immune responses. These cells can be activated with the prototypic lipid α-galactosylceramide (α-GalCer), stimulating interferon gamma (IFN-γ) production and cytokine secretion, which contribute to the enhancement of T cell activation. Methods We evaluated the antitumor effect of a combination of dendritic cells (DCs) and tumor cells with the iNKT cell agonist α-GalCer in a therapeutic model of B cell lymphoma. iNKT, NK and T cell phenotype was determined by flow cytometry. Serum cytokines were analyzed by Luminex technology. Significant differences between survival curves were assessed by the log-rank test. For all other data, Mann–Whitney test was used to analyze the differences between groups. Results This vaccine induced a potent (100% survival), long-lasting and tumor-specific antitumor immune response, that was associated with an increase of both Th1 cytokines and IFN-γ secreting iNKT cells (4.59 ± 0.41% vs. 0.92 ± 0.12% in control group; p = 0.01) and T cells (CD4 IFN-γ+: 3.75 ± 0.59% vs. 0.66 ± 0.18% p = 0.02; CD8 IFN-γ+: 10.61 ± 0.84% vs. 0.47 ± 0.03% p = 0.002). Importantly, natural killer (NK) cells played a critical role in the antitumor effect observed after vaccination. Conclusions This study provides clinically relevant data for the development of iNKT-cell based immunotherapy treatments for patients with B cell malignancies.
Collapse
|
38
|
Zamora-Chimal J, Hernández-Ruiz J, Becker I. NKT cells in leishmaniasis. Immunobiology 2017; 222:641-646. [DOI: 10.1016/j.imbio.2016.11.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 11/30/2016] [Indexed: 10/20/2022]
|
39
|
Exley MA, Friedlander P, Alatrakchi N, Vriend L, Yue S, Sasada T, Zeng W, Mizukami Y, Clark J, Nemer D, LeClair K, Canning C, Daley H, Dranoff G, Giobbie-Hurder A, Hodi FS, Ritz J, Balk SP. Adoptive Transfer of Invariant NKT Cells as Immunotherapy for Advanced Melanoma: A Phase I Clinical Trial. Clin Cancer Res 2017; 23:3510-3519. [PMID: 28193627 DOI: 10.1158/1078-0432.ccr-16-0600] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 04/12/2016] [Accepted: 01/24/2017] [Indexed: 02/06/2023]
Abstract
Purpose: Invariant NKT cells (iNKT) are innate-like CD1d-restricted T cells with immunoregulatory activity in diseases including cancer. iNKT from advanced cancer patients can have reversible defects including IFNγ production, and iNKT IFNγ production may stratify for survival. Previous clinical trials using iNKT cell activating ligand α-galactosylceramide have shown clinical responses. Therefore, a phase I clinical trial was performed of autologous in vitro expanded iNKT cells in stage IIIB-IV melanoma.Experimental Design: Residual iNKT cells [<0.05% of patient peripheral blood mononuclear cell (PBMC)] were purified from autologous leukapheresis product using an antibody against the iNKT cell receptor linked to magnetic microbeads. iNKT cells were then expanded with CD3 mAb and IL2 in vitro to obtain up to approximately 109 cells.Results: Expanded iNKT cells produced IFNγ, but limited or undetectable IL4 or IL10. Three iNKT infusions each were completed on 9 patients, and produced only grade 1-2 toxicities. The 4th patient onward received systemic GM-CSF with their second and third infusions. Increased numbers of iNKT cells were seen in PBMCs after some infusions, particularly when GM-CSF was also given. IFNγ responses to α-galactosylceramide were increased in PBMCs from some patients after infusions, and delayed-type hypersensitivity responses to Candida increased in 5 of 8 evaluated patients. Three patients have died, three were progression-free at 53, 60, and 65 months, three received further treatment and were alive at 61, 81, and 85 months. There was no clear correlation between outcome and immune parameters.Conclusions: Autologous in vitro expanded iNKT cells are a feasible and safe therapy, producing Th1-like responses with antitumor potential. Clin Cancer Res; 23(14); 3510-9. ©2017 AACR.
