54 results on '"Lotze, Michael T"'
Search Results
2. SITC Clinical Immuno-Oncology Network (SCION) commentary on measurement and interpretation of essential biomarkers in early clinical trials.
- Author
-
Lotze MT, Cottrell T, Bifulco C, Chow L, Cope L, Gnjatic S, Maecker HT, and Yeong Poh Shen J
- Subjects
- Humans, Biomarkers, Medical Oncology, Clinical Trials as Topic, Neoplasms diagnosis, Neoplasms therapy
- Abstract
Competing Interests: Competing interests: MTL—Ownership Interest: iRepertoire; Consulting Fees: iRepertoire; KSQ; BioCytics; Zia Pharma; Appel Sauce (pending); Agenus (pending). TC—Consulting Fees: Jannsen. CB—Ownership Interest: PrimeVax; Grant Funding to Institution: Illumina; Consulting Fees: Sanofi, Agilent, Roche, Incendia, PrimeVax, BioAI, Lunaphore. LCh—Consulting Fees: EMD Serono, Jazz Pharmaceuticals, AstraZeneca, Daiichi Sankyo, Ipsen, Nanobiotix, Merck, Sanofi-Genzyme, Regeneron, Bluprint Therapeutics, Beigene, Novartis, Elicio Therapeutics, Alkermes, Gilead, Cullinan-Apollo Corp. SG—Grant Funding to Institution: Regeneron, Boehringer-Ingelheim, Janssen R&D, Genentech, Takeda, Bristol Myers Squibb, Celgene; Patents: Co-inventor on an issued patent for multiplex immunohistochemistry to characterize tumors and treatment responses. The technology is filed through Icahn School of Medicine at Mount Sinai (ISMMS) and is currently unlicensed, Inventor of technology to detect autoantibodies to GM-CSF, a cytokine that promotes myeloid cell development and maturation. The technology is filed through the Icahn School of Medicine at Mount Sinai (ISMMS) and is currently unlicensed, Inventor on a patent application NY-ESO-1 Polypeptides filed through Mount Sinai and Ludwig Institute. This technology is currently unlicensed. HTM—Consulting Fees: Caris Life Sciences, Cytek Biosciences; Stock Holdings: Caris Life Sciences, Cytek Biosciences. LCo and JYPS—Nothing to disclose. SITC Staff (PJI and ER)—Nothing to disclose.
- Published
- 2024
- Full Text
- View/download PDF
3. The multifunctional protein HMGB1: 50 years of discovery.
- Author
-
Tang D, Kang R, Zeh HJ, and Lotze MT
- Subjects
- Humans, Cell Death, Immunity, Innate, Signal Transduction, HMGB1 Protein chemistry, HMGB1 Protein metabolism, Neoplasms
- Abstract
Fifty years since the initial discovery of HMGB1 in 1973 as a structural protein of chromatin, HMGB1 is now known to regulate diverse biological processes depending on its subcellular or extracellular localization. These functions include promoting DNA damage repair in the nucleus, sensing nucleic acids and inducing innate immune responses and autophagy in the cytosol and binding protein partners in the extracellular environment and stimulating immunoreceptors. In addition, HMGB1 is a broad sensor of cellular stress that balances cell death and survival responses essential for cellular homeostasis and tissue maintenance. HMGB1 is also an important mediator secreted by immune cells that is involved in a range of pathological conditions, including infectious diseases, ischaemia-reperfusion injury, autoimmunity, cardiovascular and neurodegenerative diseases, metabolic disorders and cancer. In this Review, we discuss the signalling mechanisms, cellular functions and clinical relevance of HMGB1 and describe strategies to modify its release and biological activities in the setting of various diseases., (© 2023. Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
4. Parkin Deficiency Suppresses Antigen Presentation to Promote Tumor Immune Evasion and Immunotherapy Resistance.
- Author
-
Perales-Linares R, Leli NM, Mohei H, Beghi S, Rivera OD, Kostopoulos N, Giglio A, George SS, Uribe-Herranz M, Costabile F, Pierini S, Pustylnikov S, Skoufos G, Barash Y, Hatzigeorgiou AG, Koumenis C, Maity A, Lotze MT, and Facciabene A
- Subjects
- Animals, Humans, Mice, Immunotherapy, Proto-Oncogene Proteins c-akt, Tumor Escape, Ubiquitin-Protein Ligases genetics, Ubiquitin-Protein Ligases metabolism, Antigen Presentation, Neoplasms genetics, Neoplasms therapy, Neoplasms metabolism
- Abstract
Parkin is an E3 ubiquitin ligase, which plays a key role in the development of Parkinson disease. Parkin defects also occur in numerous cancers, and a growing body of evidence indicates that Parkin functions as a tumor suppressor that impedes a number of cellular processes involved in tumorigenesis. Here, we generated murine and human models that closely mimic the advanced-stage tumors where Parkin deficiencies are found to provide deeper insights into the tumor suppressive functions of Parkin. Loss of Parkin expression led to aggressive tumor growth, which was associated with poor tumor antigen presentation and limited antitumor CD8+ T-cell infiltration and activation. The effect of Parkin deficiency on tumor growth was lost following depletion of CD8+ T cells. In line with previous findings, Parkin deficiency was linked with mitochondria-associated metabolic stress, PTEN degradation, and enhanced Akt activation. Increased Akt signaling led to dysregulation of antigen presentation, and treatment with the Akt inhibitor MK2206-2HCl restored antigen presentation in Parkin-deficient tumors. Analysis of data from patients with clear cell renal cell carcinoma indicated that Parkin expression was downregulated in tumors and that low expression correlated with reduced overall survival. Furthermore, low Parkin expression correlated with reduced patient response to immunotherapy. Overall, these results identify a role for Parkin deficiency in promoting tumor immune evasion that may explain the poor prognosis associated with loss of Parkin across multiple types of cancer., Significance: Parkin prevents immune evasion by regulating tumor antigen processing and presentation through the PTEN/Akt network, which has important implications for immunotherapy treatments in patients with Parkin-deficient tumors., (©2023 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
5. AllergoOncology: Danger signals in allergology and oncology: A European Academy of Allergy and Clinical Immunology (EAACI) Position Paper.
- Author
-
Bergmann C, Poli A, Agache I, Bianchini R, Bax HJ, Castells M, Crescioli S, Dombrowicz D, Ferastraoaru D, Fiebiger E, Gould HJ, Hartmann K, Izquierdo E, Jordakieva G, Josephs DH, Jutel M, Levi-Schaffer F, de Las Vecillas L, Lotze MT, Osborn G, Pascal M, Redegeld F, Rosenstreich D, Roth-Walter F, Schmidt-Weber C, Shamji M, Steveling EH, Turner MC, Untersmayr E, Jensen-Jarolim E, and Karagiannis SN
- Subjects
- Humans, Immunity, Inflammation, Signal Transduction, Hypersensitivity diagnosis, Hypersensitivity etiology, Hypersensitivity therapy, Neoplasms etiology, Neoplasms therapy
- Abstract
The immune system interacts with many nominal 'danger' signals, endogenous danger-associated (DAMP), exogenous pathogen (PAMP) and allergen (AAMP)-associated molecular patterns. The immune context under which these are received can promote or prevent immune activating or inflammatory mechanisms and may orchestrate diverse immune responses in allergy and cancer. Each can act either by favouring a respective pathology or by supporting the immune response to confer protective effects, depending on acuity or chronicity. In this Position Paper under the collective term danger signals or DAMPs, PAMPs and AAMPs, we consider their diverse roles in allergy and cancer and the connection between these in AllergoOncology. We focus on their interactions with different immune cells of the innate and adaptive immune system and how these promote immune responses with juxtaposing clinical outcomes in allergy and cancer. While danger signals present potential targets to overcome inflammatory responses in allergy, these may be reconsidered in relation to a history of allergy, chronic inflammation and autoimmunity linked to the risk of developing cancer, and with regard to clinical responses to anti-cancer immune and targeted therapies. Cross-disciplinary insights in AllergoOncology derived from dissecting clinical phenotypes of common danger signal pathways may improve allergy and cancer clinical outcomes., (© 2022 The Authors. Allergy published by European Academy of Allergy and Clinical Immunology and John Wiley & Sons Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
6. Characteristics of Malignant Pleural Effusion Resident CD8 + T Cells from a Heterogeneous Collection of Tumors.
- Author
-
Dhupar R, Okusanya OT, Eisenberg SH, Monaco SE, Ruffin AT, Liu D, Luketich JD, Kammula US, Bruno TC, Lotze MT, and Soloff AC
- Subjects
- Aged, Aged, 80 and over, CD8-Positive T-Lymphocytes metabolism, CD8-Positive T-Lymphocytes pathology, Cell Differentiation, Female, Gene Expression Regulation, Neoplastic, Humans, Interleukin-7 Receptor alpha Subunit metabolism, L-Lactate Dehydrogenase metabolism, Lactic Acid metabolism, Leukocyte Common Antigens metabolism, Male, Middle Aged, Myeloid-Derived Suppressor Cells metabolism, Myeloid-Derived Suppressor Cells pathology, Neoplasm Staging, Neoplasms complications, Neoplasms immunology, Pleural Effusion, Malignant etiology, Pleural Effusion, Malignant immunology, Receptors, CCR7 metabolism, Tumor Cells, Cultured, CD8-Positive T-Lymphocytes cytology, Coculture Techniques methods, Interferon-gamma metabolism, Neoplasms pathology, Pleural Effusion, Malignant pathology
- Abstract
While T cell-based cancer immunotherapies have shown great promise, there remains a need to understand how individual metastatic tumor environments impart local T cell dysfunction. At advanced stages, cancers that metastasize to the pleural space can result in a malignant pleural effusion (MPE) that harbors abundant tumor and immune cells, often exceeding 10
8 leukocytes per liter. Unlike other metastatic sites, MPEs are readily and repeatedly accessible via indwelling catheters, providing an opportunity to study the interface between tumor dynamics and immunity. In the current study, we examined CD8+ T cells within MPEs collected from patients with heterogeneous primary tumors and at various stages in treatment to determine (1) if these cells possess anti-tumor activity following removal from the MPE, (2) factors in the MPE that may contribute to their dysfunction, and (3) the phenotypic changes in T cell populations that occur following ex vivo expansion. Co-cultures of CD8+ T cells with autologous CD45- tumor containing cells demonstrated cytotoxicity ( p = 0.030) and IFNγ production ( p = 0.003) that inversely correlated with percent of myeloid derived suppressor cells, lactate, and lactate dehydrogenase (LDH) within the MPE. Ex vivo expansion of CD8+ T cells resulted in progressive differentiation marked by distinct populations expressing decreased CD45RA, CCR7, CD127, and increased inhibitory receptors. These findings suggest that MPEs may be a source of tumor-reactive T cells and that the cellular and acellular components suppress optimal function.- Published
