101
|
Sass SN, Ramsey KD, Egan SM, Wang J, Cortes Gomez E, Gollnick SO. Tumor-associated myeloid cells promote tumorigenesis of non-tumorigenic human and murine prostatic epithelial cell lines. Cancer Immunol Immunother 2018; 67:873-883. [PMID: 29502208 PMCID: PMC5951898 DOI: 10.1007/s00262-018-2143-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 02/26/2018] [Indexed: 11/29/2022]
Abstract
The etiology of prostate cancer is poorly understood, but it is a multi-step process that has been linked to environmental factors that induce inflammation within the gland. Glands of prostate cancer patients frequently contain multiple zones of disease at various stages of progression. The factors that drive disease progression from an indolent benign stage to aggressive disease are not well-defined. Prostate inflammation and carcinoma are associated with high levels of myeloid cell infiltration; these cells are linked to disease progression in other cancers, but their role in prostate cancer is unclear. To determine whether myeloid cells contribute to prostate cancer progression, the ability of prostate tumor-associated CD11b+ cells (TAMC) to drive prostate epithelial cell tumorigenesis was tested. Co-culture of CD11b+ TAMC with non-tumorigenic genetically primed prostate epithelial cells resulted in stable transformation and induction of tumorigenesis. RNA sequencing identified the IL-1α pathway as a potential molecular mechanism responsible for tumor promotion by TAMC. Inhibition of IL-1α delayed growth of TAMC-induced tumors. Further analysis showed that IL-1α inhibition led to decreased angiogenesis within tumors, suggesting that IL-1α promotes prostate tumor progression, potentially through augmentation of angiogenesis.
Collapse
Affiliation(s)
- Stephanie N Sass
- Department of Immunology, Roswell Park Cancer Institute, Elm and Carlton Sts, Buffalo, NY, 14263, USA
| | - Kimberley D Ramsey
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Elm and Carlton Sts, Buffalo, NY, 14263, USA
| | - Shawn M Egan
- Department of Immunology, Roswell Park Cancer Institute, Elm and Carlton Sts, Buffalo, NY, 14263, USA
| | - Jianmin Wang
- Department of Biostatistics and Bioinformatics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Eduardo Cortes Gomez
- Department of Biostatistics and Bioinformatics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Sandra O Gollnick
- Department of Immunology, Roswell Park Cancer Institute, Elm and Carlton Sts, Buffalo, NY, 14263, USA.
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Elm and Carlton Sts, Buffalo, NY, 14263, USA.
| |
Collapse
|
102
|
A module of inflammatory cytokines defines resistance of colorectal cancer to EGFR inhibitors. Oncotarget 2018; 7:72167-72183. [PMID: 27708224 PMCID: PMC5342152 DOI: 10.18632/oncotarget.12354] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 09/21/2016] [Indexed: 12/13/2022] Open
Abstract
Epidermal Growth Factor Receptor (EGFR) activates a robust signalling network to which colon cancer tumours often become addicted. Cetuximab, one of the monoclonal antibodies targeting this pathway, is employed to treat patients with colorectal cancer. However, many patients are intrinsically refractory to this treatment, and those who respond develop secondary resistance along time. Mechanisms of cancer cell resistance include either acquisition of new mutations or non genomic activation of alternative signalling routes. In this study, we employed a colon cancer model to assess potential mechanisms driving resistance to cetuximab. Resistant cells displayed increased ability to grow in suspension as colonspheres and this phenotype was associated with poorly organized structures. Factors secreted from resistant cells were causally involved in sustaining resistance, indeed administration to parental cells of conditioned medium collected from resistant cells was sufficient to reduce cetuximab efficacy. Among secreted factors, we report herein that a signature of inflammatory cytokines, including IL1A, IL1B and IL8, which are produced following EGFR pathway activation, was associated with the acquisition of an unresponsive phenotype to cetuximab in vitro. This signature correlated with lack of response to EGFR targeting also in patient-derived tumour xenografts. Collectively, these results highlight the contribution of inflammatory cytokines to reduced sensitivity to EGFR blockade and suggest that inhibition of this panel of cytokines in combination with cetuximab might yield an effective treatment strategy for CRC patients refractory to anti-EGFR targeting.
Collapse
|
103
|
Symptoms as Patient-Reported Outcomes in Cancer Patients Undergoing Immunotherapies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 995:165-182. [PMID: 30539512 DOI: 10.1007/978-3-030-02505-2_9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cancer therapies are toxic. Newer oncological treatments such as immunotherapy produce unconventional adverse events that are collectively referred to as immune-related adverse events (irAEs). These irAEs are clinician-rated and typically reported via tabulation of adverse events from the National Cancer Institute's Common Terminology Criteria for Adverse Events (CTCAE). However, the symptomatic effects of treatment and the severity of disease are best reported by the patient themselves. Although many pivotal trials for immunotherapeutic agents include health-related quality-of-life measures, symptom-focused assessments are more proximal to the effects of treatment and disease burden. This chapter discusses how best to measure symptoms, describes the desirable properties of a psychometrically valid symptom assessment tool, reviews available symptom assessment tools, provides methods to assist in the interpretation of PRO data, elucidates the feasibility and benefit of incorporating PRO in several cancer cohorts, describes the current use of PROs in immunotherapy, and identifies areas where further research are needed to enhance the use of PROs in cancer patients undergoing immunotherapy.
Collapse
|
104
|
Dinarello CA. Introduction to the interleukin-1 family of cytokines and receptors: Drivers of innate inflammation and acquired immunity. Immunol Rev 2018; 281:5-7. [PMID: 29248001 PMCID: PMC5750395 DOI: 10.1111/imr.12624] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Charles A Dinarello
- Department of Medicine, University of Colorado, Aurora, CO, USA
- Department of Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
105
|
Ciombor KK, Bekaii-Saab T. A Comprehensive Review of Sequencing and Combination Strategies of Targeted Agents in Metastatic Colorectal Cancer. Oncologist 2018; 23:25-34. [PMID: 29021377 PMCID: PMC5759820 DOI: 10.1634/theoncologist.2017-0203] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 09/11/2017] [Indexed: 02/07/2023] Open
Abstract
The emergence of targeted therapies for the treatment of metastatic colorectal cancer (mCRC) has considerably improved survival, but has also resulted in a dilemma of identifying the optimal sequence and combination of various agents in the mCRC treatment landscape. A number of cytotoxic agents, including irinotecan, oxaliplatin, 5-fluorouracil, capecitabine, and TAS-102, are available for treatment of mCRC. Additionally, whereas patients harboring rat sarcoma viral oncogene homolog (RAS)-wild type mCRC can be treated with the anti-epidermal growth factor receptor antibodies cetuximab and panitumumab or antiangiogenic agents (bevacizumab, ziv-aflibercept, and ramucirumab), patients with RAS-mutant mCRC are limited to antiangiogenic agents as biologic options. Regorafenib, a multikinase inhibitor, can be used in both RAS subgroups. As such, the recommended sequence of therapies that should be received by each subgroup must also be considered separately. This review provides an overview of recent clinical data for approved and investigational targeted therapies that have been studied across different mCRC treatment lines and patient subgroups. It also examines emerging trends in the treatment landscape for mCRC, including treatment with immune checkpoint inhibitors and the utilization of genomic profiling. IMPLICATIONS FOR PRACTICE Currently, there are no established guidelines for optimal sequencing of cytotoxic or targeted agents in metastatic colorectal cancer (mCRC). This review provides a snapshot of the current mCRC treatment paradigm and examines the latest clinical data that support the utilization of several targeted agents alone or in combination with backbone chemotherapy across different lines of treatment and patient populations, highlighting recommendations for their usage. Recent advances in the treatment landscape are also summarized, including genomic profiling and preliminary results with immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Kristen K Ciombor
- Division of Hematology and Oncology, Department of Internal Medicine, Vanderbilt University Medical Center/Vanderbilt-Ingram Cancer Center, Nashville, Tennessee, USA
| | - Tanios Bekaii-Saab
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Mayo Clinic Cancer Center, Phoenix, Arizona, USA
| |
Collapse
|
106
|
Abstract
The interleukin (IL)-1 family of cytokines is currently comprised of 11 members that have pleiotropic functions in inflammation and cancer. IL-1α and IL-1β were the first members of the IL-1 family to be described, and both signal via the same receptor, IL-1R. Over the last decade, much progress has been made in our understanding of biogenesis of IL-1β and its functions in human diseases. Studies from our laboratory and others have highlighted the critical role of nod-like receptors (NLRs) and multi-protein complexes known as inflammasomes in the regulation of IL-1β maturation. Recent studies have increased our appreciation of the role played by IL-1α in inflammatory diseases and cancer. However, the mechanisms that regulate the production of IL-1α and its bioavailability are relatively understudied. In this review, we summarize the distinctive roles played by IL-1α in inflammatory diseases and cancer. We also discuss our current knowledge about the mechanisms that control IL-1α biogenesis and activity, and the major unanswered questions in its biology.
Collapse
Affiliation(s)
- Ankit Malik
- Department of Immunology St. Jude Children’s Research Hospital, Memphis, TN 38105
| | | |
Collapse
|
107
|
Theut Riis P, Thorlacius LR, Jemec GB. Investigational drugs in clinical trials for Hidradenitis Suppurativa. Expert Opin Investig Drugs 2017; 27:43-53. [PMID: 29188733 DOI: 10.1080/13543784.2018.1412430] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Hidradenitis suppurativa is a chronic skin disease with a significant unmet need for treatment options. Randomized controlled trials are few and only a single drug (adalimumab) has Hidradenitis as a registered indication. AREAS COVERED The clinicaltrials.gov and the EudraCT clinical trials register for reported trials on Hidradenitis Suppurativa was searched on the 22-06-2017. Trials for upcoming new drugs for HS are reported focusing on drugs in phase I and II trials.The clinicaltrials.gov and the EudraCT clinical trials register for reported trials on Hidradenitis Suppurativa was searched on the 22-06-2017. Trials for upcoming new drugs for HS are reported focusing on drugs in phase I and II trials. EXPERT OPINION Currently, MABp1, Secukinumab, CJM112, Apremilast and IFX-1 are being investigated in Phase I and II trials and offer theoretical and promising new treatment options. A trial with the drug MEDI8968 has been terminated with disappointing results. Metformin, Botulinum Toxin B, Provodine, Benzoyl Peroxide and intralesional triamcinolone are being tested as well. Treatment of Hidradenitis remains a challenge and quality RTCs are needed. Studies indicates a range of potential targets for therapy such as interleukin-1 and interleukin-17, but 'broad-spectrum' immunosuppressants like phosphodiesterase-4 inhibitors are being examined as well. A range of outcomes, including Physician Global Assessment, Sartorius scores and hidradenitis suppurativa clinical response are used in these trials, making future meta-analysis of the data difficult.