Collapse
Affiliation(s)
- Mark A Exley
- Hematology/Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts. .,Gastroenterology, Brigham & Women's Hospital, Harvard Medical School, Boston, Massachusetts.,University of Manchester, Manchester, United Kingdom
| | - Phillip Friedlander
- Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Nadia Alatrakchi
- Hematology/Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Lianne Vriend
- Hematology/Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Simon Yue
- Hematology/Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Tetsuro Sasada
- Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.,Cancer Vaccine Center, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Wanyong Zeng
- Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.,Cancer Vaccine Center, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Yo Mizukami
- Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Justice Clark
- Hematology/Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - David Nemer
- Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | | | - Christine Canning
- Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.,Cancer Vaccine Center, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Heather Daley
- Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.,Cancer Vaccine Center, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Glenn Dranoff
- Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.,Cancer Vaccine Center, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Anita Giobbie-Hurder
- Biostatistics & Computational Biology, Dana Farber Cancer Institute, Harvard School of Public Health, Boston, Massachusetts
| | - F Stephen Hodi
- Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.,Cancer Vaccine Center, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Jerome Ritz
- Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.,Cancer Vaccine Center, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Steven P Balk
- Hematology/Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
40
|
Abstract
Conventional vaccine adjuvants enhance peptide-specific T-cell and B-cell responses by modifying peptide stability or uptake or by binding to pattern-recognition receptors on antigen-presenting cells (APCs). This article discusses the application of a distinct mechanism of adjuvant activity: the activation of type I, or invariant, natural killer T (iNKT) cells to drive cellular and humoral immune responses. Using a semi-invariant T-cell receptor (TCR), iNKT cells recognize glycolipid antigens presented on cluster of differentiation (CD)-1d molecules. When their ligands are presented in concert with peptides, iNKT cells can provide T-cell help, 'licensing' APCs to augment peptide-specific T-cell and antibody responses. We discuss the potential benefits and limitations of exploiting iNKT cells as 'universal helpers' to enhance vaccine responses for the treatment and prevention of cancer and infectious diseases.
Collapse
Affiliation(s)
- Mary Speir
- Malaghan Institute of Medical Research, PO Box 7060, Wellington, 6242, New Zealand
| | - Ian F Hermans
- Malaghan Institute of Medical Research, PO Box 7060, Wellington, 6242, New Zealand.
- School of Biological Sciences, Victoria University Wellington, PO Box 600, Wellington, 6140, New Zealand.
- Maurice Wilkins Centre, Private Bag 92019, Auckland, New Zealand.
| | - Robert Weinkove
- Malaghan Institute of Medical Research, PO Box 7060, Wellington, 6242, New Zealand.
- Wellington Blood and Cancer Centre, Wellington Hospital, Private Bag 7902, Wellington, 6242, New Zealand.
- Department of Pathology and Molecular Medicine, University of Otago Wellington, Wellington, 6021, New Zealand.
| |
Collapse
|
41
|
Chang KM, Liu M. Chronic hepatitis B: immune pathogenesis and emerging immunotherapeutics. Curr Opin Pharmacol 2016; 30:93-105. [PMID: 27570126 DOI: 10.1016/j.coph.2016.07.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 07/21/2016] [Accepted: 07/25/2016] [Indexed: 12/22/2022]
Abstract
Hepatitis B virus (HBV) evades, subverts, activates and regulates host immune components, thereby impacting its natural history and disease pathogenesis. Recent advances in our understanding of immune interactions in chronic viral infection and tumor therapy are applicable to chronic hepatitis B (CHB). With recent successes of tumor immunotherapy, there is a renewed interest in exploring immunotherapeutics in achieving sustained and functional cure of chronic hepatitis B. In this review, we discuss aspects of host innate and adaptive immune regulatory and pathogenic responses relevant for HBV infection. We also highlight several immune modulatory approaches in clinical development to treat CHB.
