1
|
Srivastava P, Rütter M, Antoniraj G, Ventura Y, David A. Dendritic Cell-Targeted Nanoparticles Enhance T Cell Activation and Antitumor Immune Responses by Boosting Antigen Presentation and Blocking PD-L1 Pathways. ACS APPLIED MATERIALS & INTERFACES 2024; 16:53577-53590. [PMID: 39344665 DOI: 10.1021/acsami.4c12821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Dendritic cells (DCs) within the tumor microenvironment (TME) have an insufficient capacity to activate T cells through antigen presentation. Furthermore, the programmed cell-death ligand 1 (PD-L1), abundantly expressed on tumor-associated DCs, binds the programmed cell-death 1 (PD-1)-positive T cells and suppresses their immune function. The binding of PD-L1 to CD80 (B7.1) on the same DC via cis-interactions further prevents T cell costimulation through CD28. Here, we present a strategy to simultaneously promote antigen cross-presentation and block the inhibitory interactions of PD-L1 on DCs to amplify T cell-mediated antitumor responses within the TME. Mesoporous silica nanoparticles (MSNPs) were loaded with clotrimazole (CLT) to boost MHC II-mediated antigen presentation by DCs, surface-modified with mannose to target CD206 on DCs, and then decorated with PD-L1 binding peptide (PDL1bp) to block PD-L1-mediated interactions. PDL1bp was cleaved from the mannosylated and CLT-loaded MSNPs (MSNP-MaN/CLT) under conditions simulating the TME and tethered to PD-L1 to reverse CD80 sequestration on DC2.4 cells. The blocking of PD-L1 by PDL1bp-decorated NPs (MSNP-MaN-PDL1bp) increased the cellular interactions between DC2.4 and EL4 T cells and the amount of IL-2 secretion. The MSNP-MaN/CLT were taken up rapidly by DC2.4 cells, promoted MHC II presentation of hen egg lysozyme (HEL), and increased IL-2 production from HEL antigen-primed 3A9 T cells, which was further enhanced by PDL1bp. In vivo investigation revealed that administration of the CLT-loaded and PDL1bp-functionalized MSNPs remarkably inhibited subcutaneous B16-F10 melanoma tumor growth when compared with anti-PD-L1 therapy. MSNP-MaN-PDL1bp/CLT treatment upregulated the levels of effector molecules such as granzyme B and proinflammatory cytokines (IFNγ and INFα) in the tumor tissue, indicating antitumoral T cell responses. This strategy of utilizing nanoparticles to trigger DC activation while promoting T cell stimulation can be used to amplify the antitumor T cell responses and represents a promising alternative to anti-PD-L1 immunotherapy.
Collapse
Affiliation(s)
- Prateek Srivastava
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Marie Rütter
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Gover Antoniraj
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Yvonne Ventura
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Ayelet David
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| |
Collapse
|
2
|
Zeng T, Zang W, Xiao H, Jiang Y, Lin S, Wang M, Li S, Li L, Li C, Lu C, Yang H. Carrier-Free Nanovaccine: An Innovative Strategy for Ultrahigh Melanoma Neoantigen Loading. ACS NANO 2023; 17:18114-18127. [PMID: 37695697 DOI: 10.1021/acsnano.3c04887] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/13/2023]
Abstract
In personalized cancer immunotherapy, developing an effective neoantigen nanovaccine with high immunogenicity is a significant challenge. Traditional nanovaccine delivery systems often require nanocarriers, which can hinder the delivery of the neoantigen and cause significant toxicity. In this study, we present an innovative strategy of carrier-free nanovaccine achieved through direct self-assembly of 2'-fluorinated CpG (2'F-CpG) with melanoma neoantigen peptide (Obsl1). Molecular dynamics simulations demonstrated that the introduction of a fluorine atom into CpG increases the noncovalent interaction between 2'F-CpG and Obsl1, which enhanced the loading of Obsl1 on 2'F-CpG, resulting in the spontaneous formation of a hybrid 2'F-CpG/Obsl1 nanovaccine. This nanovaccine without extra nanocarriers showed ultrahigh Obsl1 loading up to 83.19 wt %, increasing the neoantigen peptide uptake by antigen-presenting cells (APCs). In C57BL/6 mice models, we demonstrated the long-term preventive and therapeutic effects of the prepared 2'F-CpG/Obsl1 nanovaccine against B16F10 melanoma. Immunocellular analysis revealed that the nanovaccine activated innate and adaptive immune responses to cancer cells. Hence, this study established a simple, safe, and effective preparation strategy for a carrier-free neoantigen nanovaccine, which could be adapted for the future design of personalized cancer vaccines in clinical settings.