Collapse
Affiliation(s)
- Peter Theut Riis
- a Department of Dermatology , University Hospital Zealand , Roskilde , Denmark.,b Health Sciences Faculty , University of Copenhagen , Copenhagen , Denmark
| | - Linnea R Thorlacius
- a Department of Dermatology , University Hospital Zealand , Roskilde , Denmark.,b Health Sciences Faculty , University of Copenhagen , Copenhagen , Denmark
| | - Gregor B Jemec
- a Department of Dermatology , University Hospital Zealand , Roskilde , Denmark.,b Health Sciences Faculty , University of Copenhagen , Copenhagen , Denmark
| |
Collapse
|
108
|
Abstract
PURPOSE OF REVIEW In spite of its relevance, treatments for the cancer anorexia and cachexia syndrome (CACS) are not available. One of the agents that recently reached phase III clinical trials is anamorelin. Its development, along with that of other agents for this indication, will be reviewed here, with a focus on the gaps in the current knowledge and future directions. RECENT FINDINGS In spite of several targets showing promising results in early development, their difficulties obtaining regulatory approval underscore the need to reconsider the current strategies in drug development and the challenges in the field of CACS. SUMMARY Further research is needed in order to meet the challenges of developing treatments for CACS. Preclinical studies should expand our understanding about key regulators of appetite, muscle, and energy metabolism in this setting using models that can be translated reliably to humans. Clinical research efforts should focus on validating the entry criteria, endpoints, outcomes, and the potential synergistic effects and interaction between different targets, nutrition, and exercise interventions. Clinical meaningfulness and significance should be taken into account in the design of clinical trials. It is essential that all key stakeholders are included in the design of future strategies.
Collapse
Affiliation(s)
- Jose M. Garcia
- Geriatric Research, Education and Clinical Center (GRECC), VA Puget Sound Health Care System, Seattle, WA
- Department of Medicine, Division of Gerontology & Geriatric Medicine, University of Washington School of Medicine, Seattle, WA
| |
Collapse
|
109
|
The state-of-play and future of antibody therapeutics. Adv Drug Deliv Rev 2017; 122:2-19. [PMID: 27916504 DOI: 10.1016/j.addr.2016.11.004] [Citation(s) in RCA: 208] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 11/26/2016] [Accepted: 11/28/2016] [Indexed: 12/22/2022]
Abstract
It has been over four decades since the development of monoclonal antibodies (mAbs) using a hybridoma cell line was first reported. Since then more than thirty therapeutic antibodies have been marketed, mostly as oncology, autoimmune and inflammatory therapeutics. While antibodies are very efficient, their cost-effectiveness has always been discussed owing to their high costs, accumulating to more than one billion dollars from preclinical development through to market approval. Because of this, therapeutic antibodies are inaccessible to some patients in both developed and developing countries. The growing interest in biosimilar antibodies as affordable versions of therapeutic antibodies may provide alternative treatment options as well potentially decreasing costs. As certain markets begin to capitalize on this opportunity, regulatory authorities continue to refine the requirements for demonstrating quality, efficacy and safety of biosimilar compared to originator products. In addition to biosimilars, innovations in antibody engineering are providing the opportunity to design biobetter antibodies with improved properties to maximize efficacy. Enhancing effector function, antibody drug conjugates (ADC) or targeting multiple disease pathways via multi-specific antibodies are being explored. The manufacturing process of antibodies is also moving forward with advancements relating to host cell production and purification processes. Studies into the physical and chemical degradation pathways of antibodies are contributing to the design of more stable proteins guided by computational tools. Moreover, the delivery and pharmacokinetics of antibody-based therapeutics are improving as optimized formulations are pursued through the implementation of recent innovations in the field.
Collapse
|
110
|
Muscular Dystrophies and Cancer Cachexia: Similarities in Chronic Skeletal Muscle Degeneration. J Funct Morphol Kinesiol 2017. [DOI: 10.3390/jfmk2040039] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
111
|
Libby P. Interleukin-1 Beta as a Target for Atherosclerosis Therapy: Biological Basis of CANTOS and Beyond. J Am Coll Cardiol 2017; 70:2278-2289. [PMID: 29073957 DOI: 10.1016/j.jacc.2017.09.028] [Citation(s) in RCA: 432] [Impact Index Per Article: 61.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 09/07/2017] [Accepted: 09/10/2017] [Indexed: 02/07/2023]
Abstract
Inflammatory pathways drive atherogenesis and link conventional risk factors to atherosclerosis and its complications. One inflammatory mediator has come to the fore as a therapeutic target in cardiovascular disease. The experimental and clinical evidence reviewed here support interleukin-1 beta (IL-1β) as both a local vascular and systemic contributor in this regard. Intrinsic vascular wall cells and lesional leukocytes alike can produce this cytokine. Local stimuli in the plaque favor the generation of active IL-1β through the action of a molecular assembly known as the inflammasome. Clinically applicable interventions that interfere with IL-1 action can improve cardiovascular outcomes, ushering in a new era of anti-inflammatory therapies for atherosclerosis. The translational path described here illustrates how advances in basic vascular biology may transform therapy. Biomarker-directed application of anti-inflammatory interventions promises to help us achieve a more precise and personalized allocation of therapy for our cardiovascular patients.
Collapse
Affiliation(s)
- Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
112
|
Gong J, Wu D, Chuang J, Tuli R, Simard J, Hendifar A. Moving Beyond Conventional Clinical Trial End Points in Treatment-refractory Metastatic Colorectal Cancer: A Composite Quality-of-life and Symptom Control End Point. Clin Ther 2017; 39:2135-2145. [PMID: 29079389 DOI: 10.1016/j.clinthera.2017.09.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 09/29/2017] [Accepted: 09/29/2017] [Indexed: 12/13/2022]
Abstract
PURPOSE This review highlights the evidence supporting symptom control and quality-of-life (QOL) measures as predictors of survival in treatment-refractory metastatic colorectal cancer (mCRC) and describes a composite symptom control and QOL end point recently reported in a Phase III trial that may serve as a more reasonable end point of efficacy in this population. METHODS A literature search was conducted using MEDLINE to identify clinical studies (including case series and observational, retrospective, and prospective studies) that reported the predictive value of QOL measures for survival in mCRC. The search was limited by the following key words: quality of life, survival, and colorectal cancer. We then performed a second search limited to studies of randomized and Phase III design in mCRC to identify studies that used QOL assessments as their primary end points. A manual search was also performed to include additional studies of potential relevance. FINDINGS There is increasing evidence to support that symptom control and QOL measures are predictors of survival in treatment-refractory mCRC and can serve as an alternative but equally as important end point to survival in this population. A recent large, randomized Phase III trial using a composite primary end point of lean body mass, pain, anorexia, and fatigue reported the feasibility in evaluating benefit in mCRC beyond conventional clinical trial end points. IMPLICATIONS Future studies in treatment-refractory mCRC may be better served by evaluating improvement in symptom control and QOL, which may otherwise serve as the best predictor of survival in last-line treatment settings.
Collapse
Affiliation(s)
- Jun Gong
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, California
| | - Daniel Wu
- Department of Internal Medicine, Harbor-UCLA Medical Center, Torrance, California
| | - Jeremy Chuang
- Department of Internal Medicine, Harbor-UCLA Medical Center, Torrance, California
| | - Richard Tuli
- Gastrointestinal and Neuroendocrine Malignancies, Samuel Oschin Cancer Center, Cedars-Sinai Medical Center, Los Angeles, California
| | | | - Andrew Hendifar
- Gastrointestinal and Neuroendocrine Malignancies, Samuel Oschin Cancer Center, Cedars-Sinai Medical Center, Los Angeles, California.
| |
Collapse
|
113
|
Ridker PM, MacFadyen JG, Thuren T, Everett BM, Libby P, Glynn RJ. Effect of interleukin-1β inhibition with canakinumab on incident lung cancer in patients with atherosclerosis: exploratory results from a randomised, double-blind, placebo-controlled trial. Lancet 2017; 390:1833-1842. [PMID: 28855077 DOI: 10.1016/s0140-6736(17)32247-x] [Citation(s) in RCA: 909] [Impact Index Per Article: 129.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 08/02/2017] [Accepted: 08/03/2017] [Indexed: 02/08/2023]
Abstract
BACKGROUND Inflammation in the tumour microenvironment mediated by interleukin 1β is hypothesised to have a major role in cancer invasiveness, progression, and metastases. We did an additional analysis in the Canakinumab Anti-inflammatory Thrombosis Outcomes Study (CANTOS), a randomised trial of the role of interleukin-1β inhibition in atherosclerosis, with the aim of establishing whether inhibition of a major product of the Nod-like receptor protein 3 (NLRP3) inflammasome with canakinumab might alter cancer incidence. METHODS We did a randomised, double-blind, placebo-controlled trial of canakinumab in 10 061 patients with atherosclerosis who had had a myocardial infarction, were free of previously diagnosed cancer, and had concentrations of high-sensitivity C-reactive protein (hsCRP) of 2 mg/L or greater. To assess dose-response effects, patients were randomly assigned by computer-generated codes to three canakinumab doses (50 mg, 150 mg, and 300 mg, subcutaneously every 3 months) or placebo. Participants were followed up for incident cancer diagnoses, which were adjudicated by an oncology endpoint committee masked to drug or dose allocation. Analysis was by intention to treat. The trial is registered with ClinicalTrials.gov, NCT01327846. The trial is closed (the last patient visit was in June, 2017). FINDINGS Baseline concentrations of hsCRP (median 6·0 mg/L vs 4·2 mg/L; p<0·0001) and interleukin 6 (3·2 vs 2·6 ng/L; p<0·0001) were significantly higher among participants subsequently diagnosed with lung cancer than among those not diagnosed with cancer. During median follow-up of 3·7 years, compared with placebo, canakinumab was associated with dose-dependent reductions in concentrations of hsCRP of 26-41% and of interleukin 6 of 25-43% (p<0·0001 for all comparisons). Total cancer mortality (n=196) was significantly lower in the pooled canakinumab group than in the placebo group (p=0·0007 for trend across groups), but was significantly lower than placebo only in the 300 mg group individually (hazard ratio [HR] 0·49 [95% CI 0·31-0·75]; p=0·0009). Incident lung cancer (n=129) was significantly less frequent in the 150 mg (HR 0·61 [95% CI 0·39-0·97]; p=0·034) and 300 mg groups (HR 0·33 [95% CI 0·18-0·59]; p<0·0001; p<0·0001 for trend across groups). Lung cancer mortality was significantly less common in the canakinumab 300 mg group than in the placebo group (HR 0·23 [95% CI 0·10-0·54]; p=0·0002) and in the pooled canakinumab population than in the placebo group (p=0·0002 for trend across groups). Fatal infections or sepsis were significantly more common in the canakinumab groups than in the placebo group. All-cause mortality did not differ significantly between the canakinumab and placebo groups (HR 0·94 [95% CI 0·83-1·06]; p=0·31). INTERPRETATION Our hypothesis-generating data suggest the possibility that anti-inflammatory therapy with canakinumab targeting the interleukin-1β innate immunity pathway could significantly reduce incident lung cancer and lung cancer mortality. Replication of these data in formal settings of cancer screening and treatment is required. FUNDING Novartis Pharmaceuticals.
