1
|
Rana N, Chaudhary PK, Prasad R, Sankar M. Photodynamic Evaluation of A 2BC Aminoporphyrins: Synthesis, Characterization, and Cellular Impact. ACS APPLIED BIO MATERIALS 2025. [PMID: 40421616 DOI: 10.1021/acsabm.5c00411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2025]
Abstract
This study focuses on the design and evaluation of a series of A2BC aminoporphyrins, featuring electron-donating substituents like pyrene, carbazole, and phenothiazine to enhance their photophysical and biological performance. Detailed characterization through spectroscopic methods, single-crystal X-ray diffraction, and computational analyses revealed insights into their electronic structure and planarity. Photophysical investigations revealed characteristic Soret and Q bands, along with tunable fluorescence and excited-state lifetimes influenced by the meso substituents. Biological evaluation was conducted using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assays and fluorescence microscopy to assess the photodynamic therapeutic efficacy against T24 bladder cancer cells. The porphyrins exhibited pronounced photocytotoxicity upon 660 nm light activation, attributed to reactive oxygen species (ROS) generation. Cellular analysis, including acridine orange/ethidium bromide and 4',6-diamidino-2-phenylindole staining, confirmed apoptosis induction through chromatin condensation and nuclear fragmentation. The findings highlight the potential of these porphyrins as effective photosensitizers for photodynamic therapy, demonstrating enhanced stability and ROS generation efficiency.
Collapse
Affiliation(s)
- Nivedita Rana
- Department of Chemistry, Indian Institute of Technology Roorkee, Roorkee 247667, India
| | - Pankaj Kumar Chaudhary
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, India
| | - Ramasare Prasad
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, India
| | - Muniappan Sankar
- Department of Chemistry, Indian Institute of Technology Roorkee, Roorkee 247667, India
| |
Collapse
|
2
|
Lee KW, Yam JWP, Mao X. Dendritic Cell Vaccines: A Shift from Conventional Approach to New Generations. Cells 2023; 12:2147. [PMID: 37681880 PMCID: PMC10486560 DOI: 10.3390/cells12172147] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/21/2023] [Accepted: 08/23/2023] [Indexed: 09/09/2023] Open
Abstract
In the emerging era of cancer immunotherapy, immune checkpoint blockades (ICBs) and adoptive cell transfer therapies (ACTs) have gained significant attention. However, their therapeutic efficacies are limited due to the presence of cold type tumors, immunosuppressive tumor microenvironment, and immune-related side effects. On the other hand, dendritic cell (DC)-based vaccines have been suggested as a new cancer immunotherapy regimen that can address the limitations encountered by ICBs and ACTs. Despite the success of the first generation of DC-based vaccines, represented by the first FDA-approved DC-based therapeutic cancer vaccine Provenge, several challenges remain unsolved. Therefore, new DC vaccine strategies have been actively investigated. This review addresses the limitations of the currently most adopted classical DC vaccine and evaluates new generations of DC vaccines in detail, including biomaterial-based, immunogenic cell death-inducing, mRNA-pulsed, DC small extracellular vesicle (sEV)-based, and tumor sEV-based DC vaccines. These innovative DC vaccines are envisioned to provide a significant breakthrough in cancer immunotherapy landscape and are expected to be supported by further preclinical and clinical studies.
Collapse
Affiliation(s)
- Kyu-Won Lee
- Department of Pathology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong; (K.-W.L.); (J.W.P.Y.)
| | - Judy Wai Ping Yam
- Department of Pathology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong; (K.-W.L.); (J.W.P.Y.)
- State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong
| | - Xiaowen Mao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao
| |
Collapse
|
3
|
Aaes TL, Vandenabeele P. The intrinsic immunogenic properties of cancer cell lines, immunogenic cell death, and how these influence host antitumor immune responses. Cell Death Differ 2021; 28:843-860. [PMID: 33214663 PMCID: PMC7937679 DOI: 10.1038/s41418-020-00658-y] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 10/19/2020] [Accepted: 10/22/2020] [Indexed: 01/30/2023] Open
Abstract
Modern cancer therapies often involve the combination of tumor-directed cytotoxic strategies and generation of a host antitumor immune response. The latter is unleashed by immunotherapies that activate the immune system generating a more immunostimulatory tumor microenvironment and a stronger tumor antigen-specific immune response. Studying the interaction between antitumor cytotoxic therapies, dying cancer cells, and the innate and adaptive immune system requires appropriate experimental tumor models in mice. In this review, we discuss the immunostimulatory and immunosuppressive properties of cancer cell lines commonly used in immunogenic cell death (ICD) studies being apoptosis or necroptosis. We will especially focus on the antigenic component of immunogenicity. While in several cancer cell lines the epitopes of endogenously expressed tumor antigens are known, these intrinsic epitopes are rarely determined in experimental apoptotic or necroptotic ICD settings. Instead by far the most ICD research studies investigate the antigenic response against exogenously expressed model antigens such as ovalbumin or retroviral epitopes (e.g., AH1). In this review, we will argue that the immune response against endogenous tumor antigens and the immunopeptidome profile of cancer cell lines affect the eventual biological readouts in the typical prophylactic tumor vaccination type of experiments used in ICD research, and we will propose additional methods involving immunopeptidome profiling, major histocompatibility complex molecule expression, and identification of tumor-infiltrating immune cells to document intrinsic immunogenicity following different cell death modalities.