- 2020
- Full Text
- View/download PDF
7. The Adaptome as Biomarker for Assessing Cancer Immunity and Immunotherapy.
- Author
-
Han J and Lotze MT
- Subjects
- Adaptive Immunity, Biomarkers, Tumor genetics, High-Throughput Nucleotide Sequencing, Humans, Immunotherapy, Multiplex Polymerase Chain Reaction, Neoplasms immunology, Neoplasms therapy, Tumor Escape, Neoplasms genetics, Receptors, Antigen, B-Cell genetics, Receptors, Antigen, T-Cell genetics
- Abstract
In terms of diagnosing and treating diseases, our adaptive immune system is the "best doctor." It carries out these tasks with unmatched precision, with the help of both T and B cell receptors, our most diverse set of genes, distinguishing one individual from another. It does this by generating autologous extraordinary diversity in the receptors, ranging from 10
15 to 1025 for each chain of the rearranged receptors. By combining multiplex PCR and next-generation sequencing (NGS), we have developed high throughput methods to study adaptive immunity. The adaptome is the sum-total of expressed T and B cell receptor genes in a sample, composed of seven chains, including the alpha/beta and gamma/delta chains for T cells, and heavy/lambda or kappa chains for B cells. Immune repertoire is the sum-total of the individual clonotypes within one chain, including individual complementarity-determining regions (CDR) 3 sequences. In order to reflect the breadth and depth of the true adaptome, the following criteria assessing any method needs to be ascertained: (1) Methods need to be inclusive and quantitative; (2) Analysis should consider what questions need to be addressed and whether bulk or single cell sequencing provide the best tools for assessing the underlying biology and addressing important questions; (3) Measures of clonal diversity are key to understand the underlying structure and providence of the repertoire; and (4) Convergent evolution may allow a surprising degree of homologous or identical CDR3s associated with individual disease entities, creating hope for novel diagnostics and/or disease burden assessments. Integrating studies of the peripheral blood, lymph nodes, and tumor allows dynamic interrogation of the alterations occurring with age, treatment, and response to emergent and established therapies.- Published
- 2020
- Full Text
- View/download PDF
8. Clockophagy is a novel selective autophagy process favoring ferroptosis.
- Author
-
Yang M, Chen P, Liu J, Zhu S, Kroemer G, Klionsky DJ, Lotze MT, Zeh HJ, Kang R, and Tang D
- Subjects
- Animals, Apoptosis genetics, Biological Clocks genetics, Gene Expression Regulation, Neoplastic genetics, HL-60 Cells, Heterografts, Humans, Hypoxia-Inducible Factor 1, alpha Subunit genetics, Hypoxia-Inducible Factor-Proline Dioxygenases genetics, Iron metabolism, Lipid Peroxidation genetics, Lysosomes genetics, Mice, Neoplasms pathology, Reactive Oxygen Species metabolism, Sequestosome-1 Protein genetics, Signal Transduction genetics, ARNTL Transcription Factors genetics, Autophagy genetics, Ferroptosis genetics, Neoplasms genetics
- Abstract
Ferroptosis is a form of nonapoptotic regulated cell death driven by iron-dependent lipid peroxidation. Autophagy involves a lysosomal degradation pathway that can either promote or impede cell death. A high level of autophagy has been associated with ferroptosis, but the mechanisms underpinning this relationship are largely elusive. We characterize the contribution of autophagy to ferroptosis in human cancer cell lines and mouse tumor models. We show that "clockophagy," the selective degradation of the core circadian clock protein ARNTL by autophagy, is critical for ferroptosis. We identify SQSTM1 as a cargo receptor responsible for autophagic ARNTL degradation. ARNTL inhibits ferroptosis by repressing the transcription of Egln2 , thus activating the prosurvival transcription factor HIF1A. Genetic or pharmacological interventions blocking ARNTL degradation or inhibiting EGLN2 activation diminished, whereas destabilizing HIF1A facilitated, ferroptotic tumor cell death. Thus, our findings reveal a new pathway, initiated by the autophagic removal of ARNTL, that facilitates ferroptosis induction.
- Published
- 2019
- Full Text
- View/download PDF
9. Johnny on the Spot-Chronic Inflammation Is Driven by HMGB1.
- Author
-
Gorgulho CM, Romagnoli GG, Bharthi R, and Lotze MT
- Subjects
- Alarmins metabolism, Autoimmune Diseases immunology, Autoimmune Diseases metabolism, Chronic Disease, Exosomes immunology, Exosomes metabolism, HMGB1 Protein metabolism, Humans, Infections metabolism, Inflammation metabolism, Myeloid-Derived Suppressor Cells immunology, Myeloid-Derived Suppressor Cells metabolism, Neoplasms metabolism, Alarmins immunology, HMGB1 Protein immunology, Infections immunology, Inflammation immunology, Neoplasms immunology
- Abstract
Although much has been made of the role of HMGB1 acting as an acute damage associated molecular pattern (DAMP) molecule, prompting the response to tissue damage or injury, it is also released at sites of chronic inflammation including sites of infection, autoimmunity, and cancer. As such, the biology is distinguished from homeostasis and acute inflammation by the recruitment and persistence of myeloid derived suppressor cells, T regulatory cells, fibrosis and/or exuberant angiogenesis depending on the antecedents and the other individual inflammatory partners that HMGB1 binds and focuses, including IL-1β, CXCL12/SDF1, LPS, DNA, RNA, and sRAGE. High levels of HMGB1 released into the extracellular milieu and its persistence in the microenvironment can contribute to the pathogenesis of many if not all autoimmune disorders and is a key factor that drives inflammation further and worsens symptoms. HMGB1 is also pivotal in the maintenance of chronic inflammation and a "wound healing" type of immune response that ultimately contributes to the onset of carcinogenesis and tumor progression. Exosomes carrying HMGB1 and other instructive molecules are released and shape the response of various cells in the chronic inflammatory environment. Understanding the defining roles of REDOX, DAMPs and PAMPs, and the host response in chronic inflammation requires an alternative means for positing HMGB1's central role in limiting and focusing inflammation, distinguishing chronic from acute inflammation.
- Published
- 2019
- Full Text
- View/download PDF
10. Toward a comprehensive view of cancer immune responsiveness: a synopsis from the SITC workshop.
- Author
-
Bedognetti D, Ceccarelli M, Galluzzi L, Lu R, Palucka K, Samayoa J, Spranger S, Warren S, Wong KK, Ziv E, Chowell D, Coussens LM, De Carvalho DD, DeNardo DG, Galon J, Kaufman HL, Kirchhoff T, Lotze MT, Luke JJ, Minn AJ, Politi K, Shultz LD, Simon R, Thórsson V, Weidhaas JB, Ascierto ML, Ascierto PA, Barnes JM, Barsan V, Bommareddy PK, Bot A, Church SE, Ciliberto G, De Maria A, Draganov D, Ho WS, McGee HM, Monette A, Murphy JF, Nisticò P, Park W, Patel M, Quigley M, Radvanyi L, Raftopoulos H, Rudqvist NP, Snyder A, Sweis RF, Valpione S, Zappasodi R, Butterfield LH, Disis ML, Fox BA, Cesano A, and Marincola FM
- Subjects
- Advisory Committees, Animals, Biomarkers, Tumor genetics, Biomarkers, Tumor immunology, Congresses as Topic, Disease Models, Animal, Humans, Medical Oncology organization & administration, Neoplasms genetics, Neoplasms immunology, Societies, Medical organization & administration, Treatment Outcome, Tumor Microenvironment genetics, Immunotherapy, Neoplasms therapy, Tumor Microenvironment immunology
- Abstract
Tumor immunology has changed the landscape of cancer treatment. Yet, not all patients benefit as cancer immune responsiveness (CIR) remains a limitation in a considerable proportion of cases. The multifactorial determinants of CIR include the genetic makeup of the patient, the genomic instability central to cancer development, the evolutionary emergence of cancer phenotypes under the influence of immune editing, and external modifiers such as demographics, environment, treatment potency, co-morbidities and cancer-independent alterations including immune homeostasis and polymorphisms in the major and minor histocompatibility molecules, cytokines, and chemokines. Based on the premise that cancer is fundamentally a disorder of the genes arising within a cell biologic process, whose deviations from normality determine the rules of engagement with the host's response, the Society for Immunotherapy of Cancer (SITC) convened a task force of experts from various disciplines including, immunology, oncology, biophysics, structural biology, molecular and cellular biology, genetics, and bioinformatics to address the complexity of CIR from a holistic view. The task force was launched by a workshop held in San Francisco on May 14-15, 2018 aimed at two preeminent goals: 1) to identify the fundamental questions related to CIR and 2) to create an interactive community of experts that could guide scientific and research priorities by forming a logical progression supported by multiple perspectives to uncover mechanisms of CIR. This workshop was a first step toward a second meeting where the focus would be to address the actionability of some of the questions identified by working groups. In this event, five working groups aimed at defining a path to test hypotheses according to their relevance to human cancer and identifying experimental models closest to human biology, which include: 1) Germline-Genetic, 2) Somatic-Genetic and 3) Genomic-Transcriptional contributions to CIR, 4) Determinant(s) of Immunogenic Cell Death that modulate CIR, and 5) Experimental Models that best represent CIR and its conversion to an immune responsive state. This manuscript summarizes the contributions from each group and should be considered as a first milestone in the path toward a more contemporary understanding of CIR. We appreciate that this effort is far from comprehensive and that other relevant aspects related to CIR such as the microbiome, the individual's recombined T cell and B cell receptors, and the metabolic status of cancer and immune cells were not fully included. These and other important factors will be included in future activities of the taskforce. The taskforce will focus on prioritization and specific actionable approach to answer the identified questions and implementing the collaborations in the follow-up workshop, which will be held in Houston on September 4-5, 2019.