Collapse
Affiliation(s)
- Paul M Ridker
- Center for Cardiovascular Disease Prevention, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Jean G MacFadyen
- Center for Cardiovascular Disease Prevention, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Tom Thuren
- Novartis Pharmaceuticals, East Hanover, NJ, USA; Novartis Pharmaceuticals, Basel, Switzerland
| | - Brendan M Everett
- Center for Cardiovascular Disease Prevention, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Peter Libby
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Robert J Glynn
- Center for Cardiovascular Disease Prevention, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
114
|
Abstract
Cachexia is a metabolic syndrome driven by inflammation and characterized by loss of muscle with or without loss of fat mass. In cancer cachexia, the tumor burden and host response induce increased inflammation, decreased anabolic tone, and suppressed appetite leading to the clinical presentation of reduced body weight and quality of life (QOL). There is no approved treatment for cancer cachexia, and commonly used nutritional and anti-inflammatory strategies alone have proven ineffective for management of symptoms. Several other pharmacological agents are currently in development and have shown promise as a clinical strategy in early-phase trials. Recently, it has been proposed that multimodal strategies, with an anabolic focus, initiated early in the disease/treatment progression may provide the most therapeutic potential for symptom management. Here we review the data from recent clinical trials in cancer cachexia including pharmacological, exercise, and nutritional interventions.
Collapse
Affiliation(s)
- Lindsey J Anderson
- Geriatric Research, Education and Clinical Center (GRECC), VA Puget Sound Health Care System, Seattle, WA, 98108, USA
| | - Eliette D Albrecht
- Geriatric Research, Education and Clinical Center (GRECC), VA Puget Sound Health Care System, Seattle, WA, 98108, USA.,Yale University, New Haven, CT, 06520, USA
| | - Jose M Garcia
- Geriatric Research, Education and Clinical Center (GRECC), VA Puget Sound Health Care System, Seattle, WA, 98108, USA. .,Department of Medicine, Division of Gerontology & Geriatric Medicine, University of Washington School of Medicine, Seattle, WA, USA.
| |
Collapse
|
115
|
Rider P, Voronov E, Dinarello CA, Apte RN, Cohen I. Alarmins: Feel the Stress. THE JOURNAL OF IMMUNOLOGY 2017; 198:1395-1402. [PMID: 28167650 DOI: 10.4049/jimmunol.1601342] [Citation(s) in RCA: 128] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 09/27/2016] [Indexed: 12/20/2022]
Abstract
Over the last decade, danger-associated molecular pattern molecules, or alarmins, have been recognized as signaling mediators of sterile inflammatory responses after trauma and injury. In contrast with the accepted passive release models suggested by the "danger hypothesis," it was recently shown that alarmins can also directly sense and report damage by signaling to the environment when released from live cells undergoing physiological stress, even without loss of subcellular compartmentalization. In this article, we review the involvement of alarmins such as IL-1α, IL-33, IL-16, and high-mobility group box 1 in cellular and physiological stress, and suggest a novel activity of these molecules as central initiators of sterile inflammation in response to nonlethal stress, a function we denote "stressorins." We highlight the role of posttranslational modifications of stressorins as key regulators of their activity and propose that targeted inhibition of stressorins or their modifiers could serve as attractive new anti-inflammatory treatments for a broad range of diseases.
Collapse
Affiliation(s)
- Peleg Rider
- Department of Clinical Biochemistry and Pharmacology, Ben Gurion University of the Negev, 84105 Beer-Sheva, Israel
| | - Elena Voronov
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben Gurion University of the Negev, 84105 Beer-Sheva, Israel
| | | | - Ron N Apte
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben Gurion University of the Negev, 84105 Beer-Sheva, Israel
| | - Idan Cohen
- Faculty of Medicine, Galilee Medical Center, Nahariya Hospital, 22100 Nahariya, Israel
| |
Collapse
|
116
|
Shinko D, Diakos CI, Clarke SJ, Charles KA. Cancer-Related Systemic Inflammation: The Challenges and Therapeutic Opportunities for Personalized Medicine. Clin Pharmacol Ther 2017; 102:599-610. [PMID: 28699186 DOI: 10.1002/cpt.789] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 06/23/2017] [Accepted: 07/06/2017] [Indexed: 12/15/2022]
Abstract
Over the last decade there has been significant progress towards the development of personalized or "precision" medicine for many patients with cancer. However, there still remain subpopulations of cancer patients that do not possess a tumor mutation profile that is successfully targeted by the newer molecular anticancer drugs and further personalized approaches are needed. The presence of cancer-related systemic inflammation represents an underappreciated subpopulation of cancer patients needing personalized therapy. For ∼25% of all advanced cancer patients, regardless of histological subtype, the patients with systemic inflammation have significantly poorer response to chemotherapy and also shorter overall survival compared to those cancer patients without inflammation. The development of cancer-related systemic inflammation involves interactions between host and tumor cells that are potential new drug targets in cancer chemotherapy. In this review we discuss the challenges and clinical opportunities to develop new therapeutic strategies for this underappreciated drug target.
Collapse
Affiliation(s)
- Diana Shinko
- Discipline of Pharmacology, Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Connie I Diakos
- Northern Sydney Cancer Centre, Royal North Shore Hospital, St Leonards, NSW, Australia.,Sydney Medical School, University of Sydney, Sydney, NSW, Australia.,Bill Walsh Translational Research Laboratories, Kolling Institute of Medical Research, St Leonards, NSW, Australia
| | - Stephen J Clarke
- Northern Sydney Cancer Centre, Royal North Shore Hospital, St Leonards, NSW, Australia.,Sydney Medical School, University of Sydney, Sydney, NSW, Australia.,Bill Walsh Translational Research Laboratories, Kolling Institute of Medical Research, St Leonards, NSW, Australia
| | - Kellie A Charles
- Discipline of Pharmacology, Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
117
|
Abstract
INTRODUCTION Inflammation is an established process in colorectal cancer development and a hallmark of progression, and pro-inflammatory cytokines have been implicated in the morbidity and functional compromise associated with malignancy. MABp1, described as a first-in-class true human antibody against interleukin-1α, has undergone clinical trial evaluation in a number of indications, recently completing late phase clinical trial testing under Fast Track designation for cancer anorexia-cachexia syndrome in colorectal cancer patients. To date, MABp1 has been evaluated as a novel therapeutic strategy to ameliorate phenotypic factors associated with poor prognosis in colorectal cancer patients. Areas covered: In this review, the authors discuss the clinical trial data available to date for this antibody in colorectal cancer, including novel clinical trial endpoints utilized to evaluate sarcopenia and inflammation, as well as the proposed role of interleukin-1α antagonism in leading to improved patient outcomes. Expert opinion: There is a multitude of antibodies in therapeutic development in oncology, and MABp1 is a novel class of antibody which has been safely tolerated to date. Clinical studies of this agent suggest a significant improvement in lean body mass, though additional results evaluating the impact of targeting inflammation as a strategy to delay disease progression in this population are awaited.
Collapse
Affiliation(s)
- Geraldine O'Sullivan Coyne
- a Developmental Therapeutics Clinic, Division of Cancer Treatment and Diagnosis , National Cancer Institute, National Institutes of Health , Bethesda , MD , USA
| | - Mauricio Burotto
- b Medical Oncology Service , Clinica Alemana De Santiago,universidad Del Desarrollo , Santiago , Chile
| |
Collapse
|
118
|
Thomsen M, Guren MG, Skovlund E, Glimelius B, Hjermstad MJ, Johansen JS, Kure E, Sorbye H, Pfeiffer P, Christoffersen T, Guren TK, Tveit KM. Health-related quality of life in patients with metastatic colorectal cancer, association with systemic inflammatory response and RAS and BRAF mutation status. Eur J Cancer 2017; 81:26-35. [PMID: 28595137 DOI: 10.1016/j.ejca.2017.04.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 04/23/2017] [Accepted: 04/29/2017] [Indexed: 01/22/2023]
Abstract
BACKGROUND The aim of this study was to evaluate the effect of cetuximab on health-related quality of life (HRQoL) in the NORDIC-VII trial on metastatic colorectal cancer (mCRC), and to assess HRQoL in relation to RAS and BRAF mutation status and inflammatory biomarkers. PATIENT AND METHODS HRQoL was assessed using the European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire Core 30 (QLQ-C30) at baseline, after every fourth cycle of chemotherapy, and at the end of treatment. HRQoL during 12 cycles of chemotherapy was evaluated over time, compared between treatment arms, and assessed for association with tumour mutation status and inflammatory markers. RESULTS QLQ-C30 was completed by 512 patients (90%) before start of treatment. HRQoL variables were well balanced across treatment arms at baseline, and no statistically significant differences during treatment were seen. Patients with BRAF-mutated tumours reported poorer HRQoL at baseline and subsequent time points than patients with RAS-mutated or RAS/BRAF wild-type tumours. Patients with high serum interleukin-6 (IL-6) or C-reactive protein (CRP) had markedly impaired HRQoL compared to patients with normal levels. There was a statistically significant association between reduction in IL-6 and CRP levels and improvement in HRQoL during treatment from baseline to cycle 4. CONCLUSION The addition of cetuximab to chemotherapy did not affect HRQoL in mCRC patients. Patients with BRAF-mutated tumours have both a worse prognosis and a poor HRQoL. The associations between levels of systemic inflammatory markers and reduced HRQoL suggest that the patients might benefit from anti-inflammatory treatment.
Collapse
Affiliation(s)
- Maria Thomsen
- Department of Oncology, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Marianne Grønlie Guren
- Department of Oncology, Oslo University Hospital, Oslo, Norway; K.G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Oslo, Norway
| | - Eva Skovlund
- Department of Public Health and Nursing, NTNU, Trondheim, Norway; Mental and Physical Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Bengt Glimelius
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Marianne Jensen Hjermstad
- Regional Advisory Unit for Palliative Care, Department of Oncology, Oslo University Hospital, Oslo, Norway; European Palliative Care Research Centre, Department of Cancer Research and Molecular Medicine, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Julia S Johansen
- Department of Oncology, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
| | - Elin Kure
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Halfdan Sorbye
- Department of Oncology, Haukeland University Hospital, and Clinical Science, University of Bergen, Bergen, Norway
| | - Per Pfeiffer
- Department of Oncology, Odense University Hospital, Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Thoralf Christoffersen
- Department of Pharmacology, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Tormod Kyrre Guren
- Department of Oncology, Oslo University Hospital, Oslo, Norway; K.G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Oslo, Norway
| | - Kjell Magne Tveit
- Department of Oncology, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway; K.G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
119
|
Fenini G, Contassot E, French LE. Potential of IL-1, IL-18 and Inflammasome Inhibition for the Treatment of Inflammatory Skin Diseases. Front Pharmacol 2017; 8:278. [PMID: 28588486 PMCID: PMC5438978 DOI: 10.3389/fphar.2017.00278] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 05/02/2017] [Indexed: 11/13/2022] Open
Abstract
In 2002, intracellular protein complexes known as the inflammasomes were discovered and were shown to have a crucial role in the sensing of intracellular pathogen- and danger-associated molecular patterns (PAMPs and DAMPs). Activation of the inflammasomes results in the processing and subsequent secretion of the pro-inflammatory cytokines IL-1β and IL-18. Several autoinflammatory disorders such as cryopyrin-associated periodic syndromes and Familial Mediterranean Fever have been associated with mutations of genes encoding inflammasome components. Moreover, the importance of IL-1 has been reported for an increasing number of autoinflammatory skin diseases including but not limited to deficiency of IL-1 receptor antagonist, mevalonate kinase deficiency and PAPA syndrome. Recent findings have revealed that excessive IL-1 release induced by harmful stimuli likely contributes to the pathogenesis of common dermatological diseases such as acne vulgaris or seborrheic dermatitis. A key pathogenic feature of these diseases is IL-1β-induced neutrophil recruitment to the skin. IL-1β blockade may therefore represent a promising therapeutic approach. Several case reports and clinical trials have demonstrated the efficacy of IL-1 inhibition in the treatment of these skin disorders. Next to the recombinant IL-1 receptor antagonist (IL-1Ra) Anakinra and the soluble decoy Rilonacept, the anti-IL-1α monoclonal antibody MABp1 and anti-IL-1β Canakinumab but also Gevokizumab, LY2189102 and P2D7KK, offer valid alternatives to target IL-1. Although less thoroughly investigated, an involvement of IL-18 in the development of cutaneous inflammatory disorders is also suspected. The present review describes the role of IL-1 in diseases with skin involvement and gives an overview of the relevant studies discussing the therapeutic potential of modulating the secretion and activity of IL-1 and IL-18 in such diseases.
