1
|
Chen T, Wang M, Chen Y, Cao Y, Liu Y. Advances in predictive biomarkers associated with immunotherapy in extensive-stage small cell lung cancer. Cell Biosci 2024; 14:117. [PMID: 39267195 PMCID: PMC11391723 DOI: 10.1186/s13578-024-01283-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 08/05/2024] [Indexed: 09/14/2024] Open
Abstract
Small cell lung cancer (SCLC) is a highly malignant and poor-prognosis cancer, with most cases diagnosed at the extensive stage (ES). Amidst a landscape marked by limited progress in treatment modalities for ES-SCLC over the past few decades, the integration of immune checkpoint inhibitors (ICIs) with platinum-based chemotherapy has provided a milestone approach for improving prognosis, emerging as the new standard for initial therapy in ES-SCLC. However, only a minority of SCLC patients can benefit from ICIs, which frequently come with varying degrees of immune-related adverse events (irAEs). Therefore, it is crucial to investigate predictive biomarkers to screen potential beneficiaries of ICIs, mitigate the risk of side effects, and improve treatment precision. This review summarized potential biomarkers for predicting ICI response in ES-SCLC, with a primary focus on markers sourced from tumor tissue or peripheral blood samples. The former mainly included PD-L1 expression, tumor mutational burden (TMB), along with cellular or molecular components related to the tumor microenvironment (TME) and antigen presentation machinery (APM), molecular subtypes of SCLC, and inflammatory gene expression profiles. Circulating biomarkers predominantly comprised circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), cytokines, plasma autoantibodies, inflammation-related parameters, and blood TMB. We synthesized and analyzed the research progress of these potential markers. Notably, investigations into PD-L1 expression and TMB have been the most extensive, exhibiting preliminary predictive efficacy in salvage immunotherapy; however, consistent conclusions have yet to be reached across studies. Additionally, novel predictive markers developed based on TME composition, APM, transcriptomic and genomic features provide promising tools for precision immunotherapy. Circulating biomarkers offer the advantages of convenience, non-invasiveness, and a comprehensive reflection of tumor molecular characteristics. They may serve as alternative options for predicting immunotherapy efficacy in SCLC. However, there is a scarcity of studies, and the significant heterogeneity in research findings warrants attention.
Collapse
Affiliation(s)
- Tong Chen
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Mingzhao Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Yanchao Chen
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Yang Cao
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Yutao Liu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China.
| |
Collapse
|
2
|
Zekeridou A. Paraneoplastic Neurologic Disorders. Continuum (Minneap Minn) 2024; 30:1021-1051. [PMID: 39088287 DOI: 10.1212/con.0000000000001449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/03/2024]
Abstract
OBJECTIVE This article reviews the clinical presentations, neural antibody associations, and oncologic accompaniments of paraneoplastic neurologic syndromes and neurologic autoimmunity in the context of immune checkpoint inhibitor (ICI) cancer immunotherapy. LATEST DEVELOPMENTS Neural antibody discovery has improved the diagnosis of paraneoplastic neurologic syndromes. Neural antibodies also delineate the underlying disease pathophysiology and thus inform outcomes and treatments. Neural antibodies specific for extracellular proteins have pathogenic potential, whereas antibodies specific for intracellular targets are biomarkers of a cytotoxic T-cell immune response. A recent update in paraneoplastic neurologic syndrome criteria suggests high- and intermediate-risk phenotypes as well as neural antibodies to improve diagnostic accuracy in patients with paraneoplastic neurologic syndromes; a score was created based on this categorization. The introduction of ICI cancer immunotherapy has led to an increase in cancer-related neurologic autoimmunity with distinct clinical phenotypes. ESSENTIAL POINTS Paraneoplastic neurologic syndromes reflect an ongoing immunologic response to cancer mediated by effector T cells or antibodies. Paraneoplastic neurologic syndromes can present with manifestations at any level of the neuraxis, and neural antibodies aid diagnosis, focus cancer screening, and inform prognosis and therapy. In patients with high clinical suspicion of a paraneoplastic neurologic syndrome, cancer screening and treatment should be undertaken, regardless of the presence of a neural antibody. ICI therapy has led to immune-mediated neurologic complications. Recognition and treatment lead to improved outcomes.
Collapse
|
3
|
Moakley DF, Zhang C. Oncogenic NOVA1 expression dysregulates alternative splicing in breast cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.08.602566. [PMID: 39026722 PMCID: PMC11257507 DOI: 10.1101/2024.07.08.602566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Neuro-Oncological Ventral Antigen 1 (NOVA1) is best known for its role in mediating an alternative splicing (AS) program in neurons, yet was first discovered as an antigen expressed in breast tumors, causing rare autoimmune reactions and paraneoplastic neurological disorders (PNDs). The PND model suggests a plausible role of the tumor antigen expression in tumor suppression, whereas it has emerged that NOVA may function as an oncogene in a variety of cancers. In addition, whether NOVA mediates AS in breast cancer remains unanswered. Here we examine the AS profiles of breast invasive carcinoma (BRCA) tumor samples and demonstrate that ectopic NOVA1 expression led to the activation of neuron-like splicing patterns in many genes, including exons targeted by NOVA in the brain. The splicing dysregulation is especially prevalent in cell periphery and cytoskeleton genes related to cell-cell communication, actin-based movement, and neuronal functions. We find that NOVA1-mediated AS is most prominent in Luminal A tumors and high NOVA1 expression in this subtype is associated with poorer prognosis. Our results suggest that ectopic NOVA1 in tumors has regulatory activity affecting pathways with high relevance to tumor progression and that this might be a more general mechanism for PND antigens.
Collapse
Affiliation(s)
- Daniel F Moakley
- Department of Systems Biology, Columbia University, New York, NY 10032, USA
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032, USA
| | - Chaolin Zhang
- Department of Systems Biology, Columbia University, New York, NY 10032, USA
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032, USA
| |
Collapse
|
4
|
Mikkelsen AW, Nilsson AC, Tenstad HB, Lillevang ST, Asgari N. Initial screening for neuronal autoantibodies and their putative impact on survival in patients with small-cell lung cancer. Thorac Cancer 2024; 15:1350-1356. [PMID: 38703039 PMCID: PMC11168912 DOI: 10.1111/1759-7714.15318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/11/2024] [Accepted: 04/16/2024] [Indexed: 05/06/2024] Open
Abstract
INTRODUCTION Small-cell lung cancer (SCLC) may be associated with neuronal autoantibodies and paraneoplastic neurological syndromes. It has been suggested that neuronal autoantibodies, especially antineuronal nuclear antibody type 1 (Hu) autoantibodies, are associated with longer survival of patients with SCLC. The objective of this study was to determine the frequency and distribution of neuronal autoantibodies at the time of diagnosis of SCLC patients and assess survival rates in relation to autoimmunity. METHODS In this retrospective study, serum from 40 patients with biopsy-proven SCLC at the time of diagnosis was studied prior to treatment. The sera originated from a cancer registry at the Oncology Department, Vejle Hospital from 2007 to 2010. The sera were analyzed blindly to clinical status for the presence of neuronal autoantibodies. Medical records were reviewed for neurological symptoms. RESULTS Neuronal autoantibodies were detected in 22/40 (55%) of the SCLC patients. A broad range of neurological symptoms was recorded in 28/40 (70%) patients, of which 14/28 (50%) were positive for neuronal autoantibodies. The most frequently detected autoantibodies were Hu (7/40, 17.5%) followed by GAD65 (6/22, 15.0%). Striational and P/Q- or N-type voltage-gated calcium channel antibodies were less common, with each found in five patients (12.5%). Eight patients (20%) had coexisting autoantibodies. Autoantibody-positivity was not associated with survival. CONCLUSION Neuronal autoantibodies were at time of diagnosis found in approximately half of the treatment-naïve SCLC patients. Neither autoantibody positivity at diagnosis nor neurological manifestations correlated with survival and their clinical importance requires further studies in larger, prospective cohorts.
