151
|
Balan S, Saxena M, Bhardwaj N. Dendritic cell subsets and locations. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 348:1-68. [PMID: 31810551 DOI: 10.1016/bs.ircmb.2019.07.004] [Citation(s) in RCA: 215] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Dendritic cells (DCs) are a unique class of immune cells that act as a bridge between innate and adaptive immunity. The discovery of DCs by Cohen and Steinman in 1973 laid the foundation for DC biology, and the advances in the field identified different versions of DCs with unique properties and functions. DCs originate from hematopoietic stem cells, and their differentiation is modulated by Flt3L. They are professional antigen-presenting cells that patrol the environmental interphase, sites of infection, or infiltrate pathological tissues looking for antigens that can be used to activate effector cells. DCs are critical for the initiation of the cellular and humoral immune response and protection from infectious diseases or tumors. DCs can take up antigens using specialized surface receptors such as endocytosis receptors, phagocytosis receptors, and C type lectin receptors. Moreover, DCs are equipped with an array of extracellular and intracellular pattern recognition receptors for sensing different danger signals. Upon sensing the danger signals, DCs get activated, upregulate costimulatory molecules, produce various cytokines and chemokines, take up antigen and process it and migrate to lymph nodes where they present antigens to both CD8 and CD4 T cells. DCs are classified into different subsets based on an integrated approach considering their surface phenotype, expression of unique and conserved molecules, ontogeny, and functions. They can be broadly classified as conventional DCs consisting of two subsets (DC1 and DC2), plasmacytoid DCs, inflammatory DCs, and Langerhans cells.
Collapse
Affiliation(s)
- Sreekumar Balan
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| | - Mansi Saxena
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Nina Bhardwaj
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Parker Institute for Cancer Immunotherapy, San Francisco, CA, United States
| |
Collapse
|
152
|
Glycan-Modified Apoptotic Melanoma-Derived Extracellular Vesicles as Antigen Source for Anti-Tumor Vaccination. Cancers (Basel) 2019; 11:cancers11091266. [PMID: 31466401 PMCID: PMC6769957 DOI: 10.3390/cancers11091266] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 08/16/2019] [Accepted: 08/21/2019] [Indexed: 02/06/2023] Open
Abstract
Tumors that lack T cell infiltration are less likely to respond to immune checkpoint inhibition and could benefit from cancer vaccination for the initiation of anti-tumor T cell responses. An attractive vaccine strategy is in vivo targeting of dendritic cells (DCs), key initiators of antigen-specific T cell responses. In this study we generated tumor-derived apoptotic extracellular vesicles (ApoEVs), which are potentially an abundant source of tumor-specific neo-antigens and other tumor-associated antigens (TAAs), and which can be manipulated to express DC-targeting ligands for efficient antigen delivery. Our data demonstrates that by specifically modifying the glycocalyx of tumor cells, high-mannose glycans can be expressed on their cell surface and on extracellular vesicles derived after the induction of apoptosis. High-mannose glycans are the natural ligands of dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin (DC-SIGN), a dendritic cell associated C-type lectin receptor (CLR), which has the ability to efficiently internalize its cargo and direct it to both major histocompatibility complex (MHC)-I and MHC-II pathways for the induction of CD8+ and CD4+ T cell responses, respectively. Compared to unmodified ApoEVs, ApoEVs carrying DC-SIGN ligands are internalized to a higher extent, resulting in enhanced priming of tumor-specific CD8+ T cells. This approach thus presents a promising vaccination strategy in support of T cell-based immunotherapy of cancer.
Collapse
|
153
|
Doix B, Trempolec N, Riant O, Feron O. Low Photosensitizer Dose and Early Radiotherapy Enhance Antitumor Immune Response of Photodynamic Therapy-Based Dendritic Cell Vaccination. Front Oncol 2019; 9:811. [PMID: 31508370 PMCID: PMC6718637 DOI: 10.3389/fonc.2019.00811] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 08/08/2019] [Indexed: 12/22/2022] Open
Abstract
Recent studies have highlighted the potential of photodynamic therapy (PDT) to induce immunogenic cell death (ICD). The clinical use of photosensitizers (PS) to stimulate an anticancer immune response, and not to sterilize tumor cells, may however require some optimizations. Here, we examined how the dose of PS and the scheduling of PDT influence the generation of danger-associated molecular patterns proteins (DAMPs) and favor T cell antitumor activity. We found that upon photoactivation, a low dose of the non-porphyrinic PS OR141 was more prone than higher doses to induce DAMPs in vitro and to inhibit squamous cell carcinoma growth in mice. We further used PDT-killed cancer cells to prime dendritic cells (DC) and stimulate their maturation to evaluate whether the timing of their injection could influence the antitumor effects of radiotherapy. While PDT-based DC vaccination administered before radiotherapy failed to increase tumor growth inhibition, DC injection in the peri-radiotherapy period led to significant tumor growth delay, emphasizing the importance of the coincidence of T cell activation and alterations of the tumor bed. In conclusion, the use of OR141 as a bona fide ICD inducer led us to unravel both the non-linear relationship between PS concentration and PDT-induced antitumor immune response, and the value of an optimal timing of PDT when co-administered with conventional anticancer therapies. This study therefore stresses the necessity of adapting the clinical use of PDT when the goal is to promote an immune response and identifies PDT-based DC vaccination as a suitable modality to reach such objective.
Collapse
Affiliation(s)
- Bastien Doix
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Brussels, Belgium
| | - Natalia Trempolec
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Brussels, Belgium
| | - Olivier Riant
- Institute of Condensed Matter and Nanosciences, Molecular Chemistry, Materials and Catalysis (IMCN/MOST), UCLouvain, Louvain-la-Neuve, Belgium
| | - Olivier Feron
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Brussels, Belgium
| |
Collapse
|
154
|
Sun F, Cui L, Li T, Chen S, Song J, Li D. Oxaliplatin induces immunogenic cells death and enhances therapeutic efficacy of checkpoint inhibitor in a model of murine lung carcinoma. J Recept Signal Transduct Res 2019; 39:208-214. [DOI: 10.1080/10799893.2019.1655050] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Fengfei Sun
- The Second Clinical Medical College of North Sichuan Medical College, Sichuan, China
| | - Lijuan Cui
- Suining Central Hospital, Sichuan, China
| | - Tingting Li
- The Second Clinical Medical College of North Sichuan Medical College, Sichuan, China
| | - Silin Chen
- The Second Clinical Medical College of North Sichuan Medical College, Sichuan, China
| | - Junmei Song
- The Second Clinical Medical College of North Sichuan Medical College, Sichuan, China
| | - Dezhi Li
- The Second Clinical Medical College of North Sichuan Medical College, Sichuan, China
| |
Collapse
|
155
|
Donohoe C, Senge MO, Arnaut LG, Gomes-da-Silva LC. Cell death in photodynamic therapy: From oxidative stress to anti-tumor immunity. Biochim Biophys Acta Rev Cancer 2019; 1872:188308. [PMID: 31401103 DOI: 10.1016/j.bbcan.2019.07.003] [Citation(s) in RCA: 230] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 07/25/2019] [Accepted: 07/25/2019] [Indexed: 01/11/2023]
Abstract
Photodynamic therapy is a promising approach for cancer treatment that relies on the administration of a photosensitizer followed by tumor illumination. The generated oxidative stress may activate multiple mechanisms of cell death which are counteracted by cells through adaptive stress responses that target homeostasis rescue. The present renaissance of PDT was leveraged by the acknowledgment that this therapy has an immediate impact locally, in the illumination volume, but that subsequently it may also elicit immune responses with systemic impact. The investigation of the mechanisms of cell death under the oxidative stress of PDT is of paramount importance to understand how the immune system is activated and, ultimately, to make PDT a more appealing/relevant therapeutic option.
Collapse
Affiliation(s)
- Claire Donohoe
- CQC, Coimbra Chemistry Center, University of Coimbra, Portugal; Medicinal Chemistry, Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, Trinity College Dublin, The University of Dublin, St. James's Hospital, Dublin 8, Ireland
| | - Mathias O Senge
- Medicinal Chemistry, Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, Trinity College Dublin, The University of Dublin, St. James's Hospital, Dublin 8, Ireland
| | - Luís G Arnaut
- CQC, Coimbra Chemistry Center, University of Coimbra, Portugal
| | | |
Collapse
|
156
|
Tatsuno K, Yamazaki T, Hanlon D, Han P, Robinson E, Sobolev O, Yurter A, Rivera-Molina F, Arshad N, Edelson RL, Galluzzi L. Extracorporeal photochemotherapy induces bona fide immunogenic cell death. Cell Death Dis 2019; 10:578. [PMID: 31371700 PMCID: PMC6675789 DOI: 10.1038/s41419-019-1819-3] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 07/15/2019] [Indexed: 12/12/2022]
Abstract
Extracorporeal photochemotherapy (ECP) is employed for the management of cutaneous T cell lymphoma (CTCL). ECP involves the extracorporeal exposure of white blood cells (WBCs) to a photosensitizer, 8-methoxypsoralen (8-MOP), in the context of ultraviolet A (UVA) radiation, followed by WBC reinfusion. Historically, the therapeutic activity of ECP has been attributed to selective cytotoxicity on circulating CTCL cells. However, only a fraction of WBCs is exposed to ECP, and 8-MOP is inactive in the absence of UVA light, implying that other mechanisms underlie the anticancer effects of ECP. Recently, ECP has been shown to enable the physiological differentiation of monocytes into dendritic cells (DCs) that efficiently cross-present tumor-associated antigens (TAAs) to CD8+ T lymphocytes to initiate cognate immunity. However, the source of TAAs and immunostimulatory signals for such DCs remains to be elucidated. Here, we demonstrate that 8-MOP plus UVA light reduces melanoma cell viability along with the emission of ICD-associated danger signals including calreticulin (CALR) exposure on the cell surface and secretion of ATP, high mobility group box 1 (HMGB1) and type I interferon (IFN). Consistently, melanoma cells succumbing to 8-MOP plus UVA irradiation are efficiently engulfed by monocytes, ultimately leading to cross-priming of CD8+ T cells against cancer. Moreover, malignant cells killed by 8-MOP plus UVA irradiation in vitro vaccinate syngeneic immunocompetent mice against living cancer cells of the same type, and such a protection is lost when cancer cells are depleted of calreticulin or HMGB1, as well as in the presence of an ATP-degrading enzyme or antibodies blocking type I IFN receptors. ECP induces bona fide ICD, hence simultaneously providing monocytes with abundant amounts of TAAs and immunostimulatory signals that are sufficient to initiate cognate anticancer immunity.
Collapse
Affiliation(s)
- Kazuki Tatsuno
- Department of Dermatology, Yale School of Medicine, New Haven, CT, USA
| | - Takahiro Yamazaki
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Douglas Hanlon
- Department of Dermatology, Yale School of Medicine, New Haven, CT, USA
| | - Patrick Han
- Department of Dermatology, Yale School of Medicine, New Haven, CT, USA
| | - Eve Robinson
- Department of Dermatology, Yale School of Medicine, New Haven, CT, USA
| | - Olga Sobolev
- Department of Dermatology, Yale School of Medicine, New Haven, CT, USA
| | - Alp Yurter
- Department of Dermatology, Yale School of Medicine, New Haven, CT, USA
| | | | - Najla Arshad
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Richard L Edelson
- Department of Dermatology, Yale School of Medicine, New Haven, CT, USA. .,Comprehensive Cancer Center, Yale University School of Medicine, New Haven, CT, USA.
| | - Lorenzo Galluzzi
- Department of Dermatology, Yale School of Medicine, New Haven, CT, USA. .,Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA. .,Sandra and Edward Meyer Cancer Center, New York, NY, USA. .,Université Paris Descartes/Paris V, Paris, France.
| |
Collapse
|
157
|
Adams S, Gatti-Mays ME, Kalinsky K, Korde LA, Sharon E, Amiri-Kordestani L, Bear H, McArthur HL, Frank E, Perlmutter J, Page DB, Vincent B, Hayes JF, Gulley JL, Litton JK, Hortobagyi GN, Chia S, Krop I, White J, Sparano J, Disis ML, Mittendorf EA. Current Landscape of Immunotherapy in Breast Cancer: A Review. JAMA Oncol 2019; 5:1205-1214. [PMID: 30973611 DOI: 10.1001/jamaoncol.2018.7147] [Citation(s) in RCA: 266] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Importance There is tremendous interest in using immunotherapy to treat breast cancer, as evidenced by the more than 290 clinical trials ongoing at the time of this narrative review. The objective of this review is to describe the current status of immunotherapy in breast cancer, highlighting its potential in both early-stage and metastatic disease. Observations After searching ClinicalTrials.gov on April 24, 2018, and PubMed up to June 30, 2018, to identify breast cancer immunotherapy trials, we found that immune checkpoint blockade (ICB) is the most investigated form of immunotherapy in breast cancer. Use of ICB as monotherapy has achieved objective responses in patients with breast cancer, with higher rates seen when administered in earlier lines of therapy. For responding patients, those responses are durable. More recent data suggest clinical efficacy when ICB is given in combination with chemotherapy. Ongoing studies are evaluating combination strategies pairing ICB with additional chemotherapeutic agents, targeted therapy, vaccines, and local ablative therapies to enhance response. To date, robust predictive biomarkers for response to ICB have not been established. Conclusions and Relevance It is anticipated that combination therapy strategies will be the way forward for immunotherapy in breast cancer, with an improved understanding of tumor, microenvironment, and host factors informing treatment combination decisions. Thoughtful study design incorporating appropriate end points and correlative studies will be critical in identifying optimal strategies for enhancing the immune response against breast tumors.