Collapse
Affiliation(s)
- Tania Løve Aaes
- grid.11486.3a0000000104788040Unit for Cell Clearance in Health and Disease, VIB Center for Inflammation Research, Ghent, Belgium ,grid.5342.00000 0001 2069 7798Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium ,Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Peter Vandenabeele
- grid.5342.00000 0001 2069 7798Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium ,Cancer Research Institute Ghent (CRIG), Ghent, Belgium ,grid.11486.3a0000000104788040Unit of Molecular Signaling and Cell Death, VIB Center for Inflammation Research, Ghent, Belgium
| |
Collapse
|
4
|
Mekonnen ZA, Masavuli MG, Yu W, Gummow J, Whelan DM, Al-Delfi Z, Torresi J, Gowans EJ, Grubor-Bauk B. Enhanced T Cell Responses Induced by a Necrotic Dendritic Cell Vaccine, Expressing HCV NS3. Front Microbiol 2020; 11:559105. [PMID: 33343515 PMCID: PMC7739890 DOI: 10.3389/fmicb.2020.559105] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 10/28/2020] [Indexed: 12/21/2022] Open
Abstract
A vaccine that induces potent, broad and sustained cell-mediated immunity, resulting in effective memory has the potential to restrict hepatitis C (HCV) virus infection. Early, multi-functional CD4+ and CD8+ T cell responses against non-structural protein 3 (NS3) have been associated with HCV clearance. Necrotic cells generate strong immune responses and represent a major antigenic source used by dendritic cells (DC) for processing and presentation, but there is conflicting evidence as to their immunogenicity in vaccination. Immunization with DC loaded with viral antigens has been done in the past, but to date the immunogenicity of live vs. necrotic DC vaccines has not been investigated. We developed a DC2.4 cell line stably expressing HCV NS3, and compared the NS3-specific responses of live vs. necrotic NS3 DC. Vaccination of mice with necrotic NS3 DC increased the breadth of T-cell responses and enhanced the production of IL-2, TNF-α, and IFN-γ by effector memory CD4+ and CD8+T cells, compared to mice vaccinated with live NS3 DC. A single dose of necrotic NS3 DC vaccine induced a greater influx and activation of cross-presenting CD11c+ CD8α+ DC and necrosis-sensing Clec9A+ DC in the draining lymph nodes. Furthermore, using a hydrodynamic challenge model necrotic NS3 DC vaccination resulted in enhanced clearance of NS3-positive hepatocytes from the livers of vaccinated mice. Taken together, the data demonstrate that necrotic DC represent a novel and exciting vaccination strategy capable of inducing broad and multifunctional T cell memory.
Collapse
Affiliation(s)
- Zelalem A Mekonnen
- Viral Immunology Group, Discipline of Surgery, Basil Hetzel Institute for Translational Medicine, University of Adelaide, Adelaide, SA, Australia
| | - Makutiro G Masavuli
- Viral Immunology Group, Discipline of Surgery, Basil Hetzel Institute for Translational Medicine, University of Adelaide, Adelaide, SA, Australia
| | - Wenbo Yu
- Viral Immunology Group, Discipline of Surgery, Basil Hetzel Institute for Translational Medicine, University of Adelaide, Adelaide, SA, Australia.,Centre for Cancer Biology, University of South Australia, Adelaide, SA, Australia
| | - Jason Gummow
- Gene Silencing and Expression Laboratory, Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
| | - Dawn M Whelan
- Viral Immunology Group, Discipline of Surgery, Basil Hetzel Institute for Translational Medicine, University of Adelaide, Adelaide, SA, Australia
| | - Zahraa Al-Delfi
- Viral Immunology Group, Discipline of Surgery, Basil Hetzel Institute for Translational Medicine, University of Adelaide, Adelaide, SA, Australia
| | - Joseph Torresi
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Eric J Gowans
- Viral Immunology Group, Discipline of Surgery, Basil Hetzel Institute for Translational Medicine, University of Adelaide, Adelaide, SA, Australia
| | - Branka Grubor-Bauk
- Viral Immunology Group, Discipline of Surgery, Basil Hetzel Institute for Translational Medicine, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
5
|
Sobhanimonfared F, Bamdad T, Sadigh ZA, Choobin H. Virus specific tolerance enhanced efficacy of cancer immuno-virotherapy. Microb Pathog 2019; 140:103957. [PMID: 31891795 DOI: 10.1016/j.micpath.2019.103957] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 12/22/2019] [Accepted: 12/27/2019] [Indexed: 01/14/2023]
Abstract
BACKGROUND Activation of the immune system to fight cancer is a major goal in immunology and oncology. Although cancer treatment using oncolytic viruses shows promising results, virus mediated oncolysis induces a weak anti-tumor immune response. Upon application of viruses, immune responses against the virus play a significant role in limiting tumor virotherapy. Although suppression of host immunity increases the efficacy of virotherapy against the tumor, but inhibits anti-tumor immune responses. Induction of viral specific tolerance before viral replication may cause the virus to efficiently replicate in tumor cells without affecting the immune responses against tumor antigens. Investigation of the combined strategy of virotherapy and immunotherapy using irradiated tumor cells along with IL-2 and interferon-alpha in virus specific tolerant mice was the goal of this study. MATERIALS AND METHODS For tolerance induction, the newborn mice were injected with vesicular stomatitis virus (VSV) subcutaneously. After injection of TC-1 tumor cells to adult tolerant mice and formation of a tumor, irradiated TC-1 cells along with IL-2 and Interferon-alpha expression plasmid were injected twice in mice and followed by virotherapy. Size of tumors and CTL activity against the virus and tumor cells were measured. RESULT The results showed increased efficacy of virotherapy in combination with immune-stimulators and tumor cells injection in tolerant mice compared to normal mice. CONCLUSION Specific tolerance against the oncolytic virus enhances the efficacy of virotherapy both in monotherapy and in combination with immunotherapy.
Collapse
Affiliation(s)
- Fatemeh Sobhanimonfared
- Department of Virology, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Taravat Bamdad
- Department of Virology, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Zohreh Azita Sadigh
- Human Viral Vaccine Department, Razi Vaccine and Serum Research Institute, Karaj, Iran.
| | - Hamzeh Choobin
- Department of Virology, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
6
|
Madsen SJ, Christie C, Huynh K, Peng Q, Uzal FA, Krasieva TB, Hirschberg H. Limiting glioma development by photodynamic therapy-generated macrophage vaccine and allo-stimulation: an in vivo histological study in rats. JOURNAL OF BIOMEDICAL OPTICS 2018; 23:1-7. [PMID: 29417766 PMCID: PMC5802332 DOI: 10.1117/1.jbo.23.2.028001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 01/19/2018] [Indexed: 06/08/2023]
Abstract
Immunotherapy of brain tumors involves the stimulation of an antitumor immune response. This type of therapy can be targeted specifically to tumor cells thus sparing surrounding normal brain. Due to the presence of the blood-brain barrier, the brain is relatively isolated from the systemic circulation and, as such, the initiation of significant immune responses is more limited than other types of cancers. The purpose of this study was to show that the efficacy of tumor primed antigen presenting macrophage (MaF98) vaccines can be increased by: (1) photodynamic therapy (PDT) of the priming tumor cells and (2) intracranial injection of allogeneic glioma cells directly into the tumor site. Experiments were conducted in an in vivo brain tumor development model using Fischer rats and F98 (syngeneic) and BT4C (allogeneic) glioma cells. The results showed that immunization with Ma (acting as antigen-presenting cells), primed with PDT-treated tumor cells (MaF98), significantly slowed but did not prevent the growth of F98-induced tumors in the brain. Complete suppression of tumor development was obtained via MaF98 inoculation combined with direct intracranial injection of allogeneic glioma cells. No deleterious effects were noted in any of the animals during the 14-day observation period.