Collapse
Affiliation(s)
- Gabriele Fenini
- Department of Dermatology, University Hospital ZurichZurich, Switzerland
| | - Emmanuel Contassot
- Department of Dermatology, University Hospital ZurichZurich, Switzerland
| | - Lars E French
- Department of Dermatology, University Hospital ZurichZurich, Switzerland
| |
Collapse
|
120
|
Affiliation(s)
- Jiajie Hou
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Rd, Shanghai 200127, China
| | - Tim F Greten
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, Building 10, Bethesda, Maryland 20892, USA
| | - Qiang Xia
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Rd, Shanghai 200127, China
| |
Collapse
|
121
|
Aversa Z, Costelli P, Muscaritoli M. Cancer-induced muscle wasting: latest findings in prevention and treatment. Ther Adv Med Oncol 2017; 9:369-382. [PMID: 28529552 PMCID: PMC5424865 DOI: 10.1177/1758834017698643] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 02/14/2017] [Indexed: 12/17/2022] Open
Abstract
Cancer cachexia is a severe and disabling clinical condition that frequently accompanies the development of many types of cancer. Muscle wasting is the hallmark of cancer cachexia and is associated with serious clinical consequences such as physical impairment, poor quality of life, reduced tolerance to treatments and shorter survival. Cancer cachexia may evolve through different stages of clinical relevance, namely pre-cachexia, cachexia and refractory cachexia. Given its detrimental clinical consequences, it appears mandatory to prevent and/or delay the progression of cancer cachexia to its refractory stage by implementing the early recognition and treatment of the nutritional and metabolic alterations occurring during cancer. Research on the molecular mechanisms underlying muscle wasting during cancer cachexia has expanded in the last few years, allowing the identification of several potential therapeutic targets and the development of many promising drugs. Several of these agents have already reached the clinical evaluation, but it is becoming increasingly evident that a single therapy may not be completely successful in the treatment of cancer-related muscle wasting, given its multifactorial and complex pathogenesis. This suggests that early and structured multimodal interventions (including targeted nutritional supplementation, physical exercise and pharmacological interventions) are necessary to prevent and/or treat the devastating consequences of this cancer comorbidity, and future research should focus on this approach.
Collapse
Affiliation(s)
- Zaira Aversa
- Department of Clinical Medicine, Sapienza University of Rome, Italy
| | - Paola Costelli
- Department of Clinical and Biological Sciences, University of Turin, Italy
| | - Maurizio Muscaritoli
- Department of Clinical Medicine, Sapienza, University of Rome, Viale dell’Università 37, 00185 Rome, Italy
| |
Collapse
|
122
|
Burns JC, Koné-Paut I, Kuijpers T, Shimizu C, Tremoulet A, Arditi M. Review: Found in Translation: International Initiatives Pursuing Interleukin-1 Blockade for Treatment of Acute Kawasaki Disease. Arthritis Rheumatol 2017; 69:268-276. [PMID: 27792871 PMCID: PMC5274552 DOI: 10.1002/art.39975] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Accepted: 10/27/2016] [Indexed: 12/19/2022]
Abstract
The decision to move forward with three clinical trials of IL-1 blockade for treatment of acute Kawasaki disease is a case study in translational science. These trials were born on the one hand from transcriptome studies of host response during the acute disease coupled with animal model investigations of key immune signaling pathways and, on the other hand, out of clinical desperation to intervene in patients with severe inflammation in the setting of acute Kawasaki disease. The convergence of laboratory science and clinical observations led to the clinical trials described here and serves as a model for how such observations can be translated into new therapies.
Collapse
Affiliation(s)
- Jane C. Burns
- Department of Pediatrics, University of California San Diego and Rady Children’s Hospital San Diego. 9500 Gilman Dr. La Jolla, CA 92093-0641
| | - Isabelle Koné-Paut
- Isabelle Koné-Paut, Pediatric Rheumatology Department, National Reference Center for Autoinflammatory Diseases (CEREMAI), Bicêtre Hospital, APHP, University of Paris Sud, France
| | - Taco Kuijpers
- Academic Medical Center Emma Children's Hospital University of Amsterdam, Amsterdam, 1105 AZ Amsterdam, Netherlands
| | - Chisato Shimizu
- Department of Pediatrics, University of California San Diego and Rady Children’s Hospital San Diego. 9500 Gilman Dr. La Jolla, CA 92093-0641
| | - Adriana Tremoulet
- Department of Pediatrics, University of California San Diego and Rady Children’s Hospital San Diego. 9500 Gilman Dr. La Jolla, CA 92093-0641
| | - Moshe Arditi
- Departments of Pediatrics and Biomedical Sciences, Cedars Sinai Medical Center and UCLA School of Medicine, Los Angeles, CA 90048
| |
Collapse
|
123
|
Hickish T, Andre T, Wyrwicz L, Saunders M, Sarosiek T, Kocsis J, Nemecek R, Rogowski W, Lesniewski-Kmak K, Petruzelka L, Apte RN, Mohanty P, Stecher M, Simard J, de Gramont A. MABp1 as a novel antibody treatment for advanced colorectal cancer: a randomised, double-blind, placebo-controlled, phase 3 study. Lancet Oncol 2017; 18:192-201. [PMID: 28094194 DOI: 10.1016/s1470-2045(17)30006-2] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 10/07/2016] [Accepted: 10/19/2016] [Indexed: 12/29/2022]
Abstract
BACKGROUND MABp1, an antibody that targets interleukin 1α, has been associated with antitumour activity and relief of debilitating symptoms in patients with advanced colorectal cancer. We sought to establish the effect of MABp1 with a new primary endpoint in patients with advanced colorectal cancer. METHODS Eligible patients for the double-blind phase of this ongoing, placebo-controlled, randomised, phase 3 trial, had metastatic or unresectable disease, Eastern Cooperative Oncology Group performance status score 1 or 2, systemic inflammation, weight loss, and other disease-related morbidities associated with poor prognosis, and were refractory to oxaliplatin and irinotecan. Patients were randomly assigned 2:1 to receive either MABp1 or placebo. Randomisation codes were obtained from a centrally held list via an interactive web response system. Patients received an intravenous infusion of 7·5 mg/kg MABp1 or placebo given every 2 weeks for 8 weeks. The primary endpoint was assessed in patients who received at least one dose of MABp1 or placebo (modified intention-to-treat population), and was a composite of stable or increased lean body mass and stability or improvement in two of three symptoms (pain, fatigue, or anorexia) at week 8 compared with baseline measurements. This study is registered with ClinicalTrials.gov, number NCT02138422. FINDINGS Patients were enrolled between May 20, 2014, and Sept 2, 2015. The double-blind phase of the study was completed on Nov 3, 2015. Of 333 patients randomly assigned treatment, 207 received at least one dose of MABp1 and 102 at least one dose of placebo. 68 (33%) and 19 (19%) patients, respectively, achieved the primary endpoint (relative risk 1·76, 95% CI 1·12-2·77, p=0·0045). The most common grade 3-4 adverse events in the MABp1 group compared with in the placebo group were anaemia (eight [4%] of 207 vs five [5%] of 102 patients), increased concentration of alkaline phosphatase (nine [4%] vs two [2%]), fatigue (six [3%] vs seven [7%]), and increased concentration of aspartate aminotransferase (six [3%] vs two [2%]). After 8 weeks, 17 (8%) patients in the MABp1 group and 11 (11%) in the placebo group had died, but no death was judged to be related to treatment. The incidence of serious adverse events was not significantly different in the MABp1 group and placebo groups (47 [23%] vs 33 [32%], p=0·07). INTERPRETATION The primary endpoint was a useful means of measuring clinical performance in patients. MABp1 might represent a new standard in the management of advanced colorectal cancer. FUNDING XBiotech.
Collapse
Affiliation(s)
- Tamas Hickish
- Poole Hospital NHS Foundation Trust, Poole, Dorset, UK; Oncology Department, Royal Bournemouth Hospital NHS Foundation Trust Bournemouth, UK; Department of Oncology, Bournemouth University, Bournemouth, UK.