Collapse
Affiliation(s)
- Sylvia Adams
- Perlmutter Cancer Center, NYU School of Medicine, New York, New York
| | - Margaret E Gatti-Mays
- Laboratory of Tumor Immunology and Biology, National Cancer Institute, Bethesda, Maryland
| | | | - Larissa A Korde
- Clinical Investigations Branch, Cancer Therapy and Evaluation Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, Maryland
| | - Elad Sharon
- Investigational Drug Branch, Cancer Therapy and Evaluation Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, Maryland
| | | | - Harry Bear
- Virginia Commonwealth University, Massey Cancer Center, Richmond
| | | | - Elizabeth Frank
- Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - David B Page
- Providence Cancer Institute, Earle A. Chiles Research Institute, Portland, Oregon
| | - Benjamin Vincent
- Department of Medicine, Division of Hematology/Oncology, Lineberger Comprehensive Cancer Center, Curriculum in Bioinformatics and Computational Biology, University of North Carolina at Chapel Hill
| | - Jennifer F Hayes
- Coordinating Center for Clinical Trials, National Cancer Institute, Rockville, Maryland
| | - James L Gulley
- Genitourinary Malignancy Branch, National Cancer Institute, Bethesda, Maryland
| | | | | | - Stephen Chia
- British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Ian Krop
- Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Julia White
- The Ohio State University Comprehensive Cancer Center, Columbus
| | - Joseph Sparano
- Montefiore Einstein Center for Cancer Care, New York, New York
| | - Mary L Disis
- Fred Hutchinson Cancer Research Center, Seattle, Washington.,Editor, JAMA Oncology
| | - Elizabeth A Mittendorf
- Department of Surgery, Brigham and Women's Hospital, Boston, Massachusetts.,Breast Oncology Program, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts
| |
Collapse
|
158
|
Targeting photodynamic and photothermal therapy to the endoplasmic reticulum enhances immunogenic cancer cell death. Nat Commun 2019; 10:3349. [PMID: 31350406 PMCID: PMC6659660 DOI: 10.1038/s41467-019-11269-8] [Citation(s) in RCA: 668] [Impact Index Per Article: 111.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 07/02/2019] [Indexed: 12/27/2022] Open
Abstract
Immunogenic cell death (ICD)-associated immunogenicity can be evoked through reactive oxygen species (ROS) produced via endoplasmic reticulum (ER) stress. In this study, we generate a double ER-targeting strategy to realize photodynamic therapy (PDT) photothermal therapy (PTT) immunotherapy. This nanosystem consists of ER-targeting pardaxin (FAL) peptides modified-, indocyanine green (ICG) conjugated- hollow gold nanospheres (FAL-ICG-HAuNS), together with an oxygen-delivering hemoglobin (Hb) liposome (FAL-Hb lipo), designed to reverse hypoxia. Compared with non-targeting nanosystems, the ER-targeting naosystem induces robust ER stress and calreticulin (CRT) exposure on the cell surface under near-infrared (NIR) light irradiation. CRT, a marker for ICD, acts as an ‘eat me’ signal to stimulate the antigen presenting function of dendritic cells. As a result, a series of immunological responses are activated, including CD8+ T cell proliferation and cytotoxic cytokine secretion. In conclusion, ER-targeting PDT-PTT promoted ICD-associated immunotherapy through direct ROS-based ER stress and exhibited enhanced anti-tumour efficacy. Reactive oxygen species induced by endoplasmic reitculum stress can be exploited for cancer therapy. Here, nanoparticles are targetted to the endoplasmic reticulum and, when accompanied by PDT, produce stress resulting in calreticulin exposure on the cell surface, which activates dendritic cells.
Collapse
|
159
|
Li X, Liu Z, Zhang A, Han C, Shen A, Jiang L, Boothman DA, Qiao J, Wang Y, Huang X, Fu YX. NQO1 targeting prodrug triggers innate sensing to overcome checkpoint blockade resistance. Nat Commun 2019; 10:3251. [PMID: 31324798 PMCID: PMC6642086 DOI: 10.1038/s41467-019-11238-1] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 07/03/2019] [Indexed: 12/31/2022] Open
Abstract
Lack of proper innate sensing inside tumor microenvironment (TME) limits T cell-targeted immunotherapy. NAD(P)H:quinone oxidoreductase 1 (NQO1) is highly enriched in multiple tumor types and has emerged as a promising target for direct tumor-killing. Here, we demonstrate that NQO1-targeting prodrug β-lapachone triggers tumor-selective innate sensing leading to T cell-dependent tumor control. β-Lapachone is catalyzed and bioactivated by NQO1 to generate ROS in NQO1high tumor cells triggering oxidative stress and release of the damage signals for innate sensing. β-Lapachone-induced high mobility group box 1 (HMGB1) release activates the host TLR4/MyD88/type I interferon pathway and Batf3 dendritic cell-dependent cross-priming to bridge innate and adaptive immune responses against the tumor. Furthermore, targeting NQO1 is very potent to trigger innate sensing for T cell re-activation to overcome checkpoint blockade resistance in well-established tumors. Our study reveals that targeting NQO1 potently triggers innate sensing within TME that synergizes with immunotherapy to overcome adaptive resistance. Improper innate sensing within the tumor microenvironment limits immunotherapy success. Here, the authors show that targeting NQO1 triggers immunogenic innate sensing to reactivate T cells and overcome immune checkpoint blockade resistance.
Collapse
Affiliation(s)
- Xiaoguang Li
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA.,School of Public Health, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Zhida Liu
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Anli Zhang
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Chuanhui Han
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Aijun Shen
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Lingxiang Jiang
- Department of Radiation Oncology, Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, 46022, USA
| | - David A Boothman
- Department of Biochemistry and Molecular Biology, Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Jian Qiao
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Yang Wang
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Xiumei Huang
- Department of Radiation Oncology, Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, 46022, USA.
| | - Yang-Xin Fu
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA.
| |
Collapse
|
160
|
Sprooten J, Ceusters J, Coosemans A, Agostinis P, De Vleeschouwer S, Zitvogel L, Kroemer G, Galluzzi L, Garg AD. Trial watch: dendritic cell vaccination for cancer immunotherapy. Oncoimmunology 2019; 8:e1638212. [PMID: 31646087 PMCID: PMC6791419 DOI: 10.1080/2162402x.2019.1638212] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 06/26/2019] [Indexed: 12/12/2022] Open
Abstract
Dendritic- cells (DCs) have received considerable attention as potential targets for the development of anticancer vaccines. DC-based anticancer vaccination relies on patient-derived DCs pulsed with a source of tumor-associated antigens (TAAs) in the context of standardized maturation-cocktails, followed by their reinfusion. Extensive evidence has confirmed that DC-based vaccines can generate TAA-specific, cytotoxic T cells. Nonetheless, clinical efficacy of DC-based vaccines remains suboptimal, reflecting the widespread immunosuppression within tumors. Thus, clinical interest is being refocused on DC-based vaccines as combinatorial partners for T cell-targeting immunotherapies. Here, we summarize the most recent preclinical/clinical development of anticancer DC vaccination and discuss future perspectives for DC-based vaccines in immuno-oncology.
Collapse
Affiliation(s)
- Jenny Sprooten
- Cell Death Research & Therapy (CDRT) unit, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Jolien Ceusters
- Department of Oncology, Laboratory of Tumor Immunology and Immunotherapy, ImmunOvar Research Group, KU Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - An Coosemans
- Department of Oncology, Laboratory of Tumor Immunology and Immunotherapy, ImmunOvar Research Group, KU Leuven, Leuven Cancer Institute, Leuven, Belgium
- Department of Gynecology and Obstetrics, UZ Leuven, Leuven, Belgium
| | - Patrizia Agostinis
- Cell Death Research & Therapy (CDRT) unit, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
- Center for Cancer Biology (CCB), VIB, Leuven, Belgium
| | - Steven De Vleeschouwer
- Research Group Experimental Neurosurgery and Neuroanatomy, KU Leuven, Leuven, Belgium
- Department of Neurosurgery, UZ Leuven, Leuven, Belgium
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- INSERM, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
- Université Paris Sud/Paris XI, Le Kremlin-Bicêtre, France
| | - Guido Kroemer
- Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, INSERM U1138, Centre de Recherche des Cordeliers, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
- Suzhou Institute for Systems Medicine, Chinese Academy of Sciences, Suzhou, China
- Department of Women’s and Children’s Health, Karolinska University Hospital, Stockholm, Sweden
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
- Department of Dermatology, Yale School of Medicine, New Haven, CT, USA
- Université de Paris Descartes, Paris, France
| | - Abhishek D. Garg
- Cell Death Research & Therapy (CDRT) unit, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
161
|
Chen KTJ, Gilabert-Oriol R, Bally MB, Leung AWY. Recent Treatment Advances and the Role of Nanotechnology, Combination Products, and Immunotherapy in Changing the Therapeutic Landscape of Acute Myeloid Leukemia. Pharm Res 2019; 36:125. [PMID: 31236772 PMCID: PMC6591181 DOI: 10.1007/s11095-019-2654-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Accepted: 06/01/2019] [Indexed: 12/17/2022]
Abstract
Acute myeloid leukemia (AML) is the most common acute leukemia that is becoming more prevalent particularly in the older (65 years of age or older) population. For decades, "7 + 3" remission induction therapy with cytarabine and an anthracycline, followed by consolidation therapy, has been the standard of care treatment for AML. This stagnancy in AML treatment has resulted in less than ideal treatment outcomes for AML patients, especially for elderly patients and those with unfavourable profiles. Over the past two years, six new therapeutic agents have received regulatory approval, suggesting that a number of obstacles to treating AML have been addressed and the treatment landscape for AML is finally changing. This review outlines the challenges and obstacles in treating AML and highlights the advances in AML treatment made in recent years, including Vyxeos®, midostaurin, gemtuzumab ozogamicin, and venetoclax, with particular emphasis on combination treatment strategies. We also discuss the potential utility of new combination products such as one that we call "EnFlaM", which comprises an encapsulated nanoformulation of flavopiridol and mitoxantrone. Finally, we provide a review on the immunotherapeutic landscape of AML, discussing yet another angle through which novel treatments can be designed to further improve treatment outcomes for AML patients.
Collapse
Affiliation(s)
- Kent T J Chen
- Department of Experimental Therapeutics, BC Cancer Research Centre, Vancouver, British Columbia, Canada
- Department of Interdisciplinary Oncology, BC Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Roger Gilabert-Oriol
- Department of Experimental Therapeutics, BC Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Marcel B Bally
- Department of Experimental Therapeutics, BC Cancer Research Centre, Vancouver, British Columbia, Canada.
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada.
- Cuprous Pharmaceuticals Inc., Vancouver, British Columbia, Canada.
- Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada.
| | - Ada W Y Leung
- Department of Experimental Therapeutics, BC Cancer Research Centre, Vancouver, British Columbia, Canada
- Cuprous Pharmaceuticals Inc., Vancouver, British Columbia, Canada
- Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
162
|
Sprooten J, Agostinis P, Garg AD. Type I interferons and dendritic cells in cancer immunotherapy. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 348:217-262. [PMID: 31810554 DOI: 10.1016/bs.ircmb.2019.06.001] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Type I interferons (IFNs) facilitate cancer immunosurveillance, antitumor immunity and antitumor efficacy of conventional cell death-inducing therapies (chemotherapy/radiotherapy) as well as immunotherapy. Moreover, it is clear that dendritic cells (DCs) play a significant role in aiding type I IFN-driven immunity. Owing to these antitumor properties several immunotherapies involving, or inducing, type I IFNs have received considerable clinical attention, e.g., recombinant IFNα2 or agonists targeting pattern recognition receptor (PRR) pathways like Toll-like receptors (TLRs), cGAS-STING or RIG-I/MDA5/MAVS. A series of preclinical and clinical evidence concurs that the success of anticancer therapy hinges on responsiveness of both cancer cells and DCs to type I IFNs. In this article, we discuss this link between type I IFNs and DCs in the context of cancer biology, with particular attention to mechanisms behind type I IFN production, their impact on DC driven anticancer immunity, and the implications of this for cancer immunotherapy, including DC-based vaccines.
Collapse
Affiliation(s)
- Jenny Sprooten
- Cell Death Research & Therapy (CDRT) Unit, Department for Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Patrizia Agostinis
- Cell Death Research & Therapy (CDRT) Unit, Department for Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium; Center for Cancer Biology (CCB), VIB, Leuven, Belgium
| | - Abhishek D Garg
- Cell Death Research & Therapy (CDRT) Unit, Department for Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium.
| |
Collapse
|
163
|
Fang S, Agostinis P, Salven P, Garg AD. Decoding cancer cell death-driven immune cell recruitment: An in vivo method for site-of-vaccination analyses. Methods Enzymol 2019; 636:185-207. [PMID: 32178819 DOI: 10.1016/bs.mie.2019.04.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Anticancer vaccines have recently received renewed attention for immunotherapy of at least a subset of cancer-types. Such vaccines mostly involve either killed cancer or tumor cells alone, or combinations thereof with specific (co-incubated) innate immune cells. In recent years, the immunogenic characteristics of the dead or dying cancer cells have emerged as decisive factors behind the success of anticancer vaccines. This has amplified the importance of accounting for immunology of cell death while preparing anticancer vaccines. This, in turn, has increased the emphasis on the immune reactions at the site-of-vaccination since the therapeutic efficacy of the killed cancer/tumor cell vaccines is contingent upon the nature and characteristics of these reactions at the site-of-injection. In this article, we present a systematic methodology that exploits the murine ear pinna model to study differential immune cell recruitment by dead/dying cancer cells injected in vivo, thereby modeling the site-of-injection relevant for anticancer vaccines.
