51
|
Abstract
Immune checkpoint inhibitors (ICIs), used to treat many advanced cancers, activate the immune system to elicit an antitumor response. ICIs can also cause immune-related adverse events (irAEs) when nontumor tissues are affected by excess inflammation and autoimmunity. Rheumatic irAEs include inflammatory arthritis, myositis, sicca syndrome, polymyalgia rheumatica, and several other rare phenotypes. Treating rheumatic irAEs requires balancing the desire to decrease off-target inflammation while not negatively impacting the antitumor immune response. In this review, treatment recommendations for rheumatic irAEs have been discussed. Pathogenesis of rheumatic irAEs has been briefly reviewed. Knowledge about the effects of corticosteroids and steroid-sparing agents on tumor responses has been detailed to give context for treatment decisions. Recommendations ultimately depend not only on the clinical presentation and severity of the irAE but also on the goals of cancer treatment. Finally, how to safely use ICI therapy in patients with preexisting autoimmune diseases is considered.
Collapse
Affiliation(s)
- Pankti Reid
- University of Chicago, Department of Medicine, Section of Rheumatology, 5841 South Maryland Ave. MC 0930, Chicago, IL, 60637, USA.
| | - Laura C Cappelli
- Johns Hopkins University, Department of Medicine, Division of Rheumatology, 5501 Hopkins Bayview Circle, Suite 1B1, Baltimore, MD, 21224, USA.
| |
Collapse
|
52
|
Pean De Ponfilly – Sotier M, Seror R, Nocturne G, Besson FL. 18F-FDG PET molecular imaging: A relevant tool to investigate chronic inflammatory rheumatisms in clinical practice? Front Med (Lausanne) 2022; 9:1070445. [PMID: 36530882 PMCID: PMC9748427 DOI: 10.3389/fmed.2022.1070445] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 11/15/2022] [Indexed: 12/31/2023] Open
Abstract
18F-Labeled Fluorodeoxyglucose-Positron Emission Tomography (18F-FDG PET) is a molecular imaging tool commonly used in practice for the assessment of many cancers. Thanks to its properties, its use has been progressively extended to numerous inflammatory conditions, including chronic inflammatory rheumatism (CIR) such as rheumatoid arthritis (RA), spondylarthritis (SpAs) and polymyalgia rheumatica (PMR). 18F-FDG PET is currently not recommended for the diagnostic of CIRs. However, this whole-body imaging tool has emerged in clinical practice, providing a general overview of systemic involvement occurring in CIRs. Numerous studies have highlighted the capacity of 18F-FDG PET to detect articular and extra articular involvements in RA and PMR. However, the lack of specificity of 18F-FDG limits its use for diagnosis purpose. Finally, the key question is the definition of the best way to integrate this whole-body imaging tool in the patient's management workflow.
Collapse
Affiliation(s)
| | - Raphaële Seror
- Rheumatology, AP-HP. Université Paris-Saclay, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- Université Paris-Saclay, INSERM, CEA, Centre de Recherche en Immunologie des Infections Virales et des Maladies Auto-immunes, Le Kremlin-Bicêtre, France
| | - Gaetane Nocturne
- Rheumatology, AP-HP. Université Paris-Saclay, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- Université Paris-Saclay, INSERM, CEA, Centre de Recherche en Immunologie des Infections Virales et des Maladies Auto-immunes, Le Kremlin-Bicêtre, France
| | - Florent L. Besson
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- Biophysics and Nuclear Medicine-Molecular Imaging, AP-HP. Université Paris-Saclay, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, Orsay, France
| |
Collapse
|
53
|
Okuzumi S, Kamata H, Emoto K, Shimizu T, Otake S, Irie H, Chubachi S, Ikemura S, Yasuda H, Fukunaga K. Successful Steroid Treatment of Pembrolizumab-induced Agranulocytosis that Developed after Splenectomy in a Patient with Non-small-cell Lung Cancer: A Case Report. Intern Med 2022. [PMID: 36450461 PMCID: PMC10400385 DOI: 10.2169/internalmedicine.0278-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Neutropenia, a rare immune-related adverse event, affects patients receiving treatment with immune checkpoint inhibitors (ICIs). We herein report a case of pembrolizumab-induced agranulocytosis. An 83-year-old man was diagnosed with advanced-stage lung carcinoma concomitant with splenomegaly complicated by hypersplenism, causing pancytopenia. To avoid the risk of bone marrow suppression due to cytotoxic chemotherapy, pembrolizumab monotherapy was chosen. However, the patient developed agranulocytosis despite the resolution of pancytopenia through splenectomy performed after the fourth pembrolizumab cycle. Neutrophil counts improved after steroid treatment but not after granulocyte colony-stimulating factor treatment. This case demonstrated that neutropenia can sometimes develop abruptly after several ICI therapy cycles.
Collapse
Affiliation(s)
- Shinichi Okuzumi
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Japan
| | - Hirofumi Kamata
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Japan
| | - Katsura Emoto
- Division of Diagnostic Pathology, Keio University School of Medicine, Japan
| | - Takayuki Shimizu
- Division of Hematology, Department of Medicine, Keio University School of Medicine, Japan
| | - Shiro Otake
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Japan
| | - Hidehiro Irie
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Japan
| | - Shotaro Chubachi
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Japan
| | - Shinnosuke Ikemura
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Japan
| | - Hiroyuki Yasuda
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Japan
| | | |
Collapse
|
54
|
Gatto M, Bjursten S, Jonsson CA, Agelii ML, Jonell C, McGrath S, Lönnblom E, Sareila O, Holmdahl R, Rudin A, Levin M, Gjertsson I. Early Increase of Circulating Transitional B Cells and Autoantibodies to Joint-Related Proteins in Patients With Metastatic Melanoma Developing Checkpoint Inhibitor-Induced Inflammatory Arthritis. Arthritis Rheumatol 2022; 75:856-863. [PMID: 36409578 DOI: 10.1002/art.42406] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 09/20/2022] [Accepted: 11/08/2022] [Indexed: 11/23/2022]
Abstract
OBJECTIVE To investigate potential associations between B cell-related immunologic changes and development of inflammatory arthritis (IA) after treatment with immune checkpoint inhibitors (ICIs). METHODS Patients who developed ICI-induced IA (ICI-IA) and patients who did not develop immune-related adverse events (non-IRAE) after receiving ICIs to treat metastatic melanoma were consecutively recruited. Blood samples were collected at the time of ICI-IA occurrence and at different time points during treatment. Peripheral blood B cell subsets during ICI treatment were analyzed by flow cytometry. Rheumatoid factor, anti-citrullinated protein antibodies, and antibodies against joint-related proteins were measured. RESULTS Proportions of CD19+ B cells were higher in patients with ICI-IA (n = 7) compared to patients with non-IRAE (n = 15) (median 11.7% [interquartile range (IQR) 9.7-16.2%] versus 8.1% [IQR 5.7-11.0%]; P = 0.03). The proportion and absolute numbers of transitional CD19+CD10+CD24high CD38high B cells were increased in patients with ICI-IA compared to non-IRAE patients (median 8.1% [IQR 4.9-12.1%] versus 3.6% [IQR 1.9-4.9%]; median 10.7 cells/μl [IQR 8.9-19.6] versus 4.4 cells/μl [IQR 2.3-6.6]; P < 0.01 for both). In addition, higher levels of transitional B cells were associated with development of ICI-IA (odds ratio 2.25 [95% confidence interval 1.03-4.9], P = 0.04). Transitional B cells increased before the onset of overt ICI-IA and decreased between the active and quiescent stages of ICI-IA (P = 0.02). Autoantibodies to type II collagen epitopes were detected in up to 43% of ICI-IA patients compared to none of the non-IRAE patients (P = 0.02). CONCLUSION Development of ICI-IA is accompanied by an increase in transitional B cells and by production of autoantibodies to joint-related proteins. Monitoring of B cell-driven abnormalities upon ICI treatment may help earlier recognition of ICI-IA.
Collapse
Affiliation(s)
- Mariele Gatto
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden, and Department of Medicine, Unit of Rheumatology, University of Padova, Italy
| | - Sara Bjursten
- Department of Oncology, Sahlgrenska University Hospital, and Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Charlotte A Jonsson
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Monica Leu Agelii
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Caroline Jonell
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Sarah McGrath
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Erik Lönnblom
- Department of Medical Biochemistry and Biophysics, Section for Medical Inflammation Research, Karolinska Institute, Stockholm, Sweden
| | - Outi Sareila
- Department of Medical Biochemistry and Biophysics, Section for Medical Inflammation Research, Karolinska Institute, Stockholm, Sweden
| | - Rikard Holmdahl
- Department of Medical Biochemistry and Biophysics, Section for Medical Inflammation Research, Karolinska Institute, Stockholm, Sweden
| | - Anna Rudin
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Max Levin
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden, Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden, and Wallenberg Laboratory for Cardiovascular Research, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Inger Gjertsson
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
55
|
Benesova K, Kraus FV, Carvalho RA, Lorenz H, Hörth CH, Günther J, Klika KD, Graf J, Diekmann L, Schank T, Christopoulos P, Hassel JC, Lorenz HM, Souto-Carneiro M. Distinct immune-effector and metabolic profile of CD8 + T cells in patients with autoimmune polyarthritis induced by therapy with immune checkpoint inhibitors. Ann Rheum Dis 2022; 81:1730-1741. [PMID: 35922125 PMCID: PMC9664113 DOI: 10.1136/ard-2022-222451] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 07/16/2022] [Indexed: 02/07/2023]
Abstract
OBJECTIVES Rheumatic immune-related adverse events (irAE) such as (poly)arthritis in patients undergoing immune checkpoint inhibitor (ICI) treatment pose a major clinical challenge. ICI therapy improves CD8+ T cell (CD8) function, but CD8 contributes to chronic inflammation in autoimmune arthritis (AA). Thus, we investigated whether immune functional and metabolic changes in CD8 explain the development of musculoskeletal irAE in ICI-treated patients. METHODS Peripheral CD8 obtained from ICI-treated patients with and without arthritis irAEs and from AA patients with and without a history of malignancy were stimulated in media containing 13C-labelled glucose with and without tofacitinib or infliximab. Changes in metabolism, immune-mediator release, expression of effector cell-surface molecules and inhibition of tumour cell growth were quantified. RESULTS CD8 from patients with irAE showed significantly lower frequency and expression of cell-surface molecule characteristic for activation, effector-functions, homing, exhaustion and apoptosis and reduced release of cytotoxic and proinflammatory immune mediators compared with CD8 from ICI patients who did not develop irAE. This was accompanied by a higher glycolytic rate and ATP production. Gene-expression analysis of pre-ICI-treated CD8 revealed several differentially expressed transcripts in patients who later developed arthritis irAEs. In vitro tofacitinib or infliximab treatment did not significantly change the immune-metabolic profile nor the capacity to release cytolytic mediators that inhibit the growth of the human lung cancer cell line H838. CONCLUSIONS Our study shows that CD8 from ICI-treated patients who develop a musculoskeletal irAE has a distinct immune-effector and metabolic profile from those that remain irAE free. This specific irAE profile overlaps with the one observed in CD8 from AA patients and may prove useful for novel therapeutic strategies to manage ICI-induced irAEs.
Collapse
Affiliation(s)
- Karolina Benesova
- Department of Internal Medicine V Hematology Oncology Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Franziska Viktoria Kraus
- Department of Internal Medicine V Hematology Oncology Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Rui A Carvalho
- Department of Life Sciences, University of Coimbra Faculty of Sciences and Technology, Coimbra, Portugal
| | - Holger Lorenz
- Imaging Facility, Center of Molecular Biology (ZMBH), Universität Heidelberg, Heidelberg, Germany
| | - Christian H Hörth
- Imaging Facility, Center of Molecular Biology (ZMBH), Universität Heidelberg, Heidelberg, Germany
| | - Janine Günther
- Department of Internal Medicine V Hematology Oncology Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Karel D Klika
- Molecular and Structural Biology, German Cancer Research Centre, Heidelberg, Germany
| | - Jürgen Graf
- Nuclear Magnetic Resonance Laboratory, Institute of Organic Chemistry, Universität Heidelberg, Heidelberg, Germany
| | - Leonore Diekmann
- Department of Internal Medicine V Hematology Oncology Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Timo Schank
- Department of Dermatology, University Hospital Heidelberg, Heidelberg, Germany
- National Center of Tumor Diseases, Heidelberg, Germany
| | - Petros Christopoulos
- National Center of Tumor Diseases, Heidelberg, Germany
- Department of Thoracic Oncology, University Hospital Heidelberg, Heidelberg, Germany
| | - Jessica C Hassel
- Department of Dermatology, University Hospital Heidelberg, Heidelberg, Germany
- National Center of Tumor Diseases, Heidelberg, Germany
| | - Hanns-Martin Lorenz
- Department of Internal Medicine V Hematology Oncology Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Margarida Souto-Carneiro
- Department of Internal Medicine V Hematology Oncology Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
56
|
Hsu ML, Murray JC, Psoter KJ, Zhang J, Barasa D, Brahmer JR, Ettinger DS, Forde PM, Hann CL, Lam VK, Levy B, Marrone KA, Patel T, Peterson V, Sagorsky S, Turner M, Anagnostou V, Naidoo J, Feliciano JL. Clinical Features, Survival, and Burden of Toxicities in Survivors More Than One Year After Lung Cancer Immunotherapy. Oncologist 2022; 27:971-981. [PMID: 35972337 PMCID: PMC9632301 DOI: 10.1093/oncolo/oyac140] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 06/01/2022] [Indexed: 01/05/2023] Open
Abstract
INTRODUCTION Anti-PD-(L)1 immune checkpoint inhibitors (ICI) improve survival in patients with advanced non-small cell lung cancer (aNSCLC). The clinical features, survival, and burden of toxicities of patients with aNSCLC alive >1 year from ICI initiation are poorly understood. MATERIALS AND METHODS We defined ICI survivors as patients alive >1 year after ICI start and retrospectively reviewed demographics, treatment, and immune-related adverse events (irAEs). Long-term irAEs were defined as ongoing irAEs lasting >1 year; burden of toxicity measures were based on percentage of days a patient experienced toxicity. Using linear and logistic regression, we evaluated association between demographics and disease characteristics with burden of toxicity. RESULTS We identified 114 ICI survivors from 317 patients with aNSCLC. Half (52%) experienced an irAE of any grade, and 23.7% developed long-term irAEs. More ICI survivors with irAES in the first year had never smoked (P = .018) or received ICIs as frontline therapy (P = .015). The burden of toxicity in the first year significantly correlated with the burden of toxicity afterward (ρ = 0.72; P < .001). No patients with progressive disease had a high burden of toxicity, and they experienced 30.6% fewer days with toxicity than those with stable disease. Increased duration of therapy was associated with higher odds of experiencing toxicity. Half of ICI survivors with irAEs were still receiving treatment for unresolved irAEs at time of death or last follow-up. CONCLUSION Significant proportions of ICI survivors have unresolved long-term toxicities. These data support a growing need to understand long-term toxicity to optimize management of those treated with ICIs.