Collapse
Affiliation(s)
- Steen J. Madsen
- University of Nevada, Department of Health Physics and Diagnostic Sciences, Las Vegas, Nevada, United States
| | - Catherine Christie
- University of California, Beckman Laser Institute and Medical Clinic, Irvine, California, United States
| | - Khoi Huynh
- University of California, Beckman Laser Institute and Medical Clinic, Irvine, California, United States
| | - Qian Peng
- University of Oslo, Pathology Clinic, Rikshospitalet-Radiumhospitalet HF Medical Center, Montebello, Oslo, Norway
| | - Francisco A. Uzal
- University of California, School of Veterinary Medicine, Davis, San Bernardino, California, United States
| | - Tatiana B. Krasieva
- University of California, Beckman Laser Institute and Medical Clinic, Irvine, California, United States
| | - Henry Hirschberg
- University of California, Beckman Laser Institute and Medical Clinic, Irvine, California, United States
| |
Collapse
|
7
|
Affiliation(s)
- Thaiz Rivera Vargas
- Centre de Recherche; INSERM U1231; Facultés de Médecine et de Pharmacie; Dijon France
- Faculté de Médecine; Université de Bourgogne Franche comté; Dijon France
| | - Lionel Apetoh
- Centre de Recherche; INSERM U1231; Facultés de Médecine et de Pharmacie; Dijon France
- Faculté de Médecine; Université de Bourgogne Franche comté; Dijon France
- Centre Georges François Leclerc; Dijon France
| |
Collapse
|
8
|
Boosting Tumor-Specific Immunity Using PDT. Cancers (Basel) 2016; 8:cancers8100091. [PMID: 27782066 PMCID: PMC5082381 DOI: 10.3390/cancers8100091] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 10/04/2016] [Indexed: 01/06/2023] Open
Abstract
Photodynamic therapy (PDT) is a cancer treatment with a long-standing history. It employs the application of nontoxic components, namely a light-sensitive photosensitizer and visible light, to generate reactive oxygen species (ROS). These ROS lead to tumor cell destruction, which is accompanied by the induction of an acute inflammatory response. This inflammatory process sends a danger signal to the innate immune system, which results in activation of specific cell types and release of additional inflammatory mediators. Activation of the innate immune response is necessary for subsequent induction of the adaptive arm of the immune system. This includes the priming of tumor-specific cytotoxic T lymphocytes (CTL) that have the capability to directly recognize and kill cells which display an altered self. The past decades have brought increasing appreciation for the importance of the generation of an adaptive immune response for long-term tumor control and induction of immune memory to combat recurrent disease. This has led to considerable effort to elucidate the immune effects PDT treatment elicits. In this review we deal with the progress which has been made during the past 20 years in uncovering the role of PDT in the induction of the tumor-specific immune response, with special emphasis on adaptive immunity.
Collapse
|
9
|
Takemura R, Takaki H, Okada S, Shime H, Akazawa T, Oshiumi H, Matsumoto M, Teshima T, Seya T. PolyI:C-Induced, TLR3/RIP3-Dependent Necroptosis Backs Up Immune Effector-Mediated Tumor Elimination In Vivo. Cancer Immunol Res 2015; 3:902-914. [PMID: 25898986 DOI: 10.1158/2326-6066.cir-14-0219] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 04/07/2015] [Indexed: 01/17/2023]
Abstract
Double-stranded RNA directly acts on fibroblast and myeloid lineages to induce necroptosis as in TNFα. Here, we investigated whether this type of cell death occurred in cancer cells in response to polyinosinic-polycytidylic acid (polyI:C) and the pan-caspase inhibitor z-Val-Ala-Asp fluromethyl ketone (zVAD). We found that the colon cancer cell line CT26 is highly susceptible to necroptosis, as revealed by staining with annexin V/propidium iodide. CT26 cells possess RNA sensors, TLR3 and MDA5, which are upregulated by interferon (IFN)-inducing pathways and linked to receptor-interacting protein kinase (RIP) 1/3 activation via TICAM-1 or MAVS adaptor, respectively. Although exogenously added polyI:C alone marginally induced necroptosis in CT26 cells, a combined regimen of polyI:C and zVAD induced approximately 50% CT26 necroptosis in vitro without secondary effects of TNFα or type I IFNs. CT26 necroptosis depended on the TLR3-TICAM-1-RIP3 axis in the tumor cells to produce reactive oxygen species, but not on MDA5, MAVS, or the caspases/inflammasome activation. However, the RNA-derived necroptosis was barely reproduced in vivo in a CT26 tumor-implanted Balb/c mouse model with administration of polyI:C + zVAD. Significant shrinkage of CT26 tumors was revealed only when polyI:C (100 μg) was injected intraperitoneally and zVAD (1 mg) subcutaneously into tumor-bearing mice that were depleted of cytotoxic T lymphocytes and natural killer cells. The results were confirmed with immune-compromised mice with no lymphocytes. Although necroptosis-induced tumor growth retardation appears mechanistically complicated and dependent on the injection routes of polyI:C and zVAD, anti-caspase reagent directed to tumor cells will make RNA adjuvant immunotherapy more effective by modulating the formation of the tumoricidal microenvironment and dendritic cell-inducing antitumor immune system.