Collapse
Affiliation(s)
- Shentong Fang
- Wihuri Research Institute and Translational Cancer Medicine, University of Helsinki, Helsinki, Finland
| | - Patrizia Agostinis
- Department for Cellular and Molecular Medicine, Cell Death Research & Therapy (CDRT) Unit, KU Leuven, Leuven, Belgium; Center for Cancer Biology (CCB), VIB, Leuven, Belgium
| | - Petri Salven
- Department of Pathology, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Abhishek D Garg
- Department for Cellular and Molecular Medicine, Cell Death Research & Therapy (CDRT) Unit, KU Leuven, Leuven, Belgium.
| |
Collapse
|
164
|
Aznar MA, Planelles L, Perez-Olivares M, Molina C, Garasa S, Etxeberría I, Perez G, Rodriguez I, Bolaños E, Lopez-Casas P, Rodriguez-Ruiz ME, Perez-Gracia JL, Marquez-Rodas I, Teijeira A, Quintero M, Melero I. Immunotherapeutic effects of intratumoral nanoplexed poly I:C. J Immunother Cancer 2019; 7:116. [PMID: 31046839 PMCID: PMC6498680 DOI: 10.1186/s40425-019-0568-2] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 03/15/2019] [Indexed: 12/30/2022] Open
Abstract
Poly I:C is a powerful immune adjuvant as a result of its agonist activities on TLR-3, MDA5 and RIG-I. BO-112 is a nanoplexed formulation of Poly I:C complexed with polyethylenimine that causes tumor cell apoptosis showing immunogenic cell death features and which upon intratumoral release results in more prominent tumor infiltration by T lymphocytes. Intratumoral treatment with BO-112 of subcutaneous tumors derived from MC38, 4 T1 and B16-F10 leads to remarkable local disease control dependent on type-1 interferon and gamma-interferon. Some degree of control of non-injected tumor lesions following BO-112 intratumoral treatment was found in mice bearing bilateral B16-OVA melanomas, an activity which was enhanced with co-treatment with systemic anti-CD137 and anti-PD-L1 mAbs. More abundant CD8+ T lymphocytes were found in B16-OVA tumor-draining lymph nodes and in the tumor microenvironment following intratumoral BO-112 treatment, with enhanced numbers of tumor antigen-specific cytotoxic T lymphocytes. Genome-wide transcriptome analyses of injected tumor lesions were consistent with a marked upregulation of the type-I interferon pathway. Inspired by these data, intratumorally delivered BO-112 is being tested in cancer patients (NCT02828098).
Collapse
Affiliation(s)
- M Angela Aznar
- Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pio XII, 55, 31008, Pamplona, Spain.
| | | | | | - Carmen Molina
- Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pio XII, 55, 31008, Pamplona, Spain
| | - Saray Garasa
- Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pio XII, 55, 31008, Pamplona, Spain
| | - Iñaki Etxeberría
- Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pio XII, 55, 31008, Pamplona, Spain
| | - Guiomar Perez
- Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pio XII, 55, 31008, Pamplona, Spain
| | - Inmaculada Rodriguez
- Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pio XII, 55, 31008, Pamplona, Spain
| | - Elixabet Bolaños
- Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pio XII, 55, 31008, Pamplona, Spain
| | | | - Maria E Rodriguez-Ruiz
- Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pio XII, 55, 31008, Pamplona, Spain
| | - Jose L Perez-Gracia
- Clínica Universidad de Navarra, Pamplona, Spain.,CIBERONC, Madrid, Spain.,IDISNA, Instituto de investigación de Navarra, Pamplona, Spain
| | - Ivan Marquez-Rodas
- Medical Oncology Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Alvaro Teijeira
- Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pio XII, 55, 31008, Pamplona, Spain.,CIBERONC, Madrid, Spain.,IDISNA, Instituto de investigación de Navarra, Pamplona, Spain
| | | | - Ignacio Melero
- Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pio XII, 55, 31008, Pamplona, Spain. .,Clínica Universidad de Navarra, Pamplona, Spain. .,CIBERONC, Madrid, Spain. .,IDISNA, Instituto de investigación de Navarra, Pamplona, Spain.
| |
Collapse
|
165
|
Kepp O, Sauvat A, Leduc M, Forveille S, Liu P, Zhao L, Bezu L, Xie W, Zitvogel L, Kroemer G. A fluorescent biosensor-based platform for the discovery of immunogenic cancer cell death inducers. Oncoimmunology 2019; 8:1606665. [PMID: 31413915 DOI: 10.1080/2162402x.2019.1606665] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 04/09/2019] [Accepted: 04/09/2019] [Indexed: 12/15/2022] Open
Abstract
Systemic anticancer immunity can be reinstated via the induction of immunogenic cell death (ICD) in malignant cells. Thus, certain classes of cytotoxic compounds, for example, anthracyclines, oxaliplatin and taxanes are endowed with the capacity to act on cancer cells to ignite premortem stress pathways that lead to the surface exposure of calreticulin (CALR) and the cellular release of adenosine triphosphate, annexin A1, high mobility group B1 and type-1 interferons. Altogether, these alterations constitute the hallmarks of ICD. Here we report the design of a discovery pipeline for the identification of novel ICD inducers by means of a phenotypic screening platform. The use of fluorescent biosensors as proxies for the manifestation of ICD hallmarks has enabled the exploration of large collections of chemical compounds by automatized screening routines. Imaging-based assessment and phenotypic selection led to the identification of potential ICD inducers that could be validated further in vitro and in vivo, confirming that bona fide ICD inducers possess the capacity to induce immunological long-term memory and to confer resistance against rechallenge with syngeneic tumors. Machine learning algorithms analyzing the physicochemical properties of ICD inducers can assist in the preselection of compounds with potential ICD-stimulatory properties, further accelerating the screening efforts designed to develop new immunotherapeutic agents.
Collapse
Affiliation(s)
- Oliver Kepp
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Université Paris Descartes, Paris, France.,Université Pierre et Marie Curie, Paris, France.,Faculty of Medicine, University of Paris Sud, Kremlin-Bicêtre, France
| | - Allan Sauvat
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Université Paris Descartes, Paris, France.,Université Pierre et Marie Curie, Paris, France.,Faculty of Medicine, University of Paris Sud, Kremlin-Bicêtre, France
| | - Marion Leduc
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Université Paris Descartes, Paris, France.,Université Pierre et Marie Curie, Paris, France.,Faculty of Medicine, University of Paris Sud, Kremlin-Bicêtre, France
| | - Sabrina Forveille
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Université Paris Descartes, Paris, France.,Université Pierre et Marie Curie, Paris, France.,Faculty of Medicine, University of Paris Sud, Kremlin-Bicêtre, France
| | - Peng Liu
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Université Paris Descartes, Paris, France.,Université Pierre et Marie Curie, Paris, France.,Faculty of Medicine, University of Paris Sud, Kremlin-Bicêtre, France
| | - Liwei Zhao
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Université Paris Descartes, Paris, France.,Université Pierre et Marie Curie, Paris, France.,Faculty of Medicine, University of Paris Sud, Kremlin-Bicêtre, France
| | - Lucillia Bezu
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Université Paris Descartes, Paris, France.,Université Pierre et Marie Curie, Paris, France.,Faculty of Medicine, University of Paris Sud, Kremlin-Bicêtre, France.,Service anesthésie-réanimation, Hôpital Européen Georges Pompidou, Paris, France
| | - Wei Xie
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Université Paris Descartes, Paris, France.,Université Pierre et Marie Curie, Paris, France.,Faculty of Medicine, University of Paris Sud, Kremlin-Bicêtre, France
| | - Laurence Zitvogel
- Faculty of Medicine, University of Paris Sud, Kremlin-Bicêtre, France.,Institut de Cancérologie, Gustave Roussy Cancer Campus (GRCC), Villejuif, France.,INSERM, Villejuif, France.,Center of Clinical Investigations, Villejuif, France
| | - Guido Kroemer
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Université Paris Descartes, Paris, France.,Université Pierre et Marie Curie, Paris, France.,Faculty of Medicine, University of Paris Sud, Kremlin-Bicêtre, France.,Pôle de Biologie, Hôpital Européen Georges Pompidou, Paris, France.,Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
166
|
Gao J, Deng F, Jia W. Inhibition of Indoleamine 2,3-Dioxygenase Enhances the Therapeutic Efficacy of Immunogenic Chemotherapeutics in Breast Cancer. J Breast Cancer 2019; 22:196-209. [PMID: 31281723 PMCID: PMC6597411 DOI: 10.4048/jbc.2019.22.e23] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 04/12/2019] [Indexed: 01/19/2023] Open
Abstract
Purpose Breast cancer has become a major public health threat in the current society. Anthracycline doxorubicin (DOX) is a widely used drug in breast cancer chemotherapy. We aimed to investigate the immunogenic death of breast tumor cells caused by DOX, and detect the effects of combination of DOX and a small molecule inhibitor in tumor engrafted mouse model. Methods We used 4T1 breast cancer cells to examine the anthracycline DOX-mediated immunogenic death of breast tumor cells by assessing the calreticulin exposure and adenosine triphosphate and high mobility group box 1 release. Using 4T1 tumor cell-engrafted mouse model, we also detected the expression of indoleamine 2,3-dioxygenase (IDO) in tumor tissues after DOX treatment and further explored whether the specific small molecule IDO1 inhibitor NLG919 combined with DOX, can exhibit better therapeutic effects on breast cancer. Results DOX induced immunogenic cell death of murine breast cancer cells 4T1 as well as the upregulation of IDO1. We also found that treatment with NLG919 enhanced kynurenine inhibition in a dose-dependent manner. IDO1 inhibition reversed CD8+ T cell suppression mediated by IDO-expressing 4T1 murine breast cancer cells. Compared to the single agent or control, combination of DOX and NLG919 significantly inhibited the tumor growth, indicating that the 2 drugs exhibit synergistic effect. The combination therapy also increased the expression of transforming growth factor-β, while lowering the expressions of interleukin-12p70 and interferon-γ. Conclusion Compared to single agent therapy, combination of NLG919 with DOX demonstrated better therapeutic effects in 4T1 murine breast tumor model. IDO inhibition by NLG919 enhanced the therapeutic efficacy of DOX in breast cancer, achieving synergistic effect.
Collapse
Affiliation(s)
- Jian Gao
- Department of Clinical Medicine, Clinical Medical College of Shandong University, Jinan, China.,Department of General Surgery, the First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Fusheng Deng
- Department of General Surgery, the First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Weidong Jia
- Department of Hepatic Surgery, the First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
167
|
Stress responses in stromal cells and tumor homeostasis. Pharmacol Ther 2019; 200:55-68. [PMID: 30998941 DOI: 10.1016/j.pharmthera.2019.04.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 04/10/2019] [Indexed: 02/07/2023]
Abstract
In most (if not all) solid tumors, malignant cells are outnumbered by their non-malignant counterparts, including immune, endothelial and stromal cells. However, while the mechanisms whereby cancer cells adapt to microenvironmental perturbations have been studied in great detail, relatively little is known on stress responses in non-malignant compartments of the tumor microenvironment. Here, we discuss the mechanisms whereby cancer-associated fibroblasts and other cellular components of the tumor stroma react to stress in the context of an intimate crosstalk with malignant, endothelial and immune cells, and how such crosstalk influences disease progression and response to treatment.
Collapse
|
168
|
Serrano-Del Valle A, Anel A, Naval J, Marzo I. Immunogenic Cell Death and Immunotherapy of Multiple Myeloma. Front Cell Dev Biol 2019; 7:50. [PMID: 31041312 PMCID: PMC6476910 DOI: 10.3389/fcell.2019.00050] [Citation(s) in RCA: 135] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 03/19/2019] [Indexed: 12/24/2022] Open
Abstract
Over the past decades, immunotherapy has demonstrated a prominent clinical efficacy in a wide variety of human tumors. For many years, apoptosis has been considered a non-immunogenic or tolerogenic process whereas necrosis or necroptosis has long been acknowledged to play a key role in inflammation and immune-related processes. However, the new concept of “immunogenic cell death” (ICD) has challenged this traditional view and has granted apoptosis with immunogenic abilities. This paradigm shift offers clear implications in designing novel anti-cancer therapeutic approaches. To date, several screening studies have been carried out to discover bona fide ICD inducers and reveal the inherent capacity of a wide variety of drugs to induce cell death-associated exposure of danger signals and to bring about in vivo anti-cancer immune responses. Recent shreds of evidence place ER stress at the core of all the scenarios where ICD occur. Furthermore, ER stress and the unfolded protein response (UPR) have emerged as important targets in different human cancers. Notably, in multiple myeloma (MM), a lethal plasma cell disorder, the elevated production of immunoglobulins leaves these cells heavily reliant on the survival arm of the UPR. For that reason, drugs that disrupt ER homeostasis and engage ER stress-associated cell death, such as proteasome inhibitors, which are currently used for the treatment of MM, as well as novel ER stressors are intended to be promising therapeutic agents in MM. This not only holds true for their capacity to induce cell death, but also to their potential ability to activate the immunogenic arm of the ER stress response, with the ensuing exposure of danger signals. We provide here an overview of the up-to-date knowledge regarding the cell death mechanisms involved in situations of ER stress with a special focus on the connections with the drug-induced ER stress pathways that evoke ICD. We will also discuss how this could assist in optimizing and developing better immunotherapeutic approaches, especially in MM treatment.
