51
|
Broz ML, Krummel MF. The emerging understanding of myeloid cells as partners and targets in tumor rejection. Cancer Immunol Res 2016; 3:313-9. [PMID: 25847968 DOI: 10.1158/2326-6066.cir-15-0041] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Myeloid cells are the most prominent among cells capable of presenting tumor-derived antigens to T cells and thereby maintaining the latter in an activated state. Myeloid populations of the tumor microenvironment prominently include monocytes and neutrophils (sometimes loosely grouped as myeloid-derived suppressor cells), macrophages, and dendritic cells. Although intratumoral myeloid populations, as a whole, have long been considered nonstimulatory or suppressive, it has only recently been appreciated that not all tumor-infiltrating myeloid cells are made equal. Because of advances in high-dimensional flow cytometry as well as more robust transcriptional profiling, we now also understand that the subsets of the tumor-myeloid compartment are far more diverse and notably even contain a rare population of stimulatory dendritic cells. As all of these myeloid populations represent major T-cell-interacting partners for incoming tumor-reactive cytotoxic T lymphocytes, understanding the distinctions in their lineage and function reveals and guides numerous therapeutic avenues targeting these antigen-presenting cells. In this Cancer Immunology at the Crossroads overview, we review the recent progress in this rapidly evolving field and advance the hypothesis that the antigen-presenting compartment within tumor microenvironments may contain significant numbers of potent allies to be leveraged for immune-based tumor clearance.
Collapse
Affiliation(s)
- Miranda L Broz
- Department of Pathology, University of California at San Francisco, San Francisco, California
| | - Matthew F Krummel
- Department of Pathology, University of California at San Francisco, San Francisco, California.
| |
Collapse
|
52
|
Bougherara H, Mansuet-Lupo A, Alifano M, Ngô C, Damotte D, Le Frère-Belda MA, Donnadieu E, Peranzoni E. Real-Time Imaging of Resident T Cells in Human Lung and Ovarian Carcinomas Reveals How Different Tumor Microenvironments Control T Lymphocyte Migration. Front Immunol 2015; 6:500. [PMID: 26528284 PMCID: PMC4600956 DOI: 10.3389/fimmu.2015.00500] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 09/14/2015] [Indexed: 12/30/2022] Open
Abstract
T cells play a key role in the battle against cancer. To perform their antitumor activities, T cells need to adequately respond to tumor antigens by establishing contacts with either malignant cells or antigen-presenting cells. These latter functions rely on a series of migratory steps that go from entry of T cells into the tumor followed by their locomotion in the tumor stroma. Our knowledge of how T cells migrate within tumors mainly comes from experiments performed in mouse models. Whereas such systems have greatly advanced our understanding, they do not always faithfully recapitulate the disease observed in cancer patients. We previously described a technique based on tissue slices that enables to track with real-time imaging microscopy the motile behavior of fluorescent T cells plated onto fresh sections of human lung tumors. We have now refined this approach to monitor the locomotion of resident tumor-infiltrating CD8 T cells labeled with fluorescently coupled antibodies. Using this approach, our findings reveal that CD8 T cells accumulate in the stroma of ovarian and lung carcinomas but move slowly in this compartment. Conversely, even though less populated, tumors islets were found to be zones of faster migration for resident CD8 T cells. We also confirm the key role played by collagen fibers, which, by their orientation, spacing and density, control the distribution and migration of resident CD8 T cells within the tumor stroma. We have subsequently demonstrated that, under some physical tissue constraints, CD8 T cells exhibited a mode of migration characterized by alternate forward and backward movements. In sum, using an ex vivo assay to track CD8 T cells in fresh human tumor tissues, we have identified the extracellular matrix as a major stromal component in influencing T cell migration, thereby impacting the control of tumor growth. This approach will aid in the development and testing of novel immunotherapy strategies to promote T cell migration in tumors.
