1
|
Ottaiano A, Santorsola M, Capuozzo M, Scala S. Balancing immunotherapy and corticosteroids in cancer treatment: dilemma or paradox? Oncologist 2025; 30:oyaf045. [PMID: 40163690 PMCID: PMC11957262 DOI: 10.1093/oncolo/oyaf045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 12/19/2024] [Indexed: 04/02/2025] Open
Abstract
Corticosteroids are widely used to prevent and treat chemotherapy-induced nausea and vomiting (CINV) due to their pleiotropic biological effects. However, concerns have been raised about their immunosuppressive properties when combined with immunotherapy. Specifically, their potential impact on the efficacy of immunotherapy, mainly immune checkpoint inhibitors (ICIs), remains a subject of debate. This manuscript discusses the mechanisms by which corticosteroids mitigate CINV, the challenges associated with their concurrent use with immunotherapy, and emerging therapeutic strategies evaluating dexamethasone-free regimens. A careful balance must be struck in corticosteroid use to effectively manage CINV while optimizing the outcomes of immunotherapy.
Collapse
Affiliation(s)
- Alessandro Ottaiano
- SSD-Innovative Therapies for Abdominal Metastases, Istituto Nazionale Tumori di Napoli, IRCCS Fondazione “G. Pascale,”80131 Naples, Italy
| | - Mariachiara Santorsola
- SSD-Innovative Therapies for Abdominal Metastases, Istituto Nazionale Tumori di Napoli, IRCCS Fondazione “G. Pascale,”80131 Naples, Italy
| | | | - Stefania Scala
- Microenvironment Molecular Targets, Istituto Nazionale Tumori di Napoli, IRCCS Fondazione “G. Pascale,”80131 Naples, Italy
| |
Collapse
|
2
|
Yoshino M, Ishihara H, Nemoto Y, Mizoguchi S, Ikeda T, Nakayama T, Fukuda H, Yoshida K, Iizuka J, Shimmura H, Hashimoto Y, Kondo T, Takagi T. Survival Impact of Glucocorticoid Administration for Adverse Events During Immune Checkpoint Inhibitor Combination Therapy in Patients with Previously Untreated Advanced Renal Cell Carcinoma. Target Oncol 2024; 19:623-633. [PMID: 38819770 DOI: 10.1007/s11523-024-01069-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/07/2024] [Indexed: 06/01/2024]
Abstract
BACKGROUND The impact of glucocorticoid administration for adverse events (AEs), including immune-related AEs, on the effectiveness of immune checkpoint inhibitor (ICI) combination therapy for advanced renal cell carcinoma (RCC) remains unknown. OBJECTIVES To clarify the prognostic impact of glucocorticoid use for AEs during first-line ICI combination therapy for advanced RCC. PATIENTS AND METHODS We retrospectively evaluated data from 194 patients who received dual ICI combination therapy [i.e., immunotherapy (IO)-IO] or combinations of ICIs with tyrosine kinase inhibitors (TKIs) as first-line therapy. The patients were divided into two groups according to the history of glucocorticoid administration in each treatment group. Survival based on glucocorticoid administration was assessed. RESULTS A total of 101 (52.0%) and 93 (48.0%) patients received IO-IO and IO-TKI combination therapy, respectively. Glucocorticoids were administered to 46 (46%) and 22 (24%) patients in the IO-IO and IO-TKI groups, respectively. In the IO-IO group, progression-free survival (PFS) and overall survival (OS) were significantly longer in patients with glucocorticoid administration than in those without administration (median PFS: 14.4 versus 3.45 months, p = 0.0005; median OS: 77.6 versus 33.9 months, p = 0.0025). Multivariable analysis showed that glucocorticoid administration was an independent predictor of longer PFS (hazard ratio: 0.43, p = 0.0005) and OS (hazard ratio: 0.35, p = 0.0067) after adjustment for covariates. In the IO-TKI group, neither PFS nor OS significantly differed between patients treated with and without glucocorticoid administration (PFS: p = 0.0872, OS: p = 0.216). CONCLUSIONS Glucocorticoid administration did not negatively impact the effectiveness of ICI combination therapy for RCC, prompting glucocorticoid treatment use when AEs develop.
Collapse
Affiliation(s)
- Maki Yoshino
- Department of Urology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, Japan
| | - Hiroki Ishihara
- Department of Urology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, Japan.
| | - Yuki Nemoto
- Department of Urology, Joban Hospital, 57 Kaminodai, Jobankamiyunagayamachi, Iwaki, Fukushima, Japan
| | - Shinsuke Mizoguchi
- Department of Urology, Saiseikai Kazo Hospital, 1680 Kamitakayanagi, Kazo, Saitama, Japan
| | - Takashi Ikeda
- Department of Urology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, Japan
| | - Takayuki Nakayama
- Department of Urology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, Japan
| | - Hironori Fukuda
- Department of Urology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, Japan
| | - Kazuhiko Yoshida
- Department of Urology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, Japan
| | - Junpei Iizuka
- Department of Urology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, Japan
| | - Hiroaki Shimmura
- Department of Urology, Joban Hospital, 57 Kaminodai, Jobankamiyunagayamachi, Iwaki, Fukushima, Japan
| | - Yasunobu Hashimoto
- Department of Urology, Saiseikai Kawaguchi General Hospital, 5-11-5 Nishikawaguchi, Kawaguchi, Saitama, Japan
| | - Tsunenori Kondo
- Department of Urology, Tokyo Women's Medical University Adachi Medical Center, 4-33-1 Kouhoku, Adachi-ku, Tokyo, Japan
| | - Toshio Takagi
- Department of Urology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, Japan
| |
Collapse
|
3
|
Li S, Tang T, Han J, Liu W, Chen R, Deng H, Jian T, Fu Z. The reduced mortality of malignant melanoma at the population level is mainly attributable to treatment advances for the past decade. Cancer Epidemiol 2024; 88:102515. [PMID: 38176331 DOI: 10.1016/j.canep.2023.102515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/26/2023] [Accepted: 12/15/2023] [Indexed: 01/06/2024]
Abstract
BACKGROUND Cutaneous malignant melanoma (CMM) causes most skin cancer deaths in the United States (US). The mortality has been decreasing in the US population. We hypothesize that this population-level reduction is mainly attributable to the treatment advances, rather than the successful primary and secondary prevention. METHODS Using data from the Surveillance, Epidemiology, and End Results (SEER) databases, we collected the incidence, incidence-based mortality (IBM), and 5-year survival (5-YS) rates of CMM from 1994 to 2019. Trends by stage and sex were examined by joinpoint regression analyses and age-period-cohort analyses. RESULTS The overall incidence of CMM rose by 1.6% yearly from 1994 to 2006 (95% confidence interval [CI]: 0.9% to 2.2%) and then increased with a numerical trend. And we projected the incidence will continue to increase until 2029. In contrast, the IBM for all CMM has decreased yearly by 2.8% (95% CI: -3.9% to -1.8%) since 2010 after continuously increasing by 3.8% annually (95% CI: 3.2% to 4.4%) from 1996 to 2010. For early-stage (localized and regional) CMM, we found the incidence since 2005 plateaued without further increase, while the incidence for CMM at distant stage continuously increased by 1.4% per year (95% CI: 0.9% to 2.0%). Improvements in 5-YS were observed over the study period for all CMM and were most obvious in distant stage. And significant period effects were noted around the year 2010. CONCLUSION This study demonstrated improved survival and reduced mortality of CMM at the US population level since 2010, which were consistent with the introduction of novel therapies. Encouraging effects of primary prevention among adolescents in the most recent cohorts were found. However, the plateaued overall incidence and early diagnosis rates indicated that advances in primary and secondary prevention are very much needed to further control the burden of CMM.
Collapse
Affiliation(s)
- Si Li
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Tian Tang
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jianglong Han
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Wenmin Liu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Ruyan Chen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Haiyu Deng
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Tingting Jian
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Zhenming Fu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| |
Collapse
|
4
|
Goodman RS, Johnson DB, Balko JM. Corticosteroids and Cancer Immunotherapy. Clin Cancer Res 2023; 29:2580-2587. [PMID: 36648402 PMCID: PMC10349688 DOI: 10.1158/1078-0432.ccr-22-3181] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/22/2022] [Accepted: 01/04/2023] [Indexed: 01/18/2023]
Abstract
Despite revolutionizing cancer management, immunotherapies dysregulate the immune system, leading to immune-mediated adverse events. These common and potentially dangerous toxicities are often treated with corticosteroids, which are among the most prescribed drugs in oncology for a wide range of cancer and noncancer indications. While steroids exert several mechanisms to reduce immune activity, immunotherapies, such as immune checkpoint inhibitors (ICI), are designed to enhance the immune system's inherent antitumor activity. Because ICI requires an intact and robust immune response, the immunosuppressive properties of steroids have led to a widespread concern that they may interfere with antitumor responses. However, the existing data of the effect of systemic steroids on immunotherapy efficacy remain somewhat conflicted and unclear. To inform clinical decision-making and improve outcomes, we review the impact of steroids on antitumor immunity, recent advances in the knowledge of their impact on ICI efficacy in unique populations and settings, associated precautions, and steroid-sparing treatment approaches.
Collapse
Affiliation(s)
| | - Douglas B. Johnson
- Department of Hematology/Oncology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Justin M. Balko
- Department of Medicine, Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
5
|
Rao Ullur A, Côté G, Pelletier K, Kitchlu A. Immunotherapy in oncology and the kidneys: a clinical review of the evaluation and management of kidney immune-related adverse events. Clin Kidney J 2023; 16:939-951. [PMID: 37261008 PMCID: PMC10229281 DOI: 10.1093/ckj/sfad014] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Indexed: 11/07/2023] Open
Abstract
Immune checkpoint inhibitors (ICI) are now widely used in the treatment of many cancers, and currently represent the standard of care for multiple malignancies. These agents enhance the T cell immune response to target cancer tissues, and have demonstrated considerable benefits for cancer outcomes. However, despite these improved outcomes, there are important kidney immune-related adverse events (iRAEs) associated with ICI. Acute tubulo-interstitial nephritis remains the most frequent kidney iRAE, however glomerular lesions and electrolytes disturbances are increasingly being recognized and reported. In this review, we summarize clinical features and identify risk factors for kidney iRAEs, and discuss the current understanding of pathophysiologic mechanisms. We highlight the evidence basis for guideline-recommended management of ICI-related kidney injury as well as gaps in current knowledge. We advocate for judicious use of kidney biopsy to identify ICI-associated kidney injury, and early use of corticosteroid treatment where appropriate. Selected patients may also be candidates for re-challenge with ICI therapy after a kidney iRAE, in view of current data on recurrent rates of kidney injury. Risk of benefits of re-challenge must be considered on an individual considering patient preferences and prognosis. Lastly, we review current knowledge of ICI use in the setting of patients with end-stage kidney disease, including kidney transplant recipients and those receiving dialysis, which suggest that these patients should not be summarily excluded from the potential benefits of these cancer therapies.
Collapse
Affiliation(s)
- Avinash Rao Ullur
- Division of Nephrology, Department of Medicine, University Health Network, University of Toronto, Toronto, Canada
| | - Gabrielle Côté
- Division of Nephrology, Department of Medicine, CHU de Québec, Université Laval, Quebec City, Canada
| | - Karyne Pelletier
- Department of Medicine, Hôpital du Sacré-Coeur de Montréal, Faculty of Medicine, Université de Montréal, Montréal, Canada
| | - Abhijat Kitchlu
- Division of Nephrology, Department of Medicine, University Health Network, University of Toronto, Toronto, Canada
| |
Collapse
|
6
|
Liu JC, Yu HJ. A Review of the Pharmacokinetic Characteristics of Immune Checkpoint Inhibitors and Their Clinical Impact Factors. Pharmgenomics Pers Med 2023; 16:29-36. [PMID: 36714524 PMCID: PMC9880024 DOI: 10.2147/pgpm.s391756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 01/06/2023] [Indexed: 01/21/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) have been shown to be significant in improving the overall survival rate in certain malignancies with poor prognoses. However, only 20-40% of patients achieve long-term benefits, highlighting the relevance of the factors that influence the treatment, which can help clinicians improve their results and guide the development of new immune checkpoint therapies. In this study, the current pharmacokinetic aspects associated with the ICIs and the factors influencing clinical efficacy were characterised, including in terms of drug metabolism, drug clearance, hormonal effects and immunosuppressive effects.
Collapse
Affiliation(s)
- Jun-Chen Liu
- Department of Clinical Pharmacy, The First People’s Hospital of Jiande, Jiande, People’s Republic of China
| | - Hong-Jing Yu
- Department of Clinical Pharmacy, The First People’s Hospital of Jiande, Jiande, People’s Republic of China,Correspondence: Hong-Jing Yu, Department of Medical Oncology, The First People’s Hospital of jiande, No. 599 Yanzhou Avenue, Xin’anjiang street, Jiande, Zhejiang, 311600, People’s Republic of China, Tel +86 15869196365, Fax +86-571-64721520, Email
| |
Collapse
|
7
|
Fathi M, Razavi SM, Sojoodi M, Ahmadi A, Ebrahimi F, Namdar A, Hojjat-Farsangi M, Gholamin S, Jadidi-Niaragh F. Targeting the CTLA-4/B7 axes in glioblastoma: preclinical evidence and clinical interventions. Expert Opin Ther Targets 2022; 26:949-961. [PMID: 36527817 DOI: 10.1080/14728222.2022.2160703] [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] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Glioblastoma Multiforme (GBM) is one of the fatal cancers of the Central Nervous System (CNS). A variety of reasons exist for why previous immunotherapy strategies, especially Immune Checkpoint Blockers (ICBs), did not work in treating GBM patients. The cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) is a key immune checkpoint receptor. Its overexpression in cancer and immune cells causes tumor cell progression. CTLA-4 suppresses anti-tumor responses inside the GBM tumor-immune microenvironment. AREAS COVERED It has been attempted to explain the immunobiology of CTLA-4 as well as its interaction with different immune cells and cancer cells that lead to GBM progression. Additionally, CTLA-4 targeting studies have been reviewed and CTLA-4 combination therapy, as a promising therapeutic target and strategy for GBM immunotherapy, is recommended. EXPERT OPINION CTLA-4 could be a possible supplement for future cancer immunotherapies of GBM. However, many challenges remain such as the high toxicity of CTLA-4 blockers, and the unresponsiveness of most patients to immunotherapy. For the future clinical success of CTLA-4 blocker therapy, combination approaches with other targeted treatments would be a potentially effective strategy. Going forward, predictive biomarkers can be used to reduce trial timelines and increase the chance of success.
