1
|
Lai YG, Liao HT, Chen YH, Huang SW, Liou YH, Wu ZQ, Liao NS. cGAS and STING in Host Myeloid Cells Are Essential for Effective Cyclophosphamide Treatment of Advanced Breast Cancer. Cancers (Basel) 2025; 17:1130. [PMID: 40227734 PMCID: PMC11987962 DOI: 10.3390/cancers17071130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/21/2025] [Accepted: 03/25/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND/OBJECTIVES Cyclophosphamide (CTX) treatment in vivo kills proliferating tumor cells by DNA crosslinking; however, the suppression of tumor growth by CTX in several murine models requires CD8+ T cells. Given that CTX induces DNA damage and type I interferon (IFN-I), we investigated the role of host cGAS and STING in the anti-tumor effect of CTX in vivo. METHODS A metastasized EO771 breast cancer model with chromosomal instability and bone marrow (BM) chimera approach were used in this study. RESULTS We found that CTX therapy induces long-term survival of the mice, with this outcome being dependent on CD8+ T cells and cGAS/STING of BM-derived cells. Furthermore, the STING of type 1 conventional dendritic cells (cDC1s) and LysM+ cells and the IFN-I response of non-cDC1 myeloid cells are essential for CTX efficacy. We also found that the cGAS and STING of BM-derived cells positively modulate intratumoral exhausted and stem-cell-like CD8+ T cell populations under CTX treatment, with the latter only being affected by cGAS. CONCLUSIONS Our study demonstrates that the CD8+-T-cell-dependent anti-tumor mechanisms of CTX critically involve the cGAS-STING-IFN-I axis, IFN-I response, and STING-independent cGAS function in host myeloid cells. These findings suggest the deployment of CTX in treating advanced solid tumor to bypass the often-failed IFN-I production by tumor cells due to the chronic activation of intrinsic cGAS-STING caused by chromosomal instability.
Collapse
Affiliation(s)
- Yein-Gei Lai
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan; (Y.-G.L.); (H.-T.L.); (Y.-H.C.); (S.-W.H.); (Y.-H.L.); (Z.-Q.W.)
| | - Hao-Ting Liao
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan; (Y.-G.L.); (H.-T.L.); (Y.-H.C.); (S.-W.H.); (Y.-H.L.); (Z.-Q.W.)
- Department of Life Sciences, National Central University, Taoyuan 320, Taiwan
| | - Yung-Hsiang Chen
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan; (Y.-G.L.); (H.-T.L.); (Y.-H.C.); (S.-W.H.); (Y.-H.L.); (Z.-Q.W.)
| | - Shih-Wen Huang
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan; (Y.-G.L.); (H.-T.L.); (Y.-H.C.); (S.-W.H.); (Y.-H.L.); (Z.-Q.W.)
| | - Yae-Huei Liou
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan; (Y.-G.L.); (H.-T.L.); (Y.-H.C.); (S.-W.H.); (Y.-H.L.); (Z.-Q.W.)
| | - Zhen-Qi Wu
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan; (Y.-G.L.); (H.-T.L.); (Y.-H.C.); (S.-W.H.); (Y.-H.L.); (Z.-Q.W.)
| | - Nan-Shih Liao
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan; (Y.-G.L.); (H.-T.L.); (Y.-H.C.); (S.-W.H.); (Y.-H.L.); (Z.-Q.W.)
| |
Collapse
|
2
|
Delahousse J, Molina L, Paci A. "Cyclophosphamide and analogues; a matter of dose and schedule for dual anticancer activities". Cancer Lett 2024; 598:217119. [PMID: 39002693 DOI: 10.1016/j.canlet.2024.217119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/15/2024]
Abstract
Cyclophosphamide and ifosfamide are major alkylating agents but their therapeutics uses are limiting by the toxicity due to several toxicities. Indeed conventional chemotherapies are generally used with the maximum tolerated dose. In contrast, metronomic schedule aims to get a minimum dose for efficacy with a good safety. Depending on the dose, their mechanisms of action are different and offer a dual activity: at high dose, cyclophosphamide is mainly used in graft conditioning for its immunosuppressive properties, while at metronomic dose it is used as an immunoactive agent. Currently, at metronomic dose, cyclophosphamide is studied in clinic against various types of cancer, alone or in combination with others anticancer drugs (anti-angiogenic, immune-modulating agents, immune checkpoints blockers, vaccines, radiotherapy, others conventional anticancer agents), as a nth-line or first-line treatment. More than three quarters of clinical studies show promising results, mostly in breast, ovarian and prostate cancers. Taking advantage of the immune system, use dual antitumor action's chemotherapy is clearly a therapeutic strategy that deserves to be confirmed in order to improve the efficacy/toxicity balance of anticancer treatments, and to use CPM or analogues as a standard of care.
Collapse
Affiliation(s)
| | - Leonardo Molina
- Gustave Roussy, Department of Pharmacology, Villejuif, France
| | - Angelo Paci
- Gustave Roussy, Department of Pharmacology, Villejuif, France; Pharmacokinetics Department, Faculté de Pharmacie, Université Paris-Saclay, Gif-sur-Yvette, France
| |
Collapse
|
3
|
Tilsed CM, Morales MLO, Zemek RM, Gordon BA, Piggott MJ, Nowak AK, Fisher SA, Lake RA, Lesterhuis WJ. Tretinoin improves the anti-cancer response to cyclophosphamide, in a model-selective manner. BMC Cancer 2024; 24:203. [PMID: 38350880 PMCID: PMC10865642 DOI: 10.1186/s12885-024-11915-5] [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: 08/18/2023] [Accepted: 01/23/2024] [Indexed: 02/15/2024] Open
Abstract
BACKGROUND Chemotherapy is included in treatment regimens for many solid cancers, but when administered as a single agent it is rarely curative. The addition of immune checkpoint therapy to standard chemotherapy regimens has improved response rates and increased survival in some cancers. However, most patients do not respond to treatment and immune checkpoint therapy can cause severe side effects. Therefore, there is a need for alternative immunomodulatory drugs that enhance chemotherapy. METHODS We used gene expression data from cyclophosphamide (CY) responders and non-responders to identify existing clinically approved drugs that could phenocopy a chemosensitive tumor microenvironment (TME), and tested combination treatments in multiple murine cancer models. RESULTS The vitamin A derivative tretinoin was the top predicted upstream regulator of response to CY. Tretinoin pre-treatment induced an inflammatory, interferon-associated TME, with increased infiltration of CD8 + T cells, sensitizing the tumor to subsequent chemotherapy. However, while combination treatment significantly improved survival and cure rate in a CD4+ and CD8+ T cell dependent manner in AB1-HA murine mesothelioma, this effect was model-selective, and could not be replicated using other cell lines. CONCLUSIONS Despite the promising data in one model, the inability to validate the efficacy of combination treatment in multiple cancer models deprioritizes tretinoin/cyclophosphamide combination therapy for clinical translation.
Collapse
Affiliation(s)
- Caitlin M Tilsed
- National Centre for Asbestos Related Diseases, 6009, Nedlands, WA, Australia
- School of Biomedical Sciences, University of Western Australia, 6009, Crawley, WA, Australia
- Institute for Respiratory Health, 6101, Perth, WA, Australia
| | | | - Rachael M Zemek
- Telethon Kids Institute, University of Western Australia, 6872, West Perth, WA, Australia
| | - Brianna A Gordon
- School of Molecular Sciences, University of Western Australia, 6009, Crawley, WA, Australia
| | - Matthew J Piggott
- School of Molecular Sciences, University of Western Australia, 6009, Crawley, WA, Australia
| | - Anna K Nowak
- National Centre for Asbestos Related Diseases, 6009, Nedlands, WA, Australia
- School of Biomedical Sciences, University of Western Australia, 6009, Crawley, WA, Australia
- Institute for Respiratory Health, 6101, Perth, WA, Australia
- Department of Medical Oncology, Sir Charles Gairdner Hospital, 6009, Nedlands, WA, Australia
| | - Scott A Fisher
- National Centre for Asbestos Related Diseases, 6009, Nedlands, WA, Australia
- School of Biomedical Sciences, University of Western Australia, 6009, Crawley, WA, Australia
- Institute for Respiratory Health, 6101, Perth, WA, Australia
| | - Richard A Lake
- National Centre for Asbestos Related Diseases, 6009, Nedlands, WA, Australia
- School of Biomedical Sciences, University of Western Australia, 6009, Crawley, WA, Australia
- Institute for Respiratory Health, 6101, Perth, WA, Australia
| | - W Joost Lesterhuis
- National Centre for Asbestos Related Diseases, 6009, Nedlands, WA, Australia.
- School of Biomedical Sciences, University of Western Australia, 6009, Crawley, WA, Australia.
- Institute for Respiratory Health, 6101, Perth, WA, Australia.
- Telethon Kids Institute, University of Western Australia, 6872, West Perth, WA, Australia.
| |
Collapse
|
4
|
Zareian N, Eremin O, Pandha H, Baird R, Kwatra V, Funingana G, Verma C, Choy D, Hargreaves S, Moghimi P, Shepherd A, Lobo DN, Eremin J, Farzaneh F, Kordasti S, Spicer J. A phase 1 trial of human telomerase reverse transcriptase (hTERT) vaccination combined with therapeutic strategies to control immune-suppressor mechanisms. Exp Biol Med (Maywood) 2024; 249:10021. [PMID: 38463391 PMCID: PMC10911124 DOI: 10.3389/ebm.2024.10021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 01/04/2024] [Indexed: 03/12/2024] Open
Abstract
The presence of inhibitory immune cells and difficulty in generating activated effector T cells remain obstacles to development of effective cancer vaccines. We designed a vaccine regimen combining human telomerase reverse transcriptase (hTERT) peptides with concomitant therapies targeting regulatory T cells (Tregs) and cyclooxygenase-2 (COX2)-mediated immunosuppression. This Phase 1 trial combined an hTERT-derived 7-peptide library, selected to ensure presentation by both HLA class-I and class-II in 90% of patients, with oral low-dose cyclophosphamide (to modulate Tregs) and the COX2 inhibitor celecoxib. Adjuvants were Montanide and topical TLR-7 agonist, to optimise antigen presentation. The primary objective was determination of the safety and tolerability of this combination therapy, with anti-cancer activity, immune response and detection of antigen-specific T cells as additional endpoints. Twenty-nine patients with advanced solid tumours were treated. All were multiply-pretreated, and the majority had either colorectal or prostate cancer. The most common adverse events were injection-site reactions, fatigue and nausea. Median progression-free survival was 9 weeks, with no complete or partial responses, but 24% remained progression-free for ≥6 months. Immunophenotyping showed post-vaccination expansion of CD4+ and CD8+ T cells with effector phenotypes. The in vitro re-challenge of T cells with hTERT peptides, TCR sequencing, and TCR similarity index analysis demonstrated the expansion following vaccination of oligoclonal T cells with specificity for hTERT. However, a population of exhausted PD-1+ cytotoxic T cells was also expanded in vaccinated patients. This vaccine combination regimen was safe and associated with antigen-specific immunological responses. Clinical activity could be improved in future by combination with anti-PD1 checkpoint inhibition to address the emergence of an exhausted T cell population.
Collapse
Affiliation(s)
- Nahid Zareian
- School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| | - Oleg Eremin
- Nottingham Digestive Diseases Centre, NIHR Nottingham Biomedical Research Centre, Queen's Medical Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Nottingham, United Kingdom
| | - Hardev Pandha
- Department of Microbiology and Cellular Sciences, University of Surrey, Guildford, United Kingdom
| | - Richard Baird
- Cancer Research UK Cambridge Centre, Cambridge, United Kingdom
| | - Vineet Kwatra
- School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| | | | - Chandan Verma
- Nottingham Digestive Diseases Centre, NIHR Nottingham Biomedical Research Centre, Queen's Medical Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Nottingham, United Kingdom
| | - Desmond Choy
- School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| | - Steven Hargreaves
- Research Department of Pathology, UCL Cancer Institute, Faculty of Medical Sciences, University College London (UCL), London, United Kingdom
| | - Pejvak Moghimi
- The Institute of Structural and Molecular Biology (ISMB), Birkbeck, University of London, London, United Kingdom
| | - Adrian Shepherd
- The Institute of Structural and Molecular Biology (ISMB), Birkbeck, University of London, London, United Kingdom
| | - Dileep N Lobo
- Nottingham Digestive Diseases Centre, NIHR Nottingham Biomedical Research Centre, Queen's Medical Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Nottingham, United Kingdom
- MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, Queen's Medical Centre, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Jennifer Eremin
- Nottingham Digestive Diseases Centre, NIHR Nottingham Biomedical Research Centre, Queen's Medical Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Nottingham, United Kingdom
| | - Farzin Farzaneh
- School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| | - Shahram Kordasti
- School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| | - James Spicer
- School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| |
Collapse
|
5
|
CD4 + T cells drive an inflammatory, TNF-α/IFN-rich tumor microenvironment responsive to chemotherapy. Cell Rep 2022; 41:111874. [PMID: 36577370 DOI: 10.1016/j.celrep.2022.111874] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 08/08/2022] [Accepted: 12/02/2022] [Indexed: 12/28/2022] Open
Abstract
While chemotherapy remains the first-line treatment for many cancers, it is still unclear what distinguishes responders from non-responders. Here, we characterize the chemotherapy-responsive tumor microenvironment in mice, using RNA sequencing on tumors before and after cyclophosphamide, and compare the gene expression profiles of responders with progressors. Responsive tumors have an inflammatory and highly immune infiltrated pre-treatment tumor microenvironment characterized by the enrichment of pathways associated with CD4+ T cells, interferons (IFNs), and tumor necrosis factor alpha (TNF-α). The same gene expression profile is associated with response to cyclophosphamide-based chemotherapy in patients with breast cancer. Finally, we demonstrate that tumors can be sensitized to cyclophosphamide and 5-FU chemotherapy by pre-treatment with recombinant TNF-α, IFNγ, and poly(I:C). Thus, a CD4+ T cell-inflamed pre-treatment tumor microenvironment is necessary for response to chemotherapy, and this state can be therapeutically attained by targeted immunotherapy.
Collapse
|
6
|
Tilsed CM, Fisher SA, Nowak AK, Lake RA, Lesterhuis WJ. Cancer chemotherapy: insights into cellular and tumor microenvironmental mechanisms of action. Front Oncol 2022; 12:960317. [PMID: 35965519 PMCID: PMC9372369 DOI: 10.3389/fonc.2022.960317] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 07/01/2022] [Indexed: 12/12/2022] Open
Abstract
Chemotherapy has historically been the mainstay of cancer treatment, but our understanding of what drives a successful therapeutic response remains limited. The diverse response of cancer patients to chemotherapy has been attributed principally to differences in the proliferation rate of the tumor cells, but there is actually very little experimental data supporting this hypothesis. Instead, other mechanisms at the cellular level and the composition of the tumor microenvironment appear to drive chemotherapy sensitivity. In particular, the immune system is a critical determinant of chemotherapy response with the depletion or knock-out of key immune cell populations or immunological mediators completely abrogating the benefits of chemotherapy in pre-clinical models. In this perspective, we review the literature regarding the known mechanisms of action of cytotoxic chemotherapy agents and the determinants of response to chemotherapy from the level of individual cells to the composition of the tumor microenvironment. We then summarize current work toward the development of dynamic biomarkers for response and propose a model for a chemotherapy sensitive tumor microenvironment.