Collapse
Affiliation(s)
- Tao Zeng
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350108, People's Republic of China
| | - Weijie Zang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350108, People's Republic of China
| | - Han Xiao
- State Key Laboratory of Structure of Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences Fuzhou, Fujian 350002, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Yifan Jiang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350108, People's Republic of China
| | - Sang Lin
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350108, People's Republic of China
| | - Min Wang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350108, People's Republic of China
| | - Shiqing Li
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350108, People's Republic of China
| | - Liannishang Li
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350108, People's Republic of China
| | - Chunsen Li
- State Key Laboratory of Structure of Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences Fuzhou, Fujian 350002, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Chunhua Lu
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350108, People's Republic of China
| | - Huanghao Yang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350108, People's Republic of China
| |
Collapse
|
3
|
Bellato F, Feola S, Dalla Verde G, Bellio G, Pirazzini M, Salmaso S, Caliceti P, Cerullo V, Mastrotto F. Mannosylated Polycations Target CD206 + Antigen-Presenting Cells and Mediate T-Cell-Specific Activation in Cancer Vaccination. Biomacromolecules 2022; 23:5148-5163. [PMID: 36394394 DOI: 10.1021/acs.biomac.2c00993] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Immunotherapy is deemed one of the most powerful therapeutic approaches to treat cancer. However, limited response and tumor specificity are still major challenges to address. Herein, mannosylated polycations targeting mannose receptor- are developed as vectors for plasmid DNA (pDNA)-based vaccines to improve selective delivery of genetic material to antigen-presenting cells and enhance immune cell activation. Three diblock glycopolycations (M15A12, M29A25, and M58A45) and two triblock copolymers (M29A29B9 and M62A52B32) are generated by using mannose (M), agmatine (A), and butyl (B) derivatives to target CD206, complex nucleic acids, and favor the endosomal escape, respectively. All glycopolycations efficiently complex pDNA at N/P ratios <5, protecting the pDNA from degradation in a physiological milieu. M58A45 and M62A52B32 complexed with plasmid encoding for antigenic ovalbumin (pOVA) trigger the immune activation of cultured dendritic cells, which present the SIINFEKL antigenic peptide via specific major histocompatibility complex-I. Importantly, administration of M58A45/pOVA elicits SIINFEKL-specific T-cell response in C56BL/6 mice bearing the melanoma tumor model B16-OVA, well in line with a reduction in tumor growth. These results qualify mannosylation as an efficient strategy to target immune cells in cancer vaccination and emphasize the potential of these glycopolycations as effective delivery vehicles for nucleic acids.
Collapse
Affiliation(s)
- Federica Bellato
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131Padova, Italy
| | - Sara Feola
- Drug Research Program ImmunoViroTherapy Lab (IVT), Faculty of Pharmacy, Helsinki University, Viikinkaari 5E, 00790Helsinki, Finland.,iCAN Digital Precision Cancer Medicine Flagship, FI-00014Helsinki, Finland
| | - Gloria Dalla Verde
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131Padova, Italy
| | - Greta Bellio
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131Padova, Italy
| | - Marco Pirazzini
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/B, 35131Padova, Italy
| | - Stefano Salmaso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131Padova, Italy
| | - Paolo Caliceti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131Padova, Italy
| | - Vincenzo Cerullo
- Drug Research Program ImmunoViroTherapy Lab (IVT), Faculty of Pharmacy, Helsinki University, Viikinkaari 5E, 00790Helsinki, Finland.,iCAN Digital Precision Cancer Medicine Flagship, FI-00014Helsinki, Finland
| | - Francesca Mastrotto
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131Padova, Italy
| |
Collapse
|
4
|
Xie X, Feng Y, Zhang H, Su Q, Song T, Yang G, Li N, Wei X, Li T, Qin X, Li S, Wu C, Zhang X, Wang G, Liu Y, Yang H. Remodeling tumor immunosuppressive microenvironment via a novel bioactive nanovaccines potentiates the efficacy of cancer immunotherapy. Bioact Mater 2022; 16:107-119. [PMID: 35386322 PMCID: PMC8958467 DOI: 10.1016/j.bioactmat.2022.03.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 03/01/2022] [Accepted: 03/03/2022] [Indexed: 02/06/2023] Open
Abstract
The clinical outcomes of cancer nanovaccine have been largely impeded owing to the low antigen-specific T cell response rate and acquired resistance caused by the immunosuppressive tumor microenvironment (TME). Here, we reported a tumor acidity-responsive nanovaccine to remodel the immunosuppressive TME and expand the recruitment of tumor infiltrating lymphocytes (TILs) using hybrid micelles (HM), which encapsulated colony stimulating factor 1 receptor (CSF1-R) inhibitor BLZ-945 and indoleamine 2,3-dioxygenase (IDO) inhibitor NLG-919 in its core and displayed a model antigen ovalbumin (OVA) on its surface (denoted as BN@HM-OVA). The bioactive nanovaccine is coated with a polyethylene glycol (PEG) shell for extending nanoparticle circulation. The shell can be shed in response to the weakly acidic tumor microenvironment. The decrease in size and the increase in positive charge may cause the deep tumor penetration of drugs. We demonstrated that the bioactive nanovaccine dramatically enhance antigen presentation by dendritic cells (DCs) and drugs transportation into M1-like tumor-associated macrophages (TAMs) and tumor cells via size reduction and increasing positive charge caused by the weakly acidic TME. Such bioactive nanovaccine could remodel the immunosuppressive TME into an effector T cells favorable environment, leading to tumor growth inhibition in prophylactic and therapeutic E.G7-OVA tumor models. Furthermore, combining the bioactive nanovaccine with simultaneous anti-PD-1 antibody treatment leads to a long-term tumor inhibition, based on the optimal timing and sequence of PD-1 blockade against T cell receptor. This research provides a new strategy for the development of efficient cancer immunotherapy. A bioactive nanovaccine (BN@HM-OVA) was adopted for synergistic immunotherapy of E.G7-OVA tumors. BN@HM-OVA exhibited superior ability to induce DCs maturation and robust antigen-specific T cell responses. BN@HM-OVA contributed to a homeostasis in the tumor microenvironment ideal for antitumor vaccination. The combination treatment of BN@HM-OVA and αPD-1 achieved maximum therapeutic benefits.