Collapse
Affiliation(s)
- Mehrdad Fathi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyed-Mostafa Razavi
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Mozhdeh Sojoodi
- Division of Gastrointestinal and Oncologic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Armin Ahmadi
- Department of Chemical and Materials Engineering, The University of Alabama in Huntsville, AL, USA
| | - Farbod Ebrahimi
- Nanoparticle Process Technology, Faculty of Engineering, University of Duisburg-Essen, Duisburg, Germany
| | - Afshin Namdar
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | | | - Sharareh Gholamin
- Division of Biology and Bioengineering, California Institute of Technology, Pasadena, CA, USA
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
8
|
Howard F, Conner J, Danson S, Muthana M. Inconsistencies in Modeling the Efficacy of the Oncolytic Virus HSV1716 Reveal Potential Predictive Biomarkers for Tolerability. Front Mol Biosci 2022; 9:889395. [PMID: 35782876 PMCID: PMC9240779 DOI: 10.3389/fmolb.2022.889395] [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: 03/04/2022] [Accepted: 05/10/2022] [Indexed: 12/26/2022] Open
Abstract
Treatment with HSV1716 via intralesional administration has proven successful for melanoma patients with the hope that oncolytic virotherapy would become another weapon in the systemic anticancer therapy (SACT) arsenal. In addition to challenges surrounding the systemic delivery of oncolytic viruses (OVs), problems associated with its in vivo modeling have resulted in low predictive power, contributing to the observed disappointing clinical efficacy. As OV's efficacy is elicited through interaction with the immune system, syngeneic orthotopic mouse models offer the opportunity to study these with high reproducibility and at a lower cost; however, inbred animals display specific immune characteristics which may confound results. The systemic delivery of HSV1716 was, therefore, assessed in multiple murine models of breast cancer. Tolerability to the virus was strain-dependent with C57/Bl6, the most tolerant and Balb/c experiencing lethal side effects, when delivered intravenously. Maximum tolerated doses were not enough to demonstrate efficacy against tumor growth rates or survival of Balb/c and FVB mouse models; therefore; the most susceptible strain (Balb/c mice) was treated with immunomodulators prior to virus administration in an attempt to reduce side effects. These studies demonstrate the number of variables to consider when modeling the efficacy of OVs and the complexities involved in their interpretation for translational purposes. By reporting these observations, we have potentially revealed a role for T-cell helper polarization in viral tolerability. Importantly, these findings were translated to human studies, whereby a Th1 cytokine profile was expressed in pleural effusions of patients that responded to HSV1716 treatment for malignant pleural mesothelioma with minimal side effects, warranting further investigation as a biomarker for predictive response.
Collapse
Affiliation(s)
- Faith Howard
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
| | - Joe Conner
- Virtuu Biologics/Sorrento Therapeutics, Biocity Scotland, Newhouse, United Kingdom
| | - Sarah Danson
- Sheffield Experimental Cancer Medicine Centre and Weston Park Cancer Centre, Weston Park Hospital, University of Sheffield, Sheffield, United Kingdom
| | - Munitta Muthana
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
9
|
Graziani G, Lisi L, Tentori L, Navarra P. Monoclonal Antibodies to CTLA-4 with Focus on Ipilimumab. EXPERIENTIA SUPPLEMENTUM (2012) 2022; 113:295-350. [PMID: 35165868 DOI: 10.1007/978-3-030-91311-3_10] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The immune checkpoint cytotoxic T lymphocyte-associated antigen 4 (CTLA-4 or CD152) is a negative regulator of T-cell-mediated immune responses which plays a critical role in suppressing autoimmunity and maintaining immune homeostasis. Because of its inhibitory activity on T cells, CTLA-4 has been investigated as a drug target to induce immunostimulation, blocking the interaction with its ligands. The antitumor effects mediated by CTLA-4 blockade have been attributed to a sustained active immune response against cancer cells, due to the release of a brake on T cell activation. Ipilimumab (Yervoy, Bristol-Myers Squibb) is a fully human anti-CTLA-4 IgG1κ monoclonal antibody (mAb) that represents the first immune checkpoint inhibitor approved as monotherapy by FDA and EMA in 2011 for the treatment of unresectable/metastatic melanoma. In 2015, FDA also granted approval to ipilimumab monotherapy as adjuvant treatment of stage III melanoma to reduce the risk of tumour recurrence. The subsequent approved indications of ipilimumab for metastatic melanoma, regardless of BRAF mutational status, and other advanced/metastatic solid tumours always involve its use in association with the anti-programmed cell death protein 1 (PD-1) mAb nivolumab. Currently, ipilimumab is evaluated in ongoing clinical trials for refractory/advanced solid tumours mainly in combination with additional immunostimulating agents.
Collapse
Affiliation(s)
- Grazia Graziani
- Pharmacology Section, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.
| | - Lucia Lisi
- Section of Pharmacology, Department of Healthcare Surveillance and Bioethics, Catholic University Medical School, Catholic University of the Sacred Heart, Rome, Italy
| | - Lucio Tentori
- Pharmacology Section, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Pierluigi Navarra
- Section of Pharmacology, Department of Healthcare Surveillance and Bioethics, Catholic University Medical School, Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
10
|
Lisi L, Lacal PM, Martire M, Navarra P, Graziani G. Clinical experience with CTLA-4 blockade for cancer immunotherapy: From the monospecific monoclonal antibody ipilimumab to probodies and bispecific molecules targeting the tumor microenvironment. Pharmacol Res 2021; 175:105997. [PMID: 34826600 DOI: 10.1016/j.phrs.2021.105997] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 11/09/2021] [Accepted: 11/19/2021] [Indexed: 12/15/2022]
Abstract
The immune checkpoint cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) is an inhibitory regulator of T-cell mediated responses that has been investigated as target of monoclonal antibodies (mAbs) for cancer immunotherapy. The anti-CTLA-4 mAb ipilimumab represents the first immune checkpoint inhibitor that significantly improved overall survival in patients with unresectable/metastatic melanoma. The subsequent approved indications (often in the first-line setting) for melanoma and other advanced/metastatic solid tumors always require ipilimumab combination with nivolumab, an anti-programmed cell death protein 1 (PD-1) mAb. However, the improved clinical efficacy of the mAb combination is associated with increased immune-related adverse events, which might require treatment discontinuation even in responding patients. This drawback is expected to be overcome by the recent development of anti-CTLA-4 probodies proteolitycally activated in the tumor microenvironment and bispecific molecules targeting both CTLA-4 and PD-1, whose co-expression is characteristic of tumor-infiltrating T cells. These molecules would preferentially stimulate immune responses against the tumor, reducing toxicity toward normal tissues.
Collapse
Affiliation(s)
- Lucia Lisi
- Section of Pharmacology, Department of Healthcare surveillance and Bioethics, Catholic University Medical School, Largo F. Vito 1, 00168 Rome, Italy.
| | | | - Maria Martire
- Section of Pharmacology, Department of Healthcare surveillance and Bioethics, Catholic University Medical School, Largo F. Vito 1, 00168 Rome, Italy.
| | - Pierluigi Navarra
- Section of Pharmacology, Department of Healthcare surveillance and Bioethics, Catholic University Medical School, Largo F. Vito 1, 00168 Rome, Italy.
| | - Grazia Graziani
- IDI-IRCCS, Via dei Monti di Creta 104, 00167 Rome, Italy; Pharmacology Section, Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy.
| |
Collapse
|
11
|
Jessurun CAC, Hulsbergen AFC, de Wit AE, Tewarie IA, Snijders TJ, Verhoeff JJC, Phillips JG, Reardon DA, Mekary RA, Broekman MLD. The combined use of steroids and immune checkpoint inhibitors in brain metastasis patients: a systematic review and meta-analysis. Neuro Oncol 2021; 23:1261-1272. [PMID: 33631792 DOI: 10.1093/neuonc/noab046] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICI) have been a breakthrough for selected cancer patients, including those with brain metastases (BMs). Likewise, steroids have been an integral component of symptomatic management of BM patients. However, clinical evidence on the interaction between ICI and steroids in BM patients is conflicting and has not adequately been summarized thus far. Hence, the aim of this study was to perform a systematic literature review and meta-analysis on the association between steroid use and overall survival (OS) in BM patients receiving ICI. METHODS A systematic literature search was performed. Pooled effect estimates were calculated using random-effects models across included studies. RESULTS After screening 1145 abstracts, 15 observational studies were included. Fourteen studies reported sufficient data for meta-analysis, comprising 1102 BM patients of which 32.1% received steroids. In the steroid group, median OS ranged from 2.9 to 10.2 months. In the nonsteroid group, median OS ranged from 4.9 to 25.1 months. Pooled results demonstrated significantly worse OS (HR = 1.84, 95% CI 1.22-2.77) and systemic progression-free survival (PFS; HR = 2.00, 95% CI 1.37-2.91) in the steroid group. Stratified analysis showed a consistent effect across the melanoma subgroup; not in the lung cancer subgroup. No significant association was shown between steroid use and intracranial PFS (HR = 1.31, 95% CI 0.42-4.07). CONCLUSIONS Administration of steroids was associated with significantly worse OS and PFS in BM patients receiving ICI. Further research on dose, timing, and duration of steroids is needed to elucidate the cause of this association and optimize outcomes in BM patients receiving ICI.
Collapse
Affiliation(s)
- Charissa A C Jessurun
- Computational Neuroscience Outcomes Center (CNOC), Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Department of Neurosurgery, Leiden University Medical Center, Leiden, the Netherlands.,Department of Neurosurgery, Haaglanden Medical Center, The Hague, the Netherlands
| | - Alexander F C Hulsbergen
- Computational Neuroscience Outcomes Center (CNOC), Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Department of Neurosurgery, Leiden University Medical Center, Leiden, the Netherlands.,Department of Neurosurgery, Haaglanden Medical Center, The Hague, the Netherlands
| | - Anouk E de Wit
- Department of Psychiatry, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Ishaan A Tewarie
- Computational Neuroscience Outcomes Center (CNOC), Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Department of Neurosurgery, Leiden University Medical Center, Leiden, the Netherlands.,Department of Neurosurgery, Haaglanden Medical Center, The Hague, the Netherlands
| | - Tom J Snijders
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Joost J C Verhoeff
- Department of Radiation Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - John G Phillips
- Department of Radiation Oncology, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - David A Reardon
- Harvard Medical School, Boston, Massachusetts, USA.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Rania A Mekary
- Computational Neuroscience Outcomes Center (CNOC), Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Department of Pharmaceutical Business and Administrative Sciences, School of Pharmacy, Massachusetts College of Pharmacy and Health Sciences, Boston, Massachusetts, USA
| | - Marike L D Broekman
- Computational Neuroscience Outcomes Center (CNOC), Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Department of Neurosurgery, Leiden University Medical Center, Leiden, the Netherlands.,Department of Neurosurgery, Haaglanden Medical Center, The Hague, the Netherlands.,Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
12
|
Maslov DV, Tawagi K, Kc M, Simenson V, Yuan H, Parent C, Bamnolker A, Goel R, Blake Z, Matrana MR, Johnson DH. Timing of steroid initiation and response rates to immune checkpoint inhibitors in metastatic cancer. J Immunother Cancer 2021; 9:jitc-2020-002261. [PMID: 34226279 PMCID: PMC8258666 DOI: 10.1136/jitc-2020-002261] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2021] [Indexed: 11/17/2022] Open
Abstract
Background Corticosteroids (CS) are the mainstay of immune-related adverse effect (irAE) management, as well as for other indications in cancer treatment. Previous studies evaluating whether CS affect immune checkpoint inhibitor (CPI) efficacy compared patients receiving CS versus no CS. However, there is a paucity of clinical data evaluating the timing of concomitant CS and CPI efficacy. Methods We retrospectively collected data from patients who received CS during CPI treatment at a single institution. Patients were in two cohorts based on timing of initiation of CS (≥2 months vs <2 months after initiating CPI). Patient characteristics, irAEs, cancer type, treatment type, treatment response/progression per RECIST V.1.1, and survival data were collected. Kaplan-Meier and Cox proportional hazard regression methods estimated HRs for the primary endpoint of progression-free survival (PFS) along with overall survival (OS). Results We identified 247 patients with metastatic cancer who received CS concurrently with CPIs. The median time on CS was 1.8 months. After adjusting for treatment type, tumor type, brain metastases, and irAEs, those treated with CS ≥2 months after starting CPI had a statistically significant longer PFS (HR=0.30, p<0.001), and OS (HR 0.34, p<0.0001) than those who received CS <2 months after starting CPI. Objective response rate (ORR) for patients on CS ≥2 months was 39.8%, versus ORR for patients <2 months was 14.7% (p value =<0.001) Conclusion Our results suggest that early use of CS during CPI treatment significantly hinders CPI efficacy. This data needs to be validated prospectively. Future studies should focus on the immune mechanisms by which CSs affect T-cell function early in the CPI treatment course.
Collapse
Affiliation(s)
- Diana V Maslov
- Internal Medicine, Ochsner Medical Center-New Orleans, New Orleans, Louisiana, USA
| | - Karine Tawagi
- Hematology/Oncology, Ochsner Medical Center-New Orleans, New Orleans, Louisiana, USA
| | - Madhav Kc
- Public Health, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Victoria Simenson
- Internal Medicine, Ochsner Medical Center-New Orleans, New Orleans, Louisiana, USA
| | - Helen Yuan
- Internal Medicine, Ochsner Medical Center-New Orleans, New Orleans, Louisiana, USA
| | - Cameron Parent
- Internal Medicine, Ochsner Medical Center-New Orleans, New Orleans, Louisiana, USA
| | - Adi Bamnolker
- Internal Medicine, Ochsner Medical Center-New Orleans, New Orleans, Louisiana, USA
| | - Richa Goel
- Internal Medicine, Ochsner Medical Center-New Orleans, New Orleans, Louisiana, USA
| | - Zoe Blake
- Medical School, The University of Queensland School of Medicine, Herston, Queensland, Australia
| | - Marc R Matrana
- Hematology/Oncology, Ochsner Cancer Institute, New Orleans, Louisiana, USA
| | - Daniel H Johnson
- Hematology/Oncology, Ochsner Cancer Institute, New Orleans, Louisiana, USA
| |
Collapse
|
13
|
Passiglia F, Cetoretta V, De Filippis M, Napoli V, Novello S. Exploring the immune-checkpoint inhibitors' efficacy/tolerability in special non-small cell lung cancer (NSCLC) populations: focus on steroids and autoimmune disease. Transl Lung Cancer Res 2021; 10:2876-2889. [PMID: 34295686 PMCID: PMC8264339 DOI: 10.21037/tlcr-20-635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 10/09/2020] [Indexed: 11/18/2022]
Abstract
The advent of immune-checkpoint inhibitors targeting the programmed cell death-1 (PD-1)/programmed death ligand-1 (PD-L1) axis, both as monotherapy and in combination strategies, produced a paradigm change of the treatment algorithm for metastatic, non-oncogene addicted, non-small cell lung cancer (NSCLC) patients. Although the great efficacy and the optimal tolerability emerging from clinical studies has been confirmed for the majority of patients treated in the real-word scenario, however the potential activity and safety profile of these agents in uncommon NSCLC populations remains still controversial. Particularly, patients with previously diagnosed autoimmune disease or concomitant steroids treatment at the time of immunotherapy initiation represent two special subgroups of patients not unusual in the real-word practice, to whom the clinical implication of immune-checkpoint inhibitors administration is largely unknown. In this review we provided an updated literature overview, summarizing available evidence and reporting practical suggestions, which may guide physicians in their clinical management of these NSCLC sub-populations.