Collapse
Affiliation(s)
- Caitlin M. Tilsed
- National Centre for Asbestos Related Diseases, Institute for Respiratory Health, Nedlands, WA, Australia
- School of Biomedical Sciences, University of Western Australia, Crawley, WA, Australia
| | - Scott A. Fisher
- National Centre for Asbestos Related Diseases, Institute for Respiratory Health, Nedlands, WA, Australia
- School of Biomedical Sciences, University of Western Australia, Crawley, WA, Australia
| | - Anna K. Nowak
- National Centre for Asbestos Related Diseases, Institute for Respiratory Health, Nedlands, WA, Australia
- Medical School, University of Western Australia, Crawley, WA, Australia
- Department of Medical Oncology, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
| | - Richard A. Lake
- National Centre for Asbestos Related Diseases, Institute for Respiratory Health, Nedlands, WA, Australia
- School of Biomedical Sciences, University of Western Australia, Crawley, WA, Australia
| | - W. Joost Lesterhuis
- National Centre for Asbestos Related Diseases, Institute for Respiratory Health, Nedlands, WA, Australia
- School of Biomedical Sciences, University of Western Australia, Crawley, WA, Australia
- Telethon Kids Institute, University of Western Australia, West Perth, WA, Australia
- *Correspondence: W. Joost Lesterhuis,
| |
Collapse
|
7
|
Abstract
The nitrogen mustards are powerful cytotoxic and lymphoablative agents and have been used for more than 60 years. They are employed in the treatment of cancers, sarcomas, and hematologic malignancies. Cyclophosphamide, the most versatile of the nitrogen mustards, also has a place in stem cell transplantation and the therapy of autoimmune diseases. Adverse effects caused by the nitrogen mustards on the central nervous system, kidney, heart, bladder, and gonads remain important issues. Advances in analytical techniques have facilitated the investigation of the pharmacokinetics of the nitrogen mustards, especially the oxazaphosphorines, which are prodrugs requiring metabolic activation. Enzymes involved in the metabolism of cyclophosphamide and ifosfamide are very polymorphic, but a greater understanding of the pharmacogenomic influences on their activity has not yet translated into a personalized medicine approach. In addition to damaging DNA, the nitrogen mustards can act through other mechanisms, such as antiangiogenesis and immunomodulation. The immunomodulatory properties of cyclophosphamide are an area of current exploration. In particular, cyclophosphamide decreases the number and activity of regulatory T cells, and the interaction between cyclophosphamide and the intestinal microbiome is now recognized as an important factor. New derivatives of the nitrogen mustards continue to be assessed. Oxazaphosphorine analogs have been synthesized in attempts to both improve efficacy and reduce toxicity, with varying degrees of success. Combinations of the nitrogen mustards with monoclonal antibodies and small-molecule targeted agents are being evaluated. SIGNIFICANCE STATEMENT: The nitrogen mustards are important, well-established therapeutic agents that are used to treat a variety of diseases. Their role is continuing to evolve.
Collapse
Affiliation(s)
- Martin S Highley
- Plymouth Oncology Centre, Derriford Hospital, and Peninsula Medical School, University of Plymouth, Plymouth, United Kingdom (M.S.H.); Department of Animal Physiology and Neurobiology (B.L.) and Laboratory for Experimental Oncology (E.A.D.B.), University of Leuven, Leuven, Belgium; Oncology Department, University Hospital Antwerp, Edegem, Belgium (H.P.); and London Oncology Clinic, London, United Kingdom (P.G.H.)
| | - Bart Landuyt
- Plymouth Oncology Centre, Derriford Hospital, and Peninsula Medical School, University of Plymouth, Plymouth, United Kingdom (M.S.H.); Department of Animal Physiology and Neurobiology (B.L.) and Laboratory for Experimental Oncology (E.A.D.B.), University of Leuven, Leuven, Belgium; Oncology Department, University Hospital Antwerp, Edegem, Belgium (H.P.); and London Oncology Clinic, London, United Kingdom (P.G.H.)
| | - Hans Prenen
- Plymouth Oncology Centre, Derriford Hospital, and Peninsula Medical School, University of Plymouth, Plymouth, United Kingdom (M.S.H.); Department of Animal Physiology and Neurobiology (B.L.) and Laboratory for Experimental Oncology (E.A.D.B.), University of Leuven, Leuven, Belgium; Oncology Department, University Hospital Antwerp, Edegem, Belgium (H.P.); and London Oncology Clinic, London, United Kingdom (P.G.H.)
| | - Peter G Harper
- Plymouth Oncology Centre, Derriford Hospital, and Peninsula Medical School, University of Plymouth, Plymouth, United Kingdom (M.S.H.); Department of Animal Physiology and Neurobiology (B.L.) and Laboratory for Experimental Oncology (E.A.D.B.), University of Leuven, Leuven, Belgium; Oncology Department, University Hospital Antwerp, Edegem, Belgium (H.P.); and London Oncology Clinic, London, United Kingdom (P.G.H.)
| | - Ernst A De Bruijn
- Plymouth Oncology Centre, Derriford Hospital, and Peninsula Medical School, University of Plymouth, Plymouth, United Kingdom (M.S.H.); Department of Animal Physiology and Neurobiology (B.L.) and Laboratory for Experimental Oncology (E.A.D.B.), University of Leuven, Leuven, Belgium; Oncology Department, University Hospital Antwerp, Edegem, Belgium (H.P.); and London Oncology Clinic, London, United Kingdom (P.G.H.)
| |
Collapse
|
8
|
Proskurina AS, Ruzanova VS, Ostanin AA, Chernykh ER, Bogachev SS. Theoretical premises of a "three in one" therapeutic approach to treat immunogenic and nonimmunogenic cancers: a narrative review. Transl Cancer Res 2022; 10:4958-4972. [PMID: 35116346 PMCID: PMC8797664 DOI: 10.21037/tcr-21-919] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 10/14/2021] [Indexed: 12/12/2022]
Abstract
Objective We describe experimental and theoretical premises of a powerful cancer therapy based on the combination of three approaches. These include (I) in situ vaccination (intratumoral injections of CpG oligonucleotides and anti-OX40 antibody); (II) chronometric or metronomic low-dose cyclophosphamide (CMLD CP)-based chemotherapy; (III) cancer stem cell-eradicating therapy referred to as Karanahan (from the Sanskrit kāraṇa [“source”] + han [“to kill”]). Background In murine models, the first two approaches are particularly potent in targeting immunogenic tumors for destruction. In situ vaccination activates a fully fledged anticancer immune response via an intricate network of ligand–receptor–cytokine interactions. CMLD CP-based chemotherapy primarily targets the suppressive tumor microenvironment and activates tumor-infiltrating effectors. In contrast, Karanahan technology, being aimed at replicative machinery of tumor cells (both stem-like and committed), does not depend on tumor immunogenicity. With this technology, mice engrafted with ascites and/or solid tumors can be successfully cured. There is a significant degree of mechanistic and therapeutic overlap between these three approaches. For instance, the similarities shared between in situ vaccination and Karanahan technology include the therapeutic procedure, the cell target [antigen-presenting cells (APC) and dendritic cells (DC)], and the use of DNA-based preparations (CpG and DNAmix). Features shared between CMLD CP-based chemotherapy and Karanahan technology are the timing and the dose of the cytostatic drug administration, which lead to tumor regression. Methods The following keywords were used to search PubMed for the latest research reporting successful eradication of transplantable cancers in animal models that relied on approaches distinct from those used in the Karanahan technology: eradication of malignancy, cure cancer, complete tumor regression, permanently eradicating advanced mouse tumor, metronomic chemotherapy, in situ vaccination, immunotherapy, and others. Conclusion We hypothesize, therefore, that very potent anticancer activity can be achieved once these three therapeutic modalities are combined into a single approach. This multimodal approach is theoretically curative for any type of cancer that depends on the presence of tumor-inducing cancer stem cells, provided that the active therapeutic components are efficiently delivered into the tumor and the specific biological features of a given patient’s tumor are properly addressed. We expect this multimodal approach to be primarily applicable to late-stage or terminal cancer patients who have exhausted all treatment options as well as patients with inoperable tumors.
Collapse
Affiliation(s)
- Anastasia S Proskurina
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Vera S Ruzanova
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia.,Novosibirsk State University, Novosibirsk, Russia
| | - Alexandr A Ostanin
- Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - Elena R Chernykh
- Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - Sergey S Bogachev
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
9
|
Moysés CRS, Alvares-Saraiva AM, Perez EC, Spadacci-Morena DD, Vidôto da Costa LF, Xavier JG, Lallo MA. Mice with genetic and induced B-cell deficiency as a model for disseminated encephalitozoonosis. Comp Immunol Microbiol Infect Dis 2022; 81:101742. [PMID: 35074660 DOI: 10.1016/j.cimid.2021.101742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 11/19/2022]
Abstract
Encephalitozoon cuniculi, an intracellular pathogen, lives in a balanced relationship with immunocompetent individuals based on the activity of T lymphocytes. We previously highlighted the greater susceptibility of B-1 cell-deficient mice (XID mice) to encephalitozoonosis. This study aimed to develop a model of disseminated and severe encephalitozoonosis in mice with combined immunodeficiency to elucidate the role of B cells. To address this objective, cyclophosphamide (Cy)-treated BALB/c and XID mice were inoculated with E. cuniculi, followed by the evaluation of the immune response and histopathological lesions. Immunosuppressed BALB/c mice manifested no clinical signs with an increase in the populations of T lymphocytes and macrophages in the spleen. Immunosuppressed and infected XID mice revealed elevated T cells, macrophages populations, and pro-inflammatory cytokines levels (IFN-γ, TNF-α, and IL-6) with the presence of abdominal effusion and lesions in multiple organs. These clinical characteristics are associated with extensive and severe encephalitozoonosis. The symptoms and lesion size were reduced, whereas B-2 and CD4+ T cells populations were increased in the spleen by transferring B-2 cells adoptive to XID mice. Moreover, B-1 cells adoptive transfer upregulated the peritoneal populations of B-2 cells and macrophages but not T lymphocytes and decreased the symptoms. Herein, we speculated the consistency in the development of severe and disseminated encephalitozoonosis in mice with genetic deficiency of Bruton's tyrosine kinase (Btk) associated with Cy immunosuppression develop with that of the models with T cell deficiency. Taken together, these data emphasized the crucial role of B cells in the protective immune response against encephalitozoonosis.
Collapse
Affiliation(s)
- Carla Renata Serantoni Moysés
- Programa de Pós-Graduação em Patologia Ambiental e Experimental, Universidade Paulista - Unip, Rua José Maria Whitaker 290, CEP 05622-001 São Paulo, SP, Brazil
| | - Anuska Marcelino Alvares-Saraiva
- Programa de Pós-Graduação em Patologia Ambiental e Experimental, Universidade Paulista - Unip, Rua José Maria Whitaker 290, CEP 05622-001 São Paulo, SP, Brazil; Laboratório de Fisiopatologia, Instituto Butantan, Av. Vital Brasil 1500, CEP 05503-900 São Paulo, SP, Brazil; Programa de Pós-Graduação Interdisciplinar em Ciências da Saúde, Universidade Cruzeiro do Sul, Rua Galvão Bueno, 868, CEP 01506-000 São Paulo, SP, Brazil
| | - Elizabeth Cristina Perez
- Programa de Pós-Graduação em Patologia Ambiental e Experimental, Universidade Paulista - Unip, Rua José Maria Whitaker 290, CEP 05622-001 São Paulo, SP, Brazil
| | | | - Lidiana Flora Vidôto da Costa
- Programa de Pós-Graduação em Patologia Ambiental e Experimental, Universidade Paulista - Unip, Rua José Maria Whitaker 290, CEP 05622-001 São Paulo, SP, Brazil
| | - José Guilherme Xavier
- Programa de Pós-Graduação em Patologia Ambiental e Experimental, Universidade Paulista - Unip, Rua José Maria Whitaker 290, CEP 05622-001 São Paulo, SP, Brazil
| | - Maria Anete Lallo
- Programa de Pós-Graduação em Patologia Ambiental e Experimental, Universidade Paulista - Unip, Rua José Maria Whitaker 290, CEP 05622-001 São Paulo, SP, Brazil.
| |
Collapse
|
10
|
Dolgova EV, Andrushkevich OM, Kisaretova PE, Proskurina AS, Ritter GS, Dubatolova TD, Romanenko MV, Taranov OS, Efremov YR, Zavyalov EL, Romaschenko AV, Mishinov SV, Kirikovich SS, Levites EV, Potter EA, Ostanin AA, Chernykh ER, Roshchin SY, Bervitskiy AV, Moysak GI, Rzaev JA, Bogachev SS. Efficacy of the new therapeutic approach in curing malignant neoplasms on the model of human glioblastoma. Cancer Biol Med 2021; 18:j.issn.2095-3941.2020.0511. [PMID: 34259424 PMCID: PMC8330538 DOI: 10.20892/j.issn.2095-3941.2020.0511] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 02/08/2021] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE Glioma is a highly invasive tumor, frequently disposed in essential areas of the brain, which makes its surgical excision extremely difficult; meanwhile adjuvant therapy remains quite ineffective. METHODS In the current report, a new therapeutic approach in curing malignant neoplasms has been performed on the U87 human glioblastoma model. This approach, termed "Karanahan", is aimed at the eradication of cancer stem cells (CSCs), which were recently shown to be capable of internalizing fragments of extracellular double-stranded DNA. After being internalized, these fragments interfere in the process of repairing interstrand cross-links caused by exposure to appropriate cytostatics, and such an interference results either in elimination of CSCs or in the loss of their tumorigenic potency. Implementation of the approach requires a scheduled administration of cytostatic and complex composite double-stranded DNA preparation. RESULTS U87 cells treated in vitro in accordance with the Karanahan approach completely lost their tumorigenicity and produced no grafts upon intracerebral transplantation into immunodeficient mice. In SCID mice with developed subcutaneous grafts, the treatment resulted in reliable slowing down of tumor growth rate (P < 0.05). In the experiment with intracerebral transplantation of U87 cells followed by surgical excision of the developed graft and subsequent therapeutic treatment, the Karanahan approach was shown to reliably slow down the tumor growth rate and increase the median survival of the mice twofold relative to the control. CONCLUSIONS The effectiveness of the Karanahan approach has been demonstrated both in vitro and in vivo in treating developed subcutaneous grafts as well as orthotopic grafts after surgical excision of the tumor.
Collapse
Affiliation(s)
| | - Oleg M. Andrushkevich
- A.I. Evdokimov Moscow State University of Medicine and Dentistry, Moscow 127473, Russia
| | | | | | - Genrikh S. Ritter
- Institute of Cytology and Genetics SB RAS, Novosibirsk 630090, Russia
| | | | | | - Oleg S. Taranov
- The State Research Center of Virology and Biotechnology “Vector”, Koltsovo, Novosibirsk 630559, Russia
| | - Yaroslav R. Efremov
- Institute of Cytology and Genetics SB RAS, Novosibirsk 630090, Russia
- Novosibirsk State University, Novosibirsk 630090, Russia
| | | | | | - Sergey V. Mishinov
- First Department of Neurosurgery, Ya. L. Tsivian Novosibirsk Research Institute of Traumatology and Orthopaedics, Novosibirsk 630091, Russia
| | | | | | | | - Alexandr A. Ostanin
- Institute of Fundamental and Clinical immunology, Novosibirsk 630099, Russia
| | - Elena R. Chernykh
- Institute of Fundamental and Clinical immunology, Novosibirsk 630099, Russia
| | | | | | - Galina I. Moysak
- Novosibirsk State University, Novosibirsk 630090, Russia
- Federal Center of Neurosurgery, Novosibirsk 630048, Russia
| | - Jamil A. Rzaev
- Novosibirsk State University, Novosibirsk 630090, Russia
- Federal Center of Neurosurgery, Novosibirsk 630048, Russia
| | | |
Collapse
|
11
|
Pre-conditioning modifies the TME to enhance solid tumor CAR T cell efficacy and endogenous protective immunity. Mol Ther 2021; 29:2335-2349. [PMID: 33647456 PMCID: PMC8261088 DOI: 10.1016/j.ymthe.2021.02.024] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 01/23/2021] [Accepted: 02/24/2021] [Indexed: 12/22/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy has led to impressive clinical responses in patients with hematological malignancies; however, its effectiveness in patients with solid tumors has been limited. While CAR T cells for the treatment of advanced prostate and pancreas cancer, including those targeting prostate stem cell antigen (PSCA), are being clinically evaluated and are anticipated to show bioactivity, their safety and the impact of the immunosuppressive tumor microenvironment (TME) have not been faithfully explored preclinically. Using a novel human PSCA knockin (hPSCA-KI) immunocompetent mouse model, we evaluated the safety and therapeutic efficacy of PSCA-CAR T cells. We demonstrated that cyclophosphamide (Cy) pre-conditioning significantly modified the immunosuppressive TME and was required to uncover the efficacy of PSCA-CAR T cells in metastatic prostate and pancreas cancer models, with no observed toxicities in normal tissues with endogenous expression of PSCA. This combination dampened the immunosuppressive TME, generated pro-inflammatory myeloid and T cell signatures in tumors, and enhanced the recruitment of antigen-presenting cells, as well as endogenous and adoptively transferred T cells, resulting in long-term anti-tumor immunity.