| | - Thierry Andre
- Oncology Department, Saint Antoine Hospital, and Pierre and Marie Curie University (Paris 6), Paris, France
| | - Lucjan Wyrwicz
- Maria Sklodowska-Curie Memorial Cancer Centre and Institute of Oncology, Warsaw, Poland
| | | | | | | | - Radim Nemecek
- Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Wojciech Rogowski
- Clinical Department of Chemotherapy, Hospital Ministry of the Interior and Administration and Warmia and Mazury Oncology Centre, Olsztyn, Poland
| | | | | | - Ron N Apte
- Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | | | | | | | - Aimery de Gramont
- Oncology Department, Institut Hospitalier Franco-Britannique, Levallois-Perret, France
| |
Collapse
|
124
|
Korneev KV, Atretkhany KSN, Drutskaya MS, Grivennikov SI, Kuprash DV, Nedospasov SA. TLR-signaling and proinflammatory cytokines as drivers of tumorigenesis. Cytokine 2017; 89:127-135. [DOI: 10.1016/j.cyto.2016.01.021] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 01/26/2016] [Accepted: 01/27/2016] [Indexed: 12/29/2022]
|
125
|
Blomme A, Cusumano P, Peulen O, Bellahcène A, Castronovo V, Turtoi A. [Asporin: the protective wall against triple-negative breast cancer]. Med Sci (Paris) 2016; 32:1019-1022. [PMID: 28008845 DOI: 10.1051/medsci/20163211020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Arnaud Blomme
- Metastasis Research Laboratory, GIGA-Cancer, University of Liege, avenue de l'Hôpital 3, 4000 Liege, Belgique
| | - Pino Cusumano
- Metastasis Research Laboratory, GIGA-Cancer, University of Liege, avenue de l'Hôpital 3, 4000 Liege, Belgique - Department of senology, university hospital (CHU), University of Liège, Liège, Belgique
| | - Olivier Peulen
- Metastasis Research Laboratory, GIGA-Cancer, University of Liege, avenue de l'Hôpital 3, 4000 Liege, Belgique
| | - Akeila Bellahcène
- Metastasis Research Laboratory, GIGA-Cancer, University of Liege, avenue de l'Hôpital 3, 4000 Liege, Belgique
| | - Vincent Castronovo
- Metastasis Research Laboratory, GIGA-Cancer, University of Liege, avenue de l'Hôpital 3, 4000 Liege, Belgique
| | - Andrei Turtoi
- Metastasis Research Laboratory, GIGA-Cancer, University of Liege, avenue de l'Hôpital 3, 4000 Liege, Belgique - Institut de Recherche en Cancérologie de Montpellier ; Inserm U1194, Montpellier F-34298, France
| |
Collapse
|
126
|
Biologics for Targeting Inflammatory Cytokines, Clinical Uses, and Limitations. Int J Cell Biol 2016; 2016:9259646. [PMID: 28083070 PMCID: PMC5204077 DOI: 10.1155/2016/9259646] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 10/03/2016] [Accepted: 10/20/2016] [Indexed: 01/08/2023] Open
Abstract
Proinflammatory cytokines are potent mediators of numerous biological processes and are tightly regulated in the body. Chronic uncontrolled levels of such cytokines can initiate and derive many pathologies, including incidences of autoimmunity and cancer. Therefore, therapies that regulate the activity of inflammatory cytokines, either by supplementation of anti-inflammatory recombinant cytokines or by neutralizing them by using blocking antibodies, have been extensively used over the past decades. Over the past few years, new innovative biological agents for blocking and regulating cytokine activities have emerged. Here, we review some of the most recent approaches of cytokine targeting, focusing on anti-TNF antibodies or recombinant TNF decoy receptor, recombinant IL-1 receptor antagonist (IL-1Ra) and anti-IL-1 antibodies, anti-IL-6 receptor antibodies, and TH17 targeting antibodies. We discuss their effects as biologic drugs, as evaluated in numerous clinical trials, and highlight their therapeutic potential as well as emphasize their inherent limitations and clinical risks. We suggest that while systemic blocking of proinflammatory cytokines using biological agents can ameliorate disease pathogenesis and progression, it may also abrogate the hosts defense against infections. Moreover, we outline the rational need to develop new therapies, which block inflammatory cytokines only at sites of inflammation, while enabling their function systemically.
Collapse
|
127
|
Malik A, Sharma D, Zhu Q, Karki R, Guy CS, Vogel P, Kanneganti TD. IL-33 regulates the IgA-microbiota axis to restrain IL-1α-dependent colitis and tumorigenesis. J Clin Invest 2016; 126:4469-4481. [PMID: 27775548 PMCID: PMC5127671 DOI: 10.1172/jci88625] [Citation(s) in RCA: 136] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 09/15/2016] [Indexed: 12/14/2022] Open
Abstract
Inflammatory bowel diseases (IBD) affect over 5 million individuals in the industrialized world, with an increasing incidence rate worldwide. IBD also predisposes affected individuals to development of colorectal cancer, which is a leading cause of cancer-related deaths in adults. Mutations in genes encoding molecules in the IL-33 signaling pathway are associated with colitis and colitis-associated cancer (CAC), but how IL-33 modulates gut homeostasis is unclear. Here, we have shown that Il33-deficient mice are highly susceptible to colitis and CAC. Mechanistically, we observed that IL-33 promoted IgA production from B cells, which is important for maintaining microbial homeostasis in the intestine. Il33-deficient mice developed a dysbiotic microbiota that was characterized by increased levels of mucolytic and colitogenic bacteria. In response to chemically induced colitis, this microbial landscape promoted the release of IL-1α, which acted as a critical driver of colitis and CAC. Consequently, reconstitution of symbiotic microbiota or IL-1α ablation markedly ameliorated colitis susceptibility in Il33-deficient animals. Our results demonstrate that IL-33 promotes IgA production to maintain gut microbial homoeostasis and restrain IL-1α-dependent colitis and CAC. This study therefore highlights modulation of IL-33, IgA, IL-1α, and the microbiota as a potential therapeutic approach in the treatment of IBD and CAC.
Collapse
Affiliation(s)
- Ankit Malik
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Deepika Sharma
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Qifan Zhu
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
- Integrated Biomedical Sciences Program, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Rajendra Karki
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Clifford S. Guy
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Peter Vogel
- Animal Resources Center and the Veterinary Pathology Core, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | | |
Collapse
|
128
|
Emerging therapies for the treatment of skeletal muscle wasting in chronic obstructive pulmonary disease. Pharmacol Ther 2016; 166:56-70. [DOI: 10.1016/j.pharmthera.2016.06.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/14/2016] [Indexed: 12/18/2022]
|
129
|
Preoperative cancer cachexia and short-term outcomes following surgery. J Surg Res 2016; 205:398-406. [DOI: 10.1016/j.jss.2016.06.076] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 05/11/2016] [Accepted: 06/27/2016] [Indexed: 11/21/2022]
|
130
|
Drutskaya MS, Efimov GA, Astrakhantseva IV, Kruglov AA, Nedospasov SA. Making anti-cytokine therapy more selective: Studies in mice. Cytokine 2016; 101:33-38. [PMID: 27624866 DOI: 10.1016/j.cyto.2016.08.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 08/21/2016] [Accepted: 08/23/2016] [Indexed: 12/13/2022]
Abstract
Cytokines are involved in a wide range of functions shaping the normal immune response, yet inflammatory changes in the immune system due to dysregulated cytokine signaling may lead to the induction of autoimmunity. Cytokine inhibitors have revolutionized the treatment of many autoimmune diseases in recent years. Systemic cytokine ablation, however, is often associated with the development of adverse side effects and some patients simply do not respond to therapy. TNF, IL-1 and IL-6 are the best characterized proinflammatory cytokines considered as the main therapeutic targets for the treatment of several autoimmune and inflammatory diseases. But can anti-cytokine therapy become more selective and thus more efficient? This mini-review discusses several recently emerging paradigms and summarizes current experimental attempts to validate them in mouse studies.
Collapse
Affiliation(s)
- Marina S Drutskaya
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia
| | - Grigory A Efimov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia; National Scientific Center for Hematology, Moscow 125167, Russia; Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603950, Russia
| | | | - Andrei A Kruglov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia; Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603950, Russia; Lomonosov Moscow State University, Moscow 199991, Russia; German Rheumatism Research Center (DRFZ), Berlin 10117, Germany
| | - Sergei A Nedospasov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia; Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603950, Russia; Lomonosov Moscow State University, Moscow 199991, Russia; German Rheumatism Research Center (DRFZ), Berlin 10117, Germany.
| |
Collapse
|
131
|
Cassel JB, Del Fabbro E, Arkenau T, Higginson IJ, Hurst S, Jansen LA, Poklepovic A, Rid A, Rodón J, Strasser F, Miller FG. Phase I Cancer Trials and Palliative Care: Antagonism, Irrelevance, or Synergy? J Pain Symptom Manage 2016; 52:437-45. [PMID: 27233136 DOI: 10.1016/j.jpainsymman.2016.02.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 02/06/2016] [Accepted: 02/26/2016] [Indexed: 12/19/2022]
Abstract
This article synthesizes the presentations and conclusions of an international symposium on Phase 1 oncology trials, palliative care, and ethics held in 2014. The purpose of the symposium was to discuss the intersection of three independent trends that unfolded in the past decade. First, large-scale reviews of hundreds of Phase I trials have indicated there is a relatively low risk of serious harm and some prospect of clinical benefit that can be meaningful to patients. Second, changes in the design and analysis of Phase I trials, the introduction of "targeted" investigational agents that are generally less toxic, and an increase in Phase I trials that combine two or more agents in a novel way have changed the conduct of these trials and decreased fears and apprehensions about participation. Third, the field of palliative care in cancer has expanded greatly, offering symptom management to late-stage cancer patients, and demonstrated that it is not mutually exclusive with disease-targeted therapies or clinical research. Opportunities for collaboration and further research at the intersection of Phase 1 oncology trials and palliative care are highlighted.
Collapse
Affiliation(s)
- J Brian Cassel
- Virginia Commonwealth University, Richmond, Virginia, USA.
| | | | - Tobias Arkenau
- Sarah Cannon Research Institute and University College London, London, United Kingdom
| | - Irene J Higginson
- Cicely Saunders Institute, King's College London, London, United Kingdom
| | - Samia Hurst
- Institut d'éthique biomedicale, Centre médical universitaire, Geneva, Switzerland
| | - Lynn A Jansen
- Oregon Health and Science University, Portland, Oregon, USA
| | | | - Annette Rid
- King's College London, London, United Kingdom
| | - Jordi Rodón
- Vall d'Hebron Institut d'Oncologia, Barcelona, Spain
| | | | | |
Collapse
|
132
|
HMGB1, IL-1α, IL-33 and S100 proteins: dual-function alarmins. Cell Mol Immunol 2016; 14:43-64. [PMID: 27569562 PMCID: PMC5214941 DOI: 10.1038/cmi.2016.34] [Citation(s) in RCA: 324] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 05/16/2016] [Accepted: 05/17/2016] [Indexed: 02/08/2023] Open
Abstract
Our immune system is based on the close collaboration of the innate and adaptive immune systems for the rapid detection of any threats to the host. Recognition of pathogen-derived molecules is entrusted to specific germline-encoded signaling receptors. The same receptors have now also emerged as efficient detectors of misplaced or altered self-molecules that signal tissue damage and cell death following, for example, disruption of the blood supply and subsequent hypoxia. Many types of endogenous molecules have been shown to provoke such sterile inflammatory states when released from dying cells. However, a group of proteins referred to as alarmins have both intracellular and extracellular functions which have been the subject of intense research. Indeed, alarmins can either exert beneficial cell housekeeping functions, leading to tissue repair, or provoke deleterious uncontrolled inflammation. This group of proteins includes the high-mobility group box 1 protein (HMGB1), interleukin (IL)-1α, IL-33 and the Ca2+-binding S100 proteins. These dual-function proteins share conserved regulatory mechanisms, such as secretory routes, post-translational modifications and enzymatic processing, that govern their extracellular functions in time and space. Release of alarmins from mesenchymal cells is a highly relevant mechanism by which immune cells can be alerted of tissue damage, and alarmins play a key role in the development of acute or chronic inflammatory diseases and in cancer development.