- Published
- 2019
- Full Text
- View/download PDF
11. Different measures of HMGB1 location in cancer immunology.
- Author
-
Mendonça Gorgulho C, Murthy P, Liotta L, Espina V, and Lotze MT
- Subjects
- Animals, Antineoplastic Agents, Immunological pharmacology, Antineoplastic Agents, Immunological therapeutic use, Apoptosis drug effects, Apoptosis immunology, Autophagy immunology, Biomarkers, Tumor immunology, Biomarkers, Tumor metabolism, Carcinogenesis drug effects, Carcinogenesis immunology, Carcinogenesis pathology, Cell Nucleus immunology, Cell Nucleus metabolism, Cytosol immunology, Cytosol metabolism, Disease Progression, Extracellular Space immunology, Extracellular Space metabolism, HMGB1 Protein immunology, HMGB1 Protein metabolism, Humans, Immunogenic Cell Death drug effects, Immunogenic Cell Death immunology, Neoplasms drug therapy, Neoplasms pathology, Tumor Microenvironment immunology, Biomarkers, Tumor analysis, HMGB1 Protein analysis, Neoplasms immunology
- Abstract
HMGB1 is the most abundant non-histone nuclear protein. It regulates transcriptional access to open areas of chromatin and limits release of DNA with apoptotic death, serving to both inhibit apoptosis and promote DNA repair. When HMGB1 is translocated to the cytosol with many types of cellular stress, it is a powerful inducer of autophagy. It can also be released by activated immune cells and damaged or dying cells into the extracellular space, where it acts as a damage associated molecular pattern (DAMP) molecule, contributing to the pathogenesis and progression of cancer. Here, the most common methodologies to not only measure HMGB1 but also to effectively determine its subcellular localization, which dictates many of HMGB1's different functions, are reviewed., (© 2019 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
12. Bortezomib Treatment Sensitizes Oncolytic HSV-1-Treated Tumors to NK Cell Immunotherapy.
- Author
-
Yoo JY, Jaime-Ramirez AC, Bolyard C, Dai H, Nallanagulagari T, Wojton J, Hurwitz BS, Relation T, Lee TJ, Lotze MT, Yu JG, Zhang J, Croce CM, Yu J, Caligiuri MA, Old M, and Kaur B
- Subjects
- Animals, Caspase 3 metabolism, Caspase 7 metabolism, Cell Death drug effects, Cell Death immunology, Cell Line, Tumor, Combined Modality Therapy methods, Female, Humans, Immunotherapy methods, Killer Cells, Natural metabolism, MAP Kinase Kinase 4 metabolism, Mice, Mice, Nude, Neoplasms immunology, Neoplasms metabolism, Oncolytic Virotherapy methods, Reactive Oxygen Species metabolism, Receptor-Interacting Protein Serine-Threonine Kinases metabolism, Xenograft Model Antitumor Assays methods, Bortezomib pharmacology, Herpesvirus 1, Human immunology, Killer Cells, Natural drug effects, Killer Cells, Natural immunology, Neoplasms drug therapy, Neoplasms therapy, Oncolytic Viruses immunology
- Abstract
Purpose: Both the proteasome inhibitor bortezomib and an oncolytic herpes simplex virus-1 (oHSV)-expressing GM-CSF are currently FDA approved. Although proteasome blockade can increase oHSV replication, immunologic consequences, and consequent immunotherapy potential are unknown. In this study, we investigated the impact of bortezomib combined with oHSV on tumor cell death and sensitivity to natural killer (NK) cell immunotherapy., Experimental Design: Western blot, flow cytometry, and caspase 3/7 activity assays were used to evaluate the induction of apoptosis/autophagy and/or necroptotic cell death. Cellular and mitochondrial reactive oxygen species (ROS) production was measured using CellROX and MitoSOX. Inhibitors/shRNA-targeting ROS, JNK and RIP1 kinase (RIPK1) were used to investigate the mechanism of cell killing. The synergistic interaction between oHSV and bortezomib was calculated using a Chou-Talalay analysis. NK cells isolated from normal human blood were co-cultured with tumor cells to evaluate cellular interactions. Q-PCR, ELISA, and FACS analysis were used to evaluate NK cell activation. Intracranial tumor xenografts were used to evaluate antitumor efficacy., Results: Combination treatment with bortezomib- and oHSV-induced necroptotic cell death and increased the production of mitochondrial ROS and JNK phosphorylation. Inhibitors/shRNA of RIPK1 and JNK rescued synergistic cell killing. Combination treatment also significantly enhanced NK cell activation and adjuvant NK cell therapy of mice treated with bortezomib and oHSV improved antitumor efficacy., Conclusions: This study provides a significant rationale for triple combination therapy with bortezomib, oHSV, and NK cells to improve efficacy, in glioblastoma patients. Clin Cancer Res; 22(21); 5265-76. ©2016 AACRSee related commentary by Suryadevara et al., p. 5164., (©2016 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
13. DAMPs, ageing, and cancer: The 'DAMP Hypothesis'.
- Author
-
Huang J, Xie Y, Sun X, Zeh HJ 3rd, Kang R, Lotze MT, and Tang D
- Subjects
- Animals, Humans, Aging metabolism, Aging pathology, Neoplasms metabolism, Neoplasms pathology, Receptors, Pattern Recognition metabolism
- Abstract
Ageing is a complex and multifactorial process characterized by the accumulation of many forms of damage at the molecular, cellular, and tissue level with advancing age. Ageing increases the risk of the onset of chronic inflammation-associated diseases such as cancer, diabetes, stroke, and neurodegenerative disease. In particular, ageing and cancer share some common origins and hallmarks such as genomic instability, epigenetic alteration, aberrant telomeres, inflammation and immune injury, reprogrammed metabolism, and degradation system impairment (including within the ubiquitin-proteasome system and the autophagic machinery). Recent advances indicate that damage-associated molecular pattern molecules (DAMPs) such as high mobility group box 1, histones, S100, and heat shock proteins play location-dependent roles inside and outside the cell. These provide interaction platforms at molecular levels linked to common hallmarks of ageing and cancer. They can act as inducers, sensors, and mediators of stress through individual plasma membrane receptors, intracellular recognition receptors (e.g., advanced glycosylation end product-specific receptors, AIM2-like receptors, RIG-I-like receptors, and NOD1-like receptors, and toll-like receptors), or following endocytic uptake. Thus, the DAMP Hypothesis is novel and complements other theories that explain the features of ageing. DAMPs represent ideal biomarkers of ageing and provide an attractive target for interventions in ageing and age-associated diseases., (Copyright © 2014 Elsevier B.V. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
14. Parkinson Disease and Malignant Disease: Minding Cancer's Own Business.
- Author
-
Ganguli M and Lotze MT
- Subjects
- Female, Humans, Male, Asian People, Neoplasms ethnology, Parkinson Disease ethnology
- Published
- 2015
- Full Text
- View/download PDF
15. Nuclear DAMP complex-mediated RAGE-dependent macrophage cell death.
- Author
-
Chen R, Fu S, Fan XG, Lotze MT, Zeh HJ 3rd, Tang D, and Kang R
- Subjects
- Animals, Cell Death, Cell Line, Cell Line, Tumor, Cells, Cultured, DNA immunology, Histones immunology, Humans, Inflammation immunology, Mice, Proto-Oncogene Proteins c-akt immunology, Receptor for Advanced Glycation End Products, Toll-Like Receptor 2 immunology, Toll-Like Receptor 4 immunology, Tumor Necrosis Factor-alpha immunology, HMGB1 Protein immunology, Macrophages cytology, Macrophages immunology, Neoplasms immunology, Receptors, Immunologic immunology
- Abstract
High mobility group box 1 (HMGB1), histone, and DNA are essential nuclear components involved in the regulation of chromosome structure and function. In addition to their nuclear function, these molecules act as damage-associated molecular patterns (DAMPs) alone or together when released extracellularly. The synergistic effect of these nuclear DNA-HMGB1-histone complexes as DAMP complexes (nDCs) on immune cells remains largely unexplored. Here, we demonstrate that nDCs limit survival of macrophages (e.g., RAW264.7 and peritoneal macrophages) but not cancer cells (e.g., HCT116, HepG2 and Hepa1-6). nDCs promote production of inflammatory tumor necrosis factor α (TNFα) release, triggering reactive oxygen species-dependent apoptosis and necrosis. Moreover, the receptor for advanced glycation end products (RAGE), but not toll-like receptor (TLR)-4 and TLR-2, was required for Akt-dependent TNFα release and subsequent cell death following treatment with nDCs. Genetic depletion of RAGE by RNAi, antioxidant N-Acetyl-l-cysteine, and TNFα neutralizing antibody significantly attenuated nDC-induced cell death. These findings provide evidence supporting novel signaling mechanisms linking nDCs and inflammation in macrophage cell death., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
16. Classification of current anticancer immunotherapies.
- Author
-
Galluzzi L, Vacchelli E, Bravo-San Pedro JM, Buqué A, Senovilla L, Baracco EE, Bloy N, Castoldi F, Abastado JP, Agostinis P, Apte RN, Aranda F, Ayyoub M, Beckhove P, Blay JY, Bracci L, Caignard A, Castelli C, Cavallo F, Celis E, Cerundolo V, Clayton A, Colombo MP, Coussens L, Dhodapkar MV, Eggermont AM, Fearon DT, Fridman WH, Fučíková J, Gabrilovich DI, Galon J, Garg A, Ghiringhelli F, Giaccone G, Gilboa E, Gnjatic S, Hoos A, Hosmalin A, Jäger D, Kalinski P, Kärre K, Kepp O, Kiessling R, Kirkwood JM, Klein E, Knuth A, Lewis CE, Liblau R, Lotze MT, Lugli E, Mach JP, Mattei F, Mavilio D, Melero I, Melief CJ, Mittendorf EA, Moretta L, Odunsi A, Okada H, Palucka AK, Peter ME, Pienta KJ, Porgador A, Prendergast GC, Rabinovich GA, Restifo NP, Rizvi N, Sautès-Fridman C, Schreiber H, Seliger B, Shiku H, Silva-Santos B, Smyth MJ, Speiser DE, Spisek R, Srivastava PK, Talmadge JE, Tartour E, Van Der Burg SH, Van Den Eynde BJ, Vile R, Wagner H, Weber JS, Whiteside TL, Wolchok JD, Zitvogel L, Zou W, and Kroemer G
- Subjects
- Animals, Humans, Immunotherapy methods, Neoplasms immunology, Neoplasms therapy
- Abstract
During the past decades, anticancer immunotherapy has evolved from a promising therapeutic option to a robust clinical reality. Many immunotherapeutic regimens are now approved by the US Food and Drug Administration and the European Medicines Agency for use in cancer patients, and many others are being investigated as standalone therapeutic interventions or combined with conventional treatments in clinical studies. Immunotherapies may be subdivided into "passive" and "active" based on their ability to engage the host immune system against cancer. Since the anticancer activity of most passive immunotherapeutics (including tumor-targeting monoclonal antibodies) also relies on the host immune system, this classification does not properly reflect the complexity of the drug-host-tumor interaction. Alternatively, anticancer immunotherapeutics can be classified according to their antigen specificity. While some immunotherapies specifically target one (or a few) defined tumor-associated antigen(s), others operate in a relatively non-specific manner and boost natural or therapy-elicited anticancer immune responses of unknown and often broad specificity. Here, we propose a critical, integrated classification of anticancer immunotherapies and discuss the clinical relevance of these approaches.
- Published
- 2014
- Full Text
- View/download PDF
17. HMGB1 in cancer: good, bad, or both?
- Author
-
Kang R, Zhang Q, Zeh HJ 3rd, Lotze MT, and Tang D
- Subjects
- Apoptosis genetics, Autophagy genetics, Cell Nucleus metabolism, Cell Survival genetics, Chemokines metabolism, Cytokines metabolism, HMGB1 Protein metabolism, Humans, Intercellular Signaling Peptides and Proteins metabolism, Neoplasms pathology, Neoplasms therapy, HMGB1 Protein genetics, Neoplasms genetics, Signal Transduction genetics
- Abstract
Forty years ago, high mobility group box 1 (HMGB1) was discovered in calf thymus and named according to its electrophoretic mobility in polyacrylamide gels. Now, we know that HMGB1 performs dual functions. Inside the cell, HMGB1 is a highly conserved chromosomal protein acting as a DNA chaperone. Outside of the cell, HMGB1 is a prototypical damage-associated molecular pattern, acting with cytokines, chemokines, and growth factors. During tumor development and in cancer therapy, HMGB1 has been reported to play paradoxical roles in promoting both cell survival and death by regulating multiple signaling pathways, including inflammation, immunity, genome stability, proliferation, metastasis, metabolism, apoptosis, and autophagy. Here, we review the current knowledge of both HMGB1's oncogenic and tumor-suppressive roles and the potential strategies that target HMGB1 for the prevention and treatment of cancer., (©2013 AACR.)