Collapse
Affiliation(s)
| | - Alberto Anel
- Department of Biochemistry and Molecular and Cell Biology, University of Zaragoza, Zaragoza, Spain
| | - Javier Naval
- Department of Biochemistry and Molecular and Cell Biology, University of Zaragoza, Zaragoza, Spain
| | - Isabel Marzo
- Department of Biochemistry and Molecular and Cell Biology, University of Zaragoza, Zaragoza, Spain
| |
Collapse
|
169
|
Sorafenib alone vs. sorafenib plus GEMOX as 1 st-line treatment for advanced HCC: the phase II randomised PRODIGE 10 trial. Br J Cancer 2019; 120:896-902. [PMID: 30944458 PMCID: PMC6734663 DOI: 10.1038/s41416-019-0443-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 03/08/2019] [Accepted: 03/14/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Sorafenib remains one major first-line therapeutic options for advanced hepatocellular carcinoma (aHCC), with modest efficacy. We investigated the addition of gemcitabine and oxaliplatin (GEMOX) to sorafenib in aHCC patients. METHODS Our multicentre phase II trial randomised aHCC first-line patients to sorafenib (400 mg BID) or sorafenib-GEMOX every 2 weeks (1000 mg/m2 gemcitabine; 100 mg/m2 oxaliplatin). Primary endpoint was the 4-month progression-free survival (PFS) rate. RESULTS Ninety-four patients were randomised (sorafenib-GEMOX: n = 48; sorafenib: n = 46). Median age was 64 years, PS 0 (69%) or 1 (31%), 63% patients had cirrhosis, 29% portal vein thrombosis and 70% extra-hepatic disease. Median duration of sorafenib treatment was 4 months (1-51); median number of GEMOX cycles was 7 (1-16). The 4-month PFS rates were 64% and 61% in the sorafenib-GEMOX and sorafenib arms, respectively; median PFS and OS were 6.2 (95% CI: 3.8-6.8) and 13.5 (7.5-16.2) months, and 4.6 (3.9-6.2) months and 14.8 (12.2-22.2), respectively. The ORR/DCR were 9%/70% and 15%/77% in the sorafenib-GEMOX and sorafenib alone arms, respectively. Main toxicities were (sorafenib-GEMOX/sorafenib) neutropenia (23%/0), thrombocytopenia (33%/0), diarrhoea (18%/9), peripheral neuropathy (5%/0) and hand-foot syndrome (5%/18). CONCLUSIONS Addition of GEMOX had an inpact on ORR and was well-tolerated as frontline systemic therapy. The benefit on PFS seems moderate; no subsequent study was planned.
Collapse
|
170
|
Lévesque S, Pol JG, Ferrere G, Galluzzi L, Zitvogel L, Kroemer G. Trial watch: dietary interventions for cancer therapy. Oncoimmunology 2019; 8:1591878. [PMID: 31143510 PMCID: PMC6527263 DOI: 10.1080/2162402x.2019.1591878] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 03/05/2019] [Indexed: 12/30/2022] Open
Abstract
Dietary interventions have a profound impact on whole body metabolism, including oncometabolism (the metabolic features allowing cancer cells to proliferate) and immunometabolism (the catabolic and anabolic reactions that regulate immune responses). Recent preclinical studies demonstrated that multiple dietary changes can improve anticancer immunosurveillance of chemo-, radio- and immunotherapy. These findings have fostered the design of clinical trials evaluating the capacity of dietary interventions to synergize with treatment and hence limit tumor progression. Here, we discuss the scientific rationale for harnessing dietary interventions to improve the efficacy of anticancer therapy and present up-to-date information on clinical trials currently investigating this possibility.
Collapse
Affiliation(s)
- Sarah Lévesque
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,INSERM, U1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Université Paris-Saclay, Orsay, France.,Fondation pour la Recherche Médicale, Paris, France
| | - Jonathan G Pol
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,INSERM, U1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France
| | - Gladys Ferrere
- INSERM U1015, Villejuif, France.,CICBT507, Villejuif, France
| | - Lorenzo Galluzzi
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France.,Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Sandra and Edward Meyer Cancer Center, New York, NY, USA.,Department of Dermatology, Yale School of Medicine, New Haven, CT, USA
| | - Laurence Zitvogel
- Université Paris-Saclay, Orsay, France.,INSERM U1015, Villejuif, France.,CICBT507, Villejuif, France
| | - Guido Kroemer
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,INSERM, U1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Université Paris-Saclay, Orsay, France.,Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France.,Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.,Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
171
|
Davola ME, Mossman KL. Oncolytic viruses: how "lytic" must they be for therapeutic efficacy? Oncoimmunology 2019; 8:e1581528. [PMID: 31069150 PMCID: PMC6492965 DOI: 10.1080/2162402x.2019.1596006] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 02/22/2019] [Accepted: 03/07/2019] [Indexed: 12/21/2022] Open
Abstract
Oncolytic viruses (OVs) preferentially target and kill cancer cells without affecting healthy cells through a multi-modal mechanism of action. While historically the direct killing activity of OVs was considered the primary mode of action, initiation or augmentation of a host antitumor immune response is now considered an essential aspect of oncolytic virotherapy. To improve oncolytic virotherapy, many studies focus on increasing virus replication and spread. In this article, we open for discussion the traditional dogma that correlates replication with the efficacy of OVs, pointing out several examples that oppose this principle.
Collapse
Affiliation(s)
- Maria Eugenia Davola
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Michael DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Karen Louise Mossman
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Michael DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
172
|
Hof J, Kok K, Sijmons RH, de Jong KP. Systematic Review of the Prognostic Role of the Immune System After Surgery of Colorectal Liver Metastases. Front Oncol 2019; 9:148. [PMID: 30941301 PMCID: PMC6433783 DOI: 10.3389/fonc.2019.00148] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 02/21/2019] [Indexed: 12/11/2022] Open
Abstract
Background: The current prognostication of patient survival after surgery for colorectal liver metastases is based on clinical characteristics, but low accuracy makes it difficult to guide treatment for the individual patient. Rapidly evolving technologies have led to the expectation that biomarkers will be able to outperform the current clinical scoring systems and provide more effective personalised treatment. Two main topics prevail in cancer treatment, namely the role of the immune system and the prediction and prognostication by application of high-throughput methodology. The aim of this review is to examine the evidence for prognostic immunological and molecular markers studied in tumour tissue obtained at surgical resection for colorectal liver metastases. Methods: First we analysed immunophenotypical protein markers, that are mainly studied by immunohistochemistry. Second, we review molecular markers by analysing high-throughput studies on tumour mRNA and microRNA expression. Results: CD3, CD4, and CD8 are the most frequently studied protein markers. High intra-tumoural CD3+ T cell infiltration and low CXCR4 expression have the best association with favourable patient survival. Studies that analysed microRNA or mRNA expression data showed very little overlap in prognostic genes. Conclusions: Patient prognostication after surgery for colorectal liver metastases by analysing the immune system remains difficult. Current data are based on diverse and heterogeneous patient populations which prohibits drawing firm conclusions.
Collapse
Affiliation(s)
- Joost Hof
- Department of Hepato-Pancreato-Biliary Surgery and Liver Transplantation, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.,Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Klaas Kok
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Rolf H Sijmons
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Koert P de Jong
- Department of Hepato-Pancreato-Biliary Surgery and Liver Transplantation, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
173
|
Rapoport BL, Anderson R. Realizing the Clinical Potential of Immunogenic Cell Death in Cancer Chemotherapy and Radiotherapy. Int J Mol Sci 2019; 20:ijms20040959. [PMID: 30813267 PMCID: PMC6412296 DOI: 10.3390/ijms20040959] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 02/05/2019] [Accepted: 02/06/2019] [Indexed: 02/06/2023] Open
Abstract
Immunogenic cell death (ICD), which is triggered by exposure of tumor cells to a limited range of anticancer drugs, radiotherapy, and photodynamic therapy, represents a recent innovation in the revitalized and burgeoning field of oncoimmunnotherapy. ICD results in the cellular redistribution and extracellular release of damage-associated molecular patterns (DAMPs), which have the potential to activate and restore tumor-targeted immune responses. Although a convincing body of evidence exists with respect to the antitumor efficacy of ICD in various experimental systems, especially murine models of experimental anticancer immunotherapy, evidence for the existence of ICD in the clinical setting is less compelling. Following overviews of hallmark developments, which have sparked the revival of interest in the field of oncoimmunotherapy, types of tumor cell death and the various DAMPs most prominently involved in the activation of antitumor immune responses, the remainder of this review is focused on strategies which may potentiate ICD in the clinical setting. These include identification of tumor- and host-related factors predictive of the efficacy of ICD, the clinical utility of combinatorial immunotherapeutic strategies, novel small molecule inducers of ICD, novel and repurposed small molecule immunostimulants, as well as the critical requirement for validated biomarkers in predicting the efficacy of ICD.
Collapse
Affiliation(s)
- Bernardo L Rapoport
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa.
- The Medical Oncology Centre of Rosebank, Johannesburg 2196, South Africa.
| | - Ronald Anderson
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa.
- Institute for Cellular and Molecular Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa.
| |
Collapse
|
174
|
Mutational and Antigenic Landscape in Tumor Progression and Cancer Immunotherapy. Trends Cell Biol 2019; 29:396-416. [PMID: 30765144 DOI: 10.1016/j.tcb.2019.01.003] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 01/04/2019] [Accepted: 01/08/2019] [Indexed: 12/18/2022]
Abstract
Evolving neoplasms accumulate non-synonymous mutations at a high rate, potentially enabling the expression of antigenic epitopes that can be recognized by the immune system. Since they are not covered by central tolerance, such tumor neoantigens (TNAs) should be under robust immune control as they surge. However, genetic defects that impair cancer cell eradication by the immune system coupled with the establishment of local immunosuppression can enable TNA accumulation, which is generally associated with improved clinical sensitivity to various immunotherapies. Here, we explore how tumor-intrinsic factors and immunological processes shape the mutational and antigenic landscape of evolving neoplasms to influence clinical responses to immunotherapy, and propose strategies to achieve robust immunological control of the disease despite disabled immunosurveillance.
Collapse
|
175
|
Rodríguez-Ruiz ME, Rodríguez I, Mayorga L, Labiano T, Barbes B, Etxeberria I, Ponz-Sarvise M, Azpilikueta A, Bolaños E, Sanmamed MF, Berraondo P, Calvo FA, Barcelos-Hoff MH, Perez-Gracia JL, Melero I. TGFβ Blockade Enhances Radiotherapy Abscopal Efficacy Effects in Combination with Anti-PD1 and Anti-CD137 Immunostimulatory Monoclonal Antibodies. Mol Cancer Ther 2019; 18:621-631. [PMID: 30683810 DOI: 10.1158/1535-7163.mct-18-0558] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 11/07/2018] [Accepted: 01/15/2019] [Indexed: 12/23/2022]
Abstract
Radiotherapy can be synergistically combined with immunotherapy in mouse models, extending its efficacious effects outside of the irradiated field (abscopal effects). We previously reported that a regimen encompassing local radiotherapy in combination with anti-CD137 plus anti-PD-1 mAbs achieves potent abscopal effects against syngeneic transplanted murine tumors up to a certain tumor size. Knowing that TGFβ expression or activation increases in irradiated tissues, we tested whether TGFβ blockade may further enhance abscopal effects in conjunction with the anti-PD-1 plus anti-CD137 mAb combination. Indeed, TGFβ blockade with 1D11, a TGFβ-neutralizing mAb, markedly enhanced abscopal effects and overall treatment efficacy against subcutaneous tumors of either 4T1 breast cancer cells or large MC38 colorectal tumors. Increases in CD8 T cells infiltrating the nonirradiated lesion were documented upon combined treatment, which intensely expressed Granzyme-B as an indicator of cytotoxic effector capability. Interestingly, tumor tissue but not healthy tissue irradiation results in the presence of higher concentrations of TGFβ in the nonirradiated contralateral tumor that showed smad2/3 phosphorylation increases in infiltrating CD8 T cells. In conclusion, radiotherapy-induced TGFβ hampers abscopal efficacy even upon combination with a potent immunotherapy regimen. Therefore, TGFβ blockade in combination with radioimmunotherapy results in greater efficacy.