Collapse
Affiliation(s)
- Houcine Bougherara
- INSERM U1016, Institut Cochin , Paris , France ; CNRS, UMR8104 , Paris , France ; Université Paris Descartes, Sorbonne Paris Cité , Paris , France
| | - Audrey Mansuet-Lupo
- Université Paris Descartes, Sorbonne Paris Cité , Paris , France ; Department of Pathology, Paris Centre University Hospitals, Assistance Publique-Hôpitaux de Paris , Paris , France ; Cancer and Immune Escape, Cordeliers Research Center, INSERM U1138 , Paris , France ; University Pierre and Marie Curie , Paris , France
| | - Marco Alifano
- Université Paris Descartes, Sorbonne Paris Cité , Paris , France ; Department of Thoracic Surgery, Paris Centre University Hospitals, Assistance Publique-Hôpitaux de Paris , Paris , France
| | - Charlotte Ngô
- Department of Gynaecological and Oncological Surgery, Hôpital Européen Georges Pompidou, Université Paris Descartes, Assistance Publique-Hôpitaux de Paris , Paris , France
| | - Diane Damotte
- Université Paris Descartes, Sorbonne Paris Cité , Paris , France ; Department of Pathology, Paris Centre University Hospitals, Assistance Publique-Hôpitaux de Paris , Paris , France ; Cancer and Immune Escape, Cordeliers Research Center, INSERM U1138 , Paris , France ; University Pierre and Marie Curie , Paris , France
| | - Marie-Aude Le Frère-Belda
- Department of Pathology, Hôpital Européen Georges Pompidou, Université Paris Descartes, Assistance Publique-Hôpitaux de Paris , Paris , France
| | - Emmanuel Donnadieu
- INSERM U1016, Institut Cochin , Paris , France ; CNRS, UMR8104 , Paris , France ; Université Paris Descartes, Sorbonne Paris Cité , Paris , France
| | - Elisa Peranzoni
- INSERM U1016, Institut Cochin , Paris , France ; CNRS, UMR8104 , Paris , France ; Université Paris Descartes, Sorbonne Paris Cité , Paris , France
| |
Collapse
|
53
|
Maeda A, Kulbatski I, DaCosta RS. Emerging Applications for Optically Enabled Intravital Microscopic Imaging in Radiobiology. Mol Imaging 2015. [DOI: 10.2310/7290.2015.00022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Azusa Maeda
- From the Princess Margaret Cancer Centre, University Health Network, MaRS Centre; Techna Institute for Advancement of Technologies for Health; and Department of Medical Biophysics, University of Toronto, MaRS Centre, Toronto, ON
| | - Iris Kulbatski
- From the Princess Margaret Cancer Centre, University Health Network, MaRS Centre; Techna Institute for Advancement of Technologies for Health; and Department of Medical Biophysics, University of Toronto, MaRS Centre, Toronto, ON
| | - Ralph S. DaCosta
- From the Princess Margaret Cancer Centre, University Health Network, MaRS Centre; Techna Institute for Advancement of Technologies for Health; and Department of Medical Biophysics, University of Toronto, MaRS Centre, Toronto, ON
| |
Collapse
|
54
|
Johansson A, Hamzah J, Ganss R. More than a scaffold: Stromal modulation of tumor immunity. Biochim Biophys Acta Rev Cancer 2015; 1865:3-13. [PMID: 26071879 DOI: 10.1016/j.bbcan.2015.06.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 05/20/2015] [Accepted: 06/04/2015] [Indexed: 12/25/2022]
Abstract
Current clinical success with anti-cancer immunotherapy provides exciting new treatment opportunities. While encouraging, more needs to be done to induce durable effects in a higher proportion of patients. Increasing anti-tumor effector T cell quantity or quality alone does not necessarily correlate with therapeutic outcome. Instead, the tumor microenvironment is a critical determinant of anti-cancer responsiveness to immunotherapy and can confer profound resistance. Yet, the tumor-promoting environment - due to its enormous plasticity - also delivers the best opportunities for adjuvant therapy aiming at recruiting, priming and sustaining anti-tumor cytotoxicity. While the tumor environment as an entity is increasingly well understood, current interventions are still broad and often systemic. In contrast, tumors grow in a highly compartmentalized environment which includes the vascular/perivascular niche, extracellular matrix components and in some tumors lymph node aggregates; all of these structures harbor and instruct subsets of immune cells. Targeting and re-programming specific compartments may provide better opportunities for adjuvant immunotherapy.
Collapse
Affiliation(s)
- Anna Johansson
- Vascular Biology and Stromal Targeting, Harry Perkins Institute of Medical Research, The University of Western Australia, Centre for Medical Research, Nedlands, Western Australia 6009, Australia
| | - Juliana Hamzah
- Targeted Drug Delivery, Imaging and Therapy, Harry Perkins Institute of Medical Research, The University of Western Australia, Centre for Medical Research, Nedlands, Western Australia 6009, Australia
| | - Ruth Ganss
- Vascular Biology and Stromal Targeting, Harry Perkins Institute of Medical Research, The University of Western Australia, Centre for Medical Research, Nedlands, Western Australia 6009, Australia.