Collapse
|
12
|
Liu KX, Ioakeim-Ioannidou M, Susko MS, Rao AD, Yeap BY, Snijders AM, Ladra MM, Vogel J, Zaslowe-Dude C, Marcus KJ, Yock TI, Grassberger C, Braunstein SE, Haas-Kogan DA, Terezakis SA, MacDonald SM. A Multi-institutional Comparative Analysis of Proton and Photon Therapy-Induced Hematologic Toxicity in Patients With Medulloblastoma. Int J Radiat Oncol Biol Phys 2020; 109:726-735. [PMID: 33243479 DOI: 10.1016/j.ijrobp.2020.09.049] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 09/11/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023]
Abstract
PURPOSE This multi-institutional retrospective study sought to examine the hematologic effects of craniospinal irradiation (CSI) in pediatric patients with medulloblastoma using proton or photon therapy. METHODS AND MATERIALS Clinical and treatment characteristics were recorded for 97 pediatric patients with medulloblastoma who received CSI without concurrent chemotherapy or with concurrent single-agent vincristine from 2000 to 2017. Groups of 60 and 37 patients underwent treatment with proton-based and photon-based therapy, respectively. Overall survival was determined by Kaplan-Meier curves with log-rank test. Comparisons of blood counts at each timepoint were conducted using multiple t tests with Bonferroni corrections. Univariate and multivariate analyses of time to grade ≥3 hematologic toxicity were performed with Cox regression analyses. RESULTS Median age of patients receiving proton and photon CSI was 7.5 years (range, 3.5-22.7 years) and 9.9 years (range, 3.6-19.5 years), respectively. Most patients had a diagnosis of standard risk medulloblastoma, with 86.7% and 89.2% for the proton and photon cohorts, respectively. Median total dose to involved field or whole posterior fossa was 54.0 Gy/Gy relative biological effectiveness (RBE) and median CSI dose was 23.4 Gy/Gy(RBE) (range, 18-36 Gy/Gy[RBE]) for both cohorts. Counts were significantly higher in the proton cohort compared with the photon cohort in weeks 3 to 6 of radiation therapy (RT). Although white blood cell counts did not differ between the 2 cohorts, patients receiving proton RT had significantly higher lymphocyte counts throughout the RT course. Similar results were observed when excluding patients who received vertebral body sparing proton RT or limiting to those receiving 23.4 Gy. Only photon therapy was associated with decreased time to grade ≥3 hematologic toxicity on univariate and multivariable analyses. No difference in overall survival was observed, and lymphopenia did not predict survival. CONCLUSIONS Patients who receive CSI using proton therapy experience significantly decreased hematologic toxicity compared with those receiving photon therapy.
Collapse
Affiliation(s)
- Kevin X Liu
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Myrsini Ioakeim-Ioannidou
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Department of Radiation and Molecular Radiation Sciences, Sidney Kimmel Comprehensive Cancer Center Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Matthew S Susko
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California
| | - Avani D Rao
- Department of Radiation and Molecular Radiation Sciences, Sidney Kimmel Comprehensive Cancer Center Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Beow Y Yeap
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Antoine M Snijders
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, California
| | - Matthew M Ladra
- Department of Radiation and Molecular Radiation Sciences, Sidney Kimmel Comprehensive Cancer Center Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Jennifer Vogel
- Department of Radiation and Molecular Radiation Sciences, Sidney Kimmel Comprehensive Cancer Center Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Cierra Zaslowe-Dude
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Karen J Marcus
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Torunn I Yock
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Clemens Grassberger
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Steve E Braunstein
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California
| | - Daphne A Haas-Kogan
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Stephanie A Terezakis
- Department of Radiation and Molecular Radiation Sciences, Sidney Kimmel Comprehensive Cancer Center Johns Hopkins School of Medicine, Baltimore, Maryland; Department of Radiation Oncology, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Shannon M MacDonald
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
13
|
Bhatia K, Bhumika, Das A. Combinatorial drug therapy in cancer - New insights. Life Sci 2020; 258:118134. [PMID: 32717272 DOI: 10.1016/j.lfs.2020.118134] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/18/2020] [Accepted: 07/20/2020] [Indexed: 12/19/2022]
Abstract
Cancer can arise due to mutations in numerous pathways present in our body and thus has many alternatives for getting aggravated. Due to this attribute, it gets difficult to treat cancer patients with monotherapy alone and has a risk of not being eliminated to the full extent. This necessitates the introduction of combinatorial therapy as it employs cancer treatment using more than one method and shows a greater success rate. Combinatorial therapy involves a complementary combination of two different therapies like a combination of radio and immunotherapy or a combination of drugs that can target more than one pathway of cancer formation like combining CDK targeting drugs with Growth factors targeting drugs. In this review, we discuss the various aspects of cancer which include, its causes; four regulatory mechanisms namely: apoptosis, cyclin-dependent kinases, tumor suppressor genes, and growth factors; some of the pathways involved; treatment: monotherapy and combinatorial therapy and combinatorial drug formulation in chemotherapy. The present review gives a holistic account of the different mechanisms of therapies and also drug combinations that may serve to not only complement the monotherapy but can also surpass the resistance against monotherapy agents.
Collapse
Affiliation(s)
- Karanpreet Bhatia
- Department of Biotechnology, Delhi Technological University, Main Bawana Road, Delhi 110042, India
| | - Bhumika
- Department of Biotechnology, Delhi Technological University, Main Bawana Road, Delhi 110042, India
| | - Asmita Das
- Department of Biotechnology, Delhi Technological University, Main Bawana Road, Delhi 110042, India.
| |
Collapse
|
14
|
Klampatsa A, Leibowitz MS, Sun J, Liousia M, Arguiri E, Albelda SM. Analysis and Augmentation of the Immunologic Bystander Effects of CAR T Cell Therapy in a Syngeneic Mouse Cancer Model. MOLECULAR THERAPY-ONCOLYTICS 2020; 18:360-371. [PMID: 32802940 PMCID: PMC7417672 DOI: 10.1016/j.omto.2020.07.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 07/09/2020] [Indexed: 02/07/2023]
Abstract
The therapeutic efficacy of adoptive transfer of T cells transduced with chimeric antigen receptors (CARs) has been limited in the treatment of solid cancers, partly due to tumor antigen heterogeneity. Overcoming lack of universal tumor antigen expression would be achieved if CAR T cells could induce bystander effects. To study this process, we developed a system where CAR T cells targeting mesothelin could cure tumors containing 100% antigen-positive cells in immunocompetent mice. Using this model, we found that the CAR T cells were unable to cure tumors, even when only 10% of the tumor cells were mesothelin negative. A bystander effect was not induced by co-administration of anti-PD-1, anti-CTLA-4, or anti-TGF-β (transforming growth factor β) antibodies; agonistic CD40 antibodies; or an IDO (indoleamine 2,3-dioxygenase) inhibitor. However, pretreatment with a non-lymphodepleting dose of cyclophosphamide (CTX) prior to CAR T cells resulted in cures of tumors with up to 25% mesothelin-negative cells. The mechanism was dependent on endogenous CD8 T cells but not on basic leucine zipper transcription factor ATF-like 3 (BATF3)-dependent dendritic cells. These data suggest that CAR T cell therapy of solid tumors, in which the targeted antigen is not expressed by the vast majority of tumor cells, will not likely be successful unless combination strategies to enhance bystander effects are used.
Collapse
Affiliation(s)
- Astero Klampatsa
- Thoracic Oncology Immunotherapy Group, Division of Cancer Therapeutics, The Institute of Cancer Research, London SM2 5NG, UK
- Pulmonary, Allergy and Critical Care Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Corresponding author Astero Klampatsa, Thoracic Oncology Immunotherapy Group, Division of Cancer Therapeutics, The Institute of Cancer Research, 15 Cotswold Road, Sutton, London SM2 5NG, UK.
| | - Michael S. Leibowitz
- Pulmonary, Allergy and Critical Care Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Oncology, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Jing Sun
- Pulmonary, Allergy and Critical Care Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Maria Liousia
- Pulmonary, Allergy and Critical Care Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Evguenia Arguiri
- Pulmonary, Allergy and Critical Care Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Steven M. Albelda
- Pulmonary, Allergy and Critical Care Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
15
|
Repurposing Food and Drug Administration-Approved Drugs to Promote Antitumor Immunity. ACTA ACUST UNITED AC 2020; 25:88-99. [PMID: 30896530 DOI: 10.1097/ppo.0000000000000368] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
There has been a major resurgence of interest in immune-based approaches to treat cancer, based largely on the success of checkpoint inhibitors (anti-cytotoxic T-lymphocyte-associated antigen 4, anti-programmed cell death 1, and anti-programmed cell death ligand 1 antibodies) in several malignancies. However, not all tumors respond to checkpoint therapy, and there is clearly a need for additional approaches for enhancing tumor immunity. We summarize the critical elements necessary for mounting an efficacious T-cell response to a tumor. We cite drugs approved by the Food and Drug Administration for no-cancer indications that could be repurposed and used as part of an antitumor immune cocktail. We also list cancer drugs not initially intended to impact tumor immunity (soft repurposing) but that have been found to modulate the immune system. We highlight those drugs that might be used in combination with checkpoint inhibitors to increase response rates and survival of cancer patients. Our focus will be on drugs for which there are limited but existing human data. We cite supporting mechanistic mouse data as well. Repurposing drugs to modulate antitumor immunity is an opportunity to rapidly bring new, effective, and affordable treatments to cancer patients.
Collapse
|
16
|
Neophytou CM, Pierides C, Christodoulou MI, Costeas P, Kyriakou TC, Papageorgis P. The Role of Tumor-Associated Myeloid Cells in Modulating Cancer Therapy. Front Oncol 2020; 10:899. [PMID: 32656079 PMCID: PMC7325995 DOI: 10.3389/fonc.2020.00899] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 05/07/2020] [Indexed: 12/20/2022] Open
Abstract
Myeloid cells include various cellular subtypes that are distinguished into mononuclear and polymorphonuclear cells, derived from either common myeloid progenitor cells (CMPs) or myeloid stem cells. They play pivotal roles in innate immunity since, following invasion by pathogens, myeloid cells are recruited and initiate phagocytosis and secretion of inflammatory cytokines into local tissues. Moreover, mounting evidence suggests that myeloid cells may also regulate cancer development by infiltrating the tumor to directly interact with cancer cells or by affecting the tumor microenvironment. Importantly, mononuclear phagocytes, including macrophages and dendritic cells (DCs), can have either a positive or negative impact on the efficacy of chemotherapy, radiotherapy as well as targeted anti-cancer therapies. Tumor-associated macrophages (TAMs), profusely found in the tumor stroma, can promote resistance to chemotherapeutic drugs, such as Taxol and Paclitaxel, whereas the suppression of TAMs can lead to an improved radiotherapy outcome. On the contrary, the presence of TAMs may be beneficial for targeted therapies as they can facilitate the accumulation of large quantities of nanoparticles carrying therapeutic compounds. Tumor infiltrating DCs, however, are generally thought to enhance cytotoxic therapies, including those using anthracyclines. This review focuses on the role of tumor-infiltrating and stroma myeloid cells in modulating tumor responses to various treatments. We herein report the impact of myeloid cells in a number of therapeutic approaches across a wide range of malignancies, as well as the efforts toward the elimination of myeloid cells or the exploitation of their presence for the enhancement of therapeutic efficacy against cancer.
Collapse
Affiliation(s)
- Christiana M Neophytou
- European University Research Centre, Nicosia, Cyprus.,Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus
| | - Chryso Pierides
- The Center for the Study of Haematological Malignancies, Nicosia, Cyprus
| | | | - Paul Costeas
- The Center for the Study of Haematological Malignancies, Nicosia, Cyprus.,The Cyprus Cancer Research Institute, Nicosia, Cyprus
| | | | - Panagiotis Papageorgis
- European University Research Centre, Nicosia, Cyprus.,Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus
| |
Collapse
|
17
|
Bao L, Hao C, Wang J, Wang D, Zhao Y, Li Y, Yao W. High-Dose Cyclophosphamide Administration Orchestrates Phenotypic and Functional Alterations of Immature Dendritic Cells and Regulates Th Cell Polarization. Front Pharmacol 2020; 11:775. [PMID: 32523537 PMCID: PMC7261842 DOI: 10.3389/fphar.2020.00775] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 05/11/2020] [Indexed: 12/29/2022] Open
Abstract
High-dose cyclophosphamide (CTX) inhibits the immune response. Dendritic cells (DCs) are professional antigen presenting cells (APCs) with a crucial role in initiating immune responses and sustaining immune tolerance. The relative contribution of DCs to immunosuppression induced by high-dose CTX is not well-documented. In this study, we employed the CTX-induced immunosuppressive rat model to examine alterations in DCs. We generated and cultured monocyte-derived immature DCs (imDCs) in vitro and explored their capacity of antigen uptake, T cell priming, cytokine production, and surface marker expression following high-dose CTX. Subsequently, we co-cultured CTX-treated imDCs with Th cells to determine Th cell polarization, and further explored the Toll-like receptor/Myeloid differentiation primary response 88/Mitogen-activated protein kinase (TLR/MyD88/MAPK) pathway. Our results show reduced cell number and surface maker alterations in splenic CD103+ DCs of CTX-treated immunosuppressed rats. In vitro, high-dose CTX weakened the antigen uptake capacity and enhanced the T cell priming capacity of imDCs, in addition to triggering imDC surface marker alterations. TLR, MyD88, and MAPK expression levels, involved in mediating Th cell polarization, were also significantly elevated. Our collective findings indicate that high-dose CTX administration potentiates phenotypic and functional alterations of imDC. Such changes may contribute to the regulation of Th polarization.