Collapse
Affiliation(s)
- María E Rodríguez-Ruiz
- Department of Oncology, University of Navarra and Instituto de Investigación Sanitaria de Navarra (IdISNA), Pamplona, Spain. .,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Inmaculada Rodríguez
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Department of Cellular Therapy, University of Navarra and Instituto de Investigación Sanitaria de Navarra (IdISNA), Pamplona, Spain
| | - Lina Mayorga
- Department of Oncology, University of Navarra and Instituto de Investigación Sanitaria de Navarra (IdISNA), Pamplona, Spain
| | - Tania Labiano
- Department of Oncology, Complejo Hospitalario de Navarra, Pamplona, Spain
| | - Benigno Barbes
- Department of Oncology, University of Navarra and Instituto de Investigación Sanitaria de Navarra (IdISNA), Pamplona, Spain
| | - Iñaki Etxeberria
- Division of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra and Instituto de Investigación Sanitaria de Navarra (IdISNA), Pamplona, Spain
| | - Mariano Ponz-Sarvise
- Department of Oncology, University of Navarra and Instituto de Investigación Sanitaria de Navarra (IdISNA), Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Arantza Azpilikueta
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Division of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra and Instituto de Investigación Sanitaria de Navarra (IdISNA), Pamplona, Spain
| | - Elixabet Bolaños
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Division of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra and Instituto de Investigación Sanitaria de Navarra (IdISNA), Pamplona, Spain
| | - Miguel F Sanmamed
- Department of Oncology, University of Navarra and Instituto de Investigación Sanitaria de Navarra (IdISNA), Pamplona, Spain
| | - Pedro Berraondo
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Division of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra and Instituto de Investigación Sanitaria de Navarra (IdISNA), Pamplona, Spain
| | - Felipe A Calvo
- Department of Oncology, University of Navarra and Instituto de Investigación Sanitaria de Navarra (IdISNA), Pamplona, Spain
| | - Mary Helen Barcelos-Hoff
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, California
| | - Jose L Perez-Gracia
- Department of Oncology, University of Navarra and Instituto de Investigación Sanitaria de Navarra (IdISNA), Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Ignacio Melero
- Department of Oncology, University of Navarra and Instituto de Investigación Sanitaria de Navarra (IdISNA), Pamplona, Spain. .,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Division of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra and Instituto de Investigación Sanitaria de Navarra (IdISNA), Pamplona, Spain
| |
Collapse
|
176
|
Kenny RG, Marmion CJ. Toward Multi-Targeted Platinum and Ruthenium Drugs-A New Paradigm in Cancer Drug Treatment Regimens? Chem Rev 2019; 119:1058-1137. [PMID: 30640441 DOI: 10.1021/acs.chemrev.8b00271] [Citation(s) in RCA: 433] [Impact Index Per Article: 72.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
While medicinal inorganic chemistry has been practised for over 5000 years, it was not until the late 1800s when Alfred Werner published his ground-breaking research on coordination chemistry that we began to truly understand the nature of the coordination bond and the structures and stereochemistries of metal complexes. We can now readily manipulate and fine-tune their properties. This had led to a multitude of complexes with wide-ranging biomedical applications. This review will focus on the use and potential of metal complexes as important therapeutic agents for the treatment of cancer. With major advances in technologies and a deeper understanding of the human genome, we are now in a strong position to more fully understand carcinogenesis at a molecular level. We can now also rationally design and develop drug molecules that can either selectively enhance or disrupt key biological processes and, in doing so, optimize their therapeutic potential. This has heralded a new era in drug design in which we are moving from a single- toward a multitargeted approach. This approach lies at the very heart of medicinal inorganic chemistry. In this review, we have endeavored to showcase how a "multitargeted" approach to drug design has led to new families of metallodrugs which may not only reduce systemic toxicities associated with modern day chemotherapeutics but also address resistance issues that are plaguing many chemotherapeutic regimens. We have focused our attention on metallodrugs incorporating platinum and ruthenium ions given that complexes containing these metal ions are already in clinical use or have advanced to clinical trials as anticancer agents. The "multitargeted" complexes described herein not only target DNA but also contain either vectors to enable them to target cancer cells selectively and/or moieties that target enzymes, peptides, and intracellular proteins. Multitargeted complexes which have been designed to target the mitochondria or complexes inspired by natural product activity are also described. A summary of advances in this field over the past decade or so will be provided.
Collapse
Affiliation(s)
- Reece G Kenny
- Centre for Synthesis and Chemical Biology, Department of Chemistry , Royal College of Surgeons in Ireland , 123 St. Stephen's Green , Dublin 2 , Ireland
| | - Celine J Marmion
- Centre for Synthesis and Chemical Biology, Department of Chemistry , Royal College of Surgeons in Ireland , 123 St. Stephen's Green , Dublin 2 , Ireland
| |
Collapse
|
177
|
Lhuillier C, Galluzzi L. Preface: Dendritic cells: Master regulators of innate and adaptive immunity. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 349:xi-xvi. [PMID: 31759435 DOI: 10.1016/s1937-6448(19)30114-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Claire Lhuillier
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States.
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States; Sandra and Edward Meyer Cancer Center, New York, NY, United States; Department of Dermatology, Yale School of Medicine, New Haven, CT, United States; Université de Paris, Paris, France.
| |
Collapse
|
178
|
Qin J, Kunda NM, Qiao G, Tulla K, Prabhakar BS, Maker AV. Vaccination With Mitoxantrone-Treated Primary Colon Cancer Cells Enhances Tumor-Infiltrating Lymphocytes and Clinical Responses in Colorectal Liver Metastases. J Surg Res 2019; 233:57-64. [DOI: 10.1016/j.jss.2018.07.068] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 07/13/2018] [Accepted: 07/19/2018] [Indexed: 12/21/2022]
|
179
|
Lhuillier C, Galluzzi L. Preface-Dendritic cells: Master regulators of innate and adaptive immunity. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 348:ix-xiv. [PMID: 31810557 DOI: 10.1016/s1937-6448(19)30095-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Claire Lhuillier
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States.
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States; Sandra and Edward Meyer Cancer Center, New York, NY, United States; Department of Dermatology, Yale School of Medicine, New Haven, CT, United States; Université Paris Descartes, Paris, France.
| |
Collapse
|
180
|
Uscanga-Palomeque AC, Calvillo-Rodríguez KM, Gómez-Morales L, Lardé E, Denèfle T, Caballero-Hernández D, Merle-Béral H, Susin SA, Karoyan P, Martínez-Torres AC, Rodríguez-Padilla C. CD47 agonist peptide PKHB1 induces immunogenic cell death in T-cell acute lymphoblastic leukemia cells. Cancer Sci 2018; 110:256-268. [PMID: 30460757 PMCID: PMC6317946 DOI: 10.1111/cas.13885] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 10/19/2018] [Accepted: 11/08/2018] [Indexed: 12/22/2022] Open
Abstract
T‐cell acute lymphoblastic leukemia (T‐ALL) has a poor prognosis derived from its genetic heterogeneity, which translates to a high chemoresistance. Recently, our workgroup designed thrombospondin‐1‐derived CD47 agonist peptides and demonstrated their ability to induce cell death in chronic lymphocytic leukemia. Encouraged by these promising results, we evaluated cell death induced by PKHB1 (the first‐described serum‐stable CD47‐agonist peptide) on CEM and MOLT‐4 human cell lines (T‐ALL) and on one T‐murine tumor lymphoblast cell‐line (L5178Y‐R), also assessing caspase and calcium dependency and mitochondrial membrane potential. Additionally, we evaluated selectivity for cancer cell lines by analyzing cell death and viability of human and murine non‐tumor cells after CD47 activation. In vivo, we determined that PKHB1‐treatment in mice bearing the L5178Y‐R cell line increased leukocyte cell count in peripheral blood and lymphoid organs while recruiting leukocytes to the tumor site. To analyze whether CD47 activation induced immunogenic cell death (ICD), we evaluated damage‐associated molecular patterns (DAMP) exposure (calreticulin, CRT) and release (ATP, heat shock proteins 70 and 90, high‐mobility group box 1, CRT). Furthermore, we gave prophylactic antitumor vaccination, determining immunological memory. Our data indicate that PKHB1 induces caspase‐independent and calcium‐dependent cell death in leukemic cells while sparing non‐tumor murine and human cells. Moreover, our results show that PKHB1 can induce ICD in leukemic cells as it induces CRT exposure and DAMP release in vitro, and prophylactic vaccinations inhibit tumor establishment in vivo. Together, our results improve the knowledge of CD47 agonist peptides potential as therapeutic tools to treat leukemia.
Collapse
Affiliation(s)
| | - Kenny Misael Calvillo-Rodríguez
- College of Biology Science, Laboratory of Immunology and Virology, Autonomus University of Nuevo Leon, San Nicolas de los Garza, Mexico
| | - Luis Gómez-Morales
- College of Biology Science, Laboratory of Immunology and Virology, Autonomus University of Nuevo Leon, San Nicolas de los Garza, Mexico
| | - Eva Lardé
- CNRS, Biomolecules Laboratory, Superior Normal School, PSL University, Sorbonne University, Paris, France
| | - Thomas Denèfle
- CNRS, Biomolecules Laboratory, Superior Normal School, PSL University, Sorbonne University, Paris, France
| | - Diana Caballero-Hernández
- College of Biology Science, Laboratory of Immunology and Virology, Autonomus University of Nuevo Leon, San Nicolas de los Garza, Mexico
| | - Hélène Merle-Béral
- INSERM, UMRS 1138, Sorbonne University, University of Paris Descartes, Sorbonne Paris Cite, Center of Reserch of Cordeliers, Paris, France
| | - Santos A Susin
- INSERM, UMRS 1138, Sorbonne University, University of Paris Descartes, Sorbonne Paris Cite, Center of Reserch of Cordeliers, Paris, France
| | - Philippe Karoyan
- CNRS, Biomolecules Laboratory, Superior Normal School, PSL University, Sorbonne University, Paris, France
| | - Ana Carolina Martínez-Torres
- College of Biology Science, Laboratory of Immunology and Virology, Autonomus University of Nuevo Leon, San Nicolas de los Garza, Mexico
| | - Cristina Rodríguez-Padilla
- College of Biology Science, Laboratory of Immunology and Virology, Autonomus University of Nuevo Leon, San Nicolas de los Garza, Mexico
| |
Collapse
|
181
|
Wang SJ, Haffty B. Radiotherapy as a New Player in Immuno-Oncology. Cancers (Basel) 2018; 10:cancers10120515. [PMID: 30558196 PMCID: PMC6315809 DOI: 10.3390/cancers10120515] [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: 11/07/2018] [Revised: 12/10/2018] [Accepted: 12/11/2018] [Indexed: 12/14/2022] Open
Abstract
Recent development in radiation biology has revealed potent immunogenic properties of radiotherapy in cancer treatments. However, antitumor immune effects of radiotherapy are limited by the concomitant induction of radiation-dependent immunosuppressive effects. In the growing era of immunotherapy, combining radiotherapy with immunomodulating agents has demonstrated enhancement of radiation-induced antitumor immune activation that correlated with improved treatment outcomes. Yet, how to optimally deliver combination therapy regarding dose-fractionation and timing of radiotherapy is largely unknown. Future prospective testing to fine-tune this promising combination of radiotherapy and immunotherapy is warranted.
Collapse
Affiliation(s)
- Shang-Jui Wang
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, 195 Little Albany St., New Brunswick, NJ 08901, USA.
| | - Bruce Haffty
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, 195 Little Albany St., New Brunswick, NJ 08901, USA.
| |
Collapse
|
182
|
Iribarren K, Buque A, Mondragon L, Xie W, Lévesque S, Pol J, Zitvogel L, Kepp O, Kroemer G. Anticancer effects of anti-CD47 immunotherapy in vivo. Oncoimmunology 2018; 8:1550619. [PMID: 30723582 DOI: 10.1080/2162402x.2018.1550619] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 11/09/2018] [Accepted: 11/14/2018] [Indexed: 12/22/2022] Open
Abstract
The treatment of breast cancer largely depends on the utilization of immunogenic chemotherapeutics, which, as a common leitmotif, stimulate the exposure of calreticulin (CALR) on the surface of cancer cells, thereby facilitating their recognition by dendritic cells for the uptake of tumor-associated antigens and subsequent antigen cross-presentation to cytotoxic T cells. Breast cancer cells also express the calreticulin antagonist CD47, which inhibits tumor cell phagocytosis and consequently subverts anticancer immune responses. Here, we treated carcinogen-induced or transplantable mouse models of cancer by a CD47 blocking antibody that was at least as efficient as chemotherapy and that could be favorably combined with the anthracycline mitoxantrone in the context of carcinogen-induced orthotopic breast cancers. Monotherapy by CD47 blockade led to a reduction in tumor growth and an increase in overall survival. Of note, this treatment lead to a moderate depletion of M2 macrophages as well as close-to-complete elimination of regulatory T cells from the tumor bed, suggesting a strong favorable impact of CD47 blockade on the tumor microenvironment.
Collapse
Affiliation(s)
- Kristina Iribarren
- Institut Gustave Roussy, Villejuif, France.,University of Paris Sud and Paris Saclay, Villejuif, France.,INSERM U1138, Villejuif, France
| | - Aitziber Buque
- Institut Gustave Roussy, Villejuif, France.,University of Paris Sud and Paris Saclay, Villejuif, France.,INSERM U1138, Villejuif, France
| | - Laura Mondragon
- Institut Gustave Roussy, Villejuif, France.,University of Paris Sud and Paris Saclay, Villejuif, France.,INSERM U1138, Villejuif, France
| | - Wei Xie
- Institut Gustave Roussy, Villejuif, France.,University of Paris Sud and Paris Saclay, Villejuif, France.,INSERM U1138, Villejuif, France
| | | | | | - Laurence Zitvogel
- Institut Gustave Roussy, Villejuif, France.,INSERM U1015, Villejuif, France.,CICBT1428, Villejuif, France
| | - Oliver Kepp
- Institut Gustave Roussy, Villejuif, France.,University of Paris Sud and Paris Saclay, Villejuif, France.,INSERM U1138, Villejuif, France
| | - Guido Kroemer
- INSERM U1138, Villejuif, France.,Metabolomics Platform, Institut Gustave Roussy, Villejuif, France.,Centre de Recherche des Cordeliers, Paris, France.,Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Université Pierre et Marie Curie, Paris, France.,Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
183
|
Englinger B, Pirker C, Heffeter P, Terenzi A, Kowol CR, Keppler BK, Berger W. Metal Drugs and the Anticancer Immune Response. Chem Rev 2018; 119:1519-1624. [DOI: 10.1021/acs.chemrev.8b00396] [Citation(s) in RCA: 174] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Bernhard Englinger
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - Christine Pirker
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - Petra Heffeter
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
- Research Cluster “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, Vienna, Austria
| | - Alessio Terenzi
- Research Cluster “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, Vienna, Austria
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Strasse 42, A-1090 Vienna, Austria
| | - Christian R. Kowol
- Research Cluster “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, Vienna, Austria
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Strasse 42, A-1090 Vienna, Austria
| | - Bernhard K. Keppler
- Research Cluster “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, Vienna, Austria
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Strasse 42, A-1090 Vienna, Austria
| | - Walter Berger
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
- Research Cluster “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, Vienna, Austria
| |
Collapse
|
184
|
Felley-Bosco E. Special Issue on Mechanisms of Mesothelioma Heterogeneity: Highlights and Open Questions. Int J Mol Sci 2018; 19:ijms19113560. [PMID: 30424481 PMCID: PMC6274972 DOI: 10.3390/ijms19113560] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 11/04/2018] [Accepted: 11/11/2018] [Indexed: 01/07/2023] Open
Abstract
This editorial aims to synthesize the eleven papers that have contributed to this special issue, where the mechanisms of mesothelioma heterogeneity have been tackled from different angles.