Collapse
Affiliation(s)
- Melinda L Hsu
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
- Bloomberg-Kimmel institute for Cancer Immunotherapy, Baltimore, MD, USA
| | - Joseph C Murray
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
- Bloomberg-Kimmel institute for Cancer Immunotherapy, Baltimore, MD, USA
| | - Kevin J Psoter
- Department of Pediatrics, Johns Hopkins University, Baltimore, MD, USA
| | - Jiajia Zhang
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
- Bloomberg-Kimmel institute for Cancer Immunotherapy, Baltimore, MD, USA
| | - Durrant Barasa
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
- Bloomberg-Kimmel institute for Cancer Immunotherapy, Baltimore, MD, USA
| | - Julie R Brahmer
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
- Bloomberg-Kimmel institute for Cancer Immunotherapy, Baltimore, MD, USA
| | - David S Ettinger
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
- Bloomberg-Kimmel institute for Cancer Immunotherapy, Baltimore, MD, USA
| | - Patrick M Forde
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
- Bloomberg-Kimmel institute for Cancer Immunotherapy, Baltimore, MD, USA
| | - Christine L Hann
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
- Bloomberg-Kimmel institute for Cancer Immunotherapy, Baltimore, MD, USA
| | - Vincent K Lam
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
- Bloomberg-Kimmel institute for Cancer Immunotherapy, Baltimore, MD, USA
| | - Benjamin Levy
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
- Bloomberg-Kimmel institute for Cancer Immunotherapy, Baltimore, MD, USA
| | - Kristen A Marrone
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
- Bloomberg-Kimmel institute for Cancer Immunotherapy, Baltimore, MD, USA
| | | | | | | | - Michelle Turner
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
- Bloomberg-Kimmel institute for Cancer Immunotherapy, Baltimore, MD, USA
| | - Valsamo Anagnostou
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
- Bloomberg-Kimmel institute for Cancer Immunotherapy, Baltimore, MD, USA
| | - Jarushka Naidoo
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
- Bloomberg-Kimmel institute for Cancer Immunotherapy, Baltimore, MD, USA
- Beaumont Hospital Dublin and RCSI University of Health Sciences, Dublin, Ireland
| | - Josephine L Feliciano
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
- Bloomberg-Kimmel institute for Cancer Immunotherapy, Baltimore, MD, USA
| |
Collapse
|
57
|
Moura CA, Moura CG. Severe polymyalgia-like symptoms secondary to anti-PD1 therapy successfully managed without discontinuing checkpoint inhibitor. Ann Rheum Dis 2022; 81:e237. [PMID: 32376598 DOI: 10.1136/annrheumdis-2020-217605] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 04/20/2020] [Accepted: 04/22/2020] [Indexed: 01/09/2023]
Affiliation(s)
- Carlos Antonio Moura
- Internal Medicine and Rheumatology, Hospital Santo Antonio, Salvador, Bahia, Brazil
| | - Carlos Geraldo Moura
- Internal Medicine and Rheumatology, Hospital Santo Antonio, Salvador, Bahia, Brazil
| |
Collapse
|
58
|
De La Fuente F, Belkhir R, Henry J, Nguyen CD, Pham T, Germain V, Gavand PE, Labadie C, Briere C, Lauret A, Cardon T, Mouterde G, Bonnet I, Rouxel L, Truchetet ME, Schaeverbeke T, Richez C, Kostine M. Use of a bDMARD or tsDMARD for the management of inflammatory arthritis under checkpoint inhibitors: an observational study. RMD Open 2022; 8:rmdopen-2022-002612. [PMID: 36270747 PMCID: PMC9594531 DOI: 10.1136/rmdopen-2022-002612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 10/02/2022] [Indexed: 12/04/2022] Open
Abstract
OBJECTIVE There is limited experience regarding the use of biological disease-modifying antirheumatic drug (bDMARD) and JAK inhibitor (JAKi) for the management of immune checkpoint inhibitors (ICI)-induced inflammatory arthritis. We aimed to assess their efficacy and safety in this setting. METHODS Using the Club Rhumatismes and Inflammation French network, we conducted a multicentre, retrospective, observational study of patients with cancer diagnosed with inflammatory arthritis under ICI(s) and treated with bDMARD or JAKi. Clinical data were collected using a standardised case report form. RESULTS Twenty patients (60% men, median age 69.5 years) were included, with rheumatoid arthritis (RA)-like (n=16), polymyalgia rheumatica-like (n=2) or psoriatic arthritis-like (n=2) clinical presentation. Two patients had pre-existing RA. 90% were treated with glucocorticoids as first-line therapy and 60% received methotrexate prior to bDMARD or JAKi. Anti-interleukin-6 receptor (IL-6R) therapy was used in 13/20 patients (65%), leading to clinical improvement in 11/13 patients (85%), but one patient experienced intestinal perforation and cancer progression was noticed in 6/13 patients (46%). Tumour necrosis factor inhibitors were used in 5/20 patients (25%), with improvement in 4/5 patients (80%) and cancer progression was observed in 3/5 patients (60%). Two infections (diverticulitis and pneumonitis) were reported. Anakinra, baricitinib and ustekinumab were each used in one patient. Median duration of the bDMARD or JAKi was 17 weeks. CONCLUSION Anti-IL-6R therapy is currently the most common strategy in patients with ICI-induced inflammatory arthritis and insufficient response to glucocorticoids and methotrexate, leading to improvement in >80%. Overall, cancer progression occurred in about half of patients and whether the bDMARD/JAKi impacted the tumour response remains to be determined.
Collapse
Affiliation(s)
- Fanny De La Fuente
- Department of Rheumatology, Centre Hospitalier Universitaire de Bordeaux Groupe Hospitalier Pellegrin, Bordeaux, France
| | - Rakiba Belkhir
- Department of Rheumatology, Hôpital Bicetre, Assistance Publique-Hôpitaux de Paris, FHU CARE, Le Kremlin-Bicetre, France,Inserm UMR 1184, Universite Paris-Saclay Faculte de Medecine, Le Kremlin-Bicetre, France
| | - Julien Henry
- Department of Rheumatology, Hôpital Bicetre, Assistance Publique-Hôpitaux de Paris, FHU CARE, Le Kremlin-Bicetre, France,Inserm UMR 1184, Universite Paris-Saclay Faculte de Medecine, Le Kremlin-Bicetre, France
| | - Chi Duc Nguyen
- Department of Rheumatology, Centre Hospitalier de Bethune, Bethune, France
| | - Thao Pham
- Department of Rheumatology, Sainte-Marguerite Hospital, Assistance Publique - Hôpitaux de Marseille, Marseille, France
| | - Vincent Germain
- Department of Rheumatology, Centre Hospitalier de Pau, Pau, France
| | | | - Céline Labadie
- Department of Rheumatology, Centre Hospitalier Universitaire de Bordeaux Groupe Hospitalier Pellegrin, Bordeaux, France
| | - Claire Briere
- Department of Internal Medicine, Centre Hospitalier Intercommunal de Creteil, Creteil, France
| | - Ambre Lauret
- Department of Internal Medicine, Centre Hospitalier Intercommunal de Creteil, Creteil, France
| | - Thierry Cardon
- Department of Rheumatology, Lille University Hospital Center, Lille, France
| | - Gael Mouterde
- Departement of Rheumatology, Lapeyronie Hospital, Montpellier, France
| | - Isabelle Bonnet
- Department of Rheumatology, Hôpital Bicetre, Assistance Publique-Hôpitaux de Paris, FHU CARE, Le Kremlin-Bicetre, France,Inserm UMR 1184, Universite Paris-Saclay Faculte de Medecine, Le Kremlin-Bicetre, France
| | - Léa Rouxel
- Department of Rheumatology, Arcachon Hospital, La Teste de Buch, France
| | - Marie-Elise Truchetet
- Department of Rheumatology, Centre Hospitalier Universitaire de Bordeaux Groupe Hospitalier Pellegrin, Bordeaux, France
| | - Thierry Schaeverbeke
- Department of Rheumatology, Centre Hospitalier Universitaire de Bordeaux Groupe Hospitalier Pellegrin, Bordeaux, France
| | - Christophe Richez
- Department of Rheumatology, Centre Hospitalier Universitaire de Bordeaux Groupe Hospitalier Pellegrin, Bordeaux, France
| | - Marie Kostine
- Department of Rheumatology, Centre Hospitalier Universitaire de Bordeaux Groupe Hospitalier Pellegrin, Bordeaux, France
| |
Collapse
|
59
|
Yang Y, Huang XJ. Immune checkpoint inhibitor-associated arthritis in advanced pulmonary adenocarcinoma: A case report. World J Clin Cases 2022; 10:10701-10707. [PMID: 36312471 PMCID: PMC9602251 DOI: 10.12998/wjcc.v10.i29.10701] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/16/2022] [Accepted: 09/01/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND With the wide application of immune checkpoint inhibitors (ICIs) in cancer treatment, immune-related adverse events occur frequently, involving almost all organs and systems. The incidence of ICI-associated arthritis (IA) is unknown. In most cases, IA is not serious and non-lethal. Higher checkpoint inhibitor arthritis disease activity may be associated with cancer progression. Here, we report a severe case of IA with high arthritis disease activity in advanced pulmonary adenocarcinoma, causing permanent withdrawal of pembrolizumab, but the patient remained in complete remission (CR) 20 mo after the development of IA. CASE SUMMARY An 81-year-old smoking man was admitted to our hospital because of left chest pain for 9 mo. He was finally diagnosed with advanced pulmonary adenocarcinoma, with programmed cell death 1 ligand 1 expression of 70%. The patient responded to pembrolizumab treatment and achieved CR, but IA occurred after the 5th cycle of pembrolizumab administration. Although non-steroidal anti-inflammatory drugs and disease-modifying anti-rheumatic drugs were prescribed, arthralgia and joint swelling occurred. The symptoms of arthritis were further aggravated when immunotherapy was given again after short-term withdrawal. Clinical Disease Activity Index (CDAI) score, a traditional measure of arthritis activity, was 43. Intravenous methylprednisolone was prescribed at 20 mg/d and then tapered over the subsequent 4 wk. The symptoms of arthritis steadily improved and completely resolved 4 mo after withdrawal of pembrolizumab. A recent follow-up in June 2022 revealed satisfactory clinical recovery of arthritis and the patient remained in CR. CONCLUSION This case report highlights that early recognition of IA and appropriate treatment are critical to improving the outcome of both ICI-arthritis and lung cancer.
Collapse
Affiliation(s)
- Yan Yang
- Department of Respiratory Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| | - Xiao-Jie Huang
- Department of Respiratory Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| |
Collapse
|
60
|
Plaçais L, Dalle S, Dereure O, Trabelsi S, Dalac S, Legoupil D, Montaudié H, Arnault JP, De Quatrebarbes J, Saiag P, Brunet-Possenti F, Lesimple T, Maubec E, Aubin F, Granel-Brocard F, Grob JJ, Stoebner PE, Allayous C, Oriano B, Dutriaux C, Mortier L, Lebbe C. Risk of irAEs in patients with autoimmune diseases treated by immune checkpoint inhibitors for stage III or IV melanoma: results from a matched case-control study. Ann Rheum Dis 2022; 81:1445-1452. [PMID: 35788496 DOI: 10.1136/ard-2022-222186] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 06/15/2022] [Indexed: 11/03/2022]
Abstract
OBJECTIVE To quantify the risk of immune-related adverse events (irAEs) in patients with pre-existing autoimmune disease (pAID) treated by immune checkpoint inhibitors (ICIs) for stage III or IV melanoma. METHODS Case-control study performed on a French multicentric prospective cohort of patients with melanoma, matched for irAE risk factors and oncological staging. Risk of irAE was assessed by logistic regression. RESULTS 110 patients with pAID were included and matched with 330 controls, from March 2013 to October 2020. Over a median follow-up period of 7.2 months for cases and 6.9 months for controls, the ORs of developing all-grade and grade ≥3 irAEs among cases compared with controls were 1.91 (95% CI (1.56 to 2.27)) and 1.44 (95% CI (1.08 to 1.82)), respectively. Patients with pAID had an increased risk of multiple irAEs (OR 1.46, 95% CI (1.15 to 2.67)) and a shorter time to irAE onset. In contrast, there were no difference in irAE-related mortality nor in the rate of treatment discontinuation, and a landmark analysis revealed a better survival at 24 months among cases (p=0.02). Thirty per cent of cases experienced a pAID flare during follow-up, and baseline immunosuppression did not prevent irAE occurrence. Last, we report associations between the pAID clinical subsets and organ-specific irAEs. CONCLUSION In our study, patients with pAID were at greater risk of all-grade, severe and multiple irAEs, yet had a better 24-month survival than controls. Thus, patients with pAID should be eligible for ICI therapy but benefit from a close monitoring for irAE occurrence, especially during the first months of therapy.
Collapse
Affiliation(s)
- Léo Plaçais
- Internal Medicine and Clinical Immunology, Hopital Bicêtre, Le Kremlin-Bicêtre, Île-de-France, France
| | - Stéphane Dalle
- Dermatology, Centre Hospitalier Universitaire de Lyon, Lyon, Rhône-Alpes, France
| | - Olivier Dereure
- Dermatology, CHU Montpellier, Montpellier, Languedoc-Roussillon, France
| | - Sabiha Trabelsi
- Dermatology, CHU Grenoble Alpes, Grenoble, Auvergne-Rhone-Alpes, France
| | - Sophie Dalac
- Dermatology, CHU Dijon, Dijon, Bourgogne, France
| | | | | | - Jean-Philippe Arnault
- Dermatology, Centre Hospitalier Universitaire Amiens-Picardie, Amiens, Hauts-de-France, France
| | | | - Philippe Saiag
- Dermatology, Hôpital Ambroise Paré, Neuilly-sur-Seine, Île-de-France, France
| | | | | | - Eve Maubec
- Dermatology, Hopital Avicenne, Bobigny, France
| | | | | | - Jean-Jacques Grob
- Dermatology, Hôpital de la Timone, Marseille, Provence-Alpes-Côte d'Azu, France
| | | | - Clara Allayous
- Dermatology, CHU Saint-Louis, Paris, Île-de-France, France
| | - Bastien Oriano
- Dermatology, CHU Saint-Louis, Paris, Île-de-France, France
| | - Caroline Dutriaux
- Dermatology, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, Aquitaine, France
| | | | - Céleste Lebbe
- Dermatology, Hopital Saint-Louis, Paris, Île-de-France, France
| |
Collapse
|
61
|
Abstract
MRI is a widely available clinical tool for cancer diagnosis and treatment monitoring. MRI provides excellent soft tissue imaging, using a wide range of contrast mechanisms, and can non-invasively detect tissue metabolites. These approaches can be used to distinguish cancer from normal tissues, to stratify tumor aggressiveness, and to identify changes within both the tumor and its microenvironment in response to therapy. In this review, the role of MRI in immunotherapy monitoring will be discussed and how it could be utilized in the future to address some of the unique clinical questions that arise from immunotherapy. For example, MRI could play a role in identifying pseudoprogression, mixed response, T cell infiltration, cell tracking, and some of the characteristic immune-related adverse events associated with these agents. The factors to be considered when developing MRI imaging biomarkers for immunotherapy will be reviewed. Finally, the advantages and limitations of each approach will be discussed, as well as the challenges for future clinical translation into routine clinical care. Given the increasing use of immunotherapy in a wide range of cancers and the ability of MRI to detect the microstructural and functional changes associated with successful response to immunotherapy, the technique has great potential for more widespread and routine use in the future for these applications.