Collapse
Affiliation(s)
- Lei Bao
- Department of Occupational Health and Environmental Health, School of Public Health, Hebei Medical University, Shijiazhuang, China.,Department of Occupational Health and Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, China.,Hebei Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang, China
| | - Changfu Hao
- Department of Occupational Health and Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, China
| | - Juan Wang
- Department of Statistics, Hebei General Hospital, Shijiazhuang, China
| | - Di Wang
- Department of Occupational Health and Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, China
| | - Youliang Zhao
- Department of Occupational Health and Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, China
| | - Yiping Li
- Department of Occupational Health and Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, China
| | - Wu Yao
- Department of Occupational Health and Environmental Health, School of Public Health, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
18
|
Roghanian A, Hu G, Fraser C, Singh M, Foxall RB, Meyer MJ, Lees E, Huet H, Glennie MJ, Beers SA, Lim SH, Ashton-Key M, Thirdborough SM, Cragg MS, Chen J. Cyclophosphamide Enhances Cancer Antibody Immunotherapy in the Resistant Bone Marrow Niche by Modulating Macrophage FcγR Expression. Cancer Immunol Res 2019; 7:1876-1890. [PMID: 31451483 PMCID: PMC7780711 DOI: 10.1158/2326-6066.cir-18-0835] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 05/06/2019] [Accepted: 08/21/2019] [Indexed: 12/14/2022]
Abstract
Therapy-resistant microenvironments represent a major barrier toward effective elimination of disseminated cancer. Many hematologic and solid tumors are resistant to therapeutic antibodies in the bone marrow (BM), but not in the periphery (e.g., spleen). We previously showed that cyclophosphamide (CTX) sensitizes the BM niche to antibody therapeutics. Here, we show that (i) BM resistance was induced not only by the tumor but also by the intrinsic BM microenvironment; (ii) CTX treatment overcame both intrinsic and extrinsic resistance mechanisms by augmenting macrophage activation and phagocytosis, including significant upregulation of activating Fcγ receptors (FcγRIII and FcγRIV) and downregulation of the inhibitory receptor, FcγRIIB; and (iii) CTX synergized with cetuximab (anti-EGFR) and trastuzumab (anti-Her2) in eliminating metastatic breast cancer in the BM of humanized mice. These findings provide insights into the mechanisms by which CTX synergizes with antibody therapeutics in resistant niche-specific organs and its applicability in treating BM-resident tumors.
Collapse
Affiliation(s)
- Ali Roghanian
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts.
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- Cancer Research UK Centre, University of Southampton, Southampton, United Kindgom
| | - Guangan Hu
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Christopher Fraser
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Maneesh Singh
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Russell B Foxall
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- Cancer Research UK Centre, University of Southampton, Southampton, United Kindgom
| | - Matthew J Meyer
- Novartis Institute for Biomedical Research, Inc., Cambridge, Massachusetts
| | - Emma Lees
- Novartis Institute for Biomedical Research, Inc., Cambridge, Massachusetts
| | - Heather Huet
- Novartis Institute for Biomedical Research, Inc., Cambridge, Massachusetts
| | - Martin J Glennie
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- Cancer Research UK Centre, University of Southampton, Southampton, United Kindgom
| | - Stephen A Beers
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- Cancer Research UK Centre, University of Southampton, Southampton, United Kindgom
| | - Sean H Lim
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- Cancer Research UK Centre, University of Southampton, Southampton, United Kindgom
| | - Margaret Ashton-Key
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- Cancer Research UK Centre, University of Southampton, Southampton, United Kindgom
| | | | - Mark S Cragg
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- Cancer Research UK Centre, University of Southampton, Southampton, United Kindgom
| | - Jianzhu Chen
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts.
| |
Collapse
|
19
|
Wu J, Waxman DJ. Immunogenic chemotherapy: Dose and schedule dependence and combination with immunotherapy. Cancer Lett 2019; 419:210-221. [PMID: 29414305 DOI: 10.1016/j.canlet.2018.01.050] [Citation(s) in RCA: 234] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 01/11/2018] [Accepted: 01/12/2018] [Indexed: 12/20/2022]
Abstract
Conventional cytotoxic cancer chemotherapy is often immunosuppressive and associated with drug resistance and tumor regrowth after a short period of tumor shrinkage or growth stasis. However, certain cytotoxic cancer chemotherapeutic drugs, including doxorubicin, mitoxantrone, and cyclophosphamide, can kill tumor cells by an immunogenic cell death pathway, which activates robust innate and adaptive anti-tumor immune responses and has the potential to greatly increase the efficacy of chemotherapy. Here, we review studies on chemotherapeutic drug-induced immunogenic cell death, focusing on how the choice of a conventional cytotoxic agent and its dose and schedule impact anti-tumor immune responses. We propose a strategy for effective immunogenic chemotherapy that employs a modified metronomic schedule for drug delivery, which we term medium-dose intermittent chemotherapy (MEDIC). Striking responses have been seen in preclinical cancer models using MEDIC, where an immunogenic cancer chemotherapeutic agent is administered intermittently and at an intermediate dose, designed to impart strong and repeated cytotoxic damage to tumors, and on a schedule compatible with activation of a sustained anti-tumor immune response, thereby maximizing anti-cancer activity. We also discuss strategies for combination chemo-immunotherapy, and we outline approaches to identify new immunogenic chemotherapeutic agents for drug development.
Collapse
Affiliation(s)
- Junjie Wu
- Department of Biology, Division of Cell and Molecular Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA
| | - David J Waxman
- Department of Biology, Division of Cell and Molecular Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA.
| |
Collapse
|
20
|
Lin W, Chen M, Hong L, Zhao H, Chen Q. Crosstalk Between PD-1/PD-L1 Blockade and Its Combinatorial Therapies in Tumor Immune Microenvironment: A Focus on HNSCC. Front Oncol 2018; 8:532. [PMID: 30519541 PMCID: PMC6258806 DOI: 10.3389/fonc.2018.00532] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 10/30/2018] [Indexed: 02/05/2023] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is the sixth most common malignancy worldwide with a poor prognosis and high mortality. More than two-thirds of HNSCC patients still have no effective control of clinical progression, and the five-year survival rate is < 50%. Moreover, patients with platinum-refractory HNSCC have a median survival of < 6 months. The significant toxicity and low survival rates of current treatment strategies highlight the necessity for new treatment modalities. Recently, a large number of studies have demonstrated that programmed cell death protein-1 (PD-1) and its ligand, programmed cell death protein ligand-1 (PD-L1) play an essential role in tumor initiation and progression. PD-1/PD-L1 blockade has shown a desired and long-lasting therapeutic effect in the treatment of HNSCC and other malignancies. However, only a small number of patients with HNSCC can benefit from PD-1/PD-L1 blockade monotherapy, while the majority of patients do not respond. To overcome the unsatisfactory therapeutic effect of PD-1/PD-L1 blockade monotherapy, combining other treatment options for HNSCC (including chemotherapy, radiotherapy, targeted therapy, and immunotherapy) in the treatment scheme has become a commonly used strategy. Herein, the potential mechanisms underlying the crosstalk between PD-1/PD-L1 blockade and its combinatorial therapies for HNSCC were reviewed, and it is hoped that the improved understanding of the crosstalk process would provide further ideas for the design of a combinatorial regimen with a higher efficiency and response rate for the treatment of HNSCC and other malignancies.
Collapse
Affiliation(s)
- Weimin Lin
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Miao Chen
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Le Hong
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hang Zhao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qianming Chen
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
21
|
High-grade glioma associated immunosuppression does not prevent immune responses induced by therapeutic vaccines in combination with T reg depletion. Cancer Immunol Immunother 2018; 67:1545-1558. [PMID: 30054667 PMCID: PMC6182405 DOI: 10.1007/s00262-018-2214-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Accepted: 07/20/2018] [Indexed: 01/20/2023]
Abstract
High-grade gliomas (HGG) exert systemic immunosuppression, which is of particular importance as immunotherapeutic strategies such as therapeutic vaccines are increasingly used to treat HGGs. In a first cohort of 61 HGG patients we evaluated a panel of 30 hematological and 34 plasma biomarkers. Then, we investigated in a second cohort of 11 relapsed HGG patients receiving immunomodulation with metronomic cyclophosphamide upfront to a DC-based vaccine whether immune abnormalities persisted and whether they hampered induction of IFNγ+ T-cell responses. HGG patients from the first cohort showed increased numbers of leukocytes, neutrophils and MDSCs and in parallel reduced numbers of CD4+/CD8+ T-cells, plasmacytoid and conventional DC2s. MDSCs and T-cell alterations were more profound in WHO IV° glioma patients. Moreover, levels of MDSCs and epidermal growth factor were negatively associated with survival. Serum levels of IL-2, IL-4, IL-5 and IL-10 were altered in HGG patients, however, without any impact on clinical outcome. In the immunotherapy cohort, 6-month overall survival was 100%. Metronomic cyclophosphamide led to > 40% reduction of regulatory T cells (Treg). In parallel to Treg-depletion, MDSCs and DC subsets became indistinguishable from healthy controls, whereas T-lymphopenia persisted. Despite low T-cells, IFNγ-responses could be induced in 9/10 analyzed cases. Importantly, frequency of CD8+VLA-4+ T-cells with CNS-homing properties, but not of CD4+ VLA-4+ T-cells, increased during vaccination. Our study identifies several features of systemic immunosuppression in HGGs. Metronomic cyclophosphamide in combination with an active immunization alleviates the latter and the combined treatment allows induction of a high rate of anti-glioma immune responses.
Collapse
|
22
|
Bao L, Hao C, Liu S, Zhang L, Wang J, Wang D, Li Y, Yao W. Dendritic cells trigger imbalance of Th1/Th2 cells in silica dust exposure rat model via MHC-II, CD80, CD86 and IL-12. RSC Adv 2018; 8:26108-26115. [PMID: 35541981 PMCID: PMC9083086 DOI: 10.1039/c8ra03970d] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 07/07/2018] [Indexed: 12/24/2022] Open
Abstract
Silicosis is one of the most common occupational respiratory diseases caused by inhaling silica dust over a prolonged period of time, and the progression of silicosis is accompanied with chronic inflammation and progressive pulmonary fibrosis, in which dendritic cells (DCs), the most powerful antigen presentation cell (APC) in the immune response, play a crucial role. To investigate the role of DCs in the development of silicosis, we established an experimental silicosis rat model and examined the number of DCs and alveolar macrophages (AMs) in lung tissues using immunofluorescence over 84 days. Additionally, to obtain an overview of the immunological changes in rat lung tissues, a series of indicators including Th1/Th2 cells, IFN-γ, IL-4, MHC-II, CD80/86 and IL-12 were detected using flow cytometry and an enzyme-linked immunosorbent assay (ELISA) as well as a real-time polymerase chain reaction (PCR) assay. We observed that the number of DCs slightly increased at the inflammatory stage, and it increased significantly at the final stage of fibrosis. Polarization of Th1 cells and IFN-γ expressions were dominant during the inflammatory stage, whereas polarization of Th2 cells and IL-4 expressions were dominant during the fibrotic stage. The subsequent mechanistic study found that the expressions of MHC-II, CD80/86 and IL-12, which are the key molecules that connect DCs and Th cells, changed dynamically in the experimental silicosis rat model. The data obtained in this study indicated that the increase in DCs may contribute to polarization of Th1/Th2 cells via MHC-II, CD80/86, and IL-12 in silica dust-exposed rats.
Collapse
Affiliation(s)
- Lei Bao
- School of Public Health, Zhengzhou University No. 100 Science Avenue Zhengzhou Henan 450001 China +86-371-67781922 +86-371-67781922
| | - Changfu Hao
- School of Public Health, Zhengzhou University No. 100 Science Avenue Zhengzhou Henan 450001 China +86-371-67781922 +86-371-67781922
| | - Suna Liu
- The Third Affiliated Hospital of Zhengzhou University Zhengzhou Henan 450001 China
| | - Lin Zhang
- School of Public Health, Zhengzhou University No. 100 Science Avenue Zhengzhou Henan 450001 China +86-371-67781922 +86-371-67781922
| | - Juan Wang
- Hebei General Hospital Shijiazhuang Hebei 050000 China
| | - Di Wang
- School of Public Health, Zhengzhou University No. 100 Science Avenue Zhengzhou Henan 450001 China +86-371-67781922 +86-371-67781922
| | - Yiping Li
- School of Public Health, Zhengzhou University No. 100 Science Avenue Zhengzhou Henan 450001 China +86-371-67781922 +86-371-67781922
| | - Wu Yao
- School of Public Health, Zhengzhou University No. 100 Science Avenue Zhengzhou Henan 450001 China +86-371-67781922 +86-371-67781922
| |
Collapse
|
23
|
Systemic immune response induced by oxaliplatin-based neoadjuvant therapy favours survival without metastatic progression in high-risk rectal cancer. Br J Cancer 2018; 118:1322-1328. [PMID: 29695770 PMCID: PMC5959927 DOI: 10.1038/s41416-018-0085-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Revised: 03/20/2018] [Accepted: 03/22/2018] [Indexed: 12/22/2022] Open
Abstract
Background Systemic failure remains a challenge in rectal cancer. We investigated the possible systemic anti-tumour immune activity invoked within oxaliplatin-based neoadjuvant therapy. Methods In two high-risk patient cohorts, we assessed the circulating levels of the fms-like tyrosine kinase 3 ligand (Flt3L), a factor reflecting both therapy-induced myelosuppression and activation of tumour antigen-presenting dendritic cells, at baseline and following induction chemotherapy and sequential chemoradiotherapy, both modalities containing oxaliplatin. The primary end point was progression-free survival (PFS). Results In both cohorts, the median Flt3L level was significantly higher at completion of each sequential modality than at baseline. The 5-year PFS (most events being metastatic progression) was 68% and 71% in the two cohorts consisting of 33% and 52% T4 cases. In the principal cohort, a high Flt3L level following the induction chemotherapy was associated with low risk for a PFS event (HR: 0.15; P < 0.01). These patients also had available dose scheduling and toxicity data, revealing that oxaliplatin dose reduction during chemoradiotherapy, undertaken to maintain compliance to the radiotherapy protocol, was associated with advantageous PFS (HR: 0.47; P = 0.046). Conclusion In high-risk rectal cancer, oxaliplatin-containing neoadjuvant therapy may promote an immune response that favours survival without metastatic progression.
Collapse
|
24
|
Bryant CE, Sutherland S, Kong B, Papadimitrious MS, Fromm PD, Hart DNJ. Dendritic cells as cancer therapeutics. Semin Cell Dev Biol 2018; 86:77-88. [PMID: 29454038 DOI: 10.1016/j.semcdb.2018.02.015] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 12/14/2017] [Accepted: 02/10/2018] [Indexed: 02/06/2023]
Abstract
The ability of immune therapies to control cancer has recently generated intense interest. This therapeutic outcome is reliant on T cell recognition of tumour cells. The natural function of dendritic cells (DC) is to generate adaptive responses, by presenting antigen to T cells, hence they are a logical target to generate specific anti-tumour immunity. Our understanding of the biology of DC is expanding, and they are now known to be a family of related subsets with variable features and function. Most clinical experience to date with DC vaccination has been using monocyte-derived DC vaccines. There is now growing experience with alternative blood-derived DC derived vaccines, as well as with multiple forms of tumour antigen and its loading, a wide range of adjuvants and different modes of vaccine delivery. Key insights from pre-clinical studies, and lessons learned from early clinical testing drive progress towards improved vaccines. The potential to fortify responses with other modalities of immunotherapy makes clinically effective "second generation" DC vaccination strategies a priority for cancer immune therapists.