Collapse
Affiliation(s)
- Emanuela Felley-Bosco
- Laboratory of Molecular Oncology, University Hospital Zurich, Sternwartstrasse 14, 8091 Zürich, Switzerland.
| |
Collapse
|
185
|
Lybaert L, Vermaelen K, De Geest BG, Nuhn L. Immunoengineering through cancer vaccines – A personalized and multi-step vaccine approach towards precise cancer immunity. J Control Release 2018; 289:125-145. [DOI: 10.1016/j.jconrel.2018.09.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 09/11/2018] [Accepted: 09/12/2018] [Indexed: 02/07/2023]
|
186
|
Immunomodulatory Effects of Drugs for Effective Cancer Immunotherapy. JOURNAL OF ONCOLOGY 2018; 2018:8653489. [PMID: 30498512 PMCID: PMC6222238 DOI: 10.1155/2018/8653489] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 10/10/2018] [Indexed: 12/12/2022]
Abstract
Recent advances in cancer immunotherapy, including immune checkpoint inhibitors or adoptive T cell therapies, have contributed to better outcomes in cancer patients. However, there are still many cancers with no cure. Therefore, combinations of several treatment strategies are being explored, and enhancing anticancer immunity will play an important role to combat the disease. There have been several reports on the immune-modulatory effects of commonly used drugs, namely, statin, metformin, and angiotensin receptor blockers (ARBs), which suggest that these drugs could enhance immunity against cancer cells. Other anticancer drugs, such as anthracyclines, thalidomides, lenalidomides, and hypomethylating drugs, could also strengthen the immune system to attack cancer cells at a relatively low dose. Hence, these drugs might contribute to better outcomes in cancer patients.
Collapse
|
187
|
Alkarakooly Z, Al-Anbaky QA, Kannan K, Ali N. Metabolic reprogramming by Dichloroacetic acid potentiates photodynamic therapy of human breast adenocarcinoma MCF-7 cells. PLoS One 2018; 13:e0206182. [PMID: 30352078 PMCID: PMC6198976 DOI: 10.1371/journal.pone.0206182] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 10/07/2018] [Indexed: 01/07/2023] Open
Abstract
Aberrant glycolytic metabolism is one of the hallmarks of carcinogenesis and therefore reversal of metabolic transformation is a promising drug target in cancer treatment strategies. Dichloroacetic acid (DCA) is known to target the glycolytic pathway in cancer cells and facilitates reversal of metabolic transformation from aerobic cytosolic accumulation of pyruvic acid, "the Warburg effect", to mitochondrial oxidative phosphorylation. Recently, combination therapy particularly involving photodynamic therapy (PDT) has received considerable attention in oncology. We hypothesized that if DCA and PDT are combined, they might potentiate mitochondrial dysfunction and induce apoptosis by a reactive oxygen species (ROS) dependent pathway. We used MCF-7 cells as our in vitro model and 5-aminolevulinic acid (5-ALA) dependent PDT therapy to test our hypothesis. We found that combinatorial treatment of MCF-7 cells with PDT and DCA not only increased cell growth inhibition, but also affected mitochondrial membrane integrity perhaps via production of ROS, and enhanced apoptosis. Further, our results on ATP release during the combined treatment demonstrate that immunogenic cell death (ICD) is likely to be a potential mechanism by which PDT and DCA induce cancer cell death. Taken together, our study suggests a novel way of sensitizing MCF-7 cells for accelerated induction of apoptosis and ICD in these cells. The findings included in this study might have direct relevance in breast cancer treatment strategies.
Collapse
Affiliation(s)
- Zeiyad Alkarakooly
- Department of Biology, College of Science, University of Diyala, Diyala, Iraq
- Department of Biology, College of Arts, Letters and Sciences, University of Arkansas at Little Rock, Little Rock, Arkansas, United States of America
| | - Qudes A. Al-Anbaky
- Department of Biology, College of Science, University of Diyala, Diyala, Iraq
- Department of Biology, College of Arts, Letters and Sciences, University of Arkansas at Little Rock, Little Rock, Arkansas, United States of America
| | - Krishnaswamy Kannan
- Department of Biology, College of Arts, Letters and Sciences, University of Arkansas at Little Rock, Little Rock, Arkansas, United States of America
| | - Nawab Ali
- Department of Biology, College of Arts, Letters and Sciences, University of Arkansas at Little Rock, Little Rock, Arkansas, United States of America
| |
Collapse
|
188
|
Smith M, García-Martínez E, Pitter MR, Fucikova J, Spisek R, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Toll-like receptor agonists in cancer immunotherapy. Oncoimmunology 2018; 7:e1526250. [PMID: 30524908 PMCID: PMC6279325 DOI: 10.1080/2162402x.2018.1526250] [Citation(s) in RCA: 177] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Indexed: 12/14/2022] Open
Abstract
Toll-like receptor (TLR) agonists demonstrate therapeutic promise as immunological adjuvants for anticancer immunotherapy. To date, three TLR agonists have been approved by US regulatory agencies for use in cancer patients. Additionally, the potential of hitherto experimental TLR ligands to mediate clinically useful immunostimulatory effects has been extensively investigated over the past few years. Here, we summarize recent preclinical and clinical advances in the development of TLR agonists for cancer therapy.
Collapse
Affiliation(s)
- Melody Smith
- Department of Medicine and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Elena García-Martínez
- Hematology and Oncology Department, Hospital Universitario Morales Meseguer, Murcia, Spain
| | - Michael R. Pitter
- Department of Medicine and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jitka Fucikova
- Sotio a.c., Prague, Czech Republic
- Department of Immunology, 2nd Faculty of Medicine, University Hospital Motol, Charles University, Prague, Czech Republic
| | - Radek Spisek
- Sotio a.c., Prague, Czech Republic
- Department of Immunology, 2nd Faculty of Medicine, University Hospital Motol, Charles University, Prague, Czech Republic
| | - Laurence Zitvogel
- INSERM, U1015, Villejuif, France
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
- Université Paris Sud/Paris XI, Le Kremlin-Bicêtre, France
| | - Guido Kroemer
- Université Paris Descartes/ Paris V, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
- INSERM, U1138, Paris, France
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- Karolinska Institute, Department of Women’s and Children’s Health, Karolinska University Hospital, Stockholm, Sweden
- Pôle de Biologie, Hopitâl Européen George Pompidou, AP-HP; Paris, France
| | - Lorenzo Galluzzi
- Université Paris Descartes/ Paris V, Paris, France
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
| |
Collapse
|
189
|
Giglio P, Gagliardi M, Bernardini R, Mattei M, Cotella D, Santoro C, Piacentini M, Corazzari M. Ecto-Calreticulin is essential for an efficient immunogenic cell death stimulation in mouse melanoma. Genes Immun 2018; 20:509-513. [PMID: 30282994 DOI: 10.1038/s41435-018-0047-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 09/07/2018] [Accepted: 09/12/2018] [Indexed: 02/07/2023]
Abstract
Skin melanoma remains one of the most aggressive and difficult to treat human malignancy, with an increasing incidence every year. Although surgical resection represents the best therapeutic approach, this is only feasible in cases of early diagnosis. Furthermore, the established malignancy is resistant to all therapeutic strategies employed so far, resulting in an unacceptable patient survival rate. Although the immune-mediated therapeutic approaches, based on anti-PD1 or anti-CTLA4, are very promising and under clinical trial experimentation, they could conceal not yet fully emerged pitfalls such as the development of autoimmune diseases. Therefore, alternative therapeutic approaches are still under investigation, such as the immunogenic cell death (ICD) process. Here we show that the lack of calreticulin translocation onto mouse melanoma cell membrane prevents the stimulation of an effective ICD response in vivo.
Collapse
Affiliation(s)
- Paola Giglio
- Department of Epidemiology, National Institute for Infectious Diseases 'L. Spallanzani', Rome, Italy
| | - Mara Gagliardi
- Department of Biology, University of Rome 'Tor Vergata', Rome, Italy.,Department of Health Science (DISS), University of 'Piemonte Orientale', Novara, Italy
| | - Roberta Bernardini
- Department of Biology, Centro Servizi Interdipartimentale-STA, University of Rome 'Tor Vergata', Rome, Italy
| | - Maurizio Mattei
- Department of Biology, Centro Servizi Interdipartimentale-STA, University of Rome 'Tor Vergata', Rome, Italy
| | - Diego Cotella
- Department of Health Science (DISS), University of 'Piemonte Orientale', Novara, Italy
| | - Claudio Santoro
- Department of Health Science (DISS), University of 'Piemonte Orientale', Novara, Italy
| | - Mauro Piacentini
- Department of Epidemiology, National Institute for Infectious Diseases 'L. Spallanzani', Rome, Italy.,Department of Biology, University of Rome 'Tor Vergata', Rome, Italy
| | - Marco Corazzari
- Department of Health Science (DISS), University of 'Piemonte Orientale', Novara, Italy.
| |
Collapse
|
190
|
Osa A, Uenami T, Koyama S, Fujimoto K, Okuzaki D, Takimoto T, Hirata H, Yano Y, Yokota S, Kinehara Y, Naito Y, Otsuka T, Kanazu M, Kuroyama M, Hamaguchi M, Koba T, Futami Y, Ishijima M, Suga Y, Akazawa Y, Machiyama H, Iwahori K, Takamatsu H, Nagatomo I, Takeda Y, Kida H, Akbay EA, Hammerman PS, Wong KK, Dranoff G, Mori M, Kijima T, Kumanogoh A. Clinical implications of monitoring nivolumab immunokinetics in non-small cell lung cancer patients. JCI Insight 2018; 3:59125. [PMID: 30282824 PMCID: PMC6237460 DOI: 10.1172/jci.insight.59125] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 08/21/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND. The PD-1–blocking antibody nivolumab persists in patients several weeks after the last infusion. However, no study has systematically evaluated the maximum duration that the antibody persists on T cells or the association between this duration and residual therapeutic efficacy or potential adverse events. METHODS. To define the duration of binding and residual efficacy of nivolumab after discontinuation, we developed a simplified strategy for T cell monitoring and used it to analyze T cells from peripheral blood from 11 non–small cell lung cancer patients previously treated with nivolumab. To determine the suitability of our method for other applications, we compared transcriptome profiles between nivolumab-bound and nivolumab-unbound CD8 T cells. We also applied T cell monitoring in 2 nivolumab-treated patients who developed progressive lung tumors during long-term follow-up. RESULTS. Prolonged nivolumab binding was detected more than 20 weeks after the last infusion, regardless of the total number of nivolumab infusions (2–15 doses) or type of subsequent treatment, in 9 of the 11 cases in which long-term monitoring was possible. Ki-67 positivity, a proliferation marker, in T cells decreased in patients with progressive disease. Transcriptome profiling identified the signals regulating activation of nivolumab-bound T cells, which may contribute to nivolumab resistance. In 2 patients who restarted nivolumab, T cell proliferation markers exhibited the opposite trend and correlated with clinical response. CONCLUSIONS. Although only a few samples were analyzed, our strategy of monitoring both nivolumab binding and Ki-67 in T cells might help determine residual efficacy under various types of concurrent or subsequent treatment. TRIAL REGISTRATION. University Hospital Medical Information Network Clinical Trials Registry, UMIN000024623. FUNDING. This work was supported by Japan Society for the Promotion of Science KAKENHI (JP17K16045, JP18H05282, and JP15K09220), Japan Agency for Medical Research and Development (JP17cm0106310, JP18cm0106335 and JP18cm059042), and Core Research for Evolutional Science and Technology (JPMJCR16G2). A method for detecting nivolumab binding and T cell activation status, which could be used to predict residual efficacy and toxicity, is developed.