Collapse
Affiliation(s)
- Kyong-Mi Chang
- University of Pennsylvania Perelman School of Medicine, USA; Philadelphia Corporal Michael J. Crescenz VA Medical Center, USA.
| | - Mengfei Liu
- University of Pennsylvania Perelman School of Medicine, USA
| |
Collapse
|
42
|
Immunotherapeutic strategies targeting natural killer T cell responses in cancer. Immunogenetics 2016; 68:623-38. [PMID: 27393665 DOI: 10.1007/s00251-016-0928-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 06/22/2016] [Indexed: 12/21/2022]
Abstract
Natural killer T (NKT) cells are a unique subset of lymphocytes that bridge the innate and adaptive immune system. NKT cells possess a classic αβ T cell receptor (TCR) that is able to recognize self and foreign glycolipid antigens presented by the nonclassical class I major histocompatibility complex (MHC) molecule, CD1d. Type I NKT cells (referred to as invariant NKT cells) express a semi-invariant Vα14Jα18 TCR in mice and Vα24Jα18 TCR in humans. Type II NKT cells are CD1d-restricted T cells that express a more diverse set of TCR α chains. The two types of NKT cells often exert opposing effects especially in tumor immunity, where type II cells generally suppress tumor immunity while type I NKT cells can enhance anti-tumor immune responses. In this review, we focus on the role of NKT cells in cancer. We discuss their effector and suppressive functions, as well as describe preclinical and clinical studies utilizing therapeutic strategies focused on harnessing their potent anti-tumor effector functions, and conclude with a discussion on potential next steps for the utilization of NKT cell-targeted therapies for the treatment of cancer.
Collapse
|
43
|
Abstract
The liver is an organ that has the largest amount of natural killer T(NKT) cells, which play critical roles in the pathogenesis of liver diseases. In this article, the authors summarize recent findings about the roles of NKT cells in liver injury, inflammation, fibrosis, regeneration and cancer. In brief, NKT cells accelerate liver injury by producing pro-inflammatory cytokines and directly killing hepatocytes. NKT cells are involved in complex roles in liver fibrogenesis. For instance, NKT cells inhibit liver fibrosis via suppressing hepatic stellate cell activation and can also promote liver fibrosis via enhancing liver inflammation and injury. Inactivated or weakly activated NKT cells play a minimal role in controlling liver regeneration, whilst activated NKT cells have an inhibitory effect on liver regeneration. In liver cancer, NKT cells play both pro-tumor and anti-tumor roles in controlling tumor progress.
Collapse
Affiliation(s)
- Hua Wang
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | | |
Collapse
|
44
|
Biosurfactants Produced by Marine Microorganisms with Therapeutic Applications. Mar Drugs 2016; 14:md14020038. [PMID: 26901207 PMCID: PMC4771991 DOI: 10.3390/md14020038] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 01/29/2016] [Accepted: 02/01/2016] [Indexed: 12/20/2022] Open
Abstract
Marine microorganisms possess unique metabolic and physiological features and are an important source of new biomolecules, such as biosurfactants. Some of these surface-active compounds synthesized by marine microorganisms exhibit antimicrobial, anti-adhesive and anti-biofilm activity against a broad spectrum of human pathogens (including multi-drug resistant pathogens), and could be used instead of existing drugs to treat infections caused by them. In other cases, these biosurfactants show anti-cancer activity, which could be envisaged as an alternative to conventional therapies. However, marine biosurfactants have not been widely explored, mainly due to the difficulties associated with the isolation and growth of their producing microorganisms. Culture-independent techniques (metagenomics) constitute a promising approach to study the genetic resources of otherwise inaccessible marine microorganisms without the requirement of culturing them, and can contribute to the discovery of novel biosurfactants with significant biological activities. This paper reviews the most relevant biosurfactants produced by marine microorganisms with potential therapeutic applications and discusses future perspectives and opportunities to discover novel molecules from marine environments.