Collapse
Affiliation(s)
- Christian E Bryant
- Institute of Haematology, Royal Prince Alfred Hospital, Camperdown, NSW Australia; Dendritic Cell Research, ANZAC Research Institute, Concord, NSW Australia.
| | - Sarah Sutherland
- Dendritic Cell Research, ANZAC Research Institute, Concord, NSW Australia; Sydney Medical School, The University of Sydney, Sydney, NSW Australia
| | - Benjamin Kong
- Dendritic Cell Research, ANZAC Research Institute, Concord, NSW Australia; Sydney Medical School, The University of Sydney, Sydney, NSW Australia
| | - Michael S Papadimitrious
- Dendritic Cell Research, ANZAC Research Institute, Concord, NSW Australia; Sydney Medical School, The University of Sydney, Sydney, NSW Australia
| | - Phillip D Fromm
- Dendritic Cell Research, ANZAC Research Institute, Concord, NSW Australia; Sydney Medical School, The University of Sydney, Sydney, NSW Australia
| | - Derek N J Hart
- Institute of Haematology, Royal Prince Alfred Hospital, Camperdown, NSW Australia; Dendritic Cell Research, ANZAC Research Institute, Concord, NSW Australia; Sydney Medical School, The University of Sydney, Sydney, NSW Australia.
| |
Collapse
|
25
|
Hijikata Y, Okazaki T, Tanaka Y, Murahashi M, Yamada Y, Yamada K, Takahashi A, Inoue H, Kishimoto J, Nakanishi Y, Oda Y, Nakamura Y, Tani K. A phase I clinical trial of RNF43 peptide-related immune cell therapy combined with low-dose cyclophosphamide in patients with advanced solid tumors. PLoS One 2018; 13:e0187878. [PMID: 29293510 PMCID: PMC5749706 DOI: 10.1371/journal.pone.0187878] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Accepted: 10/26/2017] [Indexed: 12/22/2022] Open
Abstract
The objective of this study was to investigate the safety and the tolerability of combined cellular immunotherapy with low-dose cyclophosphamide (CPA) in patients with advanced solid tumors. This study targeted a novel tumor-associated antigen, ring finger protein 43 (RNF43). Eligible patients were resistant to standard therapy, HLA-A*24:02- or A*02:01-positive and exhibiting high RNF43 expression in their tumor cells. They were administered 300 mg/m2 CPA followed by autologous lymphocytes, preliminarily cultured with autologous RNF43 peptide-pulsed dendritic cells (DCs), RNF43 peptide-pulsed DCs and systemic low dose interleukin-2. The primary endpoint was safety whereas the secondary endpoint was immunological and clinical response to treatment. Ten patients, in total, were enrolled in this trial. Primarily, no adverse events greater than Grade 3 were observed. Six out of 10 patients showed stable disease (SD) on day 49, while 4 other patients showed progressive disease. In addition, one patient with SD exhibited a partial response after the second trial. The frequency of regulatory T cells (Tregs) in patients with SD significantly decreased after CPA administration. The ratio of interferon-γ-producing, tumor-reactive CD8+ T cells increased with time in patients with SD. We successfully showed that the combination of immune cell therapy and CPA was safe, might induce tumor-specific immune responses and clinical efficacy, and was accompanied by a decreased ratio of Tregs in patients with RNF43-positive advanced solid tumors.
Collapse
Affiliation(s)
- Yasuki Hijikata
- Department of Advanced Cell and Molecular Therapy, Kyushu University Hospital, Fukuoka, Japan
| | - Toshihiko Okazaki
- ARO Advanced Medical Center, Kyushu University Hospital, Fukuoka, Japan
| | - Yoshihiro Tanaka
- ARO Advanced Medical Center, Kyushu University Hospital, Fukuoka, Japan
| | - Mutsunori Murahashi
- Department of Advanced Cell and Molecular Therapy, Kyushu University Hospital, Fukuoka, Japan
| | - Yuichi Yamada
- Department of Anatomic Pathology, Pathological Sciences, Kyushu University, Fukuoka, Japan
| | - Kazunari Yamada
- Department of Advanced Cell and Molecular Therapy, Kyushu University Hospital, Fukuoka, Japan
| | - Atsushi Takahashi
- Department of Advanced Cell and Molecular Therapy, Kyushu University Hospital, Fukuoka, Japan
| | - Hiroyuki Inoue
- Department of Advanced Cell and Molecular Therapy, Kyushu University Hospital, Fukuoka, Japan
| | - Junji Kishimoto
- ARO Advanced Medical Center, Kyushu University Hospital, Fukuoka, Japan
| | - Yoichi Nakanishi
- Research Institute of Diseases of Chest, Kyushu University, Fukuoka, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Pathological Sciences, Kyushu University, Fukuoka, Japan
| | - Yusuke Nakamura
- Human genome center, Institute of medical science, University of Tokyo, Tokyo, Japan
| | - Kenzaburo Tani
- Department of Advanced Cell and Molecular Therapy, Kyushu University Hospital, Fukuoka, Japan
- Project Division of ALA Advanced Medical Research, Advanced Medical Science of Internal Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- * E-mail:
| |
Collapse
|
26
|
Ishii H, Chikamatsu K, Igarashi S, Takahashi H, Sakamoto K, Higuchi H, Tanaka S, Matsuoka T, Masuyama K. Establishment of Synergistic Chemoimmunotherapy for Head and Neck Cancer Using Peritumoral Immature Dendritic Cell Injections and Low-Dose Chemotherapies. Transl Oncol 2017; 11:132-139. [PMID: 29268186 PMCID: PMC5738236 DOI: 10.1016/j.tranon.2017.11.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 11/28/2017] [Indexed: 01/30/2023] Open
Abstract
The lack of available tumor antigens with strong immunogenicity, human leukocyte antigen restriction, and immunosuppression via regulatory T-cells (Tregs) and myeloid-derived suppressor cells are limitations for dendritic cell (DC)–based immunotherapy in patients with advanced head and neck cancer (HNC). We sought to overcome these limitations and induce effective antitumor immunity in the host. The effect of low-dose docetaxel (DTX) treatment on DC maturation was examined in an ex vivo study, and a phase I clinical trial of combination therapy with direct peritumoral immature DC (iDC) injection with OK-432 and low-dose cyclophosphamide (CTX) plus DTX was designed. Low-dose DTX did not negatively affect iDC viability and instead promoted maturation and IL-12 production. Five patients with metastatic or recurrent HNC were enrolled for the trial. All patients experienced grade 1 to 3 fevers. Intriguingly, elevated CD8+ effector T-cells and reduced Tregs were observed in four patients who completed two treatment cycles. All patients were judged to have progressive disease, but tumor regressions were observed in a subset of targeted metastatic lesions in two of five patients. Our results show that the combination of direct peritumoral iDC injection with OK-432 and low-dose CTX plus DTX is well tolerated and should give rise to changing the immune profile of T-cell subsets and improvement of immunosuppression in advanced HNC patients. Additionally, our ex vivo data on the effect of low-dose DTX treatment on DC maturation may contribute to developing new combination therapies with low-dose chemotherapy and immunotherapy.
Collapse
Affiliation(s)
- Hiroki Ishii
- Department of Otolaryngology, Head and Neck Surgery, University of Yamanashi.
| | - Kazuaki Chikamatsu
- Department of Otolaryngology, Head and Neck Surgery, University of Yamanashi; Department of Otolaryngology-Head and Neck Surgery, Gunma University Graduate School of Medicine
| | - Satoshi Igarashi
- Department of Otolaryngology, Head and Neck Surgery, University of Yamanashi
| | - Hideyuki Takahashi
- Department of Otolaryngology-Head and Neck Surgery, Gunma University Graduate School of Medicine
| | - Kaname Sakamoto
- Department of Otolaryngology, Head and Neck Surgery, University of Yamanashi
| | - Hiroji Higuchi
- Division of Transfusion Medicine and Cell Therapy, University of Yamanashi Hospital
| | - Shota Tanaka
- Department of Otolaryngology, Head and Neck Surgery, University of Yamanashi
| | - Tomokazu Matsuoka
- Department of Otolaryngology, Head and Neck Surgery, University of Yamanashi
| | - Keisuke Masuyama
- Department of Otolaryngology, Head and Neck Surgery, University of Yamanashi.
| |
Collapse
|
27
|
Scurr M, Pembroke T, Bloom A, Roberts D, Thomson A, Smart K, Bridgeman H, Adams R, Brewster A, Jones R, Gwynne S, Blount D, Harrop R, Hills R, Gallimore A, Godkin A. Low-Dose Cyclophosphamide Induces Antitumor T-Cell Responses, which Associate with Survival in Metastatic Colorectal Cancer. Clin Cancer Res 2017; 23:6771-6780. [PMID: 28855352 PMCID: PMC5769815 DOI: 10.1158/1078-0432.ccr-17-0895] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 07/18/2017] [Accepted: 08/25/2017] [Indexed: 01/04/2023]
Abstract
Purpose: Anticancer T-cell responses can control tumors, but immunosuppressive mechanisms in vivo prevent their function. The role of regulatory T cells (Tregs) in metastatic colorectal cancer is unclear. We have previously shown depletion of Tregs enhances colorectal cancer-specific effector T-cell responses. Low-dose cyclophosphamide targets Tregs in animal models and some human studies; however, the effect of cyclophosphamide in metastatic colorectal cancer is unknown.Experimental Design: Fifty-five patients with metastatic colorectal cancer were enrolled in a phase I/II trial and randomly assigned to receive 2-week-long courses of low-dose (50 mg twice a day) cyclophosphamide or not. The absolute number, phenotype, and antitumor function of peripheral blood-derived lymphocyte subsets were monitored throughout treatment, as well as during 18-month follow-up.Results: Initially, cyclophosphamide reduced proliferation in all lymphocyte subsets; however, a rapid mobilization of effector T cells overcame this decrease, leading to increased absolute T-cell numbers. In contrast, a reduction in proportional and absolute Treg, B-cell, and NK-cell numbers occurred. The expansion and subsequent activation of effector T cells was focused on tumor-specific T cells, producing both granzyme B and IFNγ. Cyclophosphamide-treated patients demonstrating the most enhanced IFNγ+ tumor-specific T-cell responses exhibited a significant delay in tumor progression [HR = 0.29; 95% confidence interval (CI), 0.12-0.69; P = 0.0047), compared with nonresponders and no-treatment controls.Conclusions: Cyclophosphamide-induced Treg depletion is mirrored by a striking boost in antitumor immunity. This study provides the first direct evidence of the benefit of naturally primed T cells in patients with metastatic colorectal cancer. Our results also support the concept that nonmutated self-antigens may act as useful targets for immunotherapies. Clin Cancer Res; 23(22); 6771-80. ©2017 AACR.
Collapse
Affiliation(s)
- Martin Scurr
- Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom
| | - Tom Pembroke
- Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom
| | - Anja Bloom
- Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom
| | - David Roberts
- Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom
| | - Amanda Thomson
- Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom
| | - Kathryn Smart
- Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom
| | - Hayley Bridgeman
- Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom
| | - Richard Adams
- Velindre NHS Trust, Whitchurch, Cardiff, United Kingdom
| | | | - Robert Jones
- Velindre NHS Trust, Whitchurch, Cardiff, United Kingdom
| | - Sarah Gwynne
- South West Wales Cancer Centre, Swansea, United Kingdom
| | | | | | - Robert Hills
- Cancer Trials Unit (Translational Statistics), Cardiff University, Cardiff, United Kingdom
| | - Awen Gallimore
- Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom
| | - Andrew Godkin
- Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom.
- Department of Gastroenterology and Hepatology, University Hospital of Wales, Heath Park, Cardiff, United Kingdom
| |
Collapse
|
28
|
Scurr M, Pembroke T, Bloom A, Roberts D, Thomson A, Smart K, Bridgeman H, Adams R, Brewster A, Jones R, Gwynne S, Blount D, Harrop R, Wright M, Hills R, Gallimore A, Godkin A. Effect of Modified Vaccinia Ankara-5T4 and Low-Dose Cyclophosphamide on Antitumor Immunity in Metastatic Colorectal Cancer: A Randomized Clinical Trial. JAMA Oncol 2017; 3:e172579. [PMID: 28880972 PMCID: PMC5824319 DOI: 10.1001/jamaoncol.2017.2579] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 06/14/2017] [Indexed: 01/04/2023]
Abstract
IMPORTANCE The success of immunotherapy with checkpoint inhibitors is not replicated in most cases of colorectal cancer; therefore, different strategies are urgently required. The oncofetal antigen 5T4 is expressed in more than 90% of cases of metastatic colorectal cancer (mCRC). Preliminary data using modified vaccinia Ankara-5T4 (MVA-5T4) in mCRC demonstrated that it safely induced serologic and T-cell responses. OBJECTIVE To determine whether antitumor immunity in mCRC could be increased using MVA-5T4, metronomic low-dose cyclophosphamide, or a combination of both treatments. DESIGN, SETTING, AND PARTICIPANTS In this randomized clinical trial, 55 patients with inoperable mCRC and prior stable disease after standard chemotherapy were enrolled at a single center and randomized to watch and wait (n = 9), cyclophosphamide treatment only (n = 9), MVA-5T4 only (n = 19), and a combination of MVA-5T4 and cyclophosphamide (n = 18). Patients were enrolled and treated from July 9, 2012, through February 8, 2016, and follow-up was completed on December 13, 2016. Data were analyzed based on intention to treat. INTERVENTIONS Patients randomized to a cyclophosphamide group received 50 mg twice daily on treatment days 1 to 7 and 15 to 21. Patients randomized to a MVA-5T4 group received an intramuscular injection at a dose of 1 × 109 50% tissue culture infectious dose on treatment days 22, 36, 50, 64, 78, and 106. MAIN OUTCOMES AND MEASURES The predefined primary end point was the magnitude of anti-5T4 immune responses (5T4-specific T-cell and antibody levels) generated at treatment week 7. Secondary end points included analysis of the kinetics of anti-5T4 responses, progression-free survival (PFS), and overall survival (OS). RESULTS Fifty-two patients (38 men and 14 women; mean [SD] age, 64.2 [10.1] years) were included in the study analysis. The 5T4-specific antibody immune responses were significantly increased in the MVA-5T4 (83.41 [36.09] relative units [RU]; P = .02) and combination treatment (65.81 [16.68] RU; P = .002) groups compared with no treatment (20.09 [7.20] RU). Cyclophosphamide depleted regulatory T cells in 24 of 27 patients receiving MVA-5T4, independently prolonging PFS (5.0 vs 2.5 months; hazard ratio [HR], 0.48; 95% CI, 0.21-1.11; P = .09). MVA-5T4 doubled baseline anti-5T4 responses in 16 of 35 patients, resulting in significantly prolonged PFS (5.6 vs 2.4 months; HR, 0.21; 95% CI, 0.09-0.47; P < .001) and OS (20.0 vs 10.3 months; HR, 0.32; 95% CI, 0.14-0.74; P = .008). No grade 3 or 4 adverse events were observed. CONCLUSIONS AND RELEVANCE This initial randomized clinical immunotherapy study demonstrates a significant survival benefit in mCRC. Prior depletion of regulatory T cells by cyclophosphamide did not increase immune responses generated by MVA-5T4 vaccination; however, cyclophosphamide and MVA-5T4 each independently induced beneficial antitumor immune responses, resulting in prolonged survival without toxic effects. Larger clinical trials are planned to further validate these data. TRIAL REGISTRATION isrctn.org Identifier: ISRCTN54669986.