| |
Collapse
|
55
|
Roth MD, Harui A. Human tumor infiltrating lymphocytes cooperatively regulate prostate tumor growth in a humanized mouse model. J Immunother Cancer 2015; 3:12. [PMID: 25901284 PMCID: PMC4404579 DOI: 10.1186/s40425-015-0056-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 03/06/2015] [Indexed: 12/17/2022] Open
Abstract
Background The complex interactions that occur between human tumors, tumor infiltrating lymphocytes (TIL) and the systemic immune system are likely to define critical factors in the host response to cancer. While conventional animal models have identified an array of potential anti-tumor therapies, mouse models often fail to translate into effective human treatments. Our goal is to establish a humanized tumor model as a more effective pre-clinical platform for understanding and manipulating TIL. Methods The immune system in NOD/SCID/IL-2Rγnull (NSG) mice was reconstituted by the co-administration of human peripheral blood lymphocytes (PBL) or subsets (CD4+ or CD8+) and autologous human dendritic cells (DC), and animals simultaneously challenged by implanting human prostate cancer cells (PC3 line). Tumor growth was evaluated over time and the phenotype of recovered splenocytes and TIL characterized by flow cytometry and immunohistochemistry (IHC). Serum levels of circulating cytokines and chemokines were also assessed. Results A tumor-bearing huPBL-NSG model was established in which human leukocytes reconstituted secondary lymphoid organs and promoted the accumulation of TIL. These TIL exhibited a unique phenotype when compared to splenocytes with a predominance of CD8+ T cells that exhibited increased expression of CD69, CD56, and an effector memory phenotype. TIL from huPBL-NSG animals closely matched the features of TIL recovered from primary human prostate cancers. Human cytokines were readily detectible in the serum and exhibited a different profile in animals implanted with PBL alone, tumor alone, and those reconstituted with both. Immune reconstitution slowed but could not eliminate tumor growth and this effect required the presence of CD4+ T cell help. Conclusions Simultaneous implantation of human PBL, DC and tumor results in a huPBL-NSG model that recapitulates the development of human TIL and allows an assessment of tumor and immune system interaction that cannot be carried out in humans. Furthermore, the capacity to manipulate individual features and cell populations provides an opportunity for hypothesis testing and outcome monitoring in a humanized system that may be more relevant than conventional mouse models.
Collapse
Affiliation(s)
- Michael D Roth
- Division of Pulmonary & Critical Care, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1690 USA
| | - Airi Harui
- Division of Pulmonary & Critical Care, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1690 USA
| |
Collapse
|
56
|
Tran Janco JM, Lamichhane P, Karyampudi L, Knutson KL. Tumor-infiltrating dendritic cells in cancer pathogenesis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2015; 194:2985-91. [PMID: 25795789 PMCID: PMC4369768 DOI: 10.4049/jimmunol.1403134] [Citation(s) in RCA: 329] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Dendritic cells (DCs) play a pivotal role in the tumor microenvironment, which is known to affect disease progression in many human malignancies. Infiltration by mature, active DCs into the tumors confers an increase in immune activation and recruitment of disease-fighting immune effector cells and pathways. DCs are the preferential target of infiltrating T cells. However, tumor cells have means of suppressing DC function or of altering the tumor microenvironment in such a way that immune-suppressive DCs are recruited. Advances in understanding these changes have led to promising developments in cancer-therapeutic strategies targeting tumor-infiltrating DCs to subdue their immunosuppressive functions and enhance their immune-stimulatory capacity.
Collapse
Affiliation(s)
| | - Purushottam Lamichhane
- Department of Immunology, Mayo Clinic, Rochester, MN 55906; and Cancer Vaccines and Immune Therapies Program, Vaccine and Gene Therapy Institute, Port St. Lucie, FL 34987
| | - Lavakumar Karyampudi
- Cancer Vaccines and Immune Therapies Program, Vaccine and Gene Therapy Institute, Port St. Lucie, FL 34987
| | - Keith L Knutson
- Department of Immunology, Mayo Clinic, Rochester, MN 55906; and Cancer Vaccines and Immune Therapies Program, Vaccine and Gene Therapy Institute, Port St. Lucie, FL 34987
| |
Collapse
|
57
|
Zhang GQ, Li F, Sun SJ, Hu Y, Wang G, Wang Y, Cui XX, Jiao SC. Adoptive Immunotherapy for Small Cell Lung Cancer by Expanded Activated Autologous Lymphocytes: a Retrospective Clinical Analysis. Asian Pac J Cancer Prev 2015; 16:1487-94. [DOI: 10.7314/apjcp.2015.16.4.1487] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
58
|
McDonnell AM, Joost Lesterhuis W, Khong A, Nowak AK, Lake RA, Currie AJ, Robinson BW. Restoration of defective cross-presentation in tumors by gemcitabine. Oncoimmunology 2015; 4:e1005501. [PMID: 26155402 PMCID: PMC4485774 DOI: 10.1080/2162402x.2015.1005501] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Revised: 12/29/2014] [Accepted: 12/30/2014] [Indexed: 02/03/2023] Open
Abstract
Tumor antigen cross-presentation by dendritic cells (DCs) to specific CD8+ T cells is central to antitumor immunity. Although highly efficient in draining lymph nodes, it is defective within the tumor site itself. Importantly, an immunogenic chemotherapy, gemcitabine, reverses this defect, allowing the potential re-stimulation of cytotoxic T lymphocytes within tumor sites.