Collapse
|
45
|
Gaoatswe G, Kent BD, Corrigan MA, Nolan G, Hogan AE, McNicholas WT, O'Shea D. Invariant Natural Killer T Cell Deficiency and Functional Impairment in Sleep Apnea: Links to Cancer Comorbidity. Sleep 2015; 38:1629-34. [PMID: 26414901 DOI: 10.5665/sleep.5062] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 05/28/2015] [Indexed: 11/03/2022] Open
Abstract
STUDY OBJECTIVES Emerging evidence links obstructive sleep apnea (OSA) with increased cancer incidence and mortality. Invariant natural killer T (iNKT) cells play an important role in cancer immunity. We hypothesized that patients with OSA have low number of circulating invariant natural killer T (iNKT) cells, which may also be functionally impaired. This study aims to evaluate the frequency of circulating iNKT cells in OSA. DESIGN We evaluated the frequency of circulating iNKT cells by flow cytometry in 33 snorers being assessed for possible OSA. Using iNKT cell lines, we also evaluated the effect of exposure to hypoxia over 24 hours on apoptosis, cytotoxicity, and cytokine production. SETTING Teaching hospital based sleep unit and research laboratory. PATIENTS Thirty-three snorers were evaluated: 9 with no OSA (apnea-hypopnea frequency [AHI] < 5/h), 12 with mild-moderate OSA (AHI 5-30) and 12 with severe OSA (AHI > 30). MEASUREMENTS AND RESULTS Patients with severe OSA had considerably fewer iNKT cells (0.18%) compared to patients with mild-moderate (0.24%) or no OSA (0.35%), P = 0.0026. The frequency of iNKT cells correlated negatively with apnea-hypopnea index (r = -0.58, P = 0.001), oxygen desaturation index (r = -0.58, P = 0.0003), and SpO2% < 90% (r = -0.5407, P = 0.005). The frequency of iNKT cells increased following 12 months of nCPAP therapy (P = 0.015). Hypoxia resulted in increased apoptosis (P = 0.016) and impaired cytotoxicity (P = 0.035). CONCLUSION Patients with obstructive sleep apnea (OSA) have significantly reduced levels of circulating invariant natural killer T (iNKT) cells and hypoxia leads to impaired iNKT cell function. These observations may partly explain the increased cancer risk reported in patients with OSA.
Collapse
Affiliation(s)
- Gadintshware Gaoatswe
- Obesity Immunology Group, Education & Research Center, St Vincent's University Hospital, University College Dublin, Dublin, Ireland
| | - Brian D Kent
- Pulmonary and Sleep Disorders Unit, St Vincent's University Hospital, Dublin, and School of Medicine and Medical Sciences, University College Dublin, Dublin, Ireland
| | - Michelle A Corrigan
- Obesity Immunology Group, Education & Research Center, St Vincent's University Hospital, University College Dublin, Dublin, Ireland
| | - Geraldine Nolan
- Pulmonary and Sleep Disorders Unit, St Vincent's University Hospital, Dublin, and School of Medicine and Medical Sciences, University College Dublin, Dublin, Ireland
| | - Andrew E Hogan
- Obesity Immunology Group, Education & Research Center, St Vincent's University Hospital, University College Dublin, Dublin, Ireland.,Obesity Immunology Group, National Children's Research Center, Dublin, Ireland
| | - Walter T McNicholas
- Pulmonary and Sleep Disorders Unit, St Vincent's University Hospital, Dublin, and School of Medicine and Medical Sciences, University College Dublin, Dublin, Ireland
| | - Donal O'Shea
- Obesity Immunology Group, Education & Research Center, St Vincent's University Hospital, University College Dublin, Dublin, Ireland
| |
Collapse
|
46
|
Van Rhijn I, Godfrey DI, Rossjohn J, Moody DB. Lipid and small-molecule display by CD1 and MR1. Nat Rev Immunol 2015; 15:643-54. [PMID: 26388332 PMCID: PMC6944187 DOI: 10.1038/nri3889] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The antigen-presenting molecules CD1 and MHC class I-related protein (MR1) display lipids and small molecules to T cells. The antigen display platforms in the four CD1 proteins are laterally asymmetrical, so that the T cell receptor (TCR)-binding surfaces are comprised of roofs and portals, rather than the long grooves seen in the MHC antigen-presenting molecules. TCRs can bind CD1 proteins with left-sided or right-sided footprints, creating unexpected modes of antigen recognition. The use of tetramers of human CD1a, CD1b, CD1c or MR1 proteins now allows detailed analysis of the human T cell repertoire, which has revealed new invariant TCRs that bind CD1b molecules and are different from those that define natural killer T cells and mucosal-associated invariant T cells.