Collapse
Affiliation(s)
- Martin Scurr
- Division of Infection and Immunity, Cardiff University, Cardiff, Wales
| | - Tom Pembroke
- Division of Infection and Immunity, Cardiff University, Cardiff, Wales
| | - Anja Bloom
- Division of Infection and Immunity, Cardiff University, Cardiff, Wales
| | - David Roberts
- Division of Infection and Immunity, Cardiff University, Cardiff, Wales
| | - Amanda Thomson
- Division of Infection and Immunity, Cardiff University, Cardiff, Wales
| | - Kathryn Smart
- Division of Infection and Immunity, Cardiff University, Cardiff, Wales
| | - Hayley Bridgeman
- Division of Infection and Immunity, Cardiff University, Cardiff, Wales
| | - Richard Adams
- Velindre Cancer Centre, National Health Service (NHS) Trust, Cardiff, Wales
| | - Alison Brewster
- Velindre Cancer Centre, National Health Service (NHS) Trust, Cardiff, Wales
| | - Robert Jones
- Velindre Cancer Centre, National Health Service (NHS) Trust, Cardiff, Wales
| | - Sarah Gwynne
- South West Wales Cancer Centre, Singleton Hospital, NHS Trust, Swansea
| | | | | | - Melissa Wright
- Centre for Trials Research, Cardiff University, Cardiff, Wales
| | - Robert Hills
- Centre for Trials Research, Cardiff University, Cardiff, Wales
| | - Awen Gallimore
- Division of Infection and Immunity, Cardiff University, Cardiff, Wales
| | - Andrew Godkin
- Division of Infection and Immunity, Cardiff University, Cardiff, Wales
- Department of Gastroenterology and Hepatology, University Hospital of Wales, Cardiff
| |
Collapse
|
29
|
Constantino J, Gomes C, Falcão A, Neves BM, Cruz MT. Dendritic cell-based immunotherapy: a basic review and recent advances. Immunol Res 2017; 65:798-810. [DOI: 10.1007/s12026-017-8931-1] [Citation(s) in RCA: 145] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
30
|
Krais JJ, Virani N, McKernan PH, Nguyen Q, Fung KM, Sikavitsas VI, Kurkjian C, Harrison RG. Antitumor Synergism and Enhanced Survival with a Tumor Vasculature-Targeted Enzyme Prodrug System, Rapamycin, and Cyclophosphamide. Mol Cancer Ther 2017; 16:1855-1865. [PMID: 28522586 DOI: 10.1158/1535-7163.mct-16-0263] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 01/17/2017] [Accepted: 05/09/2017] [Indexed: 11/16/2022]
Abstract
Mutant cystathionine gamma-lyase was targeted to phosphatidylserine exposed on tumor vasculature through fusion with Annexin A1 or Annexin A5. Cystathionine gamma-lyase E58N, R118L, and E338N mutations impart nonnative methionine gamma-lyase activity, resulting in tumor-localized generation of highly toxic methylselenol upon systemic administration of nontoxic selenomethionine. The described therapeutic system circumvents systemic toxicity issues using a novel drug delivery/generation approach and avoids the administration of nonnative proteins and/or DNA required with other enzyme prodrug systems. The enzyme fusion exhibits strong and stable in vitro binding with dissociation constants in the nanomolar range for both human and mouse breast cancer cells and in a cell model of tumor vascular endothelium. Daily administration of the therapy suppressed growth of highly aggressive triple-negative murine 4T1 mammary tumors in immunocompetent BALB/cJ mice and MDA-MB-231 tumors in SCID mice. Treatment did not result in the occurrence of negative side effects or the elicitation of neutralizing antibodies. On the basis of the vasculature-targeted nature of the therapy, combinations with rapamycin and cyclophosphamide were evaluated. Rapamycin, an mTOR inhibitor, reduces the prosurvival signaling of cells in a hypoxic environment potentially exacerbated by a vasculature-targeted therapy. IHC revealed, unsurprisingly, a significant hypoxic response (increase in hypoxia-inducible factor 1 α subunit, HIF1A) in the enzyme prodrug-treated tumors and a dramatic reduction of HIF1A upon rapamycin treatment. Cyclophosphamide, an immunomodulator at low doses, was combined with the enzyme prodrug therapy and rapamycin; this combination synergistically reduced tumor volumes, inhibited metastatic progression, and enhanced survival. Mol Cancer Ther; 16(9); 1855-65. ©2017 AACR.
Collapse
Affiliation(s)
- John J Krais
- School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma
| | - Needa Virani
- School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma
| | - Patrick H McKernan
- School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma
| | - Quang Nguyen
- School of Chemical, Biological and Materials Engineering, University of Oklahoma, Norman, Oklahoma
| | - Kar-Ming Fung
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Vassilios I Sikavitsas
- School of Chemical, Biological and Materials Engineering, University of Oklahoma, Norman, Oklahoma
| | - Carla Kurkjian
- Oncology/Hematology Section, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Roger G Harrison
- School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma. .,School of Chemical, Biological and Materials Engineering, University of Oklahoma, Norman, Oklahoma
| |
Collapse
|
31
|
Li J, Chen T, Yuan C, Zhao G, xu M, Li X, Cao J, Xing L. Effect of intravenous immunoglobulin on the function of Treg cells derived from immunosuppressed mice with Pseudomonas aeruginosa pneumonia. PLoS One 2017; 12:e0176843. [PMID: 28481908 PMCID: PMC5421750 DOI: 10.1371/journal.pone.0176843] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 04/18/2017] [Indexed: 12/31/2022] Open
Abstract
AIM The present study aimed to investigate the effect of intravenous immunoglobulin (IVIG) on regulatory T (Treg) cells derived from immunosuppressed mice with Pseudomonas aeruginosa (PA) pneumonia. METHODS A total of 108 BALB/c mice were randomly divided into the following groups: control group (Control), immunosuppressed group (IS), PA pneumonia group (PA), PA pneumonia in immunosuppressed group (IS + PA), PA pneumonia with IVIG treatment in immunocompetent group (PA + IVIG) and PA pneumonia with IVIG treatment in immunosuppressed group (IS + PA + IVIG). Each group comprised 18 mice. The combined PA pneumonia in immunosuppressed model and the treatment models were established. The mice in each group were sacrificed at 4, 8, and 24 h time points. The general condition and pathological changes in the lung tissues of the mice were monitored. Reverse transcription-polymerase chain reaction was used to detect the forkhead box P3 (FOXP3) mRNA relative expression level in the lung tissues. The enzyme-linked immunosorbent assay was used to detect the serum concentration of active transforming growth factor beta (TGF-β). RESULTS No inflammatory response were exhibited in the lung tissues of the mice in Control group and IS group, while varying degrees of acute lung injury were revealed in the mice in PA group, IS + PA group, PA + IVIG group and IS + PA + IVIG group. Lung tissue injury was most apparent at the 8 h time point, and it indicated the greatest effect in IS + PA group. Whereas tissue damages were alleviated in PA + IVIG group and IS + PA + IVIG group compared with IS + PA group. In addition, tissue damage lessened in PA + IVIG group compared with PA group and IS + PA + IVIG group. FOXP3 mRNA expression levels in the lung tissues and the serum concentration of TGF-β were lower in IS group, PA group, IS + PA group and IS + PA + IVIG group at the 4, 8 and 24 h time points, respectively compared with Control group. FOXP3 mRNA expression levels decreased in PA + IVIG group at the 4h time point and TGF-β serum concentrations decreased at the 4 and 8h time points compared with Control group, and subsequently increased. CONCLUSIONS In the immunosuppred model with PA pneumonia, the immune system was greatly compromised. IVIG partially restored the immunosuppressed functions of Treg cells, suppressed the overactivated immune system and ameliorated the development of the disease.
Collapse
Affiliation(s)
- Junlu Li
- Department of Respiratory Intensive Care Unit (RICU), The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Tingsang Chen
- Department of Respiratory Intensive Care Unit (RICU), The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Congcong Yuan
- Department of Respiratory Intensive Care Unit (RICU), The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Guoqiang Zhao
- School of basic medical sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Min xu
- Department of Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaoyan Li
- Department of Respiratory Intensive Care Unit (RICU), The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jie Cao
- Department of Respiratory Intensive Care Unit (RICU), The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lihua Xing
- Department of Respiratory Intensive Care Unit (RICU), The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- * E-mail:
| |
Collapse
|
32
|
Herrera FG, Bourhis J, Coukos G. Radiotherapy combination opportunities leveraging immunity for the next oncology practice. CA Cancer J Clin 2017; 67:65-85. [PMID: 27570942 DOI: 10.3322/caac.21358] [Citation(s) in RCA: 356] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Approximately one-half of patients with newly diagnosed cancer and many patients with persistent or recurrent tumors receive radiotherapy (RT), with the explicit goal of eliminating tumors through direct killing. The current RT dose and schedule regimens have been empirically developed. Although early clinical studies revealed that RT could provoke important responses not only at the site of treatment but also on remote, nonirradiated tumor deposits-the so-called "abscopal effect"- the underlying mechanisms were poorly understood and were not therapeutically exploited. Recent work has elucidated the immune mechanisms underlying these effects and has paved the way for developing combinations of RT with immune therapy. In the wake of recent therapeutic breakthroughs in the field of immunotherapy, rational combinations of immunotherapy with RT could profoundly change the standard of care for many tumor types in the next decade. Thus, a deep understanding of the immunologic effects of RT is urgently needed to design the next generation of therapeutic combinations. Here, the authors review the immune mechanisms of tumor radiation and summarize the preclinical and clinical evidence on immunotherapy-RT combinations. Furthermore, a framework is provided for the practicing clinician and the clinician investigator to guide the development of novel combinations to more rapidly advance this important field. CA Cancer J Clin 2017;67:65-85. © 2016 American Cancer Society.
Collapse
Affiliation(s)
- Fernanda G Herrera
- Radiation Oncologist, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
- Instructor, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Jean Bourhis
- Professor, Chief of Radiation Oncology Service, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - George Coukos
- Professor, Director, Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
- Director, Ludwig Institute for Cancer Research, University of Lausanne Branch, Lausanne, Switzerland
| |
Collapse
|
33
|
Qi S, Li H, Lu L, Qi Z, Liu L, Chen L, Shen G, Fu L, Luo Q, Zhang Z. Long-term intravital imaging of the multicolor-coded tumor microenvironment during combination immunotherapy. eLife 2016; 5. [PMID: 27855783 PMCID: PMC5173323 DOI: 10.7554/elife.14756] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 10/17/2016] [Indexed: 12/22/2022] Open
Abstract
The combined-immunotherapy of adoptive cell therapy (ACT) and cyclophosphamide (CTX) is one of the most efficient treatments for melanoma patients. However, no synergistic effects of CTX and ACT on the spatio-temporal dynamics of immunocytes in vivo have been described. Here, we visualized key cell events in immunotherapy-elicited immunoreactions in a multicolor-coded tumor microenvironment, and then established an optimal strategy of metronomic combined-immunotherapy to enhance anti-tumor efficacy. Intravital imaging data indicated that regulatory T cells formed an 'immunosuppressive ring' around a solid tumor. The CTX-ACT combined-treatment elicited synergistic immunoreactions in tumor areas, which included relieving the immune suppression, triggering the transient activation of endogenous tumor-infiltrating immunocytes, increasing the accumulation of adoptive cytotoxic T lymphocytes, and accelerating the infiltration of dendritic cells. These insights into the spatio-temporal dynamics of immunocytes are beneficial for optimizing immunotherapy and provide new approaches for elucidating the mechanisms underlying the involvement of immunocytes in cancer immunotherapy. DOI:http://dx.doi.org/10.7554/eLife.14756.001 Melanoma is a form of skin cancer that is particularly difficult to treat. A new approach that has shown a lot of promise in treating many different cancers, including melanoma, is called “immunotherapy”. This technique harnesses the immune system – the body’s natural defences that help to protect against infections and disease – to combat cancer. One powerful type of immunotherapy involves injecting patients with cells called lymphocytes, which form part of the immune system. This is known as adoptive cell therapy and can activate the immune system to fight cancer, helping to shrink tumors. This treatment can be made even more powerful by combining it with a drug called cyclophosphamide and this combination, known as CTX-ACT, is currently one of the most efficient treatments for melanoma. Yet, little information is available to indicate why this treatment is so effective. Using mice implanted with melanoma cells, Qi, Li et al. sought to understand how CTX-ACT treatment works, with the goal of optimising it to increase its success. The results showed that a protective barrier of immune cells that suppresses the anti-tumor immune response – called an “immunosuppressive ring” – surrounds untreated tumors. CTX-ACT treatment can breakdown these rings, helping to reactivate the anti-tumor immune reaction in the tumors. This allows both the injected and mouse’s own immune cells to move into the tumor and destroy cancer cells. Qi, Li et al. used their findings to optimise treatment and succeeded in controlling tumor growth in the mice for several weeks. These new insights could be used to improve current immunotherapies, and offer new approaches for investigating the involvement of immune cells in the treatment of a wide range of different cancers. DOI:http://dx.doi.org/10.7554/eLife.14756.002
Collapse
Affiliation(s)
- Shuhong Qi
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Li
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, China
| | - Lisen Lu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, China
| | - Zhongyang Qi
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Liu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, China
| | - Lu Chen
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, China.,Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guanxin Shen
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ling Fu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, China
| | - Qingming Luo
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, China
| | - Zhihong Zhang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China.,MoE Key Laboratory for Biomedical Photonics, Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
34
|
Rini BI, Stenzl A, Zdrojowy R, Kogan M, Shkolnik M, Oudard S, Weikert S, Bracarda S, Crabb SJ, Bedke J, Ludwig J, Maurer D, Mendrzyk R, Wagner C, Mahr A, Fritsche J, Weinschenk T, Walter S, Kirner A, Singh-Jasuja H, Reinhardt C, Eisen T. IMA901, a multipeptide cancer vaccine, plus sunitinib versus sunitinib alone, as first-line therapy for advanced or metastatic renal cell carcinoma (IMPRINT): a multicentre, open-label, randomised, controlled, phase 3 trial. Lancet Oncol 2016; 17:1599-1611. [DOI: 10.1016/s1470-2045(16)30408-9] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 08/01/2016] [Accepted: 08/10/2016] [Indexed: 10/20/2022]
|
35
|
Constantino J, Gomes C, Falcão A, Cruz MT, Neves BM. Antitumor dendritic cell-based vaccines: lessons from 20 years of clinical trials and future perspectives. Transl Res 2016; 168:74-95. [PMID: 26297944 DOI: 10.1016/j.trsl.2015.07.008] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 07/25/2015] [Accepted: 07/28/2015] [Indexed: 02/06/2023]
Abstract
Dendritic cells (DCs) are versatile elements of the immune system and are best known for their unparalleled ability to initiate and modulate adaptive immune responses. During the past few decades, DCs have been the subject of numerous studies seeking new immunotherapeutic strategies against cancer. Despite the initial enthusiasm, disappointing results from early studies raised some doubts regarding the true clinical value of these approaches. However, our expanding knowledge of DC immunobiology and the definition of the optimal characteristics for antitumor immune responses have allowed a more rational development of DC-based immunotherapies in recent years. Here, after a brief overview of DC immunobiology, we sought to systematize the knowledge provided by 20 years of clinical trials, with a special emphasis on the diversity of approaches used to manipulate DCs and their consequent impact on vaccine effectiveness. We also address how new therapeutic concepts, namely the combination of DC vaccines with other anticancer therapies, are being implemented and are leveraging clinical outcomes. Finally, optimization strategies, new insights, and future perspectives on the field are also highlighted.
Collapse
Affiliation(s)
- João Constantino
- Faculty of Pharmacy and Centre for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Célia Gomes
- Faculty of Medicine, Laboratory of Pharmacology and Experimental Therapeutics, Institute for Biomedical Imaging and Life Sciences (IBILI) and Center of Investigation in Environment, Genetics and Oncobiology (CIMAGO), University of Coimbra, Coimbra, Portugal; CNC.IBILI, University of Coimbra, Coimbra, Portugal
| | - Amílcar Falcão
- Faculty of Pharmacy and Centre for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal; CNC.IBILI, University of Coimbra, Coimbra, Portugal
| | - Maria T Cruz
- Faculty of Pharmacy and Centre for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal; CNC.IBILI, University of Coimbra, Coimbra, Portugal
| | - Bruno M Neves
- Faculty of Pharmacy and Centre for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal; CNC.IBILI, University of Coimbra, Coimbra, Portugal; Department of Chemistry and QOPNA, Mass Spectrometry Centre, University of Aveiro, Aveiro, Portugal.
| |
Collapse
|
36
|
Jordan M, Waxman DJ. CpG-1826 immunotherapy potentiates chemotherapeutic and anti-tumor immune responses to metronomic cyclophosphamide in a preclinical glioma model. Cancer Lett 2015; 373:88-96. [PMID: 26655275 DOI: 10.1016/j.canlet.2015.11.029] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 11/26/2015] [Accepted: 11/27/2015] [Indexed: 02/08/2023]
Abstract
Cyclophosphamide administered on an intermittent metronomic schedule induces strong immune-dependent regression in several glioma models. Here we investigate whether this immunogenic chemotherapy can be potentiated by combination with the immune stimulatory TLR9 agonist CpG-1826. CpG-1826 treatment of GL261 gliomas implanted in immune competent mice induced tumor growth delay associated with increased tumor recruitment of macrophages and B cells. Anti-tumor responses varied between individuals, with CpG-1826 inducing robust tumor growth delay in ~50% of treated mice. Both high and low CpG-1826-responsive mice showed striking improvements when CpG-1826 was combined with cyclophosphamide treatment. Tumor-associated macrophages, B cells, dendritic cells, and cytotoxic T cells were increased, T regulatory cells were not induced, and long-term GL261 glioma regression with immune memory was achieved when CpG-1826 was combined with either single cyclophosphamide dosing (90 mg/kg) or metronomic cyclophosphamide treatment (two cycles at 45 mg/kg, spaced 12-days apart). B16F10 melanoma, a low immunogenic tumor model, also showed enhanced immune and anti-tumor responses to cyclophosphamide/CpG-1826 chemoimmunotherapy, but unlike GL261 tumors, did not regress. TLR9-based immunotherapy can thus be effectively combined with immunogenic cyclophosphamide treatment to enhance immune-based anti-tumor responses, even in poorly immunogenic cancer models.