Collapse
|
5
|
Huang T, Li S, Fang J, Li F, Tu S. Antibody-activated trans-endothelial delivery of mesoporous organosilica nanomedicine augments tumor extravasation and anti-cancer immunotherapy. Bioact Mater 2021; 6:2158-2172. [PMID: 33511314 PMCID: PMC7815474 DOI: 10.1016/j.bioactmat.2020.12.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 12/23/2020] [Accepted: 12/28/2020] [Indexed: 11/18/2022] Open
Abstract
Tumor vasculature constitutes a formidable hurdle for the efficient delivery of cancer nanomedicine into tumors. The leverage of passive pathway through inter-endothelial gaps in tumor blood vessels might account for limited extravasation of nanomedicine into tumor microenvironment (TME). Herein, Annexin A1 antibody-installed mesoporous organosilica nanoplatforms carrying immunotherapeutics of anti-PD-L1 antibody (aPD-L1) and Indoximod are developed to target at caveolar Annexin-A1 protein of luminal endothelial cells and to trigger the active trans-endothelial transcytosis of nanomedicine mediated by caveolae. Such strategy enables rapid nanomedicine extravasation across tumor endothelium and relatively extensive accumulation in tumor interstitium. aPD-L1 and Indoximod release from aPD/IND@MON-aANN in a reduction-responsive manner and synergistically facilitate the intratumoral infiltration of cytotoxic T lymphocytes and reverse the immunosuppressive TME, thus demonstrating substantial anti-tumor efficacy in subcutaneous 4T1 breast tumors and remarkable anti-metastatic capacity to extend the survival of 4T1 tumor metastasis model. Moreover, aPD/IND@MON-aANN nanomedicine also exhibits distinct superiority over the combination therapy of free drugs to potently attenuate the progression of urethane-induced orthotopic lung cancers. Collectively, aPD/IND@MON-aANN nanoplatforms with boosted delivery efficiency via antibody-activated trans-endothelial pathway and enhanced immunotherapeutic efficacy provides perspectives for the development of cancer nanomedicines. The nanomedicine overcomes tumor vascular barrier by active transcytosis via caveolae initiated by the conjugated aANXA1. The nanoplatform responsively releases aPD-L1 and Indoximod to synergistically improve the efficacy of immunotherapy. The nanomedicine shows anti-tumor capacity in mice breast cancers and lung cancers.
Collapse
Affiliation(s)
- Tinglei Huang
- Department of Oncology, State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Shuang Li
- Department of Stomatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Jianchen Fang
- Department of Pathology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Fuli Li
- Department of Oncology, State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Shuiping Tu
- Department of Oncology, State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
- Corresponding author.
| |
Collapse
|
6
|
Cheng S, Xu C, Jin Y, Li Y, Zhong C, Ma J, Yang J, Zhang N, Li Y, Wang C, Yang Z, Wang Y. Artificial Mini Dendritic Cells Boost T Cell-Based Immunotherapy for Ovarian Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1903301. [PMID: 32274314 PMCID: PMC7141030 DOI: 10.1002/advs.201903301] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 01/15/2020] [Indexed: 05/17/2023]
Abstract
Ovarian cancer is the most lethal gynecological malignancy with high recurrence rates and low survival rates, remaining a disease of high unmet need. Cancer immunotherapy, which harnesses the potential of the immune system to attack tumors, has emerged as one of the most promising treatment options in recent years. As an important form of immunotherapy, dendritic cell (DC)-based vaccines have demonstrated the ability to induce an immune response, while clinical efficacy of DC vaccines remains unsubstantiated as long-term benefit is only reported in a restricted proportion of patients. Here, a biomimetic nanovaccine derived from DCs is developed through cell membrane coating nanotechnology. This nanovaccine, denoted "mini DC," inherits the ability of antigen presentation and T cells' stimulation from DCs and is shown to elicit enhanced activation of T cells both in vitro and in vivo. In a mouse model of ovarian cancer, mini DCs exhibit superior therapeutic and prophylactic efficacy against cancer including delayed tumor growth and reduced tumor metastasis compared with DC vaccine. These findings suggest that mini DCs may serve as a facile and potent vaccine to boost anticancer immunotherapy.