Collapse
Affiliation(s)
- Doreen Lau
- Centre for Immuno-Oncology, University of Oxford, Oxford, UK
| | - Pippa G Corrie
- Department of Oncology, Addenbrooke's Hospital, Cambridge, UK
| | | |
Collapse
|
62
|
Cappelli LC, Bingham CO, Forde PM, Anagnostou V, Brahmer J, Lipson EJ, Mammen J, Schollenberger M, Shah AA, Darrah E. Anti-RA33 antibodies are present in a subset of patients with immune checkpoint inhibitor-induced inflammatory arthritis. RMD Open 2022; 8:rmdopen-2022-002511. [PMID: 36096522 PMCID: PMC9472204 DOI: 10.1136/rmdopen-2022-002511] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 08/22/2022] [Indexed: 11/04/2022] Open
Abstract
OBJECTIVE Patients with inflammatory arthritis (IA) associated with immune checkpoint inhibitor (ICI) treatment for cancer are typically seronegative for anti-cyclic citrullinated peptide (CCP) antibodies and rheumatoid factor, but little is known about the presence of other autoantibodies in this patient population. We investigated the prevalence and characteristics of anti-RA33 antibodies in patients with ICI-induced IA. METHODS Anti-RA33 ELISAs were performed on sera from four groups of patients: 79 with ICI-induced IA, 52 with rheumatoid arthritis (RA), 35 treated with ICIs without IA during follow-up and 50 healthy controls. Anti-RA33 positivity and level, clinical and demographic data were compared across groups. RESULTS Anti-RA33 antibodies were found in 9/79 (11.4%) patients with ICI-induced IA but in 0/35 patients treated with ICIs who did not develop IA (0%; p=0.04). Of the patients positive for anti-RA33, two had sera available from before ICI treatment; anti-RA33 antibodies were present in both pre-ICI treatments. In patients with RA, 7.7% were positive for anti-RA33 antibodies as were 2% of healthy controls. In ICI-induced IA, anti-RA33 antibodies were associated with anti-CCP antibodies (p=0.001). We found no statistically significant differences in other clinical characteristics in those with and without anti-RA33 antibodies. CONCLUSIONS Anti-RA33 antibodies are present in a subset of patients with ICI-induced IA, absent in other ICI-treated patients and may be a biomarker for developing IA. Additional studies evaluating serial samples before and after ICI treatment will further establish the temporal relationship of these antibodies to IA development.
Collapse
Affiliation(s)
- Laura C Cappelli
- Division of Rheumatology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Clifton O Bingham
- Division of Rheumatology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Patrick M Forde
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Valsamo Anagnostou
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Julie Brahmer
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Evan J Lipson
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Jennifer Mammen
- Division of Endocrinology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Megan Schollenberger
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Ami A Shah
- Division of Rheumatology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Erika Darrah
- Division of Rheumatology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
63
|
Bruera S, Suarez-Almazor ME. The effects of glucocorticoids and immunosuppressants on cancer outcomes in checkpoint inhibitor therapy. Front Oncol 2022; 12:928390. [PMID: 36081549 PMCID: PMC9445222 DOI: 10.3389/fonc.2022.928390] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 07/19/2022] [Indexed: 11/13/2022] Open
Abstract
The emergence of checkpoint inhibitors has created a paradigm shift for the treatment of various malignancies. However, although these therapies are associated with improved survival rates, they also carry the risk of immune-related adverse events (irAEs). Moderate to severe irAEs are typically treated with glucocorticoids, sometimes with the addition of immunosuppressants as steroid-sparing therapy. However, it is unclear how glucocorticoids and immunosuppressants may impact cancer survival and the efficacy of immune checkpoint therapy on cancer. In this narrative review, we discuss the effects of glucocorticoids and immunosuppressants including methotrexate, hydroxychloroquine, azathioprine, mycophenolate mofetil, tumor-necrosis factor (TNF)-inhibitors, interleukin-6 inhibitors, interleukin-1 inhibitors, abatacept, rituximab, and Janus kinase inhibitors (JAKi) on cancer-specific outcomes in the setting of immune checkpoint inhibitor use.
Collapse
Affiliation(s)
- Sebastian Bruera
- Section of Immunology, Allergy and Rheumatology, Baylor College of Medicine, Houston, TX, United States
| | - Maria E. Suarez-Almazor
- Department of Health Services Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- *Correspondence: Maria E. Suarez-Almazor,
| |
Collapse
|
64
|
Mirza J, Sunder SS, Karthikeyan B, Kattel S, Pokharel S, Quigley B, Sharma UC. Echocardiographic and Cardiac MRI Comparison of Longitudinal Strain and Strain Rate in Cancer Patients Treated with Immune Checkpoint Inhibitors. J Pers Med 2022; 12:1332. [PMID: 36013281 PMCID: PMC9410385 DOI: 10.3390/jpm12081332] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/10/2022] [Accepted: 08/16/2022] [Indexed: 01/07/2023] Open
Abstract
Background: Immune checkpoint inhibitor (ICI)-induced cardiac side effects in cancer patients are increasingly being recognized and can be fatal. There is no standardized cardiac imaging test to examine the effects of ICIs in myocardial morphology and function. Objective: To study the utility of echocardiography and cardiac MRI in examining regional and global changes arising from ICI-induced myocarditis and cardiomyopathy in high-risk subjects suspected to have developed ICI cardiomyopathy. Methods: We studied eight consecutive patients referred for cardiac MRI (CMR) from a comprehensive cancer center for suspected ICI-induced myocarditis and compared the data with sixteen age-matched controls. Using newly developed strain analysis algorithms, we measured myocardial strain and strain rates using echocardiography and CMR. Then, we compared the mean longitudinal strain and strain rates derived from echocardiography and CMR in the same ICI-treated cohort of patients (n = 8). They underwent both of these imaging studies with images taken 24−48 h apart and followed up prospectively within the same hospital course. Results: All our cases had preserved ejection fraction (EF) > 50%. Echocardiogram showed reduced mean systolic longitudinal strain (LS, %) (ICI: −12.381 ± 4.161; control: −19.761 ± 1.925; p < 0.001), peak systolic strain rate (SRS, s−1) (ICI: −0.597 ± 0.218; control: −0.947 ± 0.135; p = 0.002) and early diastolic strain rate (SRE, s−1) (ICI: 0.562 ± 0.295; control: 1.073 ± 0.228; p = 0.002) in ICI-treated cases. Direct comparison between the echocardiogram vs. CMR obtained within the same hospital course demonstrated strong a correlation of LS scores (r = 0.83, p = 0.012) and SRS scores (r = 0.71, p = 0.048). The Bland−Altman plots showed that 95% of the data points fitted within the ±1.96 SD of the mean difference, suggesting an agreement among these two imaging modalities. Conclusion: In this feasibility cohort study, both echocardiography- and CMR-based strain indices illustrate changes in myocardial contractility and relaxation suggestive of ICI-induced cardiomyopathy. Our data, after validation in a larger cohort, can form the basis of myocardial imaging in cancer patients treated with ICIs.
Collapse
Affiliation(s)
- Jibran Mirza
- Department of Medicine, Division of Cardiology, Jacob’s School of Medicine and Biomedical Sciences, Buffalo, NY 14203, USA
| | - Sunitha Shyam Sunder
- Department of Medicine, Division of Cardiology, Jacob’s School of Medicine and Biomedical Sciences, Buffalo, NY 14203, USA
| | - Badri Karthikeyan
- Department of Medicine, Division of Cardiology, Jacob’s School of Medicine and Biomedical Sciences, Buffalo, NY 14203, USA
| | - Sharma Kattel
- Department of Medicine, Division of Cardiology, Jacob’s School of Medicine and Biomedical Sciences, Buffalo, NY 14203, USA
| | - Saraswati Pokharel
- Department of Pathology and Laboratory Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA
| | - Brian Quigley
- Clinical and Research Institute on Addictions, University at Buffalo, Buffalo, NY 14203, USA
| | - Umesh C. Sharma
- Department of Medicine, Division of Cardiology, Jacob’s School of Medicine and Biomedical Sciences, Buffalo, NY 14203, USA
| |
Collapse
|
65
|
Delayed-Onset Inflammatory Optic Nerve Sheath Mass and Perineuritis Following Cessation of Ipilimumab and Nivolumab Therapy. Ophthalmic Plast Reconstr Surg 2022; 38:e119-e122. [DOI: 10.1097/iop.0000000000002170] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
66
|
Robles-Alonso V, Martínez-Valle F, Borruel N. Co Treatment With Biologic Agents and Immunotherapy in the Setting of irAEs of Difficult Management. Front Med (Lausanne) 2022; 9:906098. [PMID: 35847803 PMCID: PMC9279607 DOI: 10.3389/fmed.2022.906098] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/27/2022] [Indexed: 12/19/2022] Open
Abstract
In recent years, immunotherapy has become an important pillar of cancer treatment, with high response rates regardless of tumor histology or baseline mutations, sometime in patients without any alternative of treatment. Moreover, these treatments are moving from later line therapies to front-line therapies in the metastasic setting. However, immune activation associated with immune check-point inhibitors (ICI) is not selective and a large variety of immune-related adverse events, with an increasing frequency, have been associated with anti-PD1, anti-PD-1/L-1 and anti-CTLA-4 agents. In clinical trials, and sometimes also in real life practice, patients who develop severe toxicities on ICI-based therapies are usually not allowed to resume ICI once their disease progresses, because of the chance of developing severe irAEs on rechallenge with immunotherapies. Moreover, patients with irAEs suffer important side effects due to the high dose corticosteroids that are used to treat them. Therapy with ICI is sometimes the only alternative for certain patients, and for this reason co treatment with classic (DMARDS) or biologic immunosuppression therapy and ICI must be considered. Co-treatment with this type of immunosuppressant drugs, apart from allowing the maintenance of ICI therapy, drive to a lesser use of corticosteroids, with an improvement of the safety and quality of life of the patients. Such a tailored scheme of treatment is mostly an expert opinion based on recommendation and currently there is scarce evidence supporting it. Herein we present comprehensive, current recommendations and real-world data on the use of co-treatment with ICI and DMARDS and biologic immunosuppression.
Collapse
Affiliation(s)
- Virginia Robles-Alonso
- Crohn's and Colitis Attention Unit, Digestive System Service, Hospital Vall d'Hebron, Barcelona, Spain
| | - Fernando Martínez-Valle
- Systemic Autoimmune Diseases Unit, Internal Medicine Department, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- *Correspondence: Fernando Martínez-Valle
| | - Natalia Borruel
- Crohn's and Colitis Attention Unit, Digestive System Service, Hospital Vall d'Hebron, Barcelona, Spain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| |
Collapse
|
67
|
Gómez-Puerta JA, Lobo-Prat D, Perez-García C, Ponce A, Frade-sosa B, Millán Arciniegas AM, Ojeda F, Ruiz-Esquide V, Corominas H. Clinical Patterns and Follow-Up of Inflammatory Arthritis and Other Immune-Related Adverse Events Induced by Checkpoint Inhibitors. A Multicenter Study. Front Med (Lausanne) 2022; 9:888377. [PMID: 35783644 PMCID: PMC9240301 DOI: 10.3389/fmed.2022.888377] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 05/12/2022] [Indexed: 01/04/2023] Open
Abstract
Objectives To describe different clinical patterns of rheumatic immune-related adverse events (irAEs) induced by immune checkpoint inhibitors (ICI) and their rheumatic and oncologic outcomes. Methods We classified clinical syndromes according to five different categories: non-inflammatory arthralgias (NIA), rheumatoid arthritis (RA)-like, psoriatic arthritis (PsA)-like, polymyalgia rheumatica (PMR)-like, and a miscellaneous group of patients with other syndromes. We conducted a baseline visit and then follow-up in order to determine their clinical pattern, treatment response, and outcome. Results We included 73 patients (64% male) with a mean age of 66.1 ± 11.6 years. Main underlying diagnosis was lung carcinoma in 29 (39%) patients, melanoma in 20 (27%), and renal-urothelial cancer in 11 (15%). Main ICI included Pembrolizumab in 24 (32%), Nivolumab 17 (23%), and Atezolizumab 7 (9 %). Seventeen out of seventy-three patients had an underlying rheumatic disease before ICI treatment. Fourteen patients developed other irAEs before or simultaneously with rheumatic syndromes. Main rheumatic irAEs included: RA-like in 31 (42.4%), NIA in 19 (26.0%), PMR-like in 10 (13.7%), and PsA-like in 5 (6.8%), among others. Median time from ICI to irAEs was 5 months (IQR 3–9). Those patients who received combined therapy, had a trend for an earlier presentation than those who received monotherapy (4.3 months IQR 1.85–17 vs. 6 months IQR 3–9.25, p = NS). Mean follow-up time was 14.0 ± 10.8 (SD, months). At the last visit, 47 % were taking glucocorticoids and 11% DMARD therapy. At the last visit, 13 (17.8%) patients remained with persistent arthritis, 19 (26%) had intermittent flares, and 39 (53.4%) had a self-limited pattern. Only in 15.1% of patients ICI therapy was discontinued. Conclusions We described different patterns according to treatment and irAEs. Combined ICI therapy had an earlier onset of symptoms. Patients who presented as RA-like, had a higher risk of persistent arthritis. After a mean follow-up of more than 1 year, one-fifth of the patients remained with persistent arthritis and 11% required DMARD therapy.