Collapse
Affiliation(s)
- Alison M McDonnell
- School of Medicine and Pharmacology; The University of Western Australia ; Nedlands, WA, Australia ; National Centre for Asbestos Related Diseases ; Nedlands, WA, Australia
| | - W Joost Lesterhuis
- School of Medicine and Pharmacology; The University of Western Australia ; Nedlands, WA, Australia ; National Centre for Asbestos Related Diseases ; Nedlands, WA, Australia
| | - Andrea Khong
- School of Medicine and Pharmacology; The University of Western Australia ; Nedlands, WA, Australia ; National Centre for Asbestos Related Diseases ; Nedlands, WA, Australia
| | - Anna K Nowak
- School of Medicine and Pharmacology; The University of Western Australia ; Nedlands, WA, Australia ; National Centre for Asbestos Related Diseases ; Nedlands, WA, Australia ; Department of Medical Oncology; Sir Charles Gairdner Hospital ; Nedlands, WA, Australia
| | - Richard A Lake
- School of Medicine and Pharmacology; The University of Western Australia ; Nedlands, WA, Australia ; National Centre for Asbestos Related Diseases ; Nedlands, WA, Australia
| | - Andrew J Currie
- School of Medicine and Pharmacology; The University of Western Australia ; Nedlands, WA, Australia ; School of Veterinary and Life Sciences, Murdoch University , Murdoch, WA, Australia
| | - Bruce Ws Robinson
- School of Medicine and Pharmacology; The University of Western Australia ; Nedlands, WA, Australia ; National Centre for Asbestos Related Diseases ; Nedlands, WA, Australia
| |
Collapse
|
59
|
Guldner IH, Zhang S. A journey to uncharted territory: new technical frontiers in studying tumor-stromal cell interactions. Integr Biol (Camb) 2015; 7:153-61. [PMID: 25500646 PMCID: PMC4324098 DOI: 10.1039/c4ib00192c] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The crosstalk between tumor cells and cells of the tumor stroma dictate malignant progression and represent an intriguing and viable anticancer therapeutic target. The successful development of therapeutics targeting tumor-stroma interactions is tied to the insight provided by basic research on such crosstalk. Tumor-stroma interactions can be transient and dynamic, and they occur within defined spatiotemporal contexts among genetically and compositionally heterogeneous populations of cells, yet methods currently applied to study the said crosstalk do not sufficiently address these features. Emerging imaging and genetic methods, however, can overcome limitations of traditional approaches and provide unprecedented insight into tumor-stroma crosstalk with unparalleled accuracy. The comprehensive data obtained by applying emerging methods will require processing and analysis by multidisciplinary teams, but the efforts will ultimately rejuvenate hope in developing novel therapies against pro-tumorigenic tumor-stroma crosstalk.
Collapse
Affiliation(s)
- Ian H Guldner
- Department of Biological Science, Harper Cancer Research Institute, University of Notre Dame, A130 Harper Hall, Notre Dame, IN 46556, USA.
| | | |
Collapse
|
60
|
McDonnell AM, Lesterhuis WJ, Khong A, Nowak AK, Lake RA, Currie AJ, Robinson BWS. Tumor-infiltrating dendritic cells exhibit defective cross-presentation of tumor antigens, but is reversed by chemotherapy. Eur J Immunol 2014; 45:49-59. [PMID: 25316312 DOI: 10.1002/eji.201444722] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2014] [Revised: 09/19/2014] [Accepted: 10/10/2014] [Indexed: 11/11/2022]
Abstract
Cross-presentation defines the unique capacity of an APC to present exogenous Ag via MHC class I molecules to CD8(+) T cells. DCs are specialized cross-presenting cells and as such have a critical role in antitumor immunity. DCs are routinely found within the tumor microenvironment, but their capacity for endogenous or therapeutically enhanced cross-presentation is not well characterized. In this study, we examined the tumor and lymph node DC cross-presentation of a nominal marker tumor Ag, HA, expressed by the murine mesothelioma tumor AB1-HA. We found that tumors were infiltrated by predominantly CD11b(+) DCs with a semimature phenotype that could not cross-present tumor Ag, and therefore, were unable to induce tumor-specific T-cell activation or proliferation. Although tumor-infiltrating DCs were able to take up, process, and cross-present exogenous cell-bound and soluble Ags, this was significantly impaired relative to lymph node DCs. Importantly, however, systemic chemotherapy using gemcitabine reversed the defect in Ag cross-presentation of tumor DCs. These data demonstrate that DC cross-presentation within the tumor microenvironment is defective, but can be reversed by chemotherapy. These results have important implications for anticancer therapy, particularly regarding the use of immunotherapy in conjunction with cytotoxic chemotherapy.