Collapse
Affiliation(s)
- Marie Jordan
- Division of Cell and Molecular Biology, Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA
| | - David J Waxman
- Division of Cell and Molecular Biology, Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA.
| |
Collapse
|
37
|
Pruneri G, Vingiani A, Bagnardi V, Rotmensz N, De Rose A, Palazzo A, Colleoni AM, Goldhirsch A, Viale G. Clinical validity of tumor-infiltrating lymphocytes analysis in patients with triple-negative breast cancer. Ann Oncol 2015; 27:249-56. [PMID: 26598540 DOI: 10.1093/annonc/mdv571] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 11/11/2015] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Although tumor-infiltrating lymphocytes (TILs) have been associated with a favorable prognosis in triple-negative breast cancer (TNBC) patients, this marker is not currently considered robust enough for entering the clinical practice. In the present study, we assessed the clinical validity of the guidelines recently issued by the International TIL Working Group in a large retrospective series of well-annotated TNBC patients. PATIENTS AND METHODS TILs were evaluated in all the full-face H&E sections from 897 consecutive TNBC (i.e. tumors with <1% of ER and PgR immunoreactivity and absence of HER2 overexpression or amplification) patients diagnosed and treated at the European Institute of Oncology between 1995 and 2010 (median follow-up 8.2 years, range 6 months to 18 years). All mononuclear cells were evaluated in the stromal area within the borders of the invasive tumor, reported as a percentage value and treated as a continuous variable in survival analysis. RESULTS The median percentage of TILs was 20%, and 21.9% of the cases had ≥50% (lymphocyte predominant breast cancer, LPBC) TILs. At univariable survival analysis, TILs were a significant predictor of better disease-free survival (DFS), distant disease-free survival (DDFS) and overall survival (OS) (P < 0.0001). Multivariable analysis confirmed that each 10% increase in TILs strongly predicted better survival, independent of patients' age, lymph node status, tumor size, histological grade, peritumoral vascular invasion and Ki-67 labeling index. Patients with LPBC had a 10-year survival rate of 71%, 84% and 96% for DFS, DDFS and OS, respectively. Stratified analysis revealed a positive correlation between TILs and OS across all the subgroups analyzed. CONCLUSION Our data support the analytical validity of the recently issued TILs evaluation guidelines in the clinical practice.
Collapse
Affiliation(s)
- G Pruneri
- Department of Pathology, Biobank for Translational Medicine Unit University of Milan, School of Medicine, Milan
| | - A Vingiani
- Division of Pathology University of Milan, School of Medicine, Milan
| | - V Bagnardi
- Division of Epidemiology and Biostatistics Department of Statistics and Quantitative Methods, University of Milan-Bicocca, Milan, Italy
| | - N Rotmensz
- Division of Epidemiology and Biostatistics
| | | | | | | | - A Goldhirsch
- Division of Program of Breast Health (Senology), European Institute of Oncology, Milan International Breast Cancer Study Group, Bern, Switzerland
| | - G Viale
- Division of Pathology University of Milan, School of Medicine, Milan
| |
Collapse
|
38
|
Kirner A, Mayer-Mokler A, Reinhardt C. IMA901: a multi-peptide cancer vaccine for treatment of renal cell cancer. Hum Vaccin Immunother 2015; 10:3179-89. [PMID: 25625928 DOI: 10.4161/21645515.2014.983857] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Despite a major improvement in the treatment of advanced kidney cancer by the recent introduction of targeted agents such as multi-kinase inhibitors, long-term benefits are still limited and a significant unmet medical need remains for this disease. Cancer immunotherapy has shown its potential by the induction of long-lasting responses in a small subset of patients, however, the unspecific immune interventions with (high dose) cytokines used so far are associated with significant side effects. Specific cancer immunotherapy may circumvent these problems by attacking tumor cells while sparing normal tissue with the use of multi-peptide vaccination being one of the most promising strategies. We here summarize the clinical and translational data from phase I and II trials investigating IMA901. Significant associations of clinical benefit with detectable T cell responses against the IMA901 peptides and encouraging survival data in treated patients has prompted the start of a randomized, controlled phase III trial in 1st line advanced RCC with survival results expected toward the end of 2015. Potential combination strategies with the recently discovered so-called checkpoint inhibitors are also discussed.
Collapse
Key Words
- 5-FU, 5 fluorouracil
- AE, Adverse event
- CTL, Cytotoxic T-lymphocyte
- CY, Cyclophosphamide
- Cancer vaccine
- DC, Dendritic cell
- DCR, Disease control rate
- ECG, Electrocardiogram
- ELISpot, Enzyme-linked immunospot assay
- FDA, Food and Drug Administration
- GM-CSF
- HBV, Hepatitis B virus
- HLA, Human leukocyte antigen
- IFN, Interferon
- IL, Interleukin
- IMA901
- MDSC, Myeloid-derived suppressor cells
- MHC, Major histocompatibility complex
- MSKCC, Memorial Sloan Kettering Cancer Center
- NCI-CTC, National Cancer Institute-Common Toxicity Criteria
- OS, Overall survival
- PD, Progressive disease
- PFS, Progression-free survival
- PK, Pharmacokinetic
- PR, Partial response
- RCC, Renal cell carcinoma
- RECIST, Response Evaluation Criteria in Solid Tumors
- SAE, Serious adverse event
- SD, Stable disease
- TKI, Tyrosine-kinase inhibitors
- TNF, Tumor necrosis factor
- TUMAP, Tumor-associated peptides
- Tregs, Regulatory T-cells
- VEGF, Vascular endothelial growth factor
- ccRCC, Clear cell renal cell carcinoma
- checkpoint inhibitor
- cyclophosphamide
- i.d., intradermal
- immunotherapy
- intradermally
- kidney cancer
- mRNA, Messenger ribonucleic acid
- mTOR, Mammalian target of rapamycin
- mg, Milligram
- n, Number
- renal cell carcinoma
- s.c., subcutaneous, subcutaneously
- tumor-associated peptides
- vaccination
- μg, Microgram
Collapse
|
39
|
Apoptosis of tumor infiltrating effector TIM-3+CD8+ T cells in colon cancer. Sci Rep 2015; 5:15659. [PMID: 26493689 PMCID: PMC4616166 DOI: 10.1038/srep15659] [Citation(s) in RCA: 135] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2015] [Accepted: 09/29/2015] [Indexed: 12/28/2022] Open
Abstract
TIM-3 functions to enforce CD8+ T cell exhaustion, a dysfunctional state associated with the tolerization of tumor microenvironment. Here we report apoptosis of IFN-γ competent TIM-3+ population of tumor-infiltrating CD8+ T cells in colon cancer. In humans suffering from colorectal cancer, TIM-3+ population is higher in cancer tissue-resident relative to peripheral blood CD8+ T cells. Both the TIM-3+ and TIM-3- cancer tissue-resident CD8+ T cells secrete IFN-γ of comparable levels, although apoptotic cells are more in TIM-3+ compared to TIM-3- population. In mouse CT26 colon tumor model, majority of tumor-infiltrating CD8+ T cells express TIM-3 and execute cytolysis function with higher effector cytokine secretion and apoptosis in TIM-3+ compared to TIM-3- population. The tumor cells secrete galectin-9, which increases apoptosis of tumor-infiltrating CD8+ T cells. Galectin-9/TIM-3 signaling blockade with anti-TIM-3 antibody reduces the apoptosis and in addition, inhibits tumor growth in mice. The blockade increases therapeutic efficacy of cyclophosphamide to treat tumor in mice as well. These results reveal a previously unexplored role of TIM-3 on tumor-infiltrating CD8+ T cells in vivo.
Collapse
|
40
|
Karpanen T, Olweus J. T-cell receptor gene therapy--ready to go viral? Mol Oncol 2015; 9:2019-42. [PMID: 26548533 DOI: 10.1016/j.molonc.2015.10.006] [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] [Received: 09/04/2015] [Revised: 10/08/2015] [Accepted: 10/09/2015] [Indexed: 12/16/2022] Open
Abstract
T lymphocytes can be redirected to recognize a tumor target and harnessed to combat cancer by genetic introduction of T-cell receptors of a defined specificity. This approach has recently mediated encouraging clinical responses in patients with cancers previously regarded as incurable. However, despite the great promise, T-cell receptor gene therapy still faces a multitude of obstacles. Identification of epitopes that enable effective targeting of all the cells in a heterogeneous tumor while sparing normal tissues remains perhaps the most demanding challenge. Experience from clinical trials has revealed the dangers associated with T-cell receptor gene therapy and highlighted the need for reliable preclinical methods to identify potentially hazardous recognition of both intended and unintended epitopes in healthy tissues. Procedures for manufacturing large and highly potent T-cell populations can be optimized to enhance their antitumor efficacy. Here, we review the current knowledge gained from preclinical models and clinical trials using adoptive transfer of T-cell receptor-engineered T lymphocytes, discuss the major challenges involved and highlight potential strategies to increase the safety and efficacy to make T-cell receptor gene therapy a standard-of-care for large patient groups.
Collapse
Affiliation(s)
- Terhi Karpanen
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet and K.G. Jebsen Center for Cancer Immunotherapy, University of Oslo, Ullernchausseen 70, N-0379 Oslo, Norway.
| | - Johanna Olweus
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet and K.G. Jebsen Center for Cancer Immunotherapy, University of Oslo, Ullernchausseen 70, N-0379 Oslo, Norway.
| |
Collapse
|
41
|
Prospective study of cyclophosphamide, thiotepa, carboplatin combined with adoptive DC-CIK followed by metronomic cyclophosphamide therapy as salvage treatment for triple negative metastatic breast cancers patients (aged <45). Clin Transl Oncol 2015; 18:82-7. [DOI: 10.1007/s12094-015-1339-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Accepted: 06/26/2015] [Indexed: 12/26/2022]
|
42
|
|
43
|
Gershan JA, Barr KM, Weber JJ, Jing W, Johnson BD. Immune modulating effects of cyclophosphamide and treatment with tumor lysate/CpG synergize to eliminate murine neuroblastoma. J Immunother Cancer 2015; 3:24. [PMID: 26082836 PMCID: PMC4469315 DOI: 10.1186/s40425-015-0071-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 05/15/2015] [Indexed: 12/22/2022] Open
Abstract
Background Neuroblastoma is a pediatric cancer of neural crest origin. Despite aggressive treatment, mortality remains at 40 % for patients with high-risk disseminated disease, underscoring the need to test new combinations of therapies. In murine tumor models, our laboratory previously showed that T cell-mediated anti-tumor immune responses improve in the context of lymphopenia. The goal of this study was to incorporate lymphodepletion into an effective immune therapy that can be easily translated into neuroblastoma standard of care. Based on the lymphodepleting effects of cyclophosphamide, we hypothesized that cyclophosphamide would synergize with the TLR9 agonist, CpG oligodeoxynucleotide (ODN), to produce a T cell-mediated anti-neuroblastoma effect. Methods To test this hypothesis, we used the AgN2a aggressive murine model of neuroblastoma. Mice bearing subcutaneous tumors were treated with cyclophosphamide followed by treatment with tumor cell lysate mixed with CpG ODN injected at the tumor site. Results Subcutaneous neuroblastoma regressed only in mice that were treated with 100 mg/kg cyclophosphamide prior to receiving treatments of tumor lysate mixed with CpG ODN. The anti-neuroblastoma response was T cell-mediated. Synergy between cyclophosphamide and the tumor lysate/CpG ODN treatment influenced the production of anti-tumor CD8 T cell effectors, and dendritic cell homeostasis. For clinical consideration, an allogeneic tumor lysate was used effectively with this protocol to eliminate AgN2a tumor in vivo. Conclusion Synergistic immune modulating effects of cyclophosphamide and a treatment containing tumor cell lysate and CpG ODN provide T cell-mediated anti-tumor activity against murine neuroblastoma. Electronic supplementary material The online version of this article (doi:10.1186/s40425-015-0071-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jill A Gershan
- Division of Hematology/Oncology/Transplant, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226 USA
| | | | - James J Weber
- Division of Hematology/Oncology/Transplant, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226 USA
| | - Weiqing Jing
- Division of Hematology/Oncology/Transplant, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226 USA
| | - Bryon D Johnson
- Division of Hematology/Oncology/Transplant, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226 USA
| |
Collapse
|
44
|
Bowers JS, Nelson MH, Kundimi S, Bailey SR, Huff LW, Schwartz KM, Cole DJ, Rubinstein MP, Paulos CM. Dendritic Cells in Irradiated Mice Trigger the Functional Plasticity and Antitumor Activity of Adoptively Transferred Tc17 Cells via IL12 Signaling. Clin Cancer Res 2015; 21:2546-57. [PMID: 25904754 DOI: 10.1158/1078-0432.ccr-14-2294] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 02/09/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE The adoptive cell transfer (ACT) of CD8(+) T cells is a promising treatment for advanced malignancies. Lymphodepletion before ACT enhances IFNγ(+)CD8(+) T cell (Tc0)-mediated tumor regression. Yet, how lymphodepletion regulates the function and antitumor activity of IL17A(+)CD8(+) T cells (Tc17) is unknown. EXPERIMENTAL DESIGN To address this question, pmel-1 CD8(+) T cells were polarized to secrete either IL17A or IFNγ. These subsets were then infused into mice with B16F10 melanoma that were lymphoreplete [no total body irradiation (TBI)], or lymphodepleted with nonmyeloablative (5 Gy) or myeloablative (9 Gy with hematopoietic stem cell transplantation) TBI. The activation of innate immune cells and function of donor T-cell subsets were monitored in recipient mice. RESULTS Tc17 cells regress melanoma in myeloablated mice to a greater extent than in lymphoreplete or nonmyeloablated mice. TBI induced functional plasticity in Tc17 cells, causing conversion from IL17A to IFNγ producers. Additional investigation revealed that Tc17 plasticity and antitumor activity were mediated by IL12 secreted by irradiated host dendritic cells (DC). Neutralization of endogenous IL12 reduced the antitumor activity of Tc17 cells in myeloablated mice, whereas ex vivo priming with IL12 enhanced their capacity to regress melanoma in nonmyeloablated animals. This, coupled with exogenous administration of low-dose IL12, obviated the need for host preconditioning, creating curative responses in nonirradiated mice. CONCLUSIONS Our findings indicate that TBI-induced IL12 augments Tc17 cell-mediated tumor immunity and underline the substantial implications of in vitro preparation of antitumor Tc17 cells with IL12 in the design of T-cell immunotherapies.