Collapse
Affiliation(s)
- José A. Gómez-Puerta
- Rheumatology Department, Hospital Clinic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Department of Medicine, Universitat de Barcelona, Barcelona, Spain
- *Correspondence: José A. Gómez-Puerta
| | - David Lobo-Prat
- Rheumatology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | | | - Andrés Ponce
- Rheumatology Department, Hospital Clinic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Beatriz Frade-sosa
- Rheumatology Department, Hospital Clinic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | | | - Fabiola Ojeda
- Rheumatology Department, Hospital del Mar, Barcelona, Spain
| | - Virginia Ruiz-Esquide
- Rheumatology Department, Hospital Clinic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Hector Corominas
- Rheumatology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| |
Collapse
|
68
|
Lauwyck J, Schreuer M, Meric de Bellefon L, Van Erps J, Neyns B, Aspeslagh S. Severe treatment-induced inflammatory polyarthritis in advanced melanoma patients: 2 case reports. Melanoma Res 2022; 32:200-204. [PMID: 35377860 DOI: 10.1097/cmr.0000000000000814] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Immune checkpoint inhibitors (ICI) and targeted therapies form the therapeutic mainstay for v-Raf murine sarcoma viral oncogene homolog B V600-mutated metastatic melanoma. Both treatment regimens can cause inflammatory arthritis. The reported incidence of treatment-induced inflammatory arthritis is low, though presumably underestimated due to lack of awareness, clear definitions and uniform grading systems. Nevertheless, recognition is important as inflammatory arthritis can become chronic and thus affect the quality of life beyond treatment. In this short communication, we present two patients with metastatic melanoma treated with ICI and targeted therapies who develop severe polyarthritis. Based on their clinical discourse we describe standard inflammatory arthritis treatment modalities and more advanced immunomodulatory treatment options with conventional synthetic disease-modifying antirheumatic drugs (csDMARDs) or biologic DMARDs (bDMARDs). Long-term immunosuppressive treatment with glucocorticoids or DMARDs in this setting raises concerns about antitumour response and potential carcinogenic risk. Current literature on this topic is scarce, heterogeneous and retrospective. Prospective analysis of cancer patients treated with DMARDs is needed to clearly address these concerns.
Collapse
Affiliation(s)
- Justine Lauwyck
- Department of Medical Oncology, Universitair Ziekenhuis (UZ) Brussel, Laarbeeklaan, Brussel
| | - Max Schreuer
- Department of Medical Oncology, Algemeen Stedelijk Ziekenhuis (ASZ) Aalst, Merestraat, Aalst
| | - Laurent Meric de Bellefon
- Department of Rheumatology, Universitair Medisch Centrum (UMC) Sint-Pieter, Hoogstraat, Brussel, Belgium
| | - Joanna Van Erps
- Department of Medical Oncology, Algemeen Stedelijk Ziekenhuis (ASZ) Aalst, Merestraat, Aalst
| | - Bart Neyns
- Department of Medical Oncology, Universitair Ziekenhuis (UZ) Brussel, Laarbeeklaan, Brussel
| | - Sandrine Aspeslagh
- Department of Medical Oncology, Universitair Ziekenhuis (UZ) Brussel, Laarbeeklaan, Brussel
| |
Collapse
|
69
|
Chan KK, Bass AR. Monitoring and Management of the Patient with Immune Checkpoint Inhibitor-Induced Inflammatory Arthritis: Current Perspectives. J Inflamm Res 2022; 15:3105-3118. [PMID: 35642215 PMCID: PMC9148583 DOI: 10.2147/jir.s282600] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 03/18/2022] [Indexed: 11/23/2022] Open
Abstract
In this review, we draw from observational studies, treatment guidelines and our own clinical experience to describe approaches to monitoring and management of immune checkpoint inhibitor (ICI)-induced inflammatory arthritis, including polymyalgia rheumatica. This condition occurs in about 4% of ICI-treated cancer patients and can persist for a year or longer. Mild arthritis can generally be managed with non-steroidal anti-inflammatory drugs, intraarticular steroids injections and/or low dose corticosteroids. Higher grade arthritis should be brought under control with corticosteroids, but early introduction of a steroid-sparing agent is recommended to minimize steroid toxicity. In order to assess the effectiveness of any arthritis treatment, tender and swollen joint counts and patient reported measures of physical function, such as the health assessment questionnaire, should be obtained at each visit. Referral to a rheumatologist is recommended for patients with high grade arthritis to help guide the use of disease-modifying antirheumatic drugs.
Collapse
Affiliation(s)
- Karmela K Chan
- Department of Medicine, Hospital for Special Surgery/Weill Cornell Medicine, New York, NY, USA
| | - Anne R Bass
- Department of Medicine, Hospital for Special Surgery/Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
70
|
Zhang D, Deng T, Yuan W, Chen T, Jiang S. Glaucocalyxin A induces apoptosis of NSCLC cells by inhibiting the PI3K/Akt/GSK3β pathway. Clin Exp Pharmacol Physiol 2022; 49:797-804. [PMID: 35576104 DOI: 10.1111/1440-1681.13667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 04/27/2022] [Accepted: 05/08/2022] [Indexed: 11/28/2022]
Abstract
Lung cancer is one of the fastest growing malignancies in morbidity and mortality, and current therapies are in general not sufficiently effective for this deadly disease. This study characterizes the anti-cancer effects of Glaucocalyxin A (GLA) and explores the underlying mechanisms using human non-small cell lung carcinoma (NSCLC) cells. First, our data showed that GLA suppressed the viability of cancer cells, while no effect was observed in the normal bronchial epithelial cell Bease 2B cells. Second, GLA inhibited colony formation, induced apoptosis of cancer cells. Third, GLA down-regulated the expression of B-cell lymphoma-2 (Bcl-2) protein, up-regulated the expression of Bcl2-associated X protein (Bax) , and strengthened cleavage of Caspase-3 and poly ADP-ribose polymerase (PARP). Fourth, GLA also diminished mitochondrial membrane potential and inhibited phosphatidylinositol 3-kinase (PI3K)/Akt/ glycogen synthase kinase-3β (GSK3β) pathway. In addition, injection of GLA (20 mg/kg) every two days significantly inhibited A549 xenograft tumor growth, accompanied by increased apoptosis and decreased proliferation. Together, our study provides evidence that the anticancer effect of GLA in NSCLC is mediated by inducing apoptosis through inhibiting PI3K/Akt/GSK3β pathway and suggests that GLA may be used as a promising natural medicine for NSCLC therapy. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- De Zhang
- School of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Ting Deng
- School of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Wa Yuan
- School of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Tongqiang Chen
- School of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Shuping Jiang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China.,School of Basic Medicine, Gannan Medical University, Ganzhou, China.,Key Laboratory of Biomaterials and Bio-fabrication in Tissue Engineering of Jiangxi Province, Ganzhou, China
| |
Collapse
|
71
|
Subesinghe M, Bhuva S, Arumalla N, Cope A, D’Cruz D, Subesinghe S. 2-deoxy-2[18F]fluoro-D-glucose positron emission tomography-computed tomography in rheumatological diseases. Rheumatology (Oxford) 2022; 61:1769-1782. [PMID: 34463703 PMCID: PMC9071551 DOI: 10.1093/rheumatology/keab675] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 08/18/2021] [Accepted: 08/25/2021] [Indexed: 12/26/2022] Open
Abstract
2-deoxy-2[18F]fluoro-D-glucose (FDG) PET-CT has revolutionized oncological imaging. The cellular processes that make cancer cells visible on FDG PET-CT also occur in a number of inflammatory cells. Exploiting this phenomenon has led to a growth of evidence supporting the use of FDG PET-CT in a wide range of infective and inflammatory diseases. Rheumatological diseases can affect multiple sites within the musculoskeletal system alongside multi-organ extra-articular disease manifestations. Inflammation is central to these diseases, making FDG PET-CT a logical choice. In this review article we describe the various applications of FDG PET-CT in rheumatological diseases using illustrative examples to highlight the beneficial role of FDG PET-CT in each case.
Collapse
Affiliation(s)
- Manil Subesinghe
- King’s College London & Guy’s and St. Thomas’ PET Centre, Guy’s and St. Thomas’ NHS Foundation Trust
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, Faculty of Life Sciences and Medicine, King’s College London
| | - Shaheel Bhuva
- King’s College London & Guy’s and St. Thomas’ PET Centre, Guy’s and St. Thomas’ NHS Foundation Trust
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, Faculty of Life Sciences and Medicine, King’s College London
| | - Nikita Arumalla
- Department of Rheumatology, Guy’s and St. Thomas’ NHS Foundation Trust
| | - Andrew Cope
- Department of Rheumatology, Guy’s and St. Thomas’ NHS Foundation Trust
- Centre for Rheumatic Diseases, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, UK
| | - David D’Cruz
- Department of Rheumatology, Guy’s and St. Thomas’ NHS Foundation Trust
| | - Sujith Subesinghe
- Department of Rheumatology, Guy’s and St. Thomas’ NHS Foundation Trust
| |
Collapse
|
72
|
Zhong Z, Vong CT, Chen F, Tan H, Zhang C, Wang N, Cui L, Wang Y, Feng Y. Immunomodulatory potential of natural products from herbal medicines as immune checkpoints inhibitors: Helping to fight against cancer via multiple targets. Med Res Rev 2022; 42:1246-1279. [PMID: 35028953 PMCID: PMC9306614 DOI: 10.1002/med.21876] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 12/03/2021] [Accepted: 12/15/2021] [Indexed: 12/19/2022]
Abstract
Immunotherapy sheds new light to cancer treatment and is satisfied by cancer patients. However, immunotoxicity, single-source antibodies, and single-targeting stratege are potential challenges to the success of cancer immunotherapy. A huge number of promising lead compounds for cancer treatment are of natural origin from herbal medicines. The application of natural products from herbal medicines that have immunomodulatory properties could alter the landscape of immunotherapy drastically. The present study summarizes current medication for cancer immunotherapy and discusses the potential chemicals from herbal medicines as immune checkpoint inhibitors that have a broad range of immunomodulatory effects. Therefore, this review provides valuable insights into the efficacy and mechanism of actions of cancer immunotherapies, including natural products and combined treatment with immune checkpoint inhibitors, which could confer an improved clinical outcome for cancer treatment.
Collapse
Affiliation(s)
- Zhangfeng Zhong
- School of Chinese MedicineThe University of Hong KongPokfulamHong KongChina
- Macau Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical SciencesUniversity of MacauTaipaMacao SARChina
| | - Chi Teng Vong
- Macau Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical SciencesUniversity of MacauTaipaMacao SARChina
| | - Feiyu Chen
- School of Chinese MedicineThe University of Hong KongPokfulamHong KongChina
| | - Horyue Tan
- School of Chinese MedicineThe University of Hong KongPokfulamHong KongChina
| | - Cheng Zhang
- School of Chinese MedicineThe University of Hong KongPokfulamHong KongChina
| | - Ning Wang
- School of Chinese MedicineThe University of Hong KongPokfulamHong KongChina
| | - Liao Cui
- Guangdong Key Laboratory for Research and Development of Natural DrugsGuangdong Medical UniversityZhanjiangGuangdongChina
| | - Yitao Wang
- Macau Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical SciencesUniversity of MacauTaipaMacao SARChina
| | - Yibin Feng
- School of Chinese MedicineThe University of Hong KongPokfulamHong KongChina
| |
Collapse
|
73
|
Ma VT, Lao CD, Fecher LA, Schiopu E. Successful use of secukinumab in two melanoma patients with immune checkpoint inhibitor-induced inflammatory arthropathy. Immunotherapy 2022; 14:593-598. [PMID: 35416067 DOI: 10.2217/imt-2021-0274] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Immune-related adverse events (irAEs) are a major concern when treating cancer patients with immune checkpoint inhibitor (ICI) therapy. Selecting the most appropriate management of irAEs remains an ongoing challenge because prolonged use of glucocorticoids come with their own side effects and may counteract the antineoplastic effects from immunotherapy. In this case report, we present two patients with metastatic melanoma who developed symptoms of inflammatory arthritis attributed to ICI therapy. We found that treatment with secukinumab, an anti-IL-17A inhibitor, effectively managed their symptoms and did not lead to tumor progression. Our study suggests that secukinumab can be a safe and effective treatment option for ICI-induced inflammatory arthropathy.
Collapse
Affiliation(s)
- Vincent T Ma
- Department of Internal Medicine, University of Wisconsin, Division of Hematology, Medical Oncology and Palliative Care, Madison, WI, USA.,Department of Internal Medicine, University of Michigan, Division of Hematology and Oncology, Ann Arbor, MI, USA
| | - Christopher D Lao
- Department of Internal Medicine, University of Michigan, Division of Hematology and Oncology, Ann Arbor, MI, USA
| | - Leslie A Fecher
- Department of Internal Medicine, University of Michigan, Division of Hematology and Oncology, Ann Arbor, MI, USA
| | - Elena Schiopu
- Department of Internal Medicine, University of Michigan, Division of Rheumatology, Ann Arbor, MI, USA
| |
Collapse
|
74
|
Kim ST, Chu Y, Misoi M, Suarez-Almazor ME, Tayar JH, Lu H, Buni M, Kramer J, Rodriguez E, Hussain Z, Neelapu SS, Wang J, Shah AY, Tannir NM, Campbell MT, Gibbons DL, Cascone T, Lu C, Blumenschein GR, Altan M, Lim B, Valero V, Loghin ME, Tu J, Westin SN, Naing A, Garcia-Manero G, Abdel-Wahab N, Tawbi HA, Hwu P, Oliva ICG, Davies MA, Patel SP, Zou J, Futreal A, Diab A, Wang L, Nurieva R. Distinct molecular and immune hallmarks of inflammatory arthritis induced by immune checkpoint inhibitors for cancer therapy. Nat Commun 2022; 13:1970. [PMID: 35413951 PMCID: PMC9005525 DOI: 10.1038/s41467-022-29539-3] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 03/17/2022] [Indexed: 12/12/2022] Open
Abstract
Immune checkpoint inhibitors are associated with immune-related adverse events (irAEs), including arthritis (arthritis-irAE). Management of arthritis-irAE is challenging because immunomodulatory therapy for arthritis should not impede antitumor immunity. Understanding of the mechanisms of arthritis-irAE is critical to overcome this challenge, but the pathophysiology remains unknown. Here, we comprehensively analyze peripheral blood and/or synovial fluid samples from 20 patients with arthritis-irAE, and unmask a prominent Th1-CD8+ T cell axis in both blood and inflamed joints. CX3CR1hi CD8+ T cells in blood and CXCR3hi CD8+ T cells in synovial fluid, the most clonally expanded T cells, significantly share TCR repertoires. The migration of blood CX3CR1hi CD8+ T cells into joints is possibly mediated by CXCL9/10/11/16 expressed by myeloid cells. Furthermore, arthritis after combined CTLA-4 and PD-1 inhibitor therapy preferentially has enhanced Th17 and transient Th1/Th17 cell signatures. Our data provide insights into the mechanisms, predictive biomarkers, and therapeutic targets for arthritis-irAE.