Collapse
Affiliation(s)
- Francesco Passiglia
- Department of Oncology, S. Luigi Gonzaga Hospital, University of Turin, Orbassano (TO), Italy
| | - Valeria Cetoretta
- Department of Oncology, S. Luigi Gonzaga Hospital, University of Turin, Orbassano (TO), Italy
| | - Marco De Filippis
- Department of Oncology, S. Luigi Gonzaga Hospital, University of Turin, Orbassano (TO), Italy
| | - Valerio Napoli
- Department of Oncology, S. Luigi Gonzaga Hospital, University of Turin, Orbassano (TO), Italy
| | - Silvia Novello
- Department of Oncology, S. Luigi Gonzaga Hospital, University of Turin, Orbassano (TO), Italy
| |
Collapse
|
14
|
Robert C, Hwu WJ, Hamid O, Ribas A, Weber JS, Daud AI, Hodi FS, Wolchok JD, Mitchell TC, Hersey P, Dronca R, Joseph RW, Boutros C, Min L, Long GV, Schachter J, Puzanov I, Dummer R, Lin J, Ibrahim N, Diede SJ, Carlino MS, Joshua AM. Long-term safety of pembrolizumab monotherapy and relationship with clinical outcome: A landmark analysis in patients with advanced melanoma. Eur J Cancer 2020; 144:182-191. [PMID: 33360855 PMCID: PMC8388128 DOI: 10.1016/j.ejca.2020.11.010] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 10/21/2020] [Accepted: 11/04/2020] [Indexed: 12/16/2022]
Abstract
Objective: Long-term safety of pembrolizumab in melanoma was analyzed in KEYNOTE-001, KEYNOTE-002, and KEYNOTE-006. Patients and methods: Analysis involved patients who received ≥1 pembrolizumab dose. Lead-time bias was addressed via landmark analyses in patients who were progression-free before day 147. Results: Adverse events (AEs) were analyzed for 1567 patients (median follow-up, 42.4 months). Most AEs were mild/moderate; grade 3/4 treatment-related AEs occurred in 17.7% of patients. Two pembrolizumab-related deaths occurred. Any-grade immune-mediated AEs (imAEs) occurred in 23.0%, most commonly hypothyroidism (9.1%), pneumonitis (3.3%), and hyperthyroidism (3.0%); grade 3/4 imAEs occurred in 6.9% of patients. Most imAEs occurred within 16 weeks of treatment. In landmark analysis, patients who did (n = 79) versus did not (n = 384) develop imAEs had similar objective response rates (ORRs) (64.6% versus 63.0%); median time to response (TTR), 5.6 months for both; median duration of response (DOR), 20.0 versus 25.3 months; median progression-free survival (PFS), 17.0 versus 17.7 months; median overall survival (OS), not reached (NR) versus 43 months (p = 0.1104). Patients who did (n = 17) versus did not (n = 62) receive systemic corticosteroids had similar ORRs (70.6% vs. 62.9%) and median TTR(6.4 vs. 5.6 months) but numerically shorter median PFS(9.9 vs. 17.0 months); median DOR, 14.2 months versus NR; median OS, NR for both. Conclusions: These results enhance the knowledge base for pembrolizumab in advanced melanoma, with no new toxicity signals after lengthy follow-up of a large population. In landmark analyses, pembrolizumab efficacy was similar regardless of imAEs or systemic corticosteroid use. Clinical trial registry: NCT01295827, NCT01704287, NCT01866319.
Collapse
Affiliation(s)
- Caroline Robert
- Department of Oncology, Service of Dermatology, Institut de Cancérologie Gustave Roussy, 114 Rue Edouard Vaillant, Gustave Roussy, Villejuif, 94805, France; Paris-Saclay University, CNRS UMR 3348, Orsay, France.
| | - Wen-Jen Hwu
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
| | - Omid Hamid
- Department of Hematology/Oncology, The Angeles Clinic and Research Institute, a Cedars-Sinai Affiliate, 11800 Wilshire Blvd Suite 300, Los Angeles, CA, 90025, USA.
| | - Antoni Ribas
- Department of Medicine and the Jonsson Comprehensive Cancer Center, University of California, Los Angeles (UCLA), 11-934 Factor bldg., Los Angeles, CA, 90095, USA.
| | - Jeffrey S Weber
- Department of Medicine, Laura and Isaac Perlmutter Cancer Center at NYU Langone Health, 522 First Avenue, 1310 Smilow Building, New York, NY, 10016, USA.
| | - Adil I Daud
- Department of Medicine, University of California San Francisco, 1600 Divisadero St, San Francisco, CA, 94158, USA.
| | - F Stephen Hodi
- Melanoma Center and Center for Immuno-Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA.
| | - Jedd D Wolchok
- Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA.
| | - Tara C Mitchell
- Division of Hematology and Oncology, Abramson Cancer Center, University of Pennsylvania, 3400 Civic Center Blvd South Pavilion, Flr 10, Philadelphia, PA, 19104, USA.
| | - Peter Hersey
- Department of Medicine, The University of Sydney, Camperdown, NSW, 2006, Australia; Centenary Institute, Misendon Rd, Bldg 93, Camperdown, NSW, 2050, Australia; Melanoma Institute Australia, The University of Sydney, 40 Rocklands Rd, Wollstonecraft, NSW, 2065, Australia.
| | - Roxana Dronca
- Department of Hematology/Oncology, Mayo Clinic, 200 1st St. SW, Rochester, MN, 55905, USA.
| | - Richard W Joseph
- Division of Cancer Medicine, Mayo Clinic Cancer Center-Florida, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA.
| | - Celine Boutros
- Department of Oncology, Service of Dermatology, Institut de Cancérologie Gustave Roussy, 114 Rue Edouard Vaillant, Gustave Roussy, Villejuif, 94805, France.
| | - Le Min
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, 221 Longwood Avenue, Boston, MA, 02115, USA.
| | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney, 40 Rocklands Rd, Wollstonecraft, NSW, 2065, Australia; Royal North Shore Hospital, Reserve Road, St Leonards, NSW, 2065, Australia; Mater Hospital, Wollstonecraft, NSW, 2065, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia.
| | - Jacob Schachter
- Division of Oncology, Level 2 Cancer Center, Sheba Medical Center, Tel HaShomer Hospital, Emek HaEla Street 1, Tel HaShomer, Ramat Gan, 52621, Israel.
| | - Igor Puzanov
- Melanoma Section Department of Medicine, Roswell Park Comprehensive Cancer Center, 915 CSC Building, Elm & Carlton Streets, Buffalo, NY, 14263, USA.
| | - Reinhard Dummer
- Department of Dermatology, University Hospital Zürich, University of Zurich, Rämistrasse 100, Zürich, 8091, Switzerland.
| | - Jianxin Lin
- Department of Clinical Oncology, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA.
| | - Nageatte Ibrahim
- Department of Clinical Oncology, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA.
| | - Scott J Diede
- Department of Clinical Oncology, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA.
| | - Matteo S Carlino
- Department of Medicine, The University of Sydney, Camperdown, NSW, 2006, Australia; Melanoma Institute Australia, The University of Sydney, 40 Rocklands Rd, Wollstonecraft, NSW, 2065, Australia; Department of Medical Oncology, The Crown Princess Mary Cancer Centre, Westmead Hospital, 12 Moris Road, Westmead, NSW, 2145, Australia; Blacktown Hospital, 18 Blacktown Road, Blacktown, NSW, 2148, Australia.
| | - Anthony M Joshua
- Melanoma Institute Australia, The University of Sydney, 40 Rocklands Rd, Wollstonecraft, NSW, 2065, Australia; Department of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, 610 University Ave, Toronto, ON, M5G2C1, Canada; The Kinghorn Cancer Centre at St Vincent's Hospital, 370 Victoria St, Darlinghurst, NSW, 2010, Australia; St Vincent's Clinical School, UNSW Sydney, Victoria St, Darlinghurst, NSW, 2010, Australia.
| |
Collapse
|
15
|
Aldea M, Orillard E, Mansi L, Marabelle A, Scotte F, Lambotte O, Michot JM. How to manage patients with corticosteroids in oncology in the era of immunotherapy? Eur J Cancer 2020; 141:239-251. [DOI: 10.1016/j.ejca.2020.09.032] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/15/2020] [Accepted: 09/25/2020] [Indexed: 02/08/2023]
|
16
|
Prigent K, Aide N. 18F-Fludeoxyglucose PET/Computed Tomography for Assessing Tumor Response to Immunotherapy and Detecting Immune-Related Side Effects: A Checklist for the PET Reader. PET Clin 2020; 15:1-10. [PMID: 31735296 DOI: 10.1016/j.cpet.2019.08.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
After a short summary of the biological basis of the immune checkpoint inhibitors used for the treatment of nonhematologic solid tumors, the issues of pseudoprogression, hyperprogression, and immune-related side effects are discussed as well as their implications for patient management. Recommendations are provided for performing 18F-Fludeoxyglucose PET scanning, assessing tumor response, and reporting immune-related side effects, with representative clinical cases.
Collapse
Affiliation(s)
- Kevin Prigent
- Nuclear Medicine Department, University Hospital, Caen, France; Service de Médecine Nucléaire, CHU de CAEN, Avenue Côte de Nacre, Caen, France
| | - Nicolas Aide
- Service de Médecine Nucléaire, CHU de CAEN, Avenue Côte de Nacre, Caen, France; University of Normandy, France.
| |
Collapse
|
17
|
Collins M, Soularue E, Marthey L, Carbonnel F. Management of Patients With Immune Checkpoint Inhibitor-Induced Enterocolitis: A Systematic Review. Clin Gastroenterol Hepatol 2020; 18:1393-1403.e1. [PMID: 32007539 DOI: 10.1016/j.cgh.2020.01.033] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 01/23/2020] [Accepted: 01/24/2020] [Indexed: 02/07/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have improved the treatment of several cancers. These drugs increase T-cell activity and the antitumor immune response but also have immune-related adverse effects that can affect the gastrointestinal (GI) tract. These adverse effects have been observed in 7% to 30% of patients treated with ICIs. As the number of diseases treated with ICIs increases, gastroenterologists will see more patients with ICI-induced GI adverse events. We performed a systematic review of the incidence, risk factors, clinical manifestations, and management of the adverse effects of ICIs on the GI tract. Treatment with anti-cytotoxic T-lymphocyte-associated antigen-4 often causes severe enterocolitis, whereas treatment with inhibitors of programmed cell death 1 have less frequent and more diverse adverse effects. Management of patients with GI adverse effects of ICIs should involve first ruling out other disorders, followed by assessment of severity, treatment with corticosteroids, and rapid introduction of infliximab therapy for nonresponders.
Collapse
Affiliation(s)
- Michael Collins
- Department of Gastroenterology, Kremlin Bicêtre Hospital, Assistance Publique-Hopitaux de Paris, Le Kremlin Bicêtre, France; Paris Saclay University, Le Kremlin Bicêtre, France; INSERM, U1193, Paul-Brousse University Hospital, Hepatobiliary Centre, Villejuif, France
| | - Emilie Soularue
- Department of Gastroenterology, Kremlin Bicêtre Hospital, Assistance Publique-Hopitaux de Paris, Le Kremlin Bicêtre, France
| | - Lysiane Marthey
- Department of Gastroenterology, Kremlin Bicêtre Hospital, Assistance Publique-Hopitaux de Paris, Le Kremlin Bicêtre, France
| | - Franck Carbonnel
- Department of Gastroenterology, Kremlin Bicêtre Hospital, Assistance Publique-Hopitaux de Paris, Le Kremlin Bicêtre, France; Paris Saclay University, Le Kremlin Bicêtre, France; INSERM 1018, UPS, UVSQ Institut Gustave Roussy, Villejuif Cedex, France.
| |
Collapse
|
18
|
The glucocorticoids prednisone and dexamethasone differentially modulate T cell function in response to anti-PD-1 and anti-CTLA-4 immune checkpoint blockade. Cancer Immunol Immunother 2020; 69:1423-1436. [PMID: 32246174 DOI: 10.1007/s00262-020-02555-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 03/26/2020] [Indexed: 12/14/2022]
Abstract
On-treatment steroids for countering immune checkpoint inhibitor-induced inflammatory responses (irAEs) are a hallmark of cancer immunotherapy. However, the suppressive nature of steroids has raised questions regarding their ability to compromise the function of the 'proliferative burst' of effector T cells induced by immune checkpoint antibodies. We investigated the effector functions and the co-inhibitory receptor profile of stimulated peripheral blood mononuclear cells (PBMCs) pre-treated with prednisone and dexamethasone alone or in the presence of anti-PD-1/CTLA-4 antibodies. Also, clinical analysis of a patient who exhibited irAEs following combination (anti-PD-1/CTLA-4) in the presence of glucocorticoids was done. We found that prednisone in contrast to dexamethasone did not compromise T cell cytokine production (IL-2, IFN-γ and TNF-α) and proliferation in the absence or presence of anti-PD-1/CTLA-4 antibodies, when a physiological concentration was used. Neither single prednisone treatment nor co-treatment with checkpoint inhibitors impacted the expression of co-inhibitory receptors PD-1, CTLA-4, TIM-3 and LAG-3. In contrast, dexamethasone treatment promoted downregulation of LAG-3 expression by T cells. In addition, co-treatment of PD-1 + Jurkat cells with prednisone and/or dexamethasone with anti-PD-1 before stimulation significantly reduced SHP-2 phosphorylation, indicative of increased T cell function. Our findings hereby demonstrate a differential steroid effect on T cell function, which should be taken into consideration for patients undergoing immunotherapy. Also, the clinical analysis of a patient who exhibited irAEs following combination (anti-PD-1/CTLA-4) therapy indicated complete metabolic response in the presence of glucocorticoids. Therefore, concomitant use of prednisone does not appear to interfere with the function of immune checkpoint blockade.