Collapse
Affiliation(s)
| | - Anna Christine Nilsson
- Department of Clinical ImmunologyOdense University HospitalOdenseDenmark
- Department of Clinical ResearchUniversity of Southern DenmarkOdenseDenmark
| | - Helene Broch Tenstad
- Department of Clinical ImmunologyOdense University HospitalOdenseDenmark
- Department of RheumatologyOdense University HospitalOdenseDenmark
| | | | - Nasrin Asgari
- Department of NeurologySlagelse HospitalSlagelseDenmark
- Institute of Regional Health ResearchUniversity of Southern DenmarkOdenseDenmark
- Department of NeurobiologyInstitute of Molecular Medicine, University of Southern DenmarkOdenseDenmark
| |
Collapse
|
5
|
Mangioris G, Halfdanarson TR, Lennon VA, Chang BK, Dubey D, Dyck PJB, Flanagan EP, McKeon A, Mills JR, Pittock SJ, Zekeridou A. Neurological autoimmunity in patients with non-pulmonary neuroendocrine neoplasms: clinical manifestations and neural autoantibody profiles. Eur J Neurol 2024; 31:e16273. [PMID: 38466015 PMCID: PMC11235830 DOI: 10.1111/ene.16273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 02/16/2024] [Accepted: 02/21/2024] [Indexed: 03/12/2024]
Abstract
BACKGROUND AND PURPOSE Paraneoplastic neurological autoimmunity is well described with small-cell lung cancer, but information is limited for other neuroendocrine neoplasms (NENs). METHODS Adult patients with histopathologically confirmed non-pulmonary NENs, neurological autoimmunity within 5 years of NEN diagnosis, and neural antibody testing performed at the Mayo Clinic Neuroimmunology Laboratory (January 2008 to March 2023) were retrospectively identified. Control sera were available from patients with NENs without neurological autoimmunity (116). RESULTS Thirty-four patients were identified (median age 68 years, range 31-87). The most common primary tumor sites were pancreas (nine), skin (Merkel cell, eight), small bowel/duodenum (seven), and unknown (seven). Five patients received immune checkpoint inhibitor (ICI) therapy before symptom onset; symptoms preceded cancer diagnosis in 62.1% of non-ICI-treated patients. The most frequent neurological phenotypes (non-ICI-treated) were movement disorders (12; cerebellar ataxia in 10), dysautonomia (six), peripheral neuropathy (eight), encephalitis (four), and neuromuscular junction disorders (four). Neural antibodies were detected in 55.9% of patients studied (most common specificities: P/Q-type voltage-gated calcium channel [seven], muscle-type acetylcholine receptor [three], anti-neuronal nuclear antibody type 1 [three], and neuronal intermediate filaments [two]), but in only 6.9% of controls. Amongst patients receiving cancer or immunosuppressive therapy, 51.6% had partial or complete recovery. Outcomes were unfavorable in 48.3% (non-ICI-treated) and neural autoantibody positivity was associated with poor neurological outcome. DISCUSSION Neurological autoimmunity associated with non-pulmonary NENs is often multifocal and can be treatment responsive, underscoring the importance of rapid recognition and early treatment.
Collapse
Affiliation(s)
- Georgios Mangioris
- Department of Laboratory Medicine and PathologyMayo ClinicRochesterMinnesotaUSA
| | | | - Vanda A. Lennon
- Department of Laboratory Medicine and PathologyMayo ClinicRochesterMinnesotaUSA
- Department of NeurologyMayo ClinicRochesterMinnesotaUSA
- Department of ImmunologyMayo ClinicRochesterMinnesotaUSA
| | | | - Divyanshu Dubey
- Department of Laboratory Medicine and PathologyMayo ClinicRochesterMinnesotaUSA
- Department of NeurologyMayo ClinicRochesterMinnesotaUSA
- Center for Multiple Sclerosis and Autoimmune NeurologyMayo ClinicRochesterMinnesotaUSA
| | | | - Eoin P. Flanagan
- Department of Laboratory Medicine and PathologyMayo ClinicRochesterMinnesotaUSA
- Department of NeurologyMayo ClinicRochesterMinnesotaUSA
- Center for Multiple Sclerosis and Autoimmune NeurologyMayo ClinicRochesterMinnesotaUSA
| | - Andrew McKeon
- Department of Laboratory Medicine and PathologyMayo ClinicRochesterMinnesotaUSA
- Department of NeurologyMayo ClinicRochesterMinnesotaUSA
- Center for Multiple Sclerosis and Autoimmune NeurologyMayo ClinicRochesterMinnesotaUSA
| | - John R. Mills
- Department of Laboratory Medicine and PathologyMayo ClinicRochesterMinnesotaUSA
| | - Sean J. Pittock
- Department of Laboratory Medicine and PathologyMayo ClinicRochesterMinnesotaUSA
- Department of NeurologyMayo ClinicRochesterMinnesotaUSA
- Center for Multiple Sclerosis and Autoimmune NeurologyMayo ClinicRochesterMinnesotaUSA
| | - Anastasia Zekeridou
- Department of Laboratory Medicine and PathologyMayo ClinicRochesterMinnesotaUSA
- Department of NeurologyMayo ClinicRochesterMinnesotaUSA
- Center for Multiple Sclerosis and Autoimmune NeurologyMayo ClinicRochesterMinnesotaUSA
| |
Collapse
|
6
|
Solta A, Ernhofer B, Boettiger K, Megyesfalvi Z, Heeke S, Hoda MA, Lang C, Aigner C, Hirsch FR, Schelch K, Döme B. Small cells - big issues: biological implications and preclinical advancements in small cell lung cancer. Mol Cancer 2024; 23:41. [PMID: 38395864 PMCID: PMC10893629 DOI: 10.1186/s12943-024-01953-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
Current treatment guidelines refer to small cell lung cancer (SCLC), one of the deadliest human malignancies, as a homogeneous disease. Accordingly, SCLC therapy comprises chemoradiation with or without immunotherapy. Meanwhile, recent studies have made significant advances in subclassifying SCLC based on the elevated expression of the transcription factors ASCL1, NEUROD1, and POU2F3, as well as on certain inflammatory characteristics. The role of the transcription regulator YAP1 in defining a unique SCLC subset remains to be established. Although preclinical analyses have described numerous subtype-specific characteristics and vulnerabilities, the so far non-existing clinical subtype distinction may be a contributor to negative clinical trial outcomes. This comprehensive review aims to provide a framework for the development of novel personalized therapeutic approaches by compiling the most recent discoveries achieved by preclinical SCLC research. We highlight the challenges faced due to limited access to patient material as well as the advances accomplished by implementing state-of-the-art models and methodologies.
Collapse
Affiliation(s)
- Anna Solta
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Büsra Ernhofer
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Kristiina Boettiger
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Zsolt Megyesfalvi
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary
- National Koranyi Institute of Pulmonology, Budapest, Hungary
| | - Simon Heeke
- Department of Thoracic Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mir Alireza Hoda
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Christian Lang
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Division of Pulmonology, Department of Medicine II, Medical University of Vienna, Vienna, Austria
| | - Clemens Aigner
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Fred R Hirsch
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Center for Thoracic Oncology, Mount Sinai Health System, Tisch Cancer Institute, New York, NY, USA.
| | - Karin Schelch
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Balazs Döme
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary.
- National Koranyi Institute of Pulmonology, Budapest, Hungary.
- Department of Translational Medicine, Lund University, Lund, Sweden.
| |
Collapse
|
7
|
Guo H, Li W, Guo Y, Chen N, Cui J. Molecular classification of small cell lung cancer subtypes: Characteristics, prognostic factors, and clinical translation. Chin Med J (Engl) 2024; 137:130-139. [PMID: 37660289 PMCID: PMC10798698 DOI: 10.1097/cm9.0000000000002693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Indexed: 09/04/2023] Open
Abstract
ABSTRACT Small cell lung cancer (SCLC) is a highly malignant tumor with a very poor prognosis; therefore, more effective treatments are urgently needed for patients afflicted with the disease. In recent years, emerging molecular classifications based on key transcription factors of SCLC have provided more information on the tumor pathophysiology, metastasis, immune microenvironment, and acquired therapeutic resistance and reflected the intertumoral heterogeneity of the various SCLC phenotypes. Additionally, advances in genomics and single-cell sequencing analysis have further revealed the high intratumoral heterogeneity and plasticity of the disease. Herein, we review and summarize these recent lines of evidence and discuss the possible pathogenesis of SCLC.