Collapse
Affiliation(s)
- Sang T Kim
- Section of Rheumatology and Clinical Immunology, Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yanshuo Chu
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Mercy Misoi
- Department of General Internal Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Maria E Suarez-Almazor
- Section of Rheumatology and Clinical Immunology, Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jean H Tayar
- Section of Rheumatology and Clinical Immunology, Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Huifang Lu
- Section of Rheumatology and Clinical Immunology, Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Maryam Buni
- Section of Rheumatology and Clinical Immunology, Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jordan Kramer
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Department of Biology, Georgetown University, Washington, DC, 20057, USA
| | - Emma Rodriguez
- Department of Infectious Disease, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Zulekha Hussain
- Section of Rheumatology and Clinical Immunology, Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Sattva S Neelapu
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jennifer Wang
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Amishi Y Shah
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Nizar M Tannir
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Matthew T Campbell
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Don L Gibbons
- Department of Thoracic Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Tina Cascone
- Department of Thoracic Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Charles Lu
- Department of Thoracic Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - George R Blumenschein
- Department of Thoracic Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Mehmet Altan
- Department of Thoracic Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Bora Lim
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Vincente Valero
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Monica E Loghin
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Janet Tu
- Department of General Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Shannon N Westin
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Aung Naing
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Guillermo Garcia-Manero
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Noha Abdel-Wahab
- Section of Rheumatology and Clinical Immunology, Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Department of Rheumatology and Rehabilitation, Assiut University Hospitals, Faculty of Medicine, Assiut University, El Fateh, Egypt
| | - Hussein A Tawbi
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Patrick Hwu
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Isabella C Glitza Oliva
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Michael A Davies
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Sapna P Patel
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jun Zou
- Department of Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Andrew Futreal
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Adi Diab
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Linghua Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences (GSBS), Houston, TX, 77030, USA.
| | - Roza Nurieva
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences (GSBS), Houston, TX, 77030, USA.
| |
Collapse
|
75
|
Abstract
Immune checkpoint inhibitors activate the immune system to combat cancer. In doing so, however, they can cause immune-related adverse events (irAEs), including rheumatic syndromes, such as inflammatory arthritis, polymyalgia rheumatica, and myositis. This article reviews rheumatic irAEs that may be encountered in the general medicine practice and provides guidance to support prompt recognition, referral, and treatment of these patients.
Collapse
Affiliation(s)
- Nilasha Ghosh
- Hospital for Special Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Anne R Bass
- Hospital for Special Surgery, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
76
|
Gudi RR, Perez N, Karumuthil-Melethil S, Li G, Vasu C. Activation of T cell checkpoint pathways during β-cell antigen presentation by engineered dendritic cells promotes protection from type 1 diabetes. Immunology 2022; 166:341-356. [PMID: 35404483 DOI: 10.1111/imm.13476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/27/2021] [Accepted: 03/28/2022] [Indexed: 11/29/2022] Open
Abstract
Defective immune regulation has been recognized in type 1 diabetes (T1D). Immune regulatory T cell check-point receptors, which are generally upregulated on activated T cells, have been the molecules of attention as therapeutic targets for enhancing immune response in tumor therapy. Here, we show that pancreatic β-cell antigen (BcAg) presentation by engineered tolerogenic dendritic cells (tDCs) that express CTLA4 selective ligand (B7.1wa) or a combination of CTLA4, PD1 and BTLA selective ligands (B7.1wa, PD-L1, and HVEM-CRD1 respectively; multiligand-DCs) causes an increase in regulatory cytokine and T cell (Treg) responses and suppression of the effector T cell function as compared to engineered control-DCs. Non-obese diabetic (NOD) mice treated with BcAg-pulsed CTLA4-ligand-DCs and multiligand-DCs at pre-diabetic and early-hyperglycemic stages showed significantly lower degree of insulitis, higher frequencies of insulin-positive islets, profound delay in, and reversal of, hyperglycemia for a significant duration. Immune cells from the tDC treated mice not only produced lower amounts of IFNγ and higher amounts of IL10 and TGFβ1 upon BcAg challenge, but also failed to induce hyperglycemia upon adoptive transfer. While both CTLA4-ligand-DCs and multiligand-DCs were effective in inducing tolerance, multiligand-DC treatment produced an overall higher suppressive effect on effector T cell function and disease outcome. These studies show that enhanced engagement of T cell checkpoint receptors during BcAg presentation can modulate T cell function and suppress autoimmunity and progression of the disease in T1D.
Collapse
Affiliation(s)
- Radhika R Gudi
- Department of Microbiology and Immunology, College of Medicine, Medical University of South Carolina, Charleston
| | - Nicolas Perez
- Department of Surgery, College of Medicine, University of Illinois, Chicago, IL
| | | | - Gongbo Li
- Department of Surgery, College of Medicine, University of Illinois, Chicago, IL
| | - Chenthamarakshan Vasu
- Department of Microbiology and Immunology, College of Medicine, Medical University of South Carolina, Charleston.,Department of Surgery, College of Medicine, University of Illinois, Chicago, IL
| |
Collapse
|
77
|
Jensen AK, Chatzidionysiou K, Torp CK, Sørensen AS, Tenstad HB, Schäfer VS, Kostine M, Jacobsen S, Leipe J, Kragstrup TW. Comparison of immune checkpoint inhibitor-induced arthritis and reactive arthritis to inform therapeutic strategy. Biomed Pharmacother 2022; 148:112687. [PMID: 35228067 DOI: 10.1016/j.biopha.2022.112687] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/31/2022] [Accepted: 02/01/2022] [Indexed: 11/15/2022] Open
Abstract
INTRODUCTION Immune checkpoint inhibitor-induced inflammatory arthritis (ICI-IA) is a relatively new disease entity caused by ICI agents during cancer therapy. Reactive arthritis (ReA) is a well-known disease entity caused by urogenital or gastrointestinal bacterial infection or pneumonia. In this sense, ICI-IA and ReA are both defined by a reaction to a well-specified causal event. As a result, comparing these diseases may help to determine therapeutic strategies. METHODS We compared ICI-IA and ReA with special focus on pharmacological management. Specifically regarding treatment, we conducted a literature search of studies published in the PubMed database. Inclusion criteria were studies on treatment with non-steroidal anti-inflammatory drugs (NSAIDs), glucocorticoids (GC), or disease modifying antirheumatic drugs (DMARDs) in ICI-IA or ReA. During systematic selection, 21 studies evaluating ICI-IA and 14 studies evaluating ReA were included. RESULTS In ICI-IA, prospective and retrospective studies have shown effects of non-steroidal anti-inflammatory drugs (NSAIDs), glucocorticoid (GC), sulfasalazine (SSZ), methotrexate (MTX), hydroxychloroquine (HCQ) and TNFi. In ReA, retrospective studies evaluated NSAIDs and GC. A randomized controlled trial reported the effect of SSZ, and a retrospective study reported the effect of MTX and SSZ in combination with tumor necrosis factor alpha inhibition (TNFi). For both entities, small case reports show treatment effects of interleukin 6 receptor inhibition (IL-6Ri). DISCUSSION This literature review identified both similarities and differences regarding the pathogenesis and clinical features of ReA and ICI-IA. Studies on treatment reported effectiveness of NSAIDs, GC, MTX, SSZ and TNFi in both diseases. Further, small case reports showed effects of IL-6Ri.
Collapse
Affiliation(s)
- Anders Kirkegaard Jensen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Department of Neurology, Sygehus Lillebælt, Kolding, Denmark
| | - Katerina Chatzidionysiou
- Department of Medicine Solna, Karolinska Institutet, Rheumatology Unit, Karolinska University Hospital, Stockholm, Sweden
| | | | | | | | - Valentin S Schäfer
- Clinic of Internal Medicine III, Department of Oncology, Hematology, Rheumatology and Clinical Immunology, University Hospital Bonn, Bonn, Germany
| | - Marie Kostine
- Department of Rheumatology, Bordeaux, University, Hospital, France
| | - Søren Jacobsen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, Copenhagen, Denmark; Department of Rheumatology, Rigshospitalet, Copenhagen, Denmark
| | - Jan Leipe
- Division of Rheumatology, Department of Medicine V, University Hospital Mannheim, Medical Faculty Mannheim of the University of Heidelberg, Germany.
| | - Tue Wenzel Kragstrup
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark.
| |
Collapse
|
78
|
Poto R, Troiani T, Criscuolo G, Marone G, Ciardiello F, Tocchetti CG, Varricchi G. Holistic Approach to Immune Checkpoint Inhibitor-Related Adverse Events. Front Immunol 2022; 13:804597. [PMID: 35432346 PMCID: PMC9005797 DOI: 10.3389/fimmu.2022.804597] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 03/07/2022] [Indexed: 12/12/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) block inhibitory molecules, such as cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), programmed cell death protein 1 (PD-1), or its ligand, programmed cell death protein ligand 1 (PD-L1) and enhance antitumor T-cell activity. ICIs provide clinical benefits in a percentage of patients with advanced cancers, but they are usually associated with a remarkable spectrum of immune-related adverse events (irAEs) (e.g., rash, colitis, hepatitis, pneumonitis, endocrine, cardiac and musculoskeletal dysfunctions). Particularly patients on combination therapy (e.g., anti-CTLA-4 plus anti-PD-1/PD-L1) experience some form of irAEs. Different mechanisms have been postulated to explain these adverse events. Host factors such as genotype, gut microbiome and pre-existing autoimmune disorders may affect the risk of adverse events. Fatal ICI-related irAEs are due to myocarditis, colitis or pneumonitis. irAEs usually occur within the first months after ICI initiation but can develop as early as after the first dose to years after ICI initiation. Most irAEs resolve pharmacologically, but some appear to be persistent. Glucocorticoids represent the mainstay of management of irAEs, but other immunosuppressive drugs can be used to mitigate refractory irAEs. In the absence of specific trials, several guidelines, based on data from retrospective studies and expert consensus, have been published to guide the management of ICI-related irAEs.
Collapse
Affiliation(s)
- Remo Poto
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Teresa Troiani
- Medical Oncology, Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Gjada Criscuolo
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | | | - Fortunato Ciardiello
- Medical Oncology, Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | | | - Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
- World Allergy Organization (WAO) Center of Excellence, Naples, Italy
- Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council, Naples, Italy
| |
Collapse
|
79
|
Yaseen MM, Abuharfeil NM, Darmani H. CMTM6 as a master regulator of PD-L1. Cancer Immunol Immunother 2022; 71:2325-2340. [PMID: 35294592 DOI: 10.1007/s00262-022-03171-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 02/08/2022] [Indexed: 12/15/2022]
Abstract
Immune checkpoint proteins, such as programmed cell death receptor 1 (PD-1) and its ligand (PD-L1), play critical roles in the pathology of chronic inflammatory pathological conditions, particularly cancer. In addition, the activation of PD-1/PD-L1 pathway is involved in mediating resistance to certain anti-cancer chemo- and immuno-therapeutics. Unfortunately, targeting the PD-1/PD-L1 pathway by the available anti-PD-1/PD-L1 drugs can benefit only a small proportion of cancer patients. Thus, studying the factors that regulate the expression of these immune checkpoint proteins is of central importance in this context. Recent investigations have identified CMTM6 and, to a lesser extent, CMTM4, as master regulators of PD-L1 expression in various cancer cells. Understanding the mechanisms by which such proteins upregulate the expression of PD-L1 in tumor cells, and determining the potential regulators of CMTM6 expression in different types of cancers will accelerate the development of new therapeutic targets and/or lead to the enhancement of the currently available PD-1/PD-L1 blockade therapies.
Collapse
Affiliation(s)
- Mahmoud Mohammad Yaseen
- Department of Biotechnology and Genetic Engineering, Faculty of Science and Arts, Jordan University of Science and Technology, Irbid, 22110, Jordan.