Collapse
Affiliation(s)
- Ryo Takemura
- Department of Microbiology and Immunology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan. Department of Hematology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hiromi Takaki
- Department of Microbiology and Immunology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Seiji Okada
- Division of Hematopoiesis, Center for AIDS Research, Kumamoto University, Kumamoto, Japan
| | - Hiroaki Shime
- Department of Microbiology and Immunology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Takashi Akazawa
- Department of Tumor Immunology, Research Institute, Osaka Medical Center for Cancer and Cardiovascular Diseases, Osaka, Japan
| | - Hiroyuki Oshiumi
- Department of Microbiology and Immunology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Misako Matsumoto
- Department of Microbiology and Immunology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Takanori Teshima
- Department of Hematology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Tsukasa Seya
- Department of Microbiology and Immunology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan.
| |
Collapse
|
10
|
Hunn MK, Bauer E, Wood CE, Gasser O, Dzhelali M, Ancelet LR, Mester B, Sharples KJ, Findlay MP, Hamilton DA, Hermans IF. Dendritic cell vaccination combined with temozolomide retreatment: results of a phase I trial in patients with recurrent glioblastoma multiforme. J Neurooncol 2014; 121:319-29. [DOI: 10.1007/s11060-014-1635-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2014] [Accepted: 10/18/2014] [Indexed: 12/21/2022]
|
11
|
Gamrekelashvili J, Greten TF, Korangy F. Immunogenicity of necrotic cell death. Cell Mol Life Sci 2014; 72:273-83. [PMID: 25274062 DOI: 10.1007/s00018-014-1741-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 09/03/2014] [Accepted: 09/22/2014] [Indexed: 12/20/2022]
Abstract
The mode of tumor cell death has significant effects on anti-tumor immunity. Although, previously it was thought that cell death is an inert effect, different investigators have clearly shown that dying tumors can attract, activate and mature professional antigen presenting cells and dendritic cells. In addition, others and we have shown that the type of tumor cell death not only controls the presence or absence of specific tumor antigens, but also can result in immunological responses ranging from immunosuppression to anti-tumor immunity. More importantly, it is possible to enhance anti-tumor immunity both in vitro and in vivo by targeting specific molecular mechanisms such as oligopeptidases and the proteasome. These studies not only extend our knowledge on basic immunological questions and the induction of anti-tumor immunity, but also have implications for all types of cancer treatments, in which rapid tumor cell death is induced. This review is a comprehensive summary of cell death and particularly necrosis and the pivotal role it plays in anti-tumor immunity.
Collapse
|
12
|
Lecis D, De Cesare M, Perego P, Conti A, Corna E, Drago C, Seneci P, Walczak H, Colombo MP, Delia D, Sangaletti S. Smac mimetics induce inflammation and necrotic tumour cell death by modulating macrophage activity. Cell Death Dis 2013; 4:e920. [PMID: 24232096 PMCID: PMC3847325 DOI: 10.1038/cddis.2013.449] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Revised: 10/11/2013] [Accepted: 10/15/2013] [Indexed: 12/30/2022]
Abstract
Smac mimetics (SMs) comprise a class of small molecules that target members of the inhibitor of apoptosis family of pro-survival proteins, whose expression in cancer cells hinders the action of conventional chemotherapeutics. Herein, we describe the activity of SM83, a newly synthesised dimeric SM, in two cancer ascites models: athymic nude mice injected intraperitoneally with IGROV-1 human ovarian carcinoma cells and immunocompetent BALB/c mice injected with murine Meth A sarcoma cells. SM83 rapidly killed ascitic IGROV-1 and Meth A cells in vivo (prolonging mouse survival), but was ineffective against the same cells in vitro. IGROV-1 cells in nude mice were killed within the ascites by a non-apoptotic, tumour necrosis factor (TNF)-dependent mechanism. SM83 administration triggered a rapid inflammatory event characterised by host secretion of TNF, interleukin-1β and interferon-γ. This inflammatory response was associated with the reversion of the phenotype of tumour-associated macrophages from a pro-tumoural M2- to a pro-inflammatory M1-like state. SM83 treatment was also associated with a massive recruitment of neutrophils that, however, was not essential for the antitumoural activity of this compound. In BALB/c mice bearing Meth A ascites, SM83 treatment was in some cases curative, and these mice became resistant to a second injection of cancer cells, suggesting that they had developed an adaptive immune response. Altogether, these results indicate that, in vivo, SM83 modulates the immune system within the tumour microenvironment and, through its pro-inflammatory action, leads cancer cells to die by necrosis with the release of high-mobility group box-1. In conclusion, our work provides evidence that SMs could be more therapeutically active than expected by stimulating the immune system.
Collapse
Affiliation(s)
- D Lecis
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan 20133, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Giardino A, Girelli R, Frigerio I, Regi P, Cantore M, Alessandra A, Lusenti A, Salvia R, Bassi C, Pederzoli P. Triple approach strategy for patients with locally advanced pancreatic carcinoma. HPB (Oxford) 2013; 15:623-7. [PMID: 23458679 PMCID: PMC3731584 DOI: 10.1111/hpb.12027] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Accepted: 10/29/2012] [Indexed: 12/12/2022]
Abstract
BACKGROUND Radiofrequency ablation (RFA) is a relatively new technique, applied to metastatic solid tumours which, in recent studies, has been shown to be feasible and safe on locally advanced pancreatic carcinoma (LAPC). RFA can be combined with radio-chemotherapy (RCT) and intra-arterial plus systemic chemotherapy (IASC). The aim of this study was to investigate the impact on the prognosis of a multimodal approach to LAPC and define the best timing of RFA. METHODS This is a retrospective observational study of patients who have consecutively undergone RFA associated with multiple adjuvant approaches. RESULTS Between February 2007 and December 2011, 168 consecutive patients were treated by RFA, of which 107 were eligible for at least 18 months of follow-up. Forty-seven patients (group 1) underwent RFA as an up-front treatment and 60 patients as second treatment (group 2) depending on clinician choice. The median overall survival (OS) of the whole series was 25.6 months: 14.7 months in the group 1 and 25.6 months in the group 2 (P = 0.004). Those patients who received the multimodal treatment (RFA, RCT and IASC-triple approach strategy) had an OS of 34.0 months. CONCLUSIONS The multimodal approach seems to be feasible and associated with an improved longer survival rate.