- Published
- 2013
- Full Text
- View/download PDF
18. Lymphatics, lymph nodes and the immune system: barriers and gateways for cancer spread.
- Author
-
Ferris RL, Lotze MT, Leong SP, Hoon DS, and Morton DL
- Subjects
- Antigens, Neoplasm analysis, Antigens, Neoplasm genetics, B7 Antigens biosynthesis, B7 Antigens metabolism, Humans, Immune System physiology, Lymph Nodes pathology, Lymphangiogenesis, Neoplasm Micrometastasis, Tumor Microenvironment, Lymphatic Metastasis pathology, Lymphatic System pathology, Neoplasms immunology, Neoplasms pathology
- Abstract
Metastasis to the regional lymph node is the most important prognostic indicator for the outcomes of patients with sold cancer. In general, it is well recognized that cancer development is genetically determined with progression from the microenvironment of the primary tumor site, oftentimes via the SLN gateway, to the distant sites. In about 20 % of the time, the cancer cells may spread directly through the blood vascular system to the distant sites. Thus, in general, cancer progression is consistent with Hellman's spectrum theory in that development of nodal and systemic metastasis from a localized cancer growth is a progressive process. Cancer proliferation within the tumor microenvironment may give rise to increased tumor heterogeneity, which is further complicated by its continuous change through its evolution within the host in a Darwinian sense. It is crucial to understand the molecular process of lymphangiogenesis and hemangiogenesis in the tumor microenvironment with respect to the initial steps of cancer cells entering into the lymphatic and vascular systems so that rational therapy can be developed to curb the process of specific routes of metastasis. This chapter elucidates the role of lymphatics, nodal metastasis and antitumor immunity. We present novel immune targets in nodal metastases, the importance of the lymph node as a pre-metastatic niche, and immune-related proteins as biomarkers of metastasis.
- Published
- 2012
- Full Text
- View/download PDF
19. Tumor immunity times out: TIM-3 and HMGB1.
- Author
-
Tang D and Lotze MT
- Subjects
- Animals, Hepatitis A Virus Cellular Receptor 2, Humans, Dendritic Cells immunology, HMGB1 Protein immunology, Immunity, Innate, Neoplasms immunology, Nucleic Acids immunology, Receptors, Virus immunology, Tumor Microenvironment immunology
- Published
- 2012
- Full Text
- View/download PDF
20. Tumor-cell death, autophagy, and immunity.
- Author
-
Weiner LM and Lotze MT
- Subjects
- Animals, Autophagy immunology, Cell Death, Humans, Mice, Neoplasms metabolism, Neoplasms physiopathology, Adenosine Triphosphate metabolism, Autophagy physiology, Neoplasms immunology
- Published
- 2012
- Full Text
- View/download PDF
21. Defining the critical hurdles in cancer immunotherapy.
- Author
-
Fox BA, Schendel DJ, Butterfield LH, Aamdal S, Allison JP, Ascierto PA, Atkins MB, Bartunkova J, Bergmann L, Berinstein N, Bonorino CC, Borden E, Bramson JL, Britten CM, Cao X, Carson WE, Chang AE, Characiejus D, Choudhury AR, Coukos G, de Gruijl T, Dillman RO, Dolstra H, Dranoff G, Durrant LG, Finke JH, Galon J, Gollob JA, Gouttefangeas C, Grizzi F, Guida M, Håkansson L, Hege K, Herberman RB, Hodi FS, Hoos A, Huber C, Hwu P, Imai K, Jaffee EM, Janetzki S, June CH, Kalinski P, Kaufman HL, Kawakami K, Kawakami Y, Keilholtz U, Khleif SN, Kiessling R, Kotlan B, Kroemer G, Lapointe R, Levitsky HI, Lotze MT, Maccalli C, Maio M, Marschner JP, Mastrangelo MJ, Masucci G, Melero I, Melief C, Murphy WJ, Nelson B, Nicolini A, Nishimura MI, Odunsi K, Ohashi PS, O'Donnell-Tormey J, Old LJ, Ottensmeier C, Papamichail M, Parmiani G, Pawelec G, Proietti E, Qin S, Rees R, Ribas A, Ridolfi R, Ritter G, Rivoltini L, Romero PJ, Salem ML, Scheper RJ, Seliger B, Sharma P, Shiku H, Singh-Jasuja H, Song W, Straten PT, Tahara H, Tian Z, van Der Burg SH, von Hoegen P, Wang E, Welters MJ, Winter H, Withington T, Wolchok JD, Xiao W, Zitvogel L, Zwierzina H, Marincola FM, Gajewski TF, Wigginton JM, and Disis ML
- Subjects
- Humans, International Cooperation, Translational Research, Biomedical, Immunotherapy, Neoplasms therapy
- Abstract
Scientific discoveries that provide strong evidence of antitumor effects in preclinical models often encounter significant delays before being tested in patients with cancer. While some of these delays have a scientific basis, others do not. We need to do better. Innovative strategies need to move into early stage clinical trials as quickly as it is safe, and if successful, these therapies should efficiently obtain regulatory approval and widespread clinical application. In late 2009 and 2010 the Society for Immunotherapy of Cancer (SITC), convened an "Immunotherapy Summit" with representatives from immunotherapy organizations representing Europe, Japan, China and North America to discuss collaborations to improve development and delivery of cancer immunotherapy. One of the concepts raised by SITC and defined as critical by all parties was the need to identify hurdles that impede effective translation of cancer immunotherapy. With consensus on these hurdles, international working groups could be developed to make recommendations vetted by the participating organizations. These recommendations could then be considered by regulatory bodies, governmental and private funding agencies, pharmaceutical companies and academic institutions to facilitate changes necessary to accelerate clinical translation of novel immune-based cancer therapies. The critical hurdles identified by representatives of the collaborating organizations, now organized as the World Immunotherapy Council, are presented and discussed in this report. Some of the identified hurdles impede all investigators; others hinder investigators only in certain regions or institutions or are more relevant to specific types of immunotherapy or first-in-humans studies. Each of these hurdles can significantly delay clinical translation of promising advances in immunotherapy yet if overcome, have the potential to improve outcomes of patients with cancer.
- Published
- 2011
- Full Text
- View/download PDF
22. Principles and current strategies for targeting autophagy for cancer treatment.
- Author
-
Amaravadi RK, Lippincott-Schwartz J, Yin XM, Weiss WA, Takebe N, Timmer W, DiPaola RS, Lotze MT, and White E
- Subjects
- Animals, Clinical Trials, Phase III as Topic, Combined Modality Therapy, Cytoplasmic Vesicles metabolism, Humans, Immune Tolerance, Immunotherapy, Inflammation pathology, Metabolic Networks and Pathways drug effects, Neoplasms immunology, Neoplasms therapy, Proteasome Endopeptidase Complex metabolism, Signal Transduction drug effects, Antineoplastic Agents therapeutic use, Autophagy drug effects, Hydroxychloroquine therapeutic use, Molecular Targeted Therapy, Neoplasms pathology
- Abstract
Autophagy is an evolutionarily conserved, intracellular self-defense mechanism in which organelles and proteins are sequestered into autophagic vesicles that are subsequently degraded through fusion with lysosomes. Cells, thereby, prevent the toxic accumulation of damaged or unnecessary components, but also recycle these components to sustain metabolic homoeostasis. Heightened autophagy is a mechanism of resistance for cancer cells faced with metabolic and therapeutic stress, revealing opportunities for exploitation as a therapeutic target in cancer. We summarize recent developments in the field of autophagy and cancer and build upon the results presented at the Cancer Therapy Evaluation Program (CTEP) Early Drug Development meeting in March 2010. Herein, we describe our current understanding of the core components of the autophagy machinery and the functional relevance of autophagy within the tumor microenvironment, and we outline how this knowledge has informed preclinical investigations combining the autophagy inhibitor hydroxychloroquine (HCQ) with chemotherapy, targeted therapy, and immunotherapy. Finally, we describe ongoing clinical trials involving HCQ as a first generation autophagy inhibitor, as well as strategies for the development of novel, more potent, and specific inhibitors of autophagy., (©2011 AACR.)
- Published
- 2011
- Full Text
- View/download PDF
23. Zinc in innate and adaptive tumor immunity.
- Author
-
John E, Laskow TC, Buchser WJ, Pitt BR, Basse PH, Butterfield LH, Kalinski P, and Lotze MT
- Subjects
- Apoptosis, Humans, Signal Transduction, Zinc deficiency, Zinc metabolism, Adaptive Immunity immunology, Immunity, Innate immunology, Neoplasms immunology, Zinc immunology
- Abstract
Zinc is important. It is the second most abundant trace metal with 2-4 grams in humans. It is an essential trace element, critical for cell growth, development and differentiation, DNA synthesis, RNA transcription, cell division, and cell activation. Zinc deficiency has adverse consequences during embryogenesis and early childhood development, particularly on immune functioning. It is essential in members of all enzyme classes, including over 300 signaling molecules and transcription factors. Free zinc in immune and tumor cells is regulated by 14 distinct zinc importers (ZIP) and transporters (ZNT1-8). Zinc depletion induces cell death via apoptosis (or necrosis if apoptotic pathways are blocked) while sufficient zinc levels allows maintenance of autophagy. Cancer cells have upregulated zinc importers, and frequently increased zinc levels, which allow them to survive. Based on this novel synthesis, approaches which locally regulate zinc levels to promote survival of immune cells and/or induce tumor apoptosis are in order.
- Published
- 2010
- Full Text
- View/download PDF
24. Meeting report: The 13th Annual Meeting of the Translational Research Cancer Centers Consortium (TrC3); Immune Suppression and the Tumor Microenvironment, Columbus, Ohio; March 1-2, 2010.
- Author
-
Lesinski GB, Carson WE, Repasky EA, Wei WZ, Kalinski P, Lotze MT, June CH, Petros W, Muthusamy N, and Olencki T
- Subjects
- Animals, Cooperative Behavior, Humans, Immunity, Cellular, Immunosuppression Therapy, Neoplasms immunology, Regional Medical Programs, Translational Research, Biomedical, Tumor Escape, United States, Cancer Vaccines, Immunotherapy trends, Neoplasms therapy
- Abstract
The Translational Research Cancer Centers Consortium (TrC3) is a cancer immunotherapy network, established to promote biologic therapeutics in the Midwestern and Northeastern regions of The United States. The 13th Annual Meeting of the TrC3 was hosted by The Ohio State University Comprehensive Cancer Center-Arthur G. James Cancer Hospital and Richard J. Solove Research Institute and took place at The Blackwell Hotel and Conference Center in Columbus, OH on March 1-2, 2010 (http://www.osuccc.osu.edu/TrC3/index.htm). This year's theme was "Immune Suppression and the Tumor Microenvironment." The meeting consisted of 21 oral presentations, a roundtable discussion focused on enhancing collaborative relationships within the consortium, and a poster session with 54 abstracts from predoctoral or postdoctoral researchers. This annual meeting brought together more than 170 investigators from 9 regional cancer centers including: Abramson Cancer Center at The University of Pennsylvania, Barbara Ann Karmanos Cancer Institute at Wayne State University, Case Comprehensive Cancer Center, Cleveland Clinic Taussig Cancer Center, James P. Wilmot Cancer Center, Mary Babb Randolph Cancer Center at West Virginia University, The Ohio State University Comprehensive Cancer Center, Penn State Cancer Institute, Roswell Park Cancer Institute, and University of Pittsburgh Cancer Institute. The proceedings of this year's meeting are summarized in this report.