Collapse
Affiliation(s)
- Alison M McDonnell
- National Centre for Asbestos Related Diseases, School of Medicine and Pharmacology, The University of Western Australia, Nedlands, WA, Australia
| | | | | | | | | | | | | |
Collapse
|
61
|
Sharma RK, Chheda ZS, Jala VR, Haribabu B. Regulation of cytotoxic T-Lymphocyte trafficking to tumors by chemoattractants: implications for immunotherapy. Expert Rev Vaccines 2014; 14:537-49. [PMID: 25482400 DOI: 10.1586/14760584.2015.982101] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Cancer immunotherapy has recently emerged as an important treatment modality. FDA approval of provenge, ipilimumab and pembrolizumab has started to deliver on the long awaited promise of cancer immunotherapy. Many new modalities of immunotherapies targeting cytotoxic T lymphocytes (CTLs) responses, such as adoptive cell therapies and vaccines, are in advanced clinical trials. In all these immunotherapies, migration of CTLs to the tumor site is a critical step for achieving therapeutic efficacy. However, inefficient infiltration of activated CTLs into established tumors is increasingly being recognized as one of the major hurdles limiting efficacy. Mechanisms that control migration of CTLs to tumors are poorly defined. In this review, the authors discuss the chemoattractants and their receptors that have been implicated in endogenous- or immunotherapy-induced CTL recruitment to tumors and the potential for targeting these pathways for therapeutic efficacy.
Collapse
Affiliation(s)
- Rajesh K Sharma
- James Graham Brown Cancer Center, University of Louisville Health Sciences, Louisville, KY 40202, USA
| | | | | | | |
Collapse
|
62
|
Broz ML, Binnewies M, Boldajipour B, Nelson AE, Pollack JL, Erle DJ, Barczak A, Rosenblum MD, Daud A, Barber DL, Amigorena S, Van't Veer LJ, Sperling AI, Wolf DM, Krummel MF. Dissecting the tumor myeloid compartment reveals rare activating antigen-presenting cells critical for T cell immunity. Cancer Cell 2014; 26:638-52. [PMID: 25446897 PMCID: PMC4254577 DOI: 10.1016/j.ccell.2014.09.007] [Citation(s) in RCA: 797] [Impact Index Per Article: 79.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 08/28/2014] [Accepted: 09/19/2014] [Indexed: 12/13/2022]
Abstract
It is well understood that antigen-presenting cells (APCs) within tumors typically do not maintain cytotoxic T cell (CTL) function, despite engaging them. Across multiple mouse tumor models and human tumor biopsies, we have delineated the intratumoral dendritic cell (DC) populations as distinct from macrophage populations. Within these, CD103(+) DCs are extremely sparse and yet remarkably capable CTL stimulators. These are uniquely dependent on IRF8, Zbtb46, and Batf3 transcription factors and are generated by GM-CSF and FLT3L cytokines. Regressing tumors have higher proportions of these cells, T-cell-dependent immune clearance relies on them, and abundance of their transcripts in human tumors correlates with clinical outcome. This cell type presents opportunities for prognostic and therapeutic approaches across multiple cancer types.
Collapse
Affiliation(s)
- Miranda L Broz
- Department of Pathology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Mikhail Binnewies
- Department of Pathology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Bijan Boldajipour
- Department of Pathology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Amanda E Nelson
- Department of Pathology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Joshua L Pollack
- Lung Biology Center, University of California San Francisco, San Francisco, CA 94143, USA
| | - David J Erle
- Lung Biology Center, University of California San Francisco, San Francisco, CA 94143, USA
| | - Andrea Barczak
- Lung Biology Center, University of California San Francisco, San Francisco, CA 94143, USA
| | - Michael D Rosenblum
- Department of Dermatology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Adil Daud
- Melanoma Clinical Research Unit, University of California San Francisco, San Francisco, CA 94143, USA
| | - Diane L Barber
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Sebastian Amigorena
- INSERM U932, Immunity and Cancer, Institut Curie, 75248 Paris Cedex 05, France
| | - Laura J Van't Veer
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA 94143, USA
| | - Anne I Sperling
- Committee on Immunology, University of Chicago, 924 E. 57th Street, Chicago, IL 60637, USA
| | - Denise M Wolf
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA 94143, USA
| | - Matthew F Krummel
- Department of Pathology, University of California San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
63
|
Intravital Multiphoton Imaging of Cutaneous Immune Responses. J Invest Dermatol 2014; 134:2680-2684. [DOI: 10.1038/jid.2014.225] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 04/15/2014] [Accepted: 04/29/2014] [Indexed: 12/31/2022]
|
64
|
Cancer subclonal genetic architecture as a key to personalized medicine. Neoplasia 2014; 15:1410-20. [PMID: 24403863 DOI: 10.1593/neo.131972] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 12/03/2013] [Accepted: 12/03/2013] [Indexed: 02/08/2023] Open
Abstract
The future of personalized oncological therapy will likely rely on evidence-based medicine to integrate all of the available evidence to delineate the most efficacious treatment option for the patient. To undertake evidence-based medicine through use of targeted therapy regimens, identification of the specific underlying causative mutation(s) driving growth and progression of a patient's tumor is imperative. Although molecular subtyping is important for planning and treatment, intraclonal genetic diversity has been recently highlighted as having significant implications for biopsy-based prognosis. Overall, delineation of the clonal architecture of a patient's cancer and how this will impact on the selection of the most efficacious therapy remain a topic of intense interest.