Collapse
Affiliation(s)
| | | | | | | | - Jiuwei Cui
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| |
Collapse
|
8
|
Dalmau J. Changing landscape in the field of paraneoplastic neurology: Personal perspectives over a 35-year career. HANDBOOK OF CLINICAL NEUROLOGY 2024; 200:11-32. [PMID: 38494272 DOI: 10.1016/b978-0-12-823912-4.00013-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Paraneoplastic neurologic syndromes are a group of rare disorders that have fascinated neurologists for more than a century. The discovery in the 1980s that many of these disorders occurred in association with antibodies against neuronal proteins revived the interest for these diseases. This chapter first traces the history of the paraneoplastic neurologic syndromes during the era that preceded the discovery of immune mechanisms and then reviews the immunologic period during which many of these syndromes were found to be associated with antibodies against intracellular onconeuronal proteins and pathogenic cytotoxic T-cell mechanisms. Alongside these developments, investigations on the antibody-mediated disorders of the peripheral nervous system, such as the myasthenic syndromes or neuromyotonia, provided suggestions for the study of the central nervous system (CNS) syndromes. These converging areas of research culminated with the groundbreaking discovery of a new category of CNS disorders mediated by antibodies against neuronal surface proteins or receptors. These disorders are not always paraneoplastic, and the understanding of these syndromes and mechanisms has changed the landscape of neurology and neurosciences.
Collapse
Affiliation(s)
- Josep Dalmau
- IDIBAPS-Hospital Clinic, University of Barcelona, Barcelona, Spain; Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain.
| |
Collapse
|
9
|
Lastwika KJ, Kunihiro A, Solan JL, Zhang Y, Taverne LR, Shelley D, Rho JH, Randolph TW, Li CI, Grogan EL, Massion PP, Fitzpatrick AL, MacPherson D, Houghton AM, Lampe PD. Posttranslational modifications induce autoantibodies with risk prediction capability in patients with small cell lung cancer. Sci Transl Med 2023; 15:eadd8469. [PMID: 36630482 PMCID: PMC10117289 DOI: 10.1126/scitranslmed.add8469] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Small cell lung cancer (SCLC) elicits the generation of autoantibodies that result in unique paraneoplastic neurological syndromes. The mechanistic basis for the formation of such autoantibodies is largely unknown but is key to understanding their etiology. We developed a high-dimensional technique that enables detection of autoantibodies in complex with native antigens directly from patient plasma. Here, we used our platform to screen 1009 human plasma samples for 3600 autoantibody-antigen complexes, finding that plasma from patients with SCLC harbors, on average, fourfold higher disease-specific autoantibody signals compared with plasma from patients with other cancers. Across three independent SCLC cohorts, we identified a set of common but previously unknown autoantibodies that are produced in response to both intracellular and extracellular tumor antigens. We further characterized several disease-specific posttranslational modifications within extracellular proteins targeted by these autoantibodies, including citrullination, isoaspartylation, and cancer-specific glycosylation. Because most patients with SCLC have metastatic disease at diagnosis, we queried whether these autoantibodies could be used for SCLC early detection. We created a risk prediction model using five autoantibodies with an average area under the curve of 0.84 for the three cohorts that improved to 0.96 by incorporating cigarette smoke consumption in pack years. Together, our findings provide an innovative approach to identify circulating autoantibodies in SCLC with mechanistic insight into disease-specific immunogenicity and clinical utility.
Collapse
Affiliation(s)
- Kristin J Lastwika
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
- Translational Research Program, Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Andrew Kunihiro
- Translational Research Program, Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Joell L Solan
- Translational Research Program, Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Yuzheng Zhang
- Department of Biostatistics, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Lydia R Taverne
- Translational Research Program, Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - David Shelley
- Translational Research Program, Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Jung-Hyun Rho
- Translational Research Program, Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Timothy W Randolph
- Department of Biostatistics, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Christopher I Li
- Translational Research Program, Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Eric L Grogan
- Departments of Surgery, Medicine Radiology, and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Pierre P Massion
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt Ingram Cancer Center, Nashville, TN 37232, USA
| | - Annette L Fitzpatrick
- Department of Family Medicine, University of Washington, Seattle, WA 98195, USA
- Departments of Family Medicine, Epidemiology, and Global Health, University of Washington, Seattle, WA 98195, USA
| | - David MacPherson
- Translational Research Program, Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - A McGarry Houghton
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Paul D Lampe
- Translational Research Program, Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| |
Collapse
|
10
|
Challenges in the treatment of small cell lung cancer in the era of immunotherapy and molecular classification. Lung Cancer 2023; 175:88-100. [PMID: 36493578 DOI: 10.1016/j.lungcan.2022.11.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/18/2022] [Accepted: 11/20/2022] [Indexed: 11/26/2022]
Abstract
For many years the standard of care for small cell lung cancer (SCLC) has remained unchanged. Despite decades of active research, current treatment options are limited and the prognosis of patients with extended disease (ED) SCLC remains poor. The introduction of immune checkpoint inhibitors (ICIs) represents an exception and the only recent approval for ED-SCLC. However, the magnitude of benefit obtained with immunotherapy in SCLC is much more modest than that observed in other malignancies. Different pro-immunogenic or immunosuppressive features within the tumor microenvironment of SCLC may either modulate the sensitivity to immunotherapy or conversely dampen the efficacy of ICIs. Beside immunotherapy, a deeper understanding of the molecular biology of SCLC has led to the identification of new therapeutic targets for this lethal malignancy. Recent epigenetic and gene expression studies have resulted into a new molecular classification of four distinct subtypes of SCLC, defined by the relative expression of key transcription regulators and each characterized by specific therapeutic vulnerabilities. This review discusses the rationale for immunotherapy in SCLC and summarizes the main ICIs-trials in this tumor. We provide also an overview of new potential therapeutic opportunities and their integration with the new molecular classification of SCLC.
Collapse
|
11
|
Zekeridou A, Yang B, Lennon VA, Guo Y, Wu L, Lucchinetti CF, McKeon A, Pittock SJ, Flanagan EP. Anti-Neuronal Nuclear Antibody 3 Autoimmunity Targets Dachshund Homolog 1. Ann Neurol 2022; 91:670-675. [PMID: 35150165 PMCID: PMC9150768 DOI: 10.1002/ana.26320] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 01/11/2022] [Accepted: 01/31/2022] [Indexed: 11/11/2022]
Abstract
The antigen specificity of Anti-Neuronal Nuclear Antibody-type 3 (ANNA3)-IgG is unknown. We identified Dachshund-homolog 1 (DACH1) as the ANNA3 autoantigen and confirmed it by antigen-specific assays, immunohistochemical colocalization and immune absorption experiments. Patients' median age was 63.5 years (range, 49-88); 67% were female. Neurological manifestations (information available for 30 patients) included one or more of neuropathy, 12; cognitive difficulties, 11; ataxia, 8; dysautonomia, 7. Evidence of a neoplasm was present in 27 of 30 (90%), most of neuroendocrine lineage. DACH1-IgG is rare and represents a novel proposed biomarker of neurological autoimmunity and cancer. ANN NEUROL 2022;91:670-675.