- Published
- 2010
- Full Text
- View/download PDF
25. MicroRNAs in immune regulation--opportunities for cancer immunotherapy.
- Author
-
Okada H, Kohanbash G, and Lotze MT
- Subjects
- Animals, Humans, Immunity immunology, Neoplasms genetics, Ribonuclease III metabolism, T-Lymphocytes cytology, T-Lymphocytes enzymology, T-Lymphocytes immunology, Immunity genetics, Immunotherapy, MicroRNAs metabolism, Neoplasms immunology, Neoplasms therapy
- Abstract
Endogenously produced microRNAs are predicted to regulate the translation of over two-thirds all human gene transcripts. Certain microRNAs regulate expression of genes that are critically involved in both innate and adaptive immune responses. Immune cells represent a highly attractive target for microRNA gene therapy approaches, as these cells can be isolated, treated and then reintroduced into the patient. In this short review, we discuss how recent discoveries on the roles of microRNAs in immune-regulation will advance the field of cancer immunology and immunotherapy. Targets identified already in T cells include microRNAs, miR-17-92 family, miR-155, and miR-181a. In macrophages, miR-125b, miR-146, and miR-155 act as Pathogen Associated Molecular Pattern Molecule-associated microRNAs and miR-34C and miR-214 as Damage Associated Molecular Pattern Molecules-associated miRs. We have also demonstrated that the ability of tumors to serve as targets for cytolytic effectors is regulated by miR-222 and miR-339., (Copyright 2010 Elsevier Ltd. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
26. Cancer and inflammation: promise for biologic therapy.
- Author
-
Demaria S, Pikarsky E, Karin M, Coussens LM, Chen YC, El-Omar EM, Trinchieri G, Dubinett SM, Mao JT, Szabo E, Krieg A, Weiner GJ, Fox BA, Coukos G, Wang E, Abraham RT, Carbone M, and Lotze MT
- Subjects
- Adaptive Immunity, Adult, Animals, Child, Disease Models, Animal, Humans, Immunity, Innate, Inflammation, Neoplasms genetics, Neoplasms immunology, Biological Therapy trends, Neoplasms therapy
- Abstract
Cancers often arise as the end stage of inflammation in adults, but not in children. As such there is a complex interplay between host immune cells during neoplastic development, with both an ability to promote cancer and limit or eliminate it, most often complicit with the host. In humans, defining inflammation and the presence of inflammatory cells within or surrounding the tumor is a critical aspect of modern pathology. Groups defining staging for neoplasms are strongly encouraged to assess and incorporate measures of the presence of apoptosis, autophagy, and necrosis and also the nature and quality of the immune infiltrate. Both environmental and genetic factors enhance the risk of cigarette smoking, Helicobacter pylori, hepatitis B/C, human papilloma virus, solar irradiation, asbestos, pancreatitis, or other causes of chronic inflammation. Identifying suitable genetic polymorphisms in cytokines, cytokine receptors, and Toll-like receptors among other immune response genes is also seen as high value as genomic sequencing becomes less expensive. Animal models that incorporate and assess not only the genetic anlagen but also the inflammatory cells and the presence of microbial pathogens and damage-associated molecular pattern molecules are necessary. Identifying micro-RNAs involved in regulating the response to damage or injury are seen as highly promising. Although no therapeutic strategies to prevent or treat cancers based on insights into inflammatory pathways are currently approved for the common epithelial malignancies, there remains substantial interest in agents targeting COX2 or PPARgamma, ethyl pyruvate and steroids, and several novel agents on the horizon.
- Published
- 2010
- Full Text
- View/download PDF
27. miR-17-92 expression in differentiated T cells - implications for cancer immunotherapy.
- Author
-
Sasaki K, Kohanbash G, Hoji A, Ueda R, McDonald HA, Reinhart TA, Martinson J, Lotze MT, Marincola FM, Wang E, Fujita M, and Okada H
- Subjects
- Animals, Cell Death immunology, Cell Differentiation immunology, Humans, Interferon-gamma genetics, Interferon-gamma immunology, Interleukin-2 immunology, Interleukin-4 genetics, Interleukin-4 immunology, Jurkat Cells, Mice, Mice, Inbred C57BL, Mice, Transgenic, Microarray Analysis, Signal Transduction immunology, Immunotherapy methods, MicroRNAs genetics, MicroRNAs immunology, Neoplasms immunology, Neoplasms therapy, Th1 Cells cytology, Th1 Cells immunology, Th1 Cells physiology, Th2 Cells cytology, Th2 Cells immunology, Th2 Cells physiology
- Abstract
Background: Type-1 T cells are critical for effective anti-tumor immune responses. The recently discovered microRNAs (miRs) are a large family of small regulatory RNAs that control diverse aspects of cell function, including immune regulation. We identified miRs differentially regulated between type-1 and type-2 T cells, and determined how the expression of such miRs is regulated., Methods: We performed miR microarray analyses on in vitro differentiated murine T helper type-1 (Th1) and T helper type-2 (Th2) cells to identify differentially expressed miRs. We used quantitative RT-PCR to confirm the differential expression levels. We also used WST-1, ELISA, and flow cytometry to evaluate the survival, function and phenotype of cells, respectively. We employed mice transgenic for the identified miRs to determine the biological impact of miR-17-92 expression in T cells., Results: Our initial miR microarray analyses revealed that the miR-17-92 cluster is one of the most significantly over-expressed miR in murine Th1 cells when compared with Th2 cells. RT-PCR confirmed that the miR-17-92 cluster expression was consistently higher in Th1 cells than Th2 cells. Disruption of the IL-4 signaling through either IL-4 neutralizing antibody or knockout of signal transducer and activator of transcription (STAT)6 reversed the miR-17-92 cluster suppression in Th2 cells. Furthermore, T cells from tumor bearing mice and glioma patients had decreased levels of miR-17-92 when compared with cells from non-tumor bearing counterparts. CD4+ T cells derived from miR-17-92 transgenic mice demonstrated superior type-1 phenotype with increased IFN-gamma production and very late antigen (VLA)-4 expression when compared with counterparts derived from wild type mice. Human Jurkat T cells ectopically expressing increased levels of miR-17-92 cluster members demonstrated increased IL-2 production and resistance to activation-induced cell death (AICD)., Conclusion: The type-2-skewing tumor microenvironment induces the down-regulation of miR-17-92 expression in T cells, thereby diminishing the persistence of tumor-specific T cells and tumor control. Genetic engineering of T cells to express miR-17-92 may represent a promising approach for cancer immunotherapy.
- Published
- 2010
- Full Text
- View/download PDF
28. High-mobility group box 1 and cancer.
- Author
-
Tang D, Kang R, Zeh HJ 3rd, and Lotze MT
- Subjects
- Animals, Cell Nucleus metabolism, HMGB1 Protein chemistry, HMGB1 Protein genetics, Humans, NF-kappa B metabolism, Neoplasms blood supply, Neoplasms pathology, Neoplasms therapy, Receptors, Estrogen metabolism, Tumor Suppressor Proteins metabolism, HMGB1 Protein metabolism, Neoplasms metabolism
- Abstract
High-mobility group box 1 protein (HMGB1), a chromatin associated nuclear protein and extracellular damage associated molecular pattern molecule (DAMP), is an evolutionarily ancient and critical regulator of cell death and survival. Overexpression of HMGB1 is associated with each of the hallmarks of cancer including unlimited replicative potential, ability to develop blood vessels (angiogenesis), evasion of programmed cell death (apoptosis), self-sufficiency in growth signals, insensitivity to inhibitors of growth, inflammation, tissue invasion and metastasis. Our studies and those of our colleagues suggest that HMGB1 is central to cancer (abnormal wound healing) and many of the findings in normal wound healing as well. Here, we focus on the role of HMGB1 in cancer, the mechanisms by which it contributes to carcinogenesis, and therapeutic strategies based on targeting HMGB1., (Copyright 2009 Elsevier B.V. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
29. Autophagy inhibition in combination cancer treatment.
- Author
-
Livesey KM, Tang D, Zeh HJ, and Lotze MT
- Subjects
- Animals, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Drug Delivery Systems, Drug Resistance, Neoplasm, HMGB1 Protein metabolism, Humans, Antineoplastic Combined Chemotherapy Protocols pharmacology, Autophagy drug effects, Neoplasms drug therapy
- Abstract
The effective elimination of cancer cells is compromised by mechanisms of resistance. Such mechanisms have been variably ascribed to drug export transporters, more effective DNA repair mechanisms compared with healthy cells, singularly resistant stem cells, resistance to apoptosis, self-sufficiency for growth factor signaling and an angiogenic switch, as well as immunological pathways associated with T-regulatory cells, myeloid-derived suppressor cells or plasmacytoid dendritic cells. In this review, the critically important process of autophagy, which is a mechanism of cell survival in the presence of genomic injury, endoplasmic reticulum stress, oxidant stress, nutrient insufficiency and viral/bacterial infection, is explored in the setting of cancer treatment. Autophagy has recently been demonstrated as important for conferring resistance to chemotherapy, radiation therapy and immunotherapy. Compounds are now available that can reverse autophagy, including the antimalarial compounds chloroquine and hydroxychloroquine, as well as the antidepressant agent clomipramine. Other strategies for the reversal of autophagy are based on the recent observation that the cytosolic location of the chromatin-binding protein HMGB1 (high-mobility group box-1) is associated with sustained autophagy. Targeting HMGB1 using platinum-containing compounds, ethyl pyruvate or glycyrrhizin has also been used to limit autophagy. Screening for new agents is ongoing, which, coupled with conventional chemotherapeutic compounds, may usher in a new generation of autophagy-inhibiting agents.