Collapse
Affiliation(s)
| | - Roberto Girelli
- Pancreatic Unit, Casa di Cura PederzoliPeschiera del Garda (VR), Italy
| | - Isabella Frigerio
- Pancreatic Unit, Casa di Cura PederzoliPeschiera del Garda (VR), Italy
| | - Paolo Regi
- Pancreatic Unit, Casa di Cura PederzoliPeschiera del Garda (VR), Italy
| | - Maurizio Cantore
- Oncology Department, Casa di Cura PederzoliPeschiera del Garda (VR), Italy
| | | | - Annita Lusenti
- Oncology Department, Casa di Cura PederzoliPeschiera del Garda (VR), Italy
| | - Roberto Salvia
- Surgical and Oncological Department, University of VeronaVerona (VR), Italy
| | - Claudio Bassi
- Surgical and Oncological Department, University of VeronaVerona (VR), Italy
| | - Paolo Pederzoli
- Pancreatic Unit, Casa di Cura PederzoliPeschiera del Garda (VR), Italy
| |
Collapse
|
14
|
Kaczmarek A, Vandenabeele P, Krysko DV. Necroptosis: the release of damage-associated molecular patterns and its physiological relevance. Immunity 2013; 38:209-23. [PMID: 23438821 DOI: 10.1016/j.immuni.2013.02.003] [Citation(s) in RCA: 1157] [Impact Index Per Article: 96.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Indexed: 10/27/2022]
Abstract
Regulated necrosis, termed necroptosis, is negatively regulated by caspase-8 and is dependent on the kinase activity of RIPK1 and RIPK3. Necroptosis leads to rapid plasma membrane permeabilization and to the release of cell contents and exposure of damage-associated molecular patterns (DAMPs). We are only beginning to identify the necroptotic DAMPs, their modifications, and their potential role in the regulation of inflammation. In this review, we discuss the physiological relevance of necroptosis and its role in the modulation of inflammation. For example, during viral infection, RIPK3-mediated necroptosis acts as a backup mechanism to clear pathogens. Necroptosis is also involved in apparently immunologically silent maintenance of T cell homeostasis. In contrast, the induction of necroptosis in skin, intestine, systemic inflammatory response syndrome, and ischemia reperfusion injury provoke a strong inflammatory response, which might be triggered by emission of DAMPs from necroptotic cells, showing the detrimental side of necroptosis.
Collapse
Affiliation(s)
- Agnieszka Kaczmarek
- Molecular Signaling and Cell Death Unit, Department for Molecular Biomedical Research, VIB, 9052 Ghent, Belgium
| | | | | |
Collapse
|
15
|
Cantore M, Girelli R, Mambrini A, Frigerio I, Boz G, Salvia R, Giardino A, Orlandi M, Auriemma A, Bassi C. Combined modality treatment for patients with locally advanced pancreatic adenocarcinoma. Br J Surg 2012; 99:1083-8. [PMID: 22648697 DOI: 10.1002/bjs.8789] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/27/2012] [Indexed: 12/13/2022]
Abstract
BACKGROUND Radiofrequency ablation (RFA) is an emerging treatment for patients with locally advanced pancreatic carcinoma, and can be combined with radiochemotherapy and intra-arterial plus systemic chemotherapy. METHODS This observational study compared two groups of patients with locally advanced pancreatic carcinoma treated with either primary RFA (group 1) or RFA following any other primary treatment (group 2). RESULTS Between February 2007 and May 2010, 107 consecutive patients were treated with RFA. There were 47 patients in group 1 and 60 in group 2. Median overall survival was 25·6 months. Median overall survival was significantly shorter in group 1 than in group 2 (14·7 versus 25·6 months; P = 0·004) Patients treated with RFA, radiochemotherapy and intra-arterial plus systemic chemotherapy (triple-approach strategy) had a median overall survival of 34·0 months. CONCLUSION RFA after alternative primary treatment was associated with prolonged survival. This was further extended by use of a triple-approach strategy in selected patients. Further evaluation of this approach seems warranted.
Collapse
Affiliation(s)
- M Cantore
- Oncological Department, Carrara Hospital, Carrara, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Matuszewski M, Michajłowski J, Michajłowski I, Ruckermann-Dizurdzińska K, Witkowski JM, Biernat W, Krajka K. Impact of radiofrequency ablation on PBMC subpopulation in patients with renal cell carcinoma. Urol Oncol 2010; 29:724-30. [PMID: 20189845 DOI: 10.1016/j.urolonc.2009.11.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2009] [Revised: 09/07/2009] [Accepted: 11/20/2009] [Indexed: 11/30/2022]
Abstract
PURPOSE With the development of diagnostic techniques, renal cell carcinoma (RCC) is currently diagnosed in earlier stages, allowing the introduction of less invasive techniques in its management. One of the most promising new treatment methods is based on the utilization of high temperature created by radiofrequency current circulating around the needle probe introduced into the tumor. Besides the direct destruction of the cancer tissue, the treatment may induce immunologic reaction to tumor antigens released from destroyed tumor cell. This paper describes changes observed in the peripheral blood lymphocyte population after radiofrequency ablation (RFA) of RCC. METHODS Blood was tested before, and 2, 4, and 6 weeks after the RFA in 6 patients with RCC for the proportions and numbers of CD3(+), CD3(+)HLA-DR(+), CD3(+)CD4(+), CD3(+)CD8(+), and CD56(+)CD16(+) cells. The blood was stained with fluorochrome-conjugated monoclonal antibodies and percentages of cells expressing various markers were determined by flow cytometry. RESULTS In all patients, the changes were most pronounced 2 weeks after the procedure. The proportion of CD4(+) and CD8(+) lymphocytes were changed. In 1 patient, an increase in both CD4(+) and CD8(+) cells was observed. In 5 out of 6 patients, the proportion of activated (DR(+)) cells was increased over the whole follow-up period with the highest values in the second week after RFA. The percentage of the CD56(+)CD16(+) was decreased in most of the patients. CONCLUSIONS Our study confirms that in the majority of patients, RFA of the renal tumors causes significant changes in the proportion of the peripheral immune cells. We suggest that the results presented in this article shows the necessity for further studies.