Collapse
Affiliation(s)
- Akio Osa
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,Laboratory of Immunopathology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Takeshi Uenami
- Department of Thoracic Oncology, National Hospital Organization, Toneyama National Hospital, Toyonaka, Osaka, Japan
| | - Shohei Koyama
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,Laboratory of Immunopathology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Kosuke Fujimoto
- Department of Immunology and Genomics, Osaka City University Graduate School of Medicine, Osaka, Osaka, Japan.,Division of Innate Immune Regulation, International Research and Development Center for Mucosal Vaccines, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Daisuke Okuzaki
- DNA-Chip Developmental Center for Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Takayuki Takimoto
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Haruhiko Hirata
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yukihiro Yano
- Department of Thoracic Oncology, National Hospital Organization, Toneyama National Hospital, Toyonaka, Osaka, Japan
| | - Soichiro Yokota
- Department of Thoracic Oncology, National Hospital Organization, Toneyama National Hospital, Toyonaka, Osaka, Japan
| | - Yuhei Kinehara
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,Laboratory of Immunopathology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Yujiro Naito
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,Laboratory of Immunopathology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Tomoyuki Otsuka
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Masaki Kanazu
- Department of Thoracic Oncology, National Hospital Organization, Toneyama National Hospital, Toyonaka, Osaka, Japan
| | - Muneyoshi Kuroyama
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Masanari Hamaguchi
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Taro Koba
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,Laboratory of Immunopathology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Yu Futami
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,Laboratory of Immunopathology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Mikako Ishijima
- Department of Thoracic Oncology, National Hospital Organization, Toneyama National Hospital, Toyonaka, Osaka, Japan
| | - Yasuhiko Suga
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,Laboratory of Immunopathology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Yuki Akazawa
- Department of Thoracic Oncology, National Hospital Organization, Toneyama National Hospital, Toyonaka, Osaka, Japan
| | - Hirotomo Machiyama
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Kota Iwahori
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hyota Takamatsu
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,Laboratory of Immunopathology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Izumi Nagatomo
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yoshito Takeda
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hiroshi Kida
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Esra A Akbay
- Department of Pathology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| | - Peter S Hammerman
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts, USA
| | - Kwok-Kin Wong
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, New York, USA
| | - Glenn Dranoff
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts, USA
| | - Masahide Mori
- Department of Thoracic Oncology, National Hospital Organization, Toneyama National Hospital, Toyonaka, Osaka, Japan
| | - Takashi Kijima
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,Laboratory of Immunopathology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,Laboratory of Immunopathology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan.,Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
191
|
Castoldi F, Vacchelli E, Zitvogel L, Maiuri MC, Pietrocola F, Kroemer G. Systemic autophagy in the therapeutic response to anthracycline-based chemotherapy. Oncoimmunology 2018; 8:e1498285. [PMID: 30546941 PMCID: PMC6287785 DOI: 10.1080/2162402x.2018.1498285] [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: 06/04/2018] [Accepted: 07/05/2018] [Indexed: 11/17/2022] Open
Abstract
The success of chemotherapy largely depends on the anticancer immune response triggered by tumor cells that succumb to immunogenic cell death (ICD). One of the hallmarks of ICD is premortem autophagy that facilitates the release of adenosine triphosphate from dying cancer cells and acts as a chemoattractant for dendritic cell precursors. Here, we show that the immune response induced by inoculation of cancer cells undergoing ICD in response to the anthracycline mitoxantrone (MTX) can be improved by a short-term fasting regimen (48 hours of starvation) and that this effect is reversed by systemic administration of the autophagy inhibitor dimethyl α-ketoglutarate. Tumor growth reduction by MTX treatment is known to depend on autophagy induction in cancer cells as well as on an intact immune system. We compared the antitumor effects of MTX on autophagy-competent cancers implanted in wild type (WT) or partially autophagy-deficient (Becn1± or Atg4b−/-) mice. While there was no difference in the tumor growth reducing effects of MTX on tumors evolving in WT, Becn1+/- and Atg4b−/- mice, we observed an increase in the toxicity of MTX on Atg4b−/- mice. These results suggest that autophagy in cancer cells (but less so in host cells) is rate-limiting for therapeutically relevant anticancer immune responses, yet has a major role in blunting the life-threatening toxicity of chemotherapy.
Collapse
Affiliation(s)
- Francesca Castoldi
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,INSERM, U1138, Paris, France.,Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Université Paris Descartes/Paris V, Sorbonne Paris Cité.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,Université Pierre et Marie Curie, Paris, France.,Faculté de Medecine, Université Paris-Sud/Paris-Saclay, Kremlin-Bicetre, France.,Sotio a.c., Prague, Czech Republic
| | - Erika Vacchelli
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,INSERM, U1138, Paris, France.,Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Université Paris Descartes/Paris V, Sorbonne Paris Cité.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,Université Pierre et Marie Curie, Paris, France
| | - Laurence Zitvogel
- Faculté de Medecine, Université Paris-Sud/Paris-Saclay, Kremlin-Bicetre, France.,INSERM U1015, Gustave Roussy Cancer Campus, Villejuif, France.,Center of Clinical Investigations in Biotherapies of Cancer (CICBT), Villejuif, France
| | - Maria Chiara Maiuri
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,INSERM, U1138, Paris, France.,Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Université Paris Descartes/Paris V, Sorbonne Paris Cité.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,Université Pierre et Marie Curie, Paris, France
| | - Federico Pietrocola
- INSERM, U1138, Paris, France.,Department of Molecular Medicine, Institute for Research in Biomedicine, Barcelona, Spain
| | - Guido Kroemer
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,INSERM, U1138, Paris, France.,Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Université Paris Descartes/Paris V, Sorbonne Paris Cité.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,Université Pierre et Marie Curie, Paris, France.,Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris,France.,Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
192
|
Bezu L, Kepp O, Cerrato G, Pol J, Fucikova J, Spisek R, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Peptide-based vaccines in anticancer therapy. Oncoimmunology 2018; 7:e1511506. [PMID: 30524907 PMCID: PMC6279318 DOI: 10.1080/2162402x.2018.1511506] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Indexed: 12/15/2022] Open
Abstract
Peptide-based anticancer vaccination aims at stimulating an immune response against one or multiple tumor-associated antigens (TAAs) following immunization with purified, recombinant or synthetically engineered epitopes. Despite high expectations, the peptide-based vaccines that have been explored in the clinic so far had limited therapeutic activity, largely due to cancer cell-intrinsic alterations that minimize antigenicity and/or changes in the tumor microenvironment that foster immunosuppression. Several strategies have been developed to overcome such limitations, including the use of immunostimulatory adjuvants, the co-treatment with cytotoxic anticancer therapies that enable the coordinated release of damage-associated molecular patterns, and the concomitant blockade of immune checkpoints. Personalized peptide-based vaccines are also being explored for therapeutic activity in the clinic. Here, we review recent preclinical and clinical progress in the use of peptide-based vaccines as anticancer therapeutics.Abbreviations: CMP: carbohydrate-mimetic peptide; CMV: cytomegalovirus; DC: dendritic cell; FDA: Food and Drug Administration; HPV: human papillomavirus; MDS: myelodysplastic syndrome; MHP: melanoma helper vaccine; NSCLC: non-small cell lung carcinoma; ODD: orphan drug designation; PPV: personalized peptide vaccination; SLP: synthetic long peptide; TAA: tumor-associated antigen; TNA: tumor neoantigen
Collapse
Affiliation(s)
- Lucillia Bezu
- Faculty of Medicine, University of Paris Sud/Paris XI, Le Kremlin-Bicêtre, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Equipe 11 labellisée Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers,Paris, France.,U1138, INSERM, Paris, France.,Université Paris Descartes/Paris V, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France
| | - Oliver Kepp
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Equipe 11 labellisée Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers,Paris, France.,U1138, INSERM, Paris, France.,Université Paris Descartes/Paris V, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France
| | - Giulia Cerrato
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Equipe 11 labellisée Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers,Paris, France.,U1138, INSERM, Paris, France.,Université Paris Descartes/Paris V, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France
| | - Jonathan Pol
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Equipe 11 labellisée Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers,Paris, France.,U1138, INSERM, Paris, France.,Université Paris Descartes/Paris V, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France
| | - Jitka Fucikova
- Sotio, Prague, Czech Republic.,Dept. of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Radek Spisek
- Sotio, Prague, Czech Republic.,Dept. of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Laurence Zitvogel
- Faculty of Medicine, University of Paris Sud/Paris XI, Le Kremlin-Bicêtre, France.,Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France.,INSERM, U1015, Gustave Roussy Cancer Campus, Villejuif, France
| | - Guido Kroemer
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Equipe 11 labellisée Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers,Paris, France.,U1138, INSERM, Paris, France.,Université Paris Descartes/Paris V, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France.,Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.,Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| | - Lorenzo Galluzzi
- Université Paris Descartes/Paris V, Paris, France.,Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Sandra and Edward Meyer Cancer Center, New York, NY, USA
| |
Collapse
|
193
|
Dalziel M, Beers SA, Cragg MS, Crispin M. Through the barricades: overcoming the barriers to effective antibody-based cancer therapeutics. Glycobiology 2018; 28:697-712. [PMID: 29800150 DOI: 10.1093/glycob/cwy043] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 04/30/2018] [Indexed: 02/06/2023] Open
Abstract
Since the turn of the century, cancer therapy has undergone a transformation in terms of new treatment modalities and renewed optimism in achieving long-lived tumor control and even cure. This is, in large part, thanks to the widespread incorporation of monoclonal antibodies (mAbs) into standard treatment regimens. These new therapies have, across many settings, significantly contributed to improved clinical responses, patient quality of life and survival. Moreover, the flexibility of the antibody platform has led to the development of a wide range of innovative and combinatorial therapies that continue to augment the clinician's armory. Despite these successes, there is a growing awareness that in many cases mAb therapy remains suboptimal, primarily due to inherent limitations imposed by the immune system's own homeostatic controls and the immunosuppressive tumor microenvironment. Here, we discuss the principal barriers that act to constrain the tumor-killing activity of antibody-based therapeutics, particularly those involving antibody glycans, using illustrative examples from both pre-clinical and market approved mAbs. We also discuss strategies that have been, or are in development to overcome these obstacles. Finally, we outline how the growing understanding of the biological terrain in which mAbs function is shaping innovation and regulation in cancer therapeutics.
Collapse
Affiliation(s)
- Martin Dalziel
- Oxford Glycobiology Institute, Department of Biochemistry, South Parks Road, Oxford, UK
| | - Stephen A Beers
- Antibody & Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Mark S Cragg
- Antibody & Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Max Crispin
- Centre for Biological Sciences, University of Southampton, Southampton, UK
- Institute for Life Sciences, Highfield Campus, University of Southampton, Southampton, UK
| |
Collapse
|
194
|
Pol JG, Lévesque S, Workenhe ST, Gujar S, Le Boeuf F, Clements DR, Fahrner JE, Fend L, Bell JC, Mossman KL, Fucikova J, Spisek R, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Oncolytic viro-immunotherapy of hematologic and solid tumors. Oncoimmunology 2018; 7:e1503032. [PMID: 30524901 PMCID: PMC6279343 DOI: 10.1080/2162402x.2018.1503032] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Accepted: 07/15/2018] [Indexed: 02/08/2023] Open
Abstract
Oncolytic viruses selectively target and kill cancer cells in an immunogenic fashion, thus supporting the establishment of therapeutically relevant tumor-specific immune responses. In 2015, the US Food and Drug Administration (FDA) approved the oncolytic herpes simplex virus T-VEC for use in advanced melanoma patients. Since then, a plethora of trials has been initiated to assess the safety and efficacy of multiple oncolytic viruses in patients affected with various malignancies. Here, we summarize recent preclinical and clinical progress in the field of oncolytic virotherapy.