| | - Nizar Mohammad Abuharfeil
- Department of Biotechnology and Genetic Engineering, Faculty of Science and Arts, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Homa Darmani
- Department of Biotechnology and Genetic Engineering, Faculty of Science and Arts, Jordan University of Science and Technology, Irbid, 22110, Jordan
| |
Collapse
|
80
|
Martin de Fremont G, Belkhir R, Henry J, Voisin AL, Lambotte O, Besson FL, Mariette X, Nocturne G. Features of polymyalgia rheumatica-like syndrome after immune checkpoint inhibitor therapy. Ann Rheum Dis 2022; 81:e52. [PMID: 32188691 DOI: 10.1136/annrheumdis-2020-217225] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 02/28/2020] [Indexed: 11/04/2022]
Affiliation(s)
| | - Rakiba Belkhir
- Rheumatology, AP-HP, Université Paris Saclay, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Julien Henry
- Rheumatology, AP-HP, Université Paris Saclay, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Anne Laure Voisin
- Unité Fonctionnelle de Pharmacovigilance, Gustave Roussy, F-94800, Villejuif, France
| | - Olivier Lambotte
- Médecine Interne et Immunologie clinique, AP-HP.Université Paris Saclay, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
- Université Paris-Saclay, INSERM, CEA, Centre de recherche en Immunologie des infections virales et des maladies auto-immunes, Le Kremlin-Bicêtre, France
| | - Florent L Besson
- Biophysics and Nuclear Medicine, AP-HP.Université Paris Saclay, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
- UMR BioMaps, Inserm/CNRS/CEA/Université Paris-Saclay, Orsay, France
| | - Xavier Mariette
- Rheumatology, AP-HP, Université Paris Saclay, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
- Université Paris-Saclay, INSERM, CEA, Centre de recherche en Immunologie des infections virales et des maladies auto-immunes, Le Kremlin-Bicêtre, France
| | - Gaetane Nocturne
- Rheumatology, AP-HP, Université Paris Saclay, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
- Université Paris-Saclay, INSERM, CEA, Centre de recherche en Immunologie des infections virales et des maladies auto-immunes, Le Kremlin-Bicêtre, France
| |
Collapse
|
81
|
Cunningham-Bussel A, Wang J, Prisco LC, Martin LW, Vanni KM, Zaccardelli A, Nasrallah M, Gedmintas L, MacFarlane LA, Shadick NA, Awad MM, Rahma O, LeBoeuf NR, Gravallese EM, Sparks JA. Predictors of Rheumatic Immune-Related Adverse Events and De Novo Inflammatory Arthritis After Immune Checkpoint Inhibitor Treatment for Cancer. Arthritis Rheumatol 2022; 74:527-540. [PMID: 34397169 PMCID: PMC8847547 DOI: 10.1002/art.41949] [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: 05/08/2021] [Revised: 07/29/2021] [Accepted: 08/05/2021] [Indexed: 11/06/2022]
Abstract
OBJECTIVE To identify predictors of rheumatic immune-related adverse events (irAEs) following immune checkpoint inhibitor (ICI) treatment for cancer. METHODS We performed a case-control study to predict the occurrence of rheumatic irAEs in cancer patients who initiated ICI treatment at Mass General Brigham and the Dana-Farber Cancer Institute between 2011 and 2020. We screened for the presence of rheumatic irAEs by reviewing the medical records of patients evaluated by rheumatologists or those prescribed nonglucocorticoid immunomodulatory drugs after the time of ICI initiation (baseline). Review of medical records confirmed the presence of rheumatic irAEs and the indications necessitating immunomodulatory drug treatment. Controls were defined as patients who did not experience rheumatic irAEs, did not have preexisting rheumatic disease, did not have a clinical evaluation by a rheumatologist after ICI treatment, did not receive an immunomodulatory drug after ICI, did not receive systemic glucocorticoids after ICI, and survived at least 6 months after the initial ICI treatment. We used logistic regression to estimate the odds ratios (ORs) (with 95% confidence intervals [95% CIs]) for the risk of a rheumatic irAE in the presence of various baseline predictors. RESULTS A total of 8,028 ICI recipients were identified (mean age 65.5 years, 43.1% female, 31.8% with lung cancer). After ICI initiation, 404 patients (5.0%) were evaluated by rheumatologists, and 475 patients (5.9%) received an immunomodulatory drug to treat any irAEs. There were 226 confirmed rheumatic irAE cases (2.8%) and 118 de novo inflammatory arthritis cases (1.5%). Rheumatic diseases (either preexisting rheumatic diseases or rheumatic irAEs) were a common indication for immunomodulatory drug use (27.9%). Baseline predictors of rheumatic irAEs included melanoma (multivariable OR 4.06 [95% CI 2.54-6.51]) and genitourinary (GU) cancer (OR 2.22 [95% CI 1.39-3.54]), both relative to patients with lung cancer; combination ICI treatment (OR 2.35 [95% CI 1.48-3.74]), relative to patients receiving programmed death 1 inhibitor monotherapy; autoimmune disease (OR 2.04 [95% CI 1.45-2.85]) and recent glucocorticoid use (OR 2.13 [95% CI 1.51-2.98]), relative to patients not receiving a glucocorticoid, compared to the 2,312 controls without rheumatic irAEs. Predictors of de novo inflammatory arthritis were similar to those of rheumatic irAEs. CONCLUSION We identified novel predictors of rheumatic irAE development in cancer patients, including baseline presence of melanoma, baseline presence of GU tract cancer, preexisting autoimmune disease, receiving or having received combination ICI treatment, and receiving or having received glucocorticoids. The proportion of cancer patients experiencing rheumatic irAEs may be even higher than was reported in the present study, since we used stringent criteria to identify cases of rheumatic irAEs. Our findings could be used to identify cancer patients at risk of developing rheumatic irAEs and de novo inflammatory arthritis and may help further elucidate the pathogenesis of rheumatic irAEs in patients with cancer who are receiving ICI treatment.
Collapse
Affiliation(s)
- Amy Cunningham-Bussel
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Jiaqi Wang
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital, Boston, MA, USA
| | - Lauren C. Prisco
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital, Boston, MA, USA
| | - Lily W. Martin
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital, Boston, MA, USA
| | - Kathleen M.M. Vanni
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital, Boston, MA, USA
| | - Alessandra Zaccardelli
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital, Boston, MA, USA
| | - Mazen Nasrallah
- Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Boston, MA, United States
| | - Lydia Gedmintas
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Lindsey A. MacFarlane
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Nancy A. Shadick
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Mark M. Awad
- Harvard Medical School, Boston, MA, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Osama Rahma
- Harvard Medical School, Boston, MA, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Nicole R. LeBoeuf
- Harvard Medical School, Boston, MA, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Dermatology, Brigham and Women’s Hospital
| | - Ellen M. Gravallese
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Jeffrey A. Sparks
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
82
|
Aboo C, Krastrup TW, Tenstad HB, Ren J, Just SA, Ladekarl M, Stensballe A. Prediction and early diagnosis of immune-checkpoint inhibitor-induced inflammatory arthritis from molecular biomarkers – Where are we now? EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2022. [DOI: 10.1080/23808993.2022.2156785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Christopher Aboo
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
- Sino-Danish Center for Education and Research, University of Chinese Academy of Sciences, Beijing, China
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Beijing, China
| | - Tue Wenzel Krastrup
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Jie Ren
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Beijing, China
| | | | - Morten Ladekarl
- Department of Oncology and Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - Allan Stensballe
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| |
Collapse
|
83
|
Cappelli LC, Bingham CO, Braaten T, Shah AA. Response to: Correspondence on "Immune checkpoint inhibitor-induced inflammatory arthritis persists after immunotherapy cessation" by Braaten et al. Ann Rheum Dis 2022; 81:e14. [PMID: 31959594 PMCID: PMC7369218 DOI: 10.1136/annrheumdis-2019-216892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 01/09/2020] [Accepted: 01/09/2020] [Indexed: 01/03/2023]
Affiliation(s)
- Laura C Cappelli
- Rheumatology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Clifton O Bingham
- Rheumatology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Tawnie Braaten
- Rheumatology, University of Utah, Salt Lake City, Utah, USA
| | - Ami A Shah
- Rheumatology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
84
|
Ceccarelli F, Botticelli A, Gelibter AJ, Leccese I, Lucchetti R, Cortesi E, Valesini G, Marchetti P, Conti F. Correspondence on 'Immune checkpoint inhibitor-induced inflammatory arthritis persists after immunotherapy cessation' by Braaten et al. Ann Rheum Dis 2022; 81:e13. [PMID: 31907163 DOI: 10.1136/annrheumdis-2019-216867] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 12/21/2019] [Indexed: 11/03/2022]
Affiliation(s)
- Fulvia Ceccarelli
- Arthritis Center, Dipartimento di Scienze Cliniche Internistiche, Anestesiologiche e Cardiovascolari, Sapienza Università di Roma, Sapienza University of Rome, Roma, Italy
| | - Andrea Botticelli
- Oncologia, Dipartimento di Medicina Clinica e Molecolare, Sapienza University of Rome, Roma, Italy
| | - Alain Jonathan Gelibter
- Oncologia, Dipartimento di Medicina Clinica e Molecolare, Sapienza University of Rome, Roma, Italy
| | - Ilaria Leccese
- Arthritis Center, Dipartimento di Scienze Cliniche Internistiche, Anestesiologiche e Cardiovascolari, Sapienza Università di Roma, Sapienza University of Rome, Roma, Italy
| | - Ramona Lucchetti
- Arthritis Center, Dipartimento di Scienze Cliniche Internistiche, Anestesiologiche e Cardiovascolari, Sapienza Università di Roma, Sapienza University of Rome, Roma, Italy
| | - Enrico Cortesi
- Oncologia, Dipartimento di Medicina Clinica e Molecolare, Sapienza University of Rome, Roma, Italy
| | - Guido Valesini
- Arthritis Center, Dipartimento di Scienze Cliniche Internistiche, Anestesiologiche e Cardiovascolari, Sapienza Università di Roma, Sapienza University of Rome, Roma, Italy
| | - Paolo Marchetti
- Oncologia, Dipartimento di Medicina Clinica e Molecolare, Sapienza University of Rome, Roma, Italy
| | - Fabrizio Conti
- Arthritis Center, Dipartimento di Scienze Cliniche Internistiche, Anestesiologiche e Cardiovascolari, Sapienza Università di Roma, Sapienza University of Rome, Roma, Italy
| |
Collapse
|
85
|
He MF, Liang Y, Huang HH. Integrating molecular interactions and gene expression to identify biomarkers to predict response to tumor necrosis factor inhibitor therapies in rheumatoid arthritis patients. Technol Health Care 2022; 30:451-457. [PMID: 35124619 PMCID: PMC9028654 DOI: 10.3233/thc-thc228041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
BACKGROUND Targeted therapy using anti-TNF (tumor necrosis factor) is the first option for patients with rheumatoid arthritis (RA). Anti-TNF therapy, however, does not lead to meaningful clinical improvement in many RA patients. To predict which patients will not benefit from anti-TNF therapy, clinical tests should be performed prior to treatment beginning. OBJECTIVE Although various efforts have been made to identify biomarkers and pathways that may be helpful to predict the response to anti-TNF treatment, gaps remain in clinical use due to the low predictive power of the selected biomarkers. METHODS In this paper, we used a network-based computational method to identify the select the predictive biomarkers to guide the treatment of RA patients. RESULTS We select 69 genes from peripheral blood expression data from 46 subjects using a sparse network-based method. The result shows that the selected 69 genes might influence biological processes and molecular functions related to the treatment. CONCLUSIONS Our approach advances the predictive power of anti-TNF therapy response and provides new genetic markers and pathways that may influence the treatment.
Collapse
Affiliation(s)
- Min-Fan He
- Macau Institute of Systems Engineering and Collaborative Laboratory of Intelligent Science and Systems, Macau University of Science and Technology, Macau, China
- School of Mathematics and Big Data, Foshan University, Foshan, China
| | - Yong Liang
- Macau Institute of Systems Engineering and Collaborative Laboratory of Intelligent Science and Systems, Macau University of Science and Technology, Macau, China
| | - Hai-Hui Huang
- Provincial Demonstration Software Institute, Shaoguan University, Shaoguan, China
| |
Collapse
|
86
|
Ghosh N, Tiongson MD, Stewart C, Chan KK, Jivanelli B, Cappelli L, Bass AR. Checkpoint Inhibitor-Associated Arthritis: A Systematic Review of Case Reports and Case Series. J Clin Rheumatol 2021; 27:e317-e322. [PMID: 32345841 PMCID: PMC7606361 DOI: 10.1097/rhu.0000000000001370] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
OBJECTIVE We performed a systematic literature review to identify all reports of immune checkpoint inhibitor-associated inflammatory arthritis to describe it phenotypically and serologically. METHODS PubMed, Embase, and Cochrane databases were searched for reports of musculoskeletal immune-related adverse events secondary to ICI treatment. Publications were included if they provided individual patient level data regarding the pattern of joint involvement. Descriptive statistics were used to summarize results. RESULTS A total of 4339 articles were screened, of which 67 were included, encompassing 372 patients. The majority of patients had metastatic melanoma (57%), and they were treated with anti-PD1 or anti-PDL1 therapy (78%). Median time to onset of arthritis was 4 months (range, 1 day to 53 months). Forty-nine percent had polyarthritis, 17% oligoarthritis, 3% monoarthritis, 10% arthralgia, and 21% polymyalgia rheumatica. More than half of patients were described as having a "rheumatoid arthritis-like" presentation. Nine percent tested positive for rheumatoid factor or anti-cyclic citrullinated peptide antibodies. Seventy-four percent required corticosteroids, and 45% required additional medications. Sixty-three percent achieved arthritis control, and 32% were ultimately able to discontinue antirheumatic treatments. Immune checkpoint inhibitors were continued in 49%, transiently withheld in 11%, and permanently discontinued due to musculoskeletal immune-related adverse events in 13%. CONCLUSIONS Half of reported immune checkpoint inhibitor-associated arthritis cases present with polyarthritis (often RA-like), but only 9% are seropositive. Polymyalgia rheumatica is also common. Most patients respond to steroids alone, but about half require additional medications. Further studies are needed to determine long-term musculoskeletal outcomes in these patients, and the impact of arthritis treatment on cancer survival.