Collapse
|
17
|
Kumar S, Deepak P, Kumar S, Kishore D, Acharya A. Autologous Hsp70 induces antigen specific Th1 immune responses in a murine T-cell lymphoma. Immunol Invest 2010; 38:449-65. [PMID: 19811405 DOI: 10.1080/08820130902802673] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Heat Shock protein-70 derived from tumor cells is highly immunogenic and induces specific anti-tumor immune response by directly activating cytotoxic CD8(+) T cells. Additionally, Hsp70 is known to be a strong activator of antigen presenting cells and therefore, up regulates the production of pro-inflammatory cytokines and chemokines. In this study, we have shown the effect of tumor-derived Hsp70 on the induction of delayed type hypersensitivity reaction in a T cell lymphoma bearing mice. The autologous Hsp70 augments contact hypersensitivity and delayed type hypersensitivity responses in mice challenged with allergen in vehicle and antigens respectively. The adoptive transfer of splenocytes derived from Hsp70 immunized mice is able to enhance delayed type hypersensitivity response in antigen challenged normal and DL-bearing host. Furthermore, adoptive transfer of macrophages incubated with autologous Hsp70 also enhances DTH reactivity in mice. The pro-inflammatory cytokines and C-C chemokines are found to be elevated in the DTH footpad extract of antigen challenged normal and DL-bearing mice. Increased production of IFN-gamma and MIP-1alpha+/- suggest that autologous Hsp70 augments the recruitment of antigen specific Th1 cells, which further secretes pro-inflammatory cytokines and C-C chemokines mediating the hypersensitivity reaction upon challenge with antigens.
Collapse
Affiliation(s)
- Sanjay Kumar
- Centre of Advance Study in Zoology, Faculty of Science, Banaras Hindu University, Varanasi, U.P., India
| | | | | | | | | |
Collapse
|
18
|
Xing W, Wu S, Yuan X, Chen Q, Shen X, He F, Bian J, Lei P, Zhu H, Wang S, Shen G. The anti-tumor effect of human monocyte-derived dendritic cells loaded with HSV-TK/GCV induced dying cells. Cell Immunol 2008; 254:135-41. [PMID: 18834973 DOI: 10.1016/j.cellimm.2008.08.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2008] [Revised: 08/15/2008] [Accepted: 08/18/2008] [Indexed: 10/21/2022]
Abstract
Herpes simplex virus thymidine kinase (HSV-TK) gene and dendritic cells (DC) have been used as the pioneering in cancer therapy. HSV-TK gene can induce apoptosis and necrosis in tumor cells in the presence of the non-toxic prodrug ganciclovir (GCV). We investigated the anti-tumor effect of DC vaccination by introducing dying cells from HSV-TK gene treatment as an adjuvant. HepG(2)-TK cell line was established by transfecting human hepatoma cell line HepG(2) (HLA-A(2) positive) with HSV-TK gene. Dying tumor cells were generated by culturing HepG(2)-TK cells with GCV. After engulfed dying cells efficiently, immature DCs (imDC) derived from human monocytes were fully matured and elicited marked proliferation and cytotoxicity against HLA matched HepG(2) cells in autologous peripheral blood mononuclear cells (PBMC). It also implied that HepG(2) specific CTLs played an important role in the cytotoxicity which was primarily depended on Th1 responses. Given the feasibility of inducing dying cells by HSV-TK/GCV in vivo, our results suggest an effective method in clinical human hepatocellular carcinoma (HCC) treatment by an in vitro model of applying HSV-TK gene modified human tumor cells integrated with DC vaccination.
Collapse
Affiliation(s)
- Wei Xing
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, No.13 Hangkong Road, Wuhan 430030, PR China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Prestwich RJ, Harrington KJ, Pandha HS, Vile RG, Melcher AA, Errington F. Oncolytic viruses: a novel form of immunotherapy. Expert Rev Anticancer Ther 2008; 8:1581-8. [PMID: 18925850 PMCID: PMC2729453 DOI: 10.1586/14737140.8.10.1581] [Citation(s) in RCA: 140] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Oncolytic viruses are novel anticancer agents, currently under investigation in Phase I-III clinical trials. Until recently, most studies have focused on the direct antitumor properties of these viruses, although there is now an increasing body of evidence that the host immune response may be critical to the efficacy of oncolytic virotherapy. This may be mediated via innate immune effectors, adaptive antiviral immune responses eliminating infected cells or adaptive antitumor immune responses. This report summarizes preclinical and clinical evidence for the importance of immune interactions, which may be finely balanced between viral and tumor elimination. On this basis, oncolytic viruses represent a promising novel immunotherapy strategy, which may be optimally combined with existing therapeutic modalities.
Collapse
Affiliation(s)
- Robin J Prestwich
- Cancer Research UK Clinical Centre, Leeds Institute of Molecular Medicine, University of Leeds, Leeds, LS9 7TF, UK, Tel.: +44 113 343 8450, Fax: +44 113 242 9886,
| | - Kevin J Harrington
- Targeted Therapy Laboratory, The Institute of Cancer Research, Cancer Research UK, Centre for Cell and Molecular Biology, Chester Beatty, Laboratories, 237 Fulham Road, London, SW3 6JB, UK, Tel.: +44 207 153 5157, Fax: +44 207 808 2235,
| | - Hardev S Pandha
- Oncology, Postgraduate Medical School, University of Surrey, Guildford, GU2 7XX, UK, Tel.: +44 148 368 8618, Fax: +44 148 368 8558,
| | - Richard G Vile
- Cancer Research UK Clinical Centre, Leeds Institute of Molecular Medicine, University of Leeds, Leeds, LS9 7TF, UK; Molecular Medicine Program; Department of Immunology, Mayo Clinic, Rochester, Minnesota, 55905, USA, Tel.: +1 507 284 9941, Fax: +1 507 266 2122,
| | - Alan A Melcher
- Cancer Research UK Clinical Centre, Leeds Institute of Molecular Medicine, University of Leeds, Leeds, LS9 7TF, UK, Tel.: +44 113 343 8450, Fax: +44 113 242 9886,
| | - Fiona Errington
- Cancer Research UK Clinical Centre, Leeds Institute of Molecular Medicine, University of Leeds, Leeds, LS9 7TF, UK, Tel.: +44 113 343 8450, Fax: +44 113 242 9886,
| |
Collapse
|
20
|
Dendritic cell vaccines in melanoma: From promise to proof? Crit Rev Oncol Hematol 2008; 66:118-34. [DOI: 10.1016/j.critrevonc.2007.12.007] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2007] [Revised: 12/03/2007] [Accepted: 12/13/2007] [Indexed: 02/02/2023] Open
|
21
|
Apetoh L, Mignot G, Panaretakis T, Kroemer G, Zitvogel L. Immunogenicity of anthracyclines: moving towards more personalized medicine. Trends Mol Med 2008; 14:141-51. [DOI: 10.1016/j.molmed.2008.02.002] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2007] [Revised: 01/29/2008] [Accepted: 02/01/2008] [Indexed: 12/22/2022]
|
22
|
Endo Y, Sakai R, Ouchi M, Onimatsu H, Hioki M, Kagawa S, Uno F, Watanabe Y, Urata Y, Tanaka N, Fujiwara T. Virus-mediated oncolysis induces danger signal and stimulates cytotoxic T-lymphocyte activity via proteasome activator upregulation. Oncogene 2007; 27:2375-81. [PMID: 17982491 DOI: 10.1038/sj.onc.1210884] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Dendritic cells (DCs) are the most potent antigen-presenting cells and acquire cellular antigens and danger signals from dying cells to initiate antitumor immune responses via direct cell-to-cell interaction and cytokine production. The optimal forms of tumor cell death for priming DCs for the release of danger signals are not fully understood. OBP-301 (Telomelysin) is a telomerase-specific replication-competent adenovirus that induces selective E1 expression and exclusively kills human cancer cells. Here, we show that OBP-301 replication produced the endogenous danger signaling molecule, uric acid, in infected human tumor cells, which in turn stimulated DCs to produce interferon-gamma (IFN-gamma) and interleukin 12 (IL-12). Subsequently, IFN-gamma release upregulated the endogenous expression of the proteasome activator PA28 in tumor cells and resulted in the induction of cytotoxic T-lymphocytes. Our data suggest that virus-mediated oncolysis might be the effective stimulus for immature DCs to induce specific activity against human cancer cells.