Collapse
Affiliation(s)
- Shanshan Cheng
- Department of Obstetrics and GynecologyShanghai Key Laboratory of Gynecologic OncologyRenji HospitalSchool of MedicineShanghai Jiaotong UniversityShanghai200127P. R. China
| | - Cong Xu
- Department of Obstetrics and GynecologyShanghai Key Laboratory of Gynecologic OncologyRenji HospitalSchool of MedicineShanghai Jiaotong UniversityShanghai200127P. R. China
| | - Yue Jin
- Department of Obstetrics and GynecologyShanghai Key Laboratory of Gynecologic OncologyRenji HospitalSchool of MedicineShanghai Jiaotong UniversityShanghai200127P. R. China
| | - Yu Li
- Department of Obstetrics and GynecologyShanghai Key Laboratory of Gynecologic OncologyRenji HospitalSchool of MedicineShanghai Jiaotong UniversityShanghai200127P. R. China
| | - Cheng Zhong
- Department of Obstetrics and GynecologyShanghai Key Laboratory of Gynecologic OncologyRenji HospitalSchool of MedicineShanghai Jiaotong UniversityShanghai200127P. R. China
| | - Jun Ma
- Department of Obstetrics and GynecologyShanghai Key Laboratory of Gynecologic OncologyRenji HospitalSchool of MedicineShanghai Jiaotong UniversityShanghai200127P. R. China
| | - Jiani Yang
- Department of Obstetrics and GynecologyShanghai Key Laboratory of Gynecologic OncologyRenji HospitalSchool of MedicineShanghai Jiaotong UniversityShanghai200127P. R. China
| | - Nan Zhang
- Department of Obstetrics and GynecologyShanghai Key Laboratory of Gynecologic OncologyRenji HospitalSchool of MedicineShanghai Jiaotong UniversityShanghai200127P. R. China
| | - Yuan Li
- Department of Obstetrics and GynecologyShanghai Key Laboratory of Gynecologic OncologyRenji HospitalSchool of MedicineShanghai Jiaotong UniversityShanghai200127P. R. China
| | - Chao Wang
- Department of Obstetrics and GynecologyShanghai Key Laboratory of Gynecologic OncologyRenji HospitalSchool of MedicineShanghai Jiaotong UniversityShanghai200127P. R. China
| | - Zhiyou Yang
- Department of Obstetrics and GynecologyShanghai Key Laboratory of Gynecologic OncologyRenji HospitalSchool of MedicineShanghai Jiaotong UniversityShanghai200127P. R. China
| | - Yu Wang
- Department of Obstetrics and GynecologyShanghai Key Laboratory of Gynecologic OncologyRenji HospitalSchool of MedicineShanghai Jiaotong UniversityShanghai200127P. R. China
| |
Collapse
|
7
|
Conniot J, Scomparin A, Peres C, Yeini E, Pozzi S, Matos AI, Kleiner R, Moura LIF, Zupančič E, Viana AS, Doron H, Gois PMP, Erez N, Jung S, Satchi-Fainaro R, Florindo HF. Immunization with mannosylated nanovaccines and inhibition of the immune-suppressing microenvironment sensitizes melanoma to immune checkpoint modulators. NATURE NANOTECHNOLOGY 2019; 14:891-901. [PMID: 31384037 DOI: 10.1038/s41565-019-0512-0] [Citation(s) in RCA: 158] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 06/18/2019] [Indexed: 05/18/2023]
Abstract
A low response rate, acquired resistance and severe side effects have limited the clinical outcomes of immune checkpoint therapy. Here, we show that combining cancer nanovaccines with an anti-PD-1 antibody (αPD-1) for immunosuppression blockade and an anti-OX40 antibody (αOX40) for effector T-cell stimulation, expansion and survival can potentiate the efficacy of melanoma therapy. Prophylactic and therapeutic combination regimens of dendritic cell-targeted mannosylated nanovaccines with αPD-1/αOX40 demonstrate a synergism that stimulates T-cell infiltration into tumours at early treatment stages. However, this treatment at the therapeutic regimen does not result in an enhanced inhibition of tumour growth compared to αPD-1/αOX40 alone and is accompanied by an increased infiltration of myeloid-derived suppressor cells in tumours. Combining the double therapy with ibrutinib, a myeloid-derived suppressor cell inhibitor, leads to a remarkable tumour remission and prolonged survival in melanoma-bearing mice. The synergy between the mannosylated nanovaccines, ibrutinib and αPD-1/αOX40 provides essential insights to devise alternative regimens to improve the efficacy of immune checkpoint modulators in solid tumours by regulating the endogenous immune response.
Collapse
Affiliation(s)
- João Conniot
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Anna Scomparin
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Carina Peres
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Eilam Yeini
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sabina Pozzi
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ana I Matos
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Ron Kleiner
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Liane I F Moura
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Eva Zupančič
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Ana S Viana
- Center of Chemistry and Biochemistry, Faculty of Sciences, Universidade de Lisboa, Lisbon, Portugal
| | - Hila Doron
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Pedro M P Gois
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Neta Erez
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Steffen Jung
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Helena F Florindo
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
8
|
The role of CD95 and CD95 ligand in cancer. Cell Death Differ 2015; 22:549-59. [PMID: 25656654 PMCID: PMC4356349 DOI: 10.1038/cdd.2015.3] [Citation(s) in RCA: 193] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 12/27/2014] [Accepted: 01/02/2015] [Indexed: 02/07/2023] Open
Abstract
CD95 (Fas/APO-1) and its ligand, CD95L, have long been viewed as a death receptor/death ligand system that mediates apoptosis induction to maintain immune homeostasis. In addition, these molecules are important in the immune elimination of virus-infected cells and cancer cells. CD95L was, therefore, considered to be useful for cancer therapy. However, major side effects have precluded its systemic use. During the last 10 years, it has been recognized that CD95 and CD95L have multiple cancer-relevant nonapoptotic and tumor-promoting activities. CD95 and CD95L were discovered to be critical survival factors for cancer cells, and were found to protect and promote cancer stem cells. We now discuss five different ways in which inhibiting or eliminating CD95L, rather than augmenting, may be beneficial for cancer therapy alone or in combination with standard chemotherapy or immune therapy.
Collapse
|
9
|
Abstract
The conventional view of CD95 (Fas/APO-1) is that it is a dedicated apoptosis-inducing receptor with important functions in immune cell homeostasis and in viral and tumor defense. There is an emerging recognition, however, that CD95 also has multiple non-apoptotic activities. In the context of cancer, CD95 was shown to have tumor-promoting activities, and the concept of this new function of CD95 in cancer is gaining traction. Recently, we showed that not only is CD95 a growth promoter for cancer cells, but, paradoxically, when either CD95 or CD95 ligand (CD95L) is removed, that virtually all cancer cells die through a process we have named DICE (death induced by CD95R/L elimination). In this perspective, I outline a hypothesis regarding the physiological function of DICE, and why it may be possible to use induction of DICE to treat many, if not most, cancers.