Collapse
Affiliation(s)
- Anastasia Zekeridou
- Department of Neurology, Mayo Clinic, Rochester, MN
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
- Center of Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN
| | - Binxia Yang
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Vanda A. Lennon
- Department of Neurology, Mayo Clinic, Rochester, MN
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
- Center of Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN
- Department of Immunology, Mayo Clinic, Rochester
| | - Yong Guo
- Department of Neurology, Mayo Clinic, Rochester, MN
- Center of Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN
| | - Liang Wu
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Claudia F. Lucchinetti
- Department of Neurology, Mayo Clinic, Rochester, MN
- Center of Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN
| | - Andrew McKeon
- Department of Neurology, Mayo Clinic, Rochester, MN
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
- Center of Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN
| | - Sean J. Pittock
- Department of Neurology, Mayo Clinic, Rochester, MN
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
- Center of Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN
| | - Eoin P. Flanagan
- Department of Neurology, Mayo Clinic, Rochester, MN
- Center of Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN
| |
Collapse
|
12
|
Gutiérrez Pérez C, Lastra Aras E, Gómez Bravo R, Chivato Martín-Falquina I, Cuenca Zarzuela A, Rodríguez Ledesma I, García Girón C. Chronic intestinal pseudo-obstruction: diagnostic and prognostic utility of ANNA-1/Anti-Hu onconeural antibodies. REVISTA ESPANOLA DE ENFERMEDADES DIGESTIVAS 2022; 114:175-176. [PMID: 34991323 DOI: 10.17235/reed.2021.8521/2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
We propose an algorithm for the early detection of cancer from a case of paraneoplastic syndrome.
Collapse
|
13
|
Gutiérrez Pérez C, Lastra Aras E, Gómez Bravo R, Chivato Martín-Falquina I, Cuenca Zarzuela A, Rodríguez Ledesma I, García Girón C. Chronic intestinal pseudo-obstruction: diagnostic and prognostic utility of ANNA-1/Anti-Hu onconeural antibodies. REVISTA ESPANOLA DE ENFERMEDADES DIGESTIVAS 2021. [PMID: 34470446 DOI: 10.17235/reed.2021.8186/2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
A 65-year-old woman who, in the context of dyspepsia and dismotility, was diagnosed with chronic intestinal pseudo-obstruction (CIPO) in small cell lung carcinoma (SCLC). In spite of a remarkable tumor response after the combination of chemotherapy and immunotherapy, an intestinal sepsis led to the patient's sudden death.
Collapse
|
14
|
Zitvogel L, Perreault C, Finn OJ, Kroemer G. Beneficial autoimmunity improves cancer prognosis. Nat Rev Clin Oncol 2021; 18:591-602. [PMID: 33976418 DOI: 10.1038/s41571-021-00508-x] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2021] [Indexed: 02/06/2023]
Abstract
Many tumour antigens that do not arise from cancer cell-specific mutations are targets of humoral and cellular immunity despite their expression on non-malignant cells. Thus, in addition to the expected ability to detect mutations and stress-associated shifts in the immunoproteome and immunopeptidome (the sum of MHC class I-bound peptides) unique to malignant cells, the immune system also recognizes antigens expressed in non-malignant cells, which can result in autoimmune reactions against non-malignant cells from the tissue of origin. These autoimmune manifestations include, among others, vitiligo, thyroiditis and paraneoplastic syndromes, concurrent with melanoma, thyroid cancer and non-small-cell lung cancer, respectively. Importantly, despite the undesirable effects of these symptoms, such events can have prognostic value and correlate with favourable disease outcomes, suggesting 'beneficial autoimmunity'. Similarly, the occurrence of dermal and endocrine autoimmune adverse events in patients receiving immune-checkpoint inhibitors can have a positive predictive value for therapeutic outcomes. Neoplasias derived from stem cells deemed 'not essential' for survival (such as melanocytes, thyroid cells and most cells in sex-specific organs) have a particularly good prognosis, perhaps because the host can tolerate autoimmune reactions that destroy tumour cells at some cost to non-malignant tissues. In this Perspective, we discuss examples of spontaneous as well as therapy-induced autoimmunity that correlate with favourable disease outcomes and make a strong case in favour of this 'beneficial autoimmunity' being important not only in patients with advanced-stage disease but also in cancer immunosurveillance.
Collapse
Affiliation(s)
- Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France. .,Université Paris Saclay, Faculty of Medicine, Le Kremlin-Bicêtre, France. .,INSERM U1015, Gustave Roussy, Villejuif, France. .,Equipe labellisée par la Ligue contre le cancer, Villejuif, France. .,Center of Clinical Investigations in Biotherapies of Cancer (CICBT) BIOTHERIS, Villejuif, France. .,Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China.
| | - Claude Perreault
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Olivera J Finn
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Guido Kroemer
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France. .,Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China. .,Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, INSERM U1138, Centre de Recherche des Cordeliers, Institut Universitaire de France, Paris, France. .,Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France. .,Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France. .,Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
15
|
Neurological complications of immune checkpoint inhibitor cancer immunotherapy. J Neurol Sci 2021; 424:117424. [PMID: 33812689 DOI: 10.1016/j.jns.2021.117424] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 02/26/2021] [Accepted: 03/24/2021] [Indexed: 01/21/2023]
Abstract
Neurological autoimmunity is increasingly recognized as a complication of immune checkpoint inhibitor (ICI) cancer immunotherapy. ICIs act by enhancing endogenous anti-tumor immune responses and can also lead to autoimmunity affecting all organs. ICI-related neurological autoimmunity is rare, most often manifests with neuromuscular involvement and more rarely affects the central nervous system. Neurological complications often often present in the first three months of ICI treatment but can also appear after ICI discontinuation. These can occur in patients with tumors not traditionally associated with paraneoplastic neurological autoimmunity, such as melanoma and renal-cell carcinoma and should be suspected when a new neurological symptoms present while on ICI and cannot be explained by disease progression or as a consequence of metabolic dysfunction. Treatment consists of ICI discontinuation or withdrawal depending on the severity with or without immunosuppression. Generally, improvement is observed depending on the patient's baseline characteristics and neurological presentation.
Collapse
|
16
|
Farid S, Liu SV. Chemo-immunotherapy as first-line treatment for small-cell lung cancer. Ther Adv Med Oncol 2020; 12:1758835920980365. [PMID: 33414848 PMCID: PMC7750570 DOI: 10.1177/1758835920980365] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 11/23/2020] [Indexed: 12/13/2022] Open
Abstract
Small-cell lung cancer (SCLC) is a highly lethal subtype of lung cancer. Despite concerted efforts over the past several decades, there have been limited therapeutic advances. Traditional chemotherapy offers a high response rate and rapid symptomatic improvement, but its benefit is fleeting, and relapse is quick and unforgiving. Immunotherapy has delivered improved outcomes for patients with many cancers and there was compelling rationale for development in SCLC. While initial efforts with cytotoxic T-lymphocyte protein-4 inhibitors failed to improve upon chemotherapy alone, the addition of programmed death ligand-1 (PD-L1) inhibitors to first-line chemotherapy finally provided long-awaited gains in survival. Atezolizumab, when added to carboplatin and etoposide, improved both progression-free survival and overall survival. Durvalumab, when added to platinum plus etoposide, similarly improved OS. Biomarker development has stalled as PD-L1 expression and tumor mutational burden have not been useful predictive biomarkers. However, based on the significant survival improvements, both atezolizumab and durvalumab were approved by the US Food and Drug Administration to be given with first-line chemotherapy, and these regimens represent the new standards of care for SCLC.