Collapse
|
133
|
Di Paolo NC, Shayakhmetov DM. Interleukin 1α and the inflammatory process. Nat Immunol 2016; 17:906-13. [PMID: 27434011 PMCID: PMC5152572 DOI: 10.1038/ni.3503] [Citation(s) in RCA: 377] [Impact Index Per Article: 47.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 05/31/2016] [Indexed: 02/08/2023]
Abstract
Inflammation occurs after disruption of tissue homeostasis by cell stress, injury or infection and ultimately involves the recruitment and retention of cells of hematopoietic origin, which arrive at the affected sites to resolve damage and initiate repair. Interleukin 1α (IL-1α) and IL-1β are equally potent inflammatory cytokines that activate the inflammatory process, and their deregulated signaling causes devastating diseases manifested by severe acute or chronic inflammation. Although much attention has been given to understanding the biogenesis of IL-1β, the biogenesis of IL-1α and its distinctive role in the inflammatory process remain poorly defined. In this review we examine key aspects of IL-1α biology and regulation and discuss its emerging importance in the initiation and maintenance of inflammation that underlie the pathology of many human diseases.
Collapse
Affiliation(s)
- Nelson C Di Paolo
- Lowance Center for Human Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Dmitry M Shayakhmetov
- Lowance Center for Human Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
- Center for Transplantation and Immune-mediated Disorders, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
- Emory Vaccine Center, Departments of Pediatrics and Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
134
|
Abstract
Metabolic dysfunction contributes to the clinical deterioration observed in advanced cancer patients and is characterized by weight loss, skeletal muscle wasting, and atrophy of the adipose tissue. This systemic syndrome, termed cancer-associated cachexia (CAC), is a major cause of morbidity and mortality. While once attributed solely to decreased food intake, the present description of cancer cachexia is a disorder of multiorgan energy imbalance. Here we review the molecules and pathways responsible for metabolic dysfunction in CAC and the ideas that led to the current understanding.
Collapse
Affiliation(s)
- Michele Petruzzelli
- Department of Oncology, The Medical Research Council Cancer Unit, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 0QQ, United Kingdon
| | - Erwin F Wagner
- Genes, Development, and Disease Group, Cancer Cell Biology Programme, Centro Nacional de Investigaciones Oncológicas, Madrid 28029, Spain
| |
Collapse
|
135
|
George GC, Iwuanyanwu EC, Anderson KO, Yusuf A, Zinner RG, Piha-Paul SA, Tsimberidou AM, Naing A, Fu S, Janku F, Subbiah V, Cleeland CS, Mendoza TR, Hong DS. Sleep quality and its association with fatigue, symptom burden, and mood in patients with advanced cancer in a clinic for early-phase oncology clinical trials. Cancer 2016; 122:3401-3409. [PMID: 27412379 DOI: 10.1002/cncr.30182] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 04/21/2016] [Accepted: 05/16/2016] [Indexed: 11/12/2022]
Abstract
BACKGROUND Limited data exist about sleep quality for patients with advanced cancer in phase 1 clinical trials. Poor sleep quality is often not captured as an adverse event, and its association with fatigue, one of the most frequently reported adverse events, is not documented routinely. This article describes sleep quality and its relation with fatigue, symptom burden, and mood in patients recruited from an early-phase clinic for targeted therapy. METHODS Sleep, fatigue, symptom burden, and mood were assessed with the Pittsburgh Sleep Quality Index (PSQI), the Brief Fatigue Inventory, the MD Anderson Symptom Inventory (MDASI), and the Brief Profile of Mood States, respectively; the Eastern Cooperative Oncology Group (ECOG) performance status (PS) was determined from medical records. RESULTS The sample (n = 256) was 51.2% female, 90% had an ECOG PS of 0 or 1, and the mean age was 58 ± 0.8 years. Poor sleepers (global PSQI score > 5) constituted 64% of the sample. In separate multiple regression models, poor sleepers had higher levels of fatigue (P < .001), symptom burden (P < .001), and overall mood disturbance (P < .001) than good sleepers. Also, compared with good sleepers, poor sleepers had greater fatigue-related and symptom-related interference with daily activities (all P values < .001). The MDASI disturbed-sleep item correlated well with the global PSQI score (Pearson's r = 0.679, P < .001), and this suggests its usefulness as a patient-reported outcome screener of sleep quality in early-phase clinical trials clinics. CONCLUSIONS Poor sleep quality was a significant problem in the current study and was associated with greater fatigue, symptom burden, and mood disturbance. Sleep quality should be routinely assessed in patients with advanced cancer who are participating in early-phase clinical trials. Cancer 2016;122:3401-3409. © 2016 American Cancer Society.
Collapse
Affiliation(s)
- Goldy C George
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas. .,Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Eucharia C Iwuanyanwu
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Karen O Anderson
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alizeh Yusuf
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ralph G Zinner
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sarina A Piha-Paul
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Apostolia M Tsimberidou
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Aung Naing
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Siqing Fu
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Filip Janku
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Vivek Subbiah
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Charles S Cleeland
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Tito R Mendoza
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David S Hong
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
136
|
From Inflammation to Prostate Cancer: The Role of Inflammasomes. Adv Urol 2016; 2016:3140372. [PMID: 27429614 PMCID: PMC4939357 DOI: 10.1155/2016/3140372] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 05/17/2016] [Accepted: 06/09/2016] [Indexed: 12/22/2022] Open
Abstract
Inflammation-associated studies entice specific attention due to inflammation's role in multiple stages of prostate cancer development. However, mechanistic regulation of inflammation inciting prostate cancer remains largely uncharacterized. A focused class of inflammatory regulators known as inflammasomes has recently gained attention in cancer development. Inflammasomes are a multiprotein complex that drives a cascade of proinflammatory cytokines regulating various cellular activities. Inflammasomes activation is linked with infection, stress, or danger signals, which are common events within the prostate gland. In this study, we review the potential of inflammasomes in understanding the role of inflammation in prostate cancer.
Collapse
|
137
|
Miyamoto Y, Hanna DL, Zhang W, Baba H, Lenz HJ. Molecular Pathways: Cachexia Signaling-A Targeted Approach to Cancer Treatment. Clin Cancer Res 2016; 22:3999-4004. [PMID: 27340276 DOI: 10.1158/1078-0432.ccr-16-0495] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 06/09/2016] [Indexed: 12/18/2022]
Abstract
Cancer cachexia is a multifactorial syndrome characterized by an ongoing loss of skeletal muscle mass, which negatively affects quality of life and portends a poor prognosis. Numerous molecular substrates and mechanisms underlie the dysregulation of skeletal muscle synthesis and degradation observed in cancer cachexia, including proinflammatory cytokines (TNFα, IL1, and IL6), and the NF-κB, IGF1/AKT/mTOR, and myostatin/activin-SMAD pathways. Recent preclinical and clinical studies have demonstrated that anti-cachexia drugs (such as MABp1 and soluble receptor antagonist of myostatin/activin) not only prevent muscle wasting but also may prolong overall survival. In this review, we focus on the significance of cachexia signaling in patients with cancer and highlight promising drugs targeting tumor cachexia in clinical development. Clin Cancer Res; 22(16); 3999-4004. ©2016 AACR.
Collapse
Affiliation(s)
- Yuji Miyamoto
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Diana L Hanna
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Wu Zhang
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Heinz-Josef Lenz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California.
| |
Collapse
|
138
|
Wiedemann GM, Knott MML, Vetter VK, Rapp M, Haubner S, Fesseler J, Kühnemuth B, Layritz P, Thaler R, Kruger S, Ormanns S, Mayr D, Endres S, Anz D. Cancer cell-derived IL-1α induces CCL22 and the recruitment of regulatory T cells. Oncoimmunology 2016; 5:e1175794. [PMID: 27757295 DOI: 10.1080/2162402x.2016.1175794] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 03/29/2016] [Accepted: 04/04/2016] [Indexed: 01/04/2023] Open
Abstract
In cancer patients, immunosuppression through regulatory T cells (Treg) is a crucial component of tumor immune evasion and contributes to disease progression. Tumor-infiltrating Treg in particular suppress local effector T cell responses and are associated with poor prognosis in tumors such as human pancreatic cancer or hepatocellular carcinoma (HCC). The chemokine CCL22 is known to recruit Treg into the tumor tissue and many types of human tumors are known to express high levels of CCL22. The mechanisms leading to intratumoral secretion of CCL22 are so far unknown. We demonstrate here that intratumoral CCL22 is induced in tumor-infiltrating immune cells through cancer cell-derived interleukin-1 (IL-1α). In pancreatic cancer and HCC, CCL22 is produced by intratumoral dendritic cells, while the cancer cells themselves do not secrete CCL22 in vitro and in vivo. Incubation of human peripheral blood mononuclear cells (PBMC) or murine splenocytes with tumor cells or tumor cell supernatants strongly induced CCL22 secretion in vitro. Tumor cell supernatants contained IL-1 and CCL22 induction in PBMC could be specifically prevented by the IL-1 receptor antagonist anakinra or by transfection of tumor cell lines with IL-1 siRNA, leading to a suppression of Treg migration. In conclusion, we identify here tumor cell-derived IL-1α as a major inducer of the Treg attracting chemokine CCL22 in human cancer cells. Therapeutic blockade of the IL-1 pathway could represent a promising strategy to inhibit tumor-induced immunosuppression.
Collapse
Affiliation(s)
- Gabriela Maria Wiedemann
- Center of Integrated Protein Science Munich (CIPS-M), Division of Clinical Pharmacology, Klinikum der Universität München , Munich, Germany
| | - Max Martin Ludwig Knott
- Center of Integrated Protein Science Munich (CIPS-M), Division of Clinical Pharmacology, Klinikum der Universität München , Munich, Germany
| | - Viola Katharina Vetter
- Center of Integrated Protein Science Munich (CIPS-M), Division of Clinical Pharmacology, Klinikum der Universität München , Munich, Germany
| | - Moritz Rapp
- Center of Integrated Protein Science Munich (CIPS-M), Division of Clinical Pharmacology, Klinikum der Universität München , Munich, Germany
| | - Sascha Haubner
- Center of Integrated Protein Science Munich (CIPS-M), Division of Clinical Pharmacology, Klinikum der Universität München , Munich, Germany
| | - Julia Fesseler
- Center of Integrated Protein Science Munich (CIPS-M), Division of Clinical Pharmacology, Klinikum der Universität München , Munich, Germany
| | - Benjamin Kühnemuth
- Center of Integrated Protein Science Munich (CIPS-M), Division of Clinical Pharmacology, Klinikum der Universität München , Munich, Germany
| | - Patrick Layritz
- Center of Integrated Protein Science Munich (CIPS-M), Division of Clinical Pharmacology, Klinikum der Universität München , Munich, Germany
| | - Raffael Thaler
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München , Munich, Germany
| | - Stephan Kruger
- Medizinische Klinik und Poliklinik III, Klinikum der Universität München , Munich, Germany
| | - Steffen Ormanns
- Department of Pathology, Ludwig-Maximilians-Universität , Munich, Germany
| | - Doris Mayr
- Department of Pathology, Ludwig-Maximilians-Universität , Munich, Germany
| | - Stefan Endres
- Center of Integrated Protein Science Munich (CIPS-M), Division of Clinical Pharmacology, Klinikum der Universität München , Munich, Germany
| | - David Anz
- Center of Integrated Protein Science Munich (CIPS-M), Division of Clinical Pharmacology, Klinikum der Universität München, Munich, Germany; Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| |
Collapse
|
139
|
von Haehling S, Springer J. Treatment of Muscle Wasting: An Overview of Promising Treatment Targets. J Am Med Dir Assoc 2015; 16:1014-9. [DOI: 10.1016/j.jamda.2015.10.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 10/02/2015] [Indexed: 12/27/2022]
|
140
|
Abstract
Tumour-promoting inflammation is considered one of the enabling characteristics of cancer development. Chronic inflammatory disease increases the risk of some cancers, and strong epidemiological evidence exists that NSAIDs, particularly aspirin, are powerful chemopreventive agents. Tumour microenvironments contain many different inflammatory cells and mediators; targeting these factors in genetic, transplantable and inducible murine models of cancer substantially reduces the development, growth and spread of disease. Thus, this complex network of inflammation offers targets for prevention and treatment of malignant disease. Much potential exists in this area for novel cancer prevention and treatment strategies, although clinical research to support targeting of cancer-related inflammation and innate immunity in patients with advanced-stage cancer remains in its infancy. Following the initial successes of immunotherapies that modulate the adaptive immune system, we assert that inflammation and innate immunity are important targets in patients with cancer on the basis of extensive preclinical and epidemiological data. The adaptive immune response is heavily dependent on innate immunity, therefore, inhibiting some of the tumour-promoting immunosuppressive actions of the innate immune system might enhance the potential of immunotherapies that activate a nascent antitumour response.