Collapse
Affiliation(s)
- Marcus E Peter
- Northwestern University; Feinberg School of Medicine; Division Hematology/Oncology; Chicago, IL USA
| |
Collapse
|
10
|
Quatromoni JG, Suzuki E, Okusanya O, Judy BF, Bhojnagarwala P, Venegas O, Eruslanov E, Predina JD, Albelda SM, Singhal S. The timing of TGF-β inhibition affects the generation of antigen-specific CD8+ T cells. BMC Immunol 2013; 14:30. [PMID: 23865808 PMCID: PMC3725164 DOI: 10.1186/1471-2172-14-30] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Accepted: 07/02/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Transforming growth factor (TGF)-β is a potent immunosuppressive cytokine necessary for cancer growth. Animal and human studies have shown that pharmacologic inhibition of TGF-β slows the growth rate of established tumors and occasionally eradicates them altogether. We observed, paradoxically, that inhibiting TGF-β before exposing animals to tumor cells increases tumor growth kinetics. We hypothesized that TGF-β is necessary for the anti-tumor effects of cytotoxic CD8+ T lymphocytes (CTLs) during the early stages of tumor initiation. METHODS BALB/c mice were pretreated with a blocking soluble TGF-β receptor (sTGF-βR, TGF-β-blockade group, n=20) or IgG2a (Control group, n=20) before tumor inoculation. Tumor size was followed for 6 weeks. In vivo lymphocyte assays and depletion experiments were then performed to investigate the immunological basis of our results. Lastly, animals were pretreated with either sTGF-βR (n=6) or IgG2a (n=6) prior to immunization with an adenoviral vector encoding the human papillomavirus E7 gene (Ad.E7). One week later, flow cytometry was utilized to measure the number of splenic E7-specific CD8+ T cells. RESULTS Inhibition of TGF-β before the injection of tumor cells resulted in significantly larger average tumor volumes on days 11, 17, 22, 26 and 32 post tumor-inoculation (p < 0.05). This effect was due to the inhibition of CTLs, as it was not present in mice with severe combined immunodeficiency (SCID) or those depleted of CD8+ T cells. Furthermore, pretreatment with sTGF-βR inhibited tumor-specific CTL activity in a Winn Assay. Tumors grew to a much larger size when mixed with CD8+ T cells from mice pretreated with sTGF-βR than when mixed with CD8+ T cells from mice in the control group: 96 mm3 vs. 22.5 mm3, respectively (p < 0.05). In addition, fewer CD8+ T cells were generated in Ad.E7-immunized mice pretreated with sTGF-βR than in mice from the control group: 0.6% total CD8+ T cells vs. 1.9%, respectively (p < 0.05). CONCLUSIONS These studies provide the first in vivo evidence that TGF-β may be necessary for anti-tumor immune responses in certain cancers. This finding has important implications for our understanding of anti-tumor immune responses, the role of TGF-β in the immune system, and the future development of TGF-β inhibiting drugs.
Collapse
Affiliation(s)
- Jon G Quatromoni
- Division of Thoracic Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Xie FJ, Zhao P, Zhang YP, Liu FY, Nie XL, Zhu YH, Yu XM, Zheng QQ, Mao WM, Lu HY, Wei H, Huang W. Adenovirus-mediated interferon-γ gene therapy induced human pancreatic carcinoma Capan-2 cell apoptosis in vitro and in vivo. Anat Rec (Hoboken) 2013; 296:604-10. [PMID: 23401468 DOI: 10.1002/ar.22661] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Accepted: 12/19/2012] [Indexed: 12/30/2022]
Abstract
Pancreatic cancer is one of the most lethal human malignancies with a very low 5-year survival rate, which highlights urgent needs for more effective therapeutic strategies. In this study, we examined the potential therapeutic effects of an adenovirus encoding human interferon gamma (Ad-IFNγ) on pancreatic carcinoma cells Capan-2 in vitro and in vivo. The results indicated that Ad-IFNγ could significantly inhibit tumor cell growth via inducing cell apoptosis. After infection, IFNγ expressed durably and stably in xenografts, predominantly in tumor tissue, while much less in blood and liver. Thus, adenovirus-mediated intratumoral injection of human IFNγ gene could be an effective gene therapeutic system for the treatment of pancreatic carcinoma.
Collapse
Affiliation(s)
- Fa-Jun Xie
- Department of Medical Oncology, Zhejiang Cancer Hospital, HangZhou, 310022, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Kim JH, Hur JH, Lee SM, Im KS, Kim NH, Sur JH. Correlation of Foxp3 positive regulatory T cells with prognostic factors in canine mammary carcinomas. Vet J 2011; 193:222-7. [PMID: 22130461 DOI: 10.1016/j.tvjl.2011.10.022] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Revised: 10/03/2011] [Accepted: 10/27/2011] [Indexed: 10/15/2022]
Abstract
Regulatory T cells (Treg) cells play a crucial role in tumor progression by suppressing anti-tumor immunity, but are not well-documented in veterinary oncology. To identify the characteristics of Treg cells in tumor microenvironments, the numbers of Treg cells were analyzed and compared with histological prognostic factors and molecular biomarkers in canine mammary carcinoma (MC) tissues (n=37). Abundant Treg cells were associated with high histological grade and lymphatic invasion. The numbers of Treg cells infiltrating intratumoral areas markedly increased in tumors with poor prognostic factors, such as high histological grade, lymphatic invasion, and necrosis. These findings suggest that Treg cells play a role in canine MC progression. Furthermore, Treg cell numbers in intratumoral compartments may provide a potential prognostic factor when assessing canine MCs, which may in turn lead to the development of new immunologic therapeutics.