Collapse
|
19
|
Zheng K, Qiu W, Wang H, Si X, Zhang X, Zhang L, Li X. Clinical recommendations on diagnosis and treatment of immune checkpoint inhibitor-induced renal immune-related adverse events. Thorac Cancer 2020; 11:1746-1751. [PMID: 32232975 PMCID: PMC7262914 DOI: 10.1111/1759-7714.13405] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 03/05/2020] [Indexed: 12/27/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) are nowadays widely used in clinical oncology treatment, and significantly improve the prognosis of cancer patients. However, overactivation of T cells and related signaling pathways caused by ICIs can also induce immune‐related adverse effects (irAEs). Renal immune side‐effects are relatively rare, but some are serious and fatal. Acute kidney injury (AKI), diagnosed mainly by percentage increases in serum creatinine (sCr), is the most common clinical manifestation, while acute tubulointerstitial nephritis (ATIN) is the main cause of ICI‐related AKI. Urinalysis analysis and sediment evaluation, 24 hour urine protein and sCr are helpful in screening and monitoring renal irAEs. Multiple potential causes for AKI are involved during cancer therapy, and should be differentiated from the immune mechanisms of ICIs. Under these circumstances, a renal biopsy should be considered which is essential for clinical decision‐making. Steroids are an effective treatment option for renal irAEs. Most patients who experience ICI‐related ATIN achieve a partial or complete renal recovery with prompt diagnosis and treatment. Multidisciplinary collaborations of different specialists will improve the effectiveness and outcome in the management of ICI irAEs.
Collapse
Affiliation(s)
- Ke Zheng
- Department of Nephrology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Wei Qiu
- Department of Internal Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Hanping Wang
- Department of Respirology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaoyan Si
- Department of Respirology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaotong Zhang
- Department of Respirology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Li Zhang
- Department of Respirology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Xuemei Li
- Department of Nephrology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
20
|
Gupta S, Cortazar FB, Riella LV, Leaf DE. Immune Checkpoint Inhibitor Nephrotoxicity: Update 2020. KIDNEY360 2020; 1:130-140. [PMID: 35372904 PMCID: PMC8809100 DOI: 10.34067/kid.0000852019] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Immune checkpoint inhibitors (ICPIs) have transformed the landscape of oncology, but are associated with a variety of autoimmune adverse events, including AKI. ICPI-associated AKI (ICPI-AKI) is emerging as an increasingly frequent cause of AKI in patients with cancer, and poses unique diagnostic and management challenges to clinicians who care for these patients. In this review, we describe the incidence and risk factors for ICPI-AKI, including proton pump inhibitor use, CKD, and combination immunotherapy. We discuss the limitations of the various definitions used for ICPI-AKI in prior studies, and propose a novel classification system (definite, probable, and possible ICPI-AKI) that recognizes the diagnostic uncertainty inherent in many cases. We discuss the key clinicopathologic features and treatment strategies for ICPI-AKI, including the role of kidney biopsy versus empirical treatment with steroids. We also explore the under-studied area of ICPI use in the setting of solid organ transplantation, where nephrologists and oncologists must balance the risk of rejection versus treating the underlying malignancy. Finally, we summarize existing data on the role of ICPI rechallenge after an episode of ICPI-AKI.
Collapse
Affiliation(s)
- Shruti Gupta
- Division of Renal Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Frank B. Cortazar
- New York Nephrology Vasculitis and Glomerular Center, Albany, New York; and
- Division of Nephrology, Massachusetts General Hospital, Boston, Massachusetts
| | - Leonardo V. Riella
- Division of Renal Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| | - David E. Leaf
- Division of Renal Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| |
Collapse
|
21
|
Kelly WJ, Gilbert MR. Glucocorticoids and immune checkpoint inhibitors in glioblastoma. J Neurooncol 2020; 151:13-20. [PMID: 32108294 DOI: 10.1007/s11060-020-03439-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 02/14/2020] [Indexed: 12/19/2022]
Abstract
PURPOSE Immunotherapy, activation of the immune system to target tumor cells, represents a paradigm shift in the treatment of cancer. Immune checkpoint therapies, which target immunomodulatory molecules expressed on T-lymphocytes, have demonstrated improved survival in a variety of malignancies. However, benefit in glioblastoma, the most common and devastating malignant brain tumor, remains to be seen. With several recent clinical trials failing to show efficacy of immunotherapy, concerns have been raised regarding the impact of glucocorticoid use in this patient population that may impair the ability for immune checkpoint inhibitors to affect a response. METHODS For this article we examined the mechanism by which immune checkpoint inhibitors activate, and glucocorticoids impair, T-lymphocyte function. RESULTS In this context, we review the clinical data of immune checkpoint inhibitors in glioblastoma as well as the impact glucocorticoids have on immune checkpoint inhibitor efficacy. Finally, we highlight key questions that remain in the field, and the potential benefit of further research for central nervous system tumors. CONCLUSION More information on the extent, character and duration of glucocorticoids on patients treated with PD-(L)1 will better inform both clinical management and novel therapeutic development of immunotherapy in patients with CNS malignancies.
Collapse
Affiliation(s)
- William J Kelly
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Building 82, Room 235, 9030 Old Georgetown Road, Bethesda, MD, 20892, USA
| | - Mark R Gilbert
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Building 82, Room 235, 9030 Old Georgetown Road, Bethesda, MD, 20892, USA.
| |
Collapse
|
22
|
Abstract
PURPOSE OF REVIEW This review addresses our current knowledge of immune-mediated colitis (IMC) and offers a practical guide to its management. RECENT FINDINGS Due to the similarity in clinical, endoscopic, and histologic findings between IMC and inflammatory bowel disease (IBD), gastroenterologists have tailored their approach to IMC management to that of IBD. Immune checkpoint inhibitors (ICIs) are monoclonal antibodies that augment the T-cell anti-tumor response of the immune system and have demonstrated their importance in the treatment of a wide range of malignancies. With the growing benefits of ICIs, there are immune-related adverse events (irAEs) that mirror many known autoimmune diseases. Diarrhea and IMC are the most common and severe irAEs noted. No standardized guidelines exist in the management of these irAEs.
Collapse
Affiliation(s)
- Tara Menon
- The Ohio State University Inflammatory Bowel Disease Center, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| | - Anita Afzali
- The Ohio State University Inflammatory Bowel Disease Center, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| |
Collapse
|
23
|
Touhami S, Audo I, Terrada C, Gaudric A, LeHoang P, Touitou V, Bodaghi B. Neoplasia and intraocular inflammation: From masquerade syndromes to immunotherapy-induced uveitis. Prog Retin Eye Res 2019; 72:100761. [DOI: 10.1016/j.preteyeres.2019.05.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 04/16/2019] [Accepted: 05/01/2019] [Indexed: 12/18/2022]
|
24
|
Aston WJ, Hope DE, Cook AM, Boon L, Dick I, Nowak AK, Lake RA, Lesterhuis WJ. Dexamethasone differentially depletes tumour and peripheral blood lymphocytes and can impact the efficacy of chemotherapy/checkpoint blockade combination treatment. Oncoimmunology 2019; 8:e1641390. [PMID: 31646089 PMCID: PMC6791454 DOI: 10.1080/2162402x.2019.1641390] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 07/03/2019] [Accepted: 07/04/2019] [Indexed: 10/26/2022] Open
Abstract
Dexamethasone is a synthetic glucocorticoid commonly used for the prevention and management of side effects in cancer patients undergoing chemotherapy. While it is effective as an anti-emetic and in preventing hypersensitivity reactions, dexamethasone depletes peripheral blood lymphocytes and impacts immune responses. The effect of dexamethasone on the number and quality of tumour-infiltrating leukocytes has not been reported. To address this, we calibrated the dose in two different strains of mice to achieve the same extent of peripheral blood lymphocyte depletion observed in patients with cancer. Doses that caused analogous depletion of T and B lymphocytes and NK cells from the peripheral blood, elicited no change in these populations within the tumour. The expression of immune checkpoint molecules PD-1, OX40, GITR and TIM3 on tumour-infiltrating lymphocytes was not altered. We found that dexamethasone had a small but significant deleterious impact on weakly efficacious chemoimmunotherapy but had no effect when the protocol was highly efficacious. Based on these results, we predict that dexamethasone will have a modest negative influence on the overall effectiveness of chemoimmunotherapy treatment.
Collapse
Affiliation(s)
- Wayne J. Aston
- National Centre for Asbestos Related Diseases, The University of Western Australia, Nedlands, WA, Australia
- Medical School, The University of Western Australia, Nedlands, WA, Australia
| | - Danika E. Hope
- National Centre for Asbestos Related Diseases, The University of Western Australia, Nedlands, WA, Australia
- School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia
| | - Alistair M. Cook
- National Centre for Asbestos Related Diseases, The University of Western Australia, Nedlands, WA, Australia
- Medical School, The University of Western Australia, Nedlands, WA, Australia
| | | | - Ian Dick
- National Centre for Asbestos Related Diseases, The University of Western Australia, Nedlands, WA, Australia
- School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia
| | - Anna K. Nowak
- National Centre for Asbestos Related Diseases, The University of Western Australia, Nedlands, WA, Australia
- Medical School, The University of Western Australia, Nedlands, WA, Australia
- Department of Medical Oncology, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
| | - Richard A. Lake
- National Centre for Asbestos Related Diseases, The University of Western Australia, Nedlands, WA, Australia
- School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia
| | - W. Joost Lesterhuis
- National Centre for Asbestos Related Diseases, The University of Western Australia, Nedlands, WA, Australia
- School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia
| |
Collapse
|
25
|
Li H, Verma V, Brooks ED, Feng L, Zhang T, Welsh JW, Lin SH, Gomez D, Gandhi S, Heymach JV, Chang JY. Impact of Corticosteroid Administration on Outcomes Following Stereotactic Ablative Radiotherapy for Non-small-cell Lung Cancer. Clin Lung Cancer 2019; 20:e480-e488. [PMID: 31078421 DOI: 10.1016/j.cllc.2019.03.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 02/27/2019] [Accepted: 03/03/2019] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Radiotherapy produces immune-promoting effects, which may be blunted by the delivery of corticosteroids (CS). We thus aimed to evaluate the impact of CS use on recurrence and survival outcomes of patients with early stage non-small-cell lung cancer treated with stereotactic ablative radiotherapy (SABR). MATERIALS AND METHODS A prospectively registered database of patients with stage I to II (T1-3N0M0) stage non-small-cell lung cancer treated with SABR from 2004 to 2015 was queried. Concurrent CS administration was defined as receipt of CS within 2 days of the SABR course. Statistics included Kaplan-Meier survival analysis, Cox proportional hazards modeling, and cumulative incidence analysis utilizing death as a competing risk. RESULTS Of 912 patients, 87 (9.5%) received CS with their SABR course. The most common agent was prednisone (64.4%). Indications for CS use were chronic obstructive pulmonary disease in 53 cases (60.9%), chemotherapy in 7 (8.0%), arthritis in 7 (8.0%), chronic pain in 4 (4.6%), transplant-related in 3 (3.4%), and "others" in 13 (14.9%; pneumonia, asthma, anemia, etc.). The median follow-up time was 59.3 months. Compared with patients who did not receive CS, receipt of CS was associated with poorer overall survival (P = .004). However, CS administration was not associated with worse time to progression (P = .766) or any recurrence when using death as a competing risk (local P = .119, regional P = .449, distant P = .847, and any recurrence P = .708). Toxicity rates were not statistically different between cohorts. CONCLUSIONS These data do not suggest increased recurrence rates when patients undergoing SABR are administered corticosteroids. However, owing to limitations of retrospective analyses, individualized judgment is still recommended.
Collapse
Affiliation(s)
- Hongqi Li
- Department of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX; Department of Radiation Oncology, Airforce General Hospital PLA, Beijing, China
| | - Vivek Verma
- Department of Radiation Oncology, Allegheny General Hospital, Pittsburgh, PA
| | - Eric D Brooks
- Department of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Lei Feng
- Department of Biostatistics, The University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Tiening Zhang
- Department of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - James W Welsh
- Department of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Steven H Lin
- Department of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Daniel Gomez
- Department of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Saumil Gandhi
- Department of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - John V Heymach
- Department of Thoracic Head and Neck Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Joe Y Chang
- Department of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX.
| |
Collapse
|
26
|
Ueno R, Nemoto M, Uegami W, Fukuoka J, Misawa M. Pembrolizumab-induced pneumonitis with a perilymphatic nodular pattern in a lung cancer patient: A radio-pathologic correlation. Respir Med Case Rep 2019; 26:168-170. [PMID: 30671338 PMCID: PMC6327904 DOI: 10.1016/j.rmcr.2019.01.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 01/03/2019] [Accepted: 01/03/2019] [Indexed: 01/26/2023] Open
Abstract
A 66-year-old Japanese man with recurrent adenocarcinoma of the lung p-stage IIIA (pT2bN2M0; version 8) on pembrolizumab was present with gradually worsening dyspnea. Although history and physical examination were unremarkable, high-resolution CT showed the perilymphatic distribution of the pembrolizumab-induced pneumonitis. Consistent with the CT result, biopsy revealed the aggregation of the cytotoxic (CD8+) T-lymphocytes around the lymph tracts. Given the clinical, radiological and pathological findings, pembrolizumab-induced pneumonitis was confirmed. The patient was discharged after terminating the pembrolizumab with ameliorated symptoms. This report, in conjunction with existing literature, illustrates the wide variety of the pembrolizumab-induced pneumonitis and bolsters the current understanding of its pathophysiology.
Collapse
Affiliation(s)
- Ryo Ueno
- Department of Intensive Care Medicine, Kameda Medical Center, Kamogawa, Chiba, Japan
| | - Masahiro Nemoto
- Department of Pulmonary Medicine, Kameda Medical Center, Kamogawa, Chiba, Japan
| | - Wataru Uegami
- Department of Pathology, Kameda Medical Center, Kamogawa, Chiba, Japan
| | - Junya Fukuoka
- Department of Pathology, Kameda Medical Center, Kamogawa, Chiba, Japan
| | - Masafumi Misawa
- Department of Pulmonary Medicine, Kameda Medical Center, Kamogawa, Chiba, Japan
| |
Collapse
|
27
|
Izzedine H, Mathian A, Champiat S, Picard C, Mateus C, Routier E, Varga A, Malka D, Leary A, Michels J, Michot JM, Marabelle A, Lambotte O, Amoura Z, Soria JC, Kaaki S, Quellard N, Goujon JM, Brocheriou I. Renal toxicities associated with pembrolizumab. Clin Kidney J 2018; 12:81-88. [PMID: 30746132 PMCID: PMC6366307 DOI: 10.1093/ckj/sfy100] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 09/05/2018] [Indexed: 02/07/2023] Open
Abstract
Objective Expanded clinical experience with patients treated by pembrolizumab has accumulated. However, renal toxicities associated with this anti-programmed cell death 1 agent are poorly described because kidney histology is rarely sought. As a nephrology referral centre, we aimed to describe the clinic-biological and histopathological characteristics of pembrolizumab-related nephropathy and its response to treatment. Methods We conducted a monocentric large case series study, including all pembrolizumab-treated cancer patients presenting a renal toxicity addressed to our centre from 2015 to 2017. Results A total of 12 patients (7 men) out of 676 pembrolizumab-treated patients (incidence 1.77%) were included (median age 69.75 years). Patients were referred for acute kidney injury (n = 10) and/or proteinuria (n = 2). A kidney biopsy was performed in all patients, with a median duration of use of 9 months (range 1–24 months) after the beginning of treatment. Biopsy showed that four patients had acute interstitial nephritis (AIN), whereas five had acute tubular injury (ATI) alone, one had minimal change disease (MCD) and ATI, and one had MCD alone. Pembrolizumab withdrawal coupled with corticosteroid therapy was the most effective treatment for kidney function recovery. Drug reintroduction resulted in a more severe recurrence of AIN in one patient who required maintenance of pembrolizumab. Two patients died of cancer progression with one of them developing severe renal failure requiring dialysis. Conclusion In our series, ATI, AIN and MCD are the most frequent forms of kidney involvement under pembrolizumab therapy. Kidney dysfunction is usually isolated but can be severe. Use of corticosteroids in case of AIN improves the glomerular filtration rate.
Collapse
Affiliation(s)
- Hassan Izzedine
- Department of Nephrology, Peupliers Private Hospital, Ramsay Générale de Santé, Paris, France.,UPEC (Université Paris Est Créteil), INSERM U955, Institut Mondor de Recherche Biomdicale (IMRB), Equipe, Créteil, France
| | - Alexis Mathian
- Department of Internal Medicine, Pitie-Salpetriere Hospital, Paris, France
| | - Stephane Champiat
- Drug Development Department (DITEP), Gustave Roussy, Villejuif, France
| | - Cécile Picard
- Department of Pathology, Pitie-Salpetriere Hospital, Paris, France
| | - Christine Mateus
- Department of Supportive Care, Gustave Roussy, Villejuif, France
| | - Emilie Routier
- Department of Medical Oncology, Dermatology Unit, Gustave Roussy, Villejuif, France
| | - Andrea Varga
- Drug Development Department (DITEP), Gustave Roussy, Villejuif, France
| | - David Malka
- Department of Medical Oncology, Gastrointestinal Cancer Group, Gustave Roussy, Villejuif, France
| | - Alexandra Leary
- Department of Medical Oncology, Gynecology Unit, Gustave Roussy, Villejuif, France
| | - Judith Michels
- Department of Medical Oncology, Gynecology Unit, Gustave Roussy, Villejuif, France
| | - Jean-Marie Michot
- Drug Development Department (DITEP), Gustave Roussy, Villejuif, France
| | | | - Olivier Lambotte
- Department of Internal Medicine and Clinical Immunology, Bicetre University Hospital, Le Kremlin Bicêtre, France
| | - Zahir Amoura
- Department of Internal Medicine, Pitie-Salpetriere Hospital, Paris, France
| | | | - Sihem Kaaki
- Departement of Pathology, Electron Microscopy Unit, CHU Poitiers, Poitiers, France
| | - Nathalie Quellard
- Departement of Pathology, Electron Microscopy Unit, CHU Poitiers, Poitiers, France
| | - Jean-Michel Goujon
- Departement of Pathology, Electron Microscopy Unit, CHU Poitiers, Poitiers, France
| | | |
Collapse
|
28
|
Soularue E, Lepage P, Colombel JF, Coutzac C, Faleck D, Marthey L, Collins M, Chaput N, Robert C, Carbonnel F. Enterocolitis due to immune checkpoint inhibitors: a systematic review. Gut 2018; 67:2056-2067. [PMID: 30131322 DOI: 10.1136/gutjnl-2018-316948] [Citation(s) in RCA: 178] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 07/11/2018] [Accepted: 07/19/2018] [Indexed: 12/14/2022]
Abstract
Immune checkpoint inhibitors targeting cytotoxic T-lymphocyte-associated protein-4 (CTLA-4) and programmed death-1 (PD-1)/ligand are increasingly used to treat several types of cancer. These drugs enhance antitumour T-cell activity and therefore induce immune-related adverse effects (irAE), of which gastrointestinal (GI) irAE are among the most frequent and severe. This systematic literature review summarises the clinical manifestations, management and pathophysiology of GI irAE due to immune checkpoint inhibitors. GI irAE induced by anti-CTLA-4 are frequent, potentially severe and resemble IBD, whereas those induced by PD-1 blockade seem to be less frequent and clinically more diverse. Baseline symbiotic gut microbiota is associated with an enhanced antitumour response to immune checkpoint inhibitors and an increased susceptibility to developing enterocolitis, in patients treated with anti-CTLA-4. These findings open new perspectives for possible manipulation of the gut microbiota in order to better identify responders to immune checkpoint inhibitors and to increase their efficacy and safety.
Collapse
Affiliation(s)
- Emilie Soularue
- Department of Gastroenterology, Kremlin Bicêtre Hospital, Assistance Publique- Hôpitaux de Paris (AP-HP), Le Kremlin Bicêtre, France.,Faculté de Médecine, University Paris-Saclay, Le Kremlin Bicêtre, France
| | - Patricia Lepage
- Micalis Institute, INRA, AgroParisTech, University Paris-Saclay, Jouy-en- Josas, France
| | - Jean Frederic Colombel
- Helmsley Inflammatory Bowel Disease Center, Icahn Medical School of Medicine at Mount Sinai, New York, USA
| | - Clelia Coutzac
- Laboratory of Immunomonitoring in Oncology and CNRS-UMS 3655 and INSERM-US23, Villejuif, France
| | - David Faleck
- Helmsley Inflammatory Bowel Disease Center, Icahn Medical School of Medicine at Mount Sinai, New York, USA
| | - Lysiane Marthey
- Department of Gastroenterology, Kremlin Bicêtre Hospital, Assistance Publique- Hôpitaux de Paris (AP-HP), Le Kremlin Bicêtre, France
| | - Michael Collins
- Department of Gastroenterology, Kremlin Bicêtre Hospital, Assistance Publique- Hôpitaux de Paris (AP-HP), Le Kremlin Bicêtre, France.,Faculté de Médecine, University Paris-Saclay, Le Kremlin Bicêtre, France
| | - Nathalie Chaput
- Laboratory of Immunomonitoring in Oncology and CNRS-UMS 3655 and INSERM-US23, Villejuif, France.,Faculté de Pharmacie, University Paris-Saclay, Chatenay-Malabry, France
| | - Caroline Robert
- Faculté de Médecine, University Paris-Saclay, Le Kremlin Bicêtre, France.,Départment of Medecine, Dermatology Unit, Villejuif, France
| | - Franck Carbonnel
- Department of Gastroenterology, Kremlin Bicêtre Hospital, Assistance Publique- Hôpitaux de Paris (AP-HP), Le Kremlin Bicêtre, France.,Faculté de Médecine, University Paris-Saclay, Le Kremlin Bicêtre, France
| |
Collapse
|
29
|
Scott SC, Pennell NA. Early Use of Systemic Corticosteroids in Patients with Advanced NSCLC Treated with Nivolumab. J Thorac Oncol 2018; 13:1771-1775. [PMID: 29935305 DOI: 10.1016/j.jtho.2018.06.004] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 06/13/2018] [Indexed: 01/10/2023]
Abstract
INTRODUCTION Checkpoint inhibitors augment the immune system's natural surveillance mechanisms and have increasing applications in NSCLC. Immunosuppressive corticosteroids are also frequently used in this population to treat unwanted inflammation. In view of this mechanistic opposition, we investigated the interaction between nivolumab and corticosteroids in patients with advanced NSCLC. METHODS A retrospective review of the charts of 210 patients with NSCLC who were treated with nivolumab at the Cleveland Clinic was performed. Use of systemic corticosteroids (equivalent to >10 mg of prednisone/d) during nivolumab therapy was associated with the objective outcomes number of nivolumab cycles and overall survival. RESULTS In all, 66 patients (31%) received concurrent systemic corticosteroids during nivolumab therapy. The most common indications included sequelae from active or treated brain metastases (27%) and chronic obstructive pulmonary disease or other respiratory disease (21%). For patients with early exposure to steroids (within the first 30 days of nivolumab therapy) (12% [n=25]), the median number of nivolumab cycles was 2, compared with five cycles in patients who were not exposed to corticosteroids (p = 0.002). The median overall survival time for patients who received steroids during the first 30 days was 4.3 months, compared with 11 months for patients who did not receive steroids (hazard ratio for death = 2.30, 95% confidence interval: CI 1.27-4.16, p = 0.006 in multivariate analysis). CONCLUSION Nearly one-third of patients with NSCLC treated with nivolumab were prescribed concurrent corticosteroids during the course of nivolumab therapy. Patients exposed to corticosteroids during the first cycle of nivolumab received fewer total cycles of nivolumab, suggesting decreased clinical benefit, and they had shorter overall survival.
Collapse
Affiliation(s)
- Susan C Scott
- Cleveland Clinic Taussig Cancer Institute, Cleveland, Ohio
| | | |
Collapse
|
30
|
Giles AJ, Hutchinson MKND, Sonnemann HM, Jung J, Fecci PE, Ratnam NM, Zhang W, Song H, Bailey R, Davis D, Reid CM, Park DM, Gilbert MR. Dexamethasone-induced immunosuppression: mechanisms and implications for immunotherapy. J Immunother Cancer 2018; 6:51. [PMID: 29891009 PMCID: PMC5996496 DOI: 10.1186/s40425-018-0371-5] [Citation(s) in RCA: 327] [Impact Index Per Article: 46.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 05/30/2018] [Indexed: 12/18/2022] Open
Abstract
Background Corticosteroids are routinely utilized to alleviate edema in patients with intracranial lesions and are first-line agents to combat immune-related adverse events (irAEs) that arise with immune checkpoint blockade treatment. However, it is not known if or when corticosteroids can be administered without abrogating the efforts of immunotherapy. The purpose of this study was to evaluate the impact of dexamethasone on lymphocyte activation and proliferation during checkpoint blockade to provide guidance for corticosteroid use while immunotherapy is being implemented as a cancer treatment. Methods Lymphocyte proliferation, differentiation, and cytokine production were evaluated during dexamethasone exposure. Human T cells were stimulated through CD3 ligation and co-stimulated either directly by CD28 ligation or by providing CD80, a shared ligand for CD28 and CTLA-4. CTLA-4 signaling was inhibited by antibody blockade using ipilimumab which has been approved for the treatment of several solid tumors. The in vivo effects of dexamethasone during checkpoint blockade were evaluated using the GL261 syngeneic mouse intracranial model, and immune populations were profiled by flow cytometry. Results Dexamethasone upregulated CTLA-4 mRNA and protein in CD4 and CD8 T cells and blocked CD28-mediated cell cycle entry and differentiation. Naïve T cells were most sensitive, leading to a decrease of the development of more differentiated subsets. Resistance to dexamethasone was conferred by blocking CTLA-4 or providing strong CD28 co-stimulation prior to dexamethasone exposure. CTLA-4 blockade increased IFNγ expression, but not IL-2, in stimulated human peripheral blood T cells exposed to dexamethasone. Finally, we found that CTLA-4 blockade partially rescued T cell numbers in mice bearing intracranial gliomas. CTLA-4 blockade was associated with increased IFNγ-producing tumor-infiltrating T cells and extended survival of dexamethasone-treated mice. Conclusions Dexamethasone-mediated T cell suppression diminishes naïve T cell proliferation and differentiation by attenuating the CD28 co-stimulatory pathway. However, CTLA-4, but not PD-1 blockade can partially prevent some of the inhibitory effects of dexamethasone on the immune response. Electronic supplementary material The online version of this article (10.1186/s40425-018-0371-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Amber J Giles
- Neuro-Oncology Branch, CCR, NCI, National Institutes of Health, 37 Convent Dr. Bldg. 37, Rm. 1142B, Bethesda, MD, 20892, USA.
| | - Marsha-Kay N D Hutchinson
- Neuro-Oncology Branch, CCR, NCI, National Institutes of Health, 37 Convent Dr. Bldg. 37, Rm. 1142B, Bethesda, MD, 20892, USA
| | - Heather M Sonnemann
- Neuro-Oncology Branch, CCR, NCI, National Institutes of Health, 37 Convent Dr. Bldg. 37, Rm. 1142B, Bethesda, MD, 20892, USA
| | - Jinkyu Jung
- Neuro-Oncology Branch, CCR, NCI, National Institutes of Health, 37 Convent Dr. Bldg. 37, Rm. 1142B, Bethesda, MD, 20892, USA
| | - Peter E Fecci
- Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
| | - Nivedita M Ratnam
- Neuro-Oncology Branch, CCR, NCI, National Institutes of Health, 37 Convent Dr. Bldg. 37, Rm. 1142B, Bethesda, MD, 20892, USA
| | - Wei Zhang
- Neuro-Oncology Branch, CCR, NCI, National Institutes of Health, 37 Convent Dr. Bldg. 37, Rm. 1142B, Bethesda, MD, 20892, USA
| | - Hua Song
- Neuro-Oncology Branch, CCR, NCI, National Institutes of Health, 37 Convent Dr. Bldg. 37, Rm. 1142B, Bethesda, MD, 20892, USA
| | - Rolanda Bailey
- Neuro-Oncology Branch, CCR, NCI, National Institutes of Health, 37 Convent Dr. Bldg. 37, Rm. 1142B, Bethesda, MD, 20892, USA
| | - Dionne Davis
- Neuro-Oncology Branch, CCR, NCI, National Institutes of Health, 37 Convent Dr. Bldg. 37, Rm. 1142B, Bethesda, MD, 20892, USA
| | - Caitlin M Reid
- Neuro-Oncology Branch, CCR, NCI, National Institutes of Health, 37 Convent Dr. Bldg. 37, Rm. 1142B, Bethesda, MD, 20892, USA
| | - Deric M Park
- Neuro-Oncology Branch, CCR, NCI, National Institutes of Health, 37 Convent Dr. Bldg. 37, Rm. 1142B, Bethesda, MD, 20892, USA
| | - Mark R Gilbert
- Neuro-Oncology Branch, CCR, NCI, National Institutes of Health, 37 Convent Dr. Bldg. 37, Rm. 1142B, Bethesda, MD, 20892, USA
| |
Collapse
|
31
|
Martini DJ, Hamieh L, McKay RR, Harshman LC, Brandao R, Norton CK, Steinharter JA, Krajewski KM, Gao X, Schutz FA, McGregor B, Bossé D, Lalani AKA, De Velasco G, Michaelson MD, McDermott DF, Choueiri TK. Durable Clinical Benefit in Metastatic Renal Cell Carcinoma Patients Who Discontinue PD-1/PD-L1 Therapy for Immune-Related Adverse Events. Cancer Immunol Res 2018; 6:402-408. [PMID: 29437040 DOI: 10.1158/2326-6066.cir-17-0220] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 10/02/2017] [Accepted: 01/26/2018] [Indexed: 11/16/2022]
Abstract
The current standard of care for treatment of metastatic renal cell carcinoma (mRCC) patients is PD-1/PD-L1 inhibitors until progression or toxicity. Here, we characterize the clinical outcomes for 19 mRCC patients who experienced an initial clinical response (any degree of tumor shrinkage), but after immune-related adverse events (irAE) discontinued all systemic therapy. Clinical baseline characteristics, outcomes, and survival data were collected. The primary endpoint was time to progression from the date of treatment cessation (TTP). Most patients had clear cell histology and received anti-PD-1/PD-L1 therapy as second-line or later treatment. Median time on PD-1/PD-L1 therapy was 5.5 months (range, 0.7-46.5) and median TTP was 18.4 months (95% CI, 4.7-54.3) per Kaplan-Meier estimation. The irAEs included arthropathies, ophthalmopathies, myositis, pneumonitis, and diarrhea. We demonstrate that 68.4% of patients (n = 13) experienced durable clinical benefit off treatment (TTP of at least 6 months), with 36% (n = 7) of patients remaining off subsequent treatment for over a year after their last dose of anti-PD-1/PD-L1. Three patients with tumor growth found in a follow-up visit, underwent subsequent surgical intervention, and remain off systemic treatment. Nine patients (47.4%) have ongoing irAEs. Our results show that patients who benefitted clinically from anti-PD-1/PD-L1 therapy can experience sustained beneficial responses, not needing further therapies after the initial discontinuation of treatment due to irAEs. Investigation of biomarkers indicating sustained benefit to checkpoint blockers are needed. Cancer Immunol Res; 6(4); 402-8. ©2018 AACR.
Collapse
Affiliation(s)
- Dylan J Martini
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.,Emory University School of Medicine, Atlanta, Georgia
| | - Lana Hamieh
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Rana R McKay
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.,University of California San Diego, La Jolla, California
| | - Lauren C Harshman
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Raphael Brandao
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Craig K Norton
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - John A Steinharter
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | | | - Xin Gao
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Fabio A Schutz
- BP-Beneficencia Portuguesa de Sao Paulo, Sao Paulo, Brazil
| | - Bradley McGregor
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Dominick Bossé
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Aly-Khan A Lalani
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Guillermo De Velasco
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.,Hospital Universitario, Madrid, Spain
| | - M Dror Michaelson
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - David F McDermott
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Toni K Choueiri
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
32
|
Gong J, Chehrazi-Raffle A, Reddi S, Salgia R. Development of PD-1 and PD-L1 inhibitors as a form of cancer immunotherapy: a comprehensive review of registration trials and future considerations. J Immunother Cancer 2018; 6:8. [PMID: 29357948 PMCID: PMC5778665 DOI: 10.1186/s40425-018-0316-z] [Citation(s) in RCA: 943] [Impact Index Per Article: 134.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Accepted: 01/09/2018] [Indexed: 12/13/2022] Open
Abstract
Early preclinical evidence provided the rationale for programmed cell death 1 (PD-1) and programmed death ligand 1 (PD-L1) blockade as a potential form of cancer immunotherapy given that activation of the PD-1/PD-L1 axis putatively served as a mechanism for tumor evasion of host tumor antigen-specific T-cell immunity. Early-phase studies investigating several humanized monoclonal IgG4 antibodies targeting PD-1 and PD-L1 in advanced solid tumors paved way for the development of the first PD-1 inhibitors, nivolumab and pembrolizumab, approved by the Food and Drug Administration (FDA) in 2014. The number of FDA-approved agents of this class is rapidly enlarging with indications for treatment spanning across a spectrum of malignancies. The purpose of this review is to highlight the clinical development of PD-1 and PD-L1 inhibitors in cancer therapy to date. In particular, we focus on detailing the registration trials that have led to FDA-approved indications of anti-PD-1 and anti-PD-L1 therapies in cancer. As the number of PD-1/PD-L1 inhibitors continues to grow, predictive biomarkers, mechanisms of resistance, hyperprogressors, treatment duration and treatment beyond progression, immune-related toxicities, and clinical trial design are key concepts in need of further consideration to optimize the anticancer potential of this class of immunotherapy.
Collapse
Affiliation(s)
- Jun Gong
- Department of Medical Oncology, City of Hope National Medical Center, 1500 E Duarte St, Duarte, CA 91010 USA
| | - Alexander Chehrazi-Raffle
- Department of Internal Medicine, Harbor-UCLA Medical Center, 1000 W Carson St, Torrance, CA 90509 USA
| | - Srikanth Reddi
- Department of Internal Medicine, Harbor-UCLA Medical Center, 1000 W Carson St, Torrance, CA 90509 USA
| | - Ravi Salgia
- Medical Oncology and Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Building 51, Room 101, 1500 E Duarte St, Duarte, CA 91010 USA
| |
Collapse
|
33
|
Izzedine H, Mateus C, Boutros C, Robert C, Rouvier P, Amoura Z, Mathian A. Renal effects of immune checkpoint inhibitors. Nephrol Dial Transplant 2018; 32:936-942. [PMID: 28025384 DOI: 10.1093/ndt/gfw382] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 09/22/2016] [Indexed: 12/31/2022] Open
Abstract
Recent advances in immune checkpoint inhibitor (ICPI) development have led to major improvements in oncology patient outcomes. Cytotoxic T-lymphocyte antigen 4 (CTLA-4) and programmed cell death protein 1 (PD-1) are two essential immune checkpoint receptors. Ipilimumab and tremelimumab (anti-CTLA-4-blocking antibodies) and pembrolizumab and nivolumab (antibodies targeting PD-1 receptors) have already been approved by US Food and Drug Administration in several malignancies. Two different forms of ICPI-induced renal damage have been identified, including acute (granulomatous) tubulointerstitial nephritis and immune complex glomerulonephritis. The observed acute renal damage can be reversed upon ICPI drug discontinuation and renal function can recover back to normal following the introduction of systemic corticosteroid treatment. Any delay in treating this complication could result in definitive and irreversible renal injury.
Collapse
Affiliation(s)
- Hassan Izzedine
- Department of Nephrology, Monceau Park International Clinic Paris, France
| | - Christine Mateus
- Department of Oncology, Gustave Roussy Institute, Villejuif, France
| | - Céline Boutros
- Department of Oncology, Gustave Roussy Institute, Villejuif, France
| | - Caroline Robert
- Department of Oncology, Gustave Roussy Institute, Villejuif, France
| | - Philippe Rouvier
- Department of Pathology, Pitie-Salpetriere Hospital, Paris, France
| | - Zahir Amoura
- Department of Internal Medicine 2, Pitie-Salpetriere Hospital, Paris, France
| | - Alexis Mathian
- Department of Internal Medicine 2, Pitie-Salpetriere Hospital, Paris, France
| |
Collapse
|
34
|
Tanaka R, Fujisawa Y, Sae I, Maruyama H, Ito S, Hasegawa N, Sekine I, Fujimoto M. Severe hepatitis arising from ipilimumab administration, following melanoma treatment with nivolumab. Jpn J Clin Oncol 2017; 47:175-178. [PMID: 28173241 DOI: 10.1093/jjco/hyw167] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 10/03/2016] [Accepted: 10/25/2016] [Indexed: 01/08/2023] Open
Abstract
After 4 weeks of the last dose of nivolumab, a 59-year-old man with stage IV melanoma was subject to treatment with ipilimumab. After 5 weeks, the patient developed severe hepatitis, showing markedly elevated levels of both aspartate aminotransferase and alanine aminotransferase (>2000 U/l). Using pulse steroid therapy with 1000 mg/d of methylprednisolone, liver function initially improved, but then deteriorated upon dosage reduction. Subsequently, mycophenolate mofetil (MMF) was administered at a dose of 2 g/d in addition to the corticosteroid, which resulted in aspartate aminotransferase and alanine aminotransferase levels gradually improving to grade 1, and the corticosteroid dose was successfully reduced to 0.5 mg/kg/d of oral prednisolone. Liver function then remained stable when MMF was tapered. In conclusion, the use of MMF improved liver function in this patient with steroid-refractory hepatitis induced by immune checkpoint inhibitor administration.
Collapse
Affiliation(s)
- Ryota Tanaka
- Department of Dermatology, University of Tsukuba, Ibaraki, Japan
| | | | - Inoue Sae
- Department of Dermatology, University of Tsukuba, Ibaraki, Japan.,Department of Dermatology, Hitachi General Hospital, Ibaraki, Japan
| | - Hiroshi Maruyama
- Department of Dermatology, University of Tsukuba, Ibaraki, Japan
| | - Shusaku Ito
- Department of Dermatology, Hitachi General Hospital, Ibaraki, Japan
| | - Naoyuki Hasegawa
- Department of Gastroenterology, University of Tsukuba, Ibaraki, Japan
| | - Ikuo Sekine
- Department of Medical Oncology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Manabu Fujimoto
- Department of Dermatology, University of Tsukuba, Ibaraki, Japan
| |
Collapse
|
35
|
Garant A, Guilbault C, Ekmekjian T, Greenwald Z, Murgoi P, Vuong T. Concomitant use of corticosteroids and immune checkpoint inhibitors in patients with hematologic or solid neoplasms: A systematic review. Crit Rev Oncol Hematol 2017; 120:86-92. [DOI: 10.1016/j.critrevonc.2017.10.009] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 10/22/2017] [Indexed: 12/17/2022] Open
|
36
|
Song JC, Ding XL, Sun XH, Safi M, Tian J. Brain metastasis in a patient with melanoma receiving Pembrolizumab therapy: A case report and review of the literature. Medicine (Baltimore) 2017; 96:e9278. [PMID: 29390382 PMCID: PMC5815794 DOI: 10.1097/md.0000000000009278] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Revised: 11/21/2017] [Accepted: 11/23/2017] [Indexed: 11/25/2022] Open
Abstract
RATIONALE Melanoma with brain metastasis is associated with a poor prognosis and high mortality rate. As patients with this condition have been excluded from most clinical trials, data on the use of anti-programmed death 1 therapy for these patients are limited. PATIENT CONCERNS The patient was a 62-year-old man with a 10-year history of melanotic nevus in his right forearm. He was admitted to another hospital in August 2015 due to the growth of the melanotic nevus over 1 year and complaint of a mass in the right mid-axillary area. The patient had no relevant medical, surgical, or family history. DIAGNOSES The biopsy of his right axillary lymph node showed malignant melanoma. INTERVENTIONS He was subsequently treated with adjuvant high-dose interferon after dacarbazine. Numerous metastatic lesions were found in his lung, abdomen, pelvic cavity, and brain after five months later, and then Pembrolizumab was used for six cycles (2 mg/kg every 3 weeks). He experienced immunorelated adverse events and we gave him cortisol to treat immunorelated disease until pneumonia was found. OUTCOMES We observed a delayed effect after three cycles of Pembrolizumab, the intracranial lesion presented clear margins and localization, while the other lesions became much smaller. A mixed response was observed after four cycles, with still stable extracranial metastases but growing a new lesion in brain. After two additional cycles of Pembrolizumab, the treatment was stopped due to the patient's inability to pay for it and a decline in his performance status. He then received palliative treatment at a local hospital and died for severe pulmonary infection, with an overall survival time of 7 months from metastasis. LESSONS In the case reported here, a delayed and mixed response was observed after Pembrolizumab was used. Because of causing severe pulmonary infection, the use of steroids should be considered carefully when treating immunorelated adverse events. It seemed that the Pembrolizumab has a positive effect on melanoma brain metastases especially combined with other treatments. However, there are still some challenges including patient selection, predictors of response, drug tolerance, optimizing combination strategies and control of adverse effects. More carefully designed clinical trials are urgently needed.
Collapse
|
37
|
Abstract
Background: Checkpoint inhibitors (CPI) have now been established as standard agents in the management of patients with metastatic renal cell carcinoma (mRCC). Given the unique toxicity profiles of CPIs, a detailed understanding of their incidence rate and characteristics is critical. Objective: To perform a systematic review for the analysis of the incidence rate and characteristics of toxicities in mRCC patients treated with CPIs in published clinical trials. Methods: A systematic search of EMBASE (Ovid) and MEDLINE (Ovid) was conducted as per PRISMA guidelines to identify prospective clinical trials of checkpoint inhibitors in mRCC. The search method involved querying for the terms renal cell carcinoma or kidney carcinoma with any of the following: programmed cell death 1, PD-1, programmed cell death ligand 1, PD-L1, cytotoxic T-lymphocyte antigen 4, CTLA-4, immunotherapy, checkpoint inhibitor, anti-PD-1, or anti-PD-L1. Only prospective clinical trials were included. Results: The systematic review yielded 9,722 records through the MEDLINE (Ovid) and EMBASE (Ovid) databases. Ultimately, five prospective clinical trials with 722 patients were selected for inclusion. The rates of any grade adverse event (AE) and grade (G) 3-4 AEs were 79.9% and 20.9%, respectively. Regarding immune-related AEs (irAEs), the most common system affected by any grade irAE was the skin (30.89%) and the most common grade 3-4 irAE was related to the hepatic system (8.23%). Rates of AEs were similar across the CPI monotherapy clinical trials. Conclusions: The rates of AEs in mRCC patients treated with CPI is similar to rates in other cancers. AEs in mRCC are fairly consistent among monotherapy trials with PD-1 and PD-L1 inhibitors and as one would expect higher when CTLA-4 and PD-1 inhibitors are offered in combination.
Collapse
Affiliation(s)
| | - Jorge A Garcia
- Cleveland Clinic Taussig Cancer Institute, Cleveland, OH, USA.,Cleveland Clinic Glickman Urological and Kidney Institute, Cleveland, OH, USA
| |
Collapse
|
38
|
Simmons D, Lang E. The Most Recent Oncologic Emergency: What Emergency Physicians Need to Know About the Potential Complications of Immune Checkpoint Inhibitors. Cureus 2017; 9:e1774. [PMID: 29250474 PMCID: PMC5729030 DOI: 10.7759/cureus.1774] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 10/13/2017] [Indexed: 12/27/2022] Open
Abstract
Immune checkpoint inhibitors targeting cytotoxic T-lymphocyte associated protein 4 (CTLA-4) and programmable cell death protein 1 (PD-1)/PD-L1 have shown antitumor activity in cancers such as melanoma, non-small cell lung cancer, renal cell carcinoma, and urothelial cancer. Certain checkpoint inhibitors have been approved for use in Canada, and are becoming a mainstay in the treatment of melanoma and other malignancies. These drugs have a unique side effect profile and are known to cause immune-related adverse events (irAEs). These adverse events often appear to originate from an infectious etiology, when in fact they result from the enhanced immune response caused by immune checkpoint therapy. IrAEs are primarily treated with corticosteroids, which suppress the overactive immune response that is secondary to the treatment. IrAEs can occur in any organ system, but adverse events in the skin, gastrointestinal, endocrine, and pulmonary systems are among the most common. As an emergency physician, one must be familiar with these drugs and their adverse events in order to identify patients presenting with irAE and treat them accordingly. This paper provides a brief introduction to immune checkpoint inhibitors, discusses the most common irAEs relevant to emergency physicians, and gives suggestions on how to manage patients presenting to the emergency department (ED) suffering from irAEs.
Collapse
Affiliation(s)
- Duncan Simmons
- Faculty of Medicine and Dentistry, University of Alberta
| | - Eddy Lang
- Emergency Medicine, University of Calgary
| |
Collapse
|
39
|
Sheldon CA, Kharlip J, Tamhankar MA. Inflammatory Orbitopathy Associated With Ipilimumab. Ophthalmic Plast Reconstr Surg 2017; 33:S155-S158. [PMID: 26068559 DOI: 10.1097/iop.0000000000000509] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In this case report, the clinical presentation of an inflammatory orbitopathy seen following treatment with ipilimumab is described. After 3 rounds of ipilimumab (10 mg/kg) treatment for Stage III metastatic melanoma, the subject of this case report developed acute eye pain and proptosis. At initial presentation, she had marked proptosis and diffuse severe ophthalmoparesis. After treatment with high-dose steroids, over a period of 6 months, the symptoms gradually resolved fully. Although the condition may mimic thyroid-associated orbitopathy, imaging and laboratory features suggest that the orbitopathy associated with ipilimumab is not a secondary effect of thyroid dysfunction but a distinct inflammatory condition. The authors conclude that immune-related side effects can occur with biologic agents used to treat malignancies and these side-effects can involve the eye. This case illustrates the occurrence of an inflammatory orbitopathy associated with ipilimumab treatment.
Collapse
Affiliation(s)
- Claire A Sheldon
- *Scheie Eye Institute, Department of Ophthalmology, and †Departments of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, U.S.A
| | | | | |
Collapse
|
40
|
Feld E, Horn L. Emerging role of nivolumab in the management of patients with non-small-cell lung cancer: current data and future perspectives. Onco Targets Ther 2017; 10:3697-3708. [PMID: 28769573 PMCID: PMC5533488 DOI: 10.2147/ott.s97903] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Immune-checkpoint inhibitors have become valuable therapies in the treatment of patients with non-small-cell lung cancer (NSCLC). Recent clinical trials have shown promising results with regard to efficacy and toxicity profiles of these agents compared to cytotoxic chemotherapy. Nivolumab was one of the first immune-checkpoint inhibitors to demonstrate clinical activity in patients with NSCLC, and is currently approved in the US for treatment of patients with advanced squamous and nonsquamous NSCLC who have progressed on or after platinum-based chemotherapy. This review provides an update on nivolumab's pharmacology, safety, and efficacy, as established by the CheckMate trials. We also discuss specific applications and strategies for the use of nivolumab in NSCLC patients, as well as predictive biomarkers and their role in treatment selection.
Collapse
Affiliation(s)
- Emily Feld
- Department of Internal Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Leora Horn
- Department of Internal Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
41
|
Limitations and opportunities for immune checkpoint inhibitors in pediatric malignancies. Cancer Treat Rev 2017; 58:22-33. [PMID: 28622628 PMCID: PMC5524462 DOI: 10.1016/j.ctrv.2017.05.006] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 05/23/2017] [Accepted: 05/24/2017] [Indexed: 12/14/2022]
Abstract
Immune checkpoint inhibitors (ICI) have shown great promise in a wide spectrum of adult solid and hematological malignancies, achieving objective tumor responses and prolonging survival. However, there is limited clinical success amongst pediatric patients. In this review, we summarize the current understanding of ICI and present an up-to-date overview of recent and ongoing clinical trials of ICI in pediatric malignancies. In addition, we will discuss immunologic and clinical difficulties in this young population, as well as future prospects for combination of ICI with other immune-based and conventional treatments.
Collapse
|
42
|
Demlova R, Valík D, Obermannova R, ZdraŽilová-Dubská L. The safety of therapeutic monoclonal antibodies: implications for cancer therapy including immuno-checkpoint inhibitors. Physiol Res 2017; 65:S455-S462. [PMID: 28006927 DOI: 10.33549/physiolres.933525] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Monoclonal antibody-based treatment of cancer has been established as one of the most successful therapeutic strategies for both hematologic malignancies and solid tumors. In addition to targeting cancer antigens antibodies can also modulate immunological pathways that are critical to immune surveillance. Antibody therapy directed against several negative immunologic regulators (checkpoints) is demonstrating significant success in the past few years. Immune checkpoint inhibitors, ipilimumab, pembrolizumab and nivolumab, have shown significant clinical benefit in several malignancies and are already approved for advanced melanoma and squamous NSCLC. Based on their mechanism of action, these agents can exert toxicities that are unlike conventional cytotoxic chemotherapy, whose nature is close to autoimmune diseases - immune related adverse events (irAEs). In this review we focus on the spectrum of irAEs associated with immune checkpoint antibodies, discussing the pharmacological treatment strategy and possible clinical impact.
Collapse
Affiliation(s)
- R Demlova
- Department of Pharmacology, Faculty of Medicine, Brno, Czech Republic.
| | | | | | | |
Collapse
|
43
|
Genova C, Rossi G, Rijavec E, Biello F, Barletta G, Tagliamento M, Grossi F. Releasing the brake: safety profile of immune check-point inhibitors in non-small cell lung cancer. Expert Opin Drug Saf 2017; 16:573-585. [PMID: 28351171 DOI: 10.1080/14740338.2017.1313228] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Immune check-point inhibitors are now employed as single-agents in current practice for the treatment of advanced non-small cell lung cancer (NSCLC), while combinations of different inhibitors are being evaluated in clinical trials. Although the safety profile of these compounds, with particular reference to drugs targeting programmed death protein 1 (PD-1) and its ligand (PD-L1), is generally considered manageable, peculiar, immune-related toxicities may onset. Areas covered: This review focuses on the immune-related adverse events (irAEs) observed during immune check-point blockade in NSCLC and their management. The authors report the incidence of irAEs based on the currently available data involving NSCLC and provide recommendations on the general approach to irAEs, as well as indications for the most relevant site-specific events. Expert opinion: Since irAEs may involve a wide range of organs and systems and are potentially reversible if promptly treated, early diagnosis should always be achieved; this might be particularly challenging when other potential causes of toxicity are suspected, such as infections or concurrent treatments. Finally, drugs active on the PD-1/PD-L1 axis appear to be generally manageable even when they are administered to patients with relevant comorbidities, provided that adequate clinical monitoring is performed.
Collapse
Affiliation(s)
- Carlo Genova
- a Lung Cancer Unit , IRCCS AOU San Martino - IST National Cancer Research Institute , Genoa , Italy.,b Department of Internal Medicine, School of Medicine , University of Genoa , Genoa , Italy
| | - Giovanni Rossi
- a Lung Cancer Unit , IRCCS AOU San Martino - IST National Cancer Research Institute , Genoa , Italy
| | - Erika Rijavec
- a Lung Cancer Unit , IRCCS AOU San Martino - IST National Cancer Research Institute , Genoa , Italy
| | - Federica Biello
- a Lung Cancer Unit , IRCCS AOU San Martino - IST National Cancer Research Institute , Genoa , Italy
| | - Giulia Barletta
- a Lung Cancer Unit , IRCCS AOU San Martino - IST National Cancer Research Institute , Genoa , Italy
| | - Marco Tagliamento
- a Lung Cancer Unit , IRCCS AOU San Martino - IST National Cancer Research Institute , Genoa , Italy
| | - Francesco Grossi
- a Lung Cancer Unit , IRCCS AOU San Martino - IST National Cancer Research Institute , Genoa , Italy
| |
Collapse
|
44
|
Abstract
Brain metastases are a major clinical challenge occurring in up to 60% of patients suffering from metastatic melanoma. They cause significant clinical symptoms and impair the overall survival prognosis. The introduction of targeted therapies including BRAF and MEK inhibitors as well as CTLA-4 and PD-1 axis targeting immune checkpoint inhibitors have dramatically improved the treatment and prognosis of patients with extracranial metastatic melanoma. Although, similar response rates for extra- and intracranial metastases have been reported, only few data from brain metastasis specific trails are available so far. The following review will provide an overview on the currently available data on targeted therapies, remaining questions and the most important side effects in the special clinical situation of melanoma brain metastases.
Collapse
Affiliation(s)
- Anna S Berghoff
- Department of Medicine I and Comprehensive Cancer Center CNS Unit (CCC-CNS), Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Matthias Preusser
- Department of Medicine I and Comprehensive Cancer Center CNS Unit (CCC-CNS), Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.
| |
Collapse
|
45
|
Byun DJ, Wolchok JD, Rosenberg LM, Girotra M. Cancer immunotherapy - immune checkpoint blockade and associated endocrinopathies. Nat Rev Endocrinol 2017; 13:195-207. [PMID: 28106152 PMCID: PMC5629093 DOI: 10.1038/nrendo.2016.205] [Citation(s) in RCA: 487] [Impact Index Per Article: 60.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Advances in cancer therapy in the past few years include the development of medications that modulate immune checkpoint proteins. Cytotoxic T-lymphocyte antigen 4 (CTLA4) and programmed cell death protein 1 (PD1) are two co-inhibitory receptors that are expressed on activated T cells against which therapeutic blocking antibodies have reached routine clinical use. Immune checkpoint blockade can induce inflammatory adverse effects, termed immune-related adverse events (IRAEs), which resemble autoimmune disease. In this Review, we describe the current data regarding immune-related endocrinopathies, including hypophysitis, thyroid dysfunction and diabetes mellitus. We discuss the clinical management of these endocrinopathies within the context of our current understanding of the mechanisms of IRAEs.
Collapse
Affiliation(s)
- David J Byun
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, 10065 New York, USA
- Weill Cornell Medical College, 1300 York Avenue, New York, 10065 New York, USA
| | - Jedd D Wolchok
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, 10065 New York, USA
- Weill Cornell Medical College, 1300 York Avenue, New York, 10065 New York, USA
| | - Lynne M Rosenberg
- Weill Cornell Medical College, 1300 York Avenue, New York, 10065 New York, USA
| | - Monica Girotra
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, 10065 New York, USA
- Weill Cornell Medical College, 1300 York Avenue, New York, 10065 New York, USA
| |
Collapse
|
46
|
Rubin KM. Understanding Immune Checkpoint Inhibitors for Effective Patient Care. Clin J Oncol Nurs 2017; 19:709-17. [PMID: 26583635 DOI: 10.1188/15.cjon.709-717] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Immune checkpoint inhibitors represent a paradigm change in the treatment of melanoma and other advanced cancers. These agents manipulate key immune-regulating pathways to restore immune responses against tumors. The success of this approach is demonstrated by ipilimumab (Yervoy®) for the treatment of advanced melanoma, with improvement in three-year survival rates of about 20%. Newer checkpoint inhibitors targeting the programmed death-1 (PD-1) pathway have been approved and may have higher response rates and improved tolerability. OBJECTIVES This article aims to educate nurses and increase their comfort level with these new therapies. METHODS The mechanism of action of immune checkpoint inhibitors is reviewed, and insight is provided on how nurses can use this knowledge to more effectively care for patients receiving these therapies. FINDINGS The use of immuno-oncology agents is increasing. Oncology nurses must understand the basic immune mechanism of action responsible for the novel toxicity profile characterized by immune-related adverse events (irAEs) and clinical response patterns. Managing irAEs with immune checkpoint inhibitors is not necessarily more difficult than with conventional agents, but a difference does exist. Nurses and other healthcare providers must consider the underlying cause of toxicity with immune checkpoint inhibitors when making management decisions.
Collapse
Affiliation(s)
- Krista M Rubin
- Melanoma Disease Center, Massachusetts General Hospital Cancer Center in Boston
| |
Collapse
|
47
|
Abstract
Immune checkpoint inhibitors (ICIs), including antibodies targeting cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) and programmed cell death protein-1 (PD-1), have shown durable treatment responses in multiple tumor types by enhancing antitumor immunity. However, removal of self-tolerance can induce autoimmunity and produce a unique immune-driven toxicity profile, termed immune-related adverse events (irAEs). As ICIs gain approval for a growing number of indications, it is imperative clinicians increase their knowledge of and ability to manage irAEs. This review examines the etiology, presentation, kinetics, and treatment of irAEs and aims to provide practical guidance for clinicians.
Collapse
|
48
|
Gaillard SL, Secord AA, Monk B. The role of immune checkpoint inhibition in the treatment of ovarian cancer. GYNECOLOGIC ONCOLOGY RESEARCH AND PRACTICE 2016; 3:11. [PMID: 27904752 PMCID: PMC5122024 DOI: 10.1186/s40661-016-0033-6] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 11/18/2016] [Indexed: 12/18/2022]
Abstract
The introduction of immune checkpoint inhibitors has revolutionized treatment of multiple cancers and has bolstered interest in this treatment approach. So far, emerging clinical data show limited clinical efficacy of these agents in ovarian cancer with objective response rates of 10–15% with some durable responses. In this review, we present emerging clinical data of completed trials of immune checkpoint inhibitors and review ongoing studies. In addition we examine the current knowledge of the tumor microenvironment of ovarian cancers with a focus on the significance of PD-L1 expression and tumor-infiltrating lymphocytes on predicting response to immune checkpoint blockade. We evaluate approaches to improve treatment outcomes through the use of predictive biomarkers and patient selection. Finally, we review management considerations including immune related adverse events and response criteria.
Collapse
Affiliation(s)
- Stéphanie L Gaillard
- Department of Medicine, Division of Medical Oncology, Duke Cancer Institute, 200 Trent Drive, Durham, NC 27710 USA
| | - Angeles A Secord
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Duke Cancer Institute, 200 Trent Drive, Durham, NC 27710 USA
| | - Bradley Monk
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, University of Arizona College of Medicine, 2222 E. Highland Ave., Suite 400, Phoenix, AZ 85016 USA
| |
Collapse
|
49
|
Jung K, Zeng X, Bilusic M. Nivolumab-associated acute glomerulonephritis: a case report and literature review. BMC Nephrol 2016; 17:188. [PMID: 27876011 PMCID: PMC5120473 DOI: 10.1186/s12882-016-0408-2] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 11/15/2016] [Indexed: 02/05/2023] Open
Abstract
Background Immune checkpoint inhibitors are changing the landscape of oncology treatment as they are significantly improving treatment for multiple malignancies. Nivolumab, an anti-programmed death 1 antibody, is a US Food and Drug Administration-approved treatment for melanoma, non-small cell lung cancer, and kidney cancer but can result in a spectrum of autoimmune side effects. Adverse effects can occur within any organ system in the body including the colon, lung, liver, endocrine systems, or kidneys. Case presentation A 70-year-old male with clear cell kidney cancer was admitted with acute kidney injury while on nivolumab. A kidney biopsy revealed diffuse tubular injury and immune complex-mediated glomerulonephritis. Electron microscopy of the specimen showed hump-like subepithelial deposits. Nivolumab was discontinued and the patient was started on a high dose of steroids. After 5 months of systemic corticosteroids and hemodialysis, the patient’s kidney function improved to his baseline level. Despite a prolonged interruption to treatment, immunosuppressive therapy did not compromise the anticancer effects of nivolumab. Conclusion Immune-related adverse effects in the kidney can cause autoimmune glomerulonephritis as well as tubulointerstitial injury. In the literature, immune-related nephritis generally responded well to systemic corticosteroid treatment. Based on our experience, a prolonged course of a high dose of steroids and hemodialysis may be required to achieve an adequate treatment effect.
Collapse
Affiliation(s)
- Kyungsuk Jung
- Department of Medicine, Fox Chase Cancer Center, 333 Cottman Ave, Philadelphia, PA, 19111, USA.
| | - Xu Zeng
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, 3500N Broad St, Philadelphia, PA, 19140, USA
| | - Marijo Bilusic
- Department of Medical Oncology, Fox Chase Cancer Center, 333 Cottman Ave, Philadelphia, PA, 19111, USA
| |
Collapse
|
50
|
Weber JS, Hodi FS, Wolchok JD, Topalian SL, Schadendorf D, Larkin J, Sznol M, Long GV, Li H, Waxman IM, Jiang J, Robert C. Safety Profile of Nivolumab Monotherapy: A Pooled Analysis of Patients With Advanced Melanoma. J Clin Oncol 2016; 35:785-792. [PMID: 28068177 DOI: 10.1200/jco.2015.66.1389] [Citation(s) in RCA: 874] [Impact Index Per Article: 97.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Purpose We conducted a retrospective analysis to assess the safety profile of nivolumab monotherapy in patients with advanced melanoma and describe the management of adverse events (AEs) using established safety guidelines. Patients and Methods Safety data were pooled from four studies, including two phase III trials, with patients who received nivolumab 3 mg/kg once every 2 weeks. We evaluated rate of treatment-related AEs, time to onset and resolution of select AEs (those with potential immunologic etiology), and impact of select AEs and suppressive immune-modulating agents (IMs) on antitumor efficacy. Results Among 576 patients, 71% (95% CI, 67% to 75%) experienced any-grade treatment-related AEs (most commonly fatigue [25%], pruritus [17%], diarrhea [13%], and rash [13%]), and 10% (95% CI, 8% to 13%) experienced grade 3 to 4 treatment-related AEs. No drug-related deaths were reported. Select AEs (occurring in 49% of patients) were most frequently skin related, GI, endocrine, and hepatic; grade 3 to 4 select AEs occurred in 4% of patients. Median time to onset of select AEs ranged from 5 weeks for skin to 15 weeks for renal AEs. Approximately 24% of patients received systemic IMs to manage select AEs, which in most cases resolved. Adjusting for number of doses, objective response rate (ORR) was significantly higher in patients who experienced treatment-related select AEs of any grade compared with those who did not. ORRs were similar in patients who did and patients who did not receive systemic IMs. Conclusion Treatment-related AEs with nivolumab monotherapy were primarily low grade, and most resolved with established safety guidelines. Use of IMs did not affect ORR, although treatment-related select AEs of any grade were associated with higher ORR, but no progression-free survival benefit.
Collapse
Affiliation(s)
- Jeffrey S Weber
- Jeffrey S. Weber, H. Lee Moffitt Cancer Center, Tampa, FL; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY; Suzanne L. Topalian, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD; Mario Sznol, Yale University School of Medicine and Smilow Cancer Center, Yale-New Haven Hospital, New Haven, CT; Hewei Li, Ian M. Waxman, and Joel Jiang, Bristol-Myers Squibb, Princeton, NJ; Dirk Schadendorf, University of Essen, Essen, Germany; James Larkin, Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Georgina V. Long, Melanoma Institute Australia and University of Sydney, Sydney, New South Wales, Australia; and Caroline Robert, Gustave Roussy and Paris-Sud University, Villejuif-Paris Sud, France
| | - F Stephen Hodi
- Jeffrey S. Weber, H. Lee Moffitt Cancer Center, Tampa, FL; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY; Suzanne L. Topalian, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD; Mario Sznol, Yale University School of Medicine and Smilow Cancer Center, Yale-New Haven Hospital, New Haven, CT; Hewei Li, Ian M. Waxman, and Joel Jiang, Bristol-Myers Squibb, Princeton, NJ; Dirk Schadendorf, University of Essen, Essen, Germany; James Larkin, Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Georgina V. Long, Melanoma Institute Australia and University of Sydney, Sydney, New South Wales, Australia; and Caroline Robert, Gustave Roussy and Paris-Sud University, Villejuif-Paris Sud, France
| | - Jedd D Wolchok
- Jeffrey S. Weber, H. Lee Moffitt Cancer Center, Tampa, FL; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY; Suzanne L. Topalian, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD; Mario Sznol, Yale University School of Medicine and Smilow Cancer Center, Yale-New Haven Hospital, New Haven, CT; Hewei Li, Ian M. Waxman, and Joel Jiang, Bristol-Myers Squibb, Princeton, NJ; Dirk Schadendorf, University of Essen, Essen, Germany; James Larkin, Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Georgina V. Long, Melanoma Institute Australia and University of Sydney, Sydney, New South Wales, Australia; and Caroline Robert, Gustave Roussy and Paris-Sud University, Villejuif-Paris Sud, France
| | - Suzanne L Topalian
- Jeffrey S. Weber, H. Lee Moffitt Cancer Center, Tampa, FL; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY; Suzanne L. Topalian, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD; Mario Sznol, Yale University School of Medicine and Smilow Cancer Center, Yale-New Haven Hospital, New Haven, CT; Hewei Li, Ian M. Waxman, and Joel Jiang, Bristol-Myers Squibb, Princeton, NJ; Dirk Schadendorf, University of Essen, Essen, Germany; James Larkin, Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Georgina V. Long, Melanoma Institute Australia and University of Sydney, Sydney, New South Wales, Australia; and Caroline Robert, Gustave Roussy and Paris-Sud University, Villejuif-Paris Sud, France
| | - Dirk Schadendorf
- Jeffrey S. Weber, H. Lee Moffitt Cancer Center, Tampa, FL; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY; Suzanne L. Topalian, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD; Mario Sznol, Yale University School of Medicine and Smilow Cancer Center, Yale-New Haven Hospital, New Haven, CT; Hewei Li, Ian M. Waxman, and Joel Jiang, Bristol-Myers Squibb, Princeton, NJ; Dirk Schadendorf, University of Essen, Essen, Germany; James Larkin, Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Georgina V. Long, Melanoma Institute Australia and University of Sydney, Sydney, New South Wales, Australia; and Caroline Robert, Gustave Roussy and Paris-Sud University, Villejuif-Paris Sud, France
| | - James Larkin
- Jeffrey S. Weber, H. Lee Moffitt Cancer Center, Tampa, FL; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY; Suzanne L. Topalian, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD; Mario Sznol, Yale University School of Medicine and Smilow Cancer Center, Yale-New Haven Hospital, New Haven, CT; Hewei Li, Ian M. Waxman, and Joel Jiang, Bristol-Myers Squibb, Princeton, NJ; Dirk Schadendorf, University of Essen, Essen, Germany; James Larkin, Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Georgina V. Long, Melanoma Institute Australia and University of Sydney, Sydney, New South Wales, Australia; and Caroline Robert, Gustave Roussy and Paris-Sud University, Villejuif-Paris Sud, France
| | - Mario Sznol
- Jeffrey S. Weber, H. Lee Moffitt Cancer Center, Tampa, FL; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY; Suzanne L. Topalian, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD; Mario Sznol, Yale University School of Medicine and Smilow Cancer Center, Yale-New Haven Hospital, New Haven, CT; Hewei Li, Ian M. Waxman, and Joel Jiang, Bristol-Myers Squibb, Princeton, NJ; Dirk Schadendorf, University of Essen, Essen, Germany; James Larkin, Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Georgina V. Long, Melanoma Institute Australia and University of Sydney, Sydney, New South Wales, Australia; and Caroline Robert, Gustave Roussy and Paris-Sud University, Villejuif-Paris Sud, France
| | - Georgina V Long
- Jeffrey S. Weber, H. Lee Moffitt Cancer Center, Tampa, FL; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY; Suzanne L. Topalian, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD; Mario Sznol, Yale University School of Medicine and Smilow Cancer Center, Yale-New Haven Hospital, New Haven, CT; Hewei Li, Ian M. Waxman, and Joel Jiang, Bristol-Myers Squibb, Princeton, NJ; Dirk Schadendorf, University of Essen, Essen, Germany; James Larkin, Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Georgina V. Long, Melanoma Institute Australia and University of Sydney, Sydney, New South Wales, Australia; and Caroline Robert, Gustave Roussy and Paris-Sud University, Villejuif-Paris Sud, France
| | - Hewei Li
- Jeffrey S. Weber, H. Lee Moffitt Cancer Center, Tampa, FL; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY; Suzanne L. Topalian, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD; Mario Sznol, Yale University School of Medicine and Smilow Cancer Center, Yale-New Haven Hospital, New Haven, CT; Hewei Li, Ian M. Waxman, and Joel Jiang, Bristol-Myers Squibb, Princeton, NJ; Dirk Schadendorf, University of Essen, Essen, Germany; James Larkin, Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Georgina V. Long, Melanoma Institute Australia and University of Sydney, Sydney, New South Wales, Australia; and Caroline Robert, Gustave Roussy and Paris-Sud University, Villejuif-Paris Sud, France
| | - Ian M Waxman
- Jeffrey S. Weber, H. Lee Moffitt Cancer Center, Tampa, FL; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY; Suzanne L. Topalian, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD; Mario Sznol, Yale University School of Medicine and Smilow Cancer Center, Yale-New Haven Hospital, New Haven, CT; Hewei Li, Ian M. Waxman, and Joel Jiang, Bristol-Myers Squibb, Princeton, NJ; Dirk Schadendorf, University of Essen, Essen, Germany; James Larkin, Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Georgina V. Long, Melanoma Institute Australia and University of Sydney, Sydney, New South Wales, Australia; and Caroline Robert, Gustave Roussy and Paris-Sud University, Villejuif-Paris Sud, France
| | - Joel Jiang
- Jeffrey S. Weber, H. Lee Moffitt Cancer Center, Tampa, FL; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY; Suzanne L. Topalian, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD; Mario Sznol, Yale University School of Medicine and Smilow Cancer Center, Yale-New Haven Hospital, New Haven, CT; Hewei Li, Ian M. Waxman, and Joel Jiang, Bristol-Myers Squibb, Princeton, NJ; Dirk Schadendorf, University of Essen, Essen, Germany; James Larkin, Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Georgina V. Long, Melanoma Institute Australia and University of Sydney, Sydney, New South Wales, Australia; and Caroline Robert, Gustave Roussy and Paris-Sud University, Villejuif-Paris Sud, France
| | - Caroline Robert
- Jeffrey S. Weber, H. Lee Moffitt Cancer Center, Tampa, FL; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, New York, NY; Suzanne L. Topalian, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD; Mario Sznol, Yale University School of Medicine and Smilow Cancer Center, Yale-New Haven Hospital, New Haven, CT; Hewei Li, Ian M. Waxman, and Joel Jiang, Bristol-Myers Squibb, Princeton, NJ; Dirk Schadendorf, University of Essen, Essen, Germany; James Larkin, Royal Marsden National Health Service Foundation Trust, London, United Kingdom; Georgina V. Long, Melanoma Institute Australia and University of Sydney, Sydney, New South Wales, Australia; and Caroline Robert, Gustave Roussy and Paris-Sud University, Villejuif-Paris Sud, France
| |
Collapse
|