Collapse
Affiliation(s)
- Saira Farid
- Department of Internal Medicine, MedStar Washington Hospital Center, Washington, DC, USA
| | - Stephen V Liu
- Lombardi Comprehensive Cancer Center, Georgetown University Hospital, 3800 Reservoir Road NW, Washington, DC 20007, USA
| |
Collapse
|
17
|
Sechi E, Zekeridou A. Neurologic Complications of Immune Checkpoint Inhibitors in Thoracic Malignancies. J Thorac Oncol 2020; 16:381-394. [PMID: 33188910 DOI: 10.1016/j.jtho.2020.11.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 10/30/2020] [Accepted: 11/05/2020] [Indexed: 01/08/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have transformed the prognosis of cancers previously considered lethal. The spectrum of therapeutic indications is rapidly expanding, including the vast majority of thoracic malignancies. By enhancing the immune responses against cancer, the ICI treatments lead to the development of immune-related adverse events (irAEs) that may affect any organ. Severity varies from mild to fatal clinical manifestations. Neurologic involvement is relatively rare and highly heterogeneous, including central and peripheral nervous system diseases associated with neural-specific autoantibodies or not, central nervous system vasculitis, and granulomatous and demyelinating disorders. Symptoms often manifest within the first four cycles of treatment and can develop regardless of the class of ICI used. An unfavorable outcome is found in up to one-third of patients and is generally associated with the patients' clinical characteristics (e.g., age, coexistence of systemic adverse events), cancer type (e.g., lung cancer versus other), and specific clinical setting (e.g., ICI treatment in patients with preexisting paraneoplastic neurologic autoimmunity, ICI rechallenge after a first neurologic irAE). Diagnosis should be suspected in patients with new-onset neurologic symptoms while on ICI treatment which are not explained by metastatic disease or other metabolic/infectious disorders. Recommended treatment is based on clinical severity and consists of ICI discontinuation with or without immunosuppressive/immunomodulatory therapy, although alternative approaches are reasonable depending on cancer status (e.g., aggressive immunosuppression without discontinuing ICI in patients with initial cancer response). Early recognition and appropriate treatment of these neurologic irAEs are crucial for improved patient outcomes and therapeutic planning.
Collapse
Affiliation(s)
- Elia Sechi
- Department of Neurology, Mayo Clinic, Rochester, Minnesota
| | - Anastasia Zekeridou
- Department of Neurology, Mayo Clinic, Rochester, Minnesota; Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
18
|
Xu Y, Zhan P, Song Y. [Immunotherapy Advances in Small Cell Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2020; 23:989-998. [PMID: 32752583 PMCID: PMC7679219 DOI: 10.3779/j.issn.1009-3419.2020.105.02] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Small cell lung cancer (SCLC) is a "refractory cancer" characterized by rapid growth and extensive early metastasis. About 70% of patients are already in the extensive stage at the time of diagnosis. Despite the high response rate to platinum-contained first-line chemotherapy, almost all patients subsequently experienced inevitable recurrence and had poor response to second-line treatment. The high mutation load and immunogenicity of SCLC suggest that immunotherapy may be effective for SCLC patients. Over the past few years, several clinical trials have evaluated the efficacy of checkpoint inhibitors [mainly cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) and programmed death 1 (PD-1)/programmed death ligand-1 (PD-L1) inhibitors] in SCLC patients and demonstrated promising survival prospects. This article reviewed the clinical studies of immune checkpoint inhibitors (ICIs) in the first-line, maintenance and second or more line treatment of SCLC. Besides, predictive biomarkers were discussed to select suitable patients for immunotherapy effectively.
Collapse
Affiliation(s)
- Yangyang Xu
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Nanjing Medical University, Nanjing 210002, China
| | - Ping Zhan
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing 210002, China
| | - Yong Song
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Nanjing Medical University, Nanjing 210002, China.,Department of Respiratory and Critical Care Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing 210002, China
| |
Collapse
|
19
|
Liu M, Deng S, Xiao T, Gao J. NOVA1 expression is associated with clinicopathological characteristics and prognosis in patients with small cell lung cancer. Transl Cancer Res 2020; 9:4373-4382. [PMID: 35117803 PMCID: PMC8798774 DOI: 10.21037/tcr-19-2806] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 05/21/2020] [Indexed: 12/01/2022]
Abstract
BACKGROUND Small cell lung cancer (SCLC) is a highly aggressive lung malignancy which is characteristic of rapid tumor proliferation, metastasis as well as paraneoplastic neurological syndromes (PNS). Recent studies have shown that neuro-oncological ventral antigen-1 (NOVA1) plays a crucial role in the progression of various tumors. However, its effect on SCLC is still exclusive. This study was aimed to explore NOVA1 expression in tumor tissues of SCLC patients as well as its correlation with clinicopathological characteristics and cancer-specific overall survivals. METHODS In this study, the patients pathologically diagnosed with new primary SCLC were enrolled. Firstly, we compared NOVA1 expression in SCLC tissues and adjacent normal tissues by immunohistochemistry. We further investigated the association of NOVA1 expression with clinicopathological markers (especially the categories of PNS) and survival time in SCLC patients. RESULTS Our finding exhibited that NOVA1 was remarkably expressed in SCLC tumorous tissues compared with adjacent normal lung tissues (P<0.001). Additionally, NOVA1 expression was closely associated with clinicopathological characteristics in SCLC patients in terms of tumor staging(χ2=15.833; P<0.001), lymph node metastasis (χ2=9.624; P=0.002), brain metastasis (χ2=9.624; P=0.002) and PNS (χ2=5.004; P=0.024). With the COX proportional hazard regression model, we detected that high expression of NOVA1 (HR =0.445; 95% CI: 0.213-0.934; P=0.032) was an independent factor for shorter survival time. CONCLUSIONS High expression of NOVA1 is closely associated with aggressive clinicopathological characteristics including PNS as well as poor survival in SCLC patients. NOVA1 can be served as a promising predictive factor for prognosis in SCLC.
Collapse
Affiliation(s)
- Meixuan Liu
- Department of Respiratory Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Respiratory Medicine, East Clinical College, Nanjing Medical University, Nanjing, China
| | - Shuangshuang Deng
- Department of Pathology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Tianyu Xiao
- Department of Pathology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jinli Gao
- Department of Pathology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
20
|
Sung S, Shirazi M, Shu CA, Crapanzano JP, Saqi A. Pulmonary small cell carcinoma: Review, common and uncommon differentials, genomics and management. Diagn Cytopathol 2020; 48:790-803. [PMID: 32348027 DOI: 10.1002/dc.24449] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/20/2020] [Accepted: 04/15/2020] [Indexed: 12/12/2022]
Abstract
Lung cancer is the leading cause of cancer-related death worldwide. It is divided into sub-categories based upon morphology, immunostaining pattern, biology, molecular profile, and/or treatment options. Up until the early 2000s when driver mutations with targeted therapies were identified in a subset of adenocarcinomas, the most critical distinction of lung carcinomas was driven by differences in treatment between small cell carcinoma (SCC) and nonsmall cell lung carcinoma (NSCLC). The distinction between SCC and NSCLC remains critical in the 21st century for management, especially for advanced stage cancer. In the vast majority of cases, morphological features are sufficient to separate SCC from other types of lung cancers. In some instances, however, cytomorphological features and immunohistochemical overlap with other tumors, limited sample availability, and/or crush artifact pose diagnostic challenges. The aim of this review is to highlight salient features of SCC and ancillary studies to distinguish it from common and uncommon potential mimickers, as well as provide updates in genomics and management.
Collapse
Affiliation(s)
- Simon Sung
- Department of Pathology & Cell Biology, Columbia University Medical Center, New York, New York, USA
| | - Maryam Shirazi
- Feinberg School of Medicine, Northwestern Medicine, Chicago, Illinois, USA
| | - Catherine A Shu
- Columbia University Medical Center, Department of Medicine, Hematology & Oncology, New York, New York, USA
| | - John P Crapanzano
- Department of Pathology & Cell Biology, Columbia University Medical Center, New York, New York, USA
| | - Anjali Saqi
- Department of Pathology & Cell Biology, Columbia University Medical Center, New York, New York, USA
| |
Collapse
|
21
|
Gelsomino F, Lamberti G, Parisi C, Casolari L, Melotti B, Sperandi F, Ardizzoni A. The evolving landscape of immunotherapy in small-cell lung cancer: A focus on predictive biomarkers. Cancer Treat Rev 2019; 79:101887. [PMID: 31491661 DOI: 10.1016/j.ctrv.2019.08.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 07/28/2019] [Accepted: 08/06/2019] [Indexed: 12/11/2022]
Abstract
Small cell lung cancer (SCLC) was defined as a "recalcitrant cancer" because of its dismal prognosis and lack of outcome improvements in the last 30 years. Immunotherapy with checkpoint inhibitors revolutionized treatment in many cancer types and results from the IMpower133 study, a double-blind placebo-controlled phase III trial, showed overall survival benefit for atezolizumab when added to standard platinum-etoposide chemotherapy in first-line SCLC setting for the first time since years. Trials with other checkpoint inhibitors, e.g. pembrolizumab, durvalumab, nivolumab and ipilimumab, are ongoing in various settings, but, to date, there are no defined factors to identify patients who are more likely to benefit from such treatments. This review summarizes results of immunotherapy trials in SCLC for first-line, maintenance and further-line therapies for single-agents and combinations with checkpoint inhibitors. Predictive factors from these trials are reviewed in order to identify their clinical value, with particular emphasis on PD-L1 expression on both tumor cells and in stroma, especially in pembrolizumab-treated patients, and tumor mutational burden, for patients treated with the ipilimumab and nivolumab combination.