Collapse
MESH Headings
- Antigen Presentation/immunology
- Antigens, CD1/chemistry
- Antigens, CD1/immunology
- Antigens, CD1/metabolism
- Histocompatibility Antigens Class I/chemistry
- Histocompatibility Antigens Class I/immunology
- Histocompatibility Antigens Class I/metabolism
- Humans
- Lipids/chemistry
- Lipids/immunology
- Minor Histocompatibility Antigens
- Models, Molecular
- Protein Binding/immunology
- Protein Structure, Tertiary
- Receptors, Antigen, T-Cell/chemistry
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
Collapse
Affiliation(s)
- Ildiko Van Rhijn
- Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Dale I Godfrey
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria 3010, Australia
- Australian Research Council Centre of Excellence for Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Jamie Rossjohn
- Infection and Immunity Program and The Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
- Institute of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, Victoria 3800, Australia
| | - D Branch Moody
- Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
47
|
Neumann S, Young K, Compton B, Anderson R, Painter G, Hook S. Synthetic TRP2 long-peptide and α-galactosylceramide formulated into cationic liposomes elicit CD8+ T-cell responses and prevent tumour progression. Vaccine 2015; 33:5838-5844. [PMID: 26363382 DOI: 10.1016/j.vaccine.2015.08.083] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 08/28/2015] [Accepted: 08/29/2015] [Indexed: 12/22/2022]
Abstract
The lipid antigen α-galactosylceramide (α-GalCer) is a potent activator of invariant natural killer T-cells (iNKT cells) and can stimulate cytotoxic and anti-tumour immune responses. However optimal responses appear to be induced by α-GalCer when cell-based vaccines are delivered intravenously. Here we investigated if co-delivery of protein and peptide antigens along with α-GalCer in a liposomal formulation could stimulate therapeutic anti-tumour immune responses. Cationic liposomes were inherently immune-stimulatory and induced cytotoxic immune responses when delivered both by intravenous and subcutaneous injection. However, only vaccine delivered intravenously stimulated therapeutic anti-tumour immune responses to a peptide antigen. Surface modification with polyethylene glycol (PEG) did not improve immune responses to either intravenously or subcutaneously delivered vaccines. Immune responses to short and long peptide sequences (CD8 and CD4 epitopes) of the self-antigen tyrosinase-related protein 2 (TRP2) as a vaccine antigen, co-delivered with α-GalCer in either cationic liposomes or PBS were further examined. Enhanced production of IFN-γ, increased cytotoxic T-cell responses and tumour survival were observed when a long TRP2-peptide was delivered with α-GalCer in cationic liposomes.