- Published
- 2009
30. Emerging concepts in biomarker discovery; the US-Japan Workshop on Immunological Molecular Markers in Oncology.
- Author
-
Tahara H, Sato M, Thurin M, Wang E, Butterfield LH, Disis ML, Fox BA, Lee PP, Khleif SN, Wigginton JM, Ambs S, Akutsu Y, Chaussabel D, Doki Y, Eremin O, Fridman WH, Hirohashi Y, Imai K, Jacobson J, Jinushi M, Kanamoto A, Kashani-Sabet M, Kato K, Kawakami Y, Kirkwood JM, Kleen TO, Lehmann PV, Liotta L, Lotze MT, Maio M, Malyguine A, Masucci G, Matsubara H, Mayrand-Chung S, Nakamura K, Nishikawa H, Palucka AK, Petricoin EF, Pos Z, Ribas A, Rivoltini L, Sato N, Shiku H, Slingluff CL, Streicher H, Stroncek DF, Takeuchi H, Toyota M, Wada H, Wu X, Wulfkuhle J, Yaguchi T, Zeskind B, Zhao Y, Zocca MB, and Marincola FM
- Subjects
- Humans, Japan, National Cancer Institute (U.S.), Reproducibility of Results, United States, United States Food and Drug Administration, Biomarkers, Tumor immunology, Biomedical Research trends, Neoplasms drug therapy
- Abstract
Supported by the Office of International Affairs, National Cancer Institute (NCI), the "US-Japan Workshop on Immunological Biomarkers in Oncology" was held in March 2009. The workshop was related to a task force launched by the International Society for the Biological Therapy of Cancer (iSBTc) and the United States Food and Drug Administration (FDA) to identify strategies for biomarker discovery and validation in the field of biotherapy. The effort will culminate on October 28th 2009 in the "iSBTc-FDA-NCI Workshop on Prognostic and Predictive Immunologic Biomarkers in Cancer", which will be held in Washington DC in association with the Annual Meeting. The purposes of the US-Japan workshop were a) to discuss novel approaches to enhance the discovery of predictive and/or prognostic markers in cancer immunotherapy; b) to define the state of the science in biomarker discovery and validation. The participation of Japanese and US scientists provided the opportunity to identify shared or discordant themes across the distinct immune genetic background and the diverse prevalence of disease between the two Nations. Converging concepts were identified: enhanced knowledge of interferon-related pathways was found to be central to the understanding of immune-mediated tissue-specific destruction (TSD) of which tumor rejection is a representative facet. Although the expression of interferon-stimulated genes (ISGs) likely mediates the inflammatory process leading to tumor rejection, it is insufficient by itself and the associated mechanisms need to be identified. It is likely that adaptive immune responses play a broader role in tumor rejection than those strictly related to their antigen-specificity; likely, their primary role is to trigger an acute and tissue-specific inflammatory response at the tumor site that leads to rejection upon recruitment of additional innate and adaptive immune mechanisms. Other candidate systemic and/or tissue-specific biomarkers were recognized that might be added to the list of known entities applicable in immunotherapy trials. The need for a systematic approach to biomarker discovery that takes advantage of powerful high-throughput technologies was recognized; it was clear from the current state of the science that immunotherapy is still in a discovery phase and only a few of the current biomarkers warrant extensive validation. It was, finally, clear that, while current technologies have almost limitless potential, inadequate study design, limited standardization and cross-validation among laboratories and suboptimal comparability of data remain major road blocks. The institution of an interactive consortium for high throughput molecular monitoring of clinical trials with voluntary participation might provide cost-effective solutions.
- Published
- 2009
- Full Text
- View/download PDF
31. RAGE (Receptor for Advanced Glycation Endproducts), RAGE ligands, and their role in cancer and inflammation.
- Author
-
Sparvero LJ, Asafu-Adjei D, Kang R, Tang D, Amin N, Im J, Rutledge R, Lin B, Amoscato AA, Zeh HJ, and Lotze MT
- Subjects
- Animals, High Mobility Group Proteins metabolism, Humans, Ligands, Receptor for Advanced Glycation End Products, S100 Proteins metabolism, Inflammation metabolism, Neoplasms metabolism, Receptors, Immunologic metabolism
- Abstract
The Receptor for Advanced Glycation Endproducts [RAGE] is an evolutionarily recent member of the immunoglobulin super-family, encoded in the Class III region of the major histocompatability complex. RAGE is highly expressed only in the lung at readily measurable levels but increases quickly at sites of inflammation, largely on inflammatory and epithelial cells. It is found either as a membrane-bound or soluble protein that is markedly upregulated by stress in epithelial cells, thereby regulating their metabolism and enhancing their central barrier functionality. Activation and upregulation of RAGE by its ligands leads to enhanced survival. Perpetual signaling through RAGE-induced survival pathways in the setting of limited nutrients or oxygenation results in enhanced autophagy, diminished apoptosis, and (with ATP depletion) necrosis. This results in chronic inflammation and in many instances is the setting in which epithelial malignancies arise. RAGE and its isoforms sit in a pivotal role, regulating metabolism, inflammation, and epithelial survival in the setting of stress. Understanding the molecular structure and function of it and its ligands in the setting of inflammation is critically important in understanding the role of this receptor in tumor biology.
- Published
- 2009
- Full Text
- View/download PDF
32. Not just nuclear proteins: 'novel' autophagy cancer treatment targets - p53 and HMGB1.
- Author
-
Livesey KM, Tang D, Zeh HJ, and Lotze MT
- Subjects
- Animals, Apoptosis drug effects, Apoptosis physiology, Autophagy physiology, Drugs, Investigational pharmacology, HMGB1 Protein metabolism, Humans, Neoplasms metabolism, Neoplasms physiopathology, Nuclear Proteins metabolism, Tumor Suppressor Protein p53 metabolism, Autophagy drug effects, HMGB1 Protein physiology, Neoplasms drug therapy, Nuclear Proteins physiology, Tumor Suppressor Protein p53 drug effects, Tumor Suppressor Protein p53 physiology
- Published
- 2008
33. Detecting DNA: getting and begetting cancer.
- Author
-
Farkas AM, Kilgore TM, and Lotze MT
- Subjects
- DNA, Neoplasm blood, DNA, Neoplasm immunology, Humans, Neoplasms metabolism, Neoplasms pathology, DNA, Neoplasm genetics, DNA, Neoplasm metabolism, Neoplasms etiology, Neoplasms genetics
- Published
- 2007
34. A life in passing: Jonathan Gray.
- Author
-
Lotze MT and Gray J
- Subjects
- Animals, Humans, Biomedical Research economics, Neoplasms economics, Neoplasms immunology, Patients psychology, Research Support as Topic
- Published
- 2007
- Full Text
- View/download PDF
35. Inside, outside, upside down: damage-associated molecular-pattern molecules (DAMPs) and redox.
- Author
-
Rubartelli A and Lotze MT
- Subjects
- Cytokines immunology, Humans, Immunity, Innate, Inflammation metabolism, Inflammation Mediators immunology, Neoplasms immunology, Oxidation-Reduction, Cytokines metabolism, Inflammation immunology, Inflammation Mediators metabolism, Neoplasms metabolism, Proteins metabolism
- Abstract
Immune responses are initiated and perpetuated by molecules derived from microorganisms pathogen-associated molecular-pattern molecules or from the damage or death of host cells [damage-associated molecular-pattern (DAMP) molecules]. Many DAMPs are nuclear or cytosolic proteins with defined intracellular function that, when released outside the cell following tissue injury, move from a reducing to an oxidizing milieu resulting in their functional denaturation. Here, we discuss the consequences of DAMP oxidation on the outcome of acute inflammation. We also suggest that, outside the cell, DAMPs might adopt novel conformations or alter the redox of the extracellular environment to more closely mimic the internal one, thereby avoiding oxidation-mediated inactivation and promoting pathology. We propose that chronic inflammation associated with autoimmunity, chronic viral infection and cancer is probably mediated by persistent release and function of DAMPs, promoting and promoted by a disordered redox environment.
- Published
- 2007
- Full Text
- View/download PDF
36. Report on the ISBTC mini-symposium on biologic effects of targeted therapeutics.
- Author
-
Atkins MB, Carbone D, Coukos G, Dhodapkar M, Ernstoff MS, Finke J, Gajewski TF, Gollob J, Lotze MT, Storkus W, and Weiner LM
- Subjects
- Antibodies, Monoclonal immunology, Antibodies, Monoclonal metabolism, Antibodies, Monoclonal therapeutic use, Antineoplastic Agents immunology, Antineoplastic Agents metabolism, Apoptosis immunology, Combined Modality Therapy, Humans, Immunologic Factors immunology, Immunologic Factors metabolism, Immunologic Factors therapeutic use, Neoplasms genetics, Neoplasms immunology, Neoplasms metabolism, Neovascularization, Pathologic, Angiogenesis Inhibitors therapeutic use, Antineoplastic Agents therapeutic use, Immunotherapy, Neoplasms therapy
- Abstract
The International Society for Biologic Therapy of Cancer held a mini-symposium on October 26, 2006 in Los Angeles to review current information regarding the biologic effects of both standard and targeted therapies. The purpose of the mini-symposium was to describe the existing knowledge regarding various biologic effects of current therapies, identify the most relevant issues and gaps in the knowledge base and discuss the optimal means of obtaining necessary missing information. Topics discussed included: (1) The impact of antitumor monoclonal antibody therapy on antigen presentation and adaptive immunity; (2) the effects of antiangiogenic/targeted therapy of the immune system; (3) the impact of chemotherapy on angiogenesis and immune function; (4) combination of antiangiogenic and immunotherapy at the clinical level; (5) the effects of tyrosine kinase inhibitors on TH1/TH2 response and T-regulatory cells; (6) the impact of farnesyltransferase inhibitors and other targeted agents on T-cell activation; (7) the impact of epigenetic modulators on biologic properties, and (8) the impact of the nature of cell death on the immune system. The ultimate goals of this mini-symposium were to use the above information to inform and influence basic science efforts and discussions, rationally design combination treatment regimens and optimally employ correlative studies in the context of ongoing and future clinical investigations.
- Published
- 2007
- Full Text
- View/download PDF
37. High mobility group box I (HMGB1) release from tumor cells after treatment: implications for development of targeted chemoimmunotherapy.
- Author
-
Dong Xda E, Ito N, Lotze MT, Demarco RA, Popovic P, Shand SH, Watkins S, Winikoff S, Brown CK, Bartlett DL, and Zeh HJ 3rd
- Subjects
- Antineoplastic Agents therapeutic use, Apoptosis, Cell Line, Tumor, Cell Nucleus metabolism, Colonic Neoplasms, Combined Modality Therapy, Humans, Immunotherapy, Killer Cells, Lymphokine-Activated immunology, Killer Cells, Lymphokine-Activated metabolism, Melanoma, Melphalan pharmacology, Microscopy, Confocal, Necrosis, Neoplasms drug therapy, Neoplasms pathology, Oxaliplatin, Paclitaxel pharmacology, Pancreatic Neoplasms, Antineoplastic Agents pharmacology, HMGB1 Protein metabolism, Neoplasms metabolism, Neoplasms therapy, Organoplatinum Compounds pharmacology
- Abstract
We have recently demonstrated that cytolysis of human melanoma cells by immune effectors (both NK and T cells) is associated with release of the nuclear chromatin protein, high mobility group box I (HMGB1). Extracellular HMGB1 mediates a number of important functions including endothelial cell activation, stromagenesis, recruitment and activation of innate immune cells, and also dendritic cell maturation that, in the setting of cancer, lead to a chronic inflammatory response. This reparative inflammatory response promotes tumor cell survival, expansion, and metastases. Release of HMGB1 after chemotherapy-induced cytotoxicity has not been well characterized. We measured the release of HMGB1 after chemotherapy or immune cytolysis and demonstrated that this did not correlate with conventional markers of apoptosis and necrosis in several human colorectal, pancreatic, and melanoma tumor cell lines. Rather, we observed that tumor cells incubated with the platinating agent oxaliplatin, retained HMGB1 within the nucleus for significantly longer periods than other agents used at comparable cytotoxic concentrations or even with potent cytolytic cells. Thus, release of HMGB1 from dying tumor cells treated with chemotherapy or cells with lymphokine activated killer cell activity is not dependent solely on the mode of cell death. Sequestration of the damage associated molecular pattern molecule, HMGB1, may play a role in the clinical efficacy of platinating agents and suggests this as a superior agent for coupling with immunotherapeutic strategies, possibly enhancing their effectiveness.