Collapse
Affiliation(s)
- Y Endo
- Division of Surgical Oncology, Department of Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Chaput N, De Botton S, Obeid M, Apetoh L, Ghiringhelli F, Panaretakis T, Flament C, Zitvogel L, Kroemer G. Molecular determinants of immunogenic cell death: surface exposure of calreticulin makes the difference. J Mol Med (Berl) 2007; 85:1069-76. [PMID: 17891368 DOI: 10.1007/s00109-007-0214-1] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2007] [Revised: 04/23/2007] [Accepted: 04/25/2007] [Indexed: 12/17/2022]
Abstract
The treatment of cancer by chemotherapy causes tumour cell death, mostly by apoptosis. This tumour cell death may or may not elicit an immune response. At least in some cases, the efficacy of chemotherapy critically depends on the induction of immunogenic cell death that is a type of cell demise that stimulates the activation of an adaptative anti-tumour immune response, which in turn helps to eradicate residual cancer (stem) cells. Indeed, anthracyclins care more efficient in curing tumours in immunocompetent than in T cell-deficient mice. The molecular mechanism implicated in this anti-tumour T cell activation was recently discovered. Anthracyclins cause immunogenic cell death due to their specific capacity to stimulate the translocation of calreticulin to the cell surface. Calreticulin then acts as an "eat me" signal for dendritic cells, allowing them to phagocytose tumour cells and to prime tumour antigen-specific cytotoxic T cells. Importantly, non-immunogenic chemotherapy can be rendered immunogenic by adsorbing recombinant calreticulin to tumour cells or by enforcing the translocation of endogenous calreticulin to the cell surface by means of PP1/GADD34 inhibitors. This strategy could have major implications for the treatment of human cancer. Indeed, in vivo treatments with anthracyclins can cause the translocation of calreticulin to the surface of circulating tumour cells, in patients with acute myeloid leukaemia (AML). The challenge will be to determine whether the exposure of calreticulin translocation on the tumour cell surface is linked to chemotherapy-induced anti-tumour immune responses and therapeutic efficacy in human cancer.
Collapse
MESH Headings
- Animals
- Anthracyclines/pharmacology
- Anthracyclines/therapeutic use
- Antigens, Neoplasm/drug effects
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/metabolism
- Antigens, Surface/drug effects
- Antigens, Surface/immunology
- Antigens, Surface/metabolism
- Antineoplastic Agents/pharmacology
- Apoptosis/drug effects
- Apoptosis/immunology
- Calreticulin/immunology
- Calreticulin/metabolism
- Cell Line, Tumor
- Cell Membrane/drug effects
- Cell Membrane/immunology
- Cell Membrane/metabolism
- Dendritic Cells/immunology
- Humans
- Mice
- Models, Biological
- Neoplasms, Experimental/immunology
- T-Lymphocytes, Cytotoxic/immunology
Collapse
Affiliation(s)
- Nathalie Chaput
- Centre d'Investigation Clinique Biothérapie, Institut Gustave Roussy, Villejuif, France
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Abstract
Photodynamic therapy (PDT) uses non-toxic photosensitizers and harmless visible light in combination with oxygen to produce cytotoxic reactive oxygen species that kill malignant cells by apoptosis and/or necrosis, shut down the tumour microvasculature and stimulate the host immune system. In contrast to surgery, radiotherapy and chemotherapy that are mostly immunosuppressive, PDT causes acute inflammation, expression of heat-shock proteins, invasion and infiltration of the tumour by leukocytes, and might increase the presentation of tumour-derived antigens to T cells.