Collapse
Affiliation(s)
- Shanthini M Crusz
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | - Frances R Balkwill
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| |
Collapse
|
141
|
Ballak DB, Stienstra R, Tack CJ, Dinarello CA, van Diepen JA. IL-1 family members in the pathogenesis and treatment of metabolic disease: Focus on adipose tissue inflammation and insulin resistance. Cytokine 2015; 75:280-90. [PMID: 26194067 PMCID: PMC4553099 DOI: 10.1016/j.cyto.2015.05.005] [Citation(s) in RCA: 180] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Revised: 05/07/2015] [Accepted: 05/07/2015] [Indexed: 02/06/2023]
Abstract
Obesity is characterized by a chronic, low-grade inflammation that contributes to the development of insulin resistance and type 2 diabetes. Cytokines and chemokines produced by immunocompetent cells influence local as well as systemic inflammation and are therefore critical contributors to the pathogenesis of type 2 diabetes. Hence, cytokines that modulate inflammatory responses are emerging as potential targets for intervention and treatment of the metabolic consequences of obesity. The interleukin-1 (IL-1) family of cytokines and receptors are key mediators of innate inflammatory responses and exhibit both pro- and anti-inflammatory functions. During the last decades, mechanistic insights into how the IL-1 family affects the initiation and progression of obesity-induced insulin resistance have increased significantly. Here, we review the current knowledge and understanding, with emphasis on the therapeutic potential of individual members of the IL-1 family of cytokines for improving insulin sensitivity in patients with diabetes.
Collapse
Affiliation(s)
- Dov B Ballak
- Department of Medicine, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA; Department of Medicine, University of Colorado Denver, Aurora, CO, USA.
| | - Rinke Stienstra
- Department of Medicine, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Human Nutrition, Wageningen University, Wageningen, The Netherlands
| | - Cees J Tack
- Department of Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Charles A Dinarello
- Department of Medicine, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Medicine, University of Colorado Denver, Aurora, CO, USA
| | - Janna A van Diepen
- Department of Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
142
|
West NR, McCuaig S, Franchini F, Powrie F. Emerging cytokine networks in colorectal cancer. Nat Rev Immunol 2015; 15:615-29. [PMID: 26358393 DOI: 10.1038/nri3896] [Citation(s) in RCA: 266] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cytokine networks are crucial aspects of tumour immunology, particularly for colorectal cancer (CRC), in which inflammation and antitumour immunity are key determinants of disease progression. In this Review, we highlight new insights into the functions of well-known cytokines in CRC, describe recently discovered roles for a growing number of novel players, and emphasize the complexity and therapeutic implications of the cytokine milieu. We also discuss how cancer mutations and epigenetic adaptations influence the oncogenic potential of cytokines, a relatively unexplored area that could yield crucial insights into tumour immunology and facilitate the effective application of cytokine-modulatory therapies for CRC.
Collapse
Affiliation(s)
- Nathan R West
- Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7LF, UK.,Translational Gastroenterology Unit, Nuffield Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Sarah McCuaig
- Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7LF, UK.,Translational Gastroenterology Unit, Nuffield Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Fanny Franchini
- Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7LF, UK.,Translational Gastroenterology Unit, Nuffield Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Fiona Powrie
- Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7LF, UK.,Translational Gastroenterology Unit, Nuffield Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| |
Collapse
|
143
|
Madeddu C, Mantovani G, Gramignano G, Macciò A. Advances in pharmacologic strategies for cancer cachexia. Expert Opin Pharmacother 2015; 16:2163-77. [PMID: 26330024 DOI: 10.1517/14656566.2015.1079621] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Cancer cachexia is a severe inflammatory metabolic syndrome accounting for fatigue, an impairment of normal activities and, eventually, death. The loss of muscle mass associated with body weight loss is the main feature of this syndrome. AREAS COVERED The present review aims to describe the advances in the pharmacological approaches for cancer cachexia, highlighting the impact on weight loss, muscle wasting and related outcomes. EXPERT OPINION Among the pharmacological agents, attention should yet be given to the currently most widely studied drugs, such as progestogens and NSAIDs. Emerging drugs, such as ghrelin and selective androgen receptor modulators, have obtained promising results in recent randomized clinical trials. Larger sample sizes and more robust data on the effectiveness of anti-cytokine agents are needed. Any pharmacological approach to counteract cancer cachexia should always be associated with an adequate caloric intake, obtained by diet or through enteral or parenteral supplementation, if indicated. Finally, we can currently state that a combined approach that simultaneously targets the fundamental pathways involved in the pathogenesis of cancer cachexia is likely to be the most effective in terms of improvements in body weight as well as muscle wasting, function, physical performance and quality of life.
Collapse
Affiliation(s)
- Clelia Madeddu
- a 1 University of Cagliari, AOU Cagliari, Department of Medical Sciences M. Aresu , Cagliari, Italy
| | - Giovanni Mantovani
- b 2 University of Cagliari, AOU Cagliari, Department of Medical Sciences M. Aresu , Via Catalani 1b, 09100 Cagliari, Italy
| | - Giulia Gramignano
- c 3 N.S. Bonaria Hospital, Medical Oncology Unit , San Gavino, Italy
| | - Antonio Macciò
- d 4 A. Businco Hospital, Regional Referral Center for Cancer Diseases, Department of Gynecologic Oncology , Cagliari, Italy
| |
Collapse
|
144
|
Timper K, Seelig E, Tsakiris DA, Donath MY. Safety, pharmacokinetics, and preliminary efficacy of a specific anti-IL-1alpha therapeutic antibody (MABp1) in patients with type 2 diabetes mellitus. J Diabetes Complications 2015; 29:955-60. [PMID: 26139558 DOI: 10.1016/j.jdiacomp.2015.05.019] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 05/24/2015] [Accepted: 05/26/2015] [Indexed: 10/23/2022]
Abstract
AIMS The role of the IL-1 system in development of type 2 diabetes is well established. Using an IL-1 receptor antagonist, which blocks IL-1alpha and -beta activity, or by specifically neutralizing IL-1beta, several clinical studies have demonstrated improvement in insulin secretion and glycaemia. However, the role of IL-1alpha remains to be investigated. METHODS We evaluated the safety and preliminary efficacy of a neutralizing true human™ monoclonal antibody against IL-1alpha (MABp1) in an open label trial in patients with type 2 diabetes. Seven patients between 50 to 66years with type 2 diabetes mellitus were enrolled in the study. The study subjects received four biweekly intravenous infusions of MABp1 at 1.25mg/kg body weight up to day 60 and were followed up for a total of 90days. RESULTS Compared to baseline, after the 60-day period of treatment HbA1c was numerically reduced by 0.14±0.21% (p=0.15), fasting C-peptide was increased by 88% (p=0.03), pro-insulin by 48% (p=0.03) and insulin numerically increased by 74% (p=0.11). Systolic blood pressure numerically decreased by 11mmHg (p=0.2). Both HbA1c and blood pressure rebounded to baseline levels thirty days after the end of MABp1 application. Treatment with MABp1 was well tolerated, and no adverse events occurred during the study. CONCLUSION The results point to a role of IL-1alpha in type 2 diabetes and encourage further investigations. (ClinicalTrials.gov number NCT01427699).
Collapse
Affiliation(s)
- Katharina Timper
- University Hospital of Basel, Department of Endocrinology, Diabetes, and Metabolism, Basel, Switzerland.
| | - Eleonora Seelig
- University Hospital of Basel, Department of Endocrinology, Diabetes, and Metabolism, Basel, Switzerland
| | - Dimitrios A Tsakiris
- University Hospital of Basel, Department of Diagnostic Hematology, Basel, Switzerland
| | - Marc Y Donath
- University Hospital of Basel, Department of Endocrinology, Diabetes, and Metabolism, Basel, Switzerland
| |
Collapse
|
145
|
Maris P, Blomme A, Palacios AP, Costanza B, Bellahcène A, Bianchi E, Gofflot S, Drion P, Trombino GE, Di Valentin E, Cusumano PG, Maweja S, Jerusalem G, Delvenne P, Lifrange E, Castronovo V, Turtoi A. Asporin Is a Fibroblast-Derived TGF-β1 Inhibitor and a Tumor Suppressor Associated with Good Prognosis in Breast Cancer. PLoS Med 2015; 12:e1001871. [PMID: 26327350 PMCID: PMC4556693 DOI: 10.1371/journal.pmed.1001871] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 07/21/2015] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Breast cancer is a leading malignancy affecting the female population worldwide. Most morbidity is caused by metastases that remain incurable to date. TGF-β1 has been identified as a key driving force behind metastatic breast cancer, with promising therapeutic implications. METHODS AND FINDINGS Employing immunohistochemistry (IHC) analysis, we report, to our knowledge for the first time, that asporin is overexpressed in the stroma of most human breast cancers and is not expressed in normal breast tissue. In vitro, asporin is secreted by breast fibroblasts upon exposure to conditioned medium from some but not all human breast cancer cells. While hormone receptor (HR) positive cells cause strong asporin expression, triple-negative breast cancer (TNBC) cells suppress it. Further, our findings show that soluble IL-1β, secreted by TNBC cells, is responsible for inhibiting asporin in normal and cancer-associated fibroblasts. Using recombinant protein, as well as a synthetic peptide fragment, we demonstrate the ability of asporin to inhibit TGF-β1-mediated SMAD2 phosphorylation, epithelial to mesenchymal transition, and stemness in breast cancer cells. In two in vivo murine models of TNBC, we observed that tumors expressing asporin exhibit significantly reduced growth (2-fold; p = 0.01) and metastatic properties (3-fold; p = 0.045). A retrospective IHC study performed on human breast carcinoma (n = 180) demonstrates that asporin expression is lowest in TNBC and HER2+ tumors, while HR+ tumors have significantly higher asporin expression (4-fold; p = 0.001). Assessment of asporin expression and patient outcome (n = 60; 10-y follow-up) shows that low protein levels in the primary breast lesion significantly delineate patients with bad outcome regardless of the tumor HR status (area under the curve = 0.87; 95% CI 0.78-0.96; p = 0.0001). Survival analysis, based on gene expression (n = 375; 25-y follow-up), confirmed that low asporin levels are associated with a reduced likelihood of survival (hazard ratio = 0.58; 95% CI 0.37-0.91; p = 0.017). Although these data highlight the potential of asporin to serve as a prognostic marker, confirmation of the clinical value would require a prospective study on a much larger patient cohort. CONCLUSIONS Our data show that asporin is a stroma-derived inhibitor of TGF-β1 and a tumor suppressor in breast cancer. High asporin expression is significantly associated with less aggressive tumors, stratifying patients according to the clinical outcome. Future pre-clinical studies should consider options for increasing asporin expression in TNBC as a promising strategy for targeted therapy.