- Published
- 2007
- Full Text
- View/download PDF
38. Masquerader: high mobility group box-1 and cancer.
- Author
-
Ellerman JE, Brown CK, de Vera M, Zeh HJ, Billiar T, Rubartelli A, and Lotze MT
- Subjects
- Cell Movement, Cell Nucleus metabolism, HMGB1 Protein antagonists & inhibitors, HMGB1 Protein chemistry, Humans, Neoplasms drug therapy, Receptors, Cell Surface metabolism, Signal Transduction, HMGB1 Protein metabolism, Neoplasms metabolism, Neoplasms pathology
- Abstract
Since its identification a third of a century ago, the high-mobility group box-1 (HMGB1) protein has been linked to varied diverse cellular processes, including release from necrotic cells and secretion by activated macrophages engulfing apoptotic cells. Initially described as solely chromatin-associated, HMGB1 was additionally discovered in the cytoplasm of several types of cultured mammalian cells 6 years later. In addition to its intracellular role, HMGB1 has been identified extracellularly as a putative leaderless cytokine and differentiation factor. In the years since its discovery, HMGB1 has also been implicated in disease states, including Alzheimer's, sepsis, ischemia-reperfusion, arthritis, and cancer. In cancer, overexpression of HMGB1, particularly in conjunction with its receptor for advanced glycation end products, has been associated with the proliferation and metastasis of many tumor types, including breast, colon, melanoma, and others. This review focuses on current knowledge and speculation on the role of HMGB1 in the development of cancer, metastasis, and potential targets for therapy.
- Published
- 2007
- Full Text
- View/download PDF
39. Eosinophilic granulocytes and damage-associated molecular pattern molecules (DAMPs): role in the inflammatory response within tumors.
- Author
-
Lotfi R, Lee JJ, and Lotze MT
- Subjects
- Gastrointestinal Neoplasms immunology, Gastrointestinal Neoplasms therapy, HMGB1 Protein immunology, Humans, Eosinophils immunology, Immunotherapy methods, Neoplasms immunology, Neoplasms therapy
- Abstract
The development of a tumor over many years typically leads to reciprocal alternations in the host and the tumor, enabling tumor growth paradoxically in the setting of substantial necrosis and inflammation. When evaluating a tumor, it is important to assess 3 elements: (1) the quantity and quality of tumor-associated leukocytes, (2) their state of activation, and (3) tumor microenvironment. Peripheral blood eosinophilia and tumor-associated tissue eosinophilia are frequently associated with some tumor types and also found after immunotherapy with IL-2, IL-4, granulocyte-macrophage colony-stimulating factor, and antibody to CTLA-4. Within several tumor types including gastrointestinal tumors, tumor-associated tissue eosinophilia is associated with a significantly better prognosis. The converse is true in other tumor types such as differentiated oral squamous cell carcinoma. On the basis of the emergent data, tumor-associated eosinophils have at least 2 dominant nonoverlapping activities: (1) destructive effector functions potentially limiting tumor growth as well as causing recruitment and activation of other leukocytes, (2) immunoregulative and remodeling activities which suppress immune response and promote tumor proliferation. The mechanism by which eosinophils in particular are recruited into tumor tissue is largely unknown. Candidates for causing eosinophil chemotaxis into tumor tissue are the released damage-associated molecular pattern molecules (DAMPs) including the nuclear protein high mobility group box 1. High mobility group box 1 is released upon necrotic cell death and secreted by many cells, particularly during periods of nutrient, hypoxic, or oxidant stress. This overview on eosinophil biology in the context of cancer and necrosis, introduces intriguing and novel strategies targeting eosinophils to enable more effective biologic therapy for cancer patients.
- Published
- 2007
- Full Text
- View/download PDF
40. Cancer genomics: the unknown unknowns.
- Author
-
Lotze MT and Yu Y
- Subjects
- Animals, Biomarkers, Tumor genetics, Biomarkers, Tumor metabolism, Cell Transformation, Neoplastic genetics, Cell Transformation, Neoplastic metabolism, Genome, Human, Humans, MicroRNAs biosynthesis, Neoplasms metabolism, Gene Expression Regulation, Neoplastic, MicroRNAs metabolism, Neoplasms genetics
- Published
- 2006
41. Pediatric cancers are infiltrated predominantly by macrophages and contain a paucity of dendritic cells: a major nosologic difference with adult tumors.
- Author
-
Vakkila J, Jaffe R, Michelow M, and Lotze MT
- Subjects
- Adolescent, Adult, Age Factors, Aged, Child, Child, Preschool, Humans, Immunohistochemistry, Infant, Middle Aged, Neoplasms diagnosis, Neoplasms immunology, Pilot Projects, Retrospective Studies, Dendritic Cells pathology, Lymphocytes, Tumor-Infiltrating pathology, Macrophages pathology, Neoplasms pathology
- Abstract
Purpose: Adult cancer is frequently preceded by a period of prolonged chronic inflammation caused by infectious microbial agents or physical or chemical irritants. By contrast, an association between the classic pediatric neoplasias and inflammatory triggers is only rarely recognized. We hypothesized that the difference could be reflected in the inflammatory cell infiltrates of pediatric and adult cancer., Experimental Design: Three investigators retrospectively studied 27 pediatric and 13 adult cancers at first diagnosis by immunohistochemistry. Inflammatory cells were identified and counted, and their location in relation to tumor tissue was analyzed., Results: A majority of tumor-associated leukocytes (TAL) in adult tumors were located at the edges of tumor islands forming inflammatory foci between the supporting stroma and the malignant infiltrate. In contrast, TALs in pediatric tumors were scattered within the malignant tumor islands. In adult tumors, TALs were composed of diverse leukocyte types; but in pediatric tumors, the infiltrating cells were predominantly macrophages that accumulated in areas of necrosis within the tumors. The most striking feature in the pediatric tumors was the virtual absence of dendritic cells. The proportion of intratumoral dendritic cells in pediatric samples was 4.1%; whereas in adult tumors, they formed 36.9% of TALs within the tumor islands and 25.1% around the tumors., Conclusions: We conclude that TALs in pediatric cancers are composed mainly of macrophages and largely devoid of dendritic cell. The findings may provide a major nosologic difference reclassifying pediatric and adult tumors based on nominal inflammatory and noninflammatory etiologies.
- Published
- 2006
- Full Text
- View/download PDF
42. Regulatory balance between the immune response of tumor antigen-specific T-cell receptor gene-transduced CD8 T cells and the suppressive effects of tolerogenic dendritic cells.
- Author
-
Fujii S, Nishimura MI, and Lotze MT
- Subjects
- Granulocyte-Macrophage Colony-Stimulating Factor pharmacology, Humans, Immunotherapy, Interleukin-10 pharmacology, Interleukin-4 pharmacology, Lymphocyte Activation, Lymphocyte Culture Test, Mixed, Neoplasms therapy, Recombinant Proteins pharmacology, T-Lymphocytes, Cytotoxic immunology, CD8-Positive T-Lymphocytes immunology, Dendritic Cells immunology, Neoplasms immunology, Receptors, Antigen, T-Cell immunology
- Abstract
Tumor immune responses, including some immunotherapy strategies, can fail because of a number of reasons, such as poor tumor cell immunogenicity or local suppressive cytokine release by dendritic cells (DC) at tumor sites. The retroviral transfer of T-cell receptor (TCR) genes encoding tumor-specific receptors into T cells is a fascinating approach to bypass antigen-presenting cells and allow T cells to directly recognize antigen. It also allows the generation and expansion of potent antitumor cytotoxic T lymphocytes with defined cancer antigen specificities more readily than naive T cells. However, interleukin-10 (IL-10)-exposed dendritic cells (IL-10-DC) have been labeled tolerogenic because of the suppressive effects they have on T cell responses. Whether TCR gene-transduced effector CD8(+) T cells can break through suppressive functions mediated by IL-10-DC is not known. In the current study, we demonstrate the role of IL-10 in modifying the function of DC that otherwise would activate potent TCR gene-transduced T cells against tumor antigens. TCR gene-transduced T cells maintained their cytolytic activity in the presence of DC exposed to low doses of IL-10 during maturation; however, they lost this activity in an antigen-specific manner when exposed to DC matured with high doses of IL-10., ((Cancer Sci 2005; 96: 897-902).)
- Published
- 2005
- Full Text
- View/download PDF
43. Natural killer-dendritic cell cross-talk in cancer immunotherapy.
- Author
-
Kalinski P, Mailliard RB, Giermasz A, Zeh HJ, Basse P, Bartlett DL, Kirkwood JM, Lotze MT, and Herberman RB
- Subjects
- Animals, Dendritic Cells cytology, Dendritic Cells metabolism, Humans, Killer Cells, Natural cytology, Killer Cells, Natural metabolism, Receptor Cross-Talk physiology, Receptors, Immunologic physiology, Cell Communication immunology, Dendritic Cells immunology, Immunotherapy, Adoptive methods, Killer Cells, Natural immunology, Neoplasms immunology, Neoplasms therapy
- Abstract
Natural killer (NK) cells and dendritic cells (DCs), two important components of the immune system, can exchange bidirectional activating signals in a positive feedback. Myeloid DCs, the cell type specialised in the presentation of antigen and initiation of antigen-specific immune responses, have recently been documented to be involved in supporting innate immunity, promoting the production of cytokines and cytotoxicity of NK cells, and enhancing their tumouricidal activity. Natural interferon-producing cells/plasmacytoid DCs (IPCs/PDCs) play an additional role in NK cell activation. Reciprocally, NK cells, traditionally considered to be major innate effector cells, have also recently been shown to play immunoregulatory 'helper' functions, being able to activate DCs and to enhance their ability to produce pro-inflammatory cytokines, and to stimulate T helper (Th) 1 and cytotoxic T lymphocyte (CTL) responses of tumour-specific CD4+ and CD8+ T cells. Activated NK cells induce the maturation of myeloid DCs into stable type-1 polarised DCs (DC1), characterised by up to a 100-fold enhanced ability to produce IL-12p70 in response to subsequent interaction with Th cells. In addition, the ability of NK cells to kill tumour cells may facilitate the generation of tumour-related antigenic material, further accelerating the induction of tumour-specific immunity. DC1, induced by NK cells or by NK cell-related soluble factors, are stable, resistant to tumour-related suppressive factors, and demonstrate a strongly enhanced ability to induce Th1 and CTL responses in human in vitro and mouse in vivo models. Compared with the standard mature DCs that are used in clinical trials at present, human NK cell-induced DC1s act as superior inducers of anticancer CTL responses during in vitro sensitisation. This provides a strong rationale for the combined use of NK cells and DCs in the immunotherapy of patients with cancer and patients with chronic infections that are resistant to standard forms of treatment. Stage I/II clinical trials that are being implemented at present should allow evaluation of the immunological and clinical efficacy of combined NK-DC therapy of melanoma and other cancers.