Collapse
|
65
|
Benechet AP, Menon M, Khanna KM. Visualizing T Cell Migration in situ. Front Immunol 2014; 5:363. [PMID: 25120547 PMCID: PMC4114210 DOI: 10.3389/fimmu.2014.00363] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Accepted: 07/14/2014] [Indexed: 12/16/2022] Open
Abstract
Mounting a protective immune response is critically dependent on the orchestrated movement of cells within lymphoid tissues. The structure of secondary lymphoid organs regulates immune responses by promoting optimal cell-cell and cell-extracellular matrix interactions. Naïve T cells are initially activated by antigen presenting cells in secondary lymphoid organs. Following priming, effector T cells migrate to the site of infection to exert their functions. Majority of the effector cells die while a small population of antigen-specific T cells persists as memory cells in distinct anatomical locations. The persistence and location of memory cells in lymphoid and non-lymphoid tissues is critical to protect the host from re-infection. The localization of memory T cells is carefully regulated by several factors including the highly organized secondary lymphoid structure, the cellular expression of chemokine receptors and compartmentalized secretion of their cognate ligands. This balance between the anatomy and the ordered expression of cell surface and soluble proteins regulates the subtle choreography of T cell migration. In recent years, our understanding of cellular dynamics of T cells has been advanced by the development of new imaging techniques allowing in situ visualization of T cell responses. Here, we review the past and more recent studies that have utilized sophisticated imaging technologies to investigate the migration dynamics of naïve, effector, and memory T cells.
Collapse
Affiliation(s)
- Alexandre P. Benechet
- Department of Immunology, University of Connecticut Health Center, Farmington, CT, USA
| | - Manisha Menon
- Department of Immunology, University of Connecticut Health Center, Farmington, CT, USA
| | - Kamal M. Khanna
- Department of Immunology, University of Connecticut Health Center, Farmington, CT, USA
| |
Collapse
|
66
|
Abstract
The tumor microenvironment is a complex ecology of cells that evolves with and provides support to tumor cells during the transition to malignancy. Among the innate and adaptive immune cells recruited to the tumor site, macrophages are particularly abundant and are present at all stages of tumor progression. Clinical studies and experimental mouse models indicate that these macrophages generally play a protumoral role. In the primary tumor, macrophages can stimulate angiogenesis and enhance tumor cell invasion, motility, and intravasation. During monocytes and/or metastasis, macrophages prime the premetastatic site and promote tumor cell extravasation, survival, and persistent growth. Macrophages are also immunosuppressive, preventing tumor cell attack by natural killer and T cells during tumor progression and after recovery from chemo- or immunotherapy. Therapeutic success in targeting these protumoral roles in preclinical models and in early clinical trials suggests that macrophages are attractive targets as part of combination therapy in cancer treatment.
Collapse
Affiliation(s)
- Roy Noy
- Department of Developmental and Molecular Biology, Center for the Study of Reproductive Biology and Women's Health, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Jeffrey W Pollard
- Department of Developmental and Molecular Biology, Center for the Study of Reproductive Biology and Women's Health, Albert Einstein College of Medicine, New York, NY 10461, USA; MRC Centre for Reproductive Health, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, UK.
| |
Collapse
|
67
|
Abstract
The tumor microenvironment is a complex ecology of cells that evolves with and provides support to tumor cells during the transition to malignancy. Among the innate and adaptive immune cells recruited to the tumor site, macrophages are particularly abundant and are present at all stages of tumor progression. Clinical studies and experimental mouse models indicate that these macrophages generally play a protumoral role. In the primary tumor, macrophages can stimulate angiogenesis and enhance tumor cell invasion, motility, and intravasation. During monocytes and/or metastasis, macrophages prime the premetastatic site and promote tumor cell extravasation, survival, and persistent growth. Macrophages are also immunosuppressive, preventing tumor cell attack by natural killer and T cells during tumor progression and after recovery from chemo- or immunotherapy. Therapeutic success in targeting these protumoral roles in preclinical models and in early clinical trials suggests that macrophages are attractive targets as part of combination therapy in cancer treatment.
Collapse
Affiliation(s)
- Roy Noy
- Department of Developmental and Molecular Biology, Center for the Study of Reproductive Biology and Women's Health, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Jeffrey W Pollard
- Department of Developmental and Molecular Biology, Center for the Study of Reproductive Biology and Women's Health, Albert Einstein College of Medicine, New York, NY 10461, USA; MRC Centre for Reproductive Health, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, UK.
| |
Collapse
|
68
|
Abstract
To comprehend the complexity of cancer, the biological characteristics acquired during the initiation and progression of tumours were classified as the 'hallmarks of cancer'. Intravital microscopy techniques have been developed to study individual cells that acquire these crucial traits, by visualizing tissues with cellular or subcellular resolution in living animals. In this Review, we highlight the latest intravital microscopy techniques that have been used in living animals (predominantly mice) to unravel fundamental and dynamic aspects of various hallmarks of cancer. In addition, we discuss the application of intravital microscopy techniques to cancer therapy, as well as limitations and future perspectives for these techniques.