Collapse
Affiliation(s)
- Jonathan G. Pol
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- INSERM, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
| | - Sarah Lévesque
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- INSERM, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
| | - Samuel T. Workenhe
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Shashi Gujar
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
- Department of Microbiology and Immunology, Dalhousie University, NS, Canada
- Department of Biology, Dalhousie University, NS, Canada
- Centre for Innovative and Collaborative Health Sciences Research, Quality and System Performance, IWK Health Centre, Halifax, NS, Canada
| | - Fabrice Le Boeuf
- Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | | | - Jean-Eudes Fahrner
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- INSERM, Villejuif, France
- Transgene S.A., Illkirch-Graffenstaden, France
| | | | - John C. Bell
- Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Karen L. Mossman
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Jitka Fucikova
- Sotio a.c., Prague, Czech Republic
- Department of Immunology, 2nd Faculty of Medicine, University Hospital Motol, Charles University, Prague, Czech Republic
| | - Radek Spisek
- Sotio a.c., Prague, Czech Republic
- Department of Immunology, 2nd Faculty of Medicine, University Hospital Motol, Charles University, Prague, Czech Republic
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- INSERM, Villejuif, France
| | - Guido Kroemer
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- INSERM, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, Paris, France
- Department of Women’s and Children’s Health, Karolinska University Hospital, Stockholm, Sweden
| | - Lorenzo Galluzzi
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
| |
Collapse
|
195
|
Rossi C, Gilhodes J, Maerevoet M, Herbaux C, Morschhauser F, Brice P, Garciaz S, Borel C, Ysebaert L, Obéric L, Lazarovici J, Deau B, Dupuis J, Chauchet A, Abraham J, Bijou F, Stamatoullas-Bastard A, Malfuson JV, Golfier C, Laurent C, Pericart S, Traverse-Glehen A, Kanoun S, Filleron T, Casasnovas RO, Ghesquières H. Efficacy of chemotherapy or chemo-anti-PD-1 combination after failed anti-PD-1 therapy for relapsed and refractory Hodgkin lymphoma: A series from Lysa centers. Am J Hematol 2018; 93:1042-1049. [PMID: 29884994 DOI: 10.1002/ajh.25154] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 05/11/2018] [Accepted: 05/20/2018] [Indexed: 12/13/2022]
Abstract
Anti-PD-1 therapy provides high response rates in Hodgkin lymphoma (HL) patients who have relapsed or are refractory (R/R) to autologous stem cell transplantation (ASCT) and brentuximab vedotin (BV), but median progression free survival (PFS) is only one year. The efficacy of treatment following anti-PD-1 is not well known. We retrospectively investigated the efficacy of salvage therapies for unsatisfactory response to anti-PD-1 therapy, assessed by PET-CT according to the Lugano criteria, in 30 R/R HL patients. Patients were highly pre-treated before anti-PD-1 (70% received ASCT and 93% BV). Unsatisfactory responses to anti-PD1 therapy were progressive disease (PD) (n=24) and partial response (PR) (n=6). For the 24 PD patients, median anti-PD-1 related PFS was 7.5 months (95%CI, 5.7-11.6); 17 received subsequent CT alone (Group 1) and 7 received CT in addition to anti-PD-1 (Group 2). 16/24 patients (67%) obtained an objective response. In the 15 patients treated with the same CT, twelve obtained PR or complete response (CR). In Group 1, there were 7 CR (41%), 3 PR (18%), and 7 PD (41%). In Group 2, there were 4 CR (57%), 2 PR (29%), and 1 SD (14%). No unexpected toxicity was observed. Six patients who achieved response proceeded to allogeneic SCT. With a median follow-up of 12.1 months (7-14.7), the median PFS following the initiation of CT was 11 months (95%CI, 6.3; not reached) and the median of overall survival was not reached. These observations in highly pre-treated HL patients suggest that anti-PD-1 therapy might re-sensitize tumor cells to CT. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Cédric Rossi
- Cancer Research Centre of Toulouse (CRCT), UMR1037 INSERM, Université Toulouse III Paul-Sabatier, ERL5294 CNRS, Université de Toulouse, Toulouse, France
- Department of Hematology, Dijon University Hospital, Dijon, France
| | - Julia Gilhodes
- Clinical trials office, Institut universitaire du cancer Toulouse- Oncopole, Toulouse, France
| | | | - Charles Herbaux
- Department of Hematology, Unité GRITA, CHRU Claude Huriez, Lille, France
| | | | - Pauline Brice
- Department of Hematology, CHU Paris-GH St-Louis Lariboisière F-Widal - Hôpital Saint-Louis, Paris, France
| | - Sylvain Garciaz
- Department of Hematology, Institut Paoli-Calmettes, Marseille, France
| | - Cécile Borel
- Department of Hematology, Institut universitaire du cancer Toulouse- Oncopole, Toulouse, France
| | - Loïc Ysebaert
- Department of Hematology, Institut universitaire du cancer Toulouse- Oncopole, Toulouse, France
| | - Lucie Obéric
- Department of Hematology, Institut universitaire du cancer Toulouse- Oncopole, Toulouse, France
| | - Julien Lazarovici
- Department of Hematology, Institut Gustave-Roussy, Villejuif, France
| | | | - Jehan Dupuis
- Department of Hematology, CHU Henri Mondor, Créteil, France
| | | | - Julie Abraham
- Department of Hematology, CHU Limoges, Limoges, France
| | - Fontanet Bijou
- Department of Hematology, Institut Bergonié, Bordeaux, France
| | | | - Jean-Valère Malfuson
- Department of Hematology, hôpital d'instruction des armées Percy, Clamart, France
| | - Camille Golfier
- Department of Hematology, Dijon University Hospital, Dijon, France
| | - Camille Laurent
- Anatomy-pathology Department, Institut universitaire du cancer Toulouse-Oncopole, Toulouse, France
| | - Sarah Pericart
- Anatomy-pathology Department, Institut universitaire du cancer Toulouse-Oncopole, Toulouse, France
| | | | - Salim Kanoun
- Nuclear Medecine Unit, Institut universitaire du cancer Toulouse-Oncopole, Toulouse, France
| | - Thomas Filleron
- Clinical trials office, Institut universitaire du cancer Toulouse- Oncopole, Toulouse, France
| | - René-Olivier Casasnovas
- Department of Hematology, Dijon University Hospital, Dijon, France
- INSERM UMR 1231 CHU Dijon, France
| | - Hervé Ghesquières
- Department of Hematology, Centre Hospitalier Lyon Sud, Pierre-Bénite, France
| |
Collapse
|
196
|
Qiao M, Jiang T, Zhou C. Shining light on advanced NSCLC in 2017: combining immune checkpoint inhibitors. J Thorac Dis 2018; 10:S1534-S1546. [PMID: 29951304 PMCID: PMC5994489 DOI: 10.21037/jtd.2018.04.99] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 04/11/2018] [Indexed: 12/25/2022]
Abstract
The treatment landscape has changed since the immune checkpoint inhibitors were approved in the treatment of non-small cell lung cancer (NSCLC). Although the promising clinical benefit from programmed death-1/programmed death ligand-1 (PD-1/PD-L1) inhibitors was observed in the second or subsequent line treatment of patients who progressed on chemotherapy, it has a long way for single PD-1/PD-L1 inhibitor to move forward to the frontline without a predictive biomarker. Tumor response is far from satisfactory without selection and primary or acquired resistance to PD-1/PD-L1 inhibitors hampered their utility. Therefore, it is crucial to determine a strategy that can optimize the application of immune checkpoint inhibitors and increase the numbers of the responders. Multiple combination approaches based on PD-1/PD-L1 inhibitors are designed and aimed to boost anti-tumor response and benefit a broader population. In this review, we will integrate the updated clinical data to highlight the four most promising combination strategies in advance NSCLC: combination of checkpoint inhibition with chemotherapy, anti-angiogenesis, immunotherapy and radiotherapy. We further discuss the issues needed to be addressed and perspectives in the context of "combination era".
Collapse
Affiliation(s)
- Meng Qiao
- Department of Medical Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China
| | - Tao Jiang
- Department of Medical Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China
| | - Caicun Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China
| |
Collapse
|
197
|
Romano E, Rufo N, Korf H, Mathieu C, Garg AD, Agostinis P. BNIP3 modulates the interface between B16-F10 melanoma cells and immune cells. Oncotarget 2018; 9:17631-17644. [PMID: 29707136 PMCID: PMC5915144 DOI: 10.18632/oncotarget.24815] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 02/27/2018] [Indexed: 01/10/2023] Open
Abstract
The hypoxia responsive protein BNIP3, plays an important role in promoting cell death and/or autophagy, ultimately resulting in a cancer type-dependent, tumour-enhancer or tumour-suppressor activity. We previously reported that in melanoma cells, BNIP3 regulates cellular morphology, mitochondrial clearance, cellular viability and maintains protein expression of CD47, a pro-cancerous, immunosuppressive 'don't eat me' signal. Surface exposed CD47 is often up-regulated by cancer cells to avoid clearance by phagocytes and to suppress immunogenic cell death (ICD) elicited by anticancer therapies. However, whether melanoma-associated BNIP3 modulates CD47-associated immunological effects or ICD has not been explored properly. To this end, we evaluated the impact of the genetic ablation of BNIP3 (i.e. BNIP3KD) in melanoma cells, on macrophage-based phagocytosis, polarization and chemotaxis. Additionally, we tested its effects on crucial determinants of chemotherapy-induced ICD (i.e. danger signals), as well as in vivo anticancer vaccination effect. Interestingly, loss of BNIP3 reduced the expression of CD47 both in normoxic and hypoxic conditions while macrophage phagocytosis and chemotaxis were accentuated only when BNIP3KD melanoma cells were exposed to hypoxia. Moreover, when exposed to the ICD inducer mitoxantrone, the loss of melanoma cell-associated BNIP3 did not alter apoptosis induction, but significantly prevented ATP secretion and reduced phagocytic clearance of dying cells. In line with this, prophylactic vaccination experiments showed that the loss of BNIP3 tends to increase the intrinsic resistance of B16-F10 melanoma cells to ICD-associated anticancer vaccination effect in vivo. Thus, normoxic vs. hypoxic and live vs. dying cell contexts influence the ultimate immunomodulatory roles of melanoma cell-associated BNIP3.
Collapse
Affiliation(s)
- Erminia Romano
- Laboratory for Cell Death Research and Therapy (CDRT), Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Nicole Rufo
- Laboratory for Cell Death Research and Therapy (CDRT), Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Hannelie Korf
- Laboratory of Hepatology, Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
- Laboratory of Clinical and Experimental Endocrinology (CEE), Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Chantal Mathieu
- Laboratory of Clinical and Experimental Endocrinology (CEE), Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Abhishek D Garg
- Laboratory for Cell Death Research and Therapy (CDRT), Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Patrizia Agostinis
- Laboratory for Cell Death Research and Therapy (CDRT), Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
198
|
Patel SA, Minn AJ. Combination Cancer Therapy with Immune Checkpoint Blockade: Mechanisms and Strategies. Immunity 2018; 48:417-433. [PMID: 29562193 PMCID: PMC6948191 DOI: 10.1016/j.immuni.2018.03.007] [Citation(s) in RCA: 418] [Impact Index Per Article: 59.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 03/01/2018] [Accepted: 03/02/2018] [Indexed: 12/13/2022]
Abstract
The success of immune checkpoint blockade in patients with a wide variety of malignancies has changed the treatment paradigm in oncology. However, combination therapies with immune checkpoint blockade will be needed to overcome resistance and broaden the clinical utility of immunotherapy. Here we discuss a framework for rationally designing combination therapy strategies based on enhancing major discriminatory functions of the immune system that are corrupted by cancer-namely, antigenicity, adjuvanticity, and homeostatic feedback inhibition. We review recent advances on how conventional genotoxic cancer therapies, molecularly targeted therapies, epigenetic agents, and immune checkpoint inhibitors can restore these discriminatory functions. Potential barriers that can impede response despite combination therapy are also discussed.
Collapse
Affiliation(s)
- Shetal A Patel
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Parker Institute for Cancer Immunotherapy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Andy J Minn
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Parker Institute for Cancer Immunotherapy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Basser Center for BRCA, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
199
|
Versteven M, Van den Bergh JMJ, Marcq E, Smits ELJ, Van Tendeloo VFI, Hobo W, Lion E. Dendritic Cells and Programmed Death-1 Blockade: A Joint Venture to Combat Cancer. Front Immunol 2018; 9:394. [PMID: 29599770 PMCID: PMC5863527 DOI: 10.3389/fimmu.2018.00394] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 02/13/2018] [Indexed: 12/31/2022] Open
Abstract
Two decades of clinical cancer research with dendritic cell (DC)-based vaccination have proved that this type of personalized medicine is safe and has the capacity to improve survival, but monotherapy is unlikely to cure the cancer. Designed to empower the patient’s antitumor immunity, huge research efforts are set to improve the efficacy of next-generation DC vaccines and to find synergistic combinations with existing cancer therapies. Immune checkpoint approaches, aiming to breach immune suppression and evasion to reinforce antitumor immunity, have been a revelation in the immunotherapy field. Early success of therapeutic antibodies blocking the programmed death-1 (PD-1) pathway has sparked the development of novel inhibitors and combination therapies. Hence, merging immunoregulatory tumor-specific DC strategies with PD-1-targeted approaches is a promising path to explore. In this review, we focus on the role of PD-1-signaling in DC-mediated antitumor immunity. In the quest of exploiting the full potential of DC therapy, different strategies to leverage DC immunopotency by impeding PD-1-mediated immune regulation are discussed, including the most advanced research on targeted therapeutic antibodies, lessons learned from chemotherapy-induced immune activation, and more recent developments with soluble molecules and gene-silencing techniques. An overview of DC/PD-1 immunotherapy combinations that are currently under preclinical and clinical investigation substantiates the clinical potential of such combination strategies.
Collapse
Affiliation(s)
- Maarten Versteven
- Laboratory of Experimental Hematology, Faculty of Medicine and Health Sciences, Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium
| | - Johan M J Van den Bergh
- Laboratory of Experimental Hematology, Faculty of Medicine and Health Sciences, Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium
| | - Elly Marcq
- Center for Oncological Research Antwerp, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Evelien L J Smits
- Laboratory of Experimental Hematology, Faculty of Medicine and Health Sciences, Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium.,Center for Oncological Research Antwerp, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.,Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium
| | - Viggo F I Van Tendeloo
- Laboratory of Experimental Hematology, Faculty of Medicine and Health Sciences, Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium
| | - Willemijn Hobo
- Laboratory of Hematology, Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Eva Lion
- Laboratory of Experimental Hematology, Faculty of Medicine and Health Sciences, Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium.,Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Antwerp, Belgium
| |
Collapse
|
200
|
Abstract
Neutrophils are the most abundant leukocytes in the circulation, and have been regarded as first line of defense in the innate arm of the immune system. They capture and destroy invading microorganisms, through phagocytosis and intracellular degradation, release of granules, and formation of neutrophil extracellular traps after detecting pathogens. Neutrophils also participate as mediators of inflammation. The classical view for these leukocytes is that neutrophils constitute a homogenous population of terminally differentiated cells with a unique function. However, evidence accumulated in recent years, has revealed that neutrophils present a large phenotypic heterogeneity and functional versatility, which place neutrophils as important modulators of both inflammation and immune responses. Indeed, the roles played by neutrophils in homeostatic conditions as well as in pathological inflammation and immune processes are the focus of a renovated interest in neutrophil biology. In this review, I present the concept of neutrophil phenotypic and functional heterogeneity and describe several neutrophil subpopulations reported to date. I also discuss the role these subpopulations seem to play in homeostasis and disease.
Collapse
Affiliation(s)
- Carlos Rosales
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| |
Collapse
|