Collapse
Affiliation(s)
- Silke Neumann
- School of Pharmacy, University of Otago, PO Box 56, Dunedin 9054, New Zealand; Department of Pathology, Dunedin School of Medicine, University of Otago, PO Box 56, Dunedin 9054, New Zealand
| | - Katie Young
- School of Pharmacy, University of Otago, PO Box 56, Dunedin 9054, New Zealand; Department of Pathology, Dunedin School of Medicine, University of Otago, PO Box 56, Dunedin 9054, New Zealand
| | - Benji Compton
- Ferrier Research Institute, Victoria University of Wellington, PO Box 33436, Petone, 5046 Wellington, New Zealand
| | - Regan Anderson
- Ferrier Research Institute, Victoria University of Wellington, PO Box 33436, Petone, 5046 Wellington, New Zealand
| | - Gavin Painter
- Ferrier Research Institute, Victoria University of Wellington, PO Box 33436, Petone, 5046 Wellington, New Zealand
| | - Sarah Hook
- School of Pharmacy, University of Otago, PO Box 56, Dunedin 9054, New Zealand.
| |
Collapse
|
48
|
Birkholz AM, Girardi E, Wingender G, Khurana A, Wang J, Zhao M, Zahner S, Illarionov PA, Wen X, Li M, Yuan W, Porcelli SA, Besra GS, Zajonc DM, Kronenberg M. A Novel Glycolipid Antigen for NKT Cells That Preferentially Induces IFN-γ Production. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2015; 195:924-33. [PMID: 26078271 PMCID: PMC4506857 DOI: 10.4049/jimmunol.1500070] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 05/18/2015] [Indexed: 11/19/2022]
Abstract
In this article, we characterize a novel Ag for invariant NKT (iNKT) cells capable of producing an especially robust Th1 response. This glycosphingolipid, DB06-1, is similar in chemical structure to the well-studied α-galactosylceramide (αGalCer), with the only change being a single atom: the substitution of a carbonyl oxygen with a sulfur atom. Although DB06-1 is not a more effective Ag in vitro, the small chemical change has a marked impact on the ability of this lipid Ag to stimulate iNKT cells in vivo, with increased IFN-γ production at 24 h compared with αGalCer, increased IL-12, and increased activation of NK cells to produce IFN-γ. These changes are correlated with an enhanced ability of DB06-1 to load in the CD1d molecules expressed by dendritic cells in vivo. Moreover, structural studies suggest a tighter fit into the CD1d binding groove by DB06-1 compared with αGalCer. Surprisingly, when iNKT cells previously exposed to DB06-1 are restimulated weeks later, they have greatly increased IL-10 production. Therefore, our data are consistent with a model whereby augmented and or prolonged presentation of a glycolipid Ag leads to increased activation of NK cells and a Th1-skewed immune response, which may result, in part, from enhanced loading into CD1d. Furthermore, our data suggest that strong antigenic stimulation in vivo may lead to the expansion of IL-10-producing iNKT cells, which could counteract the benefits of increased early IFN-γ production.
Collapse
Affiliation(s)
- Alysia M Birkholz
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037; Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037; Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92037
| | - Enrico Girardi
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037
| | - Gerhard Wingender
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037
| | - Archana Khurana
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037
| | - Jing Wang
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037
| | - Meng Zhao
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037
| | - Sonja Zahner
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037
| | - Petr A Illarionov
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Xiangshu Wen
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
| | - Michelle Li
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
| | - Weiming Yuan
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
| | - Steven A Porcelli
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461; and Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Gurdyal S Besra
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Dirk M Zajonc
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037
| | - Mitchell Kronenberg
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037; Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92037;
| |
Collapse
|
49
|