- Published
- 2005
- Full Text
- View/download PDF
44. Workshop on cancer biometrics: identifying biomarkers and surrogates of cancer in patients: a meeting held at the Masur Auditorium, National Institutes of Health.
- Author
-
Lotze MT, Wang E, Marincola FM, Hanna N, Bugelski PJ, Burns CA, Coukos G, Damle N, Godfrey TE, Howell WM, Panelli MC, Perricone MA, Petricoin EF, Sauter G, Scheibenbogen C, Shivers SC, Taylor DL, Weinstein JN, and Whiteside TL
- Subjects
- Alternative Splicing, Biomarkers, Biomarkers, Tumor, Clinical Trials as Topic, DNA metabolism, DNA Methylation, Flow Cytometry, Gene Expression Profiling methods, Humans, Immunohistochemistry, Internet, Lymph Nodes pathology, Monitoring, Immunologic, Necrosis, Neoplasms therapy, Oligonucleotide Array Sequence Analysis, Polymorphism, Genetic, Polymorphism, Single Nucleotide, Protein Binding, Proteomics, RNA, Messenger metabolism, Reverse Transcriptase Polymerase Chain Reaction, Tissue Distribution, Computational Biology methods, Neoplasms blood, Neoplasms diagnosis
- Abstract
The current excitement about molecular targeted therapies has driven much of the recent dialog in cancer diagnosis and treatment. Particularly in the biologic therapy of cancer, identifiable antigenic T-cell targets restricted by MHC molecules and the related novel stress molecules such as MICA/B and Letal allow a degree of precision previously unknown in cancer therapy. We have previously held workshops on immunologic monitoring and angiogenesis monitoring. This workshop was designed to discuss the state of the art in identification of biomarkers and surrogates of tumor in patients with cancer, with particular emphasis on assays within the blood and tumor. We distinguish this from immunologic monitoring in the sense that it is primarily a measure of the tumor burden as opposed to the immune response to it. Recommendations for intensive investigation and targeted funding to enable such strategies were developed in seven areas: genomic analysis; detection of molecular markers in peripheral blood and lymph node by tumor capture and RT-PCR; serum, plasma, and tumor proteomics; immune polymorphisms; high content screening using flow and imaging cytometry; immunohistochemistry and tissue microarrays; and assessment of immune infiltrate and necrosis in tumors. Concrete recommendations for current application and enabling further development in cancer biometrics are summarized. This will allow a more informed, rapid, and accurate assessment of novel cancer therapies.
- Published
- 2005
- Full Text
- View/download PDF
45. Inflammation and necrosis promote tumour growth.
- Author
-
Vakkila J and Lotze MT
- Subjects
- Animals, DNA-Binding Proteins metabolism, Genes, Tumor Suppressor, Humans, Leukocytes immunology, Mutation, STAT3 Transcription Factor, Signal Transduction immunology, Signal Transduction physiology, Trans-Activators metabolism, Inflammation immunology, Necrosis, Neoplasms immunology
- Published
- 2004
- Full Text
- View/download PDF
46. Identifying biomarkers and surrogates of tumors (cancer biometrics): correlation with immunotherapies and immune cells.
- Author
-
Lotze MT and Rees RC
- Subjects
- Biomarkers, Tumor, Clinical Trials as Topic, Genomic Instability, Humans, Neoplasms genetics, Neoplasms immunology, Neoplastic Cells, Circulating, Proteomics, Reverse Transcriptase Polymerase Chain Reaction, Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization, Immunotherapy, Neoplasms therapy
- Abstract
The presence of inflammatory cells within cancer has been described for quite some time by pathologists, with generally improved outcome associated with their presence in various epithelial neoplasms. Most remarkably, this has included dendritic cells and T cells but more recently NK cells as well. Coupled with the rapid evolution of molecular technology, microarray analyses of primary tumors, serum and tumor proteomics, tumor capture analyses in the peripheral blood (together with quantitative RT-PCR), and novel histochemical markers and tissue microarrays, this provides the opportunity to establish a more effective means to study and classify into subsets various forms of cancer. Much of the current controversy in cancer diagnosis and pathologic assessment of prognosis lies in the application of these techniques in concert with other molecular tools including DNA microarrays, expression of histochemically defined cytokines, proangiogenic factors, and oncogene products, and correlating this with clinical relevance. Molecular detection technologies such as reverse transcriptase polymerase chain reaction, proteomics, and microarray analyses will be validated based on their integration with conventional cancer pathology and cancer diagnostics. Further work is needed to establish which cancer biomarkers and surrogates should be routinely measured and in which settings, and determining the appropriate sample size for such assays that can be validated in retrospective and prospective clinical studies. The ability to integrate these rapidly evolving strategies will consume much of our coordinate effort in cancer and cancer therapeutics for the near future.
- Published
- 2004
- Full Text
- View/download PDF
47. A primer on cancer immunology and immunotherapy.
- Author
-
Lotze MT and Papamichail M
- Subjects
- Antigens, Neoplasm analysis, Antigens, Neoplasm immunology, Dendritic Cells physiology, Humans, Killer Cells, Natural immunology, Neoplasms therapy, T-Lymphocytes, Cytotoxic immunology, Immunotherapy, Neoplasms immunology
- Abstract
The role of immunity in cancer has been abundantly demonstrated in murine tumor models as well as in man. Induction of clinically effective antitumor immune responses, based on this information, in patients with cancer however, remains elusive. This is not because tumors lack recognizable antigens [in fact there is evidence that there are thousands of potential novel targets in each tumor cell] but rather due to the fact that the induction of responses is not adequate nor particularly well understood. Tumors seem to be rather effective at limiting immune responses. Many of the molecularly defined antigens that have been detected on tumor cells are derived from self-proteins and as such are subject to tolerizing mechanisms. Such tumors have also developed escape mechanisms capable of evading or suppressing immune responses. Understanding the role of dendritic cells during the effector phase of the immune response and the complex interactions of stromal, immune, and tumor cells in the tumor microenvironment represent the next challenges to be understood for tumor immunology.
- Published
- 2004
- Full Text
- View/download PDF
48. Dealing with death: HMGB1 as a novel target for cancer therapy.
- Author
-
Lotze MT and DeMarco RA
- Subjects
- Animals, Cell Death drug effects, Glycation End Products, Advanced, HMGB1 Protein metabolism, Humans, Neoplasms enzymology, Neoplasms metabolism, Receptor for Advanced Glycation End Products, Antineoplastic Agents pharmacology, Drug Delivery Systems methods, HMGB1 Protein antagonists & inhibitors, HMGB1 Protein physiology, Neoplasms drug therapy, Neoplasms pathology, Receptors, Immunologic metabolism
- Abstract
High mobility group B1 (HMGB1) and its counter-receptor, receptor for advanced glycation end products (RAGE), represent suitable targets for investigation, integrating many aspects of modern biology, particularly that associated with chronic diseases involving inflammation, dysregulated cell death and cancer. Also known as amphoterin, HMGB1 was initially identified over 25 years ago as a transcriptional regulatory molecule causing DNA bending, and facilitating binding of several transcriptional complexes, in particular members of the nuclear hormone receptor family. Although loosely bound to chromatin, it is released from necrotic cells but not apoptotic cells and is actively secreted by activated macrophages in a partially tumor necrosis factor-dependent manner. It serves as a late mediator of septic death present within the serum and inflammatory sites of patients with arthritis, correlating with the inflammatory response, to signal tissue injury, causes sickness behavior, and acts as an endogenous pyrogen. Although known to interact with RAGE on endothelial cells causing activation and leukocyte recruitment, RAGE itself has most recently been shown to serve as a counter-receptor for leukocyte integrins, suggesting that signaling through this molecule is potentially important for cell adhesion and clustering as well as recruitment of inflammatory cells. Targeting the HMGB1 ligand or its receptor represents an important potential application in cancer therapeutics, given its widespread overexpression, as well as that of its receptor in virtually every tumor type carefully examined. This, coupled with its ability to accelerate tumor growth in immunodeficient murine models, suggests that it is a possible therapeutic target in patients with cancer.
- Published
- 2003
49. Advantages and limitations of particle-mediated transfection (gene gun) in cancer immuno-gene therapy using IL-10, IL-12 or B7-1 in murine tumor models.
- Author
-
Kitagawa T, Iwazawa T, Robbins PD, Lotze MT, and Tahara H
- Subjects
- Analysis of Variance, Animals, B7-1 Antigen genetics, B7-1 Antigen metabolism, Cell Line, Tumor, Cytomegalovirus, Flow Cytometry, Genetic Vectors genetics, Genetic Vectors metabolism, Interleukin-10 genetics, Interleukin-10 metabolism, Interleukin-12 genetics, Interleukin-12 metabolism, Mice, Neoplasms genetics, Transfection methods, Biolistics methods, Cytokines metabolism, Gene Expression Regulation, Neoplastic, Genetic Therapy methods, Neoplasms therapy
- Abstract
Background: Previous studies have shown that particle-mediated transfection (PMT; gene gun) is an efficient method of non-viral gene transfer. We have examined the advantages and limitations of PMT in cancer immuno-gene therapy using IL-10, IL-12 or B7-1, all of which have been shown to be effective in murine tumor models using retroviral vectors., Methods: Murine cell lines (MCA205, MCA207, NIH-3T3) were treated with in vitro PMT, liposome-mediated transfection (LMT) or retroviral transfection. In vivo PMT was also examined for in vivo experiments using C57BL/6., Results: Transfection efficiency and cytokine expression of PMT were similar to those of LMT, another non-viral approach, in culture when murine adherent cells were used. Tumor establishment of MCA205 was suppressed when they were transfected with the mIL-12 and/or mB7-1 gene using PMT in vitro. In a treatment of established tumor, vaccination with tumor cells transfected with an IL-12 gene suppressed tumor growth, whereas a B7-1 gene was not effective. When an IL-10 gene was used to supply a high level of expression for antitumor effects, neither in vitro nor in vivo PMT could suppress even tumor establishment. These failures appear to be caused by the characteristics of PMT which allows high, but transient, expression of transfected genes., Conclusions: Particle-mediated transfection is a useful non-viral transfection method in a system which does not require high-level gene expression for a prolonged time of period., (Copyright 2003 John Wiley & Sons, Ltd.)
- Published
- 2003
- Full Text
- View/download PDF
50. The critical need for cancer biometrics: quantitative, reproducible measures of cancer to define response to therapy.
- Author
-
Lotze MT
- Subjects
- Biometry methods, Drug Delivery Systems trends, Humans, Neoplasms genetics, Proteomics methods, Reproducibility of Results, Technology, Pharmaceutical methods, Technology, Pharmaceutical trends, Drug Delivery Systems methods, Neoplasms drug therapy
- Published
- 2003
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.