Collapse
Affiliation(s)
- Pamela Maris
- Metastasis Research Laboratory, GIGA–Cancer, University of Liège, Liège, Belgium
| | - Arnaud Blomme
- Metastasis Research Laboratory, GIGA–Cancer, University of Liège, Liège, Belgium
| | - Ana Perez Palacios
- Metastasis Research Laboratory, GIGA–Cancer, University of Liège, Liège, Belgium
| | - Brunella Costanza
- Metastasis Research Laboratory, GIGA–Cancer, University of Liège, Liège, Belgium
| | - Akeila Bellahcène
- Metastasis Research Laboratory, GIGA–Cancer, University of Liège, Liège, Belgium
| | - Elettra Bianchi
- Department of Pathology, University Hospital Liège, University of Liège, Liège, Belgium
| | | | - Pierre Drion
- Animal Facility, GIGA–Cardiovascular Sciences, University of Liège, Liège, Belgium
| | - Giovanna Elvi Trombino
- Metastasis Research Laboratory, GIGA–Cancer, University of Liège, Liège, Belgium
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Cosenza, Italy
| | | | - Pino G. Cusumano
- Department of Senology, University Hospital Liège, University of Liège, Liège, Belgium
| | - Sylvie Maweja
- Department of Abdominal Surgery, University of Liège, Liège, Belgium
| | - Guy Jerusalem
- Department of Medical Oncology, University Hospital Liège, University of Liège, Liège, Belgium
| | - Philippe Delvenne
- Department of Pathology, University Hospital Liège, University of Liège, Liège, Belgium
| | - Eric Lifrange
- Department of Senology, University Hospital Liège, University of Liège, Liège, Belgium
| | - Vincent Castronovo
- Metastasis Research Laboratory, GIGA–Cancer, University of Liège, Liège, Belgium
- * E-mail: (VC); (AT)
| | - Andrei Turtoi
- Metastasis Research Laboratory, GIGA–Cancer, University of Liège, Liège, Belgium
- * E-mail: (VC); (AT)
| |
Collapse
|
146
|
Rider P, Carmi Y, Yossef R, Guttman O, Eini H, Azam T, Dinarello CA, Lewis EC. IL-1 Receptor Antagonist Chimeric Protein: Context-Specific and Inflammation-Restricted Activation. THE JOURNAL OF IMMUNOLOGY 2015; 195:1705-12. [PMID: 26157171 DOI: 10.4049/jimmunol.1501168] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 06/15/2015] [Indexed: 12/20/2022]
Abstract
Both IL-1α and IL-1β are highly inflammatory cytokines mediating a wide spectrum of diseases. A recombinant form of the naturally occurring IL-1R antagonist (IL-1Ra), which blocks IL-1R1, is broadly used to treat autoimmune and autoinflammatory diseases; however, blocking IL-1 increases the risk of infection. In this study, we describe the development of a novel form of recombinant IL-1Ra, termed chimeric IL-1Ra. This molecule is a fusion of the N-terminal peptide of IL-1β and IL-1Ra, resulting in inactive IL-1Ra. Because the IL-1β N-terminal peptide contains several protease sites clustered around the caspase-1 site, local proteases at sites of inflammation can cleave chimeric IL-1Ra and turn IL-1Ra active. We demonstrate that chimeric IL-1Ra reduces IL-1-mediated inflammation in vitro and in vivo. This unique approach limits IL-1 receptor blockade to sites of inflammation, while sparing a multitude of desired IL-1-related activities, including host defense against infections and IL-1-mediated repair.
Collapse
Affiliation(s)
- Peleg Rider
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel;
| | - Yaron Carmi
- Department of Pathology, School of Medicine, Stanford University, Palo Alto, CA 95305; The Shraga Segal Department of Microbiology, Immunology and Genetics and The Cancer Research Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Rami Yossef
- The Shraga Segal Department of Microbiology, Immunology and Genetics and The Cancer Research Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Ofer Guttman
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Hadar Eini
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Tania Azam
- University of Colorado at Denver, Aurora, CO 80045; and
| | - Charles A Dinarello
- University of Colorado at Denver, Aurora, CO 80045; and Radboud University Medical Centre, Nijmegen 6500, the Netherlands
| | - Eli C Lewis
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| |
Collapse
|
147
|
Hui D, Finlay E, Buss MK, Prommer EE, Bruera E. Palliative Oncologists: Specialists in the Science and Art of Patient Care. J Clin Oncol 2015; 33:2314-7. [PMID: 25918292 DOI: 10.1200/jco.2014.60.3274] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- David Hui
- David Hui and Eduardo Bruera, The University of Texas MD Anderson Cancer Center, Houston, TX; Esmé Finlay, University of New Mexico Cancer Center, Albuquerque, NM; Mary K. Buss, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; and Eric E. Prommer, Mayo Clinic, Scottsdale, AZ.
| | - Esmé Finlay
- David Hui and Eduardo Bruera, The University of Texas MD Anderson Cancer Center, Houston, TX; Esmé Finlay, University of New Mexico Cancer Center, Albuquerque, NM; Mary K. Buss, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; and Eric E. Prommer, Mayo Clinic, Scottsdale, AZ
| | - Mary K Buss
- David Hui and Eduardo Bruera, The University of Texas MD Anderson Cancer Center, Houston, TX; Esmé Finlay, University of New Mexico Cancer Center, Albuquerque, NM; Mary K. Buss, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; and Eric E. Prommer, Mayo Clinic, Scottsdale, AZ
| | - Eric E Prommer
- David Hui and Eduardo Bruera, The University of Texas MD Anderson Cancer Center, Houston, TX; Esmé Finlay, University of New Mexico Cancer Center, Albuquerque, NM; Mary K. Buss, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; and Eric E. Prommer, Mayo Clinic, Scottsdale, AZ
| | - Eduardo Bruera
- David Hui and Eduardo Bruera, The University of Texas MD Anderson Cancer Center, Houston, TX; Esmé Finlay, University of New Mexico Cancer Center, Albuquerque, NM; Mary K. Buss, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA; and Eric E. Prommer, Mayo Clinic, Scottsdale, AZ
| |
Collapse
|
148
|
Dinarello CA. The history of fever, leukocytic pyrogen and interleukin-1. Temperature (Austin) 2015; 2:8-16. [PMID: 27226996 PMCID: PMC4843879 DOI: 10.1080/23328940.2015.1017086] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 02/05/2015] [Indexed: 02/07/2023] Open
Abstract
There has been great progress in the 30 y since the reporting in 1984 of the cDNA for interleukin1 (IL1) β in the human and IL1α in the mouse. However, the history of IL1 begins in the early 1940s with investigations into the nature of an endogenous fever-producing protein released rabbit peritoneal neutrophils. Most researchers in immunology today are unaware that the field of cytokines, particularly the field of inflammatory cytokines. Toll-like receptors and innate immunity traces back to studies on fever. Researchers in infectious diseases wanted to know about an endogenous protein that caused fever, independent of infection. The endogenous fever-producing protein was called by various names: granulocyte, endogenous or leukocytic pyrogen. It is a fascinating and sometimes controversial story for biology and medicine and for the treatment of inflammatory diseases. Few imagined that this fever-producing protein would play such a major role in nearly every cell and in most diseases. This paper reviews the true background and milestones of interleukin1 from the purification of leukocytic pyrogen to the first cDNA of IL1β and the validation of cytokine biology from ill-defined factors to its present day importance.
Collapse
Affiliation(s)
- Charles A Dinarello
- Department of Medicine; Division of Infectious Diseases; University of Colorado Denver; Aurora, CO USA; Department of Internal Medicine; Radboud University Medical Center; Nijmegen, The Netherlands
| |
Collapse
|
149
|
|
150
|
Xilonix, a novel true human antibody targeting the inflammatory cytokine interleukin-1 alpha, in non-small cell lung cancer. Invest New Drugs 2015; 33:621-31. [PMID: 25822109 DOI: 10.1007/s10637-015-0226-6] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 03/04/2015] [Indexed: 01/11/2023]
Abstract
BACKGROUND Advanced non-small cell lung cancer (NSCLC) patients were treated as part of a Phase I dose escalation and expansion study evaluating a true human monoclonal antibody targeting IL-1α (Xilonix), which is intended to modulate the malignant phenotype-inhibiting tumor growth, spread and offering relief of symptoms. METHODS Sixteen NSCLC patients were included. Patients failed a median of 4 chemotherapy regimens, including 10/16 failing anti-EGFR therapy. Disease progression was evaluated using a multi-modal approach: tumor response, patient reported outcomes (EORTC-QLQC30), and lean body mass (LBM). Patients received infusions every 2 or 3 weeks until progression, and were followed 24 months to assess survival. RESULTS There were no infusion reactions, dose-limiting toxicities, or deaths due to therapy. Albeit not statistically significant, there was a trend in IL-6 (-2.6 ± 18.5 (0.1 [-2.8-2.4]), platelet counts (-11 ± 54 (-4[-36.0-1.0]), CRP (-3.3 ± 30.2 (0.4 [-10.7-1.8]) and LBM (1.0 ± 2.5 (0.4 [-0.5-2.6]). Self-reported outcomes revealed reductions in pain, fatigue and improvement in appetite. Median survival was 7.6 (IQR 4.4-11.5) months, stratification based on prior anti-EGFR therapy revealed a median survival of 9.4 months (IQR 7.6-12.5) for those pretreated (N = 10) versus a survival of 4.8 months (IQR 4.3-5.7) for those without (N = 6, logrank p = 0.187). CONCLUSION Xilonix was well tolerated, with gains in LBM and improvement in symptoms suggesting a clinically important response. Although not statistically significant, the survival outcomes observed for patients with and without prior anti-EGFR therapy raises intriguing questions about the potential synergy of IL-1α blockade and anti-EGFR therapy. Further study for this agent in NSCLC is warranted.
Collapse
|