Collapse
Affiliation(s)
- J H Kim
- Department of Veterinary Pathology, Small Animal Tumor Diagnostic Center, College of Veterinary Medicine, Konkuk University, 1 Hwayang-dong, Kwangjin-gu, Seoul 143-701, Republic of Korea
| | | | | | | | | | | |
Collapse
|
13
|
Mempel TR, Bauer CA. Intravital imaging of CD8+ T cell function in cancer. Clin Exp Metastasis 2008; 26:311-27. [PMID: 18665448 DOI: 10.1007/s10585-008-9196-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2008] [Accepted: 07/14/2008] [Indexed: 12/11/2022]
Abstract
Recent technological advances in photonics are making intravital microscopy (IVM) an increasingly powerful approach for the mechanistic exploration of biological processes in the physiological context of complex native tissue environments. Direct, dynamic and multiparametric visualization of immune cell behavior in living animals at cellular and subcellular resolution has already proved its utility in auditing basic immunological concepts established through conventional approaches and has also generated new hypotheses that can conversely be complemented and refined by traditional experimental methods. The insight that outgrowing tumors must not necessarily have evaded recognition by the adaptive immune system, but can escape rejection by actively inducing a state of immunological tolerance calls for a detailed investigation of the cellular and molecular mechanisms by which the anti-cancer response is subverted. Along with molecular imaging techniques that provide dynamic information at the population level, IVM can be expected to make a critical contribution to this effort by allowing the observation of immune cell behavior in vivo at single cell-resolution. We review here how IVM-based investigation can help to clarify the role of cytotoxic T lymphocytes (CTL) in the immune response against cancer and identify the ways by which their function might be impaired through tolerogenic mechanisms.
Collapse
Affiliation(s)
- Thorsten R Mempel
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA.
| | | |
Collapse
|
14
|
Hallermalm K, Johansson S, Bråve A, Ek M, Engström G, Boberg A, Gudmundsdotter L, Blomberg P, Mellstedt H, Stout R, Liu MA, Wahren B. Pre-clinical evaluation of a CEA DNA prime/protein boost vaccination strategy against colorectal cancer. Scand J Immunol 2007; 66:43-51. [PMID: 17587345 DOI: 10.1111/j.1365-3083.2007.01945.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In preparation for a clinical trial in patients diagnosed with colorectal cancer, a vaccination strategy targeting the carcinoembryonic antigen (CEA) was evaluated in mice using a GMP-produced plasmid DNA vaccine, CEA66, encoding a truncated form of the tumour-associated antigen, CEA. The GMP-produced CEA DNA vaccine was also evaluated for toxicity. Repeated intradermal administration of the GMP-produced vaccine using a novel needle-free jet injection device (Biojector) induced robust CD4 and CD8 T-cell responses in mice, and did not result in any vaccine-related toxicity. In a heterologous DNA prime/protein boost setting, cellular immune responses were of higher magnitude in animals primed with CEA66 DNA than in animals receiving repeated doses of recombinant CEA protein. These responses were further enhanced if recombinant murine granulocyte-macrophage colony-stimulating factor was given as an adjuvant prior to vaccination. In contrast to repeated administration of recombinant CEA protein as a single modality vaccine, the heterologous CEA66 DNA prime/rCEA boost vaccination strategy resulted in a qualitatively broader immune response, and supports clinical testing of this vaccination regimen in humans.
Collapse
Affiliation(s)
- K Hallermalm
- Department of Microbiology and Tumor and Cell Biology, Karolinska Institutet & Swedish Institute for Infectious Disease ControlCenter for Molecular Medicine, Karolinska InstitutetVecura, Cell & Gene Therapy Center, Karolinska SjukhusetCancer Center Karolinska, Karolinska Sjukhuset, Stockholm, SwedenBioject Medical Technologies Inc., Tualatin, OR, USAProTherImmune, Lafayette, CA, USA
| | - S Johansson
- Department of Microbiology and Tumor and Cell Biology, Karolinska Institutet & Swedish Institute for Infectious Disease ControlCenter for Molecular Medicine, Karolinska InstitutetVecura, Cell & Gene Therapy Center, Karolinska SjukhusetCancer Center Karolinska, Karolinska Sjukhuset, Stockholm, SwedenBioject Medical Technologies Inc., Tualatin, OR, USAProTherImmune, Lafayette, CA, USA
| | - A Bråve
- Department of Microbiology and Tumor and Cell Biology, Karolinska Institutet & Swedish Institute for Infectious Disease ControlCenter for Molecular Medicine, Karolinska InstitutetVecura, Cell & Gene Therapy Center, Karolinska SjukhusetCancer Center Karolinska, Karolinska Sjukhuset, Stockholm, SwedenBioject Medical Technologies Inc., Tualatin, OR, USAProTherImmune, Lafayette, CA, USA
| | - M Ek
- Department of Microbiology and Tumor and Cell Biology, Karolinska Institutet & Swedish Institute for Infectious Disease ControlCenter for Molecular Medicine, Karolinska InstitutetVecura, Cell & Gene Therapy Center, Karolinska SjukhusetCancer Center Karolinska, Karolinska Sjukhuset, Stockholm, SwedenBioject Medical Technologies Inc., Tualatin, OR, USAProTherImmune, Lafayette, CA, USA