Collapse
Affiliation(s)
- Francesco Gelsomino
- Department of Medical Oncology, Policlinico di Sant'Orsola University Hospital, Via P. Albertoni 15, 40138 Bologna, Italy.
| | - Giuseppe Lamberti
- Department of Experimental, Diagnostic and Specialty Medicine, Policlinico di Sant'Orsola University Hospital, Via P. Albertoni 15, 40138 Bologna, Italy.
| | - Claudia Parisi
- Department of Experimental, Diagnostic and Specialty Medicine, Policlinico di Sant'Orsola University Hospital, Via P. Albertoni 15, 40138 Bologna, Italy
| | - Laura Casolari
- Department of Medical Oncology, Policlinico di Sant'Orsola University Hospital, Via P. Albertoni 15, 40138 Bologna, Italy
| | - Barbara Melotti
- Department of Medical Oncology, Policlinico di Sant'Orsola University Hospital, Via P. Albertoni 15, 40138 Bologna, Italy.
| | - Francesca Sperandi
- Department of Medical Oncology, Policlinico di Sant'Orsola University Hospital, Via P. Albertoni 15, 40138 Bologna, Italy.
| | - Andrea Ardizzoni
- Department of Medical Oncology, Policlinico di Sant'Orsola University Hospital, Via P. Albertoni 15, 40138 Bologna, Italy.
| |
Collapse
|
22
|
Pavan A, Attili I, Pasello G, Guarneri V, Conte PF, Bonanno L. Immunotherapy in small-cell lung cancer: from molecular promises to clinical challenges. J Immunother Cancer 2019; 7:205. [PMID: 31383005 PMCID: PMC6683488 DOI: 10.1186/s40425-019-0690-1] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 07/24/2019] [Indexed: 12/17/2022] Open
Abstract
Management of small cell lung cancer (SCLC) has not changed over the last decades. In more recent years, alterations of DNA repair machinery and other molecular pathways have been identified in SCLC and preclinical data suggest that dysregulation of these pathways might offer new therapeutic opportunities.While immune checkpoint inhibitors (ICIs) have had a major impact on the clinical outcome of several solid tumors, including non-small cell lung cancer, the potential role of ICIs is currently under investigation in SCLC and some promising data are available. However, several clinical and biological hurdles have to be overcome and predictive markers are still eagerly needed. Knowledge of molecular pathways specifically involved in SCLC growth and treatment resistance is essential for a more rational planning of new combinations including ICIs.The present manuscript summarizes the current clinical evidence on immunotherapy in SCLC, describes the molecular bases underlying treatment resistance and discusses the potentialities and the rationale of different therapeutic combinations.
Collapse
Affiliation(s)
- A Pavan
- Medical Oncology 2, Istituto Oncologico Veneto IOV IRCCS, Via Gattamelata 64, 35100, Padova, Italia
- Department of Surgery, Oncology and Gastroenterology, Università degli Studi di Padova, Padova, Italia
| | - I Attili
- Medical Oncology 2, Istituto Oncologico Veneto IOV IRCCS, Via Gattamelata 64, 35100, Padova, Italia
- Department of Surgery, Oncology and Gastroenterology, Università degli Studi di Padova, Padova, Italia
| | - G Pasello
- Medical Oncology 2, Istituto Oncologico Veneto IOV IRCCS, Via Gattamelata 64, 35100, Padova, Italia
| | - V Guarneri
- Medical Oncology 2, Istituto Oncologico Veneto IOV IRCCS, Via Gattamelata 64, 35100, Padova, Italia
- Department of Surgery, Oncology and Gastroenterology, Università degli Studi di Padova, Padova, Italia
| | - P F Conte
- Medical Oncology 2, Istituto Oncologico Veneto IOV IRCCS, Via Gattamelata 64, 35100, Padova, Italia
- Department of Surgery, Oncology and Gastroenterology, Università degli Studi di Padova, Padova, Italia
| | - L Bonanno
- Medical Oncology 2, Istituto Oncologico Veneto IOV IRCCS, Via Gattamelata 64, 35100, Padova, Italia.
| |
Collapse
|
23
|
Huang YT, Zhang LH, Li MF, Cheng L, Zou GY, Zhou HH. A splice site mutation causing exon 6 skipping in SLC20A2 gene in a primary familial brain calcification family. Brain Res Bull 2019; 150:261-265. [PMID: 30634018 DOI: 10.1016/j.brainresbull.2019.01.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 12/24/2018] [Accepted: 01/03/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Primary familial brain calcification (PFBC) is a rare degenerative disease characterized by symmetrical bilateral calcinosis in the basal ganglia and other brain regions. It has an autosomal dominant inheritance pattern in most cases and exhibits genetic heterogeneity. Previous studies reported that SLC20A2, PDGFRB, PDGFB, XPR1 and MYORG are associated with PFBC, with SLC20A2 the main culprit. However, other mutations may also cause PFBC. Here, we performed a study to reveal the contributing mutations that gave rise to PFBC in a Chinese PFBC family. METHODS We recruited a PFBC family consisting of eight patients and eight healthy family members across three generations. Whole-exome sequencing, Sanger sequencing and RT-PCR were used to detect the genetic mutations. RESULTS Whole-exome sequencing revealed that c.730 + 1G > A of SLC20A2 was the candidate pathogenic mutation for the proband in this family. Genomic DNA PCR amplification and Sanger sequencing confirmed that all the patients from the family carried this mutation, while the healthy subjects in the family did not. Complementary DNA (cDNA) PCR amplification and Sanger sequencing confirmed that the patients had a mutation that caused exon 6 skipping in SLC20A2. CONCLUSION We identified a SLC20A2 splicing variant (c.730 + 1G > A) in a PFBC family. This mutation led to an alternative splicing event that skipped exon 6 in SLC20A2.
Collapse
Affiliation(s)
- Yuan-Tao Huang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410008, Hunan, PR China; Department of Neurology, the Brain Hospital of Hunan Province, Changsha 410008, Hunan, PR China
| | - Li-Hua Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410008, Hunan, PR China
| | - Mei-Fang Li
- Department of Otorhinolaryngology, the Brain Hospital of Hunan Province, Changsha 410008, Hunan, PR China
| | - Lin Cheng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, PR China
| | - Guo-Ying Zou
- Department of Clinical Laboratory, the Brain Hospital of Hunan Province, Changsha 410008, Hunan, PR China
| | - Hong-Hao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410008, Hunan, PR China.