Collapse
Affiliation(s)
- Nilasha Ghosh
- Hospital for Special Surgery, New York, NY
- Weill Cornell Medicine, New York, NY
| | | | | | - Karmela K. Chan
- Hospital for Special Surgery, New York, NY
- Weill Cornell Medicine, New York, NY
| | | | - Laura Cappelli
- Johns Hopkins University School of Medicine, Baltimore, MD
| | - Anne R. Bass
- Hospital for Special Surgery, New York, NY
- Weill Cornell Medicine, New York, NY
| |
Collapse
|
87
|
Vondenberg JA, Trost JR, Redford AH, Farshami FJ, Muruganandam M, Sibbitt WL. Atypical Chronic Deforming Arthritis After Cocaine-Levamisole Exposure. J Clin Rheumatol 2021; 27:S384-S386. [PMID: 32658048 DOI: 10.1097/rhu.0000000000001484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Jaime A Vondenberg
- Division of Rheumatology and School of Medicine Department of Internal Medicine University of New Mexico Health Sciences Center Albuquerque, NM
| | | | | | | | | | | |
Collapse
|
88
|
Xu PC, Luan Y, Yu SY, Xu J, Coulter DW, Kim SY. Effects of PD-1 blockade on ovarian follicles in a prepubertal female mouse. J Endocrinol 2021; 252:15-30. [PMID: 34647523 PMCID: PMC8630981 DOI: 10.1530/joe-21-0209] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 10/12/2021] [Indexed: 12/11/2022]
Abstract
Immunotherapy has emerged at the forefront of cancer treatment. Checkpoint inhibitor pembrolizumab (KEYTRUDA), a chimeric antibody which targets programmed cell death protein 1 (PD-1), has been approved by the Food and Drug Administration (FDA) for use in pediatric patients with relapsed or refractory classical Hodgkin's lymphoma. However, there is currently no published data regarding the effects of pembrolizumab on the ovary of female pediatric patients. In this study, prepubertal immunocompetent and immunodeficient female mice were injected with pembrolizumab or anti-mouse PD-1 antibody. The number of primordial follicles significantly decreased post-injection of both pembrolizumab and anti-mouse PD-1 antibody in immunocompetent mice. However, no changes in follicle numbers were observed in immunodeficient nude mice. Superovulation test and vaginal opening experiments suggest that there is no difference in the number of cumulus-oocyte complexes (COCs) and the timing of puberty onset between the control and anti-mouse PD-1 antibody treatment groups, indicating that there is no effect on short-term fertility. Elevation of pro-inflammatory cytokine TNF-α following COX-2 upregulation was observed in the ovary. CD3+ T-cell infiltration was detected within some ovarian follicles and between stromal cells of the ovaries in mice following treatment with anti-mouse PD-1 antibody. Thus, PD-1 immune checkpoint blockade affects the ovarian reserve through a mechanism possibly involving inflammation following CD3+ T-cell infiltration.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/pharmacology
- Antineoplastic Agents, Immunological/adverse effects
- Antineoplastic Agents, Immunological/pharmacology
- Cell Count
- Female
- Immune Checkpoint Inhibitors/adverse effects
- Immune Checkpoint Inhibitors/pharmacology
- Infertility, Female/chemically induced
- Infertility, Female/pathology
- Mice
- Mice, Nude
- Oocytes/cytology
- Oocytes/drug effects
- Ovarian Follicle/drug effects
- Ovarian Reserve/drug effects
- Ovary/drug effects
- Ovary/physiology
- Programmed Cell Death 1 Receptor/antagonists & inhibitors
- Programmed Cell Death 1 Receptor/immunology
- Sexual Maturation/drug effects
Collapse
Affiliation(s)
- Pauline C. Xu
- Olson Center for Women’s Health, Department of Obstetrics and Gynecology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Yi Luan
- Olson Center for Women’s Health, Department of Obstetrics and Gynecology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Seok-Yeong Yu
- Olson Center for Women’s Health, Department of Obstetrics and Gynecology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Jing Xu
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Beaverton, Oregon; Department of Obstetrics and Gynecology, School of Medicine, Oregon Health & Science University, Portland, Oregon
| | - Donald W. Coulter
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - So-Youn Kim
- Olson Center for Women’s Health, Department of Obstetrics and Gynecology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
89
|
The Genetic, Environmental, and Immunopathological Complexity of Autoantibody-Negative Rheumatoid Arthritis. Int J Mol Sci 2021; 22:ijms222212386. [PMID: 34830268 PMCID: PMC8618508 DOI: 10.3390/ijms222212386] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/15/2021] [Accepted: 11/15/2021] [Indexed: 12/19/2022] Open
Abstract
Differences in clinical presentation, response to treatment, and long-term outcomes between autoantibody-positive and -negative rheumatoid arthritis (RA) highlight the need for a better comprehension of the immunopathogenic events underlying the two disease subtypes. Whilst the drivers and perpetuators of autoimmunity in autoantibody-positive RA have started to be disclosed, autoantibody-negative RA remains puzzling, also due its wide phenotypic heterogeneity and its possible misdiagnosis. Genetic susceptibility appears to mostly rely on class I HLA genes and a number of yet unidentified non-HLA loci. On the background of such variable genetic predisposition, multiple exogeneous, endogenous, and stochastic factors, some of which are not shared with autoantibody-positive RA, contribute to the onset of the inflammatory cascade. In a proportion of the patients, the immunopathology of synovitis, at least in the initial stages, appears largely myeloid driven, with abundant production of proinflammatory cytokines and only minor involvement of cells of the adaptive immune system. Better understanding of the complexity of autoantibody-negative RA is still needed in order to open new avenues for targeted intervention and improve clinical outcomes.
Collapse
|
90
|
Dietz H, Weinmann SC, Salama AK. Checkpoint Inhibitors in Melanoma Patients with Underlying Autoimmune Disease. Cancer Manag Res 2021; 13:8199-8208. [PMID: 34754240 PMCID: PMC8572012 DOI: 10.2147/cmar.s283217] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 09/25/2021] [Indexed: 12/18/2022] Open
Abstract
The development of immune checkpoint inhibitors (ICI) has dramatically changed the clinical management of metastatic melanoma and other solid tumors. Despite exclusion from initial clinical trials, there is a growing body of retrospective data that suggest ICI can be used in patients with underlying autoimmune disease (AID) with a tolerable level of anticipated immune-related adverse events (irAEs) and a rate of severe irAEs comparable to that of patients without underlying AID. Coordination with other subspecialists and careful monitoring for irAEs is critical in safely managing these patients. Studies exploring novel approaches examining the use of targeted immunosuppressants in the prevention and management of irAEs, as well as multiple studies currently underway are aimed at establishing safe clinical practices when using ICI in patients with underlying AID.
Collapse
Affiliation(s)
- Hilary Dietz
- Division of Medical Oncology, Duke University, Durham, NC, USA
| | - Sophia C Weinmann
- Division of Rheumatology and Immunology, Duke University, Durham, NC, USA
| | - April K Salama
- Division of Medical Oncology, Duke University, Durham, NC, USA
| |
Collapse
|
91
|
Cappelli LC, Darrah E, Shah AA, Bingham CO. Patients with checkpoint inhibitor-induced inflammatory arthritis do not become seropositive for anti-cyclic citrullinated peptide when followed over time. ACR Open Rheumatol 2021; 4:83-84. [PMID: 34757698 PMCID: PMC8754020 DOI: 10.1002/acr2.11363] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/09/2021] [Accepted: 09/22/2021] [Indexed: 12/19/2022] Open
Affiliation(s)
| | - Erika Darrah
- Johns Hopkins University School of Medicine, Baltimore, MD
| | - Ami A Shah
- Johns Hopkins University School of Medicine, Baltimore, MD
| | | |
Collapse
|
92
|
Campochiaro C, Farina N, Tomelleri A, Ferrara R, Lazzari C, De Luca G, Bulotta A, Signorelli D, Palmisano A, Vignale D, Peretto G, Sala S, Esposito A, Garassino M, Gregorc V, Dagna L. Tocilizumab for the treatment of immune-related adverse events: a systematic literature review and a multicentre case series. Eur J Intern Med 2021; 93:87-94. [PMID: 34391591 DOI: 10.1016/j.ejim.2021.07.016] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/12/2021] [Accepted: 07/20/2021] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Research is moving towards a more personalized management of immune-related adverse events (irAEs) due to immune checkpoint inhibitors (ICI). Our objective was to evaluate the efficacy and safety of tocilizumab in the treatment of these clinical manifestations. METHODS A systematic literature review was performed to retrieve data about the use of tocilizumab in the treatment of irAEs. Additionally, data from cancer patients referred to our Immune-related Adverse Event Clinic and treated with tocilizumab were collected. RESULTS Our literature review identified 20 articles and 11 meeting abstracts. Data about 91 cancer patients who received tocilizumab for the treatment of irAEs were collected. In 85% of cases, this therapy was associated with clinical benefit and no case of disease progression was reported. ICI therapy was continued following irAE onset and biologic therapy initiation in only three patients. Five patients developed irAEs upon ICI initiation and were subsequently treated with tocilizumab at our Centre. At a median follow-up of eight months, tocilizumab was safely continued along with ICI in three out of five patients, and an adequate control of irAE was obtained in all cases. No significant adverse reactions to tocilizumab were reported. Only one patient experienced a disease progression 18 months after ICI discontinuation. CONCLUSION Both our systematic literature review and case series highlight the efficacy and safety of tocilizumab in the treatment of irAEs. Furthermore, they both support the possibility of a combined approach with tocilizumab and ICI, to guarantee an effective irAEs management without losing the oncologic response.
Collapse
Affiliation(s)
- Corrado Campochiaro
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS San Raffaele, via Olgettina 60, Milan, Italy; Vita-Salute San Raffaele University, via Olgettina 60, Milan, Italy.
| | - Nicola Farina
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS San Raffaele, via Olgettina 60, Milan, Italy; Vita-Salute San Raffaele University, via Olgettina 60, Milan, Italy
| | - Alessandro Tomelleri
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS San Raffaele, via Olgettina 60, Milan, Italy; Vita-Salute San Raffaele University, via Olgettina 60, Milan, Italy
| | - Roberto Ferrara
- Thoracic Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, via Venezian 1, Milan, Italy
| | - Chiara Lazzari
- Department of Oncology, IRCCS San Raffaele, via Olgettina 60, Milan, Italy
| | - Giacomo De Luca
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS San Raffaele, via Olgettina 60, Milan, Italy; Vita-Salute San Raffaele University, via Olgettina 60, Milan, Italy
| | - Alessandra Bulotta
- Department of Oncology, IRCCS San Raffaele, via Olgettina 60, Milan, Italy
| | - Diego Signorelli
- Thoracic Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, via Venezian 1, Milan, Italy
| | - Anna Palmisano
- Department of Radiology, IRCCS San Raffaele Hospital, via Olgettina 60, Milan, Italy
| | - Davide Vignale
- Department of Radiology, IRCCS San Raffaele Hospital, via Olgettina 60, Milan, Italy
| | - Giovanni Peretto
- Department of Arrhythmology, IRCCS San Raffaele Hospital, via Olgettina 60, Milan, Italy
| | - Simone Sala
- Department of Arrhythmology, IRCCS San Raffaele Hospital, via Olgettina 60, Milan, Italy
| | - Antonio Esposito
- Department of Radiology, IRCCS San Raffaele Hospital, via Olgettina 60, Milan, Italy
| | - Marina Garassino
- Thoracic Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, via Venezian 1, Milan, Italy
| | - Vanesa Gregorc
- Department of Oncology, IRCCS San Raffaele, via Olgettina 60, Milan, Italy
| | - Lorenzo Dagna
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS San Raffaele, via Olgettina 60, Milan, Italy; Vita-Salute San Raffaele University, via Olgettina 60, Milan, Italy
| |
Collapse
|
93
|
Fediw M, Lau K. Novel Cancer Therapeutics and Implications for Rehabilitation. CURRENT PHYSICAL MEDICINE AND REHABILITATION REPORTS 2021. [DOI: 10.1007/s40141-021-00323-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
94
|
Lindsay SE, Wurster L, Woolf K, Gundle KR. An Unusual Presentation of Inflammatory Shoulder Arthritis Associated with Nivolumab: A Case Report. JBJS Case Connect 2021; 11:01709767-202112000-00022. [PMID: 34669677 DOI: 10.2106/jbjs.cc.21.00301] [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: 06/13/2023]
Abstract
CASE A 68-year-old woman with a history of stage IIIC (T3bN1bM0) melanoma with metastases to her right axilla status after 10 cycles of nivolumab presented with right shoulder pain. Radiographs showed a progressive erosive glenohumeral joint lesion. The workup was negative for metastasis and infection. Her clinical and radiographic findings were consistent with erosive arthritis. The patient underwent shoulder hemiarthroplasty and experienced substantial improvements. CONCLUSION This is an unusual case of inflammatory arthritis associated with nivolumab, an antiprogrammed cell death protein 1, and is an important reminder of the musculoskeletal toxicities associated with immunotherapies.
Collapse
Affiliation(s)
- Sarah E Lindsay
- Department of Orthopaedics and Rehabilitation, Oregon Health and Science University, Portland, Oregon
| | - Lindsey Wurster
- Operative Care Division, Portland VA Medical Center, Portland, Oregon
| | - Kirsten Woolf
- Department of Pathology, Portland VA Medical Center, Portland, Oregon
| | - Kenneth R Gundle
- Department of Orthopaedics and Rehabilitation, Oregon Health and Science University, Portland, Oregon
- Operative Care Division, Portland VA Medical Center, Portland, Oregon
| |
Collapse
|
95
|
Kaulen LD, Doubrovinskaia S, Mooshage C, Jordan B, Purrucker J, Haubner C, Seliger C, Lorenz HM, Nagel S, Wildemann B, Bendszus M, Wick W, Schönenberger S. Neurological autoimmune diseases following vaccinations against SARS-CoV-2: a case series. Eur J Neurol 2021; 29:555-563. [PMID: 34668274 PMCID: PMC8652629 DOI: 10.1111/ene.15147] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 10/11/2021] [Indexed: 01/11/2023]
Abstract
BACKGROUND AND PURPOSE Population-based studies suggest that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines may trigger immune-mediated thrombotic thrombocytopenia (VITT) raising concerns for other autoimmune responses. The aim was to characterize neurological autoimmunity after SARS-CoV-2 vaccinations. METHODS In this single-centre prospective case study patients with neurological autoimmunity in temporal association (≤6 weeks) with SARS-CoV-2 vaccinations and without other triggers are reported. Clinical, laboratory and imaging data were collected with a median follow-up of 49 days. RESULTS In the study period 232,603 inhabitants from the main catchment area of our hospital (Rhein-Neckar-Kreis, county) received SARS-CoV-2 vaccinations. Twenty-one cases (new onset n = 17, flares n = 4) diagnosed a median of 11 days (range 3-23) following SARS-CoV-2 vaccinations (BNT162b2 n = 12, ChAdOx1 n = 8, mRNA-1273 n = 1) were identified. Cases included VITT with cerebral venous sinus thrombosis (n = 3), central nervous system demyelinating diseases (n = 8), inflammatory peripheral neuropathies (n = 4), myositis (n = 3), myasthenia (n = 1), limbic encephalitis (n = 1) and giant cell arteritis (n = 1). Patients were predominantly female (ratio 3.2:1) and the median age at diagnosis was 50 years (range 22-86). Therapy included administration of steroids (n = 15), intravenous immunoglobulins in patients with Guillain-Barré syndrome or VITT (n = 4), plasma exchange in cases unresponsive to steroids (n = 3) and anticoagulation in VITT. Outcomes were favourable with partial and complete remissions achieved in 71% and 24%, respectively. Two patients received their second vaccination without further aggravation of autoimmune symptoms under low-dose immunosuppressants. CONCLUSIONS In this study various neurological autoimmune disorders encountered following SARS-CoV-2 vaccinations are characterized. Given the assumed low incidence and mostly favourable outcome of autoimmune responses, the benefits of vaccinations outweigh the comparatively small risks.