| | - G Engström
- Department of Microbiology and Tumor and Cell Biology, Karolinska Institutet & Swedish Institute for Infectious Disease ControlCenter for Molecular Medicine, Karolinska InstitutetVecura, Cell & Gene Therapy Center, Karolinska SjukhusetCancer Center Karolinska, Karolinska Sjukhuset, Stockholm, SwedenBioject Medical Technologies Inc., Tualatin, OR, USAProTherImmune, Lafayette, CA, USA
| | - A Boberg
- Department of Microbiology and Tumor and Cell Biology, Karolinska Institutet & Swedish Institute for Infectious Disease ControlCenter for Molecular Medicine, Karolinska InstitutetVecura, Cell & Gene Therapy Center, Karolinska SjukhusetCancer Center Karolinska, Karolinska Sjukhuset, Stockholm, SwedenBioject Medical Technologies Inc., Tualatin, OR, USAProTherImmune, Lafayette, CA, USA
| | - L Gudmundsdotter
- Department of Microbiology and Tumor and Cell Biology, Karolinska Institutet & Swedish Institute for Infectious Disease ControlCenter for Molecular Medicine, Karolinska InstitutetVecura, Cell & Gene Therapy Center, Karolinska SjukhusetCancer Center Karolinska, Karolinska Sjukhuset, Stockholm, SwedenBioject Medical Technologies Inc., Tualatin, OR, USAProTherImmune, Lafayette, CA, USA
| | - P Blomberg
- Department of Microbiology and Tumor and Cell Biology, Karolinska Institutet & Swedish Institute for Infectious Disease ControlCenter for Molecular Medicine, Karolinska InstitutetVecura, Cell & Gene Therapy Center, Karolinska SjukhusetCancer Center Karolinska, Karolinska Sjukhuset, Stockholm, SwedenBioject Medical Technologies Inc., Tualatin, OR, USAProTherImmune, Lafayette, CA, USA
| | - H Mellstedt
- Department of Microbiology and Tumor and Cell Biology, Karolinska Institutet & Swedish Institute for Infectious Disease ControlCenter for Molecular Medicine, Karolinska InstitutetVecura, Cell & Gene Therapy Center, Karolinska SjukhusetCancer Center Karolinska, Karolinska Sjukhuset, Stockholm, SwedenBioject Medical Technologies Inc., Tualatin, OR, USAProTherImmune, Lafayette, CA, USA
| | - R Stout
- Department of Microbiology and Tumor and Cell Biology, Karolinska Institutet & Swedish Institute for Infectious Disease ControlCenter for Molecular Medicine, Karolinska InstitutetVecura, Cell & Gene Therapy Center, Karolinska SjukhusetCancer Center Karolinska, Karolinska Sjukhuset, Stockholm, SwedenBioject Medical Technologies Inc., Tualatin, OR, USAProTherImmune, Lafayette, CA, USA
| | - M A Liu
- Department of Microbiology and Tumor and Cell Biology, Karolinska Institutet & Swedish Institute for Infectious Disease ControlCenter for Molecular Medicine, Karolinska InstitutetVecura, Cell & Gene Therapy Center, Karolinska SjukhusetCancer Center Karolinska, Karolinska Sjukhuset, Stockholm, SwedenBioject Medical Technologies Inc., Tualatin, OR, USAProTherImmune, Lafayette, CA, USA
| | - B Wahren
- Department of Microbiology and Tumor and Cell Biology, Karolinska Institutet & Swedish Institute for Infectious Disease ControlCenter for Molecular Medicine, Karolinska InstitutetVecura, Cell & Gene Therapy Center, Karolinska SjukhusetCancer Center Karolinska, Karolinska Sjukhuset, Stockholm, SwedenBioject Medical Technologies Inc., Tualatin, OR, USAProTherImmune, Lafayette, CA, USA
| |
Collapse
|
15
|
Su C, Peng L, Sham J, Wang X, Zhang Q, Chua D, Liu C, Cui Z, Xue H, Wu H, Yang Q, Zhang B, Liu X, Wu M, Qian Q. Immune gene-viral therapy with triplex efficacy mediated by oncolytic adenovirus carrying an interferon-gamma gene yields efficient antitumor activity in immunodeficient and immunocompetent mice. Mol Ther 2006; 13:918-27. [PMID: 16497559 DOI: 10.1016/j.ymthe.2005.12.011] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2005] [Revised: 12/11/2005] [Accepted: 12/28/2005] [Indexed: 11/25/2022] Open
Abstract
Among numerous gene therapeutic strategies for cancer treatment, gene transfer by conditionally replicative adenovirus (CRAd) of interferon-gamma (IFN-gamma) may be useful because of the possibility that it will yield IFN-gamma-mediated antiangiogenesis, immune responses, and CRAd-mediated oncolysis. In this study, we constructed a human TERT promoter-mediated oncolytic adenovirus targeting telomerase-positive cancers and armed with a mouse or human IFN-gamma gene to generate novel immune gene-viral therapeutic systems, CNHK300-mIFN-gamma and CNHK300-hIFN-gamma, respectively. The systems can specifically target, replicate in, and lyse cancer cells, while sparing normal cells. The advantage of these systems is that the number of transgene copies and their expression increase markedly via viral replication within infected cancer cells, and replicated viral progeny can then infect additional cancer cells within the tumor mass. CNHK300-mIFN-gamma induced regression of xenografts in liver cancer models in both immunodeficient and immunocompetent mice by triplex mechanisms including selective oncolysis, antiangiogenesis, and immune responses. We conclude that combining immune gene therapy and oncolytic virotherapy can enhance antitumor efficacy as a result of synergism between CRAd oncolysis and transgene composite antitumor responses.