| |
Collapse
|
24
|
Zekeridou A, Kryzer T, Guo Y, Hassan A, Lennon V, Lucchinetti CF, Pittock S, McKeon A. Phosphodiesterase 10A IgG: A novel biomarker of paraneoplastic neurologic autoimmunity. Neurology 2019; 93:e815-e822. [PMID: 31315972 PMCID: PMC6711657 DOI: 10.1212/wnl.0000000000007971] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 05/01/2019] [Indexed: 01/23/2023] Open
Abstract
OBJECTIVE To describe a novel antibody biomarker of neurologic paraneoplastic autoimmunity specific for phosphodiesterase 10A (PDE10A), a striatum-enriched phosphodiesterase, and to characterize the clinical phenotype of patients with PDE10A immunoglobulin G (IgG). METHODS We describe 7 patients with autoantibodies specific for PDE10A identified in the Mayo Clinic Neuroimmunology Laboratory. Patient specimens (sera, 7; CSF, 4) produced identical basal ganglia-predominant synaptic staining of murine brain tissue by indirect immunofluorescence. The autoantigen was identified by immunoprecipitation and mass spectrometry as PDE10A, and confirmed by antigen-specific recombinant Western blot and cell-based assays, and immune absorption experiments. RESULTS The median patient age was 70 years (range 66-76); 4 were men. Four patients with clinical information available had movement disorders (hyperkinetic in 3 [chorea, ballismus, dystonia] and parkinsonism in 1). All patients but one had cancer (lung [adenocarcinoma 1, squamous cell carcinoma 1, poorly differentiated mesenchymal carcinoma 1], renal adenocarcinoma 2, and pancreatic adenocarcinoma 1). Two of the 7 patients developed hyperkinetic movement disorders during treatment with immune checkpoint inhibitors (nivolumab and pembrolizumab), though none of 26 cancer control patients treated with immune checkpoint inhibitors harbored PDE10A IgG in their serum. MRIs from those 2 patients with hyperkinetic movement disorders demonstrated fluid-attenuated inversion recovery/T2 basal ganglia hyperintensities, and their CSF harbored unique oligoclonal bands. One of those 2 patients had substantial improvement after corticosteroids. One patient's renal adenocarcinoma expressed PDE10A by immunohistochemistry. CONCLUSIONS PDE10A IgG defines a novel rare neurologic autoimmune syndrome and expands the spectrum of diagnosable paraneoplastic CNS disorders. The intracellular location of PDE10A suggests a T-cell-mediated pathology targeting cells displaying MHC1-bound PDE10A peptides.
Collapse
Affiliation(s)
- Anastasia Zekeridou
- From the Departments of Laboratory Medicine and Pathology (A.Z., T.K., V.L., S.P., A.M.), Neurology (A.Z., Y.G., A.H., V.L., C.F.L., S.P., A.M.), and Immunology (V.L.), Mayo Clinic, Rochester, MN.
| | - Thomas Kryzer
- From the Departments of Laboratory Medicine and Pathology (A.Z., T.K., V.L., S.P., A.M.), Neurology (A.Z., Y.G., A.H., V.L., C.F.L., S.P., A.M.), and Immunology (V.L.), Mayo Clinic, Rochester, MN
| | - Yong Guo
- From the Departments of Laboratory Medicine and Pathology (A.Z., T.K., V.L., S.P., A.M.), Neurology (A.Z., Y.G., A.H., V.L., C.F.L., S.P., A.M.), and Immunology (V.L.), Mayo Clinic, Rochester, MN
| | - Anhar Hassan
- From the Departments of Laboratory Medicine and Pathology (A.Z., T.K., V.L., S.P., A.M.), Neurology (A.Z., Y.G., A.H., V.L., C.F.L., S.P., A.M.), and Immunology (V.L.), Mayo Clinic, Rochester, MN
| | - Vanda Lennon
- From the Departments of Laboratory Medicine and Pathology (A.Z., T.K., V.L., S.P., A.M.), Neurology (A.Z., Y.G., A.H., V.L., C.F.L., S.P., A.M.), and Immunology (V.L.), Mayo Clinic, Rochester, MN
| | - Claudia F Lucchinetti
- From the Departments of Laboratory Medicine and Pathology (A.Z., T.K., V.L., S.P., A.M.), Neurology (A.Z., Y.G., A.H., V.L., C.F.L., S.P., A.M.), and Immunology (V.L.), Mayo Clinic, Rochester, MN
| | - Sean Pittock
- From the Departments of Laboratory Medicine and Pathology (A.Z., T.K., V.L., S.P., A.M.), Neurology (A.Z., Y.G., A.H., V.L., C.F.L., S.P., A.M.), and Immunology (V.L.), Mayo Clinic, Rochester, MN
| | - Andrew McKeon
- From the Departments of Laboratory Medicine and Pathology (A.Z., T.K., V.L., S.P., A.M.), Neurology (A.Z., Y.G., A.H., V.L., C.F.L., S.P., A.M.), and Immunology (V.L.), Mayo Clinic, Rochester, MN
| |
Collapse
|
25
|
Paraneoplastic neurological syndromes in the era of immune-checkpoint inhibitors. Nat Rev Clin Oncol 2019; 16:535-548. [DOI: 10.1038/s41571-019-0194-4] [Citation(s) in RCA: 129] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
26
|
Zekeridou A, Majed M, Heliopoulos I, Lennon VA. Paraneoplastic autoimmunity and small-cell lung cancer: Neurological and serological accompaniments. Thorac Cancer 2019; 10:1001-1004. [PMID: 30810271 PMCID: PMC6449272 DOI: 10.1111/1759-7714.13009] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 01/17/2019] [Accepted: 01/17/2019] [Indexed: 01/21/2023] Open
Abstract
Paraneoplastic neurological autoimmunity is often associated with small‐cell lung cancer (SCLC), a highly malignant neuroendocrine tumor. Paraneoplastic autoimmunity often correlates with longer survival. We describe the paraneoplastic neurological manifestations of patients with SCLC with and without SCLC‐predictive autoantibodies and the correlation between autoimmunity and survival. We reviewed the records of 116 patients (51% male) from the Mayo Clinic with histopathologically confirmed SCLC for whom stored serum was available for neural autoantibody testing. Cancer was limited stage in 41%; the median age at diagnosis was 64 years. Paraneoplastic neurological manifestations were recorded in 61% (decreasing frequency: peripheral neuropathy, dysautonomia, cognitive decline, cerebellar ataxia, neuromuscular junction disorder, seizures, cranial neuropathy, movement disorder, brainstem disorder, or myelopathy). Neural autoantibodies, some with pathogenic potential, were detected in the sera of SCLC patients with and without neurological autoimmunity. The most frequent among patients with neurological manifestations were: anti‐neuronal nuclear antibody‐type 1, voltage‐gated calcium channel (VGCC)‐N‐type, VGCC‐P/Q‐type, glutamic acid decarboxylase 65 (GAD65), SOX1, and muscle acetylcholine receptor (AChR); while the most common in patients without neurological manifestations were: GAD65, muscle‐AChR, and VGCC‐P/Q‐type. Neither cancer stage at diagnosis nor survival correlated with neurological manifestations or autoantibody‐positivity, except for shorter survival in patients with myelopathy. The only predictor of longer survival was limited‐stage disease at diagnosis.