Collapse
Affiliation(s)
- Saskia I J Ellenbroek
- Cancer Genomics Netherlands-Hubrecht Institute-KNAW & University Medical Centre Utrecht, Uppsalalaan 8, 3584 CT, Utrecht, The Netherlands
| | - Jacco van Rheenen
- Cancer Genomics Netherlands-Hubrecht Institute-KNAW & University Medical Centre Utrecht, Uppsalalaan 8, 3584 CT, Utrecht, The Netherlands
| |
Collapse
|
69
|
Son CH, Bae JH, Shin DY, Lee HR, Yang K, Park YS. Antitumor effect of dendritic cell loaded ex vivo and in vivo with tumor-associated antigens in lung cancer model. Immunol Invest 2014; 43:447-62. [PMID: 24654594 DOI: 10.3109/08820139.2014.884576] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Various ex vivo or in vivo loading protocols have been developed or evaluated for the delivery of tumor antigens to dendritic cells (DCs). We compared the antitumor effect of mature DCs electroporation-pulsed (EP/mDC) ex vivo with tumor cell lysate and immature DCs (iDCs) injected into the tumor apoptosed by ionizing radiation (IR/iDC) in lung cancer model. DCs were generated from bone marrow of C57BL/6 mice. Ionizing radiation (IR) was applied at a dose of 10 Gy to the tumor on the right thigh. iDCs were intratumorally injected into the irradiated tumor and EP/mDC was injected subcutaneously in the right flank. DC injection induced strong tumor-specific immunity against Lewis lung carcinoma, as compared with the tumor-bearing control and IR only treated mice. The growth of a distant tumor on the right and left flank was inhibited by IR/iDC and EP/mDC. Particularly, IR/iDC resulted in a more significant inhibition of tumor growth and prolonged survival time. It was related to increase of tumor-specific interferon-gamma, cytotoxicity, and decrease of regulatory T-cells. The results indicate that DCs electroporation-pulsed with tumor cell lysate induce a potent antitumor effect, but that iDCs intratumoral injected into the irradiated tumor induce a more potent antitumor effect.
Collapse
Affiliation(s)
- Cheol-Hun Son
- Dongnam Institute of Radiological & Medical Sciences , Busan 619-953 , South Korea
| | | | | | | | | | | |
Collapse
|
70
|
Bracci L, Schiavoni G, Sistigu A, Belardelli F. Immune-based mechanisms of cytotoxic chemotherapy: implications for the design of novel and rationale-based combined treatments against cancer. Cell Death Differ 2014; 21:15-25. [PMID: 23787994 PMCID: PMC3857622 DOI: 10.1038/cdd.2013.67] [Citation(s) in RCA: 649] [Impact Index Per Article: 64.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Revised: 05/07/2013] [Accepted: 05/14/2013] [Indexed: 02/06/2023] Open
Abstract
Conventional anticancer chemotherapy has been historically thought to act through direct killing of tumor cells. This concept stems from the fact that cytotoxic drugs interfere with DNA synthesis and replication. Accumulating evidence, however, indicates that the antitumor activities of chemotherapy also rely on several off-target effects, especially directed to the host immune system, that cooperate for successful tumor eradication. Chemotherapeutic agents stimulate both the innate and adaptive arms of the immune system through several modalities: (i) by promoting specific rearrangements on dying tumor cells, which render them visible to the immune system; (ii) by influencing the homeostasis of the hematopoietic compartment through transient lymphodepletion followed by rebound replenishment of immune cell pools; (iii) by subverting tumor-induced immunosuppressive mechanisms and (iv) by exerting direct or indirect stimulatory effects on immune effectors. Among the indirect ways of immune cell stimulation, some cytotoxic drugs have been shown to induce an immunogenic type of cell death in tumor cells, resulting in the emission of specific signals that trigger phagocytosis of cell debris and promote the maturation of dendritic cells, ultimately resulting in the induction of potent antitumor responses. Here, we provide an extensive overview of the multiple immune-based mechanisms exploited by the most commonly employed cytotoxic drugs, with the final aim of identifying prerequisites for optimal combination with immunotherapy strategies for the development of more effective treatments against cancer.