Collapse
Affiliation(s)
- Changqing Su
- Laboratory of Viral and Gene Therapy, Eastern Hepatobiliary Surgical Hospital, Second Military Medical University, Shanghai 200438, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Blohm U, Potthoff D, van der Kogel AJ, Pircher H. Solid tumors “melt” from the inside after successful CD8 T cell attack. Eur J Immunol 2006; 36:468-77. [PMID: 16385625 DOI: 10.1002/eji.200526175] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Adoptive transfer of tumor-specific T cells represents a promising approach for cancer immunotherapy. Here, we visualized the anti-tumor response of CD8 T cells from P14 TCR-transgenic mice specific for the model antigen GP33 by immunohistology. P14 T cells, adoptively transferred into tumor-bearing hosts, induced regression of established 3LL-A9(GP33) and MCA102(GP33) tumors that express GP33 as a tumor-associated model antigen. Strikingly, the visible effects of P14 T cell attack, such as the destruction of the tumor vasculature and accumulation of granulocytes, were predominantly detected inside the tumor mass. In regressing tumors, P14 T cells were found in the intact rim zone but not in central areas that were infiltrated with granulocytes and lacked CD31(+) endothelial cells. The rim of P14 T cell-treated tumors showed an increase in vascular density and decrease in hypoxia compared to untreated tumors. Hypoxic areas of tumors are known to exhibit decreased sensitivity to radiation therapy or chemotherapy. Thus, our data also imply that adoptive transfer of tumor-specific CD8 T cells might synergize with radiation therapy or chemotherapy in the elimination of solid tumors in vivo.
Collapse
MESH Headings
- Adoptive Transfer/methods
- Animals
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- Antigens, Viral/genetics
- Antigens, Viral/immunology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/pathology
- CD8-Positive T-Lymphocytes/transplantation
- Endothelial Cells/immunology
- Endothelial Cells/pathology
- Granulocytes/immunology
- Granulocytes/pathology
- Hypoxia/immunology
- Hypoxia/pathology
- Lymphocytic choriomeningitis virus/genetics
- Lymphocytic choriomeningitis virus/immunology
- Mice
- Mice, Transgenic
- Neoplasm Transplantation
- Neoplasms, Experimental/blood supply
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/pathology
- Neoplasms, Experimental/therapy
- Neovascularization, Pathologic/immunology
- Neovascularization, Pathologic/pathology
- Neovascularization, Pathologic/therapy
- Neutrophil Infiltration/genetics
- Neutrophil Infiltration/immunology
- Platelet Endothelial Cell Adhesion Molecule-1/immunology
- Radiotherapy
Collapse
Affiliation(s)
- Ulrike Blohm
- Institute of Medical Microbiology and Hygiene, Department of Immunology, University of Freiburg, Germany
| | | | | | | |
Collapse
|
17
|
Abstract
It is often assumed that tumor rejection is mainly the result of cytotoxic T lymphocytes (CTLs) killing the tumor cells. However, recent studies have demonstrated that the rejection process is not as simple as this. In some models, tumors are rejected in the absence of lytic mechanisms (e.g. perforin or Fas ligand), and in others CTLs kill tumor stromal cells that cross-present antigen. T cells with lytic function but IFN-gamma deficiency rarely reject tumors. IFN-gamma and, in some models, other T-cell cytokines such as TNF-alpha, IL-4 or IL-10 contribute to tumor rejection by inhibition of tumor stroma formation. These cytokines inhibit tumor-induced angiogenesis, probably through different cellular targets.
Collapse
Affiliation(s)
- Thomas Blankenstein
- Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Strasse 10, 13092 Berlin, Germany.
| |
Collapse
|
18
|
Abstract
For decades it has been assumed that T cells reject tumors essentially by direct killing. However, solid tumors are composed of malignant cells and a variety of different nonmalignant cells, referred to as tumor stroma. Stromal cells, such as endothelial cells, fibroblasts and inflammatory cells, often support tumor growth. Here, we discuss new findings showing that the tumor stroma is an important target during T-cell-mediated tumor rejection. Cytotoxic molecules and cytokines produced by T cells inhibit or destroy the stromal 'infrastructure', thereby withdrawing essential resources and leading to tumor infarction and subsequent T-cell-mediated elimination of residual tumor cells. These findings are important for the development of more effective and specific immunotherapies for cancer.
Collapse
Affiliation(s)
- Thomas Kammertoens
- Institute of Immunology, Charité Campus Benjamin Franklin, 12200 Berlin, Germany
| | | | | |
Collapse
|
19
|
Wysocki PJ, Karczewska-Dzionk A, Mackiewicz-Wysocka M, Mackiewicz A. Human cancer gene therapy with cytokine gene-modified cells. Expert Opin Biol Ther 2005; 4:1595-607. [PMID: 15461571 DOI: 10.1517/14712598.4.10.1595] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Cytokines can impede tumour growth and activate innate and adaptive immune responses, leading to elimination of cancer cells. For many years, it was believed that systemic administration of recombinant cytokines might become a standard treatment of different cancer types. However, due to a high toxicity of therapeutic doses and a low efficacy, even in combination with chemotherapy, this strategy is generally not accepted. On the other hand, cancer gene therapy approaches utilising cells modified with cytokine genes seem to represent a novel promising approach. For the last decade, numerous Phase I and II clinical trials evaluating different therapies based on cytokine gene-modified cells have been carried out. In the early studies, several strategies have been shown to improve clinical outcomes and induce strong antitumour immune responses. Recently, a few prospective, randomised, Phase III clinical trials have been initiated in order to finally determine the efficacy of particular cancer immunogene therapy strategies. This article reviews the present status and perspectives of clinical trials of cancer immunotherapies utilising cytokine gene-modified cells.
Collapse
Affiliation(s)
- Piotr J Wysocki
- University of Medical Sciences at GreatPoland Cancer Center, Department of Cancer Immunology, UL. Garbary 15, 61-866 Poznan, Poland
| | | | | | | |
Collapse
|