Collapse
Affiliation(s)
- Jacob S Bowers
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina. Department of Surgery, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina.
| | - Michelle H Nelson
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina. Department of Surgery, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Sreenath Kundimi
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina. Department of Surgery, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Stefanie R Bailey
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina. Department of Surgery, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Logan W Huff
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina. Department of Surgery, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Kristina M Schwartz
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina. Department of Surgery, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - David J Cole
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina. Department of Surgery, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Mark P Rubinstein
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina. Department of Surgery, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Chrystal M Paulos
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina. Department of Surgery, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina.
| |
Collapse
|
45
|
Gaya A, Akle CA, Mudan S, Grange J. The Concept of Hormesis in Cancer Therapy - Is Less More? Cureus 2015; 7:e261. [PMID: 26180685 PMCID: PMC4494563 DOI: 10.7759/cureus.261] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2015] [Indexed: 12/21/2022] Open
Abstract
There has, in recent years, been a paradigm shift in our understanding of the role of the immune system in the development of cancers. Immune dysregulation, manifesting as chronic inflammation, not only facilitates the growth and spread of tumors but prevents the host from mounting effective immune defenses against it. Many attempts are being made to develop novel immunotherapeutic strategies, but there is growing evidence that a radical reevaluation of the mode of action of chemotherapeutic agents and ionizing radiation is required in the light of advances in immunology. Based on the concept of hormesis – defined as the presence of different modes of action of therapeutic modalities at different doses – a ‘repositioning’ of chemotherapy and radiotherapy may be required in all aspects of cancer management. In the case of chemotherapy, this may involve a change from the maximum tolerated dose concept to low dose intermittent (‘metronomic’) therapy, whilst in radiation therapy, highly accurate stereotactic targeting enables ablative, antigen-releasing (immunogenic) doses of radiation to be delivered to the tumor with sparing of surrounding normal tissues. Coupled with emerging immunotherapeutic procedures, the future of cancer treatment may well lie in repositioned chemotherapy, radiotherapy, and more localized debulking surgery.
Collapse
Affiliation(s)
- Andy Gaya
- London Oncology Clinic, Guy's and St Thomas' NHS Foundation Trust
| | | | | | | |
Collapse
|
46
|
Persa E, Balogh A, Sáfrány G, Lumniczky K. The effect of ionizing radiation on regulatory T cells in health and disease. Cancer Lett 2015; 368:252-61. [PMID: 25754816 DOI: 10.1016/j.canlet.2015.03.003] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 03/02/2015] [Accepted: 03/03/2015] [Indexed: 02/07/2023]
Abstract
Treg cells are key elements of the immune system which are responsible for the immune suppressive phenotype of cancer patients. Interaction of Treg cells with conventional anticancer therapies might fundamentally influence cancer therapy response rates. Radiotherapy, apart from its direct tumor cell killing potential, has a contradictory effect on the antitumor immune response: it augments certain immune parameters, while it depresses others. Treg cells are intrinsically radioresistant due to reduced apoptosis and increased proliferation, which leads to their systemic and/or intratumoral enrichment. While physiologically Treg suppression is not enhanced by irradiation, this is not the case in a tumorous environment, where Tregs acquire a highly suppressive phenotype, which is further increased by radiotherapy. This is the reason why the interest for combined radiotherapy and immunotherapy approaches focusing on the abrogation of Treg suppression has increased in cancer therapy in the last few years. Here we summarize the basic mechanisms of Treg radiation response both in healthy and cancerous environments and discuss Treg-targeted pre-clinical and clinical immunotherapy approaches used in combination with radiotherapy. Finally, the discrepant findings regarding the predictive value of Tregs in therapy response are also reviewed.
Collapse
Affiliation(s)
- Eszter Persa
- Frédéric Joliot-Curie National Research Institute for Radiobiology and Radiohygiene, Budapest, Hungary
| | - Andrea Balogh
- Frédéric Joliot-Curie National Research Institute for Radiobiology and Radiohygiene, Budapest, Hungary
| | - Géza Sáfrány
- Frédéric Joliot-Curie National Research Institute for Radiobiology and Radiohygiene, Budapest, Hungary
| | - Katalin Lumniczky
- Frédéric Joliot-Curie National Research Institute for Radiobiology and Radiohygiene, Budapest, Hungary.
| |
Collapse
|
47
|
Soares KC, Rucki AA, Wu AA, Olino K, Xiao Q, Chai Y, Wamwea A, Bigelow E, Lutz E, Liu L, Yao S, Anders RA, Laheru D, Wolfgang CL, Edil BH, Schulick RD, Jaffee EM, Zheng L. PD-1/PD-L1 blockade together with vaccine therapy facilitates effector T-cell infiltration into pancreatic tumors. J Immunother 2015; 38:1-11. [PMID: 25415283 PMCID: PMC4258151 DOI: 10.1097/cji.0000000000000062] [Citation(s) in RCA: 318] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDA) has a poor prognosis due to late detection and resistance to conventional therapies. Published studies show that the PDA tumor microenvironment is predominantly infiltrated with immune suppressive cells and signals that if altered, would allow effective immunotherapy. However, single-agent checkpoint inhibitors including agents that alter immune suppressive signals in other human cancers such as cytotoxic T-lymphocyte antigen 4 (CTLA-4), programmed death 1 (PD-1), and its ligand PD-L1, have failed to demonstrate objective responses when given as single agents to PDA patients. We recently reported that inhibition of the CTLA-4 pathway when given together with a T cell inducing vaccine gives objective responses in metastatic PDA patients. In this study, we evaluated blockade of the PD-1/PD-L1 pathway. We found that PD-L1 is weakly expressed at a low frequency in untreated human and murine PDAs but treatment with a granulocyte macrophage colony-stimulating factor secreting PDA vaccine (GVAX) significantly upregulates PD-L1 membranous expression after treatment of tumor-bearing mice. In addition, combination therapy with vaccine and PD-1 antibody blockade improved murine survival compared with PD-1 antibody monotherapy or GVAX therapy alone. Furthermore, PD-1 blockade increased effector CD8 T lymphocytes and tumor-specific interferon-γ production of CD8 T cells in the tumor microenvironment. Immunosuppressive pathways, including regulatory T cells and CTLA-4 expression on T cells were overcome by the addition of vaccine and low-dose cyclophosphamide to PD-1 blockade. Collectively, our study supports combining PD-1 or PD-L1 antibody therapy with a T cell inducing agent for PDA treatment.
Collapse
Affiliation(s)
- Kevin C. Soares
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
- The Sidney Kimmel Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- the Skip Viragh Center for Pancreatic Cancer Research and Clinical Care, Johns Hopkins University School of Medicine, Baltimore, MD
- the Sol Goldman Pancreatic Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Agnieszka A. Rucki
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
- The Sidney Kimmel Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Annie A. Wu
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
- The Sidney Kimmel Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Kelly Olino
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
- The Sidney Kimmel Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- the Sol Goldman Pancreatic Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Qian Xiao
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
- The Sidney Kimmel Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Yi Chai
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
- The Sidney Kimmel Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Anthony Wamwea
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
- The Sidney Kimmel Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Elaine Bigelow
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
- The Sidney Kimmel Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Eric Lutz
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
- The Sidney Kimmel Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- the Skip Viragh Center for Pancreatic Cancer Research and Clinical Care, Johns Hopkins University School of Medicine, Baltimore, MD
- the Sol Goldman Pancreatic Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | | | | | - Robert A. Anders
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
- The Sidney Kimmel Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- the Sol Goldman Pancreatic Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Daniel Laheru
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
- The Sidney Kimmel Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- the Skip Viragh Center for Pancreatic Cancer Research and Clinical Care, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Christopher L. Wolfgang
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
- The Sidney Kimmel Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- the Sol Goldman Pancreatic Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Barish H. Edil
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
- The Sidney Kimmel Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- the Sol Goldman Pancreatic Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Surgery, University of Colorado School of Medicine, Aurora, Colorado
| | - Richard D. Schulick
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
- The Sidney Kimmel Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Surgery, University of Colorado School of Medicine, Aurora, Colorado
| | - Elizabeth M. Jaffee
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
- The Sidney Kimmel Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- the Skip Viragh Center for Pancreatic Cancer Research and Clinical Care, Johns Hopkins University School of Medicine, Baltimore, MD
- the Sol Goldman Pancreatic Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Lei Zheng
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
- The Sidney Kimmel Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- the Skip Viragh Center for Pancreatic Cancer Research and Clinical Care, Johns Hopkins University School of Medicine, Baltimore, MD
- the Sol Goldman Pancreatic Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
48
|
Weir GM, Hrytsenko O, Stanford MM, Berinstein NL, Karkada M, Liwski RS, Mansour M. Metronomic cyclophosphamide enhances HPV16E7 peptide vaccine induced antigen-specific and cytotoxic T-cell mediated antitumor immune response. Oncoimmunology 2014; 3:e953407. [PMID: 25960932 PMCID: PMC4368141 DOI: 10.4161/21624011.2014.953407] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2014] [Accepted: 06/24/2014] [Indexed: 12/22/2022] Open
Abstract
In clinical trials, metronomic cyclophosphamide (CPA) is increasingly being combined with vaccines to reduce tumor-induced immune suppression. Previous strategies to modulate the immune system during vaccination have involved continuous administration of low dose chemotherapy, studies that have posed unique considerations for clinical trial design. Here, we evaluated metronomic CPA in combination with a peptide vaccine targeting HPV16E7 in an HPV16-induced tumor model, focusing on the cytotoxic T-cell response and timing of low dose metronomic CPA (mCPA) treatment relative to vaccination. Mice bearing C3 tumors were given metronomic CPA on alternating weeks in combination with immunization with a DepoVax vaccine containing HPV16E749-57 peptide antigen every 3 weeks. Only the combination therapy provided significant long-term control of tumor growth. The efficacy of the vaccine was uncompromised if given at the beginning or end of a cycle of metronomic CPA. Metronomic CPA had a pronounced lymphodepletive effect on the vaccine draining lymph node, yet did not reduce the development of antigen-specific CD8+ T cells induced by vaccination. This enrichment correlated with increased cytotoxic activity in the spleen and increased expression of cytotoxic gene signatures in the tumor. Immunity could be passively transferred through CD8+ T cells isolated from tumor-bearing mice treated with the combinatorial treatment regimen. A comprehensive survey of splenocytes indicated that metronomic CPA, in the absence of vaccination, induced transient lymphodepletion marked by a selective expansion of myeloid-derived suppressor cells. These results provide important insights into the multiple mechanisms of metronomic CPA induced immune modulation in the context of a peptide cancer vaccine that may be translated into more effective clinical trial designs.
Collapse
Key Words
- CPA, cyclophosphamide
- CTL, cytotoxic T lymphocyte
- CTLA-4, cytotoxic T lymphocyte-associated protein 4
- DPX, DepoVax
- HPV, human papilloma virus
- HPV16
- IFNγ, interferon γ
- MDSC, myeloid-derived suppressor cells
- PD-1/PDCD1, programmed cell death 1
- PO, per os (oral)
- Treg, regulatory T cell
- cancer
- checkpoint inhibitors
- mCPA, metronomic low dose CPA
- metronomic cyclophosphamide
- sbCPA, single bolus low dose CPA
- translational
- vaccine
Collapse
Affiliation(s)
- Genevieve M Weir
- Immunovaccine Inc. ; Halifax; Nova Scotia, Canada ; Department of Microbiology & Immunology; Dalhousie University ; Halifax; Nova Scotia, Canada
| | - Olga Hrytsenko
- Immunovaccine Inc. ; Halifax; Nova Scotia, Canada ; Department of Biology; Dalhousie University ; Halifax; Nova Scotia, Cananda
| | - Marianne M Stanford
- Immunovaccine Inc. ; Halifax; Nova Scotia, Canada ; Department of Microbiology & Immunology; Dalhousie University ; Halifax; Nova Scotia, Canada
| | | | - Mohan Karkada
- Immunovaccine Inc. ; Halifax; Nova Scotia, Canada ; Department of Microbiology & Immunology; Dalhousie University ; Halifax; Nova Scotia, Canada
| | - Robert S Liwski
- Department of Microbiology & Immunology; Dalhousie University ; Halifax; Nova Scotia, Canada ; Division of Hematopathology; Queen Elizabeth II Health Sciences Centre ; Nova Scotia, Canada
| | - Marc Mansour
- Immunovaccine Inc. ; Halifax; Nova Scotia, Canada
| |
Collapse
|
49
|
Anguille S, Smits EL, Lion E, van Tendeloo VF, Berneman ZN. Clinical use of dendritic cells for cancer therapy. Lancet Oncol 2014; 15:e257-67. [PMID: 24872109 DOI: 10.1016/s1470-2045(13)70585-0] [Citation(s) in RCA: 537] [Impact Index Per Article: 48.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Since the mid-1990s, dendritic cells have been used in clinical trials as cellular mediators for therapeutic vaccination of patients with cancer. Dendritic cell-based immunotherapy is safe and can induce antitumour immunity, even in patients with advanced disease. However, clinical responses have been disappointing, with classic objective tumour response rates rarely exceeding 15%. Paradoxically, findings from emerging research indicate that dendritic cell-based vaccination might improve survival, advocating implementation of alternative endpoints to assess the true clinical potency of dendritic cell-based vaccination. We review the clinical effectiveness of dendritic cell-based vaccine therapy in melanoma, prostate cancer, malignant glioma, and renal cell carcinoma, and summarise the most important lessons from almost two decades of clinical studies of dendritic cell-based immunotherapy in these malignant disorders. We also address how the specialty is evolving, and which new therapeutic concepts are being translated into clinical trials to leverage the clinical effectiveness of dendritic cell-based cancer immunotherapy. Specifically, we discuss two main trends: the implementation of the next-generation dendritic cell vaccines that have improved immunogenicity, and the emerging paradigm of combination of dendritic cell vaccination with other cancer therapies.
Collapse
Affiliation(s)
- Sébastien Anguille
- Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium; Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium.
| | - Evelien L Smits
- Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium; Center for Oncological Research, University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| | - Eva Lion
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| | - Viggo F van Tendeloo
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| | - Zwi N Berneman
- Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium; Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| |
Collapse
|
50
|
Wu J, Waxman DJ. Metronomic cyclophosphamide schedule-dependence of innate immune cell recruitment and tumor regression in an implanted glioma model. Cancer Lett 2014; 353:272-80. [PMID: 25069038 DOI: 10.1016/j.canlet.2014.07.033] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 07/17/2014] [Accepted: 07/19/2014] [Indexed: 01/13/2023]
Abstract
Metronomic cyclophosphamide (CPA) treatment activates robust innate anti-tumor immunity and induces major regression of large, implanted brain tumor xenografts when administered on an intermittent, every 6-day schedule, but not on a daily low-dose or a maximum-tolerated dose CPA schedule. Here, we used an implanted GL261 glioma model to compare five intermittent metronomic CPA schedules to elucidate the kinetics and schedule dependence of innate immune cell recruitment and tumor regression. Tumor-recruited natural killer cells induced by two every 6-day treatment cycles were significantly ablated 1 day after a third CPA treatment, but largely recovered several days later. Natural killer and other tumor-infiltrating innate immune cells peaked 12 days after the last CPA treatment on the every 6-day schedule, suggesting that drug-free intervals longer than 6 days may show increased efficacy. Metronomic CPA treatments spaced 9 or 12 days apart, or on an alternating 6 and 9 day schedule, induced extensive tumor regression, similar to the 6-day schedule; however, the tumor-infiltrating natural killer cell responses were not sustained, leading to rapid resumption of tumor growth after day 24, despite ongoing metronomic CPA treatment. Increasing the CPA dose prolonged the period of tumor regression on the every 9-day schedule, but natural killer cell activation was markedly decreased. Thus, while several intermittent metronomic CPA treatment schedules can activate innate immune cell recruitment leading to major tumor regression, sustained immune and anti-tumor responses are only achieved on the 6-day schedule. However, even with this schedule, some tumors eventually relapse, indicating a need for further improvements in this immunogenic metronomic therapy.
Collapse
Affiliation(s)
- Junjie Wu
- Division of Cell and Molecular Biology, Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA
| | - David J Waxman
- Division of Cell and Molecular Biology, Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA.
| |
Collapse
|