Collapse
Affiliation(s)
- Ana P Castano
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | | |
Collapse
|
25
|
Casares N, Pequignot MO, Tesniere A, Ghiringhelli F, Roux S, Chaput N, Schmitt E, Hamai A, Hervas-Stubbs S, Obeid M, Coutant F, Métivier D, Pichard E, Aucouturier P, Pierron G, Garrido C, Zitvogel L, Kroemer G. Caspase-dependent immunogenicity of doxorubicin-induced tumor cell death. ACTA ACUST UNITED AC 2006; 202:1691-701. [PMID: 16365148 PMCID: PMC2212968 DOI: 10.1084/jem.20050915] [Citation(s) in RCA: 1144] [Impact Index Per Article: 60.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Systemic anticancer chemotherapy is immunosuppressive and mostly induces nonimmunogenic tumor cell death. Here, we show that even in the absence of any adjuvant, tumor cells dying in response to anthracyclins can elicit an effective antitumor immune response that suppresses the growth of inoculated tumors or leads to the regression of established neoplasia. Although both antracyclins and mitomycin C induced apoptosis with caspase activation, only anthracyclin-induced immunogenic cell death was immunogenic. Caspase inhibition by Z-VAD-fmk or transfection with the baculovirus inhibitor p35 did not inhibit doxorubicin (DX)-induced cell death, yet suppressed the immunogenicity of dying tumor cells in several rodent models of neoplasia. Depletion of dendritic cells (DCs) or CD8+T cells abolished the immune response against DX-treated apoptotic tumor cells in vivo. Caspase inhibition suppressed the capacity of DX-killed cells to be phagocytosed by DCs, yet had no effect on their capacity to elicit DC maturation. Freshly excised tumors became immunogenic upon DX treatment in vitro, and intratumoral inoculation of DX could trigger the regression of established tumors in immunocompetent mice. These results delineate a procedure for the generation of cancer vaccines and the stimulation of anti-neoplastic immune responses in vivo.
Collapse
Affiliation(s)
- Noelia Casares
- Centre National de la Recherche Scientifique UMR8125, Institut Gustave Roussy, F-94805 Villejuif, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Zerbini A, Pilli M, Penna A, Pelosi G, Schianchi C, Molinari A, Schivazappa S, Zibera C, Fagnoni FF, Ferrari C, Missale G. Radiofrequency thermal ablation of hepatocellular carcinoma liver nodules can activate and enhance tumor-specific T-cell responses. Cancer Res 2006; 66:1139-46. [PMID: 16424051 DOI: 10.1158/0008-5472.can-05-2244] [Citation(s) in RCA: 193] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Radiofrequency thermal ablation (RFA) destroys tumoral tissue generating a local necrosis followed by marked inflammatory response with a dense T-cell infiltrate. In this study, we tested whether hepatocellular carcinoma thermal ablation can induce or enhance T-cell responses specific for hepatocellular carcinoma-associated antigens. Peripheral blood mononuclear cells derived from 20 patients with hepatocellular carcinoma were stimulated before and a month after RFA treatment with autologous hepatocellular carcinoma-derived protein lysates obtained before and immediately after RFA treatment. The effect of thermal ablation on memory T-cell responses to recall antigens [tetanus toxoid, protein purified derivative (PPD), Escherichia coli] was also assessed. T-cell reactivity was analyzed in an IFN-gamma enzyme-linked immunospot assay and by intracellular IFN-gamma staining. Treatment was followed by a significant increase of patients responsive either to tumor antigens derived from both the untreated hepatocellular carcinoma tissue (P < 0.05) and the necrotic tumor (P < 0.01) and by a higher frequency of circulating tumor-specific T cells. T-cell responses to recall antigens were also significantly augmented. Phenotypic analysis of circulating T and natural killer cells showed an increased expression of activation and cytotoxic surface markers. However, tumor-specific T-cell responses were not associated with protection from hepatocellular carcinoma relapse. Evidence of tumor immune escape was provided in one patient by the evidence that a new nodule of hepatocellular carcinoma recurrence was not recognized by T cells obtained at the time of RFA. In conclusion, RFA treatment generates the local conditions for activating the tumor-specific T-cell response. Although this effect is not sufficient for controlling hepatocellular carcinoma, it may represent the basis for the development of an adjuvant immunotherapy in patients undergoing RFA for primary and secondary liver tumors.
Collapse
Affiliation(s)
- Alessandro Zerbini
- Unit of Infectious Diseases and Hepatology, Azienda Ospedaliero-Universitaria di Parma, Via Gramsci 14, 43100 Parma, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Chiou SH, Sheu BC, Chang WC, Huang SC, Hong-Nerng H. Current concepts of tumor-infiltrating lymphocytes in human malignancies. J Reprod Immunol 2005; 67:35-50. [PMID: 16111767 DOI: 10.1016/j.jri.2005.06.002] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Tumor-infiltrating lymphocytes (TILs) develop as manifestations of the recognition and defense against malignant cells by the host immune system. TILs were literally defined as "tumor-infiltrating lymphocytes", which a posteriori locate within the tumor tissues. Although such cells can be found, they fail to control the growth of tumor. Many have proposed diverse mechanisms for dysfunction of TILs with regard to the roles of immunosurveillance against cancer. However, only a few cancer types, e.g. melanoma, have seen the benefits brought by activating these cells for immunotherapy. Functional defects of TILs have been linked to abnormalities of signaling molecules; however, there is conflicting data. The death of TILs was attributed to expression of cancer-derived FasL, PD-1 and RCAS1, and cancer-induced activation-induced cell death (AICD). Confirmed by studies using TILs and animal models, the compromise of tumor-specific immune responses was thought to result from not only mechanisms of clonal anergy but also exhaustion and/or deletion. Furthermore, functional cytotoxic CD8(+) TILs might be rendered incompetent by cancer-induced up-regulation of inhibitory NK receptors or proximal signaling abnormalities. Additionally, immune privilege was partly attributed to recruitment of regulatory T cells to the tumor sites. The failure of IL-2 signaling, which stands at the center of T cell functionalities, had been linked to the enzymatic activity of cancer-derived matrix metalloproteinases (MMPs). Finally, the exploitation of IDO expression, an important enzyme in pregnancy-related immunosuppression, by cancer cells might play a role in tumor immunity. The disparity of cancer types, origin, developmental stages and individual genetic backgrounds likely account for differences, or even contradictions, which might be the reason why immunotherapy works only on a few cancer types. Delineating the mechanisms behind functional defects of TILs can help not only boost chances of the development of a successful cure but understand the not fully identified roles played by immune system in the face of malignancies.
Collapse
Affiliation(s)
- Shin-Heng Chiou
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, 7 Chung-Shan South Road, Taipei 100, Taiwan
| | | | | | | | | |
Collapse
|
28
|
Fast LD. Control of immune responses induced by the transfer of allogeneic white blood cells during transfusion. Transfusion 2005; 45:44S-50S. [PMID: 16086787 DOI: 10.1111/j.1537-2995.2005.00530.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Loren D Fast
- Division of Hematology/Oncology, Rhode Island Hospital, Providence, Rhode Island 02903, USA.
| |
Collapse
|