Collapse
Affiliation(s)
- L Bracci
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - G Schiavoni
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - A Sistigu
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - F Belardelli
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
71
|
Workenhe ST, Pol JG, Lichty BD, Cummings DT, Mossman KL. Combining oncolytic HSV-1 with immunogenic cell death-inducing drug mitoxantrone breaks cancer immune tolerance and improves therapeutic efficacy. Cancer Immunol Res 2013; 1:309-19. [PMID: 24777969 DOI: 10.1158/2326-6066.cir-13-0059-t] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Although antitumor activity of herpes simplex virus 1 (HSV-1) ICP0 null oncolytic vectors has been validated in murine breast cancer models, oncolytic virus treatment alone is insufficient to break immune tolerance. Thus, we investigated enhancing efficacy through combination therapy with the immunogenic cell death-inducing chemotherapeutic drug, mitoxantrone. Despite a lack of enhanced cytotoxicity in vitro, HSV-1 ICP0 null oncolytic virus KM100 with 5 μmol/L mitoxantrone provided significant survival benefit to BALB/c mice bearing Her2/neu TUBO-derived tumors. This protection was mediated by increased intratumoral infiltration of neutrophils and tumor antigen-specific CD8(+) T cells. Depletion studies verified that CD8-, CD4-, and Ly6G-expressing cells are essential for enhanced efficacy of the combination therapy. Moreover, the addition of mitoxantrone to KM100 oncolytic virus treatment broke immune tolerance in BALB-neuT mice bearing TUBO-derived tumors. This study suggests that oncolytic viruses in combination with immunogenic cell death-inducing chemotherapeutics enhance the immunogenicity of the tumor-associated antigens, breaking immunologic tolerance established toward these antigens.
Collapse
Affiliation(s)
- Samuel T Workenhe
- Authors' Affiliation: Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | | | | | | | | |
Collapse
|
72
|
Alexander S, Weigelin B, Winkler F, Friedl P. Preclinical intravital microscopy of the tumour-stroma interface: invasion, metastasis, and therapy response. Curr Opin Cell Biol 2013; 25:659-71. [PMID: 23896198 DOI: 10.1016/j.ceb.2013.07.001] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 07/01/2013] [Accepted: 07/02/2013] [Indexed: 01/10/2023]
Abstract
Key steps of cancer progression and therapy response depend upon interactions between cancer cells with the reactive tumour microenvironment. Intravital microscopy enables multi-modal and multi-scale monitoring of cancer progression as a dynamic step-wise process within anatomic and functional niches provided by the microenvironment. These niches deliver cell-derived and matrix-derived signals that enable cell subsets or single cancer cells to survive, migrate, grow, undergo dormancy, and escape immune surveillance. Beyond basic research, intravital microscopy has reached preclinical application to identify mechanisms of tumour-stroma interactions and outcome. We here summarise how n-dimensional 'dynamic histopathology' of tumours by intravital microscopy shapes mechanistic insight into cell-cell and cell-tissue interactions that underlie single-cell and collective cancer invasion, metastatic seeding at distant sites, immune evasion, and therapy responses.
Collapse
Affiliation(s)
- Stephanie Alexander
- David H. Koch Center for Applied Research of Genitourinary Cancers, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | | | | | | |
Collapse
|
73
|
Xiao-Ai-Ping, a TCM Injection, Enhances the Antigrowth Effects of Cisplatin on Lewis Lung Cancer Cells through Promoting the Infiltration and Function of CD8(+) T Lymphocytes. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:879512. [PMID: 23956781 PMCID: PMC3730189 DOI: 10.1155/2013/879512] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Revised: 06/17/2013] [Accepted: 06/28/2013] [Indexed: 12/20/2022]
Abstract
Objectives. To investigate how Xiao-Ai-Ping injection, a traditional Chinese medicine and an ancillary drug in tumor treatment, enhances the antitumor effects of cisplatin on Lewis lung cancer (LLC) cells. Methods. LLC-bearing mice were daily intraperitoneally injected with various doses of cisplatin, Xiao-Ai-Ping, or cisplatin plus Xiao-Ai-Ping, respectively. Body weight and tumor volumes were measured every three days. Results. Combination of Xiao-Ai-Ping and cisplatin yielded significantly better antigrowth and proapoptotic effects on LLC xenografts than sole drug treatment did. In addition, we found that Xiao-Ai-Ping triggered the infiltration of CD8+ T cells, a group of cytotoxic T cells, to LLC xenografts. Furthermore, the mRNA levels of interferon-γ (ifn-γ), perforin-1 (prf-1), and granzyme B (gzmb) in CD8+ T cells were significantly increased after combination treatment of Xiao-Ai-Ping and cisplatin. In vitro studies showed that Xiao-Ai-Ping markedly upregulated the mRNA levels of ifn-γ, prf-1, and gzmb in CD8+ T cells in a concentration-dependent manner, suggesting that Xiao-Ai-Ping augments the function of CD8+ T cells. Conclusions. Xiao-Ai-Ping promotes the infiltration and function of CD8+ T cells and thus enhances the antigrowth effects of cisplatin on LLC xenografts, which provides new evidence for the combination of Xiao-Ai-Ping and cisplatin in clinic in China.
Collapse
|