Nakamura T, Kuroi M, Harashima H. Influence of Endosomal Escape and Degradation of α-Galactosylceramide Loaded Liposomes on CD1d Antigen Presentation. Mol Pharm 2015; 12:2791-9. [PMID: 26107189 DOI: 10.1021/mp500704e] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Alpha-galactosylceramide (GC), a lipid antigen present on CD1d molecules, is a unique adjuvant that enables a strong antitumor effect to be induced via activation of natural killer T cells. We previously reported that a liposomal formulation of GC significantly enhanced GC presentation via CD1d and antitumor immunity. However, the influence of the intracellular fate of liposomes controlled by the lipid composition on GC presentation using GC-loaded liposomes (GC-Lip) remains unclear. In this study, we prepared a GC-Lip formulation by incorporating dioleoyl-phosphatidylethanolamine (DOPE)/cholesterol, egg phosphatidylcholine (EPC)/cholesterol, and distearoyl phosphocholine (DSPC)/cholesterol, and investigated the relationship between the intracellular trafficking of GC-Lip and GC presentation in antigen-presenting cells. When GC-Lip was prepared using DOPE, a fusogenic lipid, the endosomal escape of liposomes was enhanced, resulting in a decrease in GC presentation of CD1d, compared to the EPC based GC-Lip (EPC/GC-Lip). The stability of liposomes in endosomes/lysosomes had no influence on GC presentation. The DSPC based GC-Lip (DSPC/GC-Lip) induced GC presentation without any detectable degradation in liposomal structure, although the EPC/GC-Lip induced GC presentation with degradation of liposomal structure. The efficiency of GC presentation between EPC/GC-Lip and DSPC/GC-Lip was comparable. These GC presentations that were independent of the degradation of liposomes were dominated by saposins, sphingolipid activator proteins. Our findings reveal that GC presentation on CD1d from the fluid liposomes involves the action of saposins, regardless of whether liposome degradation occurs. This insight can be of use in terms of developing GC-Lip formulation for efficient GC presentation.
Collapse
Affiliation(s)
- Takashi Nakamura
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Moeka Kuroi
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Hideyoshi Harashima
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| |
Collapse
|
50
|
Werter IM, Schneiders FL, Scotet E, Verheul HMW, de Gruijl TD, van der Vliet HJ. Vγ9Vδ2-T cells as antigen presenting cells for iNKT cell based cancer immunotherapy. Oncoimmunology 2015; 3:e955343. [PMID: 25941612 DOI: 10.4161/21624011.2014.955343] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 07/28/2014] [Indexed: 11/19/2022] Open
Abstract
CD1d-restricted invariant natural killer T cells (iNKT) constitute an important immunoregulatory T-cell subset involved in the induction of antitumor immune responses. Here, we provide a view on the recent observation that Vγ9Vδ2-T cells, through trogocytosis of CD1d-containing membrane fragments, have the capacity to act as antigen presenting cells for iNKT.
Collapse
Key Words
- APC
- APC, antigen presenting cells
- Ag, antigens
- BTN, butyrophilin
- BrHPP, bromohydrin pyrophosphate
- CD, Cluster of differentiation
- CD1d
- DN, double negative
- GM-CSF, granulocyte-macrophage colony-stimulating factor
- IFNγ, interferon γ
- IL, interleukine
- IPP, isopentenyl pyrophosphate
- MHC, major histocompatibility complex
- NK, natural killer cells
- PBMC, peripheral blood mononuclear cells
- TNFα, tumor necrosis factor α
- Th1, Type 1 T helper
- Th2, Type 2 T helper
- Vγ9Vδ2-T cells
- i.v., intravenous
- iNKT
- iNKT, invariant natural killer T cells
- moDC, monocyte derived dendritic cells
- pAg, phosphoantigens
- trogocytosis
- α-GalCer, α-galactosylceramide
Collapse
Affiliation(s)
- Inge M Werter
- Department of Medical Oncology; VU University Medical Center ; Amsterdam, The Netherlands
| | - Famke L Schneiders
- Department of Medical Oncology; VU University Medical Center ; Amsterdam, The Netherlands
| | - Emmanuel Scotet
- Institut National de la Santé et de la Recherche Médicale; UMR892; Center de Recherche en Cancérologie Nantes-Angers ; Nantes Cedex, France
| | - Henk M W Verheul
- Department of Medical Oncology; VU University Medical Center ; Amsterdam, The Netherlands
| | - Tanja D de Gruijl
- Department of Medical Oncology; VU University Medical Center ; Amsterdam, The Netherlands
| | - Hans J van der Vliet
- Department of Medical Oncology; VU University Medical Center ; Amsterdam, The Netherlands
| |
Collapse
|