Collapse
Affiliation(s)
- Leon D Kaulen
- Department of Neurology, University Hospital Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Sofia Doubrovinskaia
- Department of Neurology, University Hospital Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Christoph Mooshage
- Department of Neuroradiology, University Hospital Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Berit Jordan
- Department of Neurology, University Hospital Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Jan Purrucker
- Department of Neurology, University Hospital Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Carmen Haubner
- Department of Neurology, University Hospital Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Corinna Seliger
- Department of Neurology, University Hospital Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Hanns-Martin Lorenz
- Division of Rheumatology, Department of Internal Medicine V, University Hospital Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Simon Nagel
- Department of Neurology, University Hospital Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Brigitte Wildemann
- Department of Neurology, University Hospital Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Martin Bendszus
- Department of Neuroradiology, University Hospital Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Wolfgang Wick
- Department of Neurology, University Hospital Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Silvia Schönenberger
- Department of Neurology, University Hospital Heidelberg, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
96
|
Reid P, Liew DF, Akruwala R, Bass AR, Chan KK. Activated osteoarthritis following immune checkpoint inhibitor treatment: an observational study. J Immunother Cancer 2021; 9:e003260. [PMID: 34526389 PMCID: PMC8444247 DOI: 10.1136/jitc-2021-003260] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/17/2021] [Indexed: 02/04/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized cancer therapy but can result in toxicities, known as immune-related adverse events (irAEs), due to a hyperactivated immune system. ICI-related inflammatory arthritis has been described in literature, but herewith we introduce and characterize post-ICI-activated osteoarthritis (ICI-aOA). We conducted a multicenter, retrospective, observational study of patients with cancer treated with ICIs and diagnosed with ICI-aOA by a rheumatologist. ICI-aOA was defined by (1) an increase in non-inflammatory joint pain after ICI initiation, (2) in joints characteristically affected by osteoarthritis, and (3) lack of inflammation on exam. Cases were graded using the Common Terminology Criteria for Adverse Events (CTCAE) V.6.0 rubric for arthralgia. Response Evaluation Criteria in Solid Tumors V.1.1 (v.4.03) guidelines determined tumor response. Results were analyzed using χ2 tests of association and multivariate logistic regression. Thirty-six patients had ICI-aOA with a mean age at time of rheumatology presentation of 66 years (51-81 years). Most patients had metastatic melanoma (10/36, 28%) and had received a PD-1/PD-L1 inhibitor monotherapy (31/36, 86%) with 5/36 (14%) combination therapy. Large joint involvement (hip/knee) was noted in 53% (19/36), small joints of hand 25% (9/36), and spine 14% (5/36). Two-thirds (24/36) suffered multiple joint involvement. Three of 36 (8%) had CTCAE grade 3, 14 (39%) grade 2 and 19 (53%) grade 1 manifestations. Symptom onset ranged from 6 days to 33.8 months with a median of 5.2 months after ICI initiation; five patients suffered from ICI-aOA after ICI cessation (0.6, 3.5, 4.4, 7.3, and 15.4 months after ICI cessation). The most common form of therapy was intra-articular corticosteroid injections only (15/36, 42%) followed by non-steroidal anti-inflammatory drugs only (7/36, 20%). Twenty patients (56%) experienced other irAEs, with rheumatic and dermatological being the most common. All three patients with high-grade ICI-aOA also had another irAE diagnosis at some point after ICI initiation. ICI-aOA should be recognized as an adverse event of ICI immunotherapy. Early referral to a rheumatologist can facilitate the distinction between ICI induced inflammatory arthritis from post-ICI mechanical arthropathy, the latter of which can be managed with local therapy that will not compromise ICI efficacy.
Collapse
Affiliation(s)
- Pankti Reid
- Medicine, University of Chicago Biological Sciences Division, Chicago, Illinois, USA
| | - David Fl Liew
- Department of Medicine, The University of Melbourne, Melbourne, Victoria, Australia
- Division of Rheumatology and Department of Clinical Pharmacology and Therapeutics, Austin Health, Heidelberg, Victoria, Australia
| | - Rajshi Akruwala
- Department of Medicine, SUNY Downstate Medical Center, New York City, New York, USA
| | - Anne R Bass
- Division of Rheumatology, Hospital for Special Surgery, New York, New York, USA
| | - Karmela K Chan
- Division of Rheumatology, Hospital for Special Surgery, New York, New York, USA
| |
Collapse
|
97
|
Inflammation and tumor progression: signaling pathways and targeted intervention. Signal Transduct Target Ther 2021; 6:263. [PMID: 34248142 PMCID: PMC8273155 DOI: 10.1038/s41392-021-00658-5] [Citation(s) in RCA: 1219] [Impact Index Per Article: 304.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 05/11/2021] [Accepted: 05/23/2021] [Indexed: 02/06/2023] Open
Abstract
Cancer development and its response to therapy are regulated by inflammation, which either promotes or suppresses tumor progression, potentially displaying opposing effects on therapeutic outcomes. Chronic inflammation facilitates tumor progression and treatment resistance, whereas induction of acute inflammatory reactions often stimulates the maturation of dendritic cells (DCs) and antigen presentation, leading to anti-tumor immune responses. In addition, multiple signaling pathways, such as nuclear factor kappa B (NF-kB), Janus kinase/signal transducers and activators of transcription (JAK-STAT), toll-like receptor (TLR) pathways, cGAS/STING, and mitogen-activated protein kinase (MAPK); inflammatory factors, including cytokines (e.g., interleukin (IL), interferon (IFN), and tumor necrosis factor (TNF)-α), chemokines (e.g., C-C motif chemokine ligands (CCLs) and C-X-C motif chemokine ligands (CXCLs)), growth factors (e.g., vascular endothelial growth factor (VEGF), transforming growth factor (TGF)-β), and inflammasome; as well as inflammatory metabolites including prostaglandins, leukotrienes, thromboxane, and specialized proresolving mediators (SPM), have been identified as pivotal regulators of the initiation and resolution of inflammation. Nowadays, local irradiation, recombinant cytokines, neutralizing antibodies, small-molecule inhibitors, DC vaccines, oncolytic viruses, TLR agonists, and SPM have been developed to specifically modulate inflammation in cancer therapy, with some of these factors already undergoing clinical trials. Herein, we discuss the initiation and resolution of inflammation, the crosstalk between tumor development and inflammatory processes. We also highlight potential targets for harnessing inflammation in the treatment of cancer.
Collapse
|
98
|
Abstract
Immune checkpoint inhibitors (ICIs) are monoclonal antibodies that target two key signalling pathways related to T cell activation and exhaustion, by binding to and inhibiting cytotoxic T lymphocyte antigen 4 (CTLA4) or PD1 and its ligand PDL1. ICIs, such as nivolumab, pembrolizumab and ipilimumab, are approved for the treatment of numerous and diverse cancer types, in various combination regimens, and are now an established cornerstone of cancer therapeutics. Toxicities induced by ICIs are autoimmune in nature and are referred to as immune-related adverse events (irAEs); these events can affect any organ system in an unpredictable fashion. Importantly, irAEs can manifest as endocrinopathies involving the thyroid (hypothyroidism or thyrotoxicosis), pituitary (hypophysitis), adrenal glands (adrenal insufficiency) and pancreas (diabetes mellitus). These events are a frequent source of acute and persistent morbidity in patients treated with ICIs and can even be fatal. Over the past few years, there has been a growing understanding of the underlying pathogenesis of irAEs that has led to the development of more effective management strategies. Herein, we review the current understanding of the pathobiology, clinical manifestations and treatment approaches to endocrine toxicities arising from ICIs.
Collapse
Affiliation(s)
- Jordan J Wright
- Department of Medicine, Vanderbilt University Medical Center and Vanderbilt Ingram Cancer Center, Nashville, TN, USA
| | - Alvin C Powers
- Department of Medicine, Vanderbilt University Medical Center and Vanderbilt Ingram Cancer Center, Nashville, TN, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
- VA Tennessee Valley Healthcare System, Nashville, TN, USA
| | - Douglas B Johnson
- Department of Medicine, Vanderbilt University Medical Center and Vanderbilt Ingram Cancer Center, Nashville, TN, USA.
| |
Collapse
|
99
|
Daniello L, Elshiaty M, Bozorgmehr F, Kuon J, Kazdal D, Schindler H, Shah R, Volckmar AL, Lusky F, Diekmann L, Liersch S, Faehling M, Muley T, Kriegsmann M, Benesova K, Stenzinger A, Thomas M, Christopoulos P. Therapeutic and Prognostic Implications of Immune-Related Adverse Events in Advanced Non-Small-Cell Lung Cancer. Front Oncol 2021; 11:703893. [PMID: 34268127 PMCID: PMC8277237 DOI: 10.3389/fonc.2021.703893] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/09/2021] [Indexed: 01/14/2023] Open
Abstract
Introduction PD-(L)1 inhibitors have improved prognosis of non-small-cell lung cancer (NSCLC), but can also cause immune-related adverse events (irAEs) that complicate management. Methods We analyzed NSCLC patients receiving PD-(L)1 inhibitors from 2012 to 2020 in a German academic center. Results IrAE showed comparable frequencies in stage IV (198/894 or 22%) vs. III (14/45 or 31%, p = 0.15), after anti-PD-(L)1 monotherapy vs. chemoimmunotherapy (139/483 vs. 58/213, p = 0.75), and across treatment lines. In stage IV, irAE occurred after 3.1 months in median, affected multiple organs (median 2) in 27/894 patients and were associated with PD-L1 positivity (25 vs. 14%, p = 0.003), lower neutrophil-to-lymphocyte ratios (29 vs. 17%, p < 0.001 for NLR dichotomized at 5), better ECOG status (26 vs. 18% for 0 vs. 1, p = 0.004), but not related to age, sex, smoking and palliative radiotherapy. Two hundred thirty two irAEs occurred mostly in endocrine glands (4.9%), lungs (4.4%), the musculoskeletal system (4.2%), colon (4.1%), liver (3.7%), and skin (2.6%), while pneumonitis was most frequent with durvalumab following definitive chemoradiation (16% or 7/45, p < 0.01). IrAE severity was grade 1 in 11%, 2 in 41%, 3 in 36%, and 4 in 11% events, while two were lethal (<1%, myocarditis and pneumonitis). Therapy was suspended in 72%, while steroids were initiated in 66% and complemented by other immunosuppressants in 6%, with longest treatment duration for rheumatic events (mean >3 months), and average cumulative prednisone doses >700 mg for all organs, except for skin. Patients developing irAE had longer progression-free (PFS) and overall survival (OS) in multivariable 12/14-week landmark analyses including ECOG status, treatment line, treatment type, PD-L1 TPS, and NLR (median PFS 17 vs. 10 months, HR = 0.68, p = 0.009; median OS 37 vs. 15 months, HR = 0.40, p < 0.001), regardless of grade. OS was longest with skin (95% at 2 years) and shortest with pneumonitis, hepatitis, neurologic, and cardiologic irAE (38, 37, 28, and 0% at 2 years, p < 0.001). Conclusions Approximately one-fourth of immunotherapy-treated NSCLC patients develop irAEs, most of which necessitate treatment suspension and steroids. Despite more frequent occurrence with PD-L1 positive tumors, lower NLR, and better ECOG PS, irAEs are independently associated with longer survival, especially when affecting the skin. Lethality is below 1%.
Collapse
Affiliation(s)
- Lea Daniello
- Department of Thoracic Oncology, Thoraxklinik and National Center for Tumor Diseases (NCT) at Heidelberg University Hospital, Heidelberg, Germany.,Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center of Lung Research (DZL), Heidelberg, Germany
| | - Mariam Elshiaty
- Department of Thoracic Oncology, Thoraxklinik and National Center for Tumor Diseases (NCT) at Heidelberg University Hospital, Heidelberg, Germany.,Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center of Lung Research (DZL), Heidelberg, Germany
| | - Farastuk Bozorgmehr
- Department of Thoracic Oncology, Thoraxklinik and National Center for Tumor Diseases (NCT) at Heidelberg University Hospital, Heidelberg, Germany.,Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center of Lung Research (DZL), Heidelberg, Germany
| | - Jonas Kuon
- Department of Thoracic Oncology, Thoraxklinik and National Center for Tumor Diseases (NCT) at Heidelberg University Hospital, Heidelberg, Germany.,Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center of Lung Research (DZL), Heidelberg, Germany
| | - Daniel Kazdal
- Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center of Lung Research (DZL), Heidelberg, Germany.,Institute of Pathology Heidelberg, Heidelberg, Germany
| | - Hannah Schindler
- Department of Thoracic Oncology, Thoraxklinik and National Center for Tumor Diseases (NCT) at Heidelberg University Hospital, Heidelberg, Germany.,Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center of Lung Research (DZL), Heidelberg, Germany
| | - Rajiv Shah
- Department of Thoracic Oncology, Thoraxklinik and National Center for Tumor Diseases (NCT) at Heidelberg University Hospital, Heidelberg, Germany.,Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center of Lung Research (DZL), Heidelberg, Germany
| | | | - Fabienne Lusky
- Department of Thoracic Oncology, Thoraxklinik and National Center for Tumor Diseases (NCT) at Heidelberg University Hospital, Heidelberg, Germany.,Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center of Lung Research (DZL), Heidelberg, Germany
| | - Leonore Diekmann
- Department of Internal Medicine V, Hematology, Oncology und Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Stephan Liersch
- Department of Pharmacy, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany
| | - Martin Faehling
- Department of Cardiology, Angiology and Pneumology, Esslingen Hospital, Esslingen, Germany
| | - Thomas Muley
- Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center of Lung Research (DZL), Heidelberg, Germany.,Translational Research Unit, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany
| | - Mark Kriegsmann
- Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center of Lung Research (DZL), Heidelberg, Germany.,Institute of Pathology Heidelberg, Heidelberg, Germany
| | - Karolina Benesova
- Department of Internal Medicine V, Hematology, Oncology und Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Albrecht Stenzinger
- Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center of Lung Research (DZL), Heidelberg, Germany.,Institute of Pathology Heidelberg, Heidelberg, Germany
| | - Michael Thomas
- Department of Thoracic Oncology, Thoraxklinik and National Center for Tumor Diseases (NCT) at Heidelberg University Hospital, Heidelberg, Germany.,Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center of Lung Research (DZL), Heidelberg, Germany
| | - Petros Christopoulos
- Department of Thoracic Oncology, Thoraxklinik and National Center for Tumor Diseases (NCT) at Heidelberg University Hospital, Heidelberg, Germany.,Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center of Lung Research (DZL), Heidelberg, Germany
| |
Collapse
|
100
|
Nebhan CA, Johnson DB. Pembrolizumab in the adjuvant treatment of melanoma: efficacy and safety. Expert Rev Anticancer Ther 2021; 21:583-590. [PMID: 33504219 PMCID: PMC8238788 DOI: 10.1080/14737140.2021.1882856] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 01/26/2021] [Indexed: 10/22/2022]
Abstract
Introduction: Regional or distant metastases from melanoma may be surgically resected but remain at high-risk of recurrence. Over the last few years, several treatments have been approved to mitigate this risk. These include anti-PD-1 agents, specifically pembrolizumab and nivolumab.Areas covered: Herein, we will discuss the landscape of pembrolizumab safety and efficacy used in the adjuvant setting for high-risk, resected melanoma. We place this in context with other available adjuvant therapies, and discuss subgroup analyses.Expert opinion: Anti-PD-1 therapy with either pembrolizumab or nivolumab has become a standard of care for patients with resected stage III or IV melanoma. In our practice, we generally offer these agents (which have comparable safety and efficacy profiles) to patients with resected stage IIIb-IV melanoma regardless of BRAF mutation status.
Collapse
Affiliation(s)
- Caroline A. Nebhan
- Department of Medicine, Vanderbilt University Medical Center and Vanderbilt Ingram Cancer Center
| | - Douglas B. Johnson
- Department of Medicine, Vanderbilt University Medical Center and Vanderbilt Ingram Cancer Center
| |
Collapse
|