Collapse
Affiliation(s)
- Anastasia Zekeridou
- Department of Neurology, Mayo Clinic, Rochester, USA.,Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, USA
| | - Masoud Majed
- Department of Neurology, Mayo Clinic, Rochester, USA
| | - Ioannis Heliopoulos
- Department of Neurology, School of Medicine, University Hospital of Alexandroupolis, Democritus University of Thrace, Alexandroupoli, Greece
| | - Vanda A Lennon
- Department of Neurology, Mayo Clinic, Rochester, USA.,Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, USA.,Department of Immunology, Mayo Clinic, Rochester, USA
| |
Collapse
|
27
|
Hardy-Werbin M, Arpí O, Taus A, Rocha P, Joseph-Pietras D, Nolan L, Danson S, Griffiths R, Lopez-Botet M, Rovira A, Albanell J, Ottensmeier CH, Arriola E. Assessment of neuronal autoantibodies in patients with small cell lung cancer treated with chemotherapy with or without ipilimumab. Oncoimmunology 2017; 7:e1395125. [PMID: 29308329 PMCID: PMC5749672 DOI: 10.1080/2162402x.2017.1395125] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 10/17/2017] [Indexed: 12/17/2022] Open
Abstract
Small-cell lung cancer (SCLC) is often associated with paraneoplastic syndromes. To assess the role of anti-neuronal autoantibodies (NAAs) as biomarkers of treatment outcome, we assessed NAAs in serial samples from SCLC patients treated with chemoimmunotherapy compared to chemotherapy alone. We evaluated 2 cohorts: in cohort 1 (C1), 47 patients received standard platinum/etoposide, and in cohort 2 (C2), 38 patients received ipilimumab, carboplatin and etoposide. Serum samples at baseline and subsequent time points were analyzed for the presence of NAAs. NAAs were detected at baseline in 25 patients (53.2%) in C1 and in 20 patients (52.6%) in C2 (most frequently anti-Sox1). NAA at baseline was associated with limited disease (75% vs 50%; p: 0.096) and better overall survival (15.1 m vs 11.7 m; p: 0.032) in C1. Thirteen patients (28.9%) showed 2 or more reactivities before treatment; this was associated with worse PFS (5.5 m vs 7.3 m; p: 0.005) in patients treated with chemoimmunotherapy. NAA titers decreased after therapy in 68.9% patients, with no differential patterns of change between cohorts. Patients whose NAA titer decreased after treatment, showed longer OS [18.5 m (95% CI: 15.8 - 21.2)] compared with those whose NAA increased [12.3 m (95% CI: 8.1 - 16.5; p 0.049)], suggesting that antibody levels correlate to tumor load. Our findings reinforce the role of NAAs as prognostic markers and tumor activity/burden in SCLC, warrant further investigation in their predictive role for immunotherapy and raise concern over the use of immunotherapy in patients with more than one anti-NAA reactivity.
Collapse
Affiliation(s)
- M. Hardy-Werbin
- Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Universitat de Barcelona, Barcelona, Spain
| | - O. Arpí
- Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - A. Taus
- Medical Oncology Department, Hospital del Mar-CIBERONC, Barcelona, Spain
| | - P. Rocha
- Medical Oncology Department, Hospital del Mar-CIBERONC, Barcelona, Spain
| | - D. Joseph-Pietras
- NIHR Experimental Cancer Medicine Centre, Southampton, United Kingdom
| | - L. Nolan
- Medical Oncology Department, University Hospital Southampton, Southampton, United Kingdom
| | - S. Danson
- Sheffield Experimental Cancer Medicine Centre, Weston Park Hospital, Sheffield, United Kingdom
| | - R. Griffiths
- Medical Oncology Department, The Clatterbridge Cancer Centre NHS Foundation Trust, Wirral, United Kingdom
| | - M. Lopez-Botet
- Immunology unit, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - A. Rovira
- Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - J. Albanell
- Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Medical Oncology Department, Hospital del Mar-CIBERONC, Barcelona, Spain
| | - C. H. Ottensmeier
- Cancer Science Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- NIHR Experimental Cancer Medicine Centre, Southampton, United Kingdom
| | - E. Arriola
- Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Medical Oncology Department, Hospital del Mar-CIBERONC, Barcelona, Spain
| |
Collapse
|
28
|
Aggelopoulou E, Evangelatos G, Tzavida K, Koulouri V, Lazaridis N. Acquired Hemophilia and Anti-Hu Paraneoplastic Neurologic Syndrome in Small Cell Lung Cancer. Am J Med Sci 2017; 354:438-439. [PMID: 29078851 DOI: 10.1016/j.amjms.2017.03.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 03/08/2017] [Accepted: 03/08/2017] [Indexed: 12/25/2022]
Affiliation(s)
- Efi Aggelopoulou
- 3rd Department of Internal Medicine, 417 Army Share Fund Hospital, Athens, Greece.
| | | | - Konstantina Tzavida
- 3rd Department of Internal Medicine, 417 Army Share Fund Hospital, Athens, Greece
| | - Vasiliki Koulouri
- 3rd Department of Internal Medicine, 417 Army Share Fund Hospital, Athens, Greece
| | - Nikolaos Lazaridis
- 3rd Department of Internal Medicine, 417 Army Share Fund Hospital, Athens, Greece
| |
Collapse
|
29
|
Arriola E, Wheater M, Galea I, Cross N, Maishman T, Hamid D, Stanton L, Cave J, Geldart T, Mulatero C, Potter V, Danson S, Woll PJ, Griffiths R, Nolan L, Ottensmeier C. Outcome and Biomarker Analysis from a Multicenter Phase 2 Study of Ipilimumab in Combination with Carboplatin and Etoposide as First-Line Therapy for Extensive-Stage SCLC. J Thorac Oncol 2016; 11:1511-21. [PMID: 27296105 PMCID: PMC5063510 DOI: 10.1016/j.jtho.2016.05.028] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 05/23/2016] [Accepted: 05/23/2016] [Indexed: 11/24/2022]
Abstract
OBJECTIVES Our aim was to evaluate the safety and efficacy of ipilimumab combined with standard first-line chemotherapy for patients with extensive-stage SCLC. METHODS Patients with chemotherapy-naive extensive-stage SCLC were treated with carboplatin and etoposide for up to six cycles. Ipilimumab, 10 mg/kg, was given on day 1 of cycles 3 to 6 and every 12 weeks. Response was assessed by the Response Evaluation Criteria in Solid Tumors (RECIST), version 1.0, and immune-related response criteria. The primary end point was 1-year progression-free survival (PFS) according to RECIST. Secondary end points included PFS according to immune-related PFS and overall survival. Autoantibody serum levels were evaluated and correlated with clinical outcomes. RESULTS A total of 42 patients were enrolled between September 2011 and April 2014; 39 were evaluable for safety and 38 for efficacy. Six of 38 patients (15.8% [95% confidence interval (CI): 7.4-30.4]) were alive and progression-free at 1-year by RECIST. Median PFS was 6.9 months (95% CI: 5.5-7.9). Median immune-related PFS was 7.3 months (95% CI: 5.5-8.8). Median overall survival was 17.0 months (95% CI: 7.9-24.3). Of the patients evaluable for response, 21 of 29 (72.4%) achieved an objective response by RECIST and 28 of 33 (84.8%) achieved an objective response by the immune-related response criteria. All patients experienced at least one adverse event; at least one grade 3 or higher toxicity developed in 35 of 39 patients (89.7%); in 27 patients (69.2%) this was related to ipilimumab. Five deaths were reported to be related to ipilimumab. Positivity of an autoimmune profile at baseline was associated with improved outcomes and severe neurological toxicity. CONCLUSIONS Ipilimumab in combination with carboplatin and etoposide might benefit a subgroup of patients with advanced SCLC. Autoantibody analysis correlates with treatment benefit and toxicity and warrants further investigation.
Collapse
Affiliation(s)
| | - Matthew Wheater
- University Hospital Southampton, Southampton, United Kingdom
| | - Ian Galea
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Nadia Cross
- Southampton Clinical Trials Unit, University of Southampton, United Kingdom
| | - Tom Maishman
- Southampton Clinical Trials Unit, University of Southampton, United Kingdom
| | - Debbie Hamid
- Southampton Clinical Trials Unit, University of Southampton, United Kingdom
| | - Louise Stanton
- Southampton Clinical Trials Unit, University of Southampton, United Kingdom
| | - Judith Cave
- University Hospital Southampton, Southampton, United Kingdom
| | - Tom Geldart
- Royal Bournemouth and Christchurch Hospitals National Health Service Trust, Bournemouth, United Kingdom
| | | | - Vannessa Potter
- Nottingham University Hospitals National Health Service Trust, Nottingham, United Kingdom
| | - Sarah Danson
- Sheffield Experimental Cancer Medicine Centre, Weston Park Hospital, Sheffield, United Kingdom
| | - Pennella J Woll
- Sheffield Experimental Cancer Medicine Centre, Weston Park Hospital, Sheffield, United Kingdom
| | - Richard Griffiths
- The Clatterbridge Cancer Centre National Health Service Foundation Trust, Wirral, United Kingdom
| | - Luke Nolan
- University Hospital Southampton, Southampton, United Kingdom
| | | |
Collapse
|