201
|
König D, Läubli H. Mechanisms of Immune-Related Complications in Cancer Patients Treated with Immune Checkpoint Inhibitors. Pharmacology 2020; 106:123-136. [PMID: 32721966 DOI: 10.1159/000509081] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 05/29/2020] [Indexed: 12/13/2022]
Abstract
Immune checkpoint inhibitors (ICIs) have changed the prognosis of many cancer patients. Blocking antibodies targeting inhibitory cytotoxic T-lymphocyte-associated antigen 4 or programmed cell death protein-1 receptors or the programmed cell death ligand-1 have led to long-lasting remissions in patients with even advanced cancers. Main side effects induced by ICIs are inflammatory complications with sometimes severe sequelae for patients. Recent studies have improved our understanding how such immune-related adverse events (irAEs) develop. Here, we summarize the current knowledge of pathomechanisms involved in the de-velopment of irAEs with a particular focus on potential pathways that could be targeted to prevent severe immune-related complications in patients treated with cancer immunotherapy.
Collapse
Affiliation(s)
- David König
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland
| | - Heinz Läubli
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland, .,Laboratory for Cancer Immunotherapy, Department of Biomedicine, University of Basel, Basel, Switzerland,
| |
Collapse
|
202
|
Differential risks of immune-related colitis among various immune checkpoint inhibitor regimens. Int Immunopharmacol 2020; 87:106770. [PMID: 32702598 DOI: 10.1016/j.intimp.2020.106770] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 06/17/2020] [Accepted: 06/30/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Colitis is a life-threatening and common immune-related adverse event in patients receiving immune checkpoint inhibitors (ICIs). Therefore, we performed a meta-analysis to assess the risk of immune-related colitis among various ICI treatment regimens. METHODS We used PubMed, EMBASE, and the Cochrane Library to retrieve phase II or III randomized controlled trials (RCTs) of ICIs that specified the number of all-grade (grade 1-5) colitis and high-grade (grade 3-5) colitis events through January 2020. The pooled relative risk (RR) and 95% confidence intervals (CIs) were calculated to compare the risk of colitis among various therapeutic regimens. RESULTS The search strategy identified 40 RCTs involving 26,893 patients. The risk of all-grade and high-grade colitis after PD-1/PD-L1 inhibitor was significantly lower than that of CTLA-4 inhibitor (0.18 and 0.14 relative risk respectively). The risk of all-grade and high-grade colitis was dose-dependent for CTLA-4 inhibitor therapy, but not for PD-1/PD-L1 inhibitor therapy. The relative risk of all-grade and high-grade colitis after combination therapy with PD-1/PD-L1 inhibitor and CTLA-4 inhibitor compared to PD-1/PD-L1 inhibitor alone was 9.25 and 12.00 respectively. No significant difference was found between PD-1/PD-L1 inhibitor combined with chemotherapy or targeted therapy and PD-1/PD-L1 inhibitor alone for either all-grade or high-grade colitis. CONCLUSIONS Our meta-analysis indicates that CTLA-4 inhibitor is associated with a higher risk of colitis compared with PD-1/PD-L1 inhibitor, whether used as a monotherapy or combination immunotherapy. Importantly, the combination of PD-1/PD-L1 inhibitor with chemotherapy or targeted therapy may not increase the risk of colitis significantly compared to PD-1/PD-L1 inhibitor alone.
Collapse
|
203
|
Polymyalgia rheumatica with normal inflammatory indices at the time of diagnosis: can we just move a step forward? Reumatologia 2020; 58:184-186. [PMID: 32684653 PMCID: PMC7362269 DOI: 10.5114/reum.2020.96549] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 04/20/2020] [Indexed: 12/31/2022] Open
Abstract
The existence of polymyalgia rheumatica (PMR) with normal inflammatory indices at the time of diagnosis still represents a diagnostic conundrum. According to the literature, some patients with PMR following immune checkpoint inhibitory therapy had normal values of both erythrocyte sedimentation rate and C-reactive protein concentrations at the time of diagnosis. In this short communication we investigated the possibility that in some patients with PMR the main pathogenic mechanism is constituted by inhibition of some checkpoints, such as programmed death receptor-1, programmed death ligand 1, and “cytotoxic” lymphocyte antigen 4. In these patients, the pathogenetic mechanisms underlying PMR can act much more upstream than commonly suggested. Also, we addressed the question of whether these patients should be considered as affected by PMR-like syndromes or by PMR subset.
Collapse
|
204
|
Song P, Zhang D, Cui X, Zhang L. Meta-analysis of immune-related adverse events of immune checkpoint inhibitor therapy in cancer patients. Thorac Cancer 2020; 11:2406-2430. [PMID: 32643323 PMCID: PMC7471041 DOI: 10.1111/1759-7714.13541] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 05/30/2020] [Accepted: 05/30/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have significant clinical efficacy in the treatment of non-small cell lung cancer (NSCLC); however, the incidence of immune-related adverse events (irAEs) of up to 50% has prevented their widespread use. With the increase in the use of ICIs alone or as combination therapy, clinicians are required to have a better understanding of irAEs and be able to manage them systematically. In this study, we aimed to assess the incidence of irAEs associated with ICIs. METHODS We searched PubMed, Embase, and the Web of Science databases, and also included relevant literature references to widen our search. The relevant data with inclusion criteria were performed using RevMan 3.6.0 for meta-analysis. We undertook a systematic literature search which included published data up to December 2019. RESULTS Overall, 147 articles and 23 761 cancer patients with 11 different ICI treatment-related (grade 1-5 and 3-5) irAEs were included in the study. There were 46 articles on pembrolizumab (6598 patients), 27 on nivolumab (3576 patients), 13 on atezolizumab (2787 patients), 12 on avelumab (3213 patients), 10 on durvalumab (1780 patients), 22 on ipilimumab (4067 patients), eight on tremelimumab (1158 patients), three on JS001 (223 patients), four on camrelizumab (SHR-1210) (178 patients), one on sintilimab (96 patients), and one on cemiplimab (85 patients). Grade 1-5 irAEs were: cytotoxic T lymphocyte antigen 4 (CTLA-4) (82.87%), programmed cell death 1 (PD-1) (71.89%), and programmed cell death ligand-1 (PD-L1) (58.95%). Subgroup analysis was: Avelumab (44.53%), durvalumab (66.63%), pembrolizumab (67.25%), atezolizumab (68.77%), nivolumab (76.25%), Ipilimumab (82.18%), and tremelimumab (86.78%). Grade 3-5 irAEs were: CTLA-4 (27.22%), PD-1(17.29%), and PD-L1(17.29%). Subgroup analysis was: Avelumab (5.86%), durvalumab (13.43%), atezolizumab (14.45%), nivolumab (15.72%), pembrolizumab (16.58%), tremelimumab (22.04%), and ipilimumab (28.27%). CONCLUSIONS This meta-analysis confirmed that anti-PD-1 and anti-PD-L1 inhibitors had a lower incidence of irAEs compared with anti-CTLA-4 inhibitors.
Collapse
Affiliation(s)
- Peng Song
- Department of Respiratory Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Dingding Zhang
- Central Research Laboratory,Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Xiaoxia Cui
- Department of Respiratory Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Li Zhang
- Department of Respiratory Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| |
Collapse
|
205
|
Gaynor N, Crown J, Collins DM. Immune checkpoint inhibitors: Key trials and an emerging role in breast cancer. Semin Cancer Biol 2020; 79:44-57. [PMID: 32623044 DOI: 10.1016/j.semcancer.2020.06.016] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 06/19/2020] [Accepted: 06/21/2020] [Indexed: 12/15/2022]
Abstract
This review focuses on immune checkpoint inhibitors - immunomodulatory agents that aim to relieve tumour-mediated immune-cell suppression. Immune checkpoint proteins can be expressed on the tumour-cell or immune-cell populations. Immune checkpoint proteins dampen the immune response by inactivating immune cells capable of tumour destruction. Blockade of immune checkpoints has shown impressive results in a range of solid cancers, particularly melanoma and non-small cell lung cancer. The potential benefit of this class of drugs is widespread across most cancer types and an unprecedented number of clinical studies are underway to examine the benefit of these agents. The aims of this review are to: provide an overview of the key early immune checkpoint inhibitor trials involving drugs targeting programmed cell death-1 (PD-1), programmed cell death ligand-1 (PD-L1) and cytotoxic T-lymphocyte antigen-4 (CTLA-4) in multiple disease types; provide an overview of emerging therapies aimed at these targets; and provide a detailed exploration of the status of immune checkpoint inhibitors in breast cancer.
Collapse
Affiliation(s)
- Nicola Gaynor
- National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland.
| | - John Crown
- National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland; Department of Medical Oncology, St Vincent's University Hospital, Elm Park, Dublin 4, Ireland.
| | - Denis M Collins
- National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland.
| |
Collapse
|
206
|
Wu AA, Bever KM, Ho WJ, Fertig EJ, Niu N, Zheng L, Parkinson RM, Durham JN, Onners B, Ferguson AK, Wilt C, Ko AH, Wang-Gillam A, Laheru DA, Anders RA, Thompson ED, Sugar EA, Jaffee EM, Le DT. A Phase II Study of Allogeneic GM-CSF-Transfected Pancreatic Tumor Vaccine (GVAX) with Ipilimumab as Maintenance Treatment for Metastatic Pancreatic Cancer. Clin Cancer Res 2020; 26:5129-5139. [PMID: 32591464 DOI: 10.1158/1078-0432.ccr-20-1025] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/20/2020] [Accepted: 06/23/2020] [Indexed: 12/17/2022]
Abstract
PURPOSE This phase II study tested granulocyte-macrophage colony-stimulating factor (GM-CSF)-allogeneic pancreatic tumor cells (GVAX) and ipilimumab in metastatic pancreatic ductal adenocarcinoma (PDA) in the maintenance setting. PATIENTS AND METHODS Patients with PDA who were treated with front-line chemotherapy consisting of 5-fluorouracil, leucovorin, irinotecan, and oxaliplatin (FOLFIRINOX) in the metastatic setting and had ongoing response or stable disease after 8-12 doses were eligible. Patients were randomized 1:1 to treatment with GVAX and ipilimumab given every 3 weeks for four doses then every 8 weeks (Arm A) or to FOLFIRINOX continuation (Arm B). The primary objective was to compare overall survival (OS) between the two arms. RESULTS Eighty-two patients were included in the final analysis (Arm A: 40; Arm B: 42). The study was stopped for futility after interim analysis. Median OS was 9.38 months [95% confidence interval (CI), 5.0-12.2] for Arm A and 14.7 months (95% CI, 11.6-20.0) for Arm B (HR, 1.75; P = 0.019). Using immune-related response criteria, two partial responses (5.7%) were observed in Arm A and four (13.8%) in Arm B. GVAX + ipilimumab promoted T-cell differentiation into effector memory phenotypes both in the periphery and in the tumor microenvironment and increased M1 macrophages in the tumor. CONCLUSIONS GVAX and ipilimumab maintenance therapy did not improve OS over continuation of chemotherapy and resulted in a numerically inferior survival in metastatic PDA. However, clinical responses and biological effects on immune cells were observed. Further study of novel combinations in the maintenance treatment of metastatic PDA is feasible.
Collapse
Affiliation(s)
- Annie A Wu
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, The Skip Viragh Center for Pancreas Cancer Clinical Research and Patient Care, and The Bloomberg-Kimmel Institute for Cancer Immunotherapy at Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Katherine M Bever
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, The Skip Viragh Center for Pancreas Cancer Clinical Research and Patient Care, and The Bloomberg-Kimmel Institute for Cancer Immunotherapy at Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Won Jin Ho
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, The Skip Viragh Center for Pancreas Cancer Clinical Research and Patient Care, and The Bloomberg-Kimmel Institute for Cancer Immunotherapy at Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Elana J Fertig
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, The Skip Viragh Center for Pancreas Cancer Clinical Research and Patient Care, and The Bloomberg-Kimmel Institute for Cancer Immunotherapy at Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Nan Niu
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, The Skip Viragh Center for Pancreas Cancer Clinical Research and Patient Care, and The Bloomberg-Kimmel Institute for Cancer Immunotherapy at Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, China
| | - Lei Zheng
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, The Skip Viragh Center for Pancreas Cancer Clinical Research and Patient Care, and The Bloomberg-Kimmel Institute for Cancer Immunotherapy at Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Rose M Parkinson
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, The Skip Viragh Center for Pancreas Cancer Clinical Research and Patient Care, and The Bloomberg-Kimmel Institute for Cancer Immunotherapy at Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jennifer N Durham
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, The Skip Viragh Center for Pancreas Cancer Clinical Research and Patient Care, and The Bloomberg-Kimmel Institute for Cancer Immunotherapy at Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Beth Onners
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, The Skip Viragh Center for Pancreas Cancer Clinical Research and Patient Care, and The Bloomberg-Kimmel Institute for Cancer Immunotherapy at Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Anna K Ferguson
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, The Skip Viragh Center for Pancreas Cancer Clinical Research and Patient Care, and The Bloomberg-Kimmel Institute for Cancer Immunotherapy at Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Cara Wilt
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, The Skip Viragh Center for Pancreas Cancer Clinical Research and Patient Care, and The Bloomberg-Kimmel Institute for Cancer Immunotherapy at Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Andrew H Ko
- Department of Medicine, Division of Hematology/Oncology, UCSF Helen Diller Family Comprehensive Cancer Center at University of California, San Francisco, California
| | - Andrea Wang-Gillam
- Department of Internal Medicine, Division of Oncology at Washington University School of Medicine, St. Louis, Missouri
| | - Daniel A Laheru
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, The Skip Viragh Center for Pancreas Cancer Clinical Research and Patient Care, and The Bloomberg-Kimmel Institute for Cancer Immunotherapy at Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Robert A Anders
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, The Skip Viragh Center for Pancreas Cancer Clinical Research and Patient Care, and The Bloomberg-Kimmel Institute for Cancer Immunotherapy at Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Elizabeth D Thompson
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, The Skip Viragh Center for Pancreas Cancer Clinical Research and Patient Care, and The Bloomberg-Kimmel Institute for Cancer Immunotherapy at Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Elizabeth A Sugar
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, The Skip Viragh Center for Pancreas Cancer Clinical Research and Patient Care, and The Bloomberg-Kimmel Institute for Cancer Immunotherapy at Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Biostatistics, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland
| | - Elizabeth M Jaffee
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, The Skip Viragh Center for Pancreas Cancer Clinical Research and Patient Care, and The Bloomberg-Kimmel Institute for Cancer Immunotherapy at Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Dung T Le
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, The Skip Viragh Center for Pancreas Cancer Clinical Research and Patient Care, and The Bloomberg-Kimmel Institute for Cancer Immunotherapy at Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
207
|
Zhao Q, Liu C, Liu H, Chen J. Advances in immune neoadjuvant/adjuvant therapy-related adverse events of non-small cell lung cancer. Asia Pac J Clin Oncol 2020; 18:171-176. [PMID: 32573077 DOI: 10.1111/ajco.13378] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 05/05/2020] [Indexed: 12/17/2022]
Abstract
Lung cancer has been the leading cause of cancer-related death for decades and years. For the patients with early stage non-small cell lung cancer, surgical resection is the mainstay of therapy. Treatment before and after surgery, such as chemotherapy, can bring benefit to these patients, improving the 5 years survival rate by 5% approximately. Recently, the advent of immunotherapy significantly improved prognosis for the patient with lung cancer. Programmed death 1 and its ligand have become a powerful treatment option for lung cancer. In this review, we will discuss the role immunotherapy played in preoperative neoadjuvant and postoperative adjuvant treatment in lung cancer.
Collapse
Affiliation(s)
- Qingchun Zhao
- Department of Lung Cancer Surgery, Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Chao Liu
- Department of Lung Cancer Surgery, Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Hongyu Liu
- Department of Lung Cancer Surgery, Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Jun Chen
- Department of Lung Cancer Surgery, Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
208
|
Sehgal K, Costa DB, Rangachari D. Extended-Interval Dosing Strategy of Immune Checkpoint Inhibitors in Lung Cancer: Will it Outlast the COVID-19 Pandemic? Front Oncol 2020; 10:1193. [PMID: 32714874 PMCID: PMC7344199 DOI: 10.3389/fonc.2020.01193] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 06/12/2020] [Indexed: 12/19/2022] Open
Abstract
Patients with lung cancer are particularly vulnerable to complications from coronavirus disease-2019 (COVID-19). Recurrent hospital visits and hospital admission are potential risk factors for acquiring infection with its causative pathogen, severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). As immune checkpoint inhibitors (ICIs) constitute the therapeutic backbone for the vast majority of patients with advanced lung cancer in the absence of actionable driver oncogenes, there have been intense discussions within the oncology community regarding risk-benefit of delaying these treatments or use of alternative extended-interval treatment strategies to minimize the risk of viral transmission secondary to unintended nosocomial exposures. In the midst of the COVID-19 pandemic, the U.S. Food and Drug Administration (FDA) granted accelerated approval for extended-interval strategy of pembrolizumab at a dose of 400 mg every 6 weeks for all already approved oncologic indications. Herein, we summarize the evidence from the in silico pharmacokinetic modeling/simulation studies supporting extended-interval dosing strategies for the ICIs used in lung cancer. We further review the evolving clinical evidence behind these approaches and predict that they will continue to be used in routine practice even long after the pandemic, particularly for patients with durable disease control.
Collapse
Affiliation(s)
- Kartik Sehgal
- Division of Medical Oncology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Daniel B. Costa
- Division of Medical Oncology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Deepa Rangachari
- Division of Medical Oncology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
209
|
Chew HY, Dolcetti R, Simpson F. Scientifically based combination therapies with immuno-oncology checkpoint inhibitors. Br J Clin Pharmacol 2020; 86:1711-1725. [PMID: 32372470 DOI: 10.1111/bcp.14338] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 04/14/2020] [Accepted: 04/27/2020] [Indexed: 12/20/2022] Open
Abstract
The discovery of immune checkpoints and their role in modulating immune response have revolutionised cancer treatment in recent years. The immune checkpoints, cytotoxic T-lymphocyte-associated protein 4, programmed cell death protein 1 and its ligand, programmed cell death-ligand 1, have been extensively studied. Currently 7 monoclonal antibodies targeting these immune checkpoints are approved for treatment of various cancers. Inhibiting immune checkpoints has shown some success in clinic, however, a proportion of patients do not benefit from this treatment. Several other inhibitory molecules, in addition to lymphocyte-associated protein 4 and programmed cell death protein 1, are known to be involved in regulating immune response. To further improve patient outcomes, studies have examined targeting these inhibitory molecules through combination therapies. This review discusses the current landscape of combination therapies of checkpoint inhibitors.
Collapse
Affiliation(s)
- Hui Yi Chew
- The University of Queensland Diamantina Institute, Brisbane, Queensland, Australia
| | - Riccardo Dolcetti
- The University of Queensland Diamantina Institute, Brisbane, Queensland, Australia
| | - Fiona Simpson
- The University of Queensland Diamantina Institute, Brisbane, Queensland, Australia
| |
Collapse
|
210
|
Vitamin D, autoimmunity and immune-related adverse events of immune checkpoint inhibitors. Arch Dermatol Res 2020; 313:1-10. [PMID: 32519001 DOI: 10.1007/s00403-020-02094-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/26/2020] [Accepted: 05/29/2020] [Indexed: 12/16/2022]
Abstract
In addition to its quintessential role in bone homeostasis, vitamin D also plays an important role in regulating the immune system. As such, many studies have demonstrated the therapeutic benefit of vitamin D in treating autoimmune diseases. This immunomodulatory activity of vitamin D has recently attracted more attention due to the rapid development of immunotherapies for cancers, including melanoma. Patients on cancer immunotherapies can suffer from immune-related adverse events (irAEs), which can involve any organ system and range from common dermatological reactions to extremely severe cases of fatal myocarditis in metastatic melanoma patients. Since there are currently no effective approaches to predict or prevent irAEs, it is attractive to potentially leverage the intriguing immunomodulatory effects of vitamin D within this context. This review will discuss recent research investigating the possibility of using vitamin D to alleviate autoimmunity and irAEs with the hope of improving outcomes for patients on cancer immunotherapies, especially within the context of dermatology.
Collapse
|
211
|
Immune checkpoint inhibitor-related dermatologic adverse events. J Am Acad Dermatol 2020; 83:1255-1268. [PMID: 32454097 DOI: 10.1016/j.jaad.2020.03.132] [Citation(s) in RCA: 261] [Impact Index Per Article: 52.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 03/25/2020] [Accepted: 03/26/2020] [Indexed: 12/17/2022]
Abstract
Immune checkpoint inhibitors have emerged as a pillar in the management of advanced malignancies. However, nonspecific immune activation may lead to immune-related adverse events, wherein the skin and its appendages are the most frequent targets. Cutaneous immune-related adverse events include a diverse group of inflammatory reactions, with maculopapular rash, pruritus, psoriasiform and lichenoid eruptions being the most prevalent subtypes. Cutaneous immune-related adverse events occur early, with maculopapular rash presenting within the first 6 weeks after the initial immune checkpoint inhibitor dose. Management involves the use of topical corticosteroids for mild to moderate (grades 1-2) rash, addition of systemic corticosteroids for severe (grade 3) rash, and discontinuation of immunotherapy with grade 4 rash. Bullous pemphigoid eruptions, vitiligo-like skin hypopigmentation/depigmentation, and psoriasiform rash are more often attributed to programmed cell death-1/programmed cell death ligand-1 inhibitors. The treatment of bullous pemphigoid eruptions is similar to the treatment of maculopapular rash and lichenoid eruptions, with the addition of rituximab in grade 3-4 rash. Skin hypopigmentation/depigmentation does not require specific dermatologic treatment aside from photoprotective measures. In addition to topical corticosteroids, psoriasiform rash may be managed with vitamin D3 analogues, narrowband ultraviolet B light phototherapy, retinoids, or immunomodulatory biologic agents. Stevens-Johnson syndrome and other severe cutaneous immune-related adverse events, although rare, have also been associated with checkpoint blockade and require inpatient care as well as urgent dermatology consultation.
Collapse
|
212
|
Shulgin B, Kosinsky Y, Omelchenko A, Chu L, Mugundu G, Aksenov S, Pimentel R, DeYulia G, Kim G, Peskov K, Helmlinger G. Dose dependence of treatment-related adverse events for immune checkpoint inhibitor therapies: a model-based meta-analysis. Oncoimmunology 2020; 9:1748982. [PMID: 32934874 PMCID: PMC7466858 DOI: 10.1080/2162402x.2020.1748982] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 01/21/2020] [Indexed: 12/12/2022] Open
Abstract
Programmed cell death-1 (PD-1) and/or cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) immune checkpoint inhibitor (ICI) treatments are associated with adverse events (AEs), which may be dependent on ICI dose. Applying a model-based meta-analysis to evaluate safety data from published clinical trials from 2005 to 2018, we analyzed the dose/exposure dependence of ICI treatment-related AE (trAE) and immune-mediated AE (imAE) rates. Unlike with PD-1 inhibitor monotherapy, CTLA-4 inhibitor monotherapy exhibited a dose/exposure dependence on most AE types evaluated. Furthermore, combination therapy with PD-1 inhibitor significantly strengthened the dependence of trAE and imAE rates on CTLA-4 inhibitor dose/exposure.
Collapse
Affiliation(s)
| | | | | | - Lulu Chu
- PK Sciences Modeling & Simulation, Novartis Institutes of BioMedical Research, Cambridge, MA, USA
| | - Ganesh Mugundu
- Clinical Pharmacology & Quantitative Pharmacology, R&D BioPharmaceuticals, AstraZeneca, Waltham, MA, USA
| | - Sergey Aksenov
- Clinical Pharmacology & Quantitative Pharmacology, R&D BioPharmaceuticals, AstraZeneca, Waltham, MA, USA
| | | | | | | | - Kirill Peskov
- M&S Decisions LLC, Moscow, Russia
- I.M.Sechenov first Moscow State Medical University of the Russian Ministry of Health, Moscow, Russia
| | - Gabriel Helmlinger
- Clinical Pharmacology & Toxicology, Obsidian Therapeutics, Cambridge, MA, USA
| |
Collapse
|
213
|
O'Connor P, Bhadbhade P, Khan Q, Williamson S. Acral vascular syndrome during an immune checkpoint inhibitor. BMJ Case Rep 2020; 13:13/5/e233463. [PMID: 32423910 DOI: 10.1136/bcr-2019-233463] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Immune checkpoint inhibitors, including antiprogrammed death cell protein 1 (anti-PD-1) and anti cytotoxic T-lymphocyte-associated protein 4 (anti-CTLA-4), have been associated with a range of autoimmune-related side effects since their introduction in cancer treatment. Small vessel digital necrosis, referred to as the acral vascular syndrome, is a rare but serious complication that can result in loss of digits. Here we present a case report of acral vascular syndrome and review possible aetiologies. A 45- year-old woman with invasive ductal carcinoma of the left breast presented to the emergency department during neoadjuvant treatment with carboplatin, docetaxel and pembrolizumab with complaints of severe pain in her right third digit. She had physical findings consistent with ischaemic necrosis and gangrene of the distal phalanx. Angiography demonstrated Raynaud's phenomenon in the distal portion of the digits. Laboratory testing showed a weakly positive RNA polymerase III antibody level. Her case resulted in surgical amputation of her affected digit after partial resolution of symptoms with prednisone, vasodilators and antibiotics.
Collapse
Affiliation(s)
- Patrick O'Connor
- University of Kansas School of Medicine, Kansas City, Kansas, USA
| | - Pooja Bhadbhade
- Department of Allergy, Clinical Immunology, and Rheumatology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Qamar Khan
- Department of Oncology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Stephen Williamson
- Department of Medical Oncology, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
214
|
Exploiting immune-dependent effects of microtubule-targeting agents to improve efficacy and tolerability of cancer treatment. Cell Death Dis 2020; 11:361. [PMID: 32398657 PMCID: PMC7217828 DOI: 10.1038/s41419-020-2567-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 04/16/2020] [Accepted: 04/24/2020] [Indexed: 02/08/2023]
Abstract
Microtubule-targeting agents (MTAs), like taxanes and vinca alkaloids, are tubulin-binding drugs that are very effective in the treatment of various types of cancers. In cell cultures, these drugs appear to affect assembly of the mitotic spindle and to delay progression through mitosis and this correlates with their ability to induce cell death. Their clinical efficacy is, however, limited by resistance and toxicity. For these reasons, other spindle-targeting drugs, affecting proteins such as certain kinesins like Eg5 and CENP-E, or kinases like Plk1, Aurora A and B, have been developed as an alternative to MTAs. However, these attempts have disappointed in the clinic since these drugs show poor anticancer activity and toxicity ahead of positive effects. In addition, whether efficacy of MTAs in cancer treatment is solely due to their ability to delay mitosis progression remains controversial. Here we discuss recent findings indicating that the taxane paclitaxel can promote a proinflammatory response by activation of innate immunity. We further describe how this can help adaptive antitumor immune response and suggest, on this basis and on the recent success of immune checkpoint inhibitors in cancer treatment, that a combination therapy based on low doses of taxanes and immune checkpoint inhibitors may be of high clinical advantage in terms of wide applicability, reduced toxicity, and increased antitumor response.
Collapse
|
215
|
Lynes JP, Nwankwo AK, Sur HP, Sanchez VE, Sarpong KA, Ariyo OI, Dominah GA, Nduom EK. Biomarkers for immunotherapy for treatment of glioblastoma. J Immunother Cancer 2020; 8:e000348. [PMID: 32474411 PMCID: PMC7264836 DOI: 10.1136/jitc-2019-000348] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/09/2020] [Indexed: 12/25/2022] Open
Abstract
Immunotherapy is a promising new therapeutic field that has demonstrated significant benefits in many solid-tumor malignancies, such as metastatic melanoma and non-small cell lung cancer. However, only a subset of these patients responds to treatment. Glioblastoma (GBM) is the most common malignant primary brain tumor with a poor prognosis of 14.6 months and few treatment advancements over the last 10 years. There are many clinical trials testing immune therapies in GBM, but patient responses in these studies have been highly variable and a definitive benefit has yet to be identified. Biomarkers are used to quantify normal physiology and physiological response to therapies. When extensively characterized and vigorously validated, they have the potential to delineate responders from non-responders for patients treated with immunotherapy in malignancies outside of the central nervous system (CNS) as well as GBM. Due to the challenges of current modalities of radiographic diagnosis and disease monitoring, identification of new predictive and prognostic biomarkers to gauge response to immune therapy for patients with GBM will be critical in the precise treatment of this highly heterogenous disease. This review will explore the current and future strategies for the identification of potential biomarkers in the field of immunotherapy for GBM, as well as highlight major challenges of adapting immune therapy for CNS malignancies.
Collapse
Affiliation(s)
- John P Lynes
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Anthony K Nwankwo
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Hannah P Sur
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Victoria E Sanchez
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Kwadwo A Sarpong
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Oluwatobi I Ariyo
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Gifty A Dominah
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Edjah K Nduom
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
216
|
Urwyler P, Earnshaw I, Bermudez M, Perucha E, Wu W, Ryan S, Mcdonald L, Karagiannis SN, Taams LS, Powell N, Cope A, Papa S. Mechanisms of checkpoint inhibition-induced adverse events. Clin Exp Immunol 2020; 200:141-154. [PMID: 31989585 PMCID: PMC7160658 DOI: 10.1111/cei.13421] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2019] [Indexed: 12/14/2022] Open
Abstract
Immune checkpoint inhibition has revolutionized the treatment of several solid cancers, most notably melanoma and non-small-cell lung cancer (NSCLC). Drugs targeting cytotoxic T lymphocyte antigen (CTLA)-4 and programmed cell death 1 (PD-1) have made their way into routine clinical use; however, this has not been without difficulties. Stimulation of the immune system to target cancer has been found to result in a reduction of self-tolerance, leading to the development of adverse effects that resemble autoimmunity. These adverse effects are erratic in their onset and severity and can theoretically affect any organ type. Several mechanisms for immune-related toxicity have been investigated over recent years; however, no consensus on the cause or prediction of toxicity has been reached. This review seeks to examine reported evidence for possible mechanisms of toxicity, methods for prediction of those at risk and a discussion of future prospects within the field.
Collapse
Affiliation(s)
- P. Urwyler
- Department of Medical OncologyGuy’s and St Thomas’ NHS Foundation TrustLondonUK
| | - I. Earnshaw
- Centre for Inflammation Biology and Cancer ImmunologyDepartment of Inflammatory BiologySchool of Immunology and Microbial SciencesFaculty of Life Sciences and MedicineKing’s College LondonLondonUK
| | - M. Bermudez
- Centre for Inflammation Biology and Cancer ImmunologyDepartment of Inflammatory BiologySchool of Immunology and Microbial SciencesFaculty of Life Sciences and MedicineKing’s College LondonLondonUK
| | - E. Perucha
- Centre for Inflammation Biology and Cancer ImmunologyDepartment of Inflammatory BiologySchool of Immunology and Microbial SciencesFaculty of Life Sciences and MedicineKing’s College LondonLondonUK
| | - W. Wu
- Centre for Inflammation Biology and Cancer ImmunologyDepartment of Inflammatory BiologySchool of Immunology and Microbial SciencesFaculty of Life Sciences and MedicineKing’s College LondonLondonUK
| | - S. Ryan
- Centre for Inflammation Biology and Cancer ImmunologyDepartment of Inflammatory BiologySchool of Immunology and Microbial SciencesFaculty of Life Sciences and MedicineKing’s College LondonLondonUK
| | - L. Mcdonald
- Oncology and Haematology Clinical TrialsGuy’s and St Thomas’ NHS Foundation TrustLondonUK
| | - S. N. Karagiannis
- St John’s Institute of DermatologySchool of Basic and Medical BiosciencesKing’s College LondonLondonUK
| | - L. S. Taams
- Centre for Inflammation Biology and Cancer ImmunologyDepartment of Inflammatory BiologySchool of Immunology and Microbial SciencesFaculty of Life Sciences and MedicineKing’s College LondonLondonUK
| | - N. Powell
- Centre for Inflammation Biology and Cancer ImmunologyDepartment of Inflammatory BiologySchool of Immunology and Microbial SciencesFaculty of Life Sciences and MedicineKing’s College LondonLondonUK
| | - A. Cope
- Centre for Inflammation Biology and Cancer ImmunologyDepartment of Inflammatory BiologySchool of Immunology and Microbial SciencesFaculty of Life Sciences and MedicineKing’s College LondonLondonUK
| | - S. Papa
- Department of Medical OncologyGuy’s and St Thomas’ NHS Foundation TrustLondonUK
- ImmunoEngineeringSchool of Cancer and Pharmaceutical SciencesFaculty of Life Sciences and MedicineKing’s College LondonLondonUK
| |
Collapse
|
217
|
Therapeutic Development of Immune Checkpoint Inhibitors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1248:619-649. [PMID: 32185726 DOI: 10.1007/978-981-15-3266-5_23] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Immune checkpoint blockade (ICB) has been proven to be an effective strategy for enhancing the effector activity of anti-tumor T cells, and checkpoint blockers targeting CTLA-4, PD-1, and PD-L1 have displayed strong and durable clinical responses in certain cancer patients. The new hope brought by ICB therapy has led to the boost in therapeutic development of ICBs in recent years. Nonetheless, the therapeutic efficacy of ICBs varies substantially among cancer types and patients, and only a proportion of cancer patients could benefit from ICBs. The emerging targets and molecules for enhancing anticancer immunity may bring additional therapeutic opportunities for cancer patients. The current challenges in the ICB therapy have been discussed, aimed to provide further strategies for maximizing the efficacy of ICB therapy.
Collapse
|
218
|
de Wolf ACMT, Herberts CA, Hoefnagel MHN. Dawn of Monitoring Regulatory T Cells in (Pre-)clinical Studies: Their Relevance Is Slowly Recognised. Front Med (Lausanne) 2020; 7:91. [PMID: 32300597 PMCID: PMC7142310 DOI: 10.3389/fmed.2020.00091] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 03/03/2020] [Indexed: 12/14/2022] Open
Abstract
Regulatory T cells (Tregs) have a prominent role in the control of immune homeostasis. Pharmacological impact on their activity or balance with effector T cells could contribute to (impaired) clinical responses or adverse events. Monitoring treatment-related effects on T cell subsets may therefore be part of (pre-)clinical studies for medicinal products. However, the extent of immune monitoring performed in studies for marketing authorisation and the degree of correspondence with data available in the public domain is not known. We evaluated the presence of T cell immunomonitoring in 46 registration dossiers of monoclonal antibodies indicated for immune-related disorders and published scientific papers. We found that the depth of Treg analysis in registration dossiers was rather small. Nevertheless, data on treatment-related Treg effects are available in public academia-driven studies (post-registration) and suggest that Tregs may act as a biomarker for clinical responses. However, public data are fragmented and obtained with heterogeneity of experimental approaches from a diversity of species and tissues. To reveal the potential added value of T cell (and particular Treg) evaluation in (pre-)clinical studies, more cell-specific data should be acquired, at least for medicinal products with an immunomodulatory mechanism. Therefore, extensive analysis of T cell subset contribution to clinical responses and the relevance of treatment-induced changes in their levels is needed. Preferably, industry and academia should work together to obtain these data in a standardised manner and to enrich our knowledge about T cell activity in disease pathogenesis and therapies. This will ultimately elucidate the necessity of T cell subset monitoring in the therapeutic benefit-risk assessment.
Collapse
|
219
|
Bellan DL, Mazepa E, Biscaia SMP, Gonçalves JP, Oliveira CC, Rossi GR, Ferreira LG, Noseda MD, Trindade ES, Duarte MER, Franco CRC. Non-Cytotoxic Sulfated Heterorhamnan from Gayralia brasiliensis Green Seaweed Reduces Driver Features of Melanoma Metastatic Progression. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2020; 22:194-206. [PMID: 31970542 DOI: 10.1007/s10126-020-09944-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 01/02/2020] [Indexed: 06/10/2023]
Abstract
Melanoma is a form of skin cancer with high mortality owing to its fast progression and metastatic capacity. The treatments available nowadays are only palliative in advanced stages of the disease. Thus, alternative therapies for cancer treatment are in demand, and molecules from natural sources, such as polysaccharides, could represent new possible therapeutic approaches. Polysaccharides of freshwater and marine algae with biological activities, such as antitumor properties, are greatly reported in the scientific literature. In the present study, a sulfated heterorhamnan obtained from the green seaweed Gayralia brasiliensis (Gb1 fraction) was chemically characterized and its biological activities in the B16-F10 murine melanoma cell line were evaluated. The Gb1 polysaccharidic fraction tested concentrations presented low or absence of cytotoxicity to B16-F10 cells and neither cell proliferation nor cell cycle were altered. Interestingly, Gb1 treatment decreased B16-F10 cells migration and invasion capabilities and CD44 labeling, showing to be a promising compound for further in vitro and in vivo antitumor studies.
Collapse
Affiliation(s)
- D L Bellan
- Department of Cellular Biology, Federal University of Paraná, Curitiba, Paraná, Brazil.
| | - E Mazepa
- Department of Biochemistry, Federal University of Paraná, Curitiba, Paraná, Brazil
| | - S M P Biscaia
- Department of Cellular Biology, Federal University of Paraná, Curitiba, Paraná, Brazil
| | - J P Gonçalves
- Department of Cellular Biology, Federal University of Paraná, Curitiba, Paraná, Brazil
| | - C C Oliveira
- Department of Cellular Biology, Federal University of Paraná, Curitiba, Paraná, Brazil
| | - G R Rossi
- Department of Cellular Biology, Federal University of Paraná, Curitiba, Paraná, Brazil
| | - L G Ferreira
- Department of Biochemistry, Federal University of Paraná, Curitiba, Paraná, Brazil
| | - M D Noseda
- Department of Biochemistry, Federal University of Paraná, Curitiba, Paraná, Brazil
| | - E S Trindade
- Department of Cellular Biology, Federal University of Paraná, Curitiba, Paraná, Brazil
| | - M E R Duarte
- Department of Biochemistry, Federal University of Paraná, Curitiba, Paraná, Brazil.
| | - C R C Franco
- Department of Cellular Biology, Federal University of Paraná, Curitiba, Paraná, Brazil.
| |
Collapse
|
220
|
Active targeted ligand-aza-BODIPY conjugate for near-infrared photodynamic therapy in melanoma. Int J Pharm 2020; 579:119189. [DOI: 10.1016/j.ijpharm.2020.119189] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 02/11/2020] [Accepted: 02/28/2020] [Indexed: 01/02/2023]
|
221
|
Fromer MW, Scoggins CR. Article Commentary: Cancer Immunotherapy for the General Surgeon. Am Surg 2020. [DOI: 10.1177/000313482008600423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Progress in the arena of cancer immunotherapy has been immense in recent years. The fact remains that most of the cancer resections in the United States are performed by general surgeons and not oncologic specialists. A busy practice in general surgery will invariably make it difficult to keep pace with such rapid advancement. This review offers a concise summary of the major concepts and trials that have driven the immunotherapy revolution and their implications for surgeons who deliver cancer care.
Collapse
Affiliation(s)
- Marc W. Fromer
- From the Division of Surgical Oncology, Hiram C. Polk, Jr., MD Department of Surgery, University of Louisville School of Medicine, Louisville, Kentucky
| | - Charles R. Scoggins
- From the Division of Surgical Oncology, Hiram C. Polk, Jr., MD Department of Surgery, University of Louisville School of Medicine, Louisville, Kentucky
| |
Collapse
|
222
|
Abstract
Treatment options for patients with metastatic melanoma have expanded rapidly since the approval of ipilimumab by the U.S. Food and Drug Administration in 2011. Cytokines such as interferon and interleukin-2 were approved in 1995 and 1998 respectively. However, the effect on survival was marginal, and the toxicity, substantial. Multiple vaccine studies likewise failed to show improvements in survival. The "Holy Grail" came with the discovery of immune checkpoints, and the first metastatic melanoma trial to show an improvement in overall survival involved the use of an immune checkpoint inhibitor against ctla-4: ipilimumab. Since then, the field of immuno-oncology has exploded, with approvals for PD-1 inhibitors and discovery, in clinical trials, of several novel checkpoints such as tim-3, lag-3, and others. In fact more than 950 novel immunotherapy drugs are currently being trialled. Recently, combinations of ctla-4 and PD-1 inhibitors have been associated with 1-year survival rates exceeding 80% and 4-year survival rates greater than 50%. In no tumour has as much progress been made in the last 5 years as in melanoma, and the efforts to unravel and exploit mechanisms used by the tumour to avoid immune detection are just beginning.
Collapse
|
223
|
Singh S, Numan A, Agrawal N, Tambuwala MM, Singh V, Kesharwani P. Role of immune checkpoint inhibitors in the revolutionization of advanced melanoma care. Int Immunopharmacol 2020; 83:106417. [PMID: 32200155 DOI: 10.1016/j.intimp.2020.106417] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/12/2020] [Accepted: 03/13/2020] [Indexed: 12/17/2022]
Abstract
Melanoma cancer is an important public health concern owing to its prevalence, high recurrence risk, treatment failures and immunosuppressive abilities. Prolonged immune system activation is the main objective of immune checkpoint inhibitors (ICIs) therapies directed against melanoma cancer. Despite the staggering advancements in approved ICIs therapy effectiveness, immune-related adverse events (imAEs) and therapeutic resistance has limited its wide application. Thus, there is a need to establish biomarkers that predict the response to ICIs and imAEs. In this review article, we provide an in-depth understanding of the role of tolerance, immunity, and immunosuppression in antitumor immune response regulation, together with ongoing clinical therapy and suggested biomarkers. These attainments advise that approved ICIs provide a novel approach to durable and prolonged response in cancer patients and will aid in the reduction of treatment cost and duration and enhance patient recovery.
Collapse
Affiliation(s)
- Sima Singh
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban, South Africa
| | - Arshid Numan
- State Key Laboratory of ASIC and System, SIST, Fudan University, 200433 Shanghai, China
| | - Nikhil Agrawal
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban, South Africa
| | - Murtaza M Tambuwala
- SAAD Centre for Pharmacy and Diabetes, School of Pharmacy and Pharmaceutical Science, Ulster University, Newtownabbey BT370QB, London, United Kingdom
| | - Vijender Singh
- School of Pharmacy, Sharda University, Greater Noida 201310, Uttar Pradesh, India
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard 110062, New Delhi, India. https://scholar.google.com/citations?user=DJkvOAQAAAAJ&hl=en
| |
Collapse
|
224
|
Remon J, Passiglia F, Ahn MJ, Barlesi F, Forde PM, Garon EB, Gettinger S, Goldberg SB, Herbst RS, Horn L, Kubota K, Lu S, Mezquita L, Paz-Ares L, Popat S, Schalper KA, Skoulidis F, Reck M, Adjei AA, Scagliotti GV. Immune Checkpoint Inhibitors in Thoracic Malignancies: Review of the Existing Evidence by an IASLC Expert Panel and Recommendations. J Thorac Oncol 2020; 15:914-947. [PMID: 32179179 DOI: 10.1016/j.jtho.2020.03.006] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 02/29/2020] [Accepted: 03/06/2020] [Indexed: 02/07/2023]
Abstract
In the past 10 years, a deeper understanding of the immune landscape of cancers, including immune evasion processes, has allowed the development of a new class of agents. The reactivation of host antitumor immune response offers the potential for long-term survival benefit in a portion of patients with thoracic malignancies. The advent of programmed cell death protein 1/programmed death ligand-1 immune checkpoint inhibitors (ICIs), both as single agents and in combination with chemotherapy, and more recently, the combination of ICI, anti-programmed cell death protein 1, and anticytotoxic T-lymphocyte antigen 4 antibody, have led to breakthrough therapeutic advances for patients with advanced NSCLC, and to a lesser extent, patients with SCLC. Encouraging activity has recently emerged in pretreated patients with thymic carcinoma (TC). Conversely, in malignant pleural mesothelioma, pivotal positive signs of activity have not been fully confirmed in randomized trials. The additive effects of chemoradiation and immunotherapy suggested intriguing potential for therapeutic synergy with combination strategies. This has led to the introduction of ICI consolidation therapy in stage III NSCLC, creating a platform for future therapeutic developments in earlier-stage disease. Despite the definitive clinical benefit observed with ICI, primary and acquired resistance represent well-known biological phenomena, which may affect the therapeutic efficacy of these agents. The development of innovative strategies to overcome ICI resistance, standardization of new patterns of ICI progression, identification of predictive biomarkers of response, optimal treatment duration, and characterization of ICI efficacy in special populations, represent crucial issues to be adequately addressed, with the aim of improving the therapeutic benefit of ICI in patients with thoracic malignancies. In this article, an international panel of experts in the field of thoracic malignancies discussed these topics, evaluating currently available scientific evidence, with the final aim of providing clinical recommendations, which may guide oncologists in their current practice and elucidate future treatment strategies and research priorities.
Collapse
Affiliation(s)
- Jordi Remon
- Department of Medical Oncology, Centro Integral Oncológico Clara Campal (HM-CIOCC), Hospital HM Delfos, HM Hospitales, Barcelona, Spain
| | - Francesco Passiglia
- Department of Oncology, University of Torino, AOU S. Luigi Gonzaga, Orbassano, Italy
| | - Myung-Ju Ahn
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Fabrice Barlesi
- Multidisciplinary Oncology and Therapeutic Innovations Department, Aix Marseille University, CNRS, INSERM, CRCM, APHM, Marseille, France
| | - Patrick M Forde
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Edward B Garon
- David Geffen School of Medicine at University of California Los Angeles, Translational Research in Oncology US Network, Los Angeles, California
| | - Scott Gettinger
- Department of Medicine (Section of Medical Oncology), Yale School of Medicine, New Haven, Connecticut
| | - Sarah B Goldberg
- Department of Medicine (Section of Medical Oncology), Yale School of Medicine, New Haven, Connecticut
| | - Roy S Herbst
- Department of Medicine (Section of Medical Oncology), Yale School of Medicine, New Haven, Connecticut
| | - Leora Horn
- Department of Hematology and Oncology, Vanderbilt Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee
| | - Kaoru Kubota
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Shun Lu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Laura Mezquita
- Department of Cancer Medicine, Gustave Roussy Cancer Campus, Villejuif, France
| | - Luis Paz-Ares
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Sanjay Popat
- Medical Oncology Department, The Royal Marsden Hospital, London, United Kingdom; Medical Oncology Department, The Institute of Cancer Research, London, United Kingdom; National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Kurt A Schalper
- Departments of Pathology and Medicine (Section of Medical Oncology), Yale School of Medicine, New Haven, Connecticut
| | - Ferdinandos Skoulidis
- Department of Thoracic and Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Martin Reck
- Lung Clinic Grosshansdorf, Airway Research Center North, German Center of Lung Research, Grosshansdorf, Germany
| | - Alex A Adjei
- Department of Oncology, Mayo Clinic, Rochester, Minnesota
| | - Giorgio V Scagliotti
- Department of Oncology, University of Torino, AOU S. Luigi Gonzaga, Orbassano, Italy.
| |
Collapse
|
225
|
Chang CY, Park H, Malone DC, Wang CY, Wilson DL, Yeh YM, Van Boemmel-Wegmann S, Lo-Ciganic WH. Immune Checkpoint Inhibitors and Immune-Related Adverse Events in Patients With Advanced Melanoma: A Systematic Review and Network Meta-analysis. JAMA Netw Open 2020; 3:e201611. [PMID: 32211869 PMCID: PMC7097702 DOI: 10.1001/jamanetworkopen.2020.1611] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Accepted: 02/02/2020] [Indexed: 12/17/2022] Open
Abstract
Importance Since 2011, immune checkpoint inhibitors (ICIs) have been effective treatment options for advanced melanoma. Little is known about how risks of immune-related adverse events (irAEs) vary by ICIs. Objective To compare the risk of irAEs across different treatment regimens for advanced melanoma using network meta-analysis. Data Sources PubMed/MEDLINE, Embase, Web of Science, and Scopus were searched for all randomized clinical trial (RCT) articles published from January 1, 2010, through June 30, 2019. Study Selection Studies included phases 2 and 3 RCTs in the treatment of advanced melanoma that compared ICIs (ipilimumab, nivolumab, and pembrolizumab) with chemotherapy drugs (eg, dacarbazine, carboplatin, and paclitaxel) or different ICI regimens. Data Extraction and Synthesis Different treatment regimens were compared using bayesian network meta-analysis with Markov chain Monte Carlo simulation with noninformative prior distribution and random-effects generalized linear models. Main Outcomes and Measures Primary outcomes were the cumulative incidence of any irAEs (regardless of severity) and severe irAEs (grades 3-5). Based on the pooled odds ratios (ORs) and 95% credible intervals (95% CrI), the probability of being associated with the lowest irAE risks was estimated for each treatment regimen. Results Nine RCTs with 8 different treatment regimens for advanced melanoma and involving a total of 5051 patients were included. Overall, the 3 ICI treatment regimens associated with the lowest risk of any or severe irAEs were pembrolizumab, 2 mg/kg, every 3 weeks; nivolumab, 3 mg/kg, every 2 weeks; and pembrolizumab, 10 mg/kg, every 3 weeks. Compared with ipilimumab, 10 mg/kg, every 3 weeks, only nivolumab, 3 mg/kg, every 2 weeks, was associated with a decreased risk for any irAEs (OR, 0.34; 95% CrI, 0.13-0.94). A decreased risk for severe irAEs was observed for ipilimumab, 3 mg/kg, every 3 weeks (OR, 0.35; 95% CrI, 0.14-0.74); pembrolizumab, 10 mg/kg, every 2 weeks (OR, 0.22; 95% CrI, 0.05-0.95) and 10 mg/kg every 3 weeks (OR, 0.20; 95% CrI, 0.06-0.68); and nivolumab, 3 mg/kg, every 2 weeks (OR, 0.20; 95% CrI, 0.07-0.48) compared with ipilimumab, 10 mg/kg, every 3 weeks. An increased risk for severe irAEs was associated with nivolumab, 1 mg/kg, every 3 weeks combined with ipilimumab, 3 mg/kg, every 3 weeks compared with other ICI regimens (ORs ranging from 4.09 [95% CrI, 1.73-10.99] to 7.40 [95% CrI, 1.12-49.29]) except ipilimumab, 10 mg/kg, every 3 weeks. Conclusions and Relevance These findings suggest that for patients with advanced melanoma at high risk of irAEs, pembrolizumab, 2 mg/kg, every 3 weeks, nivolumab, 3 mg/kg, every 2 weeks, and pembrolizumab, 10 mg/kg, every 3 weeks may be the preferred treatment regimens (with respect to irAE risks) among the ICI regimens reported, whereas ipilimumab, 10 mg/kg, every 3 weeks alone and nivolumab, 1 mg/kg, every 3 weeks combined with ipilimumab, 3 mg/kg, every 3 weeks should be used with caution. A network analysis may be valuable for clinical decision-making when evidence from head-to-head comparisons is lacking.
Collapse
Affiliation(s)
- Ching-Yuan Chang
- Department of Pharmaceutical Outcomes and Policy, University of Florida College of Pharmacy, Gainesville
- Center for Drug Evaluation and Safety, University of Florida College of Pharmacy, Gainesville
| | - Haesuk Park
- Department of Pharmaceutical Outcomes and Policy, University of Florida College of Pharmacy, Gainesville
- Center for Drug Evaluation and Safety, University of Florida College of Pharmacy, Gainesville
| | - Daniel C. Malone
- Department of Pharmacotherapy, The University of Utah College of Pharmacy, Salt Lake City
| | - Ching-Yu Wang
- Department of Pharmaceutical Outcomes and Policy, University of Florida College of Pharmacy, Gainesville
- Center for Drug Evaluation and Safety, University of Florida College of Pharmacy, Gainesville
| | - Debbie L. Wilson
- Department of Pharmaceutical Outcomes and Policy, University of Florida College of Pharmacy, Gainesville
| | - Yu-Min Yeh
- National Cheng-Kung University Hospital, Department of Internal Medicine, National Cheng Kung University College of Medicine, Tainan, Taiwan
| | - Sascha Van Boemmel-Wegmann
- Department of Pharmaceutical Outcomes and Policy, University of Florida College of Pharmacy, Gainesville
- Center for Drug Evaluation and Safety, University of Florida College of Pharmacy, Gainesville
| | - Wei-Hsuan Lo-Ciganic
- Department of Pharmaceutical Outcomes and Policy, University of Florida College of Pharmacy, Gainesville
- Center for Drug Evaluation and Safety, University of Florida College of Pharmacy, Gainesville
| |
Collapse
|
226
|
Singh BP, Marshall JL, He AR. Workup and Management of Immune-Mediated Colitis in Patients Treated with Immune Checkpoint Inhibitors. Oncologist 2020; 25:197-202. [PMID: 32162824 PMCID: PMC7066712 DOI: 10.1634/theoncologist.2018-0304] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 08/19/2019] [Indexed: 12/14/2022] Open
Abstract
As the use of immune checkpoint inhibitors for several different malignancies becomes more mainstream, their side-effect profile raises new challenges. In 2011, the Food and Drug Administration approved the first checkpoint inhibitor for the treatment of advanced melanoma, and since then, checkpoint inhibitors have demonstrated efficacy in many other tumor types. Given the frequent use of immune checkpoint inhibitors in a wide range of cancers today, the diagnosis and management of their immune-mediated toxicities need special attention. One of the most common is immune-mediated colitis. Workup and management of immune-mediated colitis can be challenging and is the purpose of this review. KEY POINTS: Rate of immune mediated colitis differ from different kind of immune checkpoint inhibitor treatment. To work up immune-mediated colitis, tests to rule out infectious etiologies of diarrhea, colonoscopy and abdominal image will help to differentiate immune mediated colitis from colitis from other etiology. Patients with mild colitis can be managed with supportive therapies alone, but more severe cases may require immunomodulators such as steroid. Refractory cases may require tumor necrosis factor (TNF) inhibitors, such as infliximab in addition to steroid treatment.
Collapse
Affiliation(s)
- Bhavana Pendurthi Singh
- Ruesch Center for the Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical CenterWashingtonDCUSA
| | - John L. Marshall
- Ruesch Center for the Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical CenterWashingtonDCUSA
| | - Aiwu Ruth He
- Ruesch Center for the Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical CenterWashingtonDCUSA
| |
Collapse
|
227
|
Presotto EM, Rastrelli G, Desideri I, Scotti V, Gunnella S, Pimpinelli N, Vaccher E, Bearz A, Di Costanzo F, Bruggia M, Mini E, Maggi M, Peri A. Endocrine toxicity in cancer patients treated with nivolumab or pembrolizumab: results of a large multicentre study. J Endocrinol Invest 2020; 43:337-345. [PMID: 31542865 DOI: 10.1007/s40618-019-01112-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 09/03/2019] [Indexed: 12/12/2022]
Abstract
INTRODUCTION The immune checkpoint inhibitors (ICPIs) agents anti-T lymphocytes-associated antigen 4 (CTLA-4) and anti-programmed cell death protein-1 (PD-1) and its ligands (PD-L1/PD-L2) have opened a new scenario in the treatment of cancer. These agents can induce immuno-related adverse events (irAEs), which may affect the endocrine system. PURPOSE The aim of this study was to analyze the occurrence and the course of endocrine irAEs in cancer patients treated with anti-PD-1 immunotherapy. METHODS This was a retrospective, multicentre study, involving cancer patients treated with the PD-1 inhibitors nivolumab or pembrolizumab at reference Oncology Centres. One hundred and seventy-nine consecutive patients with different types of cancer (mostly non-small cell lung cancer, melanoma, kidney cancer) were included in the study. Patients had received nivolumab (70.9%) or pembrolizumab (29.1%) for 2-33 months. The study evaluated clinical data records until the established date of July 15, 2018. The primary end point was the assessment of endocrine toxicity and possible predictive factors. RESULTS Endocrine toxicity occurred in 54 out of 179 patients (30.2%) and was related to thyroid dysfunction, with the exception of one case of diabetes mellitus. Thyroid toxicity occurred mostly within 2 months from the initiation of immunotherapy (83% of cases). A pre-existing thyroid dysfunction was a significant predictor of disease flare. CONCLUSIONS Thyroid alterations are frequently associated with anti PD-1 treatment in cancer patients. Regular thyroid assessment should be performed, particularly in the first months of treatment and in patients with a pre-existing thyroid disease.
Collapse
Affiliation(s)
- E M Presotto
- Endocrinology, Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, AOU Careggi, Viale Pieraccini, 6, 50139, Florence, Italy
| | - G Rastrelli
- Andrology, Female Endocrinology and Gender Incongruence Unit, Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, AOU Careggi, 50139, Florence, Italy
| | - I Desideri
- Radiation Oncology Unit, Department of Oncology and Experimental Clinical and Biomedical Sciences "Mario Serio", AOU Careggi, University of Florence, Florence, Italy
| | - V Scotti
- Radiation Oncology Unit, Department of Oncology and Experimental Clinical and Biomedical Sciences "Mario Serio", AOU Careggi, University of Florence, Florence, Italy
| | - S Gunnella
- Melanoma and Skin Cancer Unit, Tuscany Central District, Department of Health Sciences, Dermatology Unit, University of Florence, Florence, Italy
| | - N Pimpinelli
- Melanoma and Skin Cancer Unit, Tuscany Central District, Department of Health Sciences, Dermatology Unit, University of Florence, Florence, Italy
| | - E Vaccher
- Medical Oncology and Immuno-Related Tumors, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - A Bearz
- Medical Oncology and Immuno-Related Tumors, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | | | - M Bruggia
- Unit of Translational Oncology, AOU Careggi, Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - E Mini
- Unit of Translational Oncology, AOU Careggi, Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - M Maggi
- Endocrinology, Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, AOU Careggi, Viale Pieraccini, 6, 50139, Florence, Italy
| | - A Peri
- Endocrinology, Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, AOU Careggi, Viale Pieraccini, 6, 50139, Florence, Italy.
| |
Collapse
|
228
|
Betof Warner A, Postow MA. Another Victory for Immune Checkpoint Blockade in Melanoma: Adjuvant Ipilimumab Over Interferon. J Clin Oncol 2020; 38:529-531. [DOI: 10.1200/jco.19.02988] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Allison Betof Warner
- Memorial Sloan Kettering Cancer Center, New York, NY
- Weill Cornell Medical College, New York, NY
| | - Michael A. Postow
- Memorial Sloan Kettering Cancer Center, New York, NY
- Weill Cornell Medical College, New York, NY
| |
Collapse
|
229
|
Neurotoxicity associated with cancer immunotherapy: immune checkpoint inhibitors and chimeric antigen receptor T-cell therapy. Curr Opin Neurol 2020; 32:500-510. [PMID: 30893101 DOI: 10.1097/wco.0000000000000686] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
PURPOSE OF REVIEW Immune checkpoint inhibitors (ICPI) and chimeric antigen receptor T cells (CAR-T) represent novel therapies recently approved to treat a number of human cancers. As both approaches modulate the immune system, they can generate a number of immune-related adverse events (irAEs), including a large spectrum of novel neurological toxicities. These are of special interest given their potential severity and risk of compromising further oncologic treatment. We aim to provide a comprehensive review of the literature and discuss their optimal management. RECENT FINDINGS In contrast to irAEs involving other organs, neurological complications of ICPI are uncommon, may present throughout the course of treatment and involve the peripheral and central nervous system, including polyneuropathy, myositis, myasthenia gravis, demyelinating polyradiculopathy, myelitis, encephalitis and others. If started early, ICPI-related neurologic irAEs are usually responsive to steroids. In contrast, as many as 40% of patients undergoing CAR-T therapy will develop neurologic complications in the form of a cytokine-release-associated encephalopathy. It includes delirium, aphasia, tremor/myoclonus, seizure and seizure-like activity. SUMMARY irAEs associated with CAR-T and ICPI therapy constitute new entities. Early identification and treatment are essential to optimize the functional outcome and further oncologic management of the patient.
Collapse
|
230
|
Tarhini AA, Lee SJ, Hodi FS, Rao UNM, Cohen GI, Hamid O, Hutchins LF, Sosman JA, Kluger HM, Eroglu Z, Koon HB, Lawrence DP, Kendra KL, Minor DR, Lee CB, Albertini MR, Flaherty LE, Petrella TM, Streicher H, Sondak VK, Kirkwood JM. Phase III Study of Adjuvant Ipilimumab (3 or 10 mg/kg) Versus High-Dose Interferon Alfa-2b for Resected High-Risk Melanoma: North American Intergroup E1609. J Clin Oncol 2020; 38:567-575. [PMID: 31880964 PMCID: PMC7030886 DOI: 10.1200/jco.19.01381] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2019] [Indexed: 12/28/2022] Open
Abstract
PURPOSE Phase III adjuvant trials have reported significant benefits in both relapse-free survival (RFS) and overall survival (OS) for high-dose interferon alfa (HDI) and ipilimumab at 10 mg/kg (ipi10). E1609 evaluated the safety and efficacy of ipilimumab at 3 mg/kg (ipi3) and ipi10 versus HDI. PATIENTS AND METHODS E1609 was a phase III trial in patients with resected cutaneous melanoma (American Joint Committee on Cancer 7th edition stage IIIB, IIIC, M1a, or M1b). It had 2 coprimary end points: OS and RFS. A 2-step hierarchic approach first evaluated ipi3 versus HDI followed by ipi10 versus HDI. RESULTS Between May 2011 and August 2014, 1,670 adult patients were centrally randomly assigned (1:1:1) to ipi3 (n = 523), HDI (n = 636), or ipi10 (n = 511). Treatment-related adverse events grade ≥ 3 occurred in 37% of patients receiving ipi3, 79% receiving HDI, and 58% receiving ipi10, with adverse events leading to treatment discontinuation in 35%, 20%, and 54%, respectively. Comparison of ipi3 versus HDI used an intent-to-treat analysis of concurrently randomly assigned patient cases (n = 1,051) and showed significant OS difference in favor of ipi3 (hazard ratio [HR], 0.78; 95.6% repeated CI, 0.61 to 0.99; P = .044; RFS: HR, 0.85; 99.4% CI, 0.66 to 1.09; P = .065). In the second step, for ipi10 versus HDI (n = 989), trends in favor of ipi10 did not achieve statistical significance. Salvage patterns after melanoma relapse showed significantly higher rates of ipilimumab and ipilimumab/anti-programmed death 1 use in the HDI arm versus ipi3 and ipi10 (P ≤ .001). CONCLUSION Adjuvant therapy with ipi3 benefits survival versus HDI; for the first time to our knowledge in melanoma adjuvant therapy, E1609 has demonstrated a significant improvement in OS against an active control regimen. The currently approved adjuvant ipilimumab dose (ipi10) was more toxic and not superior in efficacy to HDI.
Collapse
Affiliation(s)
| | - Sandra J. Lee
- Harvard Medical School, Boston, MA
- Dana-Farber Cancer Institute, Boston, MA
| | | | - Uma N. M. Rao
- University of Pittsburgh Medical Center, Pittsburgh, PA
| | | | - Omid Hamid
- Angeles Clinic & Research Institute, Santa Monica, CA
| | | | | | | | - Zeynep Eroglu
- H. Lee Moffitt Comprehensive Cancer Center, Tampa, FL
| | | | | | | | - David R. Minor
- Sutter-California Pacific Medical Center, San Francisco, CA
| | - Carrie B. Lee
- University of North Carolina at Chapel Hill, Chapel Hill, NC
| | | | | | | | | | | | | |
Collapse
|
231
|
Current Advances in the Treatment of BRAF-Mutant Melanoma. Cancers (Basel) 2020; 12:cancers12020482. [PMID: 32092958 PMCID: PMC7072236 DOI: 10.3390/cancers12020482] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 02/02/2020] [Accepted: 02/14/2020] [Indexed: 12/12/2022] Open
Abstract
Melanoma is the most lethal form of skin cancer. Melanoma is usually curable with surgery if detected early, however, treatment options for patients with metastatic melanoma are limited and the five-year survival rate for metastatic melanoma had been 15-20% before the advent of immunotherapy. Treatment with immune checkpoint inhibitors has increased long-term survival outcomes in patients with advanced melanoma to as high as 50% although individual response can vary greatly. A mutation within the MAPK pathway leads to uncontrollable growth and ultimately develops into cancer. The most common driver mutation that leads to this characteristic overactivation in the MAPK pathway is the B-RAF mutation. Current combinations of BRAF and MEK inhibitors that have demonstrated improved patient outcomes include dabrafenib with trametinib, vemurafenib with cobimetinib or encorafenib with binimetinib. Treatment with BRAF and MEK inhibitors has met challenges as patient responses began to drop due to the development of resistance to these inhibitors which paved the way for development of immunotherapies and other small molecule inhibitor approaches to address this. Resistance to these inhibitors continues to push the need to expand our understanding of novel mechanisms of resistance associated with treatment therapies. This review focuses on the current landscape of how resistance occurs with the chronic use of BRAF and MEK inhibitors in BRAF-mutant melanoma and progress made in the fields of immunotherapies and other small molecules when used alone or in combination with BRAF and MEK inhibitors to delay or circumvent the onset of resistance for patients with stage III/IV BRAF mutant melanoma.
Collapse
|
232
|
Almutairi AR, McBride A, Slack M, Erstad BL, Abraham I. Potential Immune-Related Adverse Events Associated With Monotherapy and Combination Therapy of Ipilimumab, Nivolumab, and Pembrolizumab for Advanced Melanoma: A Systematic Review and Meta-Analysis. Front Oncol 2020; 10:91. [PMID: 32117745 PMCID: PMC7033582 DOI: 10.3389/fonc.2020.00091] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 01/17/2020] [Indexed: 12/19/2022] Open
Abstract
Background: The use of ipilimumab, nivolumab, and pembrolizumab as monotherapies or in combination has transformed the management of advanced melanoma even though these drugs are associated with a new profile of immune-related adverse events (irAEs). The incidence of irAEs from clinical trials of these agents is an important factor for clinicians when treating patients with advanced melanoma. In the current study, we aimed to profile the incidence of potential irAEs of these agents when used as monotherapy and as combination therapy. Methods: We searched the Medline, Embase, and Cochrane databases; clinicaltrials.gov; and websites of regulatory agencies in the USA, Europe, Australia, and Japan for phase 1-3 trials of ipilimumab, nivolumab, and pembrolizumab for advanced melanoma. Random effect meta-analysis was utilized to profile the incidence of potential irAEs. Results: A total of 58 reports of 35 trials including 6,331 patients with advanced melanoma and reporting irAE data were included in the meta-analyses. We found higher incidences of potential irAEs in combination therapies vs. monotherapies for most of the types of irAEs. Among the monotherapies, ipilimumab users had the most frequent incidence of potential irAEs related to the gastrointestinal system (diarrhea, 29%; and colitis, 8%) and skin (rash, 31%; pruritus, 27%; and dermatitis, 10%), with hypophysitis in 4% of the patients. The most frequent potential irAEs among nivolumab users were maculopapular rash (13%), erythema (4%), hepatitis (3%), and infusion-related reactions (3%), while they were arthralgia (12%), hypothyroidism (8%), and hyperglycemia (6%), among pembrolizumab users. Conclusion: Especially the combination therapies tend to elevate the incidence of potential irAEs. Clinicians should be vigilant about irAEs following combination therapy as well as gastrointestinal and skin irAEs following ipilimumab therapy, in addition to being aware of potential irAEs leading to hyperglycemia, thyroid, hepatic, and musculoskeletal disorders following nivolumab and pembrolizumab therapy.
Collapse
Affiliation(s)
- Abdulaali R Almutairi
- Center for Health Outcomes and PharmacoEconomic Research, College of Pharmacy, University of Arizona, Tucson, AZ, United States.,Department of Pharmacy Practice and Science, College of Pharmacy, University of Arizona, Tucson, AZ, United States
| | - Ali McBride
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Arizona, Tucson, AZ, United States.,University of Arizona Cancer Center, Tucson, AZ, United States
| | - Marion Slack
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Arizona, Tucson, AZ, United States
| | - Brian L Erstad
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Arizona, Tucson, AZ, United States
| | - Ivo Abraham
- Center for Health Outcomes and PharmacoEconomic Research, College of Pharmacy, University of Arizona, Tucson, AZ, United States.,Department of Pharmacy Practice and Science, College of Pharmacy, University of Arizona, Tucson, AZ, United States.,University of Arizona Cancer Center, Tucson, AZ, United States
| |
Collapse
|
233
|
Desnoyer A, Broutin S, Delahousse J, Maritaz C, Blondel L, Mir O, Chaput N, Paci A. Pharmacokinetic/pharmacodynamic relationship of therapeutic monoclonal antibodies used in oncology: Part 2, immune checkpoint inhibitor antibodies. Eur J Cancer 2020; 128:119-128. [PMID: 32037060 DOI: 10.1016/j.ejca.2020.01.003] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 01/02/2020] [Accepted: 01/07/2020] [Indexed: 10/25/2022]
Abstract
Immune checkpoint inhibitors are monoclonal antibodies (mAbs) directed against negative immunologic regulators that are used to restore the immune response against cancer. Approved drugs include anti-cytotoxic T-lymphocyte antigen 4 (CTLA-4), anti-programmed cell death 1 (PD-1) and anti-programmed cell death-ligand 1 (PD-L1) antibodies exhibiting pharmacokinetic (PK) characteristics typical of mAbs. Most factors such as age, sex, ethnicity, tumour burden, performance status and immunogenicity, but not body weight, do not seem to affect drug clearance clinically. However, an exposure-response relation has been described for both the efficacy and toxicity of anti-CTLA-4 and anti-PD-1 agents. The change in clearance over time is associated with overall response at least for nivolumab and pembrolizumab. Few PK/pharmacodynamic (PD) data are available for anti-PD-L1 mAbs, but time-varying clearance has been described for these drugs, and the high immunogenicity rate observed with atezolizumab may affect PK parameters and should be further studied. These data suggest the need for additional PK/PD studies. In this review, we summarise studies of the PKs of immune checkpoint inhibitors, exploring possible interactions with PD considerations.
Collapse
Affiliation(s)
- Aude Desnoyer
- University Paris-Saclay, Faculty of Pharmacy, Chatenay-Malabry, F-92290, France; Gustave Roussy Cancer Campus, Laboratory of Immunomonitoring in Oncology, Villejuif, F-94805, France.
| | - Sophie Broutin
- Gustave Roussy Cancer Campus, Department of Pharmacology, Villejuif, F-94805, France.
| | - Julia Delahousse
- Gustave Roussy Cancer Campus, Department of Pharmacology, Villejuif, F-94805, France.
| | - Christophe Maritaz
- University Paris-Saclay, Faculty of Pharmacy, Chatenay-Malabry, F-92290, France.
| | - Louis Blondel
- University Paris-Saclay, Faculty of Pharmacy, Chatenay-Malabry, F-92290, France.
| | - Olivier Mir
- Gustave Roussy Cancer Campus, Department of Ambulatory Care, Villejuif, F-94805, France.
| | - Nathalie Chaput
- University Paris-Saclay, Faculty of Pharmacy, Chatenay-Malabry, F-92290, France; Gustave Roussy Cancer Campus, Laboratory of Immunomonitoring in Oncology, Villejuif, F-94805, France.
| | - Angelo Paci
- University Paris-Saclay, Faculty of Pharmacy, Chatenay-Malabry, F-92290, France; Gustave Roussy Cancer Campus, Department of Pharmacology, Villejuif, F-94805, France.
| |
Collapse
|
234
|
Herrera-Rios D, Mughal SS, Teuber-Hanselmann S, Pierscianek D, Sucker A, Jansen P, Schimming T, Klode J, Reifenberger J, Felsberg J, Keyvani K, Brors B, Sure U, Reifenberger G, Schadendorf D, Helfrich I. Macrophages/Microglia Represent the Major Source of Indolamine 2,3-Dioxygenase Expression in Melanoma Metastases of the Brain. Front Immunol 2020; 11:120. [PMID: 32117271 PMCID: PMC7013086 DOI: 10.3389/fimmu.2020.00120] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 01/16/2020] [Indexed: 01/12/2023] Open
Abstract
The manifestation of brain metastases in patients with advanced melanoma is a common event that limits patient's survival and quality of life. The immunosuppressive properties of the brain parenchyma are very different compared to the rest of the body, making it plausible that the current success of cancer immunotherapies is specifically limited here. In melanoma brain metastases, the reciprocal interplay between immunosuppressive mediators such as indoleamine 2, 3-dioxygenase (IDO) or programmed cell death-ligand 1 (PD-L1) in the context of neoplastic transformation are far from being understood. Therefore, we analyzed the immunoreactive infiltrate (CD45, CD3, CD8, Forkhead box P3 [FoxP3], CD11c, CD23, CD123, CD68, Allograft Inflammatory factor 1[AIF-1]) and PD-L1 with respect to IDO expression and localization in melanoma brain metastases but also in matched metastases at extracranial sites to correlate intra- and interpatient data with therapy response and survival. Comparative tissue analysis identified macrophages/microglia as the major source of IDO expression in melanoma brain metastases. In contrast to the tumor infiltrating lymphocytes, melanoma cells per se exhibited low IDO expression levels paralleled by cell surface presentation of PD-L1 in intracranial metastases. Absolute numbers and pattern of IDO-expressing cells in metastases of the brain correlated with recruitment and localization of CD8+ T cells, implicating dynamic impact on the regulation of T cell function in the brain parenchyma. However, paired analysis of matched intra- and extracranial metastases identified significantly lower fractions of cytotoxic CD8+ T cells in intracranial metastases while all other immune cell populations remain unchanged. In line with the already established clinical benefit for PD-L1 expression in extracranial melanoma metastases, Kaplan-Meier analyses correlated PD-L1 expression in brain metastases with favorable outcome in advanced melanoma patients undergoing immune checkpoint therapy. In summary, our data provide new insights into the landscape of immunosuppressive factors in melanoma brain metastases that may be useful in the implication of novel therapeutic strategies for patients undergoing cancer immunotherapy.
Collapse
Affiliation(s)
- Dayana Herrera-Rios
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University Duisburg-Essen, Essen, Germany.,German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen, Germany
| | - Sadaf S Mughal
- Division of Applied Bioinfomatics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sarah Teuber-Hanselmann
- Medical Faculty, West German Cancer Center, Institute of Neuropathology, University Duisburg-Essen, Essen, Germany
| | - Daniela Pierscianek
- German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen, Germany.,Department of Neurosurgery, Medical Faculty, West German Cancer Center, University Duisburg-Essen, Essen, Germany
| | - Antje Sucker
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University Duisburg-Essen, Essen, Germany.,German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen, Germany
| | - Philipp Jansen
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University Duisburg-Essen, Essen, Germany.,German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen, Germany
| | - Tobias Schimming
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University Duisburg-Essen, Essen, Germany.,German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen, Germany
| | - Joachim Klode
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University Duisburg-Essen, Essen, Germany.,German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen, Germany
| | - Julia Reifenberger
- Department of Dermatology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Jörg Felsberg
- Medical Faculty, Institute of Neuropathology, Heinrich Heine University, Düsseldorf, Germany
| | - Kathy Keyvani
- Medical Faculty, West German Cancer Center, Institute of Neuropathology, University Duisburg-Essen, Essen, Germany
| | - Benedikt Brors
- Division of Applied Bioinfomatics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ulrich Sure
- German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen, Germany.,Department of Neurosurgery, Medical Faculty, West German Cancer Center, University Duisburg-Essen, Essen, Germany
| | - Guido Reifenberger
- German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen, Germany.,Medical Faculty, Institute of Neuropathology, Heinrich Heine University, Düsseldorf, Germany
| | - Dirk Schadendorf
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University Duisburg-Essen, Essen, Germany.,German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen, Germany
| | - Iris Helfrich
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University Duisburg-Essen, Essen, Germany.,German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen, Germany
| |
Collapse
|
235
|
Majem M, García-Martínez E, Martinez M, Muñoz-Couselo E, Rodriguez-Abreu D, Alvarez R, Arance A, Berrocal A, de la Cruz-Merino L, Lopez-Martin JA. SEOM clinical guideline for the management of immune-related adverse events in patients treated with immune checkpoint inhibitors (2019). Clin Transl Oncol 2020; 22:213-222. [PMID: 31993963 DOI: 10.1007/s12094-019-02273-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 12/16/2019] [Indexed: 12/17/2022]
Abstract
The use of immune checkpoint inhibitors has emerged as an effective treatment option for patients with several tumor types. By increasing the activity of the immune system, they can induce inflammatory side effects, which are often termed immune-related adverse events. These are pathophysiologically unique toxicities, compared with those from other anticancer therapies. In addition, the spectrum of the target organs is very broad. Immune-inflammatory adverse events can be life threatening. Prompt diagnosis and pharmacological intervention are instrumental to avoid progression to severe manifestations. Consequently, clinicians require new skills to successfully diagnose and manage these events. These SEOM guidelines have been developed with the consensus of ten medical oncologists. Relevant studies published in peer-review journals were used for the guideline elaboration. The Infectious Diseases Society of America grading system was used to assign levels of evidence and grades of recommendation.
Collapse
Affiliation(s)
- M Majem
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, c/Sant Antoni Maria Claret 167, 08025, Barcelona, Spain.
- Spanish Group for Cancer Immuno-Biotherapy, GÉTICA, Madrid, Spain.
| | - E García-Martínez
- Department of Medical Oncology and Hematology, Hospital Universitario Morales Meseguer, Murcia, Spain
- Spanish Group for Cancer Immuno-Biotherapy, GÉTICA, Madrid, Spain
| | - M Martinez
- Department of Medical Oncology, Hospital del Mar, Barcelona, Spain
| | - E Muñoz-Couselo
- Department of Medical Oncology, Melanoma and Other Skin Tumors Unit, Vall d'Hebron Hospita, Vall d'Hebron Institute of Oncology VHIO, Barcelona, Spain
| | - D Rodriguez-Abreu
- Department of Medical Oncology, C.H.U. Insular-Materno Infantil de Gran Canaria, Las Palmas, Spain
- Spanish Group for Cancer Immuno-Biotherapy, GÉTICA, Madrid, Spain
| | - R Alvarez
- Department of Medical Oncology, Hospital Virgen de la Salud, Toledo, Spain
| | - A Arance
- Department of Medical Oncology, Hospital Clínic, Barcelona, Spain
- Spanish Group for Cancer Immuno-Biotherapy, GÉTICA, Madrid, Spain
| | - A Berrocal
- Department of Medical Oncology, Consorcio Hospital General Universitario de Valencia, Valencia, Spain
| | - L de la Cruz-Merino
- Clinical Oncology Department, Hospital Universitario Virgen Macarena, Seville, Spain
- Medicine Department, Universidad de Sevilla, Sevilla, Spain
- Spanish Group for Cancer Immuno-Biotherapy, GÉTICA, Madrid, Spain
| | - J A Lopez-Martin
- Department of Medical Oncology, Hospital Universitario, 12 de Octubre, Madrid, Spain
- Clinical and Translational Oncology, Instituto de Investigación Sanitaria Hospital, 12 de Octubre, Madrid, Spain
- Spanish Group for Cancer Immuno-Biotherapy, GÉTICA, Madrid, Spain
| |
Collapse
|
236
|
Klepsch V, Pommermayr M, Humer D, Brigo N, Hermann-Kleiter N, Baier G. Targeting the orphan nuclear receptor NR2F6 in T cells primes tumors for immune checkpoint therapy. Cell Commun Signal 2020; 18:8. [PMID: 31937317 PMCID: PMC6961368 DOI: 10.1186/s12964-019-0454-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 10/04/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND NR2F6 has been proposed as an alternative cancer immune checkpoint in the effector T cell compartment. However, a realistic assessment of the in vivo therapeutic potential of NR2F6 requires acute depletion. METHODS Employing primary T cells isolated from Cas9-transgenic mice for electroporation of chemically synthesized sgRNA, we established a CRISPR/Cas9-mediated acute knockout protocol of Nr2f6 in primary mouse T cells. RESULTS Analyzing these Nr2f6CRISPR/Cas9 knockout T cells, we reproducibly observed a hyper-reactive effector phenotype upon CD3/CD28 stimulation in vitro, highly reminiscent to Nr2f6-/- T cells. Importantly, CRISPR/Cas9-mediated Nr2f6 ablation prior to adoptive cell therapy (ACT) of autologous polyclonal T cells into wild-type tumor-bearing recipient mice in combination with PD-L1 or CTLA-4 tumor immune checkpoint blockade significantly delayed MC38 tumor progression and induced superior survival, thus further validating a T cell-inhibitory function of NR2F6 during tumor progression. CONCLUSIONS These findings indicate that Nr2f6CRISPR/Cas9 knockout T cells are comparable to germline Nr2f6-/- T cells, a result providing an independent confirmation of the immune checkpoint function of lymphatic NR2F6. Taken together, CRISPR/Cas9-mediated acute Nr2f6 gene ablation in primary mouse T cells prior to ACT appeared feasible for potentiating established PD-L1 and CTLA-4 blockade therapies, thereby pioneering NR2F6 inhibition as a sensitizing target for augmented tumor regression. Video abstract.
Collapse
Affiliation(s)
- Victoria Klepsch
- Division of Translational Cell Genetics, Medical University of Innsbruck, Peter Mayr Str. 1a, A-6020, Innsbruck, Austria.
| | - Maria Pommermayr
- Division of Translational Cell Genetics, Medical University of Innsbruck, Peter Mayr Str. 1a, A-6020, Innsbruck, Austria
| | - Dominik Humer
- Division of Translational Cell Genetics, Medical University of Innsbruck, Peter Mayr Str. 1a, A-6020, Innsbruck, Austria
| | - Natascha Brigo
- Division of Translational Cell Genetics, Medical University of Innsbruck, Peter Mayr Str. 1a, A-6020, Innsbruck, Austria
- Present address: Department of Internal Medicine II, Medical University of Innsbruck, Anichstraße 35, A-6020, Innsbruck, Austria
| | - Natascha Hermann-Kleiter
- Division of Translational Cell Genetics, Medical University of Innsbruck, Peter Mayr Str. 1a, A-6020, Innsbruck, Austria
| | - Gottfried Baier
- Division of Translational Cell Genetics, Medical University of Innsbruck, Peter Mayr Str. 1a, A-6020, Innsbruck, Austria.
| |
Collapse
|
237
|
Su Z, Zhou L, Xue J, Lu Y. Integration of stereotactic radiosurgery or whole brain radiation therapy with immunotherapy for treatment of brain metastases. Chin J Cancer Res 2020; 32:448-466. [PMID: 32963458 PMCID: PMC7491544 DOI: 10.21147/j.issn.1000-9604.2020.04.03] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The prognosis of brain metastases (BM) is traditionally poor. BM are mainly treated by local radiotherapy, including stereotactic radiosurgery (SRS) or whole brain radiation therapy (WBRT). Recently, immunotherapy (i.e., immune checkpoint inhibitors, ICI) has demonstrated a survival advantage in multiple malignancies commonly associated with BM. Individually, radiotherapy and ICI both treat BM efficiently; hence, their combination seems logical. In this review, we summarize the existing preclinical and clinical evidence that supports the applicability of radiotherapy as a sensitizer of ICI for BM. Further, we discuss the optimal timing at which radiotherapy and ICI should be administered and review the safety of the combination therapy. Data from a few clinical studies suggest that combining SRS or WBRT with ICI simultaneously rather than consecutively potentially enhances brain abscopal-like responses and survival. However, there is a lack of conclusion about the definition of "simultaneous"; the cumulative toxic effect of the combined therapies also requires further study. Thus, ongoing and planned prospective trials are needed to further explore and validate the effect, safety, and optimal timing of the combination of immunotherapy with radiotherapy for patients with BM.
Collapse
Affiliation(s)
- Zhou Su
- Department of Thoracic Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.,Department of Oncology, Sichuan Mianyang 404 Hospital, Mianyang 621000, China
| | - Lin Zhou
- Department of Thoracic Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jianxin Xue
- Department of Thoracic Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - You Lu
- Department of Thoracic Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
238
|
Coleman EL, Olamiju B, Leventhal JS. The life-threatening eruptions of immune checkpoint inhibitor therapy. Clin Dermatol 2020; 38:94-104. [DOI: 10.1016/j.clindermatol.2019.10.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
239
|
Abstract
Tumor development is characterized by the accumulation of mutational and epigenetic changes that transform normal cells and survival pathways into self-sustaining cells capable of untrammeled growth. Although multiple modalities including surgery, radiation, and chemotherapy are available for the treatment of cancer, the benefits conferred are often limited. The immune system is capable of specific, durable, and adaptable responses. However, cancers hijack immune mechanisms such as negative regulatory checkpoints that have evolved to limit inflammatory and immune responses to thwart effective antitumor immunity. The development of monoclonal antibodies against inhibitory receptors expressed by immune cells has produced durable responses in a broad array of advanced malignancies and heralded a new dawn in the cancer armamentarium. However, these remarkable responses are limited to a minority of patients and indications, highlighting the need for more effective and novel approaches. Preclinical and clinical studies with immune checkpoint blockade are exploring the therapeutic potential antibody-based therapy targeting multiple inhibitory receptors. In this chapter, we discuss the current understanding of the structure, ligand specificities, function, and signaling activities of various inhibitory receptors. Additionally, we discuss the current development status of various immune checkpoint inhibitors targeting these negative immune receptors and highlight conceptual gaps in knowledge.
Collapse
|
240
|
Luís R, Brito C, Pojo M. Melanoma Metabolism: Cell Survival and Resistance to Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1219:203-223. [PMID: 32130701 DOI: 10.1007/978-3-030-34025-4_11] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cutaneous melanoma is one of the most aggressive types of cancer, presenting the highest potential to form metastases, both locally and distally, which are associated with high death rates of melanoma patients. A high somatic mutation burden is characteristic of these tumours, with most common oncogenic mutations occurring in the BRAF, NRAS and NF1 genes. These intrinsic oncogenic pathways contribute to the metabolic switch between glycolysis and oxidative phosphorylation metabolisms of melanoma, facilitating tumour progression and resulting in a high plasticity and adaptability to unfavourable conditions. Moreover, melanoma microenvironment can influence its own metabolism and reprogram several immune cell subset functions, enabling melanoma to evade the immune system. The knowledge of the biology, molecular alterations and microenvironment of melanoma has led to the development of new targeted therapies and the improvement of patient care. In this work, we reviewed the impact of melanoma metabolism in the resistance to BRAF and MEK inhibitors and immunotherapies, emphasizing the requirement to evaluate metabolic alterations upon development of novel therapeutic approaches. Here we summarized the current understanding of the impact of metabolic processes in melanomagenesis, metastasis and microenvironment, as well as the involvement of metabolic pathways in the immune modulation and resistance to targeted and immunocheckpoint therapies.
Collapse
Affiliation(s)
- Rafael Luís
- Unidade de Investigação em Patobiologia Molecular (UIPM), Instituto Português de Oncologia de Lisboa Francisco Gentil E.P.E, Lisbon, Portugal
| | - Cheila Brito
- Unidade de Investigação em Patobiologia Molecular (UIPM), Instituto Português de Oncologia de Lisboa Francisco Gentil E.P.E, Lisbon, Portugal
| | - Marta Pojo
- Unidade de Investigação em Patobiologia Molecular (UIPM), Instituto Português de Oncologia de Lisboa Francisco Gentil E.P.E, Lisbon, Portugal
| |
Collapse
|
241
|
Pu D, Yin L, Zhou Y, Li W, Huang L, Cai L, Zhou Q. Safety and efficacy of immune checkpoint inhibitors in patients with HBV/HCV infection and advanced-stage cancer: A systematic review. Medicine (Baltimore) 2020; 99:e19013. [PMID: 32000444 PMCID: PMC7004734 DOI: 10.1097/md.0000000000019013] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Cancer patients with hepatitis B or C virus (HBV/HCV) infection are commonly seen in clinical practice, however, the data of safety and efficacy of immune checkpoint inhibitors (ICIs) among them are sparse, because active HBV/HCV infected patients were generally excluded by clinical trials and the correlation between previous infection and treatment-related adverse events was rarely reported. This review is the first to summarize the results on the safety and efficacy of immune checkpoint inhibitors (ICIs) in HBV/HCV infected cancer patients. METHOD We searched literature and conference abstracts in PubMed and Embase followed the PRISMA guideline, using the keywords hepatitis B, hepatitis C, immune checkpoint inhibitor, ipilimumab, nivolumab, pembrolizumab, atezolizumab, durvalumab, avelumab, tremelimumab. Studies described patients with HBV/HCV infection treated with ICIs for advanced stage cancer were included. FINDINGS One hundred eighty six patients were identified from 14 articles (8 case reports, 4 case series, 2 trials). Eighty nine patients had HBV infection and 98 had HCV infection (1 both had HBV and HCV). The majority of patients were treated with PD-1 inhibitor monotherapy (140 of 186, 75.3%) and anti-CTLA-4 monotherapy (36 of 186, 19.4%). No treatment-related death was reported. The incidence of grade 3 or 4 hepatic transaminase elevating (HTE) in HBV and HCV infected patients were 3.4% (3/89) and 17.3% (17/98), respectively. 2.8% patients without antivirus therapy experienced virus load increasing, and 1.9% presented virus-related hepatitis. In terms of efficacy, 22 of 118 (18.6%) patients with liver cancer, 11 of 34 (32.4%) with melanoma, 1 of 6 (16.7%) with NSCLC showed objective responses (CR and PR) to ICIs in spite of lines of therapies. CONCLUSION ICIs is considered to be safe and effective in advanced cancer patients with hepatitis B or C infection, but still has possibilities to reactive hepatitis virus due to uncertain mechanisms. We recommend that those with viral hepatitis be monitored closely and treated with antiviral therapy if indicated before or during ICIs treatment.
Collapse
Affiliation(s)
- Dan Pu
- Lung Cancer Center of West China Hospital
| | - Liyuan Yin
- Lung Cancer Center of West China Hospital
| | - Yuwen Zhou
- Department of Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Wen Li
- Lung Cancer Center of West China Hospital
| | - Lin Huang
- Lung Cancer Center of West China Hospital
| | - Liang Cai
- Lung Cancer Center of West China Hospital
| | | |
Collapse
|
242
|
de Germay S, Bagheri H, Despas F, Rousseau V, Montastruc F. Abatacept in rheumatoid arthritis and the risk of cancer: a world observational post-marketing study. Rheumatology (Oxford) 2019; 59:2360-2367. [DOI: 10.1093/rheumatology/kez604] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 11/07/2019] [Indexed: 12/16/2022] Open
Abstract
Abstract
Objectives
We aimed to investigate whether abatacept used in patients for RA was associated with an increased risk of reporting overall cancer and specific cancers, including breast, lung, lymphoma, melanoma and non-melanoma skin cancer when compared with other biologic DMARDs (bDMARDs).
Methods
We performed an observational study within VigiBase, the World Health Organization’s global database of individual case safety reports, from 2007 to 2017 to compare the cases of cancer reported in RA patients exposed to abatacept with those reported in RA patients exposed to other bDMARDs. We conducted disproportionality analyses allowing the estimation of reporting odds ratios (RORs) with 95% CIs of the exposure odds among spontaneous reporting of cancers to the exposure odds among other reported adverse effects.
Results
We identified 15 846 adverse effects reported in RA patients who received abatacept and 290 568 adverse effects reported in RA patients treated with other bDMARDs. Compared with other bDMARDs, the use of abatacept was not associated with an increased risk of reporting cancer overall [ROR 0.98 (95% CI 0.91, 1.05)]. Analyses by specific cancer sites showed a significantly increased ROR for melanoma [1.58 (95% CI 1.17, 2.08)], but not for other specific cancer sites.
Conclusion
Compared with other bDMARDs, exposure to abatacept in RA patients was only significantly associated with an increased risk of reporting melanoma. This increased risk is consistent with the properties of abatacept (CTLA-4 agonist) since it has an opposite action than ipilimumab, an antibody that blocks CTLA-4 and is approved for the treatment of malignant melanoma.
Trial registration
ClinicalTrials.gov (http://clinicaltrials.gov), NCT03980639.
Collapse
Affiliation(s)
- Sibylle de Germay
- Department of Medical and Clinical Pharmacology, Centre of Pharmacovigilance and Pharmacoepidemiology, Toulouse University Hospital, Faculty of Medicine, Toulouse, France
- INSERM, UMR 1027 Pharmacoepidemiology, Assessment of Drug Utilization and Drug Safety, CIC 1426 - University Paul Sabatier Toulouse, Toulouse, France
| | - Haleh Bagheri
- Department of Medical and Clinical Pharmacology, Centre of Pharmacovigilance and Pharmacoepidemiology, Toulouse University Hospital, Faculty of Medicine, Toulouse, France
- INSERM, UMR 1027 Pharmacoepidemiology, Assessment of Drug Utilization and Drug Safety, CIC 1426 - University Paul Sabatier Toulouse, Toulouse, France
| | - Fabien Despas
- Department of Medical and Clinical Pharmacology, Centre of Pharmacovigilance and Pharmacoepidemiology, Toulouse University Hospital, Faculty of Medicine, Toulouse, France
- INSERM, UMR 1027 Pharmacoepidemiology, Assessment of Drug Utilization and Drug Safety, CIC 1426 - University Paul Sabatier Toulouse, Toulouse, France
- Clinical Unit of Cancer Pharmacology, Toulouse University Hospital, Faculty of Medicine, Toulouse, France
| | - Vanessa Rousseau
- Department of Medical and Clinical Pharmacology, Centre of Pharmacovigilance and Pharmacoepidemiology, Toulouse University Hospital, Faculty of Medicine, Toulouse, France
- INSERM, UMR 1027 Pharmacoepidemiology, Assessment of Drug Utilization and Drug Safety, CIC 1426 - University Paul Sabatier Toulouse, Toulouse, France
| | - François Montastruc
- Department of Medical and Clinical Pharmacology, Centre of Pharmacovigilance and Pharmacoepidemiology, Toulouse University Hospital, Faculty of Medicine, Toulouse, France
- INSERM, UMR 1027 Pharmacoepidemiology, Assessment of Drug Utilization and Drug Safety, CIC 1426 - University Paul Sabatier Toulouse, Toulouse, France
| |
Collapse
|
243
|
Chen C, Dorado Garcia H, Scheer M, Henssen AG. Current and Future Treatment Strategies for Rhabdomyosarcoma. Front Oncol 2019; 9:1458. [PMID: 31921698 PMCID: PMC6933601 DOI: 10.3389/fonc.2019.01458] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 12/05/2019] [Indexed: 12/31/2022] Open
Abstract
Rhabdomyosarcoma (RMS) is the most common soft tissue sarcoma in children, and can be subcategorized histologically and/or based on PAX-FOXO1 fusion gene status. Over the last four decades, there have been no significant improvements in clinical outcomes for advanced and metastatic RMS patients, underscoring a need for new treatment options for these groups. Despite significant advancements in our understanding of the genomic landscape and underlying biological mechanisms governing RMS that have informed the identification of novel therapeutic targets, development of these therapies in clinical trials has lagged far behind. In this review, we summarize the current frontline multi-modality therapy for RMS according to pediatric protocols, highlight emerging targeted therapies and immunotherapies identified by preclinical studies, and discuss early clinical trial data and the implications they hold for future clinical development.
Collapse
Affiliation(s)
- Celine Chen
- Department of Pediatric Oncology/Hematology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Heathcliff Dorado Garcia
- Department of Pediatric Oncology/Hematology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Monika Scheer
- Pediatrics 5, Klinikum Stuttgart, Olgahospital, Stuttgart, Germany
| | - Anton G. Henssen
- Department of Pediatric Oncology/Hematology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Experimental and Clinical Research Center (ECRC) of the MDC and Charité Berlin, Berlin, Germany
| |
Collapse
|
244
|
Abstract
Immunotherapy is distinct from traditional chemotherapy in that it acts on immune cells rather than cancer cells themselves. Monoclonal antibodies targeting immune checkpoints on T cells - CTLA-4 and PD-1 - and PD-L1 on the cells of immune microenvironment are now approved for clinical use in several solid tumors and hematological malignancies. This article provides a general overview of the use of checkpoint inhibitors in hematologic malignancies with a special focus in acute myeloid leukemia.
Collapse
Affiliation(s)
- Arnab Ghosh
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Pere Barba
- Hematology Department, Vall d'Hebron University Hospital-Universitat Autónoma de Barcelona, Barcelona, Spain
| | - Miguel-Angel Perales
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
245
|
Bermudez MV, Papa S. Setting the scene - a future 'epidemic' of immune-related adverse events in association with checkpoint inhibitor therapy. Rheumatology (Oxford) 2019; 58:vii1-vii6. [PMID: 31816083 PMCID: PMC6900909 DOI: 10.1093/rheumatology/kez402] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 07/25/2019] [Indexed: 12/19/2022] Open
Abstract
Cancer immune therapy with checkpoint inhibitors (CPIs) has changed the landscape of treatment for a growing number of indications. These drugs are associated with a specific mechanism of action that has profound implications for both immunology and inflammatory disease. This article looks to set the scene covering the history of CPI therapy to date and outlining the likely future developments.
Collapse
Affiliation(s)
- Maria V Bermudez
- Centre for Inflammation Biology and Cancer Immunology, King's College London, London, UK
| | - Sophie Papa
- Division of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK.,Department of Medical Oncology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| |
Collapse
|
246
|
Dancsok AR, Setsu N, Gao D, Blay JY, Thomas D, Maki RG, Nielsen TO, Demicco EG. Expression of lymphocyte immunoregulatory biomarkers in bone and soft-tissue sarcomas. Mod Pathol 2019; 32:1772-1785. [PMID: 31263176 DOI: 10.1038/s41379-019-0312-y] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 05/29/2019] [Accepted: 05/30/2019] [Indexed: 12/18/2022]
Abstract
Despite advances in our understanding of the underlying molecular drivers of sarcomas, few treatments are available with proven benefit for advanced metastatic sarcomas. Immunotherapy has value in this setting for some types of cancers, but sarcomas, with their multiplicity of rare types, have not been characterized in detail for their expression of targetable immune biomarkers. This study provides the most systematic evaluation to date of tumor-infiltrating lymphocytes and immune checkpoint biomarker expression in sarcomas. We examined by morphology and immunohistochemistry 1072 sarcoma specimens representing 22 types, in addition to 236 benign bone and soft-tissue tumors. Genomically-complex sarcoma types-those driven by mutations and/or copy-number alterations-had much higher numbers of tumor-infiltrating lymphocytes than translocation-associated sarcomas. Prior exposure to radiotherapy was associated with increased immune infiltrates. Higher lymphocytic infiltration was associated with better overall survival among the non-translocation-associated sarcomas. Expression of PD-1 and CD56 were associated with worse overall survival. LAG-3 and TIM-3, two emerging immune checkpoints, were frequently expressed in most sarcoma types. Indeed, most cases positive for PD-(L)1 coexpressed one or both of these novel biomarkers, providing a potential rationale in support for trials targeting LAG-3 and/or TIM-3 in conjunction with PD-1 inhibition.
Collapse
Affiliation(s)
- Amanda R Dancsok
- Department of Pathology and Laboratory Medicine, Vancouver Coastal Health Research Institute and University of British Columbia, Vancouver, BC, Canada
| | - Nokitaka Setsu
- Department of Anatomic Pathology, Graduate School of Medical Science, Kyushu University, Fukuoka, 812-8582, Japan
| | - Dongxia Gao
- Department of Pathology and Laboratory Medicine, Vancouver Coastal Health Research Institute and University of British Columbia, Vancouver, BC, Canada
| | - Jean-Yves Blay
- Department of Medical Oncology, Centre Léon Bérard and University Claude Bernard Lyon, Lyon, France
| | - David Thomas
- The Kinghorn Cancer Centre and Cancer Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Robert G Maki
- Northwell Health Monter Cancer Center and Cold Spring Harbor Laboratory, Lake Success, New York, NY, USA
| | - Torsten O Nielsen
- Department of Pathology and Laboratory Medicine, Vancouver Coastal Health Research Institute and University of British Columbia, Vancouver, BC, Canada.
| | - Elizabeth G Demicco
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
247
|
Lu J, Li L, Lan Y, Liang Y, Meng H. Immune checkpoint inhibitor-associated pituitary-adrenal dysfunction: A systematic review and meta-analysis. Cancer Med 2019; 8:7503-7515. [PMID: 31679184 PMCID: PMC6912062 DOI: 10.1002/cam4.2661] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 08/25/2019] [Accepted: 10/14/2019] [Indexed: 12/15/2022] Open
Abstract
With the growing use of immune checkpoint inhibitors (ICIs), case reports of rare yet life-threatening pituitary-adrenal dysfunctions, particularly for hypopituitarism, are increasingly being published. In this analysis, we focus on these events by including the most recent publications and reports from early phase I/II and phase III clinical trials and comparing the incidence and risks across different ICI regimens. PubMed, Embase, and the Cochrane Library were systematically searched from inception to April 2019 for clinical trials that reported on pituitary-adrenal dysfunction. The rates of events, odds ratios (ORs), and 95% confidence intervals (CIs) were obtained using random effects meta-analysis. The analyses included data from 160 trials involving 40 432 participants. The rate was 2.43% (95% CI, 1.73%-3.22%) for all-grade adrenal insufficiency and 3.25% (95% CI, 2.15%-4.51%) for hypophysitis. Compared with the placebo or other therapeutic regimens, ICI agents were associated with a higher incidence of serious-grade adrenal insufficiency (OR 3.19, 95% CI, 1.84 to 5.54) and hypophysitis (OR 4.77, 95% CI, 2.60 to 8.78). Among 71 serious-grade hypopituitarism instances in 12 336 patients, there was a significant association between ICIs and hypopituitarism (OR 3.62, 95% CI, 1.86 to 7.03). Substantial heterogeneity was noted across the studies for the rates of these events, which in part was attributable to the different types of ICIs and varied phases of the clinical trials. Although the rates of these events were low, the risk was increased following ICI-based treatment, particularly for CTLA-4 inhibitors, which were associated with a higher incidence of pituitary-adrenal dysfunction than PD-1/PD-L1 inhibitors.
Collapse
Affiliation(s)
- Jingli Lu
- Department of PharmacyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Henan Key Laboratory of Precision Clinical PharmacyZhengzhou UniversityZhengzhouHenanChina
| | - Lulu Li
- Department of PharmacyWuhan No.1 HospitalWuhanHubeiChina
| | - Yan Lan
- Department of PharmacyHuangshi Center HospitalHuangshiHubeiChina
| | - Yan Liang
- Department of PharmacyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Henan Key Laboratory of Precision Clinical PharmacyZhengzhou UniversityZhengzhouHenanChina
| | - Haiyang Meng
- Department of PharmacyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Henan Key Laboratory of Precision Clinical PharmacyZhengzhou UniversityZhengzhouHenanChina
| |
Collapse
|
248
|
Kamath SD, Kalyan A, Kircher S, Nimeiri H, Fought AJ, Benson A, Mulcahy M. Ipilimumab and Gemcitabine for Advanced Pancreatic Cancer: A Phase Ib Study. Oncologist 2019; 25:e808-e815. [PMID: 31740568 DOI: 10.1634/theoncologist.2019-0473] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 10/16/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) remains resistant to chemotherapy and immunotherapy individually because of its desmoplastic stroma and immunosuppressive tumor microenvironment. Synergizing cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) immune checkpoint blockade with chemotherapy could overcome these barriers. Here we present results of a phase Ib trial combining ipilimumab and gemcitabine in advanced PDAC. MATERIALS AND METHODS This was a single-institution study with a 3 + 3 dose-escalation design. The primary objective was to determine the maximum tolerated dose (MTD). Secondary objectives included determining the toxicity profile, objective response rate (ORR), median progression-free survival (PFS), and overall survival (OS). RESULTS Twenty-one patients were enrolled, 13 during dose escalation and 8 at the MTD. The median age was 66 years, 62% were female, 95% had stage IV disease, and 67% had received at least one prior line of therapy. The primary objective to establish the MTD was achieved at doses of ipilimumab 3 mg/kg and gemcitabine 1,000 mg/m2 . The most common grade 3 or 4 adverse events were anemia (48%), leukopenia (48%), and neutropenia (43%). The ORR was 14% (3/21), and seven patients had stable disease. Median response duration for the three responders was 11 months, with one response duration of 19.8 months. Median PFS was 2.78 months (95% confidence interval [CI], 1.61-4.83 months), and median OS was 6.90 months (95% CI, 2.63-9.57 months). CONCLUSION Gemcitabine and ipilimumab is a safe and tolerable regimen for PDAC with a similar response rate to gemcitabine alone. As in other immunotherapy trials, responses were relatively durable in this study. IMPLICATIONS FOR PRACTICE Gemcitabine and ipilimumab is a safe and feasible regimen for treating advanced pancreatic cancer. Although one patient in this study had a relatively durable response of nearly 20 months, adding ipilimumab to gemcitabine does not appear to be more effective than gemcitabine alone in advanced pancreatic cancer.
Collapse
Affiliation(s)
- Suneel D Kamath
- Division of Hematology and Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, USA
| | - Aparna Kalyan
- Division of Hematology and Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Developmental Therapeutics Program, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, USA
| | - Sheetal Kircher
- Division of Hematology and Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, USA
| | - Halla Nimeiri
- Division of Hematology and Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, USA
| | - Angela J Fought
- Division of Biostatistics, Department of Preventative Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Al Benson
- Division of Hematology and Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, USA
| | - Mary Mulcahy
- Division of Hematology and Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
249
|
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
|
250
|
Simeone E, Grimaldi AM, Festino L, Trojaniello C, Vitale MG, Vanella V, Palla M, Ascierto PA. Immunotherapy in metastatic melanoma: a novel scenario of new toxicities and their management. Melanoma Manag 2019; 6:MMT30. [PMID: 31871619 PMCID: PMC6920742 DOI: 10.2217/mmt-2019-0005] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 07/18/2019] [Indexed: 12/13/2022] Open
Abstract
Checkpoint inhibitors can cause an imbalance in immune tolerance that may clinically manifest as immune-related adverse events (irAEs). These events may involve many organs and tissues, including the skin, gastrointestinal (GI) tract, liver, endocrine system, kidneys, central nervous system (CNS), eyes and lungs. The incidence of irAEs appears to be lower with anti-programmed death antigen-1/programmed death antigen-ligand-1 agents than with the anti-cytotoxic T-lymphocyte-associated protein-4 antibody ipilimumab. Combined immunotherapy does not appear to be associated with novel safety signals compared with monotherapy, but more organs may be involved. Increased experience and the use of algorithms for the most common irAEs have resulted in severe toxicity and related deaths being reduced. However, continuous vigilance, especially regarding less common events, is needed to better characterize the wide spectrum of clinical manifestations.
Collapse
Affiliation(s)
- Ester Simeone
- Unit of Melanoma, Cancer Immunotherapy & Innovative Therapies Unit – Istituto Nazionale Tumori Fondazione ‘G. Pascale,’ IRCCS, 80131, Napoli, Italy
| | - Antonio M Grimaldi
- Unit of Melanoma, Cancer Immunotherapy & Innovative Therapies Unit – Istituto Nazionale Tumori Fondazione ‘G. Pascale,’ IRCCS, 80131, Napoli, Italy
| | - Lucia Festino
- Unit of Melanoma, Cancer Immunotherapy & Innovative Therapies Unit – Istituto Nazionale Tumori Fondazione ‘G. Pascale,’ IRCCS, 80131, Napoli, Italy
| | - Claudia Trojaniello
- Unit of Melanoma, Cancer Immunotherapy & Innovative Therapies Unit – Istituto Nazionale Tumori Fondazione ‘G. Pascale,’ IRCCS, 80131, Napoli, Italy
| | - Maria G Vitale
- Unit of Melanoma, Cancer Immunotherapy & Innovative Therapies Unit – Istituto Nazionale Tumori Fondazione ‘G. Pascale,’ IRCCS, 80131, Napoli, Italy
| | - Vito Vanella
- Unit of Melanoma, Cancer Immunotherapy & Innovative Therapies Unit – Istituto Nazionale Tumori Fondazione ‘G. Pascale,’ IRCCS, 80131, Napoli, Italy
| | - Marco Palla
- Unit of Melanoma, Cancer Immunotherapy & Innovative Therapies Unit – Istituto Nazionale Tumori Fondazione ‘G. Pascale,’ IRCCS, 80131, Napoli, Italy
| | - Paolo A Ascierto
- Unit of Melanoma, Cancer Immunotherapy & Innovative Therapies Unit – Istituto Nazionale Tumori Fondazione ‘G. Pascale,’ IRCCS, 80131, Napoli, Italy